1
|
Thakur A, Singh DK, Hart KD, Kis E, Gáborik Z, Denton TT, Clarke JD, Paine MF, Prasad B. From Discovery to Translation: Endogenous Substrates of OAT1 and OAT3 as Clinical Biomarkers for Renal Secretory Function. Clin Pharmacol Ther 2025. [PMID: 40401788 DOI: 10.1002/cpt.3719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
The recent ICH M12 guidance on Drug Interaction Studies encourages the use of alternate approaches for predicting drug-drug interaction (DDI) potential of new chemical entities. One approach involves the use of endogenous substrates of drug metabolizing enzymes and transporters (DMET) as biomarkers, which can assess the inhibitory potential of new chemical entities towards DMET during Phase 1 clinical studies. Thus, biomarkers could potentially eliminate the need for dedicated DDI studies with exogenous probe substrates. Metabolomics, in conjunction with in vitro and/or in vivo preclinical models or clinical studies, can be used for biomarker discovery. We developed and applied a novel metabolomics-based DMET biomarker discovery (MDBD) approach to identify and qualify biomarkers of renal organic anion transporter 1 (OAT1) and OAT3. Untargeted metabolomics of pooled plasma and urine samples (n = 16) from a pharmacokinetic DDI study using the OAT1/3 inhibitor, probenecid, yielded 153 features identified as putative OAT1/3 biomarkers. Subsequently, in vitro transporter uptake assays using processed urine samples confirmed 57 of these features as OAT1 and/or OAT3 substrates. Finally, 23 features were clinically validated as OAT1/3 biomarkers through a detailed pharmacokinetic analysis (0-24 h) of plasma and urine samples (n = 4). These biomarkers, either alone or as part of a panel, can predict OAT1/3-mediated DDIs and interindividual variability in the renal secretory clearance of organic anions across different populations, thereby enabling translational utility in clinical settings. The novel MDBD approach can be extended to discover biomarkers of enzymes and other transporters.
Collapse
Affiliation(s)
- Aarzoo Thakur
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Dilip K Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Katherine D Hart
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Emese Kis
- Charles River Laboratories Hungary Kft, Budapest, Hungary
| | | | - Travis T Denton
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
- Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, Washington, USA
- Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, Washington, USA
| | - John D Clarke
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Mary F Paine
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| |
Collapse
|
2
|
Thakur A, Singh DK, Hart KD, Kis E, Gáborik Z, Denton TT, Clarke JD, Paine MF, Prasad B. From discovery to translation: Endogenous substrates of OAT1 and OAT3 as clinical biomarkers for renal secretory function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636675. [PMID: 39975069 PMCID: PMC11838602 DOI: 10.1101/2025.02.05.636675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The recent ICH M12 guidance on Drug Interaction Studies encourages the use of alternate approaches for predicting drug-drug interaction (DDI) potential of new chemical entities. One approach involves biomarkers, which are endogenous substrates of drug metabolizing enzymes and transporters (DMET) and can be used to assess the inhibitory potential of new chemical entities during Phase 1 clinical studies. Thus, biomarkers could potentially eliminate the need for dedicated DDI studies with exogenous probe substrates. Metabolomics, in conjunction with in vitro and/or in vivo preclinical models or clinical studies, can be used for biomarker discovery. We developed and applied a novel metabolomics-based DMET biomarker discovery (MDBD) approach to identify and qualify biomarkers of renal organic anion transporter 1 (OAT1) and OAT3. Untargeted metabolomics of pooled plasma and urine samples from a pharmacokinetic DDI study using the OAT1/3 inhibitor, probenecid, yielded 153 features identified as putative OAT1/3 biomarkers. Subsequently, in vitro transporter uptake assays using processed urine samples confirmed 57 of these features as OAT1 and/or OAT3 substrates. Finally, 23 features were clinically validated as OAT1/3 biomarkers through a detailed pharmacokinetic analysis (0-24 h) of plasma and urine samples. These biomarkers, either alone or as part of a panel, can predict OAT1/3-mediated DDIs and interindividual variability in the renal secretory clearance of organic anions across different populations, thereby enabling translational utility in clinical settings. The novel MDBD approach can be extended to discover biomarkers of other transporters and enzymes. SUMMARY Using clinical and mechanistic in vitro approaches, 23 endogenous substrates of OAT1/3 were identified as potential clinical biomarkers of renal secretary elimination of organic anions.
