1
|
Stevens LJ, Zhu AZX, Chothe PP, Chowdhury SK, Donkers JM, Vaes WHJ, Knibbe CAJ, Alwayn IPJ, van de Steeg E. Evaluation of Normothermic Machine Perfusion of Porcine Livers as a Novel Preclinical Model to Predict Biliary Clearance and Transporter-Mediated Drug-Drug Interactions Using Statins. Drug Metab Dispos 2021; 49:780-789. [PMID: 34330719 DOI: 10.1124/dmd.121.000521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/23/2021] [Indexed: 11/22/2022] Open
Abstract
There is a lack of translational preclinical models that can predict hepatic handling of drugs. In this study, we aimed to evaluate the applicability of normothermic machine perfusion (NMP) of porcine livers as a novel ex vivo model to predict hepatic clearance, biliary excretion, and plasma exposure of drugs. For this evaluation, we dosed atorvastatin, pitavastatin, and rosuvastatin as model drugs to porcine livers and studied the effect of common drug-drug interactions (DDIs) on these processes. After 120 minutes of perfusion, 0.104 mg atorvastatin (n = 3), 0.140 mg pitavastatin (n = 5), or 1.4 mg rosuvastatin (n = 4) was administered to the portal vein, which was followed 120 minutes later by a second bolus of the statin coadministered with OATP perpetrator drug rifampicin (67.7 mg). After the first dose, all statins were rapidly cleared from the circulation (hepatic extraction ratio > 0.7) and excreted into the bile. Presence of human-specific atorvastatin metabolites confirmed the metabolic capacity of porcine livers. The predicted biliary clearance of rosuvastatin was found to be closer to the observed biliary clearance. A rank order of the DDI between the various systems upon coadministration with rifampicin could be observed: atorvastatin (AUC ratio 7.2) > rosuvastatin (AUC ratio 3.1) > pitavastatin (AUC ratio 2.6), which is in good agreement with the clinical DDI data. The results from this study demonstrated the applicability of using NMP of porcine livers as a novel preclinical model to study OATP-mediated DDI and its effect on hepatic clearance, biliary excretion, and plasma profile of drugs. SIGNIFICANCE STATEMENT: This study evaluated the use of normothermic machine perfusion (NMP) of porcine livers as a novel preclinical model to study hepatic clearance, biliary excretion, plasma (metabolite) profile of statins, and OATP-mediated DDI. Results showed that NMP of porcine livers is a reliable model to study OATP-mediated DDI. Overall, the rank order of DDI severity indicated in these experiments is in good agreement with clinical data, indicating the potential importance of this new ex vivo model in early drug discovery.
Collapse
Affiliation(s)
- L J Stevens
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - A Z X Zhu
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - P P Chothe
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - S K Chowdhury
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - J M Donkers
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - W H J Vaes
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - C A J Knibbe
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - I P J Alwayn
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| | - E van de Steeg
- Department of Surgery, Leiden University Medical Centre (LUMC) Transplant Center, Leiden, The Netherlands (L.J.S., I.P.J.A.); The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands (L.J.S., J.M.D., W.H.J.V., E.v.d.S.); Quantitative Solutions (A.Z.X.Z.), Department of Drug Metabolism & Pharmacokinetic (P.P.C., S.K.C.), Takeda Pharmaceutical International, Cambridge, Massachusetts; Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands (C.A.J.K.); and Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein and Utrecht, The Netherlands (C.A.J.K.)
