1
|
Ávila M, Mora Sánchez MG, Bernal Amador AS, Paniagua R. The Metabolism of Creatinine and Its Usefulness to Evaluate Kidney Function and Body Composition in Clinical Practice. Biomolecules 2025; 15:41. [PMID: 39858438 PMCID: PMC11764249 DOI: 10.3390/biom15010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Serum creatinine levels are the most used clinical marker to estimate renal function as the glomerular function rate because it is simple, fast, and inexpensive. However, creatinine has limitations, as its levels can be influenced by factors such as advanced age, physical activity, protein-rich diets, male gender, medications, and ethnicity. Serum cystatin C and its combination with serum creatinine may serve as an alternative since these factors do not affect it. Most creatinine synthesis occurs in the muscles, making it a valuable marker for assessing lean body mass within body composition. This measurement is crucial for evaluating and monitoring nutritional status in patients with chronic kidney disease. This review aimed to discuss the literature on creatinine metabolism, its advantages and disadvantages in assessing renal function, and its utility in measuring lean body mass. The variability in the creatinine generation rate among individuals should be considered when assessing the glomerular function rate.
Collapse
Affiliation(s)
- Marcela Ávila
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Hospital de Especialidades, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
| | | | | | - Ramón Paniagua
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Hospital de Especialidades, CMN SXXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
| |
Collapse
|
2
|
Sponfeldner MI, Andrikyan W, Maas R, Fromm MF. Pseudo-Worsening of Kidney Function Due to Inhibition of Renal Creatinine Secretion: Quality of Information Provided in Prescribing Information/SmPC. Clin Pharmacol Ther 2024; 116:1259-1268. [PMID: 38989645 DOI: 10.1002/cpt.3374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
Determination of serum creatinine concentrations and subsequent calculation of estimated glomerular filtration rates (eGFR) is a cornerstone of clinical medicine. Crucial clinical decisions such as drug treatment discontinuations are based on eGFR calculated from serum creatinine measurements. However, creatinine is not only filtered in the kidneys, but also actively secreted into urine. Creatinine transporters such as OCT2, OCT3, MATE1, MATE2-K, and OAT2 expressed in proximal tubular cells are responsible for active renal secretion of creatinine. Multiple drugs (e.g., oral antitumor drugs) inhibit these transporters thereby causing a pseudo-worsening of kidney function with an increase in serum creatinine concentrations and a decrease in eGFR while other methods for eGFR determination (e.g., by cystatin C) reveal normal kidney function. Since US Prescribing Information (PI) and European Summaries of Product Characteristics (SmPCs) are the most relevant source of information for physicians, we investigated the quality of information in US PI/German SmPCs of drugs with clear evidence for pseudo-worsening of kidney function. 514 drugs putatively interacting with creatinine transporters were identified. For 149 of those drugs, an increase in serum creatinine concentrations has been described. Available data confirmed the existence of pseudo-worsening of kidney function for 30 of those drugs, for the remaining 119 drugs existing data are insufficient. Only 23.5% (12/51) of the 30 drugs' PI/SmPCs contained unambiguous statements on this proven pseudo-worsening of kidney function and gave clear recommendations for clinical management. Taken together, inadequate information provided in PI or SmPCs on the pseudo-worsening of kidney function poses patients at unnecessary risks.
Collapse
Affiliation(s)
- Michael I Sponfeldner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Wahram Andrikyan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW - Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
3
|
Izzedine H, Bouderlique E, Besse B. Selpercatinib and Pseudo-Decreases in Kidney Function. N Engl J Med 2024; 390:1241-1243. [PMID: 38598583 DOI: 10.1056/nejmc2400216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
|
4
|
Chen MF, Harada G, Liu D, DeMatteo R, Falcon C, Wilhelm C, Kris MG, Drilon A, Gutgarts V. Brief Report: Tyrosine Kinase Inhibitors for Lung Cancers That Inhibit MATE-1 Can Lead to "False" Decreases in Renal Function. J Thorac Oncol 2024; 19:153-159. [PMID: 37748692 PMCID: PMC10841070 DOI: 10.1016/j.jtho.2023.09.1444] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Select tyrosine kinase inhibitors (TKIs) used to treat oncogene-driven lung cancers also inhibit MATE-1. When MATE-1 is blocked, creatinine is retained in the serum. Elevated creatinine levels raise the specter of drug-induced intrarenal insufficiency despite the lack of true renal injury. We conducted a systematic analysis of MATE-1 inhibitor (MATEi)-treated patients to comprehensively characterize this phenomenon. METHODS Patients with oncogene-driven lung cancer treated with a wide variety of MATEi TKIs (brigatinib, cabozantinib, capmatinib, crizotinib, entrectinib, lorlatinib, pralsetinib, selpercatinib, and tepotinib) were eligible for an analysis of renal dysfunction. Acute kidney injury was classified on the basis of creatinine levels (Kidney Disease: Improving Global Outcomes criteria) as stage 1 (≥1.5× but <2× baseline), stage 2 (≥2× but <3× baseline), or stage 3 (>3× baseline). When available, cystatin C, a marker of kidney function unaffected by MATE-1, was used to evaluate the glomerular filtration rate (GFR). RESULTS We identified 863 patients receiving MATEi TKIs including crizotinib (39%, n = 333), lorlatinib (17%, n = 144), cabozantinib (10%, n = 87), selpercatinib (10%, n = 82), capmatinib (9%, n = 77), brigatinib (6%, n = 53), entrectinib (5%, n = 45), tepotinib (5%, n = 41), and pralsetinib (0.1%, n = 1). Of the 90 patients (10%) with acute kidney injury, Kidney Disease: Improving Global Outcomes stages 1, 2, and 3 were observed in 72% (n = 65), 21% (n = 19), and 7% (n = 6) of patients, respectively. Concurrently drawn creatinine and cystatin C levels on TKI therapy were available for 17 patients. In most cases (n = 15 of 17), the calculated GFR was higher using cystatin C versus creatinine. The percentage of patients whose GFR was higher using cystatin C versus creatinine by less than 10 mL/min, 10 to 19 mL/min, 20 to 29 mL/min, and more than or equal to 30 mL/min was 27% (n = four of 15), 20% (n = three of 15), 20% (n = three of 15), and 33% (n = five of 15), respectively. Long-term data in three patients that spanned 3 years revealed that GFR was higher using cystatin C versus creatinine in 96% (n = 49 of 51) of all time points. Using a virtual clinical trial GFR cutoff of 40 mL/min, the percentage of eligible patients rose from 41% (n = seven of 17) using creatinine calculations to 71% (n = 12 of 17) using cystatin C calculations. CONCLUSIONS The calculated GFR in patients with cancer receiving MATEi TKIs was higher in almost all cases when using cystatin C. When serum creatinine level seems elevated in patients receiving MATE-1 inhibitors, we recommend recalculating GFR using cystatin C before searching for other etiologies of kidney injury and reducing or stopping TKI therapy.
Collapse
Affiliation(s)
- Monica F Chen
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Guilherme Harada
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Dazhi Liu
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Ray DeMatteo
- Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Christina Falcon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Clare Wilhelm
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Mark G Kris
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York; Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York.
| | - Victoria Gutgarts
- Division of Nephrology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York
| |
Collapse
|
5
|
Nakada T, Kudo T, Ito K. Quantitative Consideration of Clinical Increases in Serum Creatinine Caused by Renal Transporter Inhibition. Drug Metab Dispos 2023; 51:1114-1126. [PMID: 36859345 DOI: 10.1124/dmd.122.000969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/09/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Creatinine is a common biomarker of renal function and is secreted in the renal tubular cells via drug transporters, such as organic cation transporter 2 and multidrug and toxin extrusion (MATE) 1/2-K. To differentiate between drug-induced acute kidney injury (AKI) and drug interactions through the renal transporter, it has been examined whether these transporter inhibitions quantitatively explained increases in serum creatinine (SCr) at their clinically relevant concentrations using drugs without any changes in renal function. For such renal transporter inhibitors and recently approved tyrosine kinase inhibitors (TKIs), this mini-review describes clinical increases in SCr and inhibitory potentials against the renal transporters. Most cases of SCr elevations can be explained by considering the renal transporter inhibitions based on unbound maximum plasma concentrations, except for drugs associated with obvious changes in renal function. SCr increases for cobicistat, dolutegravir, and dronedarone, and some TKIs were significantly underestimated, and these underestimations were suggested to be associated with low plasma unbound fractions. Sensitivity analysis of SCr elevations regarding inhibitory potentials of MATE1/2-K demonstrated that typical inhibitors such as cimetidine, DX-619, pyrimethamine, and trimethoprim could give false interpretations of AKI according to the criteria based on relative or absolute levels of SCr elevations. Recent progress and current challenges of physiologically-based pharmacokinetics modeling for creatinine disposition were also summarized. Although it should be noted for the potential impact of in vitro assay designs on clinical translatability of transporter inhibitions data, mechanistic approaches could support decision-making in clinical development to differentiate between AKI and creatinine-drug interactions. SIGNIFICANCE STATEMENT: Serum creatinine (SCr) is widely used as an indicator of kidney function, but it increases due to inhibitions of renal transporters, such as multidrug and toxin extrusion protein 1/2-K despite no functional changes in the kidney. Such SCr elevations were quantitatively explained by renal transporter inhibitions except for some drugs with high protein binding. The present analysis demonstrated that clinically relevant inhibitors of the renal transporters could cause SCr elevations above levels corresponding to acute kidney injury criteria.
Collapse
Affiliation(s)
- Tomohisa Nakada
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan (T.N.) and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan (T.K., K.I.)
| | - Toshiyuki Kudo
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan (T.N.) and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan (T.K., K.I.)
| | - Kiyomi Ito
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan (T.N.) and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan (T.K., K.I.)
| |
Collapse
|
6
|
Lin K, Kong X, Tao X, Zhai X, Lv L, Dong D, Yang S, Zhu Y. Research Methods and New Advances in Drug-Drug Interactions Mediated by Renal Transporters. Molecules 2023; 28:5252. [PMID: 37446913 DOI: 10.3390/molecules28135252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/22/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
The kidney is critical in the human body's excretion of drugs and their metabolites. Renal transporters participate in actively secreting substances from the proximal tubular cells and reabsorbing them in the distal renal tubules. They can affect the clearance rates (CLr) of drugs and their metabolites, eventually influence the clinical efficiency and side effects of drugs, and may produce drug-drug interactions (DDIs) of clinical significance. Renal transporters and renal transporter-mediated DDIs have also been studied by many researchers. In this article, the main types of in vitro research models used for the study of renal transporter-mediated DDIs are membrane-based assays, cell-based assays, and the renal slice uptake model. In vivo research models include animal experiments, gene knockout animal models, positron emission tomography (PET) technology, and studies on human beings. In addition, in vitro-in vivo extrapolation (IVIVE), ex vivo kidney perfusion (EVKP) models, and, more recently, biomarker methods and in silico models are included. This article reviews the traditional research methods of renal transporter-mediated DDIs, updates the recent progress in the development of the methods, and then classifies and summarizes the advantages and disadvantages of each method. Through the sorting work conducted in this paper, it will be convenient for researchers at different learning stages to choose the best method for their own research based on their own subject's situation when they are going to study DDIs mediated by renal transporters.
