1
|
Hwang S, Lee Y, Jang Y, Cho JY, Yoon S, Chung JY. Comprehensive Evaluation of OATP- and BCRP-Mediated Drug-Drug Interactions of Methotrexate Using Physiologically-Based Pharmacokinetic Modeling. Clin Pharmacol Ther 2024; 116:1013-1022. [PMID: 38860384 DOI: 10.1002/cpt.3329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/16/2024] [Indexed: 06/12/2024]
Abstract
Methotrexate (MTX) is an antifolate agent widely used for treating conditions such as rheumatoid arthritis and hematologic cancer. This study aimed to quantitatively interpret the drug-drug interactions (DDIs) of MTX mediated by drug transporters using physiologically-based pharmacokinetic (PBPK) modeling. An open-label, randomized, 4-treatment, 6-sequence, 4-period crossover study was conducted to investigate the effects of rifampicin (RFP), an inhibitor of organic anionic transporting peptides (OATP) 1B1/3, and febuxostat (FBX), an inhibitor of breast cancer resistance protein (BCRP), on the pharmacokinetics of MTX in healthy volunteers. PBPK models of MTX, RFP, and FBX were developed based on in vitro and in vivo data, and the performance of the simulation results for final PBPK models was validated in a clinical study. In the clinical study, when MTX was co-administered with RFP or FBX, systemic exposure of MTX increased by 33% and 17%, respectively, compared with that when MTX was administered alone. When MTX was co-administered with RFP and FBX, systemic exposure increased by 52% compared with that when MTX was administered alone. The final PBPK model showed a good prediction performance for the observed clinical data. The PBPK model of MTX was well developed in this study and can be used as a potential mechanistic model to predict and evaluate drug transporter-mediated DDIs of MTX with other drugs.
Collapse
Affiliation(s)
- Sejung Hwang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University Medical Research Center, Seoul, Korea
| | - Yujin Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
| | - Yeonseo Jang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University Medical Research Center, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seonghae Yoon
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae-Yong Chung
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Bundang Hospital, Seongnam, Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Korea
| |
Collapse
|
2
|
Fujita K, Isozumi N, Zhu Q, Matsubayashi M, Taniguchi T, Arakawa H, Shirasaka Y, Mori E, Tamai I. Unique Binding Sites of Uricosuric Agent Dotinurad for Selective Inhibition of Renal Uric Acid Reabsorptive Transporter URAT1. J Pharmacol Exp Ther 2024; 390:99-107. [PMID: 38670801 DOI: 10.1124/jpet.124.002096] [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: 01/03/2024] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Dotinurad was developed as a uricosuric agent, inhibiting urate (UA) reabsorption through the UA transporter URAT1 in the kidneys. Due to its high selectivity for URAT1 among renal UA transporters, we investigated the mechanism underlying this selectivity by identifying dotinurad binding sites specific to URAT1. Dotinurad was docked to URAT1 using AutoDock4, utilizing the AlphaFold2-predicted structure. The inhibitory effects of dotinurad on wild-type and mutated URAT1 at the predicted binding sites were assessed through URAT1-mediated [14C]UA uptake in Xenopus oocytes. Nine amino acid residues in URAT1 were identified as dotinurad-binding sites. Sequence alignment with UA-transporting organic anion transporters (OATs) revealed that H142 and R487 were unique to URAT1 among renal UA-transporting OATs. For H142, IC50 values of dotinurad increased to 62, 55, and 76 nM for mutated URAT1 (H142A, H142E, and H142R, respectively) compared with 19 nM for the wild type, indicating that H142 contributes to URAT1-selective interaction with dotinurad. H142 was predicted to interact with the phenyl-hydroxyl group of dotinurad. The IC50 of the hydroxyl group methylated dotinurad (F13141) was 165 μM, 8420-fold higher than dotinurad, suggesting the interaction of H142 and the phenyl-hydroxyl group by forming a hydrogen bond. Regarding R487, URAT1-R487A exhibited a loss of activity. Interestingly, the URAT1-H142A/R487A double mutant restored UA transport activity, with the IC50 value of dotinurad for the mutant (388 nM) significantly higher than that for H142A (73.5 nM). These results demonstrate that H142 and R487 of URAT1 determine its selectivity for dotinurad, a uniqueness observed only in URAT1 among UA-transporting OATs. SIGNIFICANCE STATEMENT: Dotinurad selectively inhibits the urate reabsorption transporter URAT1 in renal urate-transporting organic ion transporters (OATs). This study demonstrates that dotinurad interacts with H142 and R487 of URAT1, located in the extracellular domain and unique among OATs when aligning amino acid sequences. Mutations in these residues reduce affinity of dotinurad for URAT1, confirming their role in conferring selective inhibition. Additionally, the interaction between dotinurad and URAT1 involving H142 is found to mediate hydrogen bonding.
Collapse
Affiliation(s)
- Kazuki Fujita
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Noriyoshi Isozumi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Qiunan Zhu
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Masaya Matsubayashi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Tetsuya Taniguchi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Yoshiyuki Shirasaka
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Eiichiro Mori
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan (K.F., Q.Z., H.A., Y.S., I.T.); Department of Future Basic Medicine (N.I., E.M.) and V-iCliniX Laboratory (E.M.), Nara Medical University, Kashihara, Japan; and Research Laboratories 2, Fuji Yakuhin Co., Ltd., Nishi-Ward, Saitama, Japan (M.M., T.T.)