| |
Collapse
|
2
|
Dahlgren D, Sjöblom M, Lennernäs H. Intestinal absorption-modifying excipients: A current update on preclinical in vivo evaluations. Eur J Pharm Biopharm 2019; 142:411-420. [DOI: 10.1016/j.ejpb.2019.07.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/27/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022]
|
3
|
Hibbins AR, Govender M, Indermun S, Kumar P, du Toit LC, Choonara YE, Pillay V. In Vitro–In Vivo Evaluation of an Oral Ghost Drug Delivery Device for the Delivery of Salmon Calcitonin. J Pharm Sci 2018; 107:1605-1614. [DOI: 10.1016/j.xphs.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/27/2018] [Accepted: 02/06/2018] [Indexed: 11/29/2022]
|
4
|
Momper JD, Tsunoda SM, Ma JD. Evaluation of Proposed In Vivo Probe Substrates and Inhibitors for Phenotyping Transporter Activity in Humans. J Clin Pharmacol 2016; 56 Suppl 7:S82-98. [DOI: 10.1002/jcph.736] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/23/2016] [Accepted: 03/07/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Jeremiah D. Momper
- University of California, San Diego; Skaggs School of Pharmacy & Pharmaceutical Sciences; La Jolla CA USA
| | - Shirley M. Tsunoda
- University of California, San Diego; Skaggs School of Pharmacy & Pharmaceutical Sciences; La Jolla CA USA
| | - Joseph D. Ma
- University of California, San Diego; Skaggs School of Pharmacy & Pharmaceutical Sciences; La Jolla CA USA
| |
Collapse
|
5
|
Malik MY, Jaiswal S, Sharma A, Shukla M, Lal J. Role of enterohepatic recirculation in drug disposition: cooperation and complications. Drug Metab Rev 2016; 48:281-327. [PMID: 26987379 DOI: 10.3109/03602532.2016.1157600] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Enterohepatic recirculation (EHC) concerns many physiological processes and notably affects pharmacokinetic parameters such as plasma half-life and AUC as well as estimates of bioavailability of drugs. Also, EHC plays a detrimental role as the compounds/drugs are allowed to recycle. An in-depth comprehension of this phenomenon and its consequences on the pharmacological effects of affected drugs is important and decisive in the design and development of new candidate drugs. EHC of a compound/drug occurs by biliary excretion and intestinal reabsorption, sometimes with hepatic conjugation and intestinal deconjugation. EHC leads to prolonged elimination half-life of the drugs, altered pharmacokinetics and pharmacodynamics. Study of the EHC of any drug is complicated due to unavailability of the apposite model, sophisticated procedures and ethical concerns. Different in vitro and in vivo methods for studies in experimental animals and humans have been devised, each having its own merits and demerits. Involvement of the different transporters in biliary excretion, intra- and inter-species, pathological and biochemical variabilities obscure the study of the phenomenon. Modeling of drugs undergoing EHC has always been intricate and exigent models have been exploited to interpret the pharmacokinetic profiles of drugs witnessing multiple peaks due to EHC. Here, we critically appraise the mechanisms of bile formation, factors affecting biliary drug elimination, methods to estimate biliary excretion of drugs, EHC, multiple peak phenomenon and its modeling.
