1
|
Ailabouni A, Prasad B. Organic cation transporters 2: Structure, regulation, functions, and clinical implications. Drug Metab Dispos 2025; 53:100044. [PMID: 40020559 DOI: 10.1016/j.dmd.2025.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/21/2025] [Indexed: 03/03/2025] Open
Abstract
The SLC22A2 gene encodes organic cation transporter 2 (OCT2), which is predominantly expressed in renal proximal tubule cells. OCT2 is critical for the active renal excretion of various cationic drugs and endogenous metabolites. OCT2 expression varies across species, with higher levels in mice and monkeys compared with humans and rats. The human OCT2 protein consists of 555 amino acids and contains 12 transmembrane domains. OCT2 functions as a uniporter, facilitating the bidirectional transport of organic cations into renal tubular cells, driven by the inside-negative membrane potential. Its expression is regulated by sex hormones, contributing to potential sex differences in Oct2 activity in rodents. OCT2 has been linked to tissue toxicity, such as cisplatin-induced nephrotoxicity. Factors such as genetic variants, age, disease states, and the coadministration of drugs, including tyrosine kinase inhibitors, contribute to interindividual variability in OCT2 activity. This, in turn, impacts the systemic exposure and elimination of drugs and endogenous substances. Regulatory agencies recommend evaluating the potential of a drug to inhibit OCT2 through in vitro and clinical drug-drug interaction (DDI) studies, often using metformin as a probe substrate. Emerging tools like transporter biomarkers and physiologically based pharmacokinetic modeling hold promise in predicting OCT2-mediated DDIs. While several OCT2 biomarkers, such as N1-methylnicotinamide, have been proposed, their reliability in predicting renal DDIs remains uncertain and requires further study. Ultimately, a better understanding of the factors influencing OCT2 activity is essential for achieving precision medicine and minimizing renal and systemic toxicity. SIGNIFICANCE STATEMENT: Organic cation transporter 2 (OCT2) is essential for the active tubular secretion of xenobiotics and endogenous cationic substances in the kidneys. This article offers a comprehensive overview of the tissue distribution, interspecies differences, and factors affecting its activity-critical for evaluating tissue toxicity and systemic exposure to cationic substances. Using OCT2 biomarkers and integrating OCT2 activity and expression data into physiologically based pharmacokinetic models are valuable tools for predicting OCT2 function and its clinical implications.
Collapse
Affiliation(s)
- Anoud Ailabouni
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington.
| |
Collapse
|
2
|
Chen X, Yu G, Wang G, Li GF. Pediatric off-label use and nonadherence management for nadolol: A mechanistic PBPK model Incorporating Ontogeny Scaling from Interracial Adults to Children. J Pharm Sci 2025:103707. [PMID: 40010493 DOI: 10.1016/j.xphs.2025.103707] [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: 11/19/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Nadolol has demonstrated its superior efficacy over other β-blockers in the treatment of specific cardiovascular diseases in children. The clinical development of nadolol for pediatric use was prioritized by Chinese healthcare authorities in May 2023 while there was a lack of clear medication instructions for children. To expedite the pediatric development of nadolol and provide insights into its off-label applications, we developed a physiologically based pharmacokinetic model incorporating mechanistic disposition knowledge. This model integrates key processes of nadolol including P-glycoprotein (P-gp) transporter mediated the absorption of efflux, multidrug and toxin extrusion protein (MATE) 1 transporter and organic cation (OCT) 2 transporter mediated active renal excretion, organic anion transporting polypeptides (OATP) 1A2 mediated transport, along with biliary excretion. The model accurately captured the pharmacokinetic profiles of nadolol in both Western and East Asian populations following a wide dose range (2-160 mg), including the plasma concentration, urine excretion, and drug-drug interactions with the P-gp inhibitor. After our good validation on interracial adult populations, simulations of nadolol pharmacokinetic profiles in the Chinese population were performed by adjusting the liver volume of the Chinese to 0.9 of the Japanese population. Then, with the consideration of physiological changes and plasma protein ontogeny in pediatrics, the nadolol model for pediatrics was also well-verified on several children aged 3 months to 121 months. Accordingly, specific optimal dosages for children across various ages and racial backgrounds with or without obesity were offered by exposure matching with adults. Multiple remedial regimen simulations were also compared to obtain the best nonadherence management in the case of missed dosages, in which resuming a regular dose as soon as possible was the most recommended.
Collapse
Affiliation(s)
- Xiang Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guo Yu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, China
| | - Guo-Fu Li
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China; Subei People's Hospital, Yangzhou, China.
| |
Collapse
|
3
|
Pernecker M, Ciarimboli G. Regulation of renal organic cation transporters. FEBS Lett 2024; 598:2328-2347. [PMID: 38831380 DOI: 10.1002/1873-3468.14943] [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: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Transporters for organic cations (OCs) facilitate exchange of positively charged molecules through the plasma membrane. Substrates for these transporters encompass neurotransmitters, metabolic byproducts, drugs, and xenobiotics. Consequently, these transporters actively contribute to the regulation of neurotransmission, cellular penetration and elimination process for metabolic products, drugs, and xenobiotics. Therefore, these transporters have significant physiological, pharmacological, and toxicological implications. In cells of renal proximal tubules, the vectorial secretion pathways for OCs involve expression of organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) on basolateral and apical membrane domains, respectively. This review provides an overview of documented regulatory mechanisms governing OCTs and MATEs. Additionally, regulation of these transporters under various pathological conditions is summarized. The expression and functionality of OCTs and MATEs are subject to diverse pre- and post-translational modifications, providing insights into their regulation in various pathological conditions. Typically, in diseases, downregulation of transporter expression is observed, probably as a protective mechanism to prevent additional damage to kidney tissue. This regulation may be attributed to the intricate network of modifications these transporters undergo, shedding light on their dynamic responses in pathological contexts.
Collapse
Affiliation(s)
- Moritz Pernecker
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| |
Collapse
|
4
|
Thakur A, Yue G, Ahire D, Mettu VS, Maghribi AA, Ford K, Peixoto L, Leeder JS, Prasad B. Sex and the Kidney Drug-Metabolizing Enzymes and Transporters: Are Preclinical Drug Disposition Data Translatable to Humans? Clin Pharmacol Ther 2024; 116:235-246. [PMID: 38711199 PMCID: PMC11218045 DOI: 10.1002/cpt.3277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/06/2024] [Indexed: 05/08/2024]
Abstract
Cross-species differences in drug transport and metabolism are linked to poor translation of preclinical pharmacokinetic and toxicology data to humans, often resulting in the failure of new chemical entities (NCEs) during clinical drug development. Specifically, inaccurate prediction of renal clearance and renal accumulation of NCEs due to differential abundance of enzymes and transporters in kidneys can lead to differences in pharmacokinetics and toxicity between experimental animals and humans. We carried out liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based protein quantification of 78 membrane drug-metabolizing enzymes and transporters (DMETs) in the kidney membrane fractions of humans, rats, and mice for characterization of cross-species and sex-dependent differences. In general, majority of DMET proteins were higher in rodents than in humans. Significant cross-species differences were observed in 30 out of 33 membrane DMET proteins quantified in all three species. Although no significant sex-dependent differences were observed in humans, the abundance of 28 and 46 membrane proteins showed significant sex dependence in rats and mice, respectively. These cross-species and sex-dependent quantitative abundance data are valuable for gaining a mechanistic understanding of drug renal disposition and accumulation. Further, these data can also be integrated into systems pharmacology tools, such as physiologically based pharmacokinetic models, to enhance the interpretation of preclinical pharmacokinetic and toxicological data.