Collapse
Affiliation(s)
- Kexin Lin
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaorui Kong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
7
|
Guo L, Fu B, Liu Y, Hao N, Ji Y, Yang H. Diuretic resistance in patients with kidney disease: Challenges and opportunities. Biomed Pharmacother 2023; 157:114058. [PMID: 36473405 DOI: 10.1016/j.biopha.2022.114058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/15/2022] [Accepted: 11/27/2022] [Indexed: 12/10/2022] Open
Abstract
Edema caused by kidney disease is called renal edema. Edema is a common symptom of many human kidney diseases. Patients with renal edema often need to take diuretics.However, After taking diuretics, patients with kidney diseases are prone to kidney congestion, decreased renal perfusion, decreased diuretics secreted by renal tubules, neuroendocrine system abnormalities, abnormal ion transporter transport, drug interaction, electrolyte disorder, and hypoproteinemia, which lead to ineffective or weakened diuretic use and increase readmission rate and mortality. The main causes and coping strategies of diuretic resistance in patients with kidney diseases were described in detail in this report. The common causes of DR included poor diet (electrolyte disturbance and hypoproteinemia due to patients' failure to limit diet according to correct sodium, chlorine, potassium, and protein level) and poor drug compliance (the patient did not take adequate doses of diuretics. true resistance occurs only if the patient takes adequate doses of diuretics, but they are not effective), changes in pharmacokinetics and pharmacodynamics, electrolyte disorders, changes in renal adaptation, functional nephron reduction, and decreased renal blood flow. Common treatment measures include increasing in the diuretic dose and/or frequency, sequential nephron blockade,using new diuretics, ultrafiltration treatment, etc. In clinical work, measures should be taken to prevent or delay the occurrence and development of DR in patients with kidney diseases according to the actual situation of patients and the mechanism of various causes. Currently, there are many studies on DR in patients with heart diseases. Although the phenomenon of DR in patients with kidney diseases is common, there is a relatively little overview of the mechanism and treatment strategy of DR in patients with kidney diseases. Therefore, this paper hopes to show the information on DR in patients with kidney diseases to clinicians and researchers and broaden the research direction and ideas to a certain extent.
Collapse
Affiliation(s)
- Luxuan Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Baohui Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yang Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Na Hao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Ji
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hongtao Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
8
|
Ma Y, Zhang M, Yang J, Zhu L, Dai J, Wu X. Characterization of the renal tubular transport of creatinine by activity-based protein profiling and transport kinetics. Eur J Pharm Sci 2023; 180:106342. [PMID: 36435354 DOI: 10.1016/j.ejps.2022.106342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/14/2022] [Accepted: 11/23/2022] [Indexed: 11/25/2022]
Abstract
Serum creatinine is widely used to adjust the dosing of drugs eliminated by the kidney in patients with renal dysfunction, as it is a readily accessible indicator of kidney function. However, there are many limitations for drug dosage adjustment based on serum creatinine levels, one of which is the limited understanding of creatinine's tubular transport. Thus, we aimed to complement and advance the renal tubular transport of creatinine by activity-based protein profiling (ABPP) and transporter-overexpression technology. Renal tubular transporters were not identified via ABPP due to the low-affinity interaction between transporters and creatinine. The uptake of isotopically labeled d3-creatinine was significantly increased in OCT2-overexpressing cell lines (p<0.01), and the Km and Vmax of d3-creatinine uptake mediated by OCT2 was 3.1 mM and 408 pmol/mg protein/min, respectively. In the OCT2-overexpressing cell lines, the IC50 of creatinine for d3-creatinine uptake was 10.3 mM, and that of the OCT2 inhibitor cimetidine for d3-creatinine uptake was 99.04 μM. Different dosages of creatinine did not affect the renal excretion of d3-creatinine in mice (p>0.05), while cimetidine significantly reduced the renal excretion of d3-creatinine (p<0.01) without affecting the glomerular filtration rate. Molecular docking in silico showed that the OCT2 amino acid GLN242 could form a hydrogen bond of 2.5 Å with creatinine, and there may be a π-π interaction between TYR362 and creatinine. A site mutation experiment demonstrated that TYR362 and GLN242 were important sites for the OCT2-creatinine interaction. These results demonstrate that OCT2 mediates the renal tubular secretion of creatinine with low affinity and is a minor contributor to creatinine secretion.
Collapse
Affiliation(s)
- Yanrong Ma
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Mingkang Zhang
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Jinru Yang
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Lin Zhu
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China
| | - Jianye Dai
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Xinan Wu
- Department of Pharmacy, the First Hospital of Lanzhou University, Lanzhou 730000 China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
9
|
Türk D, Müller F, Fromm MF, Selzer D, Dallmann R, Lehr T. Renal Transporter-Mediated Drug-Biomarker Interactions of the Endogenous Substrates Creatinine and N 1 -Methylnicotinamide: A PBPK Modeling Approach. Clin Pharmacol Ther 2022; 112:687-698. [PMID: 35527512 DOI: 10.1002/cpt.2636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/28/2022] [Indexed: 01/06/2023]
Abstract
Endogenous biomarkers for transporter-mediated drug-drug interaction (DDI) predictions represent a promising approach to facilitate and improve conventional DDI investigations in clinical studies. This approach requires high sensitivity and specificity of biomarkers for the targets of interest (e.g., transport proteins), as well as rigorous characterization of their kinetics, which can be accomplished utilizing physiologically-based pharmacokinetic (PBPK) modeling. Therefore, the objective of this study was to develop PBPK models of the endogenous organic cation transporter (OCT)2 and multidrug and toxin extrusion protein (MATE)1 substrates creatinine and N1 -methylnicotinamide (NMN). Additionally, this study aimed to predict kinetic changes of the biomarkers during administration of the OCT2 and MATE1 perpetrator drugs trimethoprim, pyrimethamine, and cimetidine. Whole-body PBPK models of creatinine and NMN were developed utilizing studies investigating creatinine or NMN exogenous administration and endogenous synthesis. The newly developed models accurately describe and predict observed plasma concentration-time profiles and urinary excretion of both biomarkers. Subsequently, models were coupled to the previously built and evaluated perpetrator models of trimethoprim, pyrimethamine, and cimetidine for interaction predictions. Increased creatinine plasma concentrations and decreased urinary excretion during the drug-biomarker interactions with trimethoprim, pyrimethamine, and cimetidine were well-described. An additional inhibition of NMN synthesis by trimethoprim and pyrimethamine was hypothesized, improving NMN plasma and urine interaction predictions. To summarize, whole-body PBPK models of creatinine and NMN were built and evaluated to better assess creatinine and NMN kinetics while uncovering knowledge gaps for future research. The models can support investigations of renal transporter-mediated DDIs during drug development.
Collapse
Affiliation(s)
- Denise Türk
- Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dominik Selzer
- Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
10
|
Sidamo T, Shibeshi W, Yimer G, Aklillu E, Engidawork E. Explorative Analysis of Treatment Outcomes of Levofloxacin- and Moxifloxacin-Based Regimens and Outcome Predictors in Ethiopian MDR-TB Patients: A Prospective Observational Cohort Study. Infect Drug Resist 2022; 14:5473-5489. [PMID: 34984005 PMCID: PMC8703047 DOI: 10.2147/idr.s342964] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/30/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose/Background Although Ethiopia is among the thirty high multi-drug resistant tuberculosis (MDR-TB) burden countries in the world, comparative therapeutic efficacy of moxifloxacin and levofloxacin has not been explored, particularly in MDR-TB patients. We therefore aimed to prospectively compare clinical outcomes and determine potential predictors of the outcomes among patients on moxifloxacin or levofloxacin-based MDR-TB drug regimens. Methods We analyzed clinical parameters and laboratory data of eighty MDR-TB patients on moxifloxacin- or levofloxacin-based regimens. The clinical outcomes were compared using the Kaplan–Meier survival functions and the outcome definitions of the 2013 World Health Organization. Monthly sputum culture conversions and a molecular line probe assay results were also assessed. Observed outcomes and patient-related variables between the two groups were compared using chi-square, Wilcoxon Rank and Fisher exact tests. We also determined the potential predictors influencing treatment outcomes of moxifloxacin and levofloxacin using Cox proportional hazard model. Results The levofloxacin-based treatment group had a lower failure rate and adverse drug events as well as better treatment success than the moxifloxacin-based group. Overall treatment success was 65%. Disaggregating the data revealed that 53.8% were cured, 11.2% completed treatment, 10.0% died, 11.2% failed, and 13.8% were lost-to-follow-up. The line probe assay result showed that 11.3% of the clinical isolates were resistant to fluoroquinolones and 3.8% were resistant to both fluoroquinolones and injectable anti-TB agents. Treatment regimen type, culture conversion rate, alcohol use, cavity lesion, serum levels of creatinine and alanine aminotransferase were independent predictors of treatment outcome. Conclusion The levofloxacin-based regimen group has a better overall treatment success than the moxifloxacin-based group among MDR-TB patients. Clinical parameters and substance use history of the patients influenced treatment outcomes. We recommend further broader clinical studies to substantiate our findings as an input to review MDR-TB treatment guidelines.
