1
|
Kumabe H, Masuda T, Ito S, Furihata T, Toda A, Mogi M, Araki N, Ohtsuki S. Proteome profile differences among human, monkey, and mouse brain microvessels and cultured brain microvascular endothelial cells. Fluids Barriers CNS 2025; 22:53. [PMID: 40448242 PMCID: PMC12124085 DOI: 10.1186/s12987-025-00650-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/04/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) expresses transporters, receptors, and tight junction proteins that regulate the exchange of substances between the blood and brain. The differences in the expression of these proteins in the BBB among different species and cultured BBB model cells should be clarified to interpret the BBB function in model animals and cells. This study aimed to elucidate species differences among humans, monkeys, and mice and in vitro-in vivo differences in the BBB proteome using deep proteomic analysis. METHODS Brain microvessels (BMVs) were isolated from frozen cerebral cortices of human and monkey and frozen mouse cerebrums. BMVs and cultured brain microvascular endothelial cells (BMECs), such as hCMEC/D3, HBMEC/ciβ, and primary BMECs, were analyzed via data-independent acquisition using liquid chromatography-mass spectrometry. RESULTS Proteomics identified 7,149-8,274 proteins in the BMV fractions and 6,657-7,534 proteins in the brain lysates. Comparative analysis revealed distinct proteomic profiles among the three species, with the human profile being more similar to that of monkeys than that of mice. The expression profile of the solute carrier organic anion transporter family was found to vary among mouse, monkey, and human BMVs. The expression levels of SLC22A6/Slc22a6 and SLC22A8/Slc22a8 were higher in mice than in monkeys and humans, whereas SLC43A3/Slc43a3 expression levels were lower in mice. The expression of amino acid transporters, such as SLC7A5 and SLC3A2/Slc3a2, was higher in BMVs, whereas that of SLC1A5/Slc1a5 and SLC38A9/Slc38a9 was higher in cultured BMECs. MFSD2A/Mfsd2a and SLC27A1/Slc27a1 were highly expressed in BMVs. The expression of tight junction proteins, particularly the claudin family, varied between BMVs and cultured BMECs and among cell lines. Specifically, the expression of claudin-5 was higher in BMVs, and claudin-11 expression was higher in cultured BMECs. CONCLUSIONS Deep proteomic analysis revealed species-specific differences in transport-related proteins in the BBB. Furthermore, in vitro and in vivo differences were observed in the transporter and claudin protein expression. This study provides a BBB proteome profile dataset and offers insights for a comprehensive understanding of BBB protein expression across species and between in vivo and in vitro conditions.
Collapse
Affiliation(s)
- Haruka Kumabe
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, 997-0017, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Tomomi Furihata
- Laboratory of Advanced Drug Development Sciences, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Akiko Toda
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd, Miyanoura, Kagoshima, 891-1394, Japan
| | - Masayuki Mogi
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd, Miyanoura, Kagoshima, 891-1394, Japan
| | - Norie Araki
- Department of Tumor Genetics and Biology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan.
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 862-0973, Japan.
| |
Collapse
|
2
|
Rysz MA, Schäfer AM, Kinzi J, Paloumpis N, In-Albon K, Schmidlin S, Seibert I, Ricklin D, Meyer Zu Schwabedissen HE. Erlotinib-A substrate and inhibitor of OATP2B1: pharmacokinetics and CYP3A-mediated metabolism in rSlco2b1 -/- and SLCO2B1 +/+ rats. Drug Metab Dispos 2025; 53:100069. [PMID: 40239314 DOI: 10.1016/j.dmd.2025.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025] Open
Abstract
The tyrosine kinase inhibitor erlotinib is recognized as a substrate of cytochrome P450 enzymes and drug transporters. Indeed, erlotinib's extensive metabolism to the active metabolite OSI-420 (desmethyl erlotinib) mainly involves CYP3A enzymes. Additionally, erlotinib is assumed to interact with the organic anion transporting polypeptide (OATP)2B1. In this study, we aimed to investigate the role of human OATP2B1 in erlotinib's metabolism through in vitro and in vivo experiments. Using Madin-Darby canine kidney cells expressing human OATP2B1 for competitive counterflow experiments, we confirmed erlotinib as inhibitor and substrate of the transporter. Moreover, in vitro transport experiments revealed higher cellular accumulation of erlotinib at pH 5.5 than that at pH 7.4. Pharmacokinetic evaluation of orally administered erlotinib in male SLCO2B1+/+ and rSlco2b1-/- rats revealed that the human OATP2B1 does not significantly alter serum levels of erlotinib or its main metabolite OSI-420, although we observed a longer mean residence time of the metabolite in humanized rats. Although there was no difference in the OSI-420:erlotinib ratio over time in SLCO2B1+/+ and rSlco2b1-/- rats, we assessed the role of CYP3A1 and CYP3A2 in the metabolism of erlotinib. In vitro experiments showed a contribution of both enzymes to the formation of OSI-420. For CYP3A1, we found significantly higher expression in liver microsomes of male SLCO2B1+/+ rats, while the knockout genotype showed significantly higher levels of CYP3A2. However, these differences did not affect the systemic exposure of erlotinib or OSI-420 in the rats. Our findings provide further insight into the role of OATP2B1 in the disposition of orally administered erlotinib. SIGNIFICANCE STATEMENT: This study confirms that erlotinib is a substrate of the human organic anion transporting polypeptide 2B1 transporter in vitro. In vivo experiments in rat models, however, showed no significant impact of organic anion transporting polypeptide 2B1 on the systemic exposure of erlotinib or its metabolite, OSI-420. Despite variations in CYP3A enzyme expression in SLCO2B1+/+ rats, the OSI-420:erlotinib ratio remained unchanged. Although SLCO2B1+/+ rats exhibited a longer mean residence time for OSI-420, this did not significantly alter overall exposure in orally treated animals.