Collapse
Affiliation(s)
- Mohd Yaseen Malik
- a Department of Pharmaceutics , National Institute of Pharmaceutical Education and Research (NIPER) , Raebareli , India ;,b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India
| | - Swati Jaiswal
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| | - Abhisheak Sharma
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India ;,d Department of Pharmaceutics and Drug Delivery, School of Pharmacy , The University of Mississippi , Oxford , USA
| | - Mahendra Shukla
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| | - Jawahar Lal
- b Pharmacokinetics & Metabolism Division , CSIR-Central Drug Research Institute , Lucknow , India ;,c Academy of Scientific and Innovative Research , New Delhi , India
| |
Collapse
|
6
|
Shen H, Su H, Liu T, Yao M, Mintier G, Li L, Fancher RM, Iyer R, Marathe P, Lai Y, Rodrigues AD. Evaluation of Rosuvastatin as an Organic Anion Transporting Polypeptide (OATP) Probe Substrate: In Vitro Transport and In Vivo Disposition in Cynomolgus Monkeys. J Pharmacol Exp Ther 2015; 353:380-91. [DOI: 10.1124/jpet.114.221804] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
7
|
Christiansen ML, Müllertz A, Garmer M, Kristensen J, Jacobsen J, Abrahamsson B, Holm R. Evaluation of the Use of Göttingen Minipigs to Predict Food Effects on the Oral Absorption of Drugs in Humans. J Pharm Sci 2015; 104:135-43. [DOI: 10.1002/jps.24270] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
|
8
|
Dahlgren D, Roos C, Sjögren E, Lennernäs H. Direct In Vivo Human Intestinal Permeability (Peff ) Determined with Different Clinical Perfusion and Intubation Methods. J Pharm Sci 2014; 104:2702-26. [PMID: 25410736 DOI: 10.1002/jps.24258] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/17/2014] [Accepted: 10/17/2014] [Indexed: 12/21/2022]
Abstract
Regional in vivo human intestinal effective permeability (Peff ) is calculated by measuring the disappearance rate of substances during intestinal perfusion. Peff is the most relevant parameter in the prediction of rate and extent of drug absorption from all parts of the intestine. Today, human intestinal perfusions are not performed on a routine basis in drug development. Therefore, it would be beneficial to increase the accuracy of the in vitro and in silico tools used to evaluate the intestinal Peff of novel drugs. This review compiles historical Peff data from 273 individual measurements of 80 substances from 61 studies performed in all parts of the human intestinal tract. These substances include: drugs, monosaccharaides, amino acids, dipeptides, vitamins, steroids, bile acids, ions, fatty acids, and water. The review also discusses the determination and prediction of Peff using in vitro and in silico methods such as quantitative structure-activity relationship, Caco-2, Ussing chamber, animal intestinal perfusion, and physiologically based pharmacokinetic (PBPK) modeling. Finally, we briefly outline how to acquire accurate human intestinal Peff data by deconvolution of plasma concentration-time profiles following regional intestinal bolus dosing.
Collapse
Affiliation(s)
- David Dahlgren
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Carl Roos
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Erik Sjögren
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
9
|
Jamei M, Bajot F, Neuhoff S, Barter Z, Yang J, Rostami-Hodjegan A, Rowland-Yeo K. A mechanistic framework for in vitro-in vivo extrapolation of liver membrane transporters: prediction of drug-drug interaction between rosuvastatin and cyclosporine. Clin Pharmacokinet 2014; 53:73-87. [PMID: 23881596 PMCID: PMC3889821 DOI: 10.1007/s40262-013-0097-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background and Objectives The interplay between liver metabolising enzymes and transporters is a complex process involving system-related parameters such as liver blood perfusion as well as drug attributes including protein and lipid binding, ionisation, relative magnitude of passive and active permeation. Metabolism- and/or transporter-mediated drug–drug interactions (mDDIs and tDDIs) add to the complexity of this interplay. Thus, gaining meaningful insight into the impact of each element on the disposition of a drug and accurately predicting drug–drug interactions becomes