Collapse
|
3
|
Armani S, Geier A, Forst T, Merle U, Alpers DH, Lunnon MW. Effect of changes in metabolic enzymes and transporters on drug metabolism in the context of liver disease: Impact on pharmacokinetics and drug-drug interactions. Br J Clin Pharmacol 2023. [PMID: 38148609 DOI: 10.1111/bcp.15990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023] Open
Abstract
Changes in the pharmacokinetic and resulting pharmacodynamic properties of drugs are common in many chronic liver diseases, leading to adverse effects, drug interactions and increased risk of over- or underdosing of medications. Structural and functional hepatic impairment can have major effects on drug metabolism and transport. This review summarizes research on the functional changes in phase I and II metabolic enzymes and in transport proteins in patients with metabolic diseases such as type 2 diabetes, metabolic dysfunction-associated steatotic liver disease, metabolic dysfunction-associated steatohepatitis and cirrhosis, providing a clinical perspective on how these changes affect drug uptake and metabolism. Generally, a decrease in expression and/or activity of many enzymes of the cytochrome P450 family (e.g. CYP2E1 and CYP3A4), and of influx and efflux transporters (e.g. organic anion-transporting polypeptide [OATP]1B1, OATP2B1, OAT2 and bile salt export pump), has been recently documented in patients with liver disease. Decreased enzyme levels often correlate with increased severity of chronic liver disease. In subjects with hepatic impairment, there is potential for strong alterations of drug pharmacokinetics due to reduced absorption, increased volume of distribution, metabolism and extraction. Due to the altered pharmacokinetics, specific drug-drug interactions are also a potential issue to consider in patients with liver disease. Given the huge burden of liver disease in western societies, there is a need to improve awareness among all healthcare professionals and patients with liver disease to ensure appropriate drug prescriptions.
Collapse
Affiliation(s)
- Sara Armani
- CRS Clinical Research Services, Mannheim, Germany
| | - Andreas Geier
- Department of Internal Medicine and Hepatology, University Hospital, Würzburg, Germany
| | - Thomas Forst
- CRS Clinical Research Services, Mannheim, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital, Heidelberg, Germany
| | - David H Alpers
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
4
|
Chothe PP, Mitra P, Nakakariya M, Ramsden D, Rotter CJ, Sandoval P, Tohyama K. Drug transporters in drug disposition - the year 2022 in review. Drug Metab Rev 2023; 55:343-370. [PMID: 37644867 DOI: 10.1080/03602532.2023.2252618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
On behalf of all the authors, I am pleased to share our third annual review on drug transporter science with an emphasis on articles published and deemed influential in signifying drug transporters' role in drug disposition in the year 2022. As the drug transporter field is rapidly evolving several key findings were noted including promising endogenous biomarkers, rhythmic activity, IVIVE approaches in transporter-mediated clearance, new modality interaction, and transporter effect on gut microbiome. As identified previously (Chothe et Cal. 2021, 2022) the goal of this review is to highlight key findings without a comprehensive overview of each article and to this end, each coauthor independently selected 1-3 peer-reviewed articles published or available online in the year 2022 (Table 1). Each article is summarized in synopsis and commentary with unbiased viewpoints by each coauthor. We strongly encourage readers to consult original articles for specifics of the study. Finally, I would like to thank all coauthors for their continued support in writing this annual review on drug transporters and invite anyone interested in contributing to future versions of this review.
Collapse
Affiliation(s)
- Paresh P Chothe
- Department of Drug Metabolism and Pharmacokinetics, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Diane Ramsden
- Department of Drug Metabolism and Pharmacokinetics, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Charles J Rotter
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), San Diego, CA, USA
| | - Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
5
|
Ryu S, Woody N, Chang G, Mathialagan S, Varma MVS. Identification of Organic Anion Transporter 2 Inhibitors: Screening, Structure-Based Analysis, and Clinical Drug Interaction Risk Assessment. J Med Chem 2022; 65:14578-14588. [PMID: 36270005 DOI: 10.1021/acs.jmedchem.2c01079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Organic anion transporter 2 (OAT2 or SLC22A7) plays an important role in the hepatic uptake and renal secretion of several endogenous compounds and drugs. The goal of this work is to understand the structure activity of OAT2 inhibition and assess clinical drug interaction risk. A single-point inhibition assay using OAT2-transfected HEK293 cells was employed to screen about 150 compounds; and concentration-dependent inhibition potency (IC50) was measured for the identified "inhibitors". Acids represented about 65% of all inhibitors, and the frequency of bases-plus-zwitterions approximately doubled for "non-inhibitors". Interestingly, 9 of 10 most potent inhibitors (low IC50) are acids (pKa ∼ 3-5). Additionally, inhibitors are significantly larger and lipophilic than non-inhibitors. In silico (binary) models were developed to identify inhibitors and non-inhibitors. Finally, in vivo risk assessed via static drug-drug interaction models identified several inhibitors with potential for renal and hepatic OAT2 inhibition at clinical doses. This is the first study assessing the global pattern of OAT2-ligand interactions.