Collapse
Affiliation(s)
- Aarzoo Thakur
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, US
| | - Guihua Yue
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, US
| | - Deepak Ahire
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, US
| | - Vijaya S. Mettu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, US
| | - Abrar Al Maghribi
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, US
| | - Kaitlyn Ford
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, US
| | - Lucia Peixoto
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, US
| | | | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, US
| |
Collapse
|
5
|
Galetin A, Brouwer KLR, Tweedie D, Yoshida K, Sjöstedt N, Aleksunes L, Chu X, Evers R, Hafey MJ, Lai Y, Matsson P, Riselli A, Shen H, Sparreboom A, Varma MVS, Yang J, Yang X, Yee SW, Zamek-Gliszczynski MJ, Zhang L, Giacomini KM. Membrane transporters in drug development and as determinants of precision medicine. Nat Rev Drug Discov 2024; 23:255-280. [PMID: 38267543 PMCID: PMC11464068 DOI: 10.1038/s41573-023-00877-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/26/2024]
Abstract
The effect of membrane transporters on drug disposition, efficacy and safety is now well recognized. Since the initial publication from the International Transporter Consortium, significant progress has been made in understanding the roles and functions of transporters, as well as in the development of tools and models to assess and predict transporter-mediated activity, toxicity and drug-drug interactions (DDIs). Notable advances include an increased understanding of the effects of intrinsic and extrinsic factors on transporter activity, the application of physiologically based pharmacokinetic modelling in predicting transporter-mediated drug disposition, the identification of endogenous biomarkers to assess transporter-mediated DDIs and the determination of the cryogenic electron microscopy structures of SLC and ABC transporters. This article provides an overview of these key developments, highlighting unanswered questions, regulatory considerations and future directions.
Collapse
Affiliation(s)
- Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK.
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Kenta Yoshida
- Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, CA, USA
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Lauren Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., Rahway, NJ, USA
| | - Raymond Evers
- Preclinical Sciences and Translational Safety, Johnson & Johnson, Janssen Pharmaceuticals, Spring House, PA, USA
| | - Michael J Hafey
- Department of Pharmacokinetics, Dynamics, Metabolism, and Bioanalytics, Merck & Co., Inc., Rahway, NJ, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, USA
| | - Pär Matsson
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Andrew Riselli
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hong Shen
- Department of Drug Metabolism and Pharmacokinetics, Bristol Myers Squibb Research and Development, Princeton, NJ, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, CT, USA
| | - Jia Yang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | | | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Ma X, Ren X, Zhang X, Wang G, Liu H, Wang L. Rutin ameliorate PFOA induced renal damage by reducing oxidative stress and improving lipid metabolism. J Nutr Biochem 2024; 123:109501. [PMID: 37890710 DOI: 10.1016/j.jnutbio.2023.109501] [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: 05/17/2023] [Revised: 10/09/2023] [Accepted: 10/22/2023] [Indexed: 10/29/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent environmental pollutant that can accumulate in the kidneys and eventually cause kidney damage. Rutin (RUTIN) is a natural flavonoid with multiple biological activities, and its use in against kidney damage has been widely studied in recent years. It is not yet known whether rutin protects against kidney damage caused by PFOA. In this study, 30 ICR mice were randomly divided into three groups: CTRL group, PFOA group and PFOA+RUTIN group. The mice were fed continuously by gavage for 28 days. Renal pathological changes were assessed by HE and PASM staining, and serum renal function and lipid indicators were measured. RNA-seq and enrichment analysis using GO, KEGG and PPI to detect differential expression of genes in treatment groups. Kidney tissue protein expression was determined by Western blot. Research has shown that rutin can improve glomerular and tubular structural damage, and increase serum CREA, HDL-C levels and decrease LDH, LDL-C levels. The expression of AQP1 and ACOT1 was up-regulated after rutin treatment. Transcriptomic analysis indicated that PFOA and rutin affect the transcriptional expression of genes related to lipid metabolism and oxidative stress, and may affected by PI3K-Akt, PPAR, NRF2/KEAP1 signaling pathways. In conclusion, rutin ameliorated renal damage caused by PFOA exposure, and this protective effect may be exerted by ameliorating oxidative stress and regulating lipid metabolism.
Collapse
Affiliation(s)
- Xinzhuang Ma
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Xijuan Ren
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Xuemin Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, PR China
| | - Guangyin Wang
- School of Public Health, Bengbu Medical College, Bengbu, PR China
| | - Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, PR China.
| | - Li Wang
- School of Public Health, Bengbu Medical College, Bengbu, PR China.
| |
Collapse
|
7
|
Thakur A, Saradhi Mettu V, Singh DK, Prasad B. Effect of probenecid on blood levels and renal elimination of furosemide and endogenous compounds in rats: Discovery of putative organic anion transporter biomarkers. Biochem Pharmacol 2023; 218:115867. [PMID: 37866801 PMCID: PMC10900896 DOI: 10.1016/j.bcp.2023.115867] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Transporter-mediated drug-drug interactions (DDIs) are assessed using probe drugs and in vitro and in vivo models during drug development. The utility of endogenous metabolites as transporter biomarkers is emerging for prediction of DDIs during early phases of clinical trials. Endogenous metabolites such as pyridoxic acid and kynurenic acid have shown potential to predict DDIs mediated by organic anion transporters (OAT1 and OAT3). However, these metabolites have not been assessed in rats as potential transporter biomarkers. We carried out a rat pharmacokinetic DDI study using probenecid and furosemide as OAT inhibitor and substrate, respectively. Probenecid administration led to a 3.8-fold increase in the blood concentrations and a 3-fold decrease in renal clearance of furosemide. High inter-individual and intra-day variability in pyridoxic acid and kynurenic acid, and no or moderate effect of probenecid administration on these metabolites suggest their limited utility for prediction of Oat-mediated DDI in rats. Therefore, rat blood and urine samples were further analysed using untargeted metabolomics. Twenty-one m/z features (out of >8000 detected features) were identified as putative biomarkers of rat Oat1 and Oat3 using a robust biomarker qualification approach. These m/z features belong to metabolic pathways such as fatty acid analogues, peptides, prostaglandin analogues, bile acid derivatives, flavonoids, phytoconstituents, and steroids, and can be used as a panel to decrease variability caused by processes other than Oats. When validated, these putative biomarkers will be useful in predicting DDIs caused by Oats in rats.
Collapse
Affiliation(s)
- Aarzoo Thakur
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Vijaya Saradhi Mettu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Dilip K Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
8
|
Dinh J, Johnson TN, Grimstein M, Lewis T. Physiologically Based Pharmacokinetics Modeling in the Neonatal Population-Current Advances, Challenges, and Opportunities. Pharmaceutics 2023; 15:2579. [PMID: 38004559 PMCID: PMC10675397 DOI: 10.3390/pharmaceutics15112579] [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: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is an approach to predicting drug pharmacokinetics, using knowledge of the human physiology involved and drug physiochemical properties. This approach is useful when predicting drug pharmacokinetics in under-studied populations, such as pediatrics. PBPK modeling is a particularly important tool for dose optimization for the neonatal population, given that clinical trials rarely include this patient population. However, important knowledge gaps exist for neonates, resulting in uncertainty with the model predictions. This review aims to outline the sources of variability that should be considered with developing a neonatal PBPK model, the data that are currently available for the neonatal ontogeny, and lastly to highlight the data gaps where further research would be needed.