very challenging. To address this, an in vitro–in vivo extrapolation (IVIVE)-linked mechanistic physiologically based pharmacokinetic (PBPK) framework for modelling liver transporters and their interplay with liver metabolising enzymes has been developed and implemented within the Simcyp Simulator®. Methods In this article an IVIVE technique for liver transporters is described and a full-body PBPK model is developed. Passive and active (saturable) transport at both liver sinusoidal and canalicular membranes are accounted for and the impact of binding and ionisation processes is considered. The model also accommodates tDDIs involving inhibition of multiple transporters. Integrating prior in vitro information on the metabolism and transporter kinetics of rosuvastatin (organic-anion transporting polypeptides OATP1B1, OAT1B3 and OATP2B1, sodium-dependent taurocholate co-transporting polypeptide [NTCP] and breast cancer resistance protein [BCRP]) with one clinical dataset, the PBPK model was used to simulate the drug disposition of rosuvastatin for 11 reported studies that had not been used for development of the rosuvastatin model. Results The simulated area under the plasma concentration–time curve (AUC), maximum concentration (Cmax) and the time to reach Cmax (tmax) values of rosuvastatin over the dose range of 10–80 mg, were within 2-fold of the observed data. Subsequently, the validated model was used to investigate the impact of coadministration of cyclosporine (ciclosporin), an inhibitor of OATPs, BCRP and NTCP, on the exposure of rosuvastatin in healthy volunteers. Conclusion The results show the utility of the model to integrate a wide range of in vitro and in vivo data and simulate the outcome of clinical studies, with implications for their design. Electronic supplementary material The online version of this article (doi:10.1007/s40262-013-0097-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Jamei
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, John Street, S2 4SU, Sheffield, UK,
| | | | | | | | | | | | | |
Collapse
|
10
|
Sjögren E, Hedeland M, Bondesson U, Lennernäs H. Effects of verapamil on the pharmacokinetics and hepatobiliary disposition of fexofenadine in pigs. Eur J Pharm Sci 2014; 57:214-23. [PMID: 24075962 DOI: 10.1016/j.ejps.2013.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 08/19/2013] [Accepted: 09/18/2013] [Indexed: 11/29/2022]
Abstract
The pharmacokinetics (PK) of fexofenadine (FEX) in pigs were investigated with the focus on exploring the interplay between hepatic transport and metabolism when administered intravenously (iv) alone or with verapamil. The in vivo pig model enabled simultaneous sampling from plasma (pre-liver, post-liver and peripheral), bile and urine. Each animal was administered FEX 35mg iv alone or with verapamil 35mg. Plasma, bile and urine were analyzed with liquid chromatography-tandem mass spectrometry. Non-compartmental analysis (NCA) was used to estimate traditional PK parameters. In addition, a physiologically based pharmacokinetic (PBPK) model consisting of 11 compartments (6 tissues +5 sample sites) was applied for mechanistic elucidation and estimation of individual PK parameters. FEX had a terminal half-life of 1.7h and a liver extraction of 3%. The fraction of the administered dose of unchanged FEX excreted into the bile was 25% and the bile exposure was more than 100 times higher than the portal vein total plasma exposure, indicating carrier-mediated (CM) disposition processes in the liver. 23% of the administered dose of FEX was excreted unchanged in the urine. An increase in FEX plasma exposure (+50%) and a decrease in renal clearance (-61%) were detected by NCA as a direct effect of concomitant administration of verapamil. However, analysis of the PBPK model also revealed that biliary clearance was significantly inhibited (-53%) by verapamil. In addition, PBPK analysis established that metabolism and CM uptake were important factors in the disposition of FEX in the liver. In conclusion, this study demonstrated that CM transport of FEX in both liver and kidneys was inhibited by a single dose of verapamil.
Collapse
Affiliation(s)
- Erik Sjögren
- Department of Pharmacy, Biopharmaceutic Research Group, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden.
