1
|
Jin YW, Ma YR, Liu YT, Yang JR, Zhang MK, Ran FL, Chen Y, Wu XA. Identification of a substrate of the renal tubular transporters for detecting drug-induced early acute kidney injury. Toxicol Sci 2024; 201:190-205. [PMID: 39041788 DOI: 10.1093/toxsci/kfae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024] Open
Abstract
Early identification of drug-induced acute kidney injury (AKI) is essential to prevent renal damage. The renal tubules are typically the first to exhibit damage, frequently accompanied by changes in renal tubular transporters. With this in mind, we have identified an endogenous substrate of the renal tubular transporters that may serve as a biomarker for early detection of drug-induced AKI. Using gentamicin- and vancomycin-induced AKI models, we found that traumatic acid (TA), an end metabolite, was rapidly increased in both AKI models. TA, a highly albumin-bound compound (96% to 100%), could not be filtered by the glomerulus and was predominantly eliminated by renal tubules via the OAT1, OAT3, OATP4C1, and P-gp transporters. Importantly, there is a correlation between elevated serum TA levels and reduced OAT1 and OAT3 levels. A clinical study showed that serum TA levels rose before an increase in serum creatinine in 13 out of 20 AKI patients in an intensive care unit setting. In addition, there was a notable rise in TA levels in the serum of individuals suffering from nephrotic syndrome, chronic renal failure, and acute renal failure. These results indicate that the decrease in renal tubular transporter expression during drug-induced AKI leads to an increase in the serum TA level, and the change in TA may serve as a monitor for renal tubular injury. Acute kidney injury (AKI) has a high clinical incidence, and if patients do not receive timely treatment and intervention, it can lead to severe consequences. During AKI, tubular damage is often the primary issue. Endogenous biomarkers of tubular damage are critical for the early diagnosis and treatment of AKI. However, there is currently a lack of reliable endogenous biomarkers for diagnosing tubular damage in clinical practice. Tubular secretion is primarily mediated by renal tubular transporters (channels), which are also impaired during tubular damage. Therefore, we aim to identify endogenous biomarkers of tubular damage from the perspective of renal tubular transporters, providing support for the early detection and intervention of AKI. TA is a substrate of multiple channels, including OAT1, OAT3, OATP4C1, and P-gp, and is primarily secreted by the renal tubules. In the early stages of rat AKI induced by GEN and VCA, serum TA levels are significantly elevated, occurring earlier than the rise in serum creatinine (SCr). Thus, TA is expected to become a potential endogenous biomarker for the early diagnosis of tubular damage.
Collapse
Affiliation(s)
- Yong-Wen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yan-Rong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yu-Ting Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jin-Ru Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Ming-Kang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Feng-Lin Ran
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Xin-An Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
2
|
Hou K, Jabeen R, Sun L, Wei J. How do Mutations of Mycobacterium Genes Cause Drug Resistance in Tuberculosis? Curr Pharm Biotechnol 2024; 25:724-736. [PMID: 37888812 DOI: 10.2174/0113892010257816230920053547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 10/28/2023]
Abstract
A steady increase in the prevalence of drug-resistant tuberculosis (DR-TB) has already been reported in Pakistan. In addition, DR-TB is gradually changing from one-drug resistance to multi-drug resistance, which is a serious challenge for tuberculosis treatment. This review provides an overview of the anti-tuberculosis drugs and focuses on the molecular mechanisms of drug resistance in Mycobacterium tuberculosis, with the hope that it will contribute to the study of drug resistance in response to the emergence of multidrug-resistant tuberculosis.
Collapse
Affiliation(s)
- Kaiying Hou
- School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Riffat Jabeen
- School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Lin Sun
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- School of Life Sciences, Henan University, Kaifeng, 475004, China
| |
Collapse
|
3
|
Hsin CH, Kuehne A, Gu Y, Jedlitschky G, Hagos Y, Gründemann D, Fuhr U. In vitro validation of an in vivo phenotyping drug cocktail for major drug transporters in humans. Eur J Pharm Sci 2023; 186:106459. [PMID: 37142000 DOI: 10.1016/j.ejps.2023.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/19/2023] [Accepted: 05/02/2023] [Indexed: 05/06/2023]
Abstract
PURPOSE Cocktails of transporter probe drugs are used in vivo to assess transporter activity and respective drug-drug interactions. An inhibitory effect of components on transporter activities should be ruled out. Here, for a clinically tested cocktail consisting of adefovir, digoxin, metformin, sitagliptin, and pitavastatin, inhibition of major transporters by individual probe substrates was investigated in vitro. METHODS Transporter transfected HEK293 cells were used in all evaluations. Cell-based assays were applied for uptake by human organic cation transporters 1/2 (hOCT1/2), organic anion transporters 1/3 (hOAT1/3), multidrug and toxin extrusion proteins 1/2K (hMATE1/2K), and organic anion transporter polypeptide 1B1 (hOATP1B1). For P-glycoprotein (hMDR1) a cell-based efflux assay was used whereas an inside-out vesicle-based assay was used for the bile salt export pump (hBSEP). All assays used standard substrates and established inhibitors (as positive controls). Inhibition experiments using clinically achievable concentrations of potential perpetrators at the relevant transporter expression site were carried out initially. If there was a significant effect, the inhibition potency (Ki) was studied in detail. RESULTS In the inhibition tests, only sitagliptin had an effect and reduced hOCT1- and hOCT2- mediated metformin uptake and hMATE2K mediated MPP+ uptake by more than 70%, 80%, and 30%, respectively. The ratios of unbound Cmax (observed clinically) to Ki of sitagliptin were low with 0.009, 0.03, and 0.001 for hOCT1, hOCT2, and hMATE2K, respectively. CONCLUSION The inhibition of hOCT2 in vitro by sitagliptin is in agreement with the borderline inhibition of renal metformin elimination observed clinically, supporting a dose reduction of sitagliptin in the cocktail.