| |
Collapse
|
3
|
Strachowska M, Robaszkiewicz A. Characteristics of anticancer activity of CBP/p300 inhibitors - Features of their classes, intracellular targets and future perspectives of their application in cancer treatment. Pharmacol Ther 2024; 257:108636. [PMID: 38521246 DOI: 10.1016/j.pharmthera.2024.108636] [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: 11/02/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024]
Abstract
Due to the contribution of highly homologous acetyltransferases CBP and p300 to transcription elevation of oncogenes and other cancer promoting factors, these enzymes emerge as possible epigenetic targets of anticancer therapy. Extensive efforts in search for small molecule inhibitors led to development of compounds targeting histone acetyltransferase catalytic domain or chromatin-interacting bromodomain of CBP/p300, as well as dual BET and CBP/p300 inhibitors. The promising anticancer efficacy in in vitro and mice models led CCS1477 and NEO2734 to clinical trials. However, none of the described inhibitors is perfectly specific to CBP/p300 since they share similarity of a key functional domains with other enzymes, which are critically associated with cancer progression and their antagonists demonstrate remarkable clinical efficacy in cancer therapy. Therefore, we revise the possible and clinically relevant off-targets of CBP/p300 inhibitors that can be blocked simultaneously with CBP/p300 thereby improving the anticancer potential of CBP/p300 inhibitors and pharmacokinetic predicting data such as absorption, distribution, metabolism, excretion (ADME) and toxicity.
Collapse
Affiliation(s)
- Magdalena Strachowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland; University of Lodz, Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, Banacha 12 /16, 90-237 Lodz, Poland.
| | - Agnieszka Robaszkiewicz
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland; Johns Hopkins University School of Medicine, Institute of Fundamental and Basic Research, 600 5(th) Street South, Saint Petersburg FL33701, United States of America.
| |
Collapse
|
4
|
Lehtisalo M, Tarkiainen EK, Neuvonen M, Holmberg M, Kiiski JI, Lapatto-Reiniluoto O, Filppula AM, Kurkela M, Backman JT, Niemi M. Ticagrelor Increases Exposure to the Breast Cancer Resistance Protein Substrate Rosuvastatin. Clin Pharmacol Ther 2024; 115:71-79. [PMID: 37786998 DOI: 10.1002/cpt.3067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ticagrelor and rosuvastatin are often used concomitantly after atherothrombotic events. Several cases of rhabdomyolysis during concomitant ticagrelor and rosuvastatin have been reported, suggesting a drug-drug interaction. We showed recently that ticagrelor inhibits breast cancer resistance protein (BCRP) and organic anion transporting polypeptide (OATP) 1B1, 1B3, and 2B1-mediated rosuvastatin transport in vitro. The aim of this study was to investigate the effects of ticagrelor on rosuvastatin pharmacokinetics in humans. In a randomized, crossover study, 9 healthy volunteers ingested a single dose of 90 mg ticagrelor or placebo, followed by a single 10 mg dose of rosuvastatin 1 hour later. Ticagrelor 90 mg or placebo were additionally administered 12, 24, and 36 hours after their first dose. Ticagrelor increased rosuvastatin area under the plasma concentration-time curve (AUC) and peak plasma concentration 2.6-fold (90% confidence intervals: 1.8-3.8 and 1.7-4.0, P = 0.001 and P = 0.003), and prolonged its half-life from 3.1 to 6.6 hours (P = 0.009). Ticagrelor also decreased the renal clearance of rosuvastatin by 11% (3%-19%, P = 0.032). The N-desmethylrosuvastatin:rosuvastatin AUC0-10h ratio remained unaffected by ticagrelor. Ticagrelor had no effect on the plasma concentrations of the endogenous OATP1B substrates glycodeoxycholate 3-O-glucuronide, glycochenodeoxycholate 3-O-glucuronide, glycodeoxycholate 3-O-sulfate, and glycochenodeoxycholate 3-O-sulfate, or the sodium-taurocholate cotransporting polypeptide substrate taurocholic acid. These data indicate that ticagrelor increases rosuvastatin concentrations more than twofold in humans, probably mainly by inhibiting intestinal BCRP. Because the risk for rosuvastatin-induced myotoxicity increases along with rosuvastatin plasma concentrations, using ticagrelor concomitantly with high doses of rosuvastatin should be avoided.
Collapse
Affiliation(s)
- Minna Lehtisalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Mikko Holmberg
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | - Johanna I Kiiski
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Outi Lapatto-Reiniluoto
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Mika Kurkela
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
5
|
Kraev KI, Geneva-Popova MG, Hristov BK, Uchikov PA, Popova-Belova SD, Kraeva MI, Basheva-Kraeva YM, Stoyanova NS, Mitkova-Hristova VT. Celebrating Versatility: Febuxostat's Multifaceted Therapeutic Application. Life (Basel) 2023; 13:2199. [PMID: 38004339 PMCID: PMC10672185 DOI: 10.3390/life13112199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Febuxostat, initially developed as a xanthine oxidase inhibitor to address hyperuricemia in gout patients, has evolved into a versatile therapeutic agent with multifaceted applications. This review provides a comprehensive overview of febuxostat's mechanism of action, its effectiveness in gout management, its cardiovascular safety profile, renal and hepatic effects, musculoskeletal applications, safety considerations, and emerging research prospects. Febuxostat's primary mechanism involves selective inhibition of xanthine oxidase, resulting in reduced uric acid production. Its pharmacokinetics require personalized dosing strategies based on individual characteristics. In gout management, febuxostat offers a compelling alternative, effectively lowering uric acid levels, relieving symptoms, and supporting long-term control, especially for patients intolerant to allopurinol. Recent studies have demonstrated its cardiovascular safety, and it exhibits minimal hepatotoxicity, making it suitable for those with liver comorbidities. Febuxostat's potential nephroprotective effects and kidney stone prevention properties are noteworthy, particularly for gout patients with renal concerns. Beyond gout, its anti-inflammatory properties hint at applications in musculoskeletal conditions and a broader spectrum of clinical contexts, including metabolic syndrome. Emerging research explores febuxostat's roles in cardiovascular health, neurological disorders, rheumatoid arthritis, and cancer therapy, driven by its anti-inflammatory and antioxidative properties. Future directions include personalized medicine, combination therapies, mechanistic insights, and ongoing long-term safety monitoring, collectively illuminating the promising landscape of febuxostat's multifaceted therapeutic potential.