Collapse
Affiliation(s)
- Jean Dinh
- Certara UK Limited, Sheffield S1 2BJ, UK; (J.D.); (T.N.J.)
| | | | - Manuela Grimstein
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Tamorah Lewis
- Pediatric Clinical Pharmacology & Toxicology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
9
|
van Groen BD, Allegaert K, Tibboel D, de Wildt SN. Innovative approaches and recent advances in the study of ontogeny of drug metabolism and transport. Br J Clin Pharmacol 2022; 88:4285-4296. [PMID: 32851677 PMCID: PMC9545189 DOI: 10.1111/bcp.14534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/10/2020] [Accepted: 08/16/2020] [Indexed: 11/30/2022] Open
Abstract
The disposition of a drug is driven by various processes, such as drug metabolism, drug transport, glomerular filtration and body composition. These processes are subject to developmental changes reflecting growth and maturation along the paediatric continuum. However, knowledge gaps exist on these changes and their clinical impact. Filling these gaps may aid better prediction of drug disposition and creation of age-appropriate dosing guidelines. We present innovative approaches to study these developmental changes in relation to drug metabolism and transport. First, analytical methods such as including liquid chromatography-mass spectrometry for proteomic analyses allow quantitation of the expressions of a wide variety of proteins, e.g. membrane transporters, in a small piece of organ tissue. The latter is specifically important for paediatric research, where tissues are scarcely available. Second, innovative study designs using radioactive labelled microtracers allowed study-without risk for the child-of the oral bioavailability of compounds used as markers for certain drug metabolism pathways. Third, the use of modelling and simulation to support dosing recommendations for children is supported by both the European Medicines Agency and the US Food and Drug Administration. This may even do away with the need for a paediatric trial. Physiologically based pharmacokinetics models, which include age-specific physiological information are, therefore, increasingly being used, not only to aid paediatric drug development but also to improve existing drug therapies.
Collapse
Affiliation(s)
- Bianca D. van Groen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC‐Sophia Children's HospitalRotterdamthe Netherlands
| | - Karel Allegaert
- Department of Development and Regeneration, KU LeuvenLeuvenBelgium
- Department of Pharmacy and Pharmaceutical Sciences, KU LeuvenLeuvenBelgium
- Department of Clinical Pharmacy, Erasmus MCRotterdamthe Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC‐Sophia Children's HospitalRotterdamthe Netherlands
| | - Saskia N. de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC‐Sophia Children's HospitalRotterdamthe Netherlands
- Department of Pharmacology and ToxicologyRadboud Institute of Health Sciences, Radboud University Medical CenterNijmegenthe Netherlands
| |
Collapse
|
10
|
Wu C, Li B, Meng S, Qie L, Zhang J, Wang G, Ren CC. Prediction for optimal dosage of pazopanib under various clinical situations using physiologically based pharmacokinetic modeling. Front Pharmacol 2022; 13:963311. [PMID: 36172188 PMCID: PMC9510668 DOI: 10.3389/fphar.2022.963311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
This study aimed to apply a physiologically based pharmacokinetic (PBPK) model to predict optimal dosing regimens of pazopanib (PAZ) for safe and effective administration when co-administered with CYP3A4 inhibitors, acid-reducing agents, food, and administered in patients with hepatic impairment. Here, we have successfully developed the population PBPK model and the predicted PK variables by this model matched well with the clinically observed data. Most ratios of prediction to observation were between 0.5 and 2.0. Suitable dosage modifications of PAZ have been identified using the PBPK simulations in various situations, i.e., 200 mg once daily (OD) or 100 mg twice daily (BID) when co-administered with the two CYP3A4 inhibitors, 200 mg BID when simultaneously administered with food or 800 mg OD when avoiding food uptake simultaneously. Additionally, the PBPK model also suggested that dosing does not need to be adjusted when co-administered with esomeprazole and administration in patients with wild hepatic impairment. Furthermore, the PBPK model also suggested that PAZ is not recommended to be administered in patients with severe hepatic impairment. In summary, the present PBPK model can determine the optimal dosing adjustment recommendations in multiple clinical uses, which cannot be achieved by only focusing on AUC linear change of PK.
Collapse
Affiliation(s)
- Chunnuan Wu
- Department of pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Bole Li
- Department of pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Shuai Meng
- Department of pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Linghui Qie
- Department of pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jie Zhang
- Department of pharmacy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- *Correspondence: Jie Zhang, ; Guopeng Wang, ; Cong Cong Ren,
| | - Guopeng Wang
- Zhongcai Health Biological Technology Development Co., Ltd., Beijing, China
- *Correspondence: Jie Zhang, ; Guopeng Wang, ; Cong Cong Ren,
| | - Cong Cong Ren
- Department of pharmacy, Liaocheng People’s Hospital, Liaocheng, China
- *Correspondence: Jie Zhang, ; Guopeng Wang, ; Cong Cong Ren,
| |
Collapse
|
11
|
Kamath A, Srinivasamurthy SK, Chowta MN, Ullal SD, Daali Y, Chakradhara Rao US. Role of Drug Transporters in Elucidating Inter-Individual Variability in Pediatric Chemotherapy-Related Toxicities and Response. Pharmaceuticals (Basel) 2022; 15:990. [PMID: 36015138 PMCID: PMC9415926 DOI: 10.3390/ph15080990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Pediatric cancer treatment has evolved significantly in recent decades. The implementation of risk stratification strategies and the selection of evidence-based chemotherapy combinations have improved survival outcomes. However, there is large interindividual variability in terms of chemotherapy-related toxicities and, sometimes, the response among this population. This variability is partly attributed to the functional variability of drug-metabolizing enzymes (DME) and drug transporters (DTS) involved in the process of absorption, distribution, metabolism and excretion (ADME). The DTS, being ubiquitous, affects drug disposition across membranes and has relevance in determining chemotherapy response in pediatric cancer patients. Among the factors affecting DTS function, ontogeny or maturation is important in the pediatric population. In this narrative review, we describe the role of drug uptake/efflux transporters in defining pediatric chemotherapy-treatment-related toxicities and responses. Developmental differences in DTS and the consequent implications are also briefly discussed for the most commonly used chemotherapeutic drugs in the pediatric population.
Collapse
Affiliation(s)
- Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Mukta N. Chowta
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Sheetal D. Ullal
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
| | - Youssef Daali
- Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Uppugunduri S. Chakradhara Rao
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal 575001, India
- CANSEARCH Research Platform in Pediatric Oncology and Hematology, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
12
|
Ahire D, Kruger L, Sharma S, Mettu VS, Basit A, Prasad B. Quantitative Proteomics in Translational Absorption, Distribution, Metabolism, and Excretion and Precision Medicine. Pharmacol Rev 2022; 74:769-796. [PMID: 35738681 PMCID: PMC9553121 DOI: 10.1124/pharmrev.121.000449] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A reliable translation of in vitro and preclinical data on drug absorption, distribution, metabolism, and excretion (ADME) to humans is important for safe and effective drug development. Precision medicine that is expected to provide the right clinical dose for the right patient at the right time requires a comprehensive understanding of population factors affecting drug disposition and response. Characterization of drug-metabolizing enzymes and transporters for the protein abundance and their interindividual as well as differential tissue and cross-species variabilities is important for translational ADME and precision medicine. This review first provides a brief overview of quantitative proteomics principles including liquid chromatography-tandem mass spectrometry tools, data acquisition approaches, proteomics sample preparation techniques, and quality controls for ensuring rigor and reproducibility in protein quantification data. Then, potential applications of quantitative proteomics in the translation of in vitro and preclinical data as well as prediction of interindividual variability are discussed in detail with tabulated examples. The applications of quantitative proteomics data in physiologically based pharmacokinetic modeling for ADME prediction are discussed with representative case examples. Finally, various considerations for reliable quantitative proteomics analysis for translational ADME and precision medicine and the future directions are discussed. SIGNIFICANCE STATEMENT: Quantitative proteomics analysis of drug-metabolizing enzymes and transporters in humans and preclinical species provides key physiological information that assists in the translation of in vitro and preclinical data to humans. This review provides the principles and applications of quantitative proteomics in characterizing in vitro, ex vivo, and preclinical models for translational research and interindividual variability prediction. Integration of these data into physiologically based pharmacokinetic modeling is proving to be critical for safe, effective, timely, and cost-effective drug development.