Collapse
Affiliation(s)
- Chih-Hsuan Hsin
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, Cologne, Germany
| | | | - Yi Gu
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, Cologne, Germany
| | - Gabriele Jedlitschky
- Department of General Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany
| | | | - Dirk Gründemann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, Cologne, Germany
| | - Uwe Fuhr
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, Cologne, Germany.
| |
Collapse
|
4
|
[11C]glyburide PET imaging for quantitative determination of the importance of Organic Anion-Transporting Polypeptide transporter function in the human liver and whole-body. Biomed Pharmacother 2022; 156:113994. [DOI: 10.1016/j.biopha.2022.113994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
|
5
|
Chunduri V, Maddi S. Role of in vitro two-dimensional (2D) and three-dimensional (3D) cell culture systems for ADME-Tox screening in drug discovery and development: a comprehensive review. ADMET & DMPK 2022; 11:1-32. [PMID: 36778905 PMCID: PMC9909725 DOI: 10.5599/admet.1513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/07/2022] [Indexed: 11/18/2022]
Abstract
Drug discovery and development have become a very time-consuming and expensive process. Preclinical animal models have become the gold standard for studying drug pharmacokinetic and toxicity parameters. However, the involvement of a huge number of animal subjects and inter-species pathophysiological variations between animals and humans has provoked a lot of debate, particularly because of ethical concerns. Although many efforts are being established by biotech and pharmaceutical companies for screening new chemical entities in vitro before preclinical trials, failures during clinical trials are still involved. Currently, a large number of two- dimensional (2D) in vitro assays have been developed and are being developed by researchers for the screening of compounds. Although these assays are helpful in screening a huge library of compounds and have shown perception, there is a significant lack in predicting human Absorption, Distribution, Metabolism, Excretion and Toxicology (ADME-Tox). As a result, these assays cannot completely replace animal models. The recent inventions in three-dimensional (3D) cell culture-based assays like organoids and micro-physiological systems have shown great potential alternative tools for predicting the compound pharmacokinetic and pharmacodynamic fate in humans. In this comprehensive review, we have summarized some of the most commonly used 2D in vitro assays and emphasized the achievements in next-generation 3D cell culture-based systems for predicting the compound ADME-Tox.
Collapse
|
6
|
Toxicokinetics of β-Amanitin in Mice and In Vitro Drug-Drug Interaction Potential. Pharmaceutics 2022; 14:pharmaceutics14040774. [PMID: 35456608 PMCID: PMC9030691 DOI: 10.3390/pharmaceutics14040774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 01/01/2023] Open
Abstract
The toxicokinetics of β-amanitin, a toxic bicyclic octapeptide present abundantly in Amanitaceae mushrooms, was evaluated in mice after intravenous (iv) and oral administration. The area under plasma concentration curves (AUC) following iv injection increased in proportion to doses of 0.2, 0.4, and 0.8 mg/kg. β-amanitin disappeared rapidly from plasma with a half-life of 18.3−33.6 min, and 52.3% of the iv dose was recovered as a parent form. After oral administration, the AUC again increased in proportion with doses of 2, 5, and 10 mg/kg. Absolute bioavailability was 7.3−9.4%, which resulted in 72.4% of fecal recovery from orally administered β-amanitin. Tissue-to-plasma AUC ratios of orally administered β-amanitin were the highest in the intestine and stomach. It also readily distributed to kidney > spleen > lung > liver ≈ heart. Distribution to intestines, kidneys, and the liver is in agreement with previously reported target organs after acute amatoxin poisoning. In addition, β-amanitin weakly or negligibly inhibited major cytochrome P450 and 5′-diphospho-glucuronosyltransferase activities in human liver microsomes and suppressed drug transport functions in mammalian cells that overexpress transporters, suggesting the remote drug interaction potentials caused by β-amanitin exposure.