Collapse
Affiliation(s)
- Sangwoo Ryu
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nathaniel Woody
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - George Chang
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sumathy Mathialagan
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Manthena V S Varma
- Medicine Design, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
6
|
Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacol Ther 2022; 238:108268. [DOI: 10.1016/j.pharmthera.2022.108268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
7
|
Zamek-Gliszczynski MJ, Sangha V, Shen H, Feng B, Wittwer MB, Varma MVS, Liang X, Sugiyama Y, Zhang L, Bendayan R. Transporters in drug development: International transporter consortium update on emerging transporters of clinical importance. Clin Pharmacol Ther 2022; 112:485-500. [PMID: 35561119 DOI: 10.1002/cpt.2644] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/08/2022] [Indexed: 11/07/2022]
Abstract
During its 4th transporter workshop in 2021, the International Transporter Consortium (ITC) provided updates on emerging clinically relevant transporters for drug development. Previously highlighted and new transporters were considered based on up-to-date clinical evidence of their importance in drug-drug interactions and potential for altered drug efficacy and safety, including drug-nutrient interactions leading to nutrient deficiencies. For the first time, folate transport pathways (PCFT, RFC, and FRα) were examined in-depth as a potential mechanism of drug-induced folate deficiency and related toxicities (e.g., neural tube defects, megaloblastic anemia). However, routine toxicology studies conducted in support of drug development appear sufficient to flag such folate deficiency toxicities, while prospective prediction from in vitro folate metabolism and transport inhibition is not well enough established to inform drug development. Previous suggestion of retrospective study of intestinal OATP2B1 inhibition to explain unexpected decreases in drug exposure were updated. Furthermore, when the absorption of a new molecular entity is more rapid and extensive than can be explained by passive permeability, evaluation of OATP2B1 transport may be considered. Emerging research on hepatic and renal OAT2 is summarized, but current understanding of the importance of OAT2 was deemed insufficient to justify specific consideration for drug development. Hepatic, renal, and intestinal MRPs (MRP2, MRP3, MRP4) were revisited. MRPs may be considered when they are suspected to be the major determinant of drug disposition (e.g., direct glucuronide conjugates); MRP2 inhibition as a mechanistic explanation for drug-induced hyperbilirubinemia remains justified. There were no major changes in recommendations from previous ITC whitepapers.
Collapse
Affiliation(s)
| | - Vishal Sangha
- Department of Pharmaceutical Sciences, University of Toronto, Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | - Hong Shen
- Drug Metabolism and PK, Bristol Myers Squibb Company, Route 206 & Province Line Road, Princeton, NJ, 08543, USA
| | - Bo Feng
- Drug Metabolism and PK, Vertex Pharmaceuticals, Inc, 50 Northern Avenue, Boston, MA, 02210, USA
| | - Matthias B Wittwer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Manthena V S Varma
- PK, Dynamics and Metabolism, Medicine Design, Pfizer Inc, Worldwide R&D, Groton, CT, 06340, USA
| | - Xiaomin Liang
- Drug Metabolism, Gilead Sciences, Inc, 333 Lakeside Drive, Foster City, CA, 94404, USA
| | - Yuichi Sugiyama
- Laboratory of Quantitative System PK/Pharmacodynamics, School of Pharmacy, Josai International University, Kioicho Campus, Tokyo, 102-0093, Japan
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, University of Toronto, Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, ON, M5S 3M2, Canada
| | | |
Collapse
|
8
|
Yee SW, Giacomini KM. Emerging Roles of the Human Solute Carrier 22 Family. Drug Metab Dispos 2021; 50:DMD-MR-2021-000702. [PMID: 34921098 PMCID: PMC9488978 DOI: 10.1124/dmd.121.000702] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 11/22/2022] Open
Abstract
The human Solute Carrier 22 family (SLC22), also termed the organic ion transporter family, consists of 28 distinct multi-membrane spanning proteins, which phylogenetically cluster together according to their charge specificity for organic cations (OCTs), organic anions (OATs) and organic zwitterion/cations (OCTNs). Some SLC22 family members are well characterized in terms of their substrates, transport mechanisms and expression patterns, as well as their roles in human physiology and pharmacology, whereas others remain orphans with no known ligands. Pharmacologically, SLC22 family members play major roles as determinants of the absorption and disposition of many prescription drugs, and several including the renal transporters, OCT2, OAT1 and OAT3 are targets for many clinically important drug-drug interactions. In addition, mutations in some of these transporters (SLC22A5 (OCTN2) and SLC22A12 (URAT1) lead to rare monogenic disorders. Genetic polymorphisms in SLC22 transporters have been associated with common human disease, drug response and various phenotypic traits. Three members in this family were deorphaned in very recently: SLC22A14, SLC22A15 and SLC22A24, and found to transport specific compounds such as riboflavin (SLC22A14), anti-oxidant zwitterions (SLC22A15) and steroid conjugates (SLC22A24). Their physiologic and pharmacological roles need further investigation. This review aims to summarize the substrates, expression patterns and transporter mechanisms of individual SLC22 family members and their roles in human disease and drug disposition and response. Gaps in our understanding of SLC22 family members are described. Significance Statement In recent years, three members of the SLC22 family of transporters have been deorphaned and found to play important roles in the transport of diverse solutes. New research has furthered our understanding of the mechanisms, pharmacological roles, and clinical impact of SLC22 transporters. This minireview provides overview of SLC22 family members of their physiologic and pharmacologic roles, the impact of genetic variants in the SLC22 family on disease and drug response, and summary of recent studies deorphaning SLC22 family members.