Collapse
Affiliation(s)
- Temesgen Sidamo
- Department of Pharmacology & Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Workineh Shibeshi
- Department of Pharmacology & Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Getnet Yimer
- Global One Health Initiative, Office of International Affairs, The Ohio State University, Columbus, OH, USA
| | - Eleni Aklillu
- Department of Laboratory of Medicine Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, C-168 141 86, Sweden
| | - Ephrem Engidawork
- Department of Pharmacology & Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
11
|
Tang J, Shen H, Zhao X, Holenarsipur VK, Mariappan TT, Zhang Y, Panfen E, Zheng J, Humphreys WG, Lai Y. Endogenous Plasma Kynurenic Acid in Human: A Newly Discovered Biomarker for Drug-Drug Interactions Involving Organic Anion Transporter 1 and 3 Inhibition. Drug Metab Dispos 2021; 49:1063-1069. [PMID: 34599018 DOI: 10.1124/dmd.121.000486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
As an expansion investigation of drug-drug interaction (DDI) from previous clinical trials, additional plasma endogenous metabolites were quantitated in the same subjects to further identify the potential biomarkers of organic anion transporter (OAT) 1/3 inhibition. In the single dose, open label, three-phase with fixed order of treatments study, 14 healthy human volunteers orally received 1000 mg probenecid alone, or 40 mg furosemide alone, or 40 mg furosemide at 1 hour after receiving 1000 mg probenecid on days 1, 8, and 15, respectively. Endogenous metabolites including kynurenic acid, xanthurenic acid, indo-3-acetic acid, pantothenic acid, p-cresol sulfate, and bile acids in the plasma were measured by liquid chromatography-tandem mass spectrometry. The Cmax of kynurenic acids was significantly increased about 3.3- and 3.7-fold over the baseline values at predose followed by the treatment of probenecid alone or in combination with furosemide respectively. In comparison with the furosemide-alone group, the Cmax and area under the plasma concentration-time curve (AUC) up to 12 hours of kynurenic acid were significantly increased about 2.4- and 2.5-fold by probenecid alone, and 2.7- and 2.9-fold by probenecid plus furosemide, respectively. The increases in Cmax and AUC of plasma kynurenic acid by probenecid are comparable to the increases of furosemide Cmax and AUC reported previously. Additionally, the plasma concentrations of xanthurenic acid, indo-3-acetic acid, pantothenic acid, and p-cresol sulfate, but not bile acids, were also significantly elevated by probenecid treatments. The magnitude of effect size analysis for known potential endogenous biomarkers demonstrated that kynurenic acid in the plasma offers promise as a superior addition for early DDI assessment involving OAT1/3 inhibition. SIGNIFICANCE STATEMENT: This article reports that probenecid, an organic anion transporter (OAT) 1 and OAT3 inhibitor, significantly increased the plasma concentrations of kynurenic acid and several uremic acids in human subjects. Of those, the increases of plasma kynurenic acid exposure are comparable to the increases of furosemide by OAT1/3 inhibition. Effect size analysis for known potential endogenous biomarkers revealed that plasma kynurenic acid is a superior addition for early drug-drug interaction assessment involving OAT1/3 inhibition.
Collapse
Affiliation(s)
- Jennifer Tang
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Hong Shen
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Xiaofeng Zhao
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Vinay K Holenarsipur
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - T Thanga Mariappan
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Yueping Zhang
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Erika Panfen
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Jim Zheng
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - W Griffith Humphreys
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| | - Yurong Lai
- Drug Metabolism, Gilead Science Inc., Foster City, California (J.T., X.Z., J.Z., Y.L.); Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, New Jersey (H.S., Y.Z., E.P., W.G.H.); and Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre (BBRC), Syngene International Ltd., Bangalore, India (V.K.H., T.T.M.)
| |
Collapse
|
12
|
Li Y, Talebi Z, Chen X, Sparreboom A, Hu S. Endogenous Biomarkers for SLC Transporter-Mediated Drug-Drug Interaction Evaluation. Molecules 2021; 26:5500. [PMID: 34576971 PMCID: PMC8466752 DOI: 10.3390/molecules26185500] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022] Open
Abstract
Membrane transporters play an important role in the absorption, distribution, metabolism, and excretion of xenobiotic substrates, as well as endogenous compounds. The evaluation of transporter-mediated drug-drug interactions (DDIs) is an important consideration during the drug development process and can guide the safe use of polypharmacy regimens in clinical practice. In recent years, several endogenous substrates of drug transporters have been identified as potential biomarkers for predicting changes in drug transport function and the potential for DDIs associated with drug candidates in early phases of drug development. These biomarker-driven investigations have been applied in both preclinical and clinical studies and proposed as a predictive strategy that can be supplanted in order to conduct prospective DDIs trials. Here we provide an overview of this rapidly emerging field, with particular emphasis on endogenous biomarkers recently proposed for clinically relevant uptake transporters.
Collapse
Affiliation(s)
| | | | | | | | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA; (Y.L.); (Z.T.); (X.C.); (A.S.)
| |
Collapse
|
13
|
Substrates and Inhibitors of Organic Cation Transporters (OCTs) and Plasma Membrane Monoamine Transporter (PMAT) and Therapeutic Implications. Handb Exp Pharmacol 2021; 266:119-167. [PMID: 34495395 DOI: 10.1007/164_2021_516] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gene products of the SLC22A gene family (hOCT1, hOCT2, and hOCT3) and of the SLC29A4 gene (hPMAT or hENT4) are all polyspecific organic cation transporters. Human OCTs (including hPMAT) are expressed in peripheral tissues such as small intestine, liver, and kidney involved in the pharmacokinetics of drugs. In the human brain, all four transporters are expressed at the blood-brain barrier (BBB), hOCT2 is additionally expressed in neurons, and hOCT3 and hPMAT in glia. More than 40% of the presently used drugs are organic cations. This chapter lists and discusses all known drugs acting as substrates or inhibitors of these four organic cation transporters, independently of whether the transporter is expressed in the central nervous system (CNS) or in peripheral tissues. Of interest is their involvement in drug absorption, distribution, and excretion as well as potential OCT-associated drug-drug interactions (DDIs), with a focus on drugs that act in the CNS.
Collapse
|
14
|
Mochizuki T, Mizuno T, Maeda K, Kusuhara H. Current progress in identifying endogenous biomarker candidates for drug transporter phenotyping and their potential application to drug development. Drug Metab Pharmacokinet 2020; 37:100358. [PMID: 33461054 DOI: 10.1016/j.dmpk.2020.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 01/23/2023]
Abstract
Drug transporters play important roles in the elimination of various compounds from the blood. Genetic variation and drug-drug interactions underlie the pharmacokinetic differences for the substrates of drug transporters. Some endogenous substrates of drug transporters have emerged as biomarkers to assess differences in drug transporter activity-not only in animals, but also in humans. Metabolomic analysis is a promising approach for identifying such endogenous substrates through their metabolites. The appropriateness of metabolites is supported by studies in vitro and in vivo, both in animals and through pharmacogenomic or drug-drug interaction studies in humans. This review summarizes current progress in identifying such endogenous biomarkers and applying them to drug transporter phenotyping.
Collapse
Affiliation(s)
- Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Japan
| | - Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Japan.
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Japan.
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Japan.
| |
Collapse
|
15
|
Abstract
The organic cation transporters (OCTs) OCT1, OCT2, OCT3, novel OCT (OCTN)1, OCTN2, multidrug and toxin exclusion (MATE)1, and MATE kidney-specific 2 are polyspecific transporters exhibiting broadly overlapping substrate selectivities. They transport organic cations, zwitterions, and some uncharged compounds and operate as facilitated diffusion systems and/or antiporters. OCTs are critically involved in intestinal absorption, hepatic uptake, and renal excretion of hydrophilic drugs. They modulate the distribution of endogenous compounds such as thiamine, L-carnitine, and neurotransmitters. Sites of expression and functions of OCTs have important impact on energy metabolism, pharmacokinetics, and toxicity of drugs, and on drug-drug interactions. In this work, an overview about the human OCTs is presented. Functional properties of human OCTs, including identified substrates and inhibitors of the individual transporters, are described. Sites of expression are compiled, and data on regulation of OCTs are presented. In addition, genetic variations of OCTs are listed, and data on their impact on transport, drug treatment, and diseases are reported. Moreover, recent data are summarized that indicate complex drug-drug interaction at OCTs, such as allosteric high-affinity inhibition of transport and substrate dependence of inhibitor efficacies. A hypothesis about the molecular mechanism of polyspecific substrate recognition by OCTs is presented that is based on functional studies and mutagenesis experiments in OCT1 and OCT2. This hypothesis provides a framework to imagine how observed complex drug-drug interactions at OCTs arise. Finally, preclinical in vitro tests that are performed by pharmaceutical companies to identify interaction of novel drugs with OCTs are discussed. Optimized experimental procedures are proposed that allow a gapless detection of inhibitory and transported drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
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
|
17
|
Scotcher D, Arya V, Yang X, Zhao P, Zhang L, Huang S, Rostami‐Hodjegan A, Galetin A. Mechanistic Models as Framework for Understanding Biomarker Disposition: Prediction of Creatinine-Drug Interactions. CPT Pharmacometrics Syst Pharmacol 2020; 9:282-293. [PMID: 32410382 PMCID: PMC7239336 DOI: 10.1002/psp4.12508] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
Creatinine is widely used as a biomarker of glomerular filtration, and, hence, renal function. However, transporter-mediated secretion also contributes to its renal clearance, albeit to a lesser degree. Inhibition of these transporters causes transient serum creatinine elevation, which can be mistaken as impaired renal function. The current study developed mechanistic models of creatinine kinetics within physiologically based framework accounting for multiple transporters involved in creatinine renal elimination, assuming either unidirectional or bidirectional-OCT2 transport (driven by electrochemical gradient). Robustness of creatinine models was assessed by predicting creatinine-drug interactions with 10 perpetrators; performance evaluation accounted for 5% intra-individual variability in serum creatinine. Models showed comparable predictive performances of the maximum steady-state effect regardless of OCT2 directionality assumptions. However, only the bidirectional-OCT2 model successfully predicted the minimal effect of ranitidine. The dynamic nature of models provides clear advantage to static approaches and most advanced framework for evaluating interplay between multiple processes in creatinine renal disposition.
Collapse
Affiliation(s)
- Daniel Scotcher
- Centre for Applied Pharmacokinetic ResearchUniversity of ManchesterManchesterUK
| | - Vikram Arya
- Office of Clinical PharmacologyOffice of Translational SciencesCentre for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Xinning Yang
- Office of Clinical PharmacologyOffice of Translational SciencesCentre for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Ping Zhao
- Office of Clinical PharmacologyOffice of Translational SciencesCentre for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
Bill & Melinda Gates FoundationSeattleWashingtonUSA
| | - Lei Zhang
- Office of Research and StandardsOffice of Generic DrugsCentre for Drug Evaluation and Research, US Food and Drug AdministrationSilver SpringMarylandUSA
| | - Shiew‐Mei Huang
- Office of Clinical PharmacologyOffice of Translational SciencesCentre for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic ResearchUniversity of ManchesterManchesterUK
- CertaraSheffieldUK
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic ResearchUniversity of ManchesterManchesterUK
| |
Collapse
|
18
|
Nozaki Y, Izumi S. Recent advances in preclinical in vitro approaches towards quantitative prediction of hepatic clearance and drug-drug interactions involving organic anion transporting polypeptide (OATP) 1B transporters. Drug Metab Pharmacokinet 2020; 35:56-70. [DOI: 10.1016/j.dmpk.2019.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/29/2019] [Accepted: 11/02/2019] [Indexed: 12/26/2022]
|
19
|
Totsuka K, Sesoko S, Fukase H, Ikushima I, Odajima M, Niwayama Y. Pharmacokinetic study of lascufloxacin in non-elderly healthy men and elderly men. J Infect Chemother 2019; 26:231-239. [PMID: 31882385 DOI: 10.1016/j.jiac.2019.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/30/2019] [Accepted: 09/14/2019] [Indexed: 01/22/2023]
Abstract
We conducted this phase I clinical study to examine the pharmacokinetic profiles and safety of lascufloxacin (LSFX), a novel quinolone antibacterial agent, in non-elderly Japanese healthy men and the effects of aging on LSFX pharmacokinetics in elderly Japanese healthy men. 1. After single-dose oral administration of LSFX 100-800 mg (capsules) to six healthy adults in fasting state, the Cmax and AUClast roughly increased in proportion to the doses. 2. After multiple-dose oral administration of LSFX 75 mg (tablets) once daily for 7 days to six healthy adults, plasma LSFX reached the steady state by Day 7. The cumulative factor of LSFX on Day 7 to Day 1 was 1.65 for the Cmax and 1.96 for the AUCtau. 3. Regarding pharmacokinetic parameters of plasma LSFX after single-dose administration of LSFX 75 mg tablets (final product) to 24 healthy adults in fed state, the Cmax was somewhat higher, 1.28 times more than that in fasting state, whereas no changes were found in the AUClast. We therefore proposed that food effects of LSFX on absorption were negligible. 4. No clinically significant safety problems of LSFX were found in a series of studies involving healthy adults conducted this time. 5. After single-dose oral administration of LSFX 200 mg (capsules) to six elderly people in fasting state, its pharmacokinetic parameters were similar to those in non-elderly people, with no significant safety concerns. Therefore, adjustment of dosage and administration was considered to be unnecessary for LSFX administration to elderly individuals.