Collapse
|
7
|
Nguyen VL, Ahn S, Hoa PQ, Long NP, Ahn S, Cho YS, Shin JG. Center for Personalized Precision Medicine for Tuberculosis: Smart Research and Development Workstation. Healthc Inform Res 2022; 28:176-180. [PMID: 35576986 PMCID: PMC9117804 DOI: 10.4258/hir.2022.28.2.176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Van Lam Nguyen
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
| | - Sangjin Ahn
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
| | - Pham Quang Hoa
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
| | - Nguyen Phuoc Long
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
| | - Sangzin Ahn
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
| | - Yong-Soon Cho
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan,
Korea
| | - Jae-Gook Shin
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan,
Korea
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan,
Korea
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan,
Korea
| |
Collapse
|
8
|
Characterization of Clofazimine as a Potential Substrate of Drug Transporter. Antimicrob Agents Chemother 2022; 66:e0215821. [PMID: 35254089 DOI: 10.1128/aac.02158-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we explored clofazimine (CFZ) as a potential substrate of uptake and efflux transporters that might be involved in CFZ disposition, using transporter gene overexpressing cell lines in vitro. The intracellular concentrations of CFZ were significantly increased in the presence of selective inhibitors of P-gp and BCRP, which include verapamil, cyclosporine-A, PSC-833, quinidine, Ko143, and daunorubicin. In a bidirectional transport assay using transwell cultures of cell lines overexpressing P-gp and BCRP, the mean efflux ratios of CFZ were found to be 4.17 ± 0.63 and 3.37 ± 1.2, respectively. The Km and maximum rate of uptake (Vmax) were estimated to be 223.3 ± 14.73 μM and 548.8 ± 87.15 pmol/min/mg protein for P-gp and 381.9 ± 25.07 μM and 5.8 ± 1.22 pmol/min/mg protein for BCRP, respectively. Among the uptake transporters screened, the CFZ uptake rate was increased 1.93 and 3.09-fold in HEK293 cell lines overexpressing OAT1 and OAT3, respectively, compared to the control cell lines, but no significant uptake was observed in cell lines overexpressing OCT1, OCT2, OATP1B1, OATP1B3, OATP2B1, or NTCP. Both OAT1- and OAT3-mediated uptake was inhibited by the selective inhibitors diclofenac, probenecid, and butanesulfonic acid. The Km and Vmax values of CFZ were estimated to be 0.63 ± 0.15 μM and 8.23 ± 1.03 pmol/min/mg protein, respectively, for OAT1 and 0.47 ± 0.1 μM and 17.81 ± 2.19 pmol/min/mg protein, respectively, for OAT3. These findings suggest that CFZ is a novel substrate of BCRP, OAT1, and OAT3 and a known substrate of P-gp in vitro.
Collapse
|
9
|
Zhang L, Sarangi V, Ho MF, Moon I, Kalari KR, Wang L, Weinshilboum RM. SLCO1B1: Application and Limitations of Deep Mutational Scanning for Genomic Missense Variant Function. Drug Metab Dispos 2021; 49:395-404. [PMID: 33658230 PMCID: PMC8042483 DOI: 10.1124/dmd.120.000264] [Citation(s) in RCA: 16] [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: 09/29/2020] [Accepted: 02/17/2021] [Indexed: 01/07/2023] Open
Abstract
SLCO1B1 (solute carrier organic anion transporter family member 1B1) is an important transmembrane hepatic uptake transporter. Genetic variants in the SLCO1B1 gene have been associated with altered protein folding, resulting in protein degradation and decreased transporter activity. Next-generation sequencing (NGS) of pharmacogenes is being applied increasingly to associate variation in drug response with genetic sequence variants. However, it is difficult to link variants of unknown significance with functional phenotypes using "one-at-a-time" functional systems. Deep mutational scanning (DMS) using a "landing pad cell-based system" is a high-throughput technique designed to analyze hundreds of gene open reading frame (ORF) missense variants in a parallel and scalable fashion. We have applied DMS to analyze 137 missense variants in the SLCO1B1 ORF obtained from the Exome Aggregation Consortium project. ORFs containing these variants were fused to green fluorescent protein and were integrated into "landing pad" cells. Florescence-activated cell sorting was performed to separate the cells into four groups based on fluorescence readout indicating protein expression at the single cell level. NGS was then performed and SLCO1B1 variant frequencies were used to determine protein abundance. We found that six variants not previously characterized functionally displayed less than 25% and another 12 displayed approximately 50% of wild-type protein expression. These results were then functionally validated by transporter studies. Severely damaging variants identified by DMS may have clinical relevance for SLCO1B1-dependent drug transport, but we need to exercise caution since the relatively small number of severely damaging variants identified raise questions with regard to the application of DMS to intrinsic membrane proteins such as organic anion transporter protein 1B1. SIGNIFICANCE STATEMENT: The functional implications of a large numbers of open reading frame (ORF) "variants of unknown significance" (VUS) in transporter genes have not been characterized. This study applied deep mutational scanning to determine the functional effects of VUS that have been observed in the ORF of SLCO1B1(s olute carrier organic anion transporter family member 1B1). Several severely damaging variants were identified, studied, and validated. These observations have implications for both the application of deep mutational scanning to intrinsic membrane proteins and for the clinical effect of drugs and endogenous compounds transported by SLCO1B1.