Collapse
Affiliation(s)
- Sook Wah Yee
- Bioengineering and Therapeutic Sciences, Univerity of California, San Francisco, United States
| | - Kathleen M Giacomini
- Bioengineering and Therapeutic Sciences, Univerity of California, San Francisco, United States
| |
Collapse
|
9
|
Neuhoff S, Harwood MD, Rostami-Hodjegan A, Achour B. Application of proteomic data in the translation of in vitro observations to associated clinical outcomes. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 39:13-22. [PMID: 34906322 DOI: 10.1016/j.ddtec.2021.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/20/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022]
Abstract
Translation of information on drug exposure and effect is facilitated by in silico models that enable extrapolation of in vitro measurements to in vivo clinical outcomes. These models integrate drug-specific data with information describing physiological processes and pathological changes, including alterations to proteins involved in drug absorption, distribution and elimination. Over the past 15 years, quantitative proteomics has contributed a wealth of protein expression data, which are currently used for a variety of systems pharmacology applications, as a complement or a surrogate for activity of the corresponding proteins. In this review, we explore current and emerging applications of targeted and global (untargeted) proteomics in translational pharmacology as well as strategies for improved integration into model-based drug development.
Collapse
Affiliation(s)
- Sibylle Neuhoff
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Matthew D Harwood
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Amin Rostami-Hodjegan
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK; Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
10
|
Campesi I, Montella A, Seghieri G, Franconi F. The Person's Care Requires a Sex and Gender Approach. J Clin Med 2021; 10:4770. [PMID: 34682891 PMCID: PMC8541070 DOI: 10.3390/jcm10204770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
There is an urgent need to optimize pharmacology therapy with a consideration of high interindividual variability and economic costs. A sex-gender approach (which considers men, women, and people of diverse gender identities) and the assessment of differences in sex and gender promote global health, avoiding systematic errors that generate results with low validity. Care for people should consider the single individual and his or her past and present life experiences, as well as his or her relationship with care providers. Therefore, intersectoral and interdisciplinary studies are urgently required. It is desirable to create teams made up of men and women to meet the needs of both. Finally, it is also necessary to build an alliance among regulatory and ethic authorities, statistics, informatics, the healthcare system and providers, researchers, the pharmaceutical and diagnostic industries, decision makers, and patients to overcome the gender gap in medicine and to take real care of a person in an appropriate manner.
Collapse
Affiliation(s)
- Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100 Sassari, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Giuseppe Seghieri
- Department of Epidemiology, Regional Health Agency of Tuscany, 50124 Florence, Italy;
| | - Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and Biosystems, 07100 Sassari, Italy;
| |
Collapse
|
11
|
Sun HL, Wu YW, Bian HG, Yang H, Wang H, Meng XM, Jin J. Function of Uric Acid Transporters and Their Inhibitors in Hyperuricaemia. Front Pharmacol 2021; 12:667753. [PMID: 34335246 PMCID: PMC8317579 DOI: 10.3389/fphar.2021.667753] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Disorders of uric acid metabolism may be associated with pathological processes in many diseases, including diabetes mellitus, cardiovascular disease, and kidney disease. These diseases can further promote uric acid accumulation in the body, leading to a vicious cycle. Preliminary studies have proven many mechanisms such as oxidative stress, lipid metabolism disorders, and rennin angiotensin axis involving in the progression of hyperuricaemia-related diseases. However, there is still lack of effective clinical treatment for hyperuricaemia. According to previous research results, NPT1, NPT4, OAT1, OAT2, OAT3, OAT4, URAT1, GLUT9, ABCG2, PDZK1, these urate transports are closely related to serum uric acid level. Targeting at urate transporters and urate-lowering drugs can enhance our understanding of hyperuricaemia and hyperuricaemia-related diseases. This review may put forward essential references or cross references to be contributed to further elucidate traditional and novel urate-lowering drugs benefits as well as provides theoretical support for the scientific research on hyperuricemia and related diseases.
Collapse
Affiliation(s)
- Hao-Lu Sun
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Yi-Wan Wu
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - He-Ge Bian
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Hui Yang
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Heng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Department of Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
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
|
13
|
Mai Y, Dou L, Yao Z, Madla CM, Gavins FKH, Taherali F, Yin H, Orlu M, Murdan S, Basit AW. Quantification of P-Glycoprotein in the Gastrointestinal Tract of Humans and Rodents: Methodology, Gut Region, Sex, and Species Matter. Mol Pharm 2021; 18:1895-1904. [PMID: 33886332 PMCID: PMC8289313 DOI: 10.1021/acs.molpharmaceut.0c00574] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Intestinal efflux
transporters affect the gastrointestinal processing
of many drugs but further data on their intestinal expression levels
are required. Relative mRNA expression and relative and absolute protein
expression data of transporters are commonly measured by real-time
polymerase chain reaction (RT-PCR), Western blot and mass spectrometry-based
targeted proteomics techniques. All of these methods, however, have
their own strengths and limitations, and therefore, validation for
optimized quantification methods is needed. As such, the identification
of the most appropriate technique is necessary to effectively translate
preclinical findings to first-in-human trials. In this study, the
mRNA expression and protein levels of the efflux transporter P-glycoprotein
(P-gp) in jejunal and ileal epithelia of 30 male and female human
subjects, and the duodenal, jejunal, ileal and colonic tissues in
48 Wistar rats were quantified using RT-PCR, Western blot and liquid
chromatography-tandem mass spectrometry (LC-MS/MS). A similar sex
difference was observed in the expression of small intestinal P-gp
in humans and Wistar rats where P-gp was higher in males than females
with an increasing trend from the proximal to the distal parts in
both species. A strong positive linear correlation was determined
between the Western blot data and LC-MS/MS data in the small intestine
of humans (R2 = 0.85). Conflicting results,
however, were shown in rat small intestinal and colonic P-gp expression
between the techniques (R2 = 0.29 and
0.05, respectively). In RT-PCR and Western blot, an internal reference
protein is experimentally required; here, beta-actin was used which
is innately variable along the intestinal tract. Quantification via
LC-MS/MS can provide data on P-gp expression without the need for
an internal reference protein and consequently, can give higher confidence
on the expression levels of P-gp along the intestinal tract. Overall,
these findings highlight similar trends between the species and suggest
that the Wistar rat is an appropriate preclinical animal model to
predict the oral drug absorption of P-gp substrates in the human small
intestine.