Collapse
Affiliation(s)
- Krasimir Iliev Kraev
- Department of Propedeutics of Internal Diseases, Medical Faculty, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | | | - Bozhidar Krasimirov Hristov
- Second Department of Internal Diseases, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | - Petar Angelov Uchikov
- Department of Special Surgery, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | | | - Maria Ilieva Kraeva
- Department of Otorhynolaryngology, Medical Faculty, Medical University of Plovdiv, 6000 Plovdiv, Bulgaria
| | - Yordanka Mincheva Basheva-Kraeva
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| | - Nina Staneva Stoyanova
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| | - Vesela Todorova Mitkova-Hristova
- Department of Ophthalmology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- University Eye Clinic, University Hospital, 4000 Plovdiv, Bulgaria
| |
Collapse
|
6
|
Lehtisalo M, Kiander W, Filppula AM, Deng F, Kidron H, Korhonen M, Sinkko J, Koivula K, Niemi M. Rhabdomyolysis during concomitant ticagrelor and rosuvastatin: A breast cancer resistance protein-mediated drug interaction? Br J Clin Pharmacol 2023; 89:2309-2315. [PMID: 36740817 DOI: 10.1111/bcp.15684] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023] Open
Abstract
We present 3 patients diagnosed with rhabdomyolysis 1-6 months after the initiation of concomitant rosuvastatin and ticagrelor medication. A literature review and Food and Drug Administration adverse event reporting system revealed >40 reports of rhabdomyolysis during concomitant ticagrelor and rosuvastatin, including 3 with a fatal outcome. We show that ticagrelor inhibits breast cancer resistance protein-, organic anion transporting polypeptide (OATP) 1B1-, 1B3- and 2B1-mediated transport of rosuvastatin in vitro with half-maximal unbound inhibitory concentrations of 0.36, 4.13, 7.5 and 3.26 μM, respectively. A static drug interaction model predicted that ticagrelor may inhibit intestinal breast cancer resistance protein and thus increase rosuvastatin plasma exposure 2.1-fold, whereas the OATP-mediated hepatic uptake of rosuvastatin should not be inhibited due to relatively low portal ticagrelor concentrations. Taken together, concomitant use of ticagrelor with rosuvastatin may increase the systemic exposure to rosuvastatin and the risk of rosuvastatin-induced rhabdomyolysis. Further studies are warranted to investigate the potential pharmacokinetic interaction between ticagrelor and rosuvastatin in humans.
Collapse
Affiliation(s)
- Minna Lehtisalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anne M Filppula
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Feng Deng
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mari Korhonen
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | | | - Kimmo Koivula
- South Karelia Central Hospital, Lappeenranta, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
7
|
Deng F, Sjöstedt N, Santo M, Neuvonen M, Niemi M, Kidron H. Novel inhibitors of breast cancer resistance protein (BCRP, ABCG2) among marketed drugs. Eur J Pharm Sci 2023; 181:106362. [PMID: 36529162 DOI: 10.1016/j.ejps.2022.106362] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Drug-drug interactions (DDIs) are a major concern for the safe use of medications. Breast cancer resistance protein (BCRP) is a clinically relevant ATP-binding cassette (ABC) transporter for drug disposition. Inhibition of BCRP increases the plasma concentrations of BCRP substrate drugs, which potentially could lead to adverse drug reactions. The aim of the present study was to identify BCRP inhibitors amongst a library of 232 commonly used drugs and anticancer drugs approved by the United States Food and Drug Administration (FDA). BCRP inhibition studies were carried out using the vesicular transport assay. We found 75 drugs that reduced the relative transport activity of BCRP to less than 25% of the vehicle control and were categorized as strong inhibitors. The concentration required for 50% inhibition (IC50) was determined for 13 strong inhibitors that were previously poorly characterized for BCRP inhibition. The IC50 ranged from 1.1 to 11 µM, with vemurafenib, dabigatran etexilate and everolimus being the strongest inhibitors. According to the drug interaction guidance documents from the FDA and the European Medicines Agency (EMA), in vivo DDI studies are warranted if the theoretical intestinal luminal concentration of a drug exceeds its IC50 by tenfold. Here, the IC50 values for eight of the drugs were 100-fold lower than their theoretical intestinal luminal concentration. Moreover, a mechanistic static model suggested that vemurafenib, bexarotene, dabigatran etexilate, rifapentine, aprepitant, and ivacaftor could almost fully inhibit intestinal BCRP, increasing the exposure of concomitantly administered rosuvastatin over 90%. Therefore, clinical studies are warranted to investigate whether these drugs cause BCRP-mediated DDIs in humans.