Collapse
Affiliation(s)
- Lingxin Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Vivekananda Sarangi
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Irene Moon
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Krishna R Kalari
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics (L.Z., M.-F.H., I.M., L.W., R.M.W.), Division of Biomedical Statistics and Informatics, Department of Health Sciences Research (V.S., K.R.K.), and Mayo Clinic Center for Individualized Medicine (L.W., R.M.W.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
10
|
Yu X, Chu Z, Li J, He R, Wang Y, Cheng C. Pharmacokinetic Drug-drug Interaction of Antibiotics Used in Sepsis Care in China. Curr Drug Metab 2021; 22:5-23. [PMID: 32990533 DOI: 10.2174/1389200221666200929115117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/17/2020] [Accepted: 07/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Many antibiotics have a high potential for interactions with drugs, as a perpetrator and/or victim, in critically ill patients, and particularly in sepsis patients. METHODS The aim of this review is to summarize the pharmacokinetic drug-drug interaction (DDI) of 45 antibiotics commonly used in sepsis care in China. Literature search was conducted to obtain human pharmacokinetics/ dispositions of the antibiotics, their interactions with drug-metabolizing enzymes or transporters, and their associated clinical drug interactions. Potential DDI is indicated by a DDI index ≥ 0.1 for inhibition or a treatedcell/ untreated-cell ratio of enzyme activity being ≥ 2 for induction. RESULTS The literature-mined information on human pharmacokinetics of the identified antibiotics and their potential drug interactions is summarized. CONCLUSION Antibiotic-perpetrated drug interactions, involving P450 enzyme inhibition, have been reported for four lipophilic antibacterials (ciprofloxacin, erythromycin, trimethoprim, and trimethoprim-sulfamethoxazole) and three antifungals (fluconazole, itraconazole, and voriconazole). In addition, seven hydrophilic antibacterials (ceftriaxone, cefamandole, piperacillin, penicillin G, amikacin, metronidazole, and linezolid) inhibit drug transporters in vitro. Despite no clinical PK drug interactions with the transporters, caution is advised in the use of these antibacterials. Eight hydrophilic antibiotics (all β-lactams; meropenem, cefotaxime, cefazolin, piperacillin, ticarcillin, penicillin G, ampicillin, and flucloxacillin), are potential victims of drug interactions due to transporter inhibition. Rifampin is reported to perpetrate drug interactions by inducing CYP3A or inhibiting OATP1B; it is also reported to be a victim of drug interactions, due to the dual inhibition of CYP3A4 and OATP1B by indinavir. In addition, three antifungals (caspofungin, itraconazole, and voriconazole) are reported to be victims of drug interactions because of P450 enzyme induction. Reports for other antibiotics acting as victims in drug interactions are scarce.