Collapse
Affiliation(s)
- Yang Mai
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K.,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Liu Dou
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K.,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Zhicheng Yao
- Department of General Surgery, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Christine M Madla
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Francesca K H Gavins
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Farhan Taherali
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Heyue Yin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Mine Orlu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Sudaxshina Murdan
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Abdul W Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K
| |
Collapse
|
14
|
Jin C, Wei L, Ohgaki R, Tominaga H, Xu M, Okuda S, Okanishi H, Kawamoto Y, He X, Nagamori S, Kanai Y. Interaction of Halogenated Tyrosine/Phenylalanine Derivatives with Organic Anion Transporter 1 in the Renal Handling of Tumor Imaging Probes. J Pharmacol Exp Ther 2020; 375:451-462. [PMID: 32981893 DOI: 10.1124/jpet.120.000235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/16/2020] [Indexed: 03/08/2025] Open
Abstract
Halogenated tyrosine/phenylalanine derivatives have been developed for use in tumor imaging and targeted alpha therapy. 3-Fluoro-α-methyl-l-tyrosine (FAMT), targeting amino acid transporter LAT1 (SLC7A5), is a cancer-specific positron emission tomography probe that exhibits high renal accumulation, which is supposed to be mediated by organic anion transporter OAT1 (SLC22A6). In the present study, we investigated the structural requirements of FAMT essential for interaction with OAT1. OAT1 transported FAMT with a K m of 171.9 μM. In structure-activity relationship analyses, removal of either the 3-halogen or 4-hydroxyl group from FAMT or its structural analog 3-iodo-α-methyl-l-tyrosine greatly decreased the interaction with OAT1, reducing the [14C]p-aminohippurate uptake inhibition and the efflux induction. By contrast, the α-methyl group, which is essential for LAT1 specificity, contributed to a lesser degree. In fluorinated tyrosine derivatives, fluorine at any position was accepted by OAT1 when there was a hydroxyl group at the ortho-position, whereas ortho-fluorine was less interactive when a hydroxyl group was at meta- or para-positions. The replacement of the ortho-fluorine with a bulky iodine atom greatly increased the interaction. In in vivo studies, probenecid decreased the renal accumulation (P < 0.001) and urinary excretion (P = 0.0012) of FAMT, whereas the plasma concentration was increased, suggesting the involvement of OAT1-mediated transepithelial organic anion excretion. LAT1-specific 2-fluoro-α-methyltyrosine, which had lower affinity for OAT1, exhibited lower renal accumulation (P = 0.0142) and higher tumor uptake (P = 0.0192) compared with FAMT. These results would provide a basis to design tumor-specific compounds that can avoid renal accumulation for tumor imaging and targeted alpha therapy. SIGNIFICANCE STATEMENT: We revealed the structural characteristics of halogenated tyrosine derivatives essential for interaction with the organic anion transporter responsible for their renal accumulation. We have confirmed that such interactions are important for renal handling and tumor uptake. The critical contribution of hydroxyl and halogen groups and their positions as well as the role of α-methyl group found in the present study may facilitate the development of tumor-specific compounds while avoiding renal accumulation for use in tumor imaging and targeted alpha therapy.
Collapse
Affiliation(s)
- Chunhuan Jin
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Ling Wei
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Hideyuki Tominaga
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Yasuharu Kawamoto
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Xin He
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Shushi Nagamori
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine (C.J., L.W., R.O., M.X., S.O., H.O., Ya.K., Yo.K.) and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI) (Yo.K.), Osaka University, Osaka, Japan; Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan (H.T.); School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China (L.W., X.H.); and Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, Nara, Japan (S.N.)
| |
Collapse
|
15
|
Achour B, Al-Majdoub ZM, Rostami-Hodjegan A, Barber J. Mass Spectrometry of Human Transporters. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:223-247. [PMID: 32084322 DOI: 10.1146/annurev-anchem-091719-024553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Transporters are key to understanding how an individual will respond to a particular dose of a drug. Two patients with similar systemic concentrations may have quite different local concentrations of a drug at the required site. The transporter profile of any individual depends upon a variety of genetic and environmental factors, including genotype, age, and diet status. Robust models (virtual patients) are therefore required and these models are data hungry. Necessary data include quantitative transporter profiles at the relevant organ. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently the most powerful method available for obtaining this information. Challenges include sourcing the tissue, isolating the hydrophobic membrane-embedded transporter proteins, preparing the samples for MS (including proteolytic digestion), choosing appropriate quantification methodology, and optimizing the LC-MS/MS conditions. Great progress has been made with all of these, especially within the last few years, and is discussed here.