Collapse
Affiliation(s)
- Kyoichi Totsuka
- Department of Internal Medicine, Kita-tama Hospital, Tokyo, Japan
| | - Shogo Sesoko
- Medical Corporation Aggregate Shinpukai Maruyama Hospital, Shizuoka, Japan
| | - Hiroyuki Fukase
- CPC Clinical Trial Hospital, Medipolis Medical Research Institute, Kagoshima, Japan
| | | | - Masaaki Odajima
- Discovery Research Headquarters, Kyorin Pharmaceutical Co., LTD, Tokyo, Japan
| | - Yutaka Niwayama
- Discovery Research Headquarters, Kyorin Pharmaceutical Co., LTD, Tokyo, Japan.
| |
Collapse
|
20
|
|
21
|
Miyake T, Mizuno T, Takehara I, Mochizuki T, Kimura M, Matsuki S, Irie S, Watanabe N, Kato Y, Ieiri I, Maeda K, Ando O, Kusuhara H. Elucidation of N 1-methyladenosine as a Potential Surrogate Biomarker for Drug Interaction Studies Involving Renal Organic Cation Transporters. Drug Metab Dispos 2019; 47:1270-1280. [PMID: 31511257 DOI: 10.1124/dmd.119.087262] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 09/07/2019] [Indexed: 11/22/2022] Open
Abstract
Endogenous substrates are emerging biomarkers for drug transporters, which serve as surrogate probes in drug-drug interaction (DDI) studies. In this study, the results of metabolome analysis using wild-type and Oct1/2 double knockout mice suggested that N 1-methyladenosine (m1A) was a novel organic cation transporter (OCT) 2 substrate. An in vitro transport study revealed that m1A is a substrate of mouse Oct1, Oct2, Mate1, human OCT1, OCT2, and multidrug and toxin exclusion protein (MATE) 2-K, but not human MATE1. Urinary excretion accounted for 77% of the systemic elimination of m1A in mice. The renal clearance (46.9 ± 4.9 ml/min per kilogram) of exogenously given m1A was decreased to near the glomerular filtration rates by Oct1/2 double knockout or Mate1 inhibition by pyrimethamine (16.6 ± 2.6 and 24.3 ± 0.6 ml/min per kilogram, respectively), accompanied by significantly higher plasma concentrations. In vivo inhibition of OCT2/MATE2-K by a single dose of 7-[(3R)-3-(1-aminocyclopropyl)pyrrolidin-1-yl]-1-[(1R,2S)-2-fluorocyclopropyl]-8-methoxy-4-oxoquinoline-3-carboxylic acid in cynomolgus monkeys resulted in the elevation of the area under the curve of m1A (1.72-fold) as well as metformin (2.18-fold). The plasma m1A concentration profile showed low diurnal and interindividual variation in healthy volunteers. The renal clearance of m1A in younger (21-45 year old) and older (65-79 year old) volunteers (244 ± 58 and 169 ± 22 ml/min per kilogram, respectively) was about 2-fold higher than the creatinine clearance. The renal clearances of m1A and creatinine were 31% and 17% smaller in older than in younger volunteers. Thus, m1A could be a surrogate probe for the evaluation of DDIs involving OCT2/MATE2-K. SIGNIFICANCE STATEMENT: Endogenous substrates can serve as surrogate probes for clinical drug-drug interaction studies involving drug transporters or enzymes. In this study, m1A was found to be a novel substrate of renal cationic drug transporters OCT2 and MATE2-K. N 1-methyladenosine was revealed to have some advantages compared to other OCT2/MATE substrates (creatinine and N 1-methylnicotinamide). The genetic or chemical impairment of OCT2 or MATE2-K caused a significant increase in the plasma m1A concentration in mice and cynomolgus monkeys due to the high contribution of tubular secretion to the net elimination of m1A. The plasma m1A concentration profile showed low diurnal and interindividual variation in healthy volunteers. Thus, m1A could be a better biomarker of variations in OCT2/MATE2-K activity caused by inhibitory drugs.
Collapse
Affiliation(s)
- Takeshi Miyake
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Issey Takehara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Miyuki Kimura
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Shunji Matsuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Shin Irie
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Nobuaki Watanabe
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Yukio Kato
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Ichiro Ieiri
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Osamu Ando
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan (Tak.M., Tad.M., Tat.M., K.M., H.K.); Biomarker Department (I.T.) and Drug Metabolism & Pharmacokinetics Research Laboratories (N.W., O.A.), Daiichi-Sankyo Co., Ltd., Tokyo, Japan; Fukuoka Mirai Hospital Clinical Research Center, Fukuoka, Japan (M.K., S.M., S.I.); Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (Y.K.); and Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (I.I.)
| |
Collapse
|
22
|
Kikuchi R, Chiou WJ, Kasai MA, de Morais SM, Bow DA. No Inhibition of MATE1/2K-Mediated Renal Creatinine Secretion Predicted With Ritonavir or Cobicistat. J Pharm Sci 2019; 108:3118-3123. [DOI: 10.1016/j.xphs.2019.04.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022]
|
23
|
Casado JL, Monsalvo M, Vizcarra P, Fontecha M, Serrano-Villar S, Moreno S. Evaluation of kidney function in HIV-infected patients receiving an antiretroviral regimen containing one or two inhibitors of the tubular secretion of creatinine. HIV Med 2019; 20:648-656. [PMID: 31321875 DOI: 10.1111/hiv.12784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVES The aim of this study was to determine the evolution of renal function in patients receiving one or two inhibitors, according to different baseline factors. Some antiretroviral drugs such as rilpivirine (RPV), dolutegravir (DTG), or cobicistat (COBI), interact with the tubular secretion of creatinine, but there are no data about their impact in renal function evaluation in patients with renal disease or when these drugs are used concomitantly. METHODS A prospective cohort study was carried out in HIV-infected patients who switched to a dual regimen including DTG, RPV or darunavir/COBI, separately or in combination. The primary endpoint was the evolution of the serum creatinine-based estimated glomerular filtration rate (eGFR-scr). A control group not receiving any transporter inhibitor was included. RESULTS A total of 288 patients on different dual regimens were included (DTG + RPV, 92; DTG + darunavir/COBI, 23; DTG, 26; COBI, 19; control group, 128). In patients receiving two transporter inhibitors, eGFR-scr decreased by a mean of -8.4 mL/min/1.73 m2 , similar to that observed with the separate use of DTG or COBI (mean of both groups, -8.6 mL/min/1.73 m2 ), while eGFR-scr improved in the control group. Similar evolution of proteinuria and tubular dysfunction was observed in all the groups, and there were no significant changes in the cystatin C-based eGFR. Mean eGFR-scr change inversely correlated with baseline eGFR-scr value (r = -0.39; P < 0.01), with a lower eGFR-scr decrease in patients with chronic kidney disease. CONCLUSIONS Similar eGFR-scr decreases were observed in patients using different antiretroviral drugs inhibiting the tubular transport of creatinine, separately or in combination, with no alterations in proteinuria or cystatin C-based eGFR. The lack of additional changes when the drugs were used in combination, and the lower impact in cases of previous chronic kidney disease, suggest that there are compensatory mechanisms for creatinine secretion.
Collapse
Affiliation(s)
- J L Casado
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| | - M Monsalvo
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| | - P Vizcarra
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| | - M Fontecha
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| | - S Serrano-Villar
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| | - S Moreno
- Department of Infectious Diseases, Ramon y Cajal Hospital, Madrid, Spain
| |
Collapse
|
24
|
Nigam SK. The SLC22 Transporter Family: A Paradigm for the Impact of Drug Transporters on Metabolic Pathways, Signaling, and Disease. Annu Rev Pharmacol Toxicol 2019; 58:663-687. [PMID: 29309257 DOI: 10.1146/annurev-pharmtox-010617-052713] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The SLC22 transporter family consists of more than two dozen members, which are expressed in the kidney, the liver, and other tissues. Evolutionary analysis indicates that SLC22 transporters fall into at least six subfamilies: OAT (organic anion transporter), OAT-like, OAT-related, OCT (organic cation transporter), OCTN (organic cation/carnitine transporter), and OCT/OCTN-related. Some-including OAT1 [SLC22A6 or NKT (novel kidney transporter)] and OAT3 (SLC22A8), as well as OCT1 (SLC22A1) and OCT2 (SLC22A2)-are widely studied drug transporters. Nevertheless, analyses of knockout mice and other data indicate that SLC22 transporters regulate key metabolic pathways and levels of signaling molecules (e.g., gut microbiome products, bile acids, tricarboxylic acid cycle intermediates, dietary flavonoids and other nutrients, prostaglandins, vitamins, short-chain fatty acids, urate, and ergothioneine), as well as uremic toxins associated with chronic kidney disease. Certain SLC22 transporters-such as URAT1 (SLC22A12) and OCTN2 (SLC22A5)-are mutated in inherited metabolic diseases. A new systems biology view of transporters is emerging. As proposed in the remote sensing and signaling hypothesis, SLC22 transporters, together with other SLC and ABC transporters, have key roles in interorgan and interorganism small-molecule communication and, together with the neuroendocrine, growth factor-cytokine, and other homeostatic systems, regulate local and whole-body homeostasis.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics and Medicine, University of California, San Diego, La Jolla, California 92093, USA;
| |
Collapse
|
25
|
Nakada T, Kudo T, Kume T, Kusuhara H, Ito K. Estimation of changes in serum creatinine and creatinine clearance caused by renal transporter inhibition in healthy subjects. Drug Metab Pharmacokinet 2019; 34:233-238. [PMID: 31176593 DOI: 10.1016/j.dmpk.2019.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 01/30/2019] [Accepted: 02/25/2019] [Indexed: 11/24/2022]
Abstract
Creatinine is excreted into urine by glomerular filtration and renal tubular secretion through drug transporters such as organic anion transporter 2 (OAT2), organic cation transporter 2 (OCT2), OCT3, multidrug and toxin extrusion protein 1 (MATE1), and MATE2-K. We aimed to investigate whether our method for estimating percentage changes in serum creatinine concentration (SCr) and creatinine clearance (CLcre) from the baseline is applicable for studying renal transporter inhibitors. We tested 14 compounds (cimetidine, cobicistat, dolutegravir, dronedarone, DX-619, famotidine, INCB039110, nizatidine, ondansetron, pyrimethamine, rabeprazole, ranolazine, trimethoprim, and vandetanib), which were reported to cause reversible changes in SCr and/or CLcre in healthy subjects excluding elderly. Percentage changes were estimated from the relative contributions of the forementioned transporters to CLcre and competitive inhibition by these compounds at their maximum plasma unbound concentrations. For 7 and 9 out of these compounds, changes in SCr and/or CLcre were estimated within 2- and 3-fold of observed values, respectively. Less than 10% changes in SCr and/or CLcre caused by cobicistat, dolutegravir, and rabeprazole were reproduced as such by our method. These findings suggest that our method can be used to estimate changes in SCr and CLcre caused by competitive inhibitions of renal drug transporters.