Collapse
Affiliation(s)
- Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Noora Sjöstedt
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mariangela Santo
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Tukholmankatu 8 C, P.O. Box 20, 00014, Finland; Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. Haartmaninkatu 8, P.O. Box 63, 00014, Finland; Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Heidi Kidron
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland. Viikinkaari 5 E, P.O. Box 56, 00014, Finland.
| |
Collapse
|
8
|
Lehtisalo M, Taskinen S, Tarkiainen EK, Neuvonen M, Viinamäki J, Paile-Hyvärinen M, Lilius TO, Tapaninen T, Backman JT, Tornio A, Niemi M. A comprehensive pharmacogenomic study indicates roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Br J Clin Pharmacol 2023; 89:242-252. [PMID: 35942816 PMCID: PMC10087178 DOI: 10.1111/bcp.15485] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
AIMS The aim was to comprehensively investigate the effects of genetic variability on the pharmacokinetics of rosuvastatin. METHODS We conducted a genome-wide association study and candidate gene analyses of single dose rosuvastatin pharmacokinetics in a prospective study (n = 159) and a cohort of previously published studies (n = 88). RESULTS In a genome-wide association meta-analysis of the prospective study and the cohort of previously published studies, the SLCO1B1 c.521 T > C (rs4149056) single nucleotide variation (SNV) associated with increased area under the plasma concentration-time curve (AUC) and peak plasma concentration of rosuvastatin (P = 1.8 × 10-12 and P = 3.2 × 10-15 ). The candidate gene analysis suggested that the ABCG2 c.421C > A (rs2231142) SNV associates with increased rosuvastatin AUC (P = .0079), while the SLCO1B1 c.388A > G (rs2306283) and SLCO2B1 c.1457C > T (rs2306168) SNVs associate with decreased rosuvastatin AUC (P = .0041 and P = .0076). Based on SLCO1B1 genotypes, we stratified the participants into poor, decreased, normal, increased and highly increased organic anion transporting polypeptide (OATP) 1B1 function groups. The OATP1B1 poor function phenotype associated with 2.1-fold (90% confidence interval 1.6-2.8, P = 4.69 × 10-5 ) increased AUC of rosuvastatin, whereas the OATP1B1 highly increased function phenotype associated with a 44% (16-62%; P = .019) decreased rosuvastatin AUC. The ABCG2 c.421A/A genotype associated with 2.2-fold (1.5-3.0; P = 2.6 × 10-4 ) increased AUC of rosuvastatin. The SLCO2B1 c.1457C/T genotype associated with 28% decreased rosuvastatin AUC (11-42%; P = .01). CONCLUSION These data suggest roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Poor SLCO1B1 or ABCG2 function genotypes may increase the risk of rosuvastatin-induced myotoxicity. Reduced doses of rosuvastatin are advisable for patients with these genotypes.
Collapse
Affiliation(s)
- Minna Lehtisalo
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Suvi Taskinen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Jenni Viinamäki
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Maria Paile-Hyvärinen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas O Lilius
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tuija Tapaninen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Janne T Backman
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Aleksi Tornio
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.,Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
9
|
Iversen DB, Andersen NE, Dalgård Dunvald A, Pottegård A, Stage TB. Drug metabolism and drug transport of the 100 most prescribed oral drugs. Basic Clin Pharmacol Toxicol 2022; 131:311-324. [PMID: 35972991 PMCID: PMC9804310 DOI: 10.1111/bcpt.13780] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 01/05/2023]
Abstract
Safe and effective use of drugs requires an understanding of metabolism and transport. We identified the 100 most prescribed drugs in six countries and conducted a literature search on in vitro data to assess contribution of Phase I and II enzymes and drug transporters to metabolism and transport. Eighty-nine of the 100 drugs undergo drug metabolism or are known substrates for drug transporters. Phase I enzymes are involved in metabolism of 67 drugs, while Phase II enzymes mediate metabolism of 18 drugs. CYP3A4/5 is the most important Phase I enzyme involved in metabolism of 43 drugs followed by CYP2D6 (23 drugs), CYP2C9 (23 drugs), CYP2C19 (22 drugs), CYP1A2 (14 drugs) and CYP2C8 (11 drugs). More than half of the drugs (54 drugs) are known substrates for drug transporters. P-glycoprotein (P-gp) is known to be involved in transport of 30 drugs, while breast cancer resistance protein (BCRP) facilitates transport of 11 drugs. A considerable proportion of drugs are subject to a combination of Phase I metabolism, Phase II metabolism and/or drug transport. We conclude that the majority of the most frequently prescribed drugs depend on drug metabolism or drug transport. Thus, understanding variability of drug metabolism and transport remains a priority.
Collapse
Affiliation(s)
- Ditte B. Iversen
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Nanna Elman Andersen
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Ann‐Cathrine Dalgård Dunvald
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Anton Pottegård
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Tore B. Stage
- Clinical Pharmacology, Pharmacy and Environmental Medicine, Department of Public HealthUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
10
|
Hashimoto Y, Michiba K, Maeda K, Kusuhara H. Quantitative prediction of pharmacokinetic properties of drugs in humans: Recent advance in in vitro models to predict the impact of efflux transporters in the small intestine and blood-brain barrier. J Pharmacol Sci 2021; 148:142-151. [PMID: 34924119 DOI: 10.1016/j.jphs.2021.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Efflux transport systems are essential to suppress the absorption of xenobiotics from the intestinal lumen and protect the critical tissues at the blood-tissue barriers, such as the blood-brain barrier. The function of drug efflux transport is dominated by various transporters. Accumulated clinical evidences have revealed that genetic variations of the transporters, together with coadministered drugs, affect the expression and/or function of transporters and subsequently the pharmacokinetics of substrate drugs. Thus, in the preclinical stage of drug development, quantitative prediction of the impact of efflux transporters as well as that of uptake transporters and metabolic enzymes on the pharmacokinetics of drugs in humans has been performed using various in vitro experimental tools. Various kinds of human-derived cell systems can be applied to the precise prediction of drug transport in humans. Mathematical modeling consisting of each intrinsic metabolic or transport process enables us to understand the disposition of drugs both at the organ level and at the level of the whole body by integrating a variety of experimental results into model parameters. This review focuses on the role of efflux transporters in the intestinal absorption and brain distribution of drugs, in addition to recent advances in predictive tools and methodologies.