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 573, SE-751 23 Uppsala, Sweden; National Veterinary Institute (SVA), Department of Chemistry, Environment and Feed Hygiene, SE-751 89 Uppsala, Sweden
| | - Ulf Bondesson
- Department of Medicinal Chemistry, Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 573, SE-751 23 Uppsala, Sweden; National Veterinary Institute (SVA), Department of Chemistry, Environment and Feed Hygiene, SE-751 89 Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmacy, Biopharmaceutic Research Group, Uppsala University, Box 580, SE-751 23 Uppsala, Sweden
| |
Collapse
|
11
|
Sjögren E, Abrahamsson B, Augustijns P, Becker D, Bolger MB, Brewster M, Brouwers J, Flanagan T, Harwood M, Heinen C, Holm R, Juretschke HP, Kubbinga M, Lindahl A, Lukacova V, Münster U, Neuhoff S, Nguyen MA, Peer AV, Reppas C, Hodjegan AR, Tannergren C, Weitschies W, Wilson C, Zane P, Lennernäs H, Langguth P. In vivo methods for drug absorption – Comparative physiologies, model selection, correlations with in vitro methods (IVIVC), and applications for formulation/API/excipient characterization including food effects. Eur J Pharm Sci 2014; 57:99-151. [PMID: 24637348 DOI: 10.1016/j.ejps.2014.02.010] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 01/11/2023]
|
12
|
Dubbelboer IR, Lilienberg E, Hedeland M, Bondesson U, Piquette-Miller M, Sjögren E, Lennernäs H. The effects of lipiodol and cyclosporin A on the hepatobiliary disposition of doxorubicin in pigs. Mol Pharm 2014; 11:1301-13. [PMID: 24558959 DOI: 10.1021/mp4007612] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Doxorubicin (DOX) emulsified in Lipiodol (LIP) is used as local palliative treatment for unresectable intermediate stage hepatocellular carcinoma. The objective of this study was to examine the poorly understood effects of the main excipient in the drug delivery system, LIP, alone or together with cyclosporin A (CsA), on the in vivo liver disposition of DOX and its active metabolite doxorubicinol (DOXol). The advanced, multi-sampling-site, acute pig model was used; samples were collected from three blood vessels (v. portae, v. hepatica and v. femoralis), bile and urine. The four treatment groups (TI-TIV) all received two intravenous 5 min infusions of DOX into an ear vein: at 0 and 200 min. Before the second dose, the pigs received a portal vein infusion of saline (TI), LIP (TII), CsA (TIII) or LIP and CsA (TIV). Concentrations of DOX and DOXol were analyzed using UPLC-MS/MS. The developed multicompartment model described the distribution of DOX and DOXol in plasma, bile and urine. LIP did not affect the pharmacokinetics of DOX or DOXol. CsA (TIII and TIV) had no effect on the plasma pharmacokinetics of DOX, but a 2-fold increase in exposure to DOXol and a significant decrease in hepatobiliary clearance of DOX and DOXol were observed. Model simulations supported that CsA inhibits 99% of canalicular biliary secretion of both DOX and DOXol, but does not affect the metabolism of DOX to DOXol. In conclusion, LIP did not directly interact with transporters, enzymes and/or biological membranes important for the hepatobiliary disposition of DOX.
Collapse
Affiliation(s)
- Ilse R Dubbelboer
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
13
|
He L, Yang Y, Guo C, Yao D, Liu HH, Sheng JJ, Zhou WP, Ren J, Liu XD, Pan GY. Opposite regulation of hepatic breast cancer resistance protein in type 1 and 2 diabetes mellitus. Eur J Pharmacol 2014; 724:185-92. [DOI: 10.1016/j.ejphar.2013.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/23/2022]
|
14
|
Lundahl A, Tevell Åberg A, Bondesson U, Lennernäs H, Hedeland M. High-resolution mass spectrometric investigation of the phase I and II metabolites of finasteride in pig plasma, urine and bile. Xenobiotica 2013; 44:498-510. [DOI: 10.3109/00498254.2013.866298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
15
|