Collapse
Affiliation(s)
- Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Laken Kruger
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Sheena Sharma
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Vijaya Saradhi Mettu
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Abdul Basit
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
13
|
Salem F, Small BG, Johnson TN. Development and application of a pediatric mechanistic kidney model. CPT Pharmacometrics Syst Pharmacol 2022; 11:854-866. [PMID: 35506351 PMCID: PMC9286721 DOI: 10.1002/psp4.12798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Pediatric physiologically‐based pharmacokinetic (P‐PBPK) models have been used to predict age related changes in the pharmacokinetics (PKs) of renally cleared drugs mainly in relation to changes in glomerular filtration rate. With emerging data on ontogeny of renal transporters, mechanistic models of renal clearance accounting for the role of active and passive secretion should be developed and evaluated. Data on age‐related physiological changes and ontogeny of renal transporters were applied into a mechanistic kidney within a P‐PBPK model. Plasma concentration–time profile and PK parameters of cimetidine, ciprofloxacin, metformin, tenofovir, and zidovudine were predicted in subjects aged 1 day to 18 years. The predicted and observed plasma concentration–time profiles and PK parameters were compared. The predicted concentration–time profile means and 5th and 95th percent intervals generally captured the observed data and variability in various studies. Overall, based on drugs and age bands, predicted to observed clearance were all within two‐fold and in 11 of 16 cases within 1.5‐fold. Predicted to observed area under the curve (AUC) and maximum plasma concentration (Cmax) were within two‐fold in 12 of 14 and 12 of 15 cases, respectively. Predictions in neonates and early infants (up to 14 weeks postnatal age) were reasonable with 15–20 predicted PK parameters within two‐fold of the observed. ciprofloxacin but not zidovudine PK predictions were sensitive to basal kidney uptake transporter ontogeny. The results indicate that a mechanistic kidney model accounting for physiology and ontogeny of renal processes and transporters can predict the PK of renally excreted drugs in children. Further data especially in neonates are required to verify the model and ontogeny profiles.
Collapse
Affiliation(s)
- Farzaneh Salem
- Drug Metabolism and Pharmacokinetics GlaxoSmithKline R&D Ware UK
| | | | | |
Collapse
|
14
|
Chothe PP, Mitra P, Nakakariya M, Ramsden D, Rotter CJ, Sandoval P, Tohyama K. Novel Insights in Drug Transporter Sciences: the Year 2021 in Review. Drug Metab Rev 2022; 54:299-317. [PMID: 35762758 DOI: 10.1080/03602532.2022.2094944] [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: 11/03/2022]
Abstract
On behalf of the team I am pleased to present the second annual 'novel insights into drug transporter sciences review' focused on peer-reviewed articles that were published in the year 2021. In compiling the articles for inclusion, preprints available in 2021 but officially published in 2022 were considered to be in scope. To support this review the contributing authors independently selected one or two articles that were thought to be impactful and of interest to the broader research community. A similar approach as published last year was adopted whereby key observations, methods and analysis of each paper is concisely summarized in the synopsis followed by a commentary highlighting the impact of the paper in understanding of drug transporters' role in drug disposition.As the goal of this review is not to provide a comprehensive overview of each paper but rather highlight important findings that are well supported by the data, the reader is encouraged to consult the original articles for additional information. Further, and keeping in line with the goals of this review, it should be noted that all authors actively contributed by writing synopsis and commentary for individual papers and no attempt was made to standardize language or writing styles. In this way, the review article is reflective of not only the diversity of the articles but also that of the contributors. I extend my thanks to the authors for their continued support and also welcome Diane Ramsden and Pallabi Mitra as contributing authors for this issue.
Collapse
Affiliation(s)
- Paresh P Chothe
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), 95 Hayden Avenue, Lexington, Massachusetts, 02421, USA
| | - Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chrome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Diane Ramsden
- Drug Metabolism and Pharmacokinetics, Oncology Research and Development, AstraZeneca, 35 Gate House Park, Waltham, Massachusetts, USA
| | - Charles J Rotter
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), 9625 Towne Centre Drive, San Diego, California, 92121, USA
| | - Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), 95 Hayden Avenue, Lexington, Massachusetts, 02421, USA
| | - Kimio Tohyama
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), 95 Hayden Avenue, Lexington, Massachusetts, 02421, USA
| |
Collapse
|
15
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
16
|
Chu X, Prasad B, Neuhoff S, Yoshida K, Leeder JS, Mukherjee D, Taskar K, Varma MVS, Zhang X, Yang X, Galetin A. Clinical Implications of Altered Drug Transporter Abundance/Function and PBPK Modeling in Specific Populations: An ITC Perspective. Clin Pharmacol Ther 2022; 112:501-526. [PMID: 35561140 DOI: 10.1002/cpt.2643] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022]
Abstract
The role of membrane transporters on pharmacokinetics (PKs), drug-drug interactions (DDIs), pharmacodynamics (PDs), and toxicity of drugs has been broadly recognized. However, our knowledge of modulation of transporter expression and/or function in the diseased patient population or specific populations, such as pediatrics or pregnancy, is still emerging. This white paper highlights recent advances in studying the changes in transporter expression and activity in various diseases (i.e., renal and hepatic impairment and cancer) and some specific populations (i.e., pediatrics and pregnancy) with the focus on clinical implications. Proposed alterations in transporter abundance and/or activity in diseased and specific populations are based on (i) quantitative transporter proteomic data and relative abundance in specific populations vs. healthy adults, (ii) clinical PKs, and emerging transporter biomarker and/or pharmacogenomic data, and (iii) physiologically-based pharmacokinetic modeling and simulation. The potential for altered PK, PD, and toxicity in these populations needs to be considered for drugs and their active metabolites in which transporter-mediated uptake/efflux is a major contributor to their absorption, distribution, and elimination pathways and/or associated DDI risk. In addition to best practices, this white paper discusses current challenges and knowledge gaps to study and quantitatively predict the effects of modulation in transporter activity in these populations, together with the perspectives from the International Transporter Consortium (ITC) on future directions.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | | | - Kenta Yoshida
- Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, California, USA
| | - James Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Dwaipayan Mukherjee
- Clinical Pharmacology & Pharmacometrics, Research & Development, AbbVie, Inc., North Chicago, Illinois, USA
| | | | - Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Ahire D, Basit A, Christopher LJ, Iyer R, Leeder JS, Prasad B. Interindividual Variability and Differential Tissue Abundance of Mitochondrial Amidoxime Reducing Component Enzymes in Humans. Drug Metab Dispos 2022; 50:191-196. [PMID: 34949674 PMCID: PMC8969132 DOI: 10.1124/dmd.121.000805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial amidoxime-reducing component (mARC) enzymes are molybdenum-containing proteins that metabolize a number of endobiotics and xenobiotics. The interindividual variability and differential tissue abundance of mARC1 and mARC2 were quantified using targeted proteomics in three types of tissue fractions: 1) pediatric liver tissue homogenates, 2) total membrane fraction of the paired liver and kidney samples from pediatric and adult donors, and 3) pooled S9 fractions of the liver, intestine, kidney, lung, and heart. The absolute levels of mARC1 and mARC2 in the pediatric liver homogenate were 40.08 ± 4.26 and 24.58 ± 4.02 pmol/mg homogenate protein, respectively, and were independent of age and sex. In the total membrane fraction of the paired liver and kidney samples, the abundance of hepatic mARC1 and mARC2 was comparable, whereas mARC2 abundance in the kidney was approximately 9-fold higher in comparison with mARC1. The analysis of the third set of samples (i.e., S9 fraction) revealed that mARC1 abundance in the kidney, intestine, and lung was 5- to 13-fold lower than the liver S9 abundance, whereas mARC2 abundance was approximately 3- and 16-fold lower in the intestine and lung than the liver S9, respectively. In contrast, the kidney mARC2 abundance in the S9 fraction was approximately 2.5-fold higher as compared with the hepatic mARC2 abundance. The abundance of mARC enzymes in the heart was below the limit of quantification (∼0.6 pmol/mg protein). The mARC enzyme abundance data presented here can be used to develop physiologically based pharmacokinetic models for the prediction of in vivo pharmacokinetics of mARC substrates. SIGNIFICANCE STATEMENT: A precise targeted quantitative proteomics method was developed and applied to quantify newly discovered drug-metabolizing enzymes, mARC1 and mARC2, in pediatric and adult tissue samples. The data suggest that mARC enzymes are ubiquitously expressed in an isoform-specific manner in the human liver, kidney, intestine, and lung, and the enzyme abundance is not associated with age and sex. These data are important for developing physiologically based pharmacokinetic models for the prediction of in vivo pharmacokinetics of mARC substrates.