Collapse
Affiliation(s)
- Xuan Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zixuan Chu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Rongrong He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaya Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chen Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
11
|
De Vocht T, Buyck C, Deferm N, Qi B, Van Brantegem P, van Vlijmen H, Snoeys J, Hoeben E, Vermeulen A, Annaert P. Identification of novel inhibitors of rat Mrp3. Eur J Pharm Sci 2021; 162:105813. [PMID: 33753214 DOI: 10.1016/j.ejps.2021.105813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/18/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
Multidrug resistance-associated protein (MRP; ABCC gene family) mediated efflux transport plays an important role in the systemic and tissue exposure profiles of many drugs and their metabolites, and also of endogenous compounds like bile acids and bilirubin conjugates. However, potent and isoform-selective inhibitors of the MRP subfamily are currently lacking. Therefore, the purpose of the present work was to identify novel rat Mrp3 inhibitors. Using 5(6)-carboxy-2',7'-dichlorofluorescein diacetate (CDFDA) as a model-(pro)substrate for Mrp3 in an oil-spin assay with primary rat hepatocytes, the extent of inhibition of CDF efflux was determined for 1584 compounds, yielding 59 hits (excluding the reference inhibitor) that were identified as new Mrp3 inhibitors. A naive Bayesian prediction model was constructed in Pipeline Pilot to elucidate physicochemical and structural features of compounds causing Mrp3 inhibition. The final Bayesian model generated common physicochemical properties of Mrp3 inhibitors. For instance, more than half of the hits contain a phenolic structure. The identified compounds have an AlogP between 2 and 4.5, between 5 to 8 hydrogen bond acceptor atoms, a molecular weight between 260 and 400, and 2 or more aromatic rings. Compared to the depleted dataset (i.e. 90% remaining compounds), the Mrp3 hit rate in the enriched set was 7.5-fold higher (i.e. 17.2% versus 2.3%). Several hits from this first screening approach were confirmed in an additional study using Mrp3 transfected inside-out membrane vesicles. In conclusion, several new and potent inhibitors of Mrp3 mediated efflux were identified in an optimized in vitro rat hepatocyte assay and confirmed using Mrp3 transfected inside-out membrane vesicles. A final naive Bayesian model was developed in an iterative way to reveal common physicochemical and structural features for Mrp3 inhibitors. The final Bayesian model will enable in silico screening of larger libraries and in vitro identification of more potent Mrp3 inhibitors.
Collapse
Affiliation(s)
- Tom De Vocht
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg, O&N2, Herestraat 49 box 921, B-3000 Leuven, Belgium
| | - Christophe Buyck
- Discovery Sciences, Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Neel Deferm
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg, O&N2, Herestraat 49 box 921, B-3000 Leuven, Belgium
| | - Bing Qi
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg, O&N2, Herestraat 49 box 921, B-3000 Leuven, Belgium
| | - Pieter Van Brantegem
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg, O&N2, Herestraat 49 box 921, B-3000 Leuven, Belgium
| | - Herman van Vlijmen
- Discovery Sciences, Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Jan Snoeys
- Drug Metabolism and Pharmacokinetics, Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Eef Hoeben
- Quantitative Sciences, Janssen Research and Development, a division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium; BioNotus GCV, Wetenschapspark Universiteit Antwerpen, Galileilaan 15, B-2845 Niel, Belgium
| | - An Vermeulen
- Quantitative Sciences, Janssen Research and Development, a division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg, O&N2, Herestraat 49 box 921, B-3000 Leuven, Belgium; BioNotus GCV, Wetenschapspark Universiteit Antwerpen, Galileilaan 15, B-2845 Niel, Belgium.
| |
Collapse
|
12
|
Izat N, Sahin S. Hepatic transporter-mediated pharmacokinetic drug-drug interactions: Recent studies and regulatory recommendations. Biopharm Drug Dispos 2021; 42:45-77. [PMID: 33507532 DOI: 10.1002/bdd.2262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 12/16/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Transporter-mediated drug-drug interactions are one of the major mechanisms in pharmacokinetic-based drug interactions and correspondingly affecting drugs' safety and efficacy. Regulatory bodies underlined the importance of the evaluation of transporter-mediated interactions as a part of the drug development process. The liver is responsible for the elimination of a wide range of endogenous and exogenous compounds via metabolism and biliary excretion. Therefore, hepatic uptake transporters, expressed on the sinusoidal membranes of hepatocytes, and efflux transporters mediating the transport from hepatocytes to the bile are determinant factors for pharmacokinetics of drugs, and hence, drug-drug interactions. In parallel with the growing research interest in this area, regulatory guidances have been updated with detailed assay models and criteria. According to well-established preclinical results, observed or expected hepatic transporter-mediated drug-drug interactions can be taken into account for clinical studies. In this paper, various methods including in vitro, in situ, in vivo, in silico approaches, and combinational concepts and several clinical studies on the assessment of transporter-mediated drug-drug interactions were reviewed. Informative and effective evaluation by preclinical tools together with the integration of pharmacokinetic modeling and simulation can reduce unexpected clinical outcomes and enhance the success rate in drug development.