Collapse
Affiliation(s)
- Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
- Certara, Princeton, New Jersey 08540, USA
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
16
|
Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)-Based Proteomics of Drug-Metabolizing Enzymes and Transporters. Molecules 2020; 25:molecules25112718. [PMID: 32545386 PMCID: PMC7321193 DOI: 10.3390/molecules25112718] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics is a powerful tool for identifying and quantifying proteins in biological samples, outperforming conventional antibody-based methods in many aspects. LC-MS/MS-based proteomics studies have revealed the protein abundances of many drug-metabolizing enzymes and transporters (DMETs) in tissues relevant to drug metabolism and disposition. Previous studies have consistently demonstrated marked interindividual variability in DMET protein expression, suggesting that varied DMET function is an important contributing factor for interindividual variability in pharmacokinetics (PK) and pharmacodynamics (PD) of medications. Moreover, differential DMET expression profiles were observed across different species and in vitro models. Therefore, caution must be exercised when extrapolating animal and in vitro DMET proteomics findings to humans. In recent years, DMET proteomics has been increasingly utilized for the development of physiologically based pharmacokinetic models, and DMET proteins have also been proposed as biomarkers for prediction of the PK and PD of the corresponding substrate drugs. In sum, despite the existence of many challenges in the analytical technology and data analysis methods of LC-MS/MS-based proteomics, DMET proteomics holds great potential to advance our understanding of PK behavior at the individual level and to optimize treatment regimens via the DMET protein biomarker-guided precision pharmacotherapy.
Collapse
|
17
|
De Sousa Mendes M, Chetty M. Are Standard Doses of Renally-Excreted Antiretrovirals in Older Patients Appropriate: A PBPK Study Comparing Exposures in the Elderly Population With Those in Renal Impairment. Drugs R D 2020; 19:339-350. [PMID: 31602556 PMCID: PMC6890626 DOI: 10.1007/s40268-019-00285-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The elderly population receives the majority of prescription drugs but are usually excluded from Phase 1 clinical trials. Alternative approaches to estimate increases in toxicity risk or decreases in efficacy are therefore needed. This study predicted the pharmacokinetics (PK) of three renally excreted antiretroviral drugs in the elderly population and compared them with known exposures in renal impairment, to evaluate the need for dosing adjustments. METHODS The performance of the physiologically based pharmacokinetic (PBPK) models for tenofovir, lamivudine and emtricitabine were verified using clinical data in young and older subjects. Models were then used to predict PK profiles in a virtual population aged 20 to 49 years (young) and a geriatric population aged 65 to 74 years (elderly). Predicted exposure in the elderly was then compared with exposure reported for different degrees of renal impairment, where doses have been defined. RESULTS An increase in exposure (AUC) with advancing age was predicted for all drugs. The mean ratio of the increase in exposure were 1.40 for emtricitabine, 1.42 for lamivudine and 1.48 for tenofovir. The majority of virtual patients had exposures that did not require dosage adjustments. About 22% of patients on tenofovir showed exposures similar to that in moderate renal impairment, where dosage reduction may be required. CONCLUSION Comparison of the exposure in the elderly with exposure observed in patients with different levels of renal impairment, indicated that a dosage adjustment may not be required in elderly patients on lamivudine, emtricitabine and the majority of the patients on tenofovir. Clinical trials to verify these predictions are essential.
Collapse
|
18
|
A novel mass spectrometry method for the absolute quantification of several cytochrome P450 and uridine 5'-diphospho-glucuronosyltransferase enzymes in the human liver. Anal Bioanal Chem 2020; 412:1729-1740. [PMID: 32030490 DOI: 10.1007/s00216-020-02445-7] [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: 10/09/2019] [Revised: 12/22/2019] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
Cytochrome P450 (CYP450) and 5'-diphosphate glucuronosyltransferases (UGT) are the two major families of drug-metabolizing enzymes in the human liver microsome (HLM). As a result of their frequent abundance fluctuation among populations, the accurate quantification of these enzymes in different individuals is important for designing patient-specific dosage regimens in the framework of precision medicine. The preparation and quantification of internal standards is an essential step for the quantitative analysis of enzymes. However, the commonly employed stable isotope labeling-based strategy (QconCAT) suffers from requiring very expensive isotopic reagents, tedious experimental procedures, and long labeling times. Furthermore, arginine-to-proline conversion during metabolic isotopic labeling compromises the quantification accuracy. Therefore, we present a new strategy that replaces stable isotope-labeled amino acids with lanthanide labeling for the preparation and quantification of QconCAT internal standard peptides, which leads to a threefold reduction in the reagent costs and a fivefold reduction in the time consumed. The absolute amount of trypsin-digested QconCAT peptides can be obtained by lanthanide labeling and inductively coupled plasma-optical emission spectrometry (ICP-OES) analysis with a high quantification accuracy (%RE < 20%). By taking advantage of the highly selective and facile ICP-OES procedure and multiplexed large-scale absolute target protein quantification using biological mass spectrometry, this strategy was successfully used for the absolute quantification of drug-metabolizing enzymes. We obtained good linearity (correlation coefficient > 0.95) over concentrations spanning 2.5 orders of magnitude with improved sensitivity (limit of quantification = 2 fmol) in nine HLM samples, indicating the potential of this method for large-scale absolute target protein quantification in clinical samples. Graphical abstract.