Collapse
Affiliation(s)
- Deepak Ahire
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| | - Abdul Basit
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| | - Lisa J Christopher
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| | - Ramaswamy Iyer
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| | - J Steven Leeder
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (D.A., A.B., B.P.); Department of Nonclinical Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey (L.J.C., R.I.); and Department of Pediatrics, Children's Mercy Hospitals and Clinics, Kansas City, Missouri (J.S.L.)
| |
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: 48] [Impact Index Per Article: 16.0] [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
|
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
|
20
|
Streekstra EJ, Russel FGM, van de Steeg E, de Wildt SN. Application of proteomics to understand maturation of drug metabolizing enzymes and transporters for the optimization of pediatric drug therapy. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 39:31-48. [PMID: 34906324 DOI: 10.1016/j.ddtec.2021.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/22/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
Drug disposition in children is different compared to adults. Growth and developmental change the processes involved in drug disposition and efficacy, including membrane transporters and drug metabolizing enzymes, but for many of these proteins, the exact changes have not been fully elucidated to date. Quantitative proteomics offers a solution to analyze many DME and DT proteins at once and can be performed with very small tissue samples, overcoming many of the challenges previously limiting research in this pediatric field. Liquid chromatography tandem mass spectrometry (LC-MS/MS) based methods for quantification of (membrane) proteins has evolved as a golden standard for proteomic analysis. The last years, big steps have been made in maturation studies of hepatic and renal drug transporters and drug metabolizing enzymes using this method. Protein and organ specific maturation patterns have been identified for the human liver and kidney, which aids pharmacological modelling and predicting drug dosing in the pediatric population. Further research should focus on other organs, like intestine and brain, as well as on innovative methods in which proteomics can be used to further overcome the limited access to pediatric tissues, including liquid biopsies and organoids. In this review there is aimed to provide an overview of available human pediatric proteomics data, discuss its challenges and provide guidance for future research.
Collapse
Affiliation(s)
- Eva J Streekstra
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands
| | | | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein 21, Nijmegen 6525 EZ, The Netherlands; Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children Hospital, Wytemaweg 50, 3011 CN Rotterdam, The Netherlands.
| |
Collapse
|
21
|
Kiss M, Mbasu R, Nicolaï J, Barnouin K, Kotian A, Mooij MG, Kist N, Wijnen RMH, Ungell AL, Cutler P, Russel FGM, de Wildt SN. Ontogeny of Small Intestinal Drug Transporters and Metabolizing Enzymes Based on Targeted Quantitative Proteomics. Drug Metab Dispos 2021; 49:1038-1046. [PMID: 34548392 DOI: 10.1124/dmd.121.000559] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/13/2021] [Indexed: 01/16/2023] Open
Abstract
Most drugs are administered to children orally. An information gap remains on the protein abundance of small intestinal drug-metabolizing enzymes (DMEs) and drug transporters (DTs) across the pediatric age range, which hinders precision dosing in children. To explore age-related differences in DMEs and DTs, surgical leftover intestinal tissues from pediatric and adult jejunum and ileum were collected and analyzed by targeted quantitative proteomics for apical sodium-bile acid transporter, breast cancer resistance protein (BCRP), monocarboxylate transporter 1 (MCT1), multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein (MRP) 2, MRP3, organic anion-transporting polypeptide 2B1, organic cation transporter 1, peptide transporter 1 (PEPT1), CYP2C19, CYP3A4, CYP3A5, UDP glucuronosyltransferase (UGT) 1A1, UGT1A10, and UGT2B7. Samples from 58 children (48 ileums, 10 jejunums, age range: 8 weeks to 17 years) and 16 adults (8 ileums, 8 jejunums) were analyzed. When comparing age groups, BCRP, MDR1, PEPT1, and UGT1A1 abundance was significantly higher in adult ileum as compared with the pediatric ileum. Jejunal BCRP, MRP2, UGT1A1, and CYP3A4 abundance was higher in the adults compared with children 0-2 years of age. Examining the data on a continuous age scale showed that PEPT1 and UGT1A1 abundance was significantly higher, whereas MCT1 and UGT2B7 abundance was lower in adult ileum as compared with the pediatric ileum. Our data contribute to the deeper understanding of the ontogeny of small intestinal drug-metabolizing enzymes and drug transporters and shows DME-, DT-, and intestinal location-specific, age-related changes. SIGNIFICANCE STATEMENT: This is the first study that describes the ontogeny of small intestinal DTs and DMEs in human using liquid chromatography with tandem mass spectrometry-based targeted quantitative proteomics. The current analysis provides a detailed picture about the maturation of DT and DME abundances in the human jejunum and ileum. The presented results supply age-related DT and DME abundance data for building more accurate PBPK models that serve to support safer and more efficient drug dosing regimens for the pediatric population.
Collapse
Affiliation(s)
- Márton Kiss
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Richard Mbasu
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Johan Nicolaï
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Karin Barnouin
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Apoorva Kotian
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Miriam G Mooij
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Nico Kist
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Rene M H Wijnen
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Anna-Lena Ungell
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Paul Cutler
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| | - Saskia N de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.K., F.G.M.R., S.N.d.W.); Development Science (R.M., K.B., A.K., P.C.), and Statistical Sciences and Innovation (N.K.), UCB BioPharma, Slough, United Kingdom; Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N., A.-L.U.); Department of Pediatrics, Willem-Alexander Children's Hospital, Leiden University Medical Centre, Leiden, The Netherlands (M.G.M.); and Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (R.M.H.W.)
| |
Collapse
|
22
|
Al-Majdoub ZM, Scotcher D, Achour B, Barber J, Galetin A, Rostami-Hodjegan A. Quantitative Proteomic Map of Enzymes and Transporters in the Human Kidney: Stepping Closer to Mechanistic Kidney Models to Define Local Kinetics. Clin Pharmacol Ther 2021; 110:1389-1400. [PMID: 34390491 DOI: 10.1002/cpt.2396] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/03/2021] [Indexed: 12/20/2022]
Abstract
The applications of translational modeling of local drug concentrations in various organs had a sharp increase over the last decade. These are part of the model-informed drug development initiative, adopted by the pharmaceutical industry and promoted by drug regulatory agencies. With respect to the kidney, the models serve as a bridge for understanding animal vs. human observations related to renal drug disposition and any consequential adverse effects. However, quantitative data on key drug-metabolizing enzymes and transporters relevant for predicting renal drug disposition are limited. Using targeted and global quantitative proteomics, we determined the abundance of multiple enzymes and transporters in 20 human kidney cortex samples. Nine enzymes and 22 transporters were quantified (8 for the first time in the kidneys). In addition, > 4,000 proteins were identified and used to form an open database. CYP2B6, CYP3A5, and CYP4F2 showed comparable, but generally low expression, whereas UGT1A9 and UGT2B7 levels were the highest. Significant correlation between abundance and activity (measured by mycophenolic acid clearance) was observed for UGT1A9 (Rs = 0.65, P = 0.004) and UGT2B7 (Rs = 0.70, P = 0.023). Expression of P-gp ≈ MATE-1 and OATP4C1 transporters were high. Strong intercorrelations were observed between several transporters (P-gp/MRP4, MRP2/OAT3, and OAT3/OAT4); no correlation in expression was apparent for functionally related transporters (OCT2/MATEs). This study extends our knowledge of pharmacologically relevant proteins in the kidney cortex, with implications on more prudent use of mechanistic kidney models under the general framework of quantitative systems pharmacology and toxicology.