Lilienberg E, Ebeling Barbier C, Nyman R, Hedeland M, Bondesson U, Axén N, Lennernäs H. Investigation of hepatobiliary disposition of doxorubicin following intrahepatic delivery of different dosage forms. Mol Pharm 2013; 11:131-44. [PMID: 24171458 DOI: 10.1021/mp4002574] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Unresectable, intermediate stage hepatocellular carcinoma (HCC) is often treated palliatively in humans by doxorubicin (DOX). The drug is administered either as a drug-emulsified-in-Lipiodol (DLIP) or as drug loaded into drug eluting beads (DEB), and both formulations are administered intrahepatically. However, several aspects of their in vivo performance in the liver are still not well-understood. In this study, DLIP and DEB were investigated regarding the local and systemic pharmacokinetics (PK) of DOX and its primary metabolite doxorubicinol (DOXol). An advanced PK-multisampling site acute in vivo pig model was used for simultaneous sampling in the portal, hepatic, and femoral veins and the bile duct. The study had a randomized, parallel design with four treatment groups (TI-TIV). TI (n = 4) was used as control and received an intravenous (i.v.) infusion of DOX as a solution. TII and TIII were given a local injection in the hepatic artery with DLIP (n = 4) or DEB (n = 4), respectively. TIV (n = 2) received local injections of DLIP in the hepatic artery and bile duct simultaneously. All samples were analyzed for concentrations of DOX and DOXol with UPLC-MS/MS. Compared to DLIP, the systemic exposure for DOX with DEB was reduced (p < 0.05), in agreement with a slower in vivo release. The approximated intracellular bioavailability of DOX during 6 h appeared to be lower for DEB than DLIP. Following i.v. infusion (55 min), DOX had a liver extraction of 41 (28-53)%, and the fraction of the dose eliminated in bile of DOX and DOXol was 20 (15-22)% and 4.2 (3.2-5.2)%, respectively. The AUCbile/AUCVP for DOX and DOXol was 640 (580-660) and 5000 (3900-5400), respectively. In conclusion, DLIP might initially deliver a higher hepatocellular concentration of DOX than DEB as a consequence of its higher in vivo release rate. Thus, DLIP delivery results in higher intracellular peak concentrations that might correlate with better anticancer effects, but also higher systemic drug exposure and safety issues.
Collapse
Affiliation(s)
- Elsa Lilienberg
- Department of Pharmacy, Uppsala University , Box 580, 751 23 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
16
|
Iusuf D, van Esch A, Hobbs M, Taylor M, Kenworthy KE, van de Steeg E, Wagenaar E, Schinkel AH. Murine Oatp1a/1b Uptake Transporters Control Rosuvastatin Systemic Exposure Without Affecting Its Apparent Liver Exposure. Mol Pharmacol 2013; 83:919-29. [DOI: 10.1124/mol.112.081927] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
17
|
Basu S, Jana S, Patel VB, Patel H. Effects of piperine, cinnamic acid and gallic acid on rosuvastatin pharmacokinetics in rats. Phytother Res 2012. [PMID: 23208983 DOI: 10.1002/ptr.4894] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to investigate the potential pharmacokinetic interactions with natural products (such as piperine (PIP), gallic acid (GA) and cinnamic acid (CA)) and rosuvastatin (RSV) (a specific breast cancer resistance protein, BCRP substrate) in rats. In Caco2 cells, the polarized transport of RSV was effectively inhibited by PIP, CA and GA at concentration of 50 μM. After per oral (p.o.) coadministration of PIP, CA and GA (10 mg/kg) significantly increased intravenous exposure (AUC(last)) of RSV (1 mg/kg) by 73.5%, 62.9% and 53.3% (p < 0.05), respectively than alone group (control). Compared with the control (alone) group, p.o. coadministration of PIP, CA and GA (10 mg/kg) significantly increased the oral exposure (AUC(last)) of RSV (5 mg/kg) by 2.0-fold, 1.83-fold (p < 0.05) and 2.34 -fold (p < 0.05), respectively. Moreover, the cumulative biliary excretion of RSV (5 mg/kg, p.o.) was significantly decreased by 53.3, 33.4 and 39.2% at the end of 8 h after p.o. co-administration of PIP, CA and GA (10 mg/kg), respectively. Taken together, these results indicate that the natural products such as PIP, CA and GA significantly inhibit RSV transport in to bile and increased the plasma exposure (AUC(last)) of RSV.