Collapse
Affiliation(s)
- Yoshiki Hashimoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuya Maeda
- Laboratory of Pharmaceutics, Kitasato University School of Pharmacy, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, 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
|
11
|
Mitsuboshi S, Niimura T, Kanda M, Ishida S, Zamami Y, Ishizawa K. Risk of Hematologic Events With Coadministration of Methotrexate and the Breast Cancer Resistance Protein Inhibitor Febuxostat. Ann Pharmacother 2021; 56:910-915. [PMID: 34726078 DOI: 10.1177/10600280211055794] [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] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The breast cancer resistance protein (BCRP) is a key drug transporter found in the liver, kidney, central nervous system, and gastrointestinal tract. Due to the wide expression of BCRP, interactions of other drugs with methotrexate (MTX) may differ in oral and intravenous MTX users, and understanding of these interactions may be useful in preventing severe adverse events. Febuxostat, a urate-lowering drug, inhibits BCRP. OBJECTIVE The objective of this study was to clarify the differences in the drug-drug interaction profiles of oral and intravenous methotrexate, associated with BCRP. METHODS We analyzed the Japanese Adverse Drug Event Report database and compared the frequency of hematologic events in patients taking oral and intravenous MTX, with or without the concomitant use of febuxostat or allopurinol. Hematologic events were defined as pancytopenia and neutropenia. Multiple logistic regression analysis was then used to identify the risk factors for hematologic events in oral and intravenous MTX users. RESULTS We identified 8 453 oral and 810 intravenous MTX users with 546 and 126 cases of hematologic events, respectively. Compared with those not using febuxostat, a disproportionate number of hematologic events was observed in intravenous MTX users concomitantly using febuxostat (P < 0.01). The multivariate logistic analysis of intravenous MTX users showed that hematologic events were significantly associated with febuxostat use (P < 0.01) and age ≥ 60 years (P < 0.01). CONCLUSION AND RELEVANCE Our findings suggest that patients being treated with intravenous MTX who concomitantly use febuxostat may be at an increased risk of hematologic events, presumably due to BCRP-mediated drug-drug interaction.
Collapse
Affiliation(s)
| | - Takahiro Niimura
- Department of Clinical Pharmacology & Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masaya Kanda
- Department of Clinical Pharmacology & Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Shunsuke Ishida
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yoshito Zamami
- Department of Clinical Pharmacology & Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology & Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
12
|
Agustina R, Masuo Y, Kido Y, Shinoda K, Ishimoto T, Kato Y. Identification of Food-Derived Isoflavone Sulfates as Inhibition Markers for Intestinal Breast Cancer Resistance Proteins. Drug Metab Dispos 2021; 49:972-984. [PMID: 34413161 DOI: 10.1124/dmd.121.000534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022] Open
Abstract
Potential inhibition of the breast cancer resistance protein (BCRP), a drug efflux transporter, is a key issue during drug development, and the use of its physiologic substrates as biomarkers can be advantageous to assess inhibition. In this study, we aimed to identify BCRP substrates by an untargeted metabolomic approach. Mice were orally administered lapatinib to inhibit BCRP in vivo, and plasma samples were assessed by liquid chromatography/time of flight/mass spectrometry with all-ion fragmentation acquisition and quantified by liquid chromatography with tandem mass spectrometry. A differential metabolomic analysis was also performed for plasma from Bcrp -/- and wild-type mice. Plasma peaks of food-derived isoflavone metabolites, daidzein sulfate (DS), and genistein sulfate (GS) increased after lapatinib administration and in Bcrp -/- mice. Administration of lapatinib and another BCRP inhibitor febuxostat increased the area under the plasma concentration-time curve (AUC) of DS, GS, and equol sulfate (ES) by 3.6- and 1.8-, 5.6- and 4.1-, and 1.6- and 4.8-fold, respectively. BCRP inhibitors also increased the AUC and maximum plasma concentration of DS and ES after coadministration with each parent compound. After adding parent compounds to the apical side of induced pluripotent stem cell-derived small intestinal epithelial-like cells, DS, GS, and ES in the basal compartment significantly increased in the presence of lapatinib and febuxostat, suggesting the inhibition of intestinal BCRP. ATP-dependent uptake of DS and ES in BCRP-expressing membrane vesicles was reduced by both inhibitors, indicating inhibition of BCRP-mediated DS and ES transport. Thus, we propose the first evidence of surrogate markers for BCRP inhibition. SIGNIFICANCE STATEMENT: This study performed untargeted metabolomics to identify substrates of BCRP/ABCG2 to assess changes in its transport activity in vivo by BCRP/ABCG2 inhibitors. Food-derived isoflavone sulfates were identified as useful markers for evaluating changes in BCRP-mediated transport in the small intestine by its inhibitors.
Collapse
Affiliation(s)
- Rina Agustina
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| | - Yasuto Kido
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| | - Kyosuke Shinoda
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (R.A., Y.M., K.S., T.I., Y.Ka.); Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia (R.A.); and Laboratory for Drug Discovery and Development, Shionogi & Co., Ltd., Toyonaka, Osaka, Japan (Y.Ki.)