Collapse
Affiliation(s)
- Tomohisa Nakada
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan; Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50 Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Toshiyuki Kudo
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Toshiyuki Kume
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50 Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| |
Collapse
|
26
|
Investigation of non-linear Mate1-mediated efflux of trimethoprim in the mouse kidney as the mechanism underlying drug-drug interactions between trimethoprim and organic cations in the kidney. Drug Metab Pharmacokinet 2019; 34:87-94. [DOI: 10.1016/j.dmpk.2018.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/01/2018] [Accepted: 08/20/2018] [Indexed: 01/30/2023]
|
27
|
Müller F, Sharma A, König J, Fromm MF. Biomarkers for In Vivo Assessment of Transporter Function. Pharmacol Rev 2018; 70:246-277. [PMID: 29487084 DOI: 10.1124/pr.116.013326] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-drug interactions are a major concern not only during clinical practice, but also in drug development. Due to limitations of in vitro-in vivo predictions of transporter-mediated drug-drug interactions, multiple clinical Phase I drug-drug interaction studies may become necessary for a new molecular entity to assess potential drug interaction liabilities. This is a resource-intensive process and exposes study participants, who frequently are healthy volunteers without benefit from study treatment, to the potential risks of a new drug in development. Therefore, there is currently a major interest in new approaches for better prediction of transporter-mediated drug-drug interactions. In particular, researchers in the field attempt to identify endogenous compounds as biomarkers for transporter function, such as hexadecanedioate, tetradecanedioate, coproporphyrins I and III, or glycochenodeoxycholate sulfate for hepatic uptake via organic anion transporting polypeptide 1B or N1-methylnicotinamide for multidrug and toxin extrusion protein-mediated renal secretion. We summarize in this review the currently proposed biomarkers and potential limitations of the substances identified to date. Moreover, we suggest criteria based on current experiences, which may be used to assess the suitability of a biomarker for transporter function. Finally, further alternatives and supplemental approaches to classic drug-drug interaction studies are discussed.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Ashish Sharma
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| |
Collapse
|
28
|
Nomani H, Khanmohamadian H, Vaisi-Raygani A, Shakiba E, Tanhapour M, Rahimi Z. Chemerin rs17173608 and vaspin rs2236242 gene variants on the risk of end stage renal disease (ESRD) and correlation with plasma malondialdehyde (MDA) level. Ren Fail 2018; 40:350-356. [PMID: 29644922 PMCID: PMC6014516 DOI: 10.1080/0886022x.2018.1459698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Introduction: End-stage renal disease (ESRD) is associated with critical kidney illness that seriously affects the lifespan. Genetic factors and oxidative stress could play critical role in the development of ESRD. We assessed the association between chemerin rs17173608 T/G and vaspin rs2236242 T/A genes variants with the risk of ESRD and their correlation with plasma malondialdehyde (MDA) level. Materials and methods: In a case-control study, 131 gender and age-matched unrelated healthy controls and 110 ESRD patients were enrolled. The chemerin rs17173608 T/G and vaspin rs2236242 T/A were detected by Tetra primer-amplification refractory mutation system-polymerase chain reaction (T-ARMS-PCR). The MDA concentration was determined by HPLC. Results: Our findings for the first time revealed that in codominant genetic model (T/G vs. T/T genotype), the T/G genotype of chemerin gene significantly had a protective role against ESRD susceptibility. Also, in the presence of chemerin G allele, the risk of ESRD decreased by 0.79-fold (p = .048) in Kurdish population of Iran. The MDA serum levels in ESRD patients carrying the chemerin T/G + G/G genotype of rs17173608 T/G and also in carriers of A/A + T/A genotype of vaspin rs2236242 T/A were significantly higher compared to those in control subjects. The overall distribution of vaspin rs2236242 T/A genotypes and alleles comparing ESRD patients and healthy subjects were not statistically significant. Conclusion: We found that the G allele of chemerin rs17173608 compared to T allele decreased the risk of ESRD, and there was a significant association between chemerin and vaspin variants with plasma MDA level in a sample of the Iranian population.
Collapse
Affiliation(s)
- Hamid Nomani
- a Department of Clinical Biochemistry , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Hamid Khanmohamadian
- a Department of Clinical Biochemistry , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Asad Vaisi-Raygani
- b Fertility and Infertility Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Ebrahim Shakiba
- a Department of Clinical Biochemistry , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Maryam Tanhapour
- b Fertility and Infertility Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Zohreh Rahimi
- a Department of Clinical Biochemistry , Kermanshah University of Medical Sciences , Kermanshah , Iran.,c Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| |
Collapse
|
29
|
Zamek-Gliszczynski MJ, Taub ME, Chothe PP, Chu X, Giacomini KM, Kim RB, Ray AS, Stocker SL, Unadkat JD, Wittwer MB, Xia C, Yee SW, Zhang L, Zhang Y. Transporters in Drug Development: 2018 ITC Recommendations for Transporters of Emerging Clinical Importance. Clin Pharmacol Ther 2018; 104:890-899. [PMID: 30091177 DOI: 10.1002/cpt.1112] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/01/2018] [Indexed: 12/16/2022]
Abstract
This white paper provides updated International Transporter Consortium (ITC) recommendations on transporters that are important in drug development following the 3rd ITC workshop. New additions include prospective evaluation of organic cation transporter 1 (OCT1) and retrospective evaluation of organic anion transporting polypeptide (OATP)2B1 because of their important roles in drug absorption, disposition, and effects. For the first time, the ITC underscores the importance of transporters involved in drug-induced vitamin deficiency (THTR2) and those involved in the disposition of biomarkers of organ function (OAT2 and bile acid transporters).
Collapse
Affiliation(s)
| | - Mitchell E Taub
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim, Ridgefield, Connecticut, USA
| | - Paresh P Chothe
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, California, USA
| | - Richard B Kim
- Division of Clinical Pharmacology, Department of Medicine, Western University, London, ON, Canada
| | - Adrian S Ray
- Clinical Research, Gilead Sciences, Foster City, California, USA
| | - Sophie L Stocker
- Department of Clinical Pharmacology & Toxicology, St Vincent's Hospital, Sydney, NSW, Australia & St Vincent's Clinical School, UNSW Sydney, NSW, Australia
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Matthias B Wittwer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Cindy Xia
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International, Cambridge, Massachusetts, USA
| | - Sook-Wah Yee
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, California, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yan Zhang
- Drug Metabolism Pharmacokinetics & Clinical Pharmacology, Incyte, Wilmington, Delaware, USA
| | | |
Collapse
|
30
|
Omote S, Matsuoka N, Arakawa H, Nakanishi T, Tamai I. Effect of tyrosine kinase inhibitors on renal handling of creatinine by MATE1. Sci Rep 2018; 8:9237. [PMID: 29915248 PMCID: PMC6006426 DOI: 10.1038/s41598-018-27672-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/05/2018] [Indexed: 01/09/2023] Open
Abstract
Creatinine is actively secreted across tubular epithelial cells via organic cation transporter 2 (OCT2) and multidrug and toxin extrusion 1 (MATE1). We previously showed that the tyrosine kinase inhibitor (TKI) crizotinib inhibits OCT2-mediated transport of creatinine. In the present work, we examined the inhibitory potency of TKIs, including crizotinib, on MATE1-mediated transport of creatinine. Then, we used the kinetic parameters estimated in this and the previous work to predict the potential impact of TKIs on serum creatinine level (SCr) via reversible inhibition of creatinine transport. Crizotinib inhibited [14C]creatinine uptake by MATE1-overexpressing cells, and the inhibitory effect increased with incubation time, being greater in the case of pre-incubation or combined pre-incubation/co-incubation (pre/co-incubation) than in the case of co-incubation alone. The inhibition was non-competitive, with K i values of 2.34 μM, 0.455 μM and 0.342 μM under co-, pre- or pre/co-incubation conditions, respectively. Similar values were obtained for inhibition of [3H]MPP+ uptake by MATE1-overexpressing cells. Gefitinib, imatinib, pazopanib, sorafenib, and sunitinib also inhibited MATE1-mediated creatinine uptake. Further, all these TKIs except pazopanib inhibited [14C]creatinine uptake by OCT2-overexpressing cells. In rat kidney slices, the ratio of unbound tissue accumulation of TKIs to extracellular concentration ranged from 2.05 to 3.93. Prediction of the influence of TKIs on SCr based on the renal creatinine clearance and plasma maximum unbound concentrations of TKIs suggested that crizotinib and imatinib might increase SCr by more than 10% in the clinical context. Accordingly, it is necessary to be cautious in diagnosing TKI-induced renal failure only on the basis of an increase of SCr.
Collapse
Affiliation(s)
- Saki Omote
- Faculty of Pharmaceutical Science, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Natsumi Matsuoka
- Faculty of Pharmaceutical Science, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Science, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Takeo Nakanishi
- Faculty of Pharmaceutical Science, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Science, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
31
|
Takehara I, Yoshikado T, Ishigame K, Mori D, Furihata KI, Watanabe N, Ando O, Maeda K, Sugiyama Y, Kusuhara H. Comparative Study of the Dose-Dependence of OATP1B Inhibition by Rifampicin Using Probe Drugs and Endogenous Substrates in Healthy Volunteers. Pharm Res 2018; 35:138. [PMID: 29748935 DOI: 10.1007/s11095-018-2416-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/22/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE To evaluate association of the dose-dependent effect of rifampicin, an OATP1B inhibitor, on the plasma concentration-time profiles among OATP1B substrates drugs and endogenous substrates. METHODS Eight healthy volunteers received atorvastatin (1 mg), pitavastatin (0.2 mg), rosuvastatin (0.5 mg), and fluvastatin (2 mg) alone or with rifampicin (300 or 600 mg) in a crossover fashion. The plasma concentrations of these OATP1B probe drugs, total and direct bilirubin, glycochenodeoxycholate-3-sulfate (GCDCA-S), and coproporphyrin I, were determined. RESULTS The most striking effect of 600 mg rifampicin was on atorvastatin (6.0-times increase) and GCDCA-S (10-times increase). The AUC0-24h of atorvastatin was reasonably correlated with that of pitavastatin (r2 = 0.73) and with the AUC0-4h of fluvastatin (r2 = 0.62) and sufficiently with the AUC0-24h of rosuvastatin (r2 = 0.32). The AUC0-24h of GCDCA-S was reasonably correlated with those of direct bilirubin (r2 = 0.74) and coproporphyrin I (r2 = 0.78), and sufficiently with that of total bilirubin (r2 = 0.30). The AUC0-24h of GCDCA-S, direct bilirubin, and coproporphyrin I were reasonably correlated with that of atorvastatin (r2 = 0.48-0.70) [corrected]. CONCLUSION These results suggest that direct bilirubin, GCDCA-S, and coproporphyrin I are promising surrogate probes for the quantitative assessment of potential OATP1B-mediated DDI.