Collapse
Affiliation(s)
- Nihan Izat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Selma Sahin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
13
|
Song I, Jeong H, Choi M, Kwon M, Shin Y, Kim JH, Lee H. Interactions between cyazofamid and human drug transporters. J Biochem Mol Toxicol 2020; 34:e22459. [DOI: 10.1002/jbt.22459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 11/17/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023]
Affiliation(s)
- Im‐Sook Song
- College of Pharmacy and Research Institute of Pharmaceutical SciencesKyungpook National UniversityDaegu Republic of Korea
| | - Hyeon‐Uk Jeong
- Drug Metabolism & Bioanalysis Laboratory, College of PharmacyThe Catholic University of KoreaBucheon Republic of Korea
| | - Min‐Koo Choi
- College of PharmacyDankook UniversityCheon‐an Republic of Korea
| | - Mihwa Kwon
- College of Pharmacy and Research Institute of Pharmaceutical SciencesKyungpook National UniversityDaegu Republic of Korea
| | - Yongho Shin
- Drug Metabolism & Bioanalysis Laboratory, College of PharmacyThe Catholic University of KoreaBucheon Republic of Korea
| | - Jeong Han Kim
- Pesticide Chemistry and Toxicology Laboratory, Department of Agricultural Biotechnology and Research Institute of Agriculture and Life SciencesSeoul National UniversitySeoul Republic of Korea
| | - Hye‐Suk Lee
- Drug Metabolism & Bioanalysis Laboratory, College of PharmacyThe Catholic University of KoreaBucheon Republic of Korea
| |
Collapse
|
14
|
Jeon JH, Lee S, Lee W, Jin S, Kwon M, Shin CH, Choi MK, Song IS. Herb-Drug Interaction of Red Ginseng Extract and Ginsenoside Rc with Valsartan in Rats. Molecules 2020; 25:E622. [PMID: 32023909 PMCID: PMC7037682 DOI: 10.3390/molecules25030622] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/14/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
The purpose of this study was to investigate the herb-drug interactions involving red ginseng extract (RGE) or ginsenoside Rc with valsartan, a substrate for organic anion transporting polypeptide (OATP/Oatp) transporters. In HEK293 cells overexpressing drug transporters, the protopanaxadiol (PPD)-type ginsenosides- Rb1, Rb2, Rc, Rd, Rg3, compound K, and Rh2-inhibited human OATP1B1 and OATP1B3 transporters (IC50 values of 7.99-68.2 µM for OATP1B1; 1.36-30.8 µM for OATP1B3), suggesting the herb-drug interaction of PPD-type ginsenosides involving OATPs. Protopanaxatriol (PPT)-type ginsenosides-Re, Rg1, and Rh1-did not inhibit OATP1B1 and OATP1B3 and all ginsenosides tested didn't inhibit OCT and OAT transporters. However, in rats, neither RGE nor Rc, a potent OATP inhibitor among PPD-type ginsenoside, changed in vivo pharmacokinetics of valsartan following repeated oral administration of RGE (1.5 g/kg/day for 7 days) or repeated intravenous injection of Rc (3 mg/kg for 5 days). The lack of in vivo herb-drug interaction between orally administered RGE and valsartan could be attributed to the low plasma concentration of PPD-type ginsenosides (5.3-48.4 nM). Even high plasma concentration of Rc did not effectively alter the pharmacokinetics of valsartan because of high protein binding and the limited liver distribution of Rc. The results, in conclusion, would provide useful information for herb-drug interaction between RGE or PPD-type ginsenosides and Oatp substrate drugs.
Collapse
Affiliation(s)
- Ji-Hyeon Jeon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (S.L.); (M.K.); (C.H.S.)
| | - Sowon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (S.L.); (M.K.); (C.H.S.)
| | - Wonpyo Lee
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea; (W.L.); (S.J.)
| | - Sojeong Jin
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea; (W.L.); (S.J.)
| | - Mihwa Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (S.L.); (M.K.); (C.H.S.)
| | - Chul Hwi Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (S.L.); (M.K.); (C.H.S.)
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea; (W.L.); (S.J.)
| | - Im-Sook Song
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (S.L.); (M.K.); (C.H.S.)
| |
Collapse
|
15
|
A Common mdr1 Gene Polymorphism is Associated With Changes in Linezolid Clearance. Ther Drug Monit 2018; 40:602-609. [DOI: 10.1097/ftd.0000000000000536] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Comprehensive Substrate Characterization of 22 Antituberculosis Drugs for Multiple Solute Carrier (SLC) Uptake Transporters In Vitro. Antimicrob Agents Chemother 2018; 62:AAC.00512-18. [PMID: 30012768 DOI: 10.1128/aac.00512-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/08/2018] [Indexed: 12/30/2022] Open
Abstract
The substrate potentials of antituberculosis drugs on solute carrier (SLC) transporters are not well characterized to date, despite a well-established understanding of their drug dispositions and pharmacokinetics. In this study, we investigated comprehensively the substrate potentials of the 22 currently available antituberculosis drugs for SLC family transporter-mediated uptake, using Xenopus laevis oocytes and stably transfected HEK-293 cells in vitro The result suggested that ethambutol, isoniazid, amoxicillin, and prothionamide act as novel substrates for the SLC transporters. In addition, in the presence of representative transporter inhibitors, the uptake of the antituberculosis drugs was markedly decreased compared with the uptake in the absence of inhibitor, suggesting involvement of the corresponding transporters. A cellular uptake study was performed, and the Km values of ethambutol were found to be 526.1 ± 15.6, 212.0 ± 20.1, 336.8 ± 20.1, and 455.0 ± 28 μM for organic cation transporter 1 (OCT1), OCT2, OCTN1, and OCTN2, respectively. Similarly, the Km of prothionamide was 805.8 ± 23.4 μM for OCT1, while the Km values of isoniazid and amoxicillin for organic anion transporter 3 (OAT3) were 233.7 ± 14.1 and 161.4 ± 10.6 μM, respectively. The estimated in vivo drug-drug interaction indexes from in vitro transporter inhibition kinetics for verapamil, probenecid, and ibuprofen against ethambutol, prothionamide, isoniazid, and amoxicillin were found to show potential for clinical drug interactions. In conclusion, this is the first study that demonstrated 22 antituberculosis drug interactions with transporters. This study will be helpful for mechanistic understanding of the disposition, drug-drug interactions, and pharmacokinetics of these antituberculosis drugs.