| |
Collapse
|
13
|
Arakawa H, Amezawa N, Kawakatsu Y, Tamai I. Renal Reabsorptive Transport of Uric Acid Precursor Xanthine by URAT1 and GLUT9. Biol Pharm Bull 2021; 43:1792-1798. [PMID: 33132325 DOI: 10.1248/bpb.b20-00597] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Xanthine and hypoxanthine are intermediate metabolites of uric acid and a source of reactive oxidative species (ROS) by xanthine oxidoreductase (XOR), suggesting that facilitating their elimination is beneficial. Since they are reabsorbed in renal proximal tubules, we investigated their reabsorption mechanism by focusing on the renal uric acid transporters URAT1 and GLUT9, and examined the effect of clinically used URAT1 inhibitor on their renal clearance when their plasma concentration is increased by XOR inhibitor. Uptake study for [3H]xanthine and [3H]hypoxanthine was performed using URAT1- and GLUT9-expressing Xenopus oocytes. Transcellular transport study for [3H]xanthine was carried out using Madin-Darby canine kidney (MDCK)II cells co-expressing URAT1 and GLUT9. In in vivo pharmacokinetic study, renal clearance of xanthine was estimated based on plasma concentration and urinary recovery. Uptake by URAT1- and GLUT9-expressing oocytes demonstrated that xanthine is a substrate of URAT1 and GLUT9, while hypoxanthine is not. Transcellular transport of xanthine in MDCKII cells co-expressing URAT1 and GLUT9 was significantly higher than those in mock cells and cells expressing URAT1 or GLUT9 alone. Furthermore, dotinurad, a URAT1 inhibitor, increased renal clearance of xanthine in rats treated with topiroxostat to inhibit XOR. It was suggested that xanthine is reabsorbed in the same manner as uric acid through URAT1 and GLUT9, while hypoxanthine is not. Accordingly, it is expected that treatment with XOR and URAT1 inhibitors will effectively decrease purine pools in the body and prevent cell injury due to ROS generated during XOR-mediated reactions.
Collapse
Affiliation(s)
- Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Natsumi Amezawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yu Kawakatsu
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
14
|
Deng F, Tuomi SK, Neuvonen M, Hirvensalo P, Kulju S, Wenzel C, Oswald S, Filppula AM, Niemi M. Comparative Hepatic and Intestinal Efflux Transport of Statins. Drug Metab Dispos 2021; 49:750-759. [PMID: 34162690 DOI: 10.1124/dmd.121.000430] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
Previous studies have shown that lipid-lowering statins are transported by various ATP-binding cassette (ABC) transporters. However, because of varying methods, it is difficult to compare the transport profiles of statins. Therefore, we investigated the transport of 10 statins or statin metabolites by six ABC transporters using human embryonic kidney cell-derived membrane vesicles. The transporter protein expression levels in the vesicles were quantified with liquid chromatography-tandem mass spectrometry and used to scale the measured clearances to tissue levels. In our study, apically expressed breast cancer resistance protein (BCRP) and P-glycoprotein (P-gp) transported atorvastatin, fluvastatin, pitavastatin, and rosuvastatin. Multidrug resistance-associated protein 3 (MRP3) transported atorvastatin, fluvastatin, pitavastatin, and, to a smaller extent, pravastatin. MRP4 transported fluvastatin and rosuvastatin. The scaled clearances suggest that BCRP contributes to 87%-91% and 84% of the total active efflux of rosuvastatin in the small intestine and the liver, respectively. For atorvastatin, the corresponding values for P-gp-mediated efflux were 43%-79% and 66%, respectively. MRP3, on the other hand, may contribute to 23%-26% and 25%-37% of total active efflux of atorvastatin, fluvastatin, and pitavastatin in jejunal enterocytes and liver hepatocytes, respectively. These data indicate that BCRP may play an important role in limiting the intestinal absorption and facilitating the biliary excretion of rosuvastatin and that P-gp may restrict the intestinal absorption and mediate the biliary excretion of atorvastatin. Moreover, the basolateral MRP3 may enhance the intestinal absorption and sinusoidal hepatic efflux of several statins. Taken together, the data show that statins differ considerably in their efflux transport profiles. SIGNIFICANCE STATEMENT: This study characterized and compared the transport of atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin acid and four atorvastatin metabolites by six ABC transporters (BCRP, MRP2, MRP3, MRP4, MRP8, P-gp). Based on in vitro findings and protein abundance data, the study concludes that BCRP, MRP3, and P-gp have a major impact in the efflux of various statins. Together with in vitro metabolism, uptake transport, and clinical data, our findings are applicable for use in comparative systems pharmacology modeling of statins.
Collapse
Affiliation(s)
- Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Suvi-Kukka Tuomi
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Neuvonen
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Päivi Hirvensalo
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Sami Kulju
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Christoph Wenzel
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Stefan Oswald
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Anne M Filppula
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Niemi
- Department of Clinical Pharmacology, Faculty of Medicine (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.), and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (F.D., S.-K.T., M.Ne, P.H., S.K., A.M.F., M.Ni.); Institute of Pharmacology, Center of Drug Absorption and Transport, University Medicine Greifswald, Greifswald, Germany (C.W., S.O.); Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany (S.O.); and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| |
Collapse
|
15
|
Filppula AM, Hirvensalo P, Parviainen H, Ivaska VE, Lönnberg KI, Deng F, Viinamäki J, Kurkela M, Neuvonen M, Niemi M. Comparative Hepatic and Intestinal Metabolism and Pharmacodynamics of Statins. Drug Metab Dispos 2021; 49:658-667. [PMID: 34045219 DOI: 10.1124/dmd.121.000406] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
This study aimed to comprehensively investigate the in vitro metabolism of statins. The metabolism of clinically relevant concentrations of atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, simvastatin, and their metabolites were investigated using human liver microsomes (HLMs), human intestine microsomes (HIMs), liver cytosol, and recombinant cytochrome P450 enzymes. We also determined the inhibitory effects of statin acids on their pharmacological target, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase. In HLMs, statin lactones were metabolized to a much higher extent than their acid forms. Atorvastatin lactone and simvastatin (lactone) showed extensive metabolism [intrinsic clearance (CLint) values of 3700 and 7400 µl/min per milligram], whereas the metabolism of the lactones of 2-hydroxyatorvastatin, 4-hydroxyatorvastatin, and pitavastatin was slower (CLint 20-840 µl/min per milligram). The acids had CLint values in the range <0.1-80 µl/min per milligram. In HIMs, only atorvastatin lactone and simvastatin (lactone) exhibited notable metabolism, with CLint values corresponding to 20% of those observed in HLMs. CYP3A4/5 and CYP2C9 were the main statin-metabolizing enzymes. The majority of the acids inhibited HMG-CoA reductase, with 50% inhibitory concentrations of 4-20 nM. The present comparison of the metabolism and pharmacodynamics of the various statins using identical methods provides a strong basis for further application, e.g., comparative systems pharmacology modeling. SIGNIFICANCE STATEMENT: The present comparison of the in vitro metabolic and pharmacodynamic properties of atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin and their metabolites using unified methodology provides a strong basis for further application. Together with in vitro drug transporter and clinical data, the present findings are applicable for use in comparative systems pharmacology modeling to predict the pharmacokinetics and pharmacological effects of statins at different dosages.