Collapse
|
19
|
Bi YA, Costales C, Mathialagan S, West M, Eatemadpour S, Lazzaro S, Tylaska L, Scialis R, Zhang H, Umland J, Kimoto E, Tess DA, Feng B, Tremaine LM, Varma MVS, Rodrigues AD. Quantitative Contribution of Six Major Transporters to the Hepatic Uptake of Drugs: "SLC-Phenotyping" Using Primary Human Hepatocytes. J Pharmacol Exp Ther 2019; 370:72-83. [PMID: 30975793 DOI: 10.1124/jpet.119.257600] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/08/2019] [Indexed: 11/22/2022] Open
Abstract
Hepatic uptake transporters [solute carriers (SLCs)], including organic anion transporting polypeptide (OATP) 1B1, OATP1B3, OATP2B1, sodium-dependent taurocholate cotransporting polypeptide (NTCP), and organic anion (OAT2) and organic cation (OCT1) transporters, play a key role in determining the systemic and liver exposure of chemically diverse drugs. Here, we established a phenotyping approach to quantify the contribution of the six SLCs, and passive diffusion, to the overall uptake using plated human hepatocytes (PHHs). First, selective inhibitor conditions were identified by screening about 20 inhibitors across the six SLCs using single-transfected human embryonic kidney 293 cells. Data implied rifamycin SV (20 µM) inhibits three OATPs, while rifampicin (5 µM) inhibits OATP1B1/1B3 only. Further, hepatitis B virus myristoylated-preS1 peptide (0.1 µM), quinidine (100 µM), and ketoprofen (100-300 µM) are relatively selective against NTCP, OCT1, and OAT2, respectively. Second, using these inhibitory conditions, the fraction transported (ft ) by the individual SLCs was characterized for 20 substrates with PHH. Generally, extended clearance classification system class 1A/3A (e.g., warfarin) and 1B/3B compounds (e.g., statins) showed predominant OAT2 and OATP1B1/1B3 contribution, respectively. OCT1-mediated uptake was prominent for class 2/4 compounds (e.g., metformin). Third, in vitro ft values were corrected using quantitative proteomics data to obtain "scaled ft " Fourth, in vitro-in vivo extrapolation of the scaled OATP1B1/1B3 ft was assessed, leveraging statin clinical drug-drug interaction data with rifampicin as the perpetrator. Finally, we outlined a novel stepwise strategy to implement phenotypic characterization of SLC-mediated hepatic uptake for new molecular entities and drugs in a drug discovery and development setting.
Collapse
Affiliation(s)
- Yi-An Bi
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Chester Costales
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Mark West
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Soraya Eatemadpour
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sarah Lazzaro
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Renato Scialis
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Hui Zhang
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - John Umland
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Emi Kimoto
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - David A Tess
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Bo Feng
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Larry M Tremaine
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
20
|
Kurzawski M, Szeląg-Pieniek S, Łapczuk-Romańska J, Wrzesiński M, Sieńko J, Oswald S, Droździk M. The reference liver - ABC and SLC drug transporters in healthy donor and metastatic livers. Pharmacol Rep 2019; 71:738-745. [PMID: 31207436 DOI: 10.1016/j.pharep.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Analysis of results and conclusions in studies dedicated to pathology of the liver are usually based on comparison of pathological liver specimens and control/reference (considered as healthy) tissues. There are two main sources of the control liver samples used as the reference livers, i.e. deceased organ donor livers and non-tumorous tissue from metastatic livers, which are also applied for drug transporter investigations. However, no information has yet been published on drug transporters in these two major types of reference livers. METHODS We explored ABC (P-gp, MRP1, MRP2, MRP3, MRP4, BCRP, BSEP) and SLC (NTCP, MCT1, OCT1, OCT3, OAT2, OATP1B1, OATP1B3, OATP2B1) family transporters expression (qPCR) and protein abundance (LC-MS/MS) in healthy donors (n = 9) and metastatic (n = 13) livers. RESULTS The analysis of mRNA content revealed significant differences in ABCB11, ABCC1, ABCG2, SLC10A1, SLC16A1, SLCO1B1 and SLCO2B1 gene expression between livers from organ donors and patients who underwent surgical resection of metastatic tumors. The protein abundance of NTCP was significantly higher, whereas of P-gp significantly lower in non-tumorous tissues from metastatic livers. Greater inter-individual variability in protein abundance of all studied transporters in subjects with metastatic colon cancer was also observed. CONCLUSIONS The results suggest that final conclusions in liver pathology studies may depend on the reference liver tissue used, especially in gene expression studies.