Collapse
Affiliation(s)
- Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Certara UK (Simcyp Division), Sheffield, UK
| |
Collapse
|
23
|
van Groen BD, Nicolaï J, Kuik AC, Van Cruchten S, van Peer E, Smits A, Schmidt S, de Wildt SN, Allegaert K, De Schaepdrijver L, Annaert P, Badée J. Ontogeny of Hepatic Transporters and Drug-Metabolizing Enzymes in Humans and in Nonclinical Species. Pharmacol Rev 2021; 73:597-678. [PMID: 33608409 DOI: 10.1124/pharmrev.120.000071] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver represents a major eliminating and detoxifying organ, determining exposure to endogenous compounds, drugs, and other xenobiotics. Drug transporters (DTs) and drug-metabolizing enzymes (DMEs) are key determinants of disposition, efficacy, and toxicity of drugs. Changes in their mRNA and protein expression levels and associated functional activity between the perinatal period until adulthood impact drug disposition. However, high-resolution ontogeny profiles for hepatic DTs and DMEs in nonclinical species and humans are lacking. Meanwhile, increasing use of physiologically based pharmacokinetic (PBPK) models necessitates availability of underlying ontogeny profiles to reliably predict drug exposure in children. In addition, understanding of species similarities and differences in DT/DME ontogeny is crucial for selecting the most appropriate animal species when studying the impact of development on pharmacokinetics. Cross-species ontogeny mapping is also required for adequate translation of drug disposition data in developing nonclinical species to humans. This review presents a quantitative cross-species compilation of the ontogeny of DTs and DMEs relevant to hepatic drug disposition. A comprehensive literature search was conducted on PubMed Central: Tables and graphs (often after digitization) in original manuscripts were used to extract ontogeny data. Data from independent studies were standardized and normalized before being compiled in graphs and tables for further interpretation. New insights gained from these high-resolution ontogeny profiles will be indispensable to understand cross-species differences in maturation of hepatic DTs and DMEs. Integration of these ontogeny data into PBPK models will support improved predictions of pediatric hepatic drug disposition processes. SIGNIFICANCE STATEMENT: Hepatic drug transporters (DTs) and drug-metabolizing enzymes (DMEs) play pivotal roles in hepatic drug disposition. Developmental changes in expression levels and activities of these proteins drive age-dependent pharmacokinetics. This review compiles the currently available ontogeny profiles of DTs and DMEs expressed in livers of humans and nonclinical species, enabling robust interpretation of age-related changes in drug disposition and ultimately optimization of pediatric drug therapy.
Collapse
Affiliation(s)
- B D van Groen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Nicolaï
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A C Kuik
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Van Cruchten
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - E van Peer
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A Smits
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Schmidt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S N de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - K Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - L De Schaepdrijver
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - P Annaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Badée
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| |
Collapse
|
24
|
Ahire DS, Basit A, Karasu M, Prasad B. Ultrasensitive Quantification of Drug-metabolizing Enzymes and Transporters in Small Sample Volume by Microflow LC-MS/MS. J Pharm Sci 2021; 110:2833-2840. [PMID: 33785352 DOI: 10.1016/j.xphs.2021.03.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 01/31/2023]
Abstract
Protein abundance data of drug-metabolizing enzymes and transporters (DMETs) are broadly applicable to the characterization of in vitro and in vivo models, in vitro to in vivo extrapolation (IVIVE), and interindividual variability prediction. However, the emerging need of DMET quantification in small sample volumes such as organ-on a chip effluent, organoids, and biopsies requires ultrasensitive protein quantification methods. We present an ultrasensitive method that relies on an optimized sample preparation approach involving acetone precipitation coupled with a microflow-based liquid chromatography-tandem mass spectrometry (µLC-MS/MS) for the DMET quantification using limited sample volume or protein concentration, i.e., liver tissues (1-100 mg), hepatocyte counts (~4000 to 1 million cells), and microsomal protein concentration (0.01-1 mg/ml). The method was applied to quantify DMETs in differential tissue S9 fractions (liver, intestine, kidney, lung, and heart) and cryopreserved human intestinal mucosa (i.e., CHIM). The method successfully quantified >75% of the target DMETs in the trypsin digests of 1 mg tissue homogenate, 15,000 hepatocytes, and 0.06 mg/ml microsomal protein concentration. The precision of DMET quantification measured as the coefficient of variation across different tissue weights, cell counts, or microsomal protein concentration was within 30%. The method confirmed significant extrahepatic abundance of non-cytochrome P450 enzymes such as dihydropyridine dehydrogenase (DPYD), epoxide hydrolases (EPXs), arylacetamide deacetylase (AADAC), paraoxonases (PONs), and glutathione S-transferases (GSTs). The ultrasensitive method developed here is applicable to characterize emerging miniaturized in vitro models and small volume biopsies. In addition, the differential tissue abundance data of the understudied DMETs will be important for physiologically-based pharmacokinetic (PBPK) modeling of drugs.
Collapse
Affiliation(s)
- Deepak Suresh Ahire
- Department of Pharmaceutical Sciences, Washington State University, 12 E Spokane Falls Blvd, Spokane, WA 99202, USA
| | - Abdul Basit
- Department of Pharmaceutical Sciences, Washington State University, 12 E Spokane Falls Blvd, Spokane, WA 99202, USA
| | - Matthew Karasu
- Department of Pharmaceutical Sciences, Washington State University, 12 E Spokane Falls Blvd, Spokane, WA 99202, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, 12 E Spokane Falls Blvd, Spokane, WA 99202, USA.
| |
Collapse
|
25
|
Naji-Talakar S, Sharma S, Martin LA, Barnhart D, Prasad B. Potential implications of DMET ontogeny on the disposition of commonly prescribed drugs in neonatal and pediatric intensive care units. Expert Opin Drug Metab Toxicol 2021; 17:273-289. [PMID: 33256492 PMCID: PMC8346204 DOI: 10.1080/17425255.2021.1858051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/27/2020] [Indexed: 10/22/2022]
Abstract
Introduction: Pediatric patients, especially neonates and infants, are more susceptible to adverse drug events as compared to adults. In particular, immature small molecule drug metabolism and excretion can result in higher incidences of pediatric toxicity than adults if the pediatric dose is not adjusted.Area covered: We reviewed the top 29 small molecule drugs prescribed in neonatal and pediatric intensive care units and compiled the mechanisms of their metabolism and excretion. The ontogeny of Phase I and II drug metabolizing enzymes and transporters (DMETs), particularly relevant to these drugs, are summarized. The potential effects of DMET ontogeny on the metabolism and excretion of the top pediatric drugs were predicted. The current regulatory requirements and recommendations regarding safe and effective use of drugs in children are discussed. A few representative examples of the use of ontogeny-informed physiologically based pharmacokinetic (PBPK) models are highlighted.Expert opinion: Empirical prediction of pediatric drug dosing based on body weight or body-surface area from the adult parameters can be inaccurate because DMETs are not mature in children and the age-dependent maturation of these proteins is different. Ontogeny-informed-PBPK modeling provides a better alternative to predict the pharmacokinetics of drugs in children.
Collapse
Affiliation(s)
- Siavosh Naji-Talakar
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Sheena Sharma
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Leslie A. Martin
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Derek Barnhart
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| |
Collapse
|
26
|
Ma HY, Chen S, Du Y. Estrogen and estrogen receptors in kidney diseases. Ren Fail 2021; 43:619-642. [PMID: 33784950 PMCID: PMC8018493 DOI: 10.1080/0886022x.2021.1901739] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are posing great threats to global health within this century. Studies have suggested that estrogen and estrogen receptors (ERs) play important roles in many physiological processes in the kidney. For instance, they are crucial in maintaining mitochondrial homeostasis and modulating endothelin-1 (ET-1) system in the kidney. Estrogen takes part in the kidney repair and regeneration via its receptors. Estrogen also participates in the regulation of phosphorus homeostasis via its receptors in the proximal tubule. The ERα polymorphisms have been associated with the susceptibilities and outcomes of several renal diseases. As a consequence, the altered or dysregulated estrogen/ERs signaling pathways may contribute to a variety of kidney diseases, including various causes-induced AKI, diabetic kidney disease (DKD), lupus nephritis (LN), IgA nephropathy (IgAN), CKD complications, etc. Experimental and clinical studies have shown that targeting estrogen/ERs signaling pathways might have protective effects against certain renal disorders. However, many unsolved problems still exist in knowledge regarding the roles of estrogen and ERs in distinct kidney diseases. Further research is needed to shed light on this area and to enable the discovery of pathway-specific therapies for kidney diseases.