Collapse
|
17
|
Amor D, Goutal S, Marie S, Caillé F, Bauer M, Langer O, Auvity S, Tournier N. Impact of rifampicin-inhibitable transport on the liver distribution and tissue kinetics of erlotinib assessed with PET imaging in rats. EJNMMI Res 2018; 8:81. [PMID: 30116910 PMCID: PMC6095934 DOI: 10.1186/s13550-018-0434-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Background Erlotinib is an epidermal growth factor receptor (EGFR)-targeting tyrosine kinase inhibitor approved for treatment of non-small cell lung cancer. The wide inter-individual pharmacokinetic (PK) variability of erlotinib may impact treatment outcome and/or toxicity. Recent in vivo studies reported a nonlinear uptake transport of erlotinib into the liver, suggesting carrier-mediated system(s) to mediate its hepatobiliary clearance. Erlotinib has been identified in vitro as a substrate of organic anion-transporting polypeptide (OATP) transporters which expression does not restrict to hepatocytes and may impact the tissue uptake of erlotinib in vivo. Results The impact of rifampicin (40 mg/kg), a potent OATP inhibitor, on the liver uptake and exposure to tissues of 11C-erlotinib was investigated in rats (4 animals per group) using positron emission tomography (PET) imaging. Tissue pharmacokinetics (PK) and corresponding exposure (area under the curve, AUC) were assessed in the liver, kidney cortex, abdominal aorta (blood pool) and the lungs. The plasma PK of parent 11C-erlotinib was also measured using arterial blood sampling to estimate the transfer rate constant (kuptake) of 11C-erlotinib from plasma into different tissues. PET images unveiled the predominant distribution of 11C-erlotinib-associated radioactivity to the liver, which gradually moved to the intestine, thus highlighting hepatobiliary clearance. 11C-erlotinib also accumulated in the kidney cortex. Rifampicin did not impact AUCaorta but reduced kuptake, liver (p < 0.001), causing a significant 27.3% decrease in liver exposure (p < 0.001). Moreover, a significant decrease in kuptake, kidney with a concomitant decrease in AUCkidney (− 30.4%, p < 0.001) were observed. Rifampicin neither affected kuptake, lung nor AUClung. Conclusions Our results suggest that 11C-erlotinib is an in vivo substrate of rOATP transporters expressed in the liver and possibly of rifampicin-inhibitable transporter(s) in the kidneys. Decreased 11C-erlotinib uptake by elimination organs did not translate into changes in systemic exposure and exposure to the lungs, which are a target tissue for erlotinib therapy.
Collapse
Affiliation(s)
- Dorra Amor
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Sébastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Solène Marie
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France. .,CEA, DRF, JOLIOT, Service Hospitalier Frédéric Joliot, F-91401, Orsay, France.