Collapse
Affiliation(s)
- Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland.
| | - Sylwia Szeląg-Pieniek
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Joanna Łapczuk-Romańska
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Maciej Wrzesiński
- Department of General and Transplantation Surgery, Marie Curie Regional Hospital, Szczecin, Poland
| | - Jerzy Sieńko
- Department of General and Transplantation Surgery, Pomeranian Medical University, Szczecin, Poland
| | - Stefan Oswald
- Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald, Germany
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
21
|
Kumar V, Salphati L, Hop CECA, Xiao G, Lai Y, Mathias A, Chu X, Humphreys WG, Liao M, Heyward S, Unadkat JD. A Comparison of Total and Plasma Membrane Abundance of Transporters in Suspended, Plated, Sandwich-Cultured Human Hepatocytes Versus Human Liver Tissue Using Quantitative Targeted Proteomics and Cell Surface Biotinylation. Drug Metab Dispos 2019; 47:350-357. [PMID: 30622164 DOI: 10.1124/dmd.118.084988] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/07/2019] [Indexed: 02/13/2025] Open
Abstract
Suspended (SH), plated (PH), and sandwich-cultured hepatocytes (SCH) are commonly used models to predict in vivo transporter-mediated hepatic uptake (SH or PH) or biliary (SCH) clearance of drugs. When doing so, the total and the plasma membrane abundance (PMA) of transporter are assumed not to differ between hepatocytes and liver tissue (LT). This assumption has never been tested. In this study, we tested this assumption by measuring the total and PMA of the transporters in human hepatocyte models versus LT (total only) from which they were isolated. Total abundance of OATP1B1/2B1/1B3, OCT1, and OAT2 was not significantly different between the hepatocytes and LT. The same was true for the PMA of these transporters across the hepatocyte models. In contrast, total abundance of the sinusoidal efflux transporter, MRP3, and the canalicular efflux transporters, MRP2 and P-gp, was significantly greater (P < 0.05) in SCH versus LT. Of the transporters tested, only the percentage of PMA of OATP1B1, P-gp, and MRP3, in SCH (82.8% ± 7.3%, 57.5% ± 10.9%, 69.3% ± 5.7%) was significantly greater (P < 0.05) than in SH (73.3% ± 6.4%, 27.4% ± 6.4%, 53.6% ± 4.1%). If the transporters measured in the plasma membrane are functional and the PMA in SH is representative of that in LT, these data suggest that SH, PH, and SCH will result in equal prediction of hepatic uptake clearance of drugs mediated by the transporters tested above. However, SCH will predict higher sinusoidal efflux and biliary clearance of drugs if the change in PMA of these transporters is not taken into consideration.
Collapse
Affiliation(s)
- Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Guangqing Xiao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Yurong Lai
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Anita Mathias
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Xiaoyan Chu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - W Griffith Humphreys
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Mingxiang Liao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Scott Heyward
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| |
Collapse
|
22
|
Kimoto E, Mathialagan S, Tylaska L, Niosi M, Lin J, Carlo AA, Tess DA, Varma MVS. Organic Anion Transporter 2-Mediated Hepatic Uptake Contributes to the Clearance of High-Permeability-Low-Molecular-Weight Acid and Zwitterion Drugs: Evaluation Using 25 Drugs. J Pharmacol Exp Ther 2018; 367:322-334. [PMID: 30135178 DOI: 10.1124/jpet.118.252049] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 03/08/2025] Open
Abstract
High-permeability-low-molecular-weight acids/zwitterions [i.e., extended clearance classification system class 1A (ECCS 1A) drugs] are considered to be cleared by metabolism with a minimal role of membrane transporters in their hepatic clearance. However, a marked disconnect in the in vitro-in vivo (IVIV) translation of hepatic clearance is often noted for these drugs. Metabolic rates measured using human liver microsomes and primary hepatocytes tend to underpredict. Here, we evaluated the role of organic anion transporter 2 (OAT2)-mediated hepatic uptake in the clearance of ECCS 1A drugs. For a set of 25 ECCS 1A drugs, in vitro transport activity was assessed using transporter-transfected cells and primary human hepatocytes. All but two drugs showed substrate affinity to OAT2, whereas four (bromfenac, entacapone, fluorescein, and nateglinide) also showed OATP1B1 activity in transfected cells. Most of these drugs (21 of 25) showed active uptake by plated human hepatocytes, with rifamycin SV (pan-transporter inhibitor) reducing the uptake by about 25%-95%. Metabolic turnover was estimated for 19 drugs after a few showed no measurable substrate depletion in liver microsomal incubations. IVIV extrapolation using in vitro data was evaluated to project human hepatic clearance of OAT2-alone substrates considering 1) uptake transport only, 2) metabolism only, and 3) transporter-enzyme interplay (extended clearance model). The transporter-enzyme interplay approach achieved improved prediction accuracy (average fold error = 1.9 and bias = 0.93) compared with the other two approaches. In conclusion, this study provides functional evidence for the role of OAT2-mediated hepatic uptake in determining the pharmacokinetics of several clinically important ECCS 1A drugs.
Collapse
Affiliation(s)
- Emi Kimoto
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Mark Niosi
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Jian Lin
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Anthony A Carlo
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - David A Tess
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|