Collapse
Affiliation(s)
- Issey Takehara
- Biomarker Department, Daiichi Sankyo Co. Ltd., Tokyo, Japan.,Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takashi Yoshikado
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama-shi, Kanagawa, 245-0066, Japan.,Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Japan
| | - Keiko Ishigame
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Japan
| | - Daiki Mori
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | | | - Nobuaki Watanabe
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Osamu Ando
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
32
|
Izumi S, Nozaki Y, Kusuhara H, Hotta K, Mochizuki T, Komori T, Maeda K, Sugiyama Y. Relative Activity Factor (RAF)-Based Scaling of Uptake Clearance Mediated by Organic Anion Transporting Polypeptide (OATP) 1B1 and OATP1B3 in Human Hepatocytes. Mol Pharm 2018; 15:2277-2288. [DOI: 10.1021/acs.molpharmaceut.8b00138] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Saki Izumi
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Yoshitane Nozaki
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical
Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan
| | - Koichiro Hotta
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Toshiki Mochizuki
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Takafumi Komori
- Drug Metabolism and Pharmacokinetics Tsukuba, Tsukuba Research
Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba-shi, Ibaraki 300-2635, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical
Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-003, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama-shi, Kanagawa 230-0045, Japan
| |
Collapse
|
33
|
Nakada T, Kudo T, Kume T, Kusuhara H, Ito K. Quantitative analysis of elevation of serum creatinine via renal transporter inhibition by trimethoprim in healthy subjects using physiologically-based pharmacokinetic model. Drug Metab Pharmacokinet 2017; 33:103-110. [PMID: 29361388 DOI: 10.1016/j.dmpk.2017.11.314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/20/2017] [Accepted: 11/28/2017] [Indexed: 12/17/2022]
Abstract
Serum creatinine (SCr) levels rise during trimethoprim therapy for infectious diseases. This study aimed to investigate whether the elevation of SCr can be quantitatively explained using a physiologically-based pharmacokinetic (PBPK) model incorporating inhibition by trimethoprim on tubular secretion of creatinine via renal transporters such as organic cation transporter 2 (OCT2), OCT3, multidrug and toxin extrusion protein 1 (MATE1), and MATE2-K. Firstly, pharmacokinetic parameters in the PBPK model of trimethoprim were determined to reproduce the blood concentration profile after a single intravenous and oral administration of trimethoprim in healthy subjects. The model was verified with datasets of both cumulative urinary excretions after a single administration and the blood concentration profile after repeated oral administration. The pharmacokinetic model of creatinine consisted of the creatinine synthesis rate, distribution volume, and creatinine clearance (CLcre), including tubular secretion via each transporter. When combining the models for trimethoprim and creatinine, the predicted increments in SCr from baseline were 29.0%, 39.5%, and 25.8% at trimethoprim dosages of 5 mg/kg (b.i.d.), 5 mg/kg (q.i.d.), and 200 mg (b.i.d.), respectively, which were comparable with the observed values. The present model analysis enabled us to quantitatively explain increments in SCr during trimethoprim treatment by its inhibition of renal transporters.
Collapse
Affiliation(s)
- Tomohisa Nakada
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan; Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50 Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Toshiyuki Kudo
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Toshiyuki Kume
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-2-50 Kawagishi, Toda-shi, Saitama 335-8505, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| |
Collapse
|
34
|
Mathialagan S, Costales C, Tylaska L, Kimoto E, Vildhede A, Johnson J, Johnson N, Sarashina T, Hashizume K, Isringhausen CD, Vermeer LMM, Wolff AR, Rodrigues AD. In vitro studies with two human organic anion transporters: OAT2 and OAT7. Xenobiotica 2017; 48:1037-1049. [DOI: 10.1080/00498254.2017.1384595] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sumathy Mathialagan
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Chester Costales
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Laurie Tylaska
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Emi Kimoto
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Anna Vildhede
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Jillian Johnson
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | - Nathaniel Johnson
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| | | | | | | | | | | | - A. David Rodrigues
- Pharmacokinetics, Dynamics, & Metabolism, Medicine Design, Pfizer Inc, Groton, CT, USA,
| |
Collapse
|
35
|
Hibma JE, Zur AA, Castro RA, Wittwer MB, Keizer RJ, Yee SW, Goswami S, Stocker SL, Zhang X, Huang Y, Brett CM, Savic RM, Giacomini KM. The Effect of Famotidine, a MATE1-Selective Inhibitor, on the Pharmacokinetics and Pharmacodynamics of Metformin. Clin Pharmacokinet 2017; 55:711-21. [PMID: 26597253 DOI: 10.1007/s40262-015-0346-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Pharmacokinetic outcomes of transporter-mediated drug-drug interactions (TMDDIs) are increasingly being evaluated clinically. The goal of our study was to determine the effects of selective inhibition of multidrug and toxin extrusion protein 1 (MATE1), using famotidine, on the pharmacokinetics and pharmacodynamics of metformin in healthy volunteers. METHODS Volunteers received metformin alone or with famotidine in a crossover design. As a positive control, the longitudinal effects of famotidine on the plasma levels of creatinine (an endogenous substrate of MATE1) were quantified in parallel. Famotidine unbound concentrations in plasma reached 1 µM, thus exceeding the in vitro concentrations that inhibit MATE1 [concentration of drug producing 50 % inhibition (IC50) 0.25 µM]. Based on current regulatory guidance, these concentrations are expected to inhibit MATE1 clinically [i.e. maximum unbound plasma drug concentration (C max,u)/IC50 >0.1]. RESULTS Consistent with MATE1 inhibition, famotidine administration significantly altered creatinine plasma and urine levels in opposing directions (p < 0.005). Interestingly, famotidine increased the estimated bioavailability of metformin [cumulative amount of unchanged drug excreted in urine from time zero to infinity (A e∞)/dose; p < 0.005] without affecting its systemic exposure [area under the plasma concentration-time curve (AUC) or maximum concentration in plasma (C max)] as a result of a counteracting increase in metformin renal clearance. Moreover, metformin-famotidine co-therapy caused a transient effect on oral glucose tolerance tests [area under the glucose plasma concentration-time curve between time zero and 0.5 h (AUCglu,0.5); p < 0.005]. CONCLUSIONS These results suggest that famotidine may improve the bioavailability and enhance the renal clearance of metformin.
Collapse
Affiliation(s)
- Jennifer E Hibma
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA.,Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, USA
| | - Arik A Zur
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Richard A Castro
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Matthias B Wittwer
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Ron J Keizer
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Srijib Goswami
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Sophie L Stocker
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | | | - Yong Huang
- Optivia Biotechnology Inc., Menlo Park, CA, USA
| | - Claire M Brett
- Department of Anesthesiology, University of California San Francisco, San Francisco, CA, USA
| | - Radojka M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, 1550 4th St, RH 584, Box 2911, San Francisco, CA, 94158, USA.
| |
Collapse
|
36
|
Lepist EI, Ray AS. Beyond drug-drug interactions: effects of transporter inhibition on endobiotics, nutrients and toxins. Expert Opin Drug Metab Toxicol 2017; 13:1075-1087. [PMID: 28847160 DOI: 10.1080/17425255.2017.1372425] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Membrane transport proteins play a central role in regulating the disposition of endobiotics, dietary nutrients and environmental toxins. The inhibition of transporters by drugs has potential physiologic consequences. The full extent of the effect of drugs on the function of transporters is poorly understood because only a small subset of the hundreds of transporters expressed in humans - primarily those mediating the rate-determining step in the elimination of specific drugs - are assessed during clinical development. Areas covered: We provide a comprehensive overview of literature reports implicating the inhibition of transporters as the mechanism for off-target effects of drugs. Expert opinion: Transporter inhibition, the mechanism of action of many marketed drugs, appears to play an underappreciated role in a number of side effects including vitamin deficiency, edema, dyslipidemia, cholestasis and gout. Cell systems more broadly expressing transporter networks and methods like unbiased metabolomics should be incorporated into the screening paradigm to expand our understanding of the impact of drugs on the physiologic function of transporters and to allow for these effects to be taken into account in drug discovery and clinical practice.