Collapse
Affiliation(s)
- Sudipta Basu
- Drug Metabolism and Pharmacokinetics -Toxicology Division, Sai Advantium Pharma Ltd, Building 1, Plot No. 2, Chrysalis Enclave, International Biotech Park, Phase - 2, Hinjewadi, Pune, 411057, India
| | | | | | | |
Collapse
|
18
|
A Physiologically Based Pharmacokinetic Model of the Minipig: Data Compilation and Model Implementation. Pharm Res 2012. [DOI: 10.1007/s11095-012-0911-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Thörn HA, Sjögren E, Dickinson PA, Lennernäs H. Binding Processes Determine the Stereoselective Intestinal and Hepatic Extraction of Verapamil in Vivo. Mol Pharm 2012; 9:3034-45. [DOI: 10.1021/mp3000875] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Helena Anna Thörn
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, Sweden
| | - Erik Sjögren
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, Sweden
| | - Paul Alfred Dickinson
- Clinical Pharmacology and Pharmacometrics, AstraZeneca R&D, Alderley Park, Macclesfield, United Kingdom
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, Sweden
| |
Collapse
|
20
|
Sjögren E, Bredberg U, Lennernäs H. The Pharmacokinetics and Hepatic Disposition of Repaglinide in Pigs: Mechanistic Modeling of Metabolism and Transport. Mol Pharm 2012; 9:823-41. [DOI: 10.1021/mp200218p] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Erik Sjögren
- Department of Pharmacy, Uppsala University, Box 580, S-751 23 Uppsala, Sweden
| | | | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Box 580, S-751 23 Uppsala, Sweden
| |
Collapse
|
21
|
Abstract
Drug-drug interactions are a serious clinical issue. An important mechanism underlying drug-drug interactions is induction or inhibition of drug transporters that mediate the cellular uptake and efflux of xenobiotics. Especially drug transporters of the small intestine, liver and kidney are major determinants of the pharmacokinetic profile of drugs. Transporter-mediated drug-drug interactions in these three organs can considerably influence the pharmacokinetics and clinical effects of drugs. In this article, we focus on probe drugs lacking significant metabolism to highlight mechanisms of interactions of selected intestinal, hepatic and renal drug transporters (e.g., organic anion transporting polypeptide [OATP] 1A2, OATP2B1, OATP1B1, OATP1B3, P-gp, organic anion transporter [OAT] 1, OAT3, breast cancer resistance protein [BCRP], organic cation transporter [OCT] 2 and multidrug and toxin extrusion protein [MATE] 1). Genotype-dependent drug-drug interactions are also discussed.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental & Clinical Pharmacology & Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | | |
Collapse
|
22
|
Thörn HA, Lundahl A, Schrickx JA, Dickinson PA, Lennernäs H. Drug metabolism of CYP3A4, CYP2C9 and CYP2D6 substrates in pigs and humans. Eur J Pharm Sci 2011; 43:89-98. [DOI: 10.1016/j.ejps.2011.03.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 02/08/2011] [Accepted: 03/18/2011] [Indexed: 11/28/2022]
|
23
|
Lundahl A, Hedeland M, Bondesson U, Lennernäs H. In Vivo Investigation in Pigs of Intestinal Absorption, Hepatobiliary Disposition, and Metabolism of the 5α-Reductase Inhibitor Finasteride and the Effects of Coadministered Ketoconazole. Drug Metab Dispos 2011; 39:847-57. [DOI: 10.1124/dmd.110.035311] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
24
|
Matsson EM, Palm JE, Eriksson UG, Bottner P, Lundahl A, Knutson L, Lennernäs H. Effects of Ketoconazole on the In Vivo Biotransformation and Hepatobiliary Transport of the Thrombin Inhibitor AZD0837 in Pigs. Drug Metab Dispos 2010; 39:239-46. [DOI: 10.1124/dmd.110.035022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
25
|
Bergman E, Hedeland M, Bondesson U, Lennernäs H. The effect of acute administration of rifampicin and imatinib on the enterohepatic transport of rosuvastatinin vivo. Xenobiotica 2010; 40:558-68. [DOI: 10.3109/00498254.2010.496498] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|