Collapse
Affiliation(s)
- Anne M Filppula
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Päivi Hirvensalo
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Heli Parviainen
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Vilma E Ivaska
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - K Ivar Lönnberg
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Feng Deng
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Jenni Viinamäki
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mika Kurkela
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Neuvonen
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| | - Mikko Niemi
- Department of Clinical Pharmacology and Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland (A.M.F., P.H., H.P., V.E.I., K.I.L., F.D., J.V., M.K., M.Ne., M.Ni.) and Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland (M.Ni.)
| |
Collapse
|
16
|
Taniguchi T, Omura K, Motoki K, Sakai M, Chikamatsu N, Ashizawa N, Takada T, Iwanaga T. Hypouricemic agents reduce indoxyl sulfate excretion by inhibiting the renal transporters OAT1/3 and ABCG2. Sci Rep 2021; 11:7232. [PMID: 33790363 PMCID: PMC8012596 DOI: 10.1038/s41598-021-86662-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/18/2021] [Indexed: 11/14/2022] Open
Abstract
Indoxyl sulfate (IS) accumulates in the body in chronic kidney disease (CKD). In the renal proximal tubules, IS excretion is mediated by OAT1/3 and ABCG2. These transporters are inhibited by some hypouricemic agents; OATs by probenecid and benzbromarone, ABCG2 by febuxostat and benzbromarone. Thus, we evaluated whether hypouricemic agents including dotinurad, a novel selective urate reabsorption inhibitor with minimal effect on OATs or ABCG2, affect IS clearance in rats. Intact and adenine-induced acute renal failure rats were orally administered hypouricemic agents, and both endogenous IS and exogenously administered stable isotope-labeled d4-IS in the plasma and kidney were measured. Our results demonstrated that OATs inhibitors, such as probenecid, suppress IS uptake into the kidney, leading to increased plasma IS concentration, whereas ABCG2 inhibitors, such as febuxostat, cause renal IS accumulation remarkably by suppressing its excretion in intact rats. The effects of these agents were reduced in adenine-induced acute renal failure rats, presumably due to substantial decrease in renal OAT1/3 and ABCG2 expression. Dotinurad did not significantly affected the clearance of IS under both conditions. Therefore, we suggest that hypouricemic agents that do not affect OATs and ABCG2 are effective therapeutic options for the treatment of hyperuricemia complicated by CKD.
Collapse
Affiliation(s)
- Tetsuya Taniguchi
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan.
| | - Koichi Omura
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| | - Keisuke Motoki
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| | - Miku Sakai
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| | - Noriko Chikamatsu
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| | - Naoki Ashizawa
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Iwanaga
- Research Laboratories 2, Fuji Yakuhin Co., Ltd., 636-1, Iida-Shinden, Nishi-ku, Saitama, Japan
| |
Collapse
|
17
|
Asad M, Asdaq SMB, Mohzari Y, Alrashed A, Alajami HN, Aljohani AO, Mushtawi AAA, Alajmi AN, Alajmi HN, Imran M, Orfali R. Pharmacokinetic and pharmacodynamic interaction of Rosuvastatin calcium with guggulipid extract in rats. Saudi J Biol Sci 2021; 28:3490-3496. [PMID: 34121889 PMCID: PMC8176130 DOI: 10.1016/j.sjbs.2021.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/17/2023] Open
Abstract
Background & objectives Rosuvastatin calcium (RC) is a potent and competitive synthetic inhibitor of HMG-CoA reductase used for the treatment of dyslipidemia. Guggulipid obtained from Commiphora mukul is used in the treatment of a wide variety of diseases such as atherosclerosis, hypercholesterolemia, rheumatism, and obesity. The present study evaluates the pharmacokinetic and pharmacodynamic interactions between RC and the standardized guggulipid extract in rats. Materials and methods The guggulipid extract was standardized for the presence of guggulsterones. The pharmacokinetic interaction was determined after a single dose administration of RC alone or in combination with the guggulipid extract or after multiple-dose administration of RC alone or RC along with the guggulipid extract for 14 days. To determine the pharmacodynamic interaction, RC and guggulipid extract were administered to hyperlipidemic rats for 14 days. The level of significance was determined using unpaired student’s t-test, one way ANOVA, the post-ANOVA Tukey test. Results Standardization of guggulipid extract showed it contains 7.5%w/w of guggulsterones. Guggulipid extract increased the bioavailability of RC in both single-dose and multiple-dose studies. Guggulipid extract reduced the rate of absorption (Ka) of RC but showed an increase in maximum serum concentration (Cmax). An in-vitro study using isolated rat intestine revealed that guggulipid extract decreased the rate of absorption of RC in the intestinal lumen. The hypolipidemic activity of RC was augmented by the guggulipid extract in hyperlipidemic rats. Interpretation & conclusion Therefore it is concluded that guggulipid extract increases the bioavailability of RC by delaying its Ka and augments its hypolipidemic action. However, it is recommended that a combination of RC with guggulipid extract should be used only after an adverse effect(s) of this combination are determined.