Collapse
Affiliation(s)
- Eve-Irene Lepist
- a Departments of Drug Metabolism , Gilead Sciences, Inc ., Foster City , CA , USA
| | - Adrian S Ray
- b Clinical Research , Gilead Sciences, Inc ., Foster City , CA , USA
| |
Collapse
|
37
|
Feng B, Varma MV. Evaluation and Quantitative Prediction of Renal Transporter-Mediated Drug-Drug Interactions. J Clin Pharmacol 2017; 56 Suppl 7:S110-21. [PMID: 27385169 DOI: 10.1002/jcph.702] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/28/2015] [Indexed: 12/22/2022]
Abstract
With numerous drugs cleared renally, inhibition of uptake transporters localized on the basolateral membrane of renal proximal tubule cells, eg, organic anion transporters (OATs) and organic cation transporters (OCTs), may lead to clinically meaningful drug-drug interactions (DDIs). Additionally, clinical evidence for the possible involvement of efflux transporters, such as P-glycoprotein (P-gp) and multidrug and toxin extrusion protein 1/2-K (MATE1/2-K), in the renal DDIs is emerging. Herein, we review recent progress regarding mechanistic understanding of transporter-mediated renal DDIs as well as the quantitative predictability of renal DDIs using static and physiologically based pharmacokinetic (PBPK) models. Generally, clinical DDI data suggest that the magnitude of plasma exposure changes attributable to renal DDIs is less than 2-fold, unlike the DDIs associated with inhibition of cytochrome P-450s and/or hepatic uptake transporters. It is concluded that although there is a need for risk assessment early in drug development, current available data imply that safety concerns related to the renal DDIs are generally low. Nevertheless, consideration must be given to the therapeutic index of the victim drug and potential risk in a specific patient population (eg, renal impairment). Finally, in vitro transporter data and clinical pharmacokinetic parameters obtained from the first-in-human studies have proven useful in support of quantitative prediction of DDIs associated with inhibition of renal secretory transporters, OATs or OCTs.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research & Development, Groton, CT, USA
| | - Manthena V Varma
- Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Global Research & Development, Groton, CT, USA
| |
Collapse
|
38
|
Posada MM, Cannady EA, Payne CD, Zhang X, Bacon JA, Pak YA, Higgins JW, Shahri N, Hall SD, Hillgren KM. Prediction of Transporter-Mediated Drug-Drug Interactions for Baricitinib. Clin Transl Sci 2017; 10:509-519. [PMID: 28749581 PMCID: PMC6402191 DOI: 10.1111/cts.12486] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/30/2017] [Indexed: 01/05/2023] Open
Abstract
Baricitinib, an oral selective Janus kinase 1 and 2 inhibitor, undergoes active renal tubular secretion. Baricitinib was not predicted to inhibit hepatic and renal uptake and efflux drug transporters, based on the ratio of the unbound maximum eliminating-organ inlet concentration and the in vitro half-maximal inhibitory concentrations (IC50 ). In vitro, baricitinib was a substrate for organic anion transporter (OAT)3, multidrug and toxin extrusion protein (MATE)2-K, P-glycoprotein (P-gp), and breast cancer resistance protein (BCRP). Probenecid, a strong OAT3 inhibitor, increased the area under the concentration-time curve from time zero to infinity (AUC[0-∞] ) of baricitinib by twofold and decreased renal clearance to 69% of control in healthy subjects. Physiologically based pharmacokinetic (PBPK) modeling reproduced the renal clearance of baricitinib and the inhibitory effect of probenecid using the in vitro IC50 value of 4.4 μM. Using ibuprofen and diclofenac in vitro IC50 values of 4.4 and 3.8 μM toward OAT3, 1.2 and 1.0 AUC(0-∞) ratios of baricitinib were predicted. These predictions suggest clinically relevant drug-drug interactions (DDIs) with ibuprofen and diclofenac are unlikely.
Collapse
Affiliation(s)
| | | | | | - Xin Zhang
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | - Y Anne Pak
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - J William Higgins
- Eli Lilly and Company, Indianapolis, Indiana, USA.,Current address: Organovo Inc., San Diego, California, USA
| | | | | | | |
Collapse
|
39
|
Rodrigues AD, Taskar KS, Kusuhara H, Sugiyama Y. Endogenous Probes for Drug Transporters: Balancing Vision With Reality. Clin Pharmacol Ther 2017; 103:434-448. [DOI: 10.1002/cpt.749] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/04/2017] [Accepted: 05/15/2017] [Indexed: 12/17/2022]
Affiliation(s)
- AD Rodrigues
- Pharmacokinetics; Dynamics & Metabolism, Medicine Design, Pfizer Inc.; Groton Connecticut USA
| | - KS Taskar
- Mechanistic Safety and Disposition; IVIVT, GlaxoSmithKline; Ware Hertfordshire UK
| | - H Kusuhara
- Laboratory of Molecular Pharmacokinetics; Graduate School of Pharmaceutical Sciences, University of Tokyo; Tokyo Japan
| | - Y Sugiyama
- RIKEN Innovation Center; Research Cluster for Innovation; RIKEN Kanagawa Japan
| |
Collapse
|
40
|
Mathialagan S, Rodrigues AD, Feng B. Evaluation of Renal Transporter Inhibition Using Creatinine as a Substrate In Vitro to Assess the Clinical Risk of Elevated Serum Creatinine. J Pharm Sci 2017; 106:2535-2541. [PMID: 28416419 DOI: 10.1016/j.xphs.2017.04.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/27/2017] [Accepted: 04/07/2017] [Indexed: 11/29/2022]
Abstract
Creatinine is a widely accepted biomarker for renal toxicity, but its renal clearance via transporter-mediated active secretion is significant. For a given new chemical entity, therefore, elevations in serum creatinine (SCr) can be caused by the inhibition of renal transporter(s) without renal toxicity. In the present study, an effort was made to assess the correlation between the inhibition of renal transporters in vitro and elevations in SCr. A total of 15 compounds were chosen based on their known effect on SCr and minimal impact on glomerular filtration rate. Their inhibition potencies against the major creatinine renal transporters, including organic cation transporter 2, organic anion transporter 2, and 2 forms of multidrug and toxin extrusion (MATE1 and MATE2K), were assessed in transporter-transfected cell lines using creatinine as a probe substrate. Collectively, the data suggest that the observed elevations in SCr can be attributed to the inhibition of renal transporter(s), but inhibition of renal transporters does not necessarily lead to elevated SCr. Thus, renal transporter inhibition data can be used to rationalize SCr changes. Additionally, differing renal transporter inhibition potencies using creatinine and metformin as probe substrates suggest that substrate-dependent inhibition exists for some compounds.
Collapse
Affiliation(s)
- Sumathy Mathialagan
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut 06340
| | - A David Rodrigues
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut 06340
| | - Bo Feng
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Pfizer Inc., Groton, Connecticut 06340.
| |
Collapse
|
41
|
Arakawa H, Omote S, Tamai I. Inhibitory Effect of Crizotinib on Creatinine Uptake by Renal Secretory Transporter OCT2. J Pharm Sci 2017; 106:2899-2903. [PMID: 28336299 DOI: 10.1016/j.xphs.2017.03.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 12/29/2022]
Abstract
Crizotinib, a tyrosine kinase inhibitor, exhibits some cases of an increase in serum creatinine levels. Creatinine is excreted by not only glomerular filtration but also active secretion by organic cation transporters such as organic cation transporter 2 (OCT2). In the present study, we evaluated in vitro inhibitory effect of crizotinib on OCT2 by directly measuring creatinine uptake by OCT2. Coincubation of crizotinib reduced uptake of [14C]creatinine by cultured HEK293 cells expressing OCT2 (HEK293/OCT2) in a concentration-dependent manner with IC50 values of 1.58 ± 0.24 μM. Preincubation or both preincubation and coincubation (preincubation/coincubation) with crizotinib showed stronger inhibitory effect on [14C]creatinine uptake compared with that in coincubation alone with IC50 values of 0.499 ± 0.076 and 0.347 ± 0.040 μM, respectively. These IC50 values of crizotinib on [3H]N-methyl-4-phenylpyridinium acetate uptake by OCT2 were 10-20 times higher than those of [14C]creatinine uptake. Furthermore, preincubation of crizotinib inhibited creatinine uptake by OCT2 in an apparently competitive manner. In conclusion, crizotinib at a clinically relevant concentration has the potential to inhibit creatinine transport by OCT2, suggesting an increase of serum creatinine levels in clinical use.
Collapse
Affiliation(s)
- Hiroshi Arakawa
- Department of Membrane Transport of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Saki Omote
- Department of Membrane Transport of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ikumi Tamai
- Department of Membrane Transport of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
42
|
Mathialagan S, Piotrowski MA, Tess DA, Feng B, Litchfield J, Varma MV. Quantitative Prediction of Human Renal Clearance and Drug-Drug Interactions of Organic Anion Transporter Substrates Using In Vitro Transport Data: A Relative Activity Factor Approach. Drug Metab Dispos 2017; 45:409-417. [DOI: 10.1124/dmd.116.074294] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 11/22/2022] Open
|
43
|
Shen H, Lai Y, Rodrigues AD. Organic Anion Transporter 2: An Enigmatic Human Solute Carrier. Drug Metab Dispos 2016; 45:228-236. [PMID: 27872146 DOI: 10.1124/dmd.116.072264] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/17/2016] [Indexed: 12/28/2022] Open
Abstract
As a member of the solute carrier 22A (SLC22A) family, organic anion transporter 2 (OAT2; SLC22A7) is emerging as an important drug transporter because of its expression in both the liver and kidney, two major eliminating organs, and its ability to transport not only a wide variety of xenobiotics but also numerous physiologically important endogenous compounds, like creatinine and cGMP. However, OAT2 has received relatively little attention compared with other OATs and solute carriers (SLCs), like organic cation transporters, sodium-dependent taurocholate cotransporting polypeptide, multidrug and toxin extrusion proteins, and organic anion-transporting polypeptides. Overall, the literature describing OAT2 is rapidly evolving, with numerous publications contradicting each other regarding the transport mechanism, tissue distribution, and transport of creatinine and cGMP, two important endogenous OAT2 substrates. Despite its status as a liver and kidney SLC, tools for assessing its activity and inhibition are lacking, and its role in drug disposition and elimination remains to be defined. The current review focuses on the available and emerging literature describing OAT2. We envision that OAT2 will gain more prominence as its expression, substrate, and inhibitor profile is investigated further and compared with other SLCs.
Collapse
Affiliation(s)
- Hong Shen
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - Yurong Lai
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| | - A David Rodrigues
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, Princeton, New Jersey (H.S., Y.L.), and Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer World Wide Research and Development, Groton, Connecticut (A.D.R.)
| |
Collapse
|
44
|
Tsuruya Y, Kato K, Sano Y, Imamura Y, Maeda K, Kumagai Y, Sugiyama Y, Kusuhara H. Investigation of Endogenous Compounds Applicable to Drug–Drug Interaction Studies Involving the Renal Organic Anion Transporters, OAT1 and OAT3, in Humans. Drug Metab Dispos 2016; 44:1925-1933. [DOI: 10.1124/dmd.116.071472] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/14/2016] [Indexed: 01/09/2023] Open
|
45
|
Hsueh CH, Yoshida K, Zhao P, Meyer TW, Zhang L, Huang SM, Giacomini KM. Identification and Quantitative Assessment of Uremic Solutes as Inhibitors of Renal Organic Anion Transporters, OAT1 and OAT3. Mol Pharm 2016; 13:3130-40. [PMID: 27467266 DOI: 10.1021/acs.molpharmaceut.6b00332] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
One of the characteristics of chronic kidney disease (CKD) is the accumulation of uremic solutes in the plasma. Less is known about the effects of uremic solutes on transporters that may play critical roles in pharmacokinetics. We evaluated the effect of 72 uremic solutes on organic anion transporter 1 and 3 (OAT1 and OAT3) using a fluorescent probe substrate, 6-carboxyfluorescein. A total of 12 and 13 solutes were identified as inhibitors of OAT1 and OAT3, respectively. Several of them inhibited OAT1 or OAT3 at clinically relevant concentrations and reduced the transport of other OAT1/3 substrates in vitro. Review of clinical studies showed that the active secretion of most drugs that are known substrates of OAT1/3 deteriorated faster than the renal filtration in CKD. Collectively, these data suggest that through inhibition of OAT1 and OAT3, uremic solutes contribute to the decline in renal drug clearance in patients with CKD.