Collapse
Affiliation(s)
- Hao-Yang Ma
- Department of Geriatrics, Second Affiliated Hospital of Zhejiang University, Hangzhou, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Chen
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Du
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
27
|
Al‐Majdoub ZM, Achour B, Couto N, Howard M, Elmorsi Y, Scotcher D, Alrubia S, El‐Khateeb E, Vasilogianni A, Alohali N, Neuhoff S, Schmitt L, Rostami‐Hodjegan A, Barber J. Mass spectrometry-based abundance atlas of ABC transporters in human liver, gut, kidney, brain and skin. FEBS Lett 2020; 594:4134-4150. [PMID: 33128234 PMCID: PMC7756589 DOI: 10.1002/1873-3468.13982] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
ABC transporters (ATP-binding cassette transporter) traffic drugs and their metabolites across membranes, making ABC transporter expression levels a key factor regulating local drug concentrations in different tissues and individuals. Yet, quantification of ABC transporters remains challenging because they are large and low-abundance transmembrane proteins. Here, we analysed 200 samples of crude and membrane-enriched fractions from human liver, kidney, intestine, brain microvessels and skin, by label-free quantitative mass spectrometry. We identified 32 (out of 48) ABC transporters: ABCD3 was the most abundant in liver, whereas ABCA8, ABCB2/TAP1 and ABCE1 were detected in all tissues. Interestingly, this atlas unveiled that ABCB2/TAP1 may have TAP2-independent functions in the brain and that biliary atresia (BA) and control livers have quite different ABC transporter profiles. We propose that meaningful biological information can be derived from a direct comparison of these data sets.
Collapse
Affiliation(s)
- Zubida M. Al‐Majdoub
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Narciso Couto
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Martyn Howard
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Yasmine Elmorsi
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Sarah Alrubia
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Chemistry DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Eman El‐Khateeb
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | | | - Noura Alohali
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Practice DepartmentCollege of PharmacyPrincess Noura Bint Abdul Rahman UniversityRiyadhSaudi Arabia
| | | | - Lutz Schmitt
- Institute of BiochemistryHeinrich Heine University DüsseldorfGermany
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Simcyp DivisionCertara UK LtdSheffieldUK
| | - Jill Barber
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| |
Collapse
|
28
|
Sharma S, Suresh Ahire D, Prasad B. Utility of Quantitative Proteomics for Enhancing the Predictive Ability of Physiologically Based Pharmacokinetic Models Across Disease States. J Clin Pharmacol 2020; 60 Suppl 1:S17-S35. [DOI: 10.1002/jcph.1709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Sheena Sharma
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| | - Deepak Suresh Ahire
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences Washington State University Spokane Washington USA
| |
Collapse
|
29
|
Salem F, Johnson TN, Hodgkinson ABJ, Ogungbenro K, Rostami‐Hodjegan A. Does "Birth" as an Event Impact Maturation Trajectory of Renal Clearance via Glomerular Filtration? Reexamining Data in Preterm and Full-Term Neonates by Avoiding the Creatinine Bias. J Clin Pharmacol 2020; 61:159-171. [PMID: 32885464 PMCID: PMC7818478 DOI: 10.1002/jcph.1725] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Glomerular filtration rate (GFR) is an important measure of renal function. Various models for its maturation have recently been compared; however, these have used markers, which are subject to different renal elimination processes. Inulin clearance data (a purer probe of GFR) collected from the literature were used to determine age‐related changes in GFR aspects of renal drug excretion in pediatrics. An ontogeny model was derived using a best‐fit model with various combinations of covariates such as postnatal age, gestational age at birth, and body weight. The model was applied to the prediction of systemic clearance of amikacin, gentamicin, vancomycin, and gadobutrol. During neonatal life, GFR increased as a function of both gestational age at birth and postnatal age, hence implying an impact of birth and a discrepancy in GFR for neonates with the same postmenstrual age depending on gestational age at birth (ie, neonates who were outside the womb longer had higher GFR, on average). The difference in GFR between pre‐term and full‐term neonates with the same postmenstrual age was negligible from beyond 1.25 years. Considering both postnatal age and gestational age at birth in GFR ontogeny models is important because postmenstrual age alone ignores the impact of birth. Most GFR models use covariates of body size in addition to age. Therefore, prediction from these models will also depend on the change in anthropometric characteristics with age. The latter may not be similar in various ethnic groups, and this makes the head‐to‐head comparison of models very challenging.
Collapse
Affiliation(s)
| | | | | | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic ResearchDivision of Pharmacy and OptometrySchool of Health SciencesFaculty of BiologyMedicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Amin Rostami‐Hodjegan
- Certara UK Ltd, Simcyp DivisionSheffieldUK
- Centre for Applied Pharmacokinetic ResearchDivision of Pharmacy and OptometrySchool of Health SciencesFaculty of BiologyMedicine and HealthManchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
30
|
Kumar AR, Prasad B, Bhatt DK, Mathialagan S, Varma MVS, Unadkat JD. In Vivo-to-In Vitro Extrapolation of Transporter-Mediated Renal Clearance: Relative Expression Factor Versus Relative Activity Factor Approach. Drug Metab Dispos 2020; 49:470-478. [PMID: 33824168 PMCID: PMC8232064 DOI: 10.1124/dmd.121.000367] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
About 30% of approved drugs are cleared predominantly by renal clearance (CLr). Of these, many are secreted by transporters. For these drugs, in vitro-to-in vivo extrapolation of transporter-mediated renal secretory clearance (CLsec,plasma) is important to prospectively predict their renal clearance and to assess the impact of drug-drug interactions and pharmacogenetics on their pharmacokinetics. Here we compared the ability of the relative expression factor (REF) and the relative activity factor (RAF) approaches to quantitatively predict the in vivo CLsec,plasma of 26 organic anion transporter (OAT) substrates assuming that OAT-mediated uptake is the rate-determining step in the CLsec,plasma of the drugs. The REF approach requires protein quantification of each transporter in the tissue (e.g., kidney) and transporter-expressing cells, whereas the RAF approach requires the use of a transporter-selective probe substrate (both in vitro and in vivo) for each transporter of interest. For the REF approach, 50% and 69% of the CLsec,plasma predictions were within 2- and 3-fold of the observed values, respectively; the corresponding values for the RAF approach were 65% and 81%. We found no significant difference between the two approaches in their predictive capability (as measured by accuracy and bias) of the CLsec,plasma or CLr of OAT drugs. We recommend that the REF and RAF approaches can be used interchangeably to predict OAT-mediated CLsec,plasma Further research is warranted to evaluate the ability of the REF or RAF approach to predict CLsec,plasma of drugs when uptake is not the rate-determining step. SIGNIFICANCE STATEMENT: This is the first direct comparison of the relative expression factor (REF) and relative activity factor (RAF) approaches to predict transporter-mediated renal clearance (CLr). The RAF, but not REF, approach requires transporter-selective probes and that the basolateral uptake is the rate-determining step in the CLr of drugs. Given that there is no difference in predictive capability of the REF and RAF approach for organic anion transporter-mediated CLr, the REF approach should be explored further to assess its ability to predict CLr when basolateral uptake is not the sole rate-determining step.