| |
Collapse
|
18
|
The inhibitory effects of eighteen front-line antibiotics on the substrate uptake mediated by human Organic anion/cation transporters, Organic anion transporting polypeptides and Oligopeptide transporters in in vitro models. Eur J Pharm Sci 2018; 115:132-143. [DOI: 10.1016/j.ejps.2018.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/07/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022]
|
19
|
Evaluation of para-Aminosalicylic Acid as a Substrate of Multiple Solute Carrier Uptake Transporters and Possible Drug Interactions with Nonsteroidal Anti-inflammatory Drugs In Vitro. Antimicrob Agents Chemother 2017; 61:AAC.02392-16. [PMID: 28223391 DOI: 10.1128/aac.02392-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/15/2017] [Indexed: 11/20/2022] Open
Abstract
para-Aminosalicylic acid (PAS) is a second-line antituberculosis drug that has been used to treat multidrug-resistant and extensively drug-resistant tuberculosis for more than 60 years. Renal secretion and glomerular filtration are the major pathways for the elimination of PAS. We comprehensively studied PAS transport by using cell lines that overexpressed various transporters and found that PAS acts as a novel substrate of an organic anionic polypeptide (OATP1B1), organic cationic transporters (OCT1 and OCT2), and organic anion transporters (OAT1 and OAT3) but is not a substrate of any ATP-binding cassette (ABC) transporters. Net PAS uptake was measured, and the transport affinities (Km values) for OATP1B1, OCT1, OCT2, OAT1, and OAT3 were found to be 50.0, 20.3, 28.7, 78.1, and 100.1 μM, respectively. The net uptake rates suggested that renal OAT1 and OAT3 play relatively major roles in PAS elimination. The representative inhibitors rifampin for OATP1B1, probenecid for OAT1 and OAT3, and verapamil for OCT1 and OCT2 greatly inhibited PAS uptake, suggesting that PAS is dependent on multiple transporters for uptake. We also evaluated nonsteroidal anti-inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs), and metformin for the inhibition of PAS uptake via these transporters. Half-maximal (50%) inhibitory concentrations (IC50s) were kinetically determined and used to predict the drug-drug interactions (DDIs) affecting these transporters' activity toward PAS. We found that rifampin, probenecid, ibuprofen, naproxen, cimetidine, and quinidine each exhibited a significant potential for in vivo DDIs with PAS. In this study, PAS was found to be a novel substrate of several transporters, and drugs that inhibit these transporters can reduce PAS elimination.
Collapse
|
20
|
Parvez MM, Kaisar N, Shin HJ, Jung JA, Shin JG. Inhibitory Interaction Potential of 22 Antituberculosis Drugs on Organic Anion and Cation Transporters of the SLC22A Family. Antimicrob Agents Chemother 2016; 60:6558-6567. [PMID: 27550354 PMCID: PMC5075059 DOI: 10.1128/aac.01151-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/12/2016] [Indexed: 11/20/2022] Open
Abstract
Twenty-two currently marketed antituberculosis drugs were comprehensively evaluated for their inhibitory effect on organic anionic transporter (OAT)- and organic cation transporter (OCT)-mediated uptake using stably transfected HEK293 cells in vitro We observed moderate to strong inhibitory effects on OAT1- and OAT3-mediated para-aminohippurate (PAH) uptake and OCT1- and OCT2-mediated N-methyl-4-phenylpylidinium acetate (MPP+) uptake. Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 μM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 μM, respectively, for OAT3. Similarly, pyrazinamide, rifabutin, and levofloxacin were observed to have inhibitory effects, with IC50 values being 36.5, 42.7, and 30.3 μM, respectively, for OCT1 and with the IC50 value for PAS being 94.2 μM for OCT2. In addition, we used zidovudine and metformin as clinically prescribed substrates of OATs and OCTs, respectively, and zidovudine and metformin uptake was also strongly inhibited by the antituberculosis drugs. Among the tested drugs, the highest drug-drug interaction (DDI) indexes were found for PAS, which were 9.3 to 13.9 for OAT1 and 12.0 to 17.7 for OAT3, and linezolid, which were 1.18 to 2.15 for OAT1 and 1.7 to 3.01 for OAT3. Similarly, the DDI indexes of pyrazinamide and levofloxacin were 0.57 and 0.30, respectively, for OCT1, and the DDI index of PAS was 3.8 for OCT2, suggesting a stronger possibility (DDI index value cutoff, >0.1) of in vivo DDIs. This is the first comprehensive report of the inhibitory potential of anti-TB drugs on OAT- and OCT-mediated uptake of prototype and clinically prescribed substrate drugs in vitro, providing an ability to predict DDIs between anti-TB drugs and other coprescribed drugs in clinical studies in vivo.
Collapse
Affiliation(s)
- M Masud Parvez
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Nazia Kaisar
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Ho Jung Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jin Ah Jung
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| |
Collapse
|
21
|
Tajima M, Kato Y, Matsumoto J, Hirosawa I, Suzuki M, Takashio Y, Yamamoto M, Nishi Y, Yamada H. Linezolid-Induced Thrombocytopenia Is Caused by Suppression of Platelet Production via Phosphorylation of Myosin Light Chain 2. Biol Pharm Bull 2016; 39:1846-1851. [DOI: 10.1248/bpb.b16-00427] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masataka Tajima
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Yoshinori Kato
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Jun Matsumoto
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Iori Hirosawa
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Mariko Suzuki
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Yuki Takashio
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Mao Yamamoto
- Department of Pharmaceutical Sciences, International University of Health and Welfare
| | - Yoshifumi Nishi
- Graduate School of Pharmaceutical Sciences, International University of Health and Welfare
| | - Harumi Yamada
- Department of Pharmaceutical Sciences, International University of Health and Welfare
- Graduate School of Pharmaceutical Sciences, International University of Health and Welfare
| |
Collapse
|