Collapse
Affiliation(s)
- Mohammed Asad
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11911, Saudi Arabia
| | | | - Yahya Mohzari
- Clinical Pharmacy Department, King Saud Medical City, Riyadh 12746, Saudi Arabia
| | - Ahmed Alrashed
- Pharmaceutical Services Administration, Inpatient Department, Main Hospital, KFMC, Riyadh 11564, Saudi Arabia
| | - Hamdan Najib Alajami
- Pharmaceutical Services Administration, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia
| | - Awad Othman Aljohani
- Pharmaceutical Services Administration, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah Ali Al Mushtawi
- Pharmaceutical Services Administration, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia
| | - Assil Najib Alajmi
- Pharmaceutical Services Administration, Health Oasis Hospital, Riyadh, Saudi Arabia
| | - Hanan Nageeb Alajmi
- Pharmaceutical Services Administration, Health Oasis Hospital, Riyadh, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, P.O. BOX 840, Rafha 91911, Saudi Arabia
| | - Raha Orfali
- Department of Pharmacognosy, Collage of Pharmacy, King Saud university, P.O.Box 22452, Riyadh 11495, Saudi Arabia
| |
Collapse
|
18
|
Identification of Two Dysfunctional Variants in the ABCG2 Urate Transporter Associated with Pediatric-Onset of Familial Hyperuricemia and Early-Onset Gout. Int J Mol Sci 2021; 22:ijms22041935. [PMID: 33669292 PMCID: PMC7920026 DOI: 10.3390/ijms22041935] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
The ABCG2 gene is a well-established hyperuricemia/gout risk locus encoding a urate transporter that plays a crucial role in renal and intestinal urate excretion. Hitherto, p.Q141K—a common variant of ABCG2 exhibiting approximately one half the cellular function compared to the wild-type—has been reportedly associated with early-onset gout in some populations. However, compared with adult-onset gout, little clinical information is available regarding the association of other uricemia-associated genetic variations with early-onset gout; the latent involvement of ABCG2 in the development of this disease requires further evidence. We describe a representative case of familial pediatric-onset hyperuricemia and early-onset gout associated with a dysfunctional ABCG2, i.e., a clinical history of three generations of one Czech family with biochemical and molecular genetic findings. Hyperuricemia was defined as serum uric acid (SUA) concentrations 420 μmol/L for men or 360 μmol/L for women and children under 15 years on two measurements, performed at least four weeks apart. The proband was a 12-year-old girl of Roma ethnicity, whose SUA concentrations were 397–405 µmol/L. Sequencing analyses focusing on the coding region of ABCG2 identified two rare mutations—c.393G>T (p.M131I) and c.706C>T (p.R236X). Segregation analysis revealed a plausible link between these mutations and hyperuricemia and the gout phenotype in family relatives. Functional studies revealed that p.M131I and p.R236X were functionally deficient and null, respectively. Our findings illustrate why genetic factors affecting ABCG2 function should be routinely considered in clinical practice as part of a hyperuricemia/gout diagnosis, especially in pediatric-onset patients with a strong family history.
Collapse
|
19
|
Fahmy UA, Aldawsari HM, Badr-Eldin SM, Ahmed OAA, Alhakamy NA, Alsulimani HH, Caraci F, Caruso G. The Encapsulation of Febuxostat into Emulsomes Strongly Enhances the Cytotoxic Potential of the Drug on HCT 116 Colon Cancer Cells. Pharmaceutics 2020; 12:E956. [PMID: 33050567 PMCID: PMC7600960 DOI: 10.3390/pharmaceutics12100956] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Febuxostat (FBX) is a drug able to inhibit xanthine oxidase and reduce uric acid production commonly used for the treatment of hyperuricemia in subjects suffering from gout. Several studies have also been directed at its use as anti-cancer drug during the last years, opening a window for its off-label use. In the present study, an optimized formulation in terms of vesicle size and drug release, obtained by encapsulation of FBX into the emulsomes (FBX-EMLs), was evaluated for its cytotoxic potential in human colorectal carcinoma (HCT 116) cells. The optimized FBX-EMLs formula had an improved half maximal inhibitory concentration (IC50), about 4-fold lower, compared to the free drug. The cell cycle analysis showed a significant inhibition of the HCT 116 cells proliferation following FBX-EMLs treatment compared to all the other conditions, with a higher number of cells accumulating on G2/M and pre-G1 phases, paralleled by a significant reduction of cells in G0/G1 and S phases. The optimized formula was also able to significantly increase the percentage of cell population in both early and late stages of apoptosis, characterized by a higher intracellular caspase-3 concentration, as well as percentage of necrotic cells. Lastly, the FBX ability to decrease the mitochondrial membrane potential was enhanced when the drug was delivered into the EMLs. In conclusion, the new formulation of FBX into EMLs improved all the parameters related to the anti-proliferative activity and the toxic potential of the drug towards colorectal cancer cells.
Collapse
Affiliation(s)
- Usama A. Fahmy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
| | - Hibah M. Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
| | - Shaimaa M. Badr-Eldin
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt
| | - Osama A. A. Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
- Advanced Drug Delivery Research Group, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Helal H. Alsulimani
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (U.A.F.); (H.M.A.); (S.M.B.-E.); (O.A.A.A.); (N.A.A.); (H.H.A.)
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, Via Conte Ruggero, 73, 94018 Troina (EN), Italy;
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Giuseppe Caruso
- Oasi Research Institute—IRCCS, Via Conte Ruggero, 73, 94018 Troina (EN), Italy;
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| |
Collapse
|