Collapse
Affiliation(s)
- Aditya R Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| | - Deepak Kumar Bhatt
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| | - Sumathy Mathialagan
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| | - Manthena V S Varma
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (A.R.K., B.P., D.K.B., J.D.U.); and Pharmacokinetics, Pharmacodynamics, and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut (S.M., M.V.S.V.)
| |
Collapse
|
31
|
Huang L, Liao J, He J, Pan S, Zhang H, Yang X, Cheng J, Chen Y, Mo Z. Single-cell profiling reveals sex diversity in human renal proximal tubules. Gene 2020; 752:144790. [PMID: 32439376 DOI: 10.1016/j.gene.2020.144790] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 01/11/2023]
Abstract
Many anatomical regions in the kidney, including proximal tubules, differ between males and females. While such differences in renal structures and functions under various physiological and pharmacological conditions have been identified, information relating to molecular mechanisms behind this gender disparity remain unknown. To understand gene expression differences in proximal tubules from human male and female kidneys, we reported on kidney cellular landscape using single-cell RNA sequencing. Differential gene expression profiles were observed in proximal tubules, between the sexes. Interestingly, the SLC22 family of anion transporters, including SLC22A6 and SLC22A8, had different expression profiles between male and female proximal tubule clusters but not sex-dependent abundance at the protein level. Moreover, in different species, we revealed a shared and species-specific differential gene expression between human and mouse kidney proximal tubules. Taken together, at single-cell resolution, this transcriptomic map represents a baseline description of gender biased genes in human kidney proximal tubules, which provide important insights for further studies of physiological differences in kidney.
Collapse
Affiliation(s)
- Lin Huang
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jinling Liao
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Juan He
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Siqiong Pan
- Department of Pathology, The Four Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiwen Cheng
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yang Chen
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
| | - Zengnan Mo
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China; Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
32
|
Zhang H, Basit A, Wolford C, Chen KF, Gaedigk A, Lin YS, Leeder JS, Prasad B. Normalized Testosterone Glucuronide as a Potential Urinary Biomarker for Highly Variable UGT2B17 in Children 7-18 Years. Clin Pharmacol Ther 2020; 107:1149-1158. [PMID: 31900930 DOI: 10.1002/cpt.1764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Abstract
UDP-glucuronosyltransferase 2B17 (UGT2B17) is a highly variable androgen-metabolizing and drug-metabolizing enzyme. UGT2B17 exhibits a unique ontogeny profile characterized by a dramatic increase in hepatic protein expression from prepubertal age to adulthood. Age, sex, copy number variation (CNV), and single nucleotide polymorphisms only explain 26% of variability in protein expression, highlighting the need for a phenotypic biomarker for predicting interindividual variability in glucuronidation of UGT2B17 substrates. Here, we propose testosterone glucuronide (TG) normalized by androsterone glucuronide (TG/AG) as a urinary UGT2B17 biomarker, and examine the associations among urinary TG/AG and age, sex, and CNV. We performed targeted metabolomics of 12 androgen conjugates with liquid-chromatography tandem mass spectrometry in 63 pediatric subjects ages 7-18 years followed over 7 visits in 3 years. Consistent with the reported developmental trajectory of UGT2B17 protein expression, urinary TG/AG is significantly associated with age, sex, and CNV. In conclusion, TG/AG shows promise as a phenotypic urinary UGT2B17 biomarker.
Collapse
Affiliation(s)
- Haeyoung Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Abdul Basit
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Chris Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kuan-Fu Chen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Yvonne S Lin
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
33
|
Protein Abundance of Clinically Relevant Drug Transporters in The Human Kidneys. Int J Mol Sci 2019; 20:ijms20215303. [PMID: 31653114 PMCID: PMC6862022 DOI: 10.3390/ijms20215303] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Renal drug transporters such as the organic cation transporters (OCTs), organic anion transporters (OATs) and multidrug resistance proteins (MRPs) play an important role in the tubular secretion of many drugs influencing their efficacy and safety. However, only little is known about the distinct protein abundance of these transporters in human kidneys, and about the impact of age and gender as potential factors of inter-subject variability in their expression and function. The aim of this study was to determine the protein abundance of MDR1, MRP1-4, BCRP, OAT1-3, OCT2-3, MATE1, PEPT1/2, and ORCTL2 by liquid chromatography-tandem mass spectrometry-based targeted proteomics in a set of 36 human cortex kidney samples (20 males, 16 females; median age 53 and 55 years, respectively). OAT1 and 3, OCT2 and ORCTL2 were found to be most abundant renal SLC transporters while MDR1, MRP1 and MRP4 were the dominating ABC transporters. Only the expression levels of MDR1 and ORCTL2 were significantly higher abundant in older donors. Moreover, we found several significant correlations between different transporters, which may indicate their functional interplay in renal vectorial transport processes. Our data may contribute to a better understanding of the molecular processes determining renal excretion of drugs.
Collapse
|
34
|
van der Made TK, Fedecostante M, Scotcher D, Rostami-Hodjegan A, Sastre Toraño J, Middel I, Koster AS, Gerritsen KG, Jankowski V, Jankowski J, Hoenderop JGJ, Masereeuw R, Galetin A. Quantitative Translation of Microfluidic Transporter in Vitro Data to in Vivo Reveals Impaired Albumin-Facilitated Indoxyl Sulfate Secretion in Chronic Kidney Disease. Mol Pharm 2019; 16:4551-4562. [PMID: 31525064 DOI: 10.1021/acs.molpharmaceut.9b00681] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Indoxyl sulfate (IxS), a highly albumin-bound uremic solute, accumulates in chronic kidney disease (CKD) due to reduced renal clearance. This study was designed to specifically investigate the role of human serum albumin (HSA) in IxS renal secretion via organic anion transporter 1 (OAT1) in a microfluidic system and subsequently apply quantitative translation of in vitro data to predict extent of change in IxS renal clearance in CKD stage IV relative to healthy. Conditionally immortalized human proximal tubule epithelial cells overexpressing OAT1 were incubated with IxS (5-200 μM) in the HSA-free medium or in the presence of either HSA or CKD-modified HSA. IxS uptake in the presence of HSA resulted in more than 20-fold decrease in OAT1 affinity (Km,u) and 37-fold greater in vitro unbound intrinsic clearance (CLint,u) versus albumin-free condition. In the presence of CKD-modified albumin, Km,u increased four-fold and IxS CLint,u decreased almost seven-fold relative to HSA. Fold-change in parameters exceeded differences in IxS binding between albumin conditions, indicating additional mechanism and facilitating role of albumin in IxS OAT1-mediated uptake. Quantitative translation of IxS in vitro OAT1-mediated CLint,u predicted a 60% decrease in IxS renal elimination as a result of CKD, in agreement with the observed data (80%). The findings of the current study emphasize the role of albumin in IxS transport via OAT1 and explored the impact of modifications in albumin on renal excretion via active secretion in CKD. For the first time, this study performed quantitative translation of transporter kinetic data generated in a novel microfluidic in vitro system to a clinically relevant setting. Knowledge gaps and future directions in quantitative translation of renal drug disposition from microphysiological systems are discussed.
Collapse
Affiliation(s)
- Thomas K van der Made
- Centre for Applied Pharmacokinetic Research, School of Health Sciences , The University of Manchester , Manchester M13 9PL , U.K
| | | | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences , The University of Manchester , Manchester M13 9PL , U.K
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences , The University of Manchester , Manchester M13 9PL , U.K.,Simcyp Division , Certara UK Limited , Sheffield S1 2BJ , U.K
| | | | | | | | - Karin G Gerritsen
- Department of Nephrology and Hypertension , University Medical Center Utrecht , Utrecht 3508 GA , The Netherlands
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research , RWTH Aachen University Hospital , Aachen 52074 , Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research , RWTH Aachen University Hospital , Aachen 52074 , Germany.,School for Cardiovascular Diseases , Maastricht University , Universiteitssingel 50 , Maastricht 6229 ER , The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences , Radboud University Medical Center , Nijmegen 6500 HB , The Netherlands
| | | | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences , The University of Manchester , Manchester M13 9PL , U.K
| |
Collapse
|