Collapse
Affiliation(s)
- Chia-Hsiang Hsueh
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco , San Francisco, California 94158, United States.,Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation & Research, US Food and Drug Administration , Silver Spring, Maryland 20993, United States
| | - Kenta Yoshida
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation & Research, US Food and Drug Administration , Silver Spring, Maryland 20993, United States
| | - Ping Zhao
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation & Research, US Food and Drug Administration , Silver Spring, Maryland 20993, United States
| | - Timothy W Meyer
- Division of Nephrology, School of Medicine, Stanford University , Stanford, California 94305, United States
| | - Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation & Research, US Food and Drug Administration , Silver Spring, Maryland 20993, United States
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation & Research, US Food and Drug Administration , Silver Spring, Maryland 20993, United States
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco , San Francisco, California 94158, United States
| |
Collapse
|
46
|
Liu HC, Goldenberg A, Chen Y, Lun C, Wu W, Bush KT, Balac N, Rodriguez P, Abagyan R, Nigam SK. Molecular Properties of Drugs Interacting with SLC22 Transporters OAT1, OAT3, OCT1, and OCT2: A Machine-Learning Approach. J Pharmacol Exp Ther 2016; 359:215-29. [PMID: 27488918 DOI: 10.1124/jpet.116.232660] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/20/2016] [Indexed: 11/22/2022] Open
Abstract
Statistical analysis was performed on physicochemical descriptors of ∼250 drugs known to interact with one or more SLC22 "drug" transporters (i.e., SLC22A6 or OAT1, SLC22A8 or OAT3, SLC22A1 or OCT1, and SLC22A2 or OCT2), followed by application of machine-learning methods and wet laboratory testing of novel predictions. In addition to molecular charge, organic anion transporters (OATs) were found to prefer interacting with planar structures, whereas organic cation transporters (OCTs) interact with more three-dimensional structures (i.e., greater SP3 character). Moreover, compared with OAT1 ligands, OAT3 ligands possess more acyclic tetravalent bonds and have a more zwitterionic/cationic character. In contrast, OCT1 and OCT2 ligands were not clearly distinquishable form one another by the methods employed. Multiple pharmacophore models were generated on the basis of the drugs and, consistent with the machine-learning analyses, one unique pharmacophore created from ligands of OAT3 possessed cationic properties similar to OCT ligands; this was confirmed by quantitative atomic property field analysis. Virtual screening with this pharmacophore, followed by transport assays, identified several cationic drugs that selectively interact with OAT3 but not OAT1. Although the present analysis may be somewhat limited by the need to rely largely on inhibition data for modeling, wet laboratory/in vitro transport studies, as well as analysis of drug/metabolite handling in Oat and Oct knockout animals, support the general validity of the approach-which can also be applied to other SLC and ATP binding cassette drug transporters. This may make it possible to predict the molecular properties of a drug or metabolite necessary for interaction with the transporter(s), thereby enabling better prediction of drug-drug interactions and drug-metabolite interactions. Furthermore, understanding the overlapping specificities of OATs and OCTs in the context of dynamic transporter tissue expression patterns should help predict net flux in a particular tissue of anionic, cationic, and zwitterionic molecules in normal and pathophysiological states.
Collapse
Affiliation(s)
- Henry C Liu
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Anne Goldenberg
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Yuchen Chen
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Christina Lun
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Wei Wu
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Kevin T Bush
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Natasha Balac
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Paul Rodriguez
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Ruben Abagyan
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| | - Sanjay K Nigam
- Departments of Bioengineering (H.C.L.), Pediatrics (A.G., Y.C., C.L., K.T.B., S.K.N.), Medicine (W.W., S.K.N.), Cellular and Molecular Medicine (S.K.N.), and Pharmacology (R.A.), and the San Diego Supercomputer Center (N.B., P.R.), University of California San Diego, La Jolla, California
| |
Collapse
|
47
|
Chu X, Bleasby K, Chan GH, Nunes I, Evers R. The Complexities of Interpreting Reversible Elevated Serum Creatinine Levels in Drug Development: Does a Correlation with Inhibition of Renal Transporters Exist? ACTA ACUST UNITED AC 2016; 44:1498-509. [PMID: 26825641 DOI: 10.1124/dmd.115.067694] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/28/2016] [Indexed: 12/19/2022]
Abstract
In humans, creatinine is formed by a multistep process in liver and muscle and eliminated via the kidney by a combination of glomerular filtration and active transport. Based on current evidence, creatinine can be taken up into renal proximal tubule cells by the basolaterally localized organic cation transporter 2 (OCT2) and the organic anion transporter 2, and effluxed into the urine by the apically localized multidrug and toxin extrusion protein 1 (MATE1) and MATE2K. Drug-induced elevation of serum creatinine (SCr) and/or reduced creatinine renal clearance is routinely used as a marker for acute kidney injury. Interpretation of elevated SCr can be complex, because such increases can be reversible and explained by inhibition of renal transporters involved in active secretion of creatinine or other secondary factors, such as diet and disease state. Distinction between these possibilities is important from a drug development perspective, as increases in SCr can result in the termination of otherwise efficacious drug candidates. In this review, we discuss the challenges associated with using creatinine as a marker for kidney damage. Furthermore, to evaluate whether reversible changes in SCr can be predicted prospectively based on in vitro transporter inhibition data, an in-depth in vitro-in vivo correlation (IVIVC) analysis was conducted for 16 drugs with in-house and literature in vitro transporter inhibition data for OCT2, MATE1, and MATE2K, as well as total and unbound maximum plasma concentration (Cmax and Cmax,u) data measured in the clinic.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Kelly Bleasby
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Grace Hoyee Chan
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Irene Nunes
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| | - Raymond Evers
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism (X.C., K.B., G.H.C., R.E.), and Global Regulatory Affairs, Oncology, Immunology, Biologics & Devices (I.N.), Merck Sharp & Dohme Corporation, Kenilworth, New Jersey
| |
Collapse
|
48
|
Shen H, Liu T, Jiang H, Titsch C, Taylor K, Kandoussi H, Qiu X, Chen C, Sukrutharaj S, Kuit K, Mintier G, Krishnamurthy P, Fancher RM, Zeng J, Rodrigues AD, Marathe P, Lai Y. Cynomolgus Monkey as a Clinically Relevant Model to Study Transport Involving Renal Organic Cation Transporters: In Vitro and In Vivo Evaluation. Drug Metab Dispos 2015; 44:238-49. [DOI: 10.1124/dmd.115.066852] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/19/2015] [Indexed: 01/12/2023] Open
|
49
|
Nigam SK, Wu W, Bush KT, Hoenig MP, Blantz RC, Bhatnagar V. Handling of Drugs, Metabolites, and Uremic Toxins by Kidney Proximal Tubule Drug Transporters. Clin J Am Soc Nephrol 2015; 10:2039-49. [PMID: 26490509 DOI: 10.2215/cjn.02440314] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 09/28/2014] [Indexed: 01/22/2023]
Abstract
The proximal tubule of the kidney plays a crucial role in the renal handling of drugs (e.g., diuretics), uremic toxins (e.g., indoxyl sulfate), environmental toxins (e.g., mercury, aristolochic acid), metabolites (e.g., uric acid), dietary compounds, and signaling molecules. This process is dependent on many multispecific transporters of the solute carrier (SLC) superfamily, including organic anion transporter (OAT) and organic cation transporter (OCT) subfamilies, and the ATP-binding cassette (ABC) superfamily. We review the basic physiology of these SLC and ABC transporters, many of which are often called drug transporters. With an emphasis on OAT1 (SLC22A6), the closely related OAT3 (SLC22A8), and OCT2 (SLC22A2), we explore the implications of recent in vitro, in vivo, and clinical data pertinent to the kidney. The analysis of murine knockouts has revealed a key role for these transporters in the renal handling not only of drugs and toxins but also of gut microbiome products, as well as liver-derived phase 1 and phase 2 metabolites, including putative uremic toxins (among other molecules of metabolic and clinical importance). Functional activity of these transporters (and polymorphisms affecting it) plays a key role in drug handling and nephrotoxicity. These transporters may also play a role in remote sensing and signaling, as part of a versatile small molecule communication network operative throughout the body in normal and diseased states, such as AKI and CKD.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Department of Medicine, Department of Pediatrics, Department of Cell & Molecular Medicine,
| | | | | | - Melanie P Hoenig
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Roland C Blantz
- Division of Nephrology-Hypertension, and Veterans Affairs San Diego Healthcare System, San Diego, California; and
| | - Vibha Bhatnagar
- Division of Family & Preventative Medicine, University of California-San Diego, La Jolla, California
| |
Collapse
|
50
|
Reese MJ, Bowers GD, Humphreys JE, Gould EP, Ford SL, Webster LO, Polli JW. Drug interaction profile of the HIV integrase inhibitor cabotegravir: assessment from in vitro studies and a clinical investigation with midazolam. Xenobiotica 2015; 46:445-56. [PMID: 26340566 DOI: 10.3109/00498254.2015.1081993] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
1. Cabotegravir (CAB; GSK1265744) is a potent HIV integrase inhibitor in clinical development as an oral lead-in tablet and long-acting injectable for the treatment and prevention of HIV infection. 2. This work investigated if CAB was a substrate for efflux transporters, the potential for CAB to interact with drug-metabolizing enzymes and transporters to cause clinical drug interactions, and the effect of CAB on the pharmacokinetics of midazolam, a CYP3A4 probe substrate, in humans. 3. CAB is a substrate for Pgp and BCRP; however, its high intrinsic membrane permeability limits the impact of these transporters on its intestinal absorption. 4. At clinically relevant concentrations, CAB did not inhibit or induce any of the CYP or UGT enzymes evaluated in vitro and had no effect on the clinical pharmacokinetics of midazolam. 5. CAB is an inhibitor of OAT1 (IC50 0.81 µM) and OAT3 (IC50 0.41 µM) but did not or only weakly inhibited Pgp, BCRP, MRP2, MRP4, MATE1, MATE2-K, OATP1B1, OATP1B3, OCT1, OCT2 or BSEP. 6. Based on regulatory guidelines and quantitative extrapolations, CAB has a low propensity to cause clinically significant drug interactions, except for coadministration with OAT1 or OAT3 substrates.
Collapse
Affiliation(s)
- Melinda J Reese
- a Drug Metabolism and Pharmacokinetics, GlaxoSmithKline , Research Triangle Park , NC , USA and
| | - Gary D Bowers
- a Drug Metabolism and Pharmacokinetics, GlaxoSmithKline , Research Triangle Park , NC , USA and
| | - Joan E Humphreys
- a Drug Metabolism and Pharmacokinetics, GlaxoSmithKline , Research Triangle Park , NC , USA and
| | | | - Susan L Ford
- b Clinical Platforms and Sciences, GlaxoSmithKline , RTP , NC , USA
| | - Lindsey O Webster
- a Drug Metabolism and Pharmacokinetics, GlaxoSmithKline , Research Triangle Park , NC , USA and
| | - Joseph W Polli
- a Drug Metabolism and Pharmacokinetics, GlaxoSmithKline , Research Triangle Park , NC , USA and
| |
Collapse
|