Collapse
Affiliation(s)
- Marta A Rysz
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Anima M Schäfer
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jonny Kinzi
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Nikolaos Paloumpis
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Katja In-Albon
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Seraina Schmidlin
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Isabell Seibert
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Daniel Ricklin
- Molecular Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | | |
Collapse
|
3
|
Taheri H, Li Y, Huang KM, Ahmed E, Jin Y, Drabison T, Yang Y, Kulp SK, Young NA, Li J, Cheng X, Corps KN, Coss CC, Vaughn JE, Lustberg MB, Sparreboom A, Hu S. OATP1B-type Transport Function Is a Determinant of Aromatase Inhibitor-Associated Arthralgia Susceptibility. CANCER RESEARCH COMMUNICATIONS 2025; 5:496-510. [PMID: 40062557 PMCID: PMC11948302 DOI: 10.1158/2767-9764.crc-24-0475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
SIGNIFICANCE AIs are effective but often discontinued because of arthralgia. This study explores the role of OATP1B transporters in AI-related side effects and the potential usage of transporter biomarkers to predict and reduce the risk of arthralgia associated with AI treatment.
Collapse
Affiliation(s)
- Hanieh Taheri
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Kevin M. Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Eman Ahmed
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Drabison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yan Yang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Nicholas A. Young
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Junan Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Kara N. Corps
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jennifer E. Vaughn
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Maryam B. Lustberg
- Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
4
|
Martinez-Guerrero LJ, Zhang X, Wright SH, Cherrington NJ. Characterization of Human Organic Anion Transporter 4 (hOAT4) and Mouse Oat5 (mOat5) As Functional Orthologs for Renal Anion Uptake and Efflux Transport. J Pharmacol Exp Ther 2024; 391:378-386. [PMID: 38627096 PMCID: PMC11585314 DOI: 10.1124/jpet.123.001979] [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: 10/26/2023] [Accepted: 04/09/2024] [Indexed: 11/21/2024] Open
Abstract
Organic anions (OAs) are compounds including drugs or toxicants that are negatively charged at physiologic pH and are typically transported by organic anion transporters (OATs). Human OAT4 (SLC22A11) is expressed in the apical membrane of renal proximal tubules. Although there is no rodent ortholog of hOAT4, rodents express Oat5 (Slc22a19), an anion exchanger that is also localized to the apical membrane of renal proximal tubule cells. The purpose of this study was to determine the functional similarity between mouse Oat5 and human OAT4. Chinese hamster ovary (CHO) cells expressing SLC22A11 or Slc22a19 were used to assess the transport characteristics of radiolabeled ochratoxin (OTA). We determined the kinetics of OTA transport; the resulting Michaelis constant (Kt) and maximal rate of mediated substrate transport (Jmax) values were very similar for both hOAT4 and mOat5: Kt 3.9 and 7.2 μM, respectively, and Jmax 4.4 and 3.9 pmol/cm2, respectively. For the profile of OTA inhibition by OAs, IC50 values were determined for several clinically important drugs and toxicants. The resulting IC50 values ranged from 9 μM for indomethacin to ∼600 μM for the diuretic hydrochlorothiazide. We measured the efflux of OTA from preloaded cells; both hOAT4 and mOat5 supported the efflux of OTA. These data support the hypothesis that OAT4 and Oat5 are functional orthologs and share selectivity for OTA both for reabsorption and secretion. SIGNIFICANCE STATEMENT: This study compares the selectivity profile between human organic anion transporter (OAT4) and mouse Oat5. Our data revealed a similar selectivity profile for ochratoxin A (OTA) reabsorption and secretion by these two transporters, thereby supporting the hypothesis that hOAT4 and mOat5, although not genetic orthologs, behave as functional orthologs for both uptake and efflux. These data will be instrumental in selecting an appropriate animal model when studying the renal disposition of anionic drugs and toxicants.
Collapse
Affiliation(s)
- Lucy J Martinez-Guerrero
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Xiaohong Zhang
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| |
Collapse
|
5
|
Du X, Liu M, Trevisi E, Ju L, Yang Y, Gao W, Song Y, Lei L, Zolzaya M, Li X, Fang Z, Liu G. Expression of hepatic genes involved in bile acid metabolism in dairy cows with fatty liver. J Dairy Sci 2024:S0022-0302(24)00833-6. [PMID: 38825110 DOI: 10.3168/jds.2023-24485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/28/2024] [Indexed: 06/04/2024]
Abstract
Bile acids are cholesterol-derived molecules that are primarily produced in the liver. In nonruminants with fatty liver, overproduction of bile acids is associated with liver injury. During the transition period, fatty liver is a metabolic disorder that can affect up to 50% of high-producing dairy cows. The purpose of this study was to provide a comprehensive evaluation on hepatic bile acid metabolism in dairy cows with fatty liver by assessing expression changes of genes involved in bile acid synthesis, export and uptake. The serum activities of aspartate aminotransferase, alanine aminotransferase and glutamate dehydrogenase and concentration of total bile acids were all greater, whereas serum concentration of total cholesterol was lower in cows with fatty liver than in healthy cows. Content of total bile acids was higher but total cholesterol was slightly lower in liver tissues from fatty liver cows than from healthy cows. The hepatic mRNA abundance of cholesterol 7a-hydroxylase (CYP7A1), hydroxy-delta-5-steroid dehydrogenase, 3 β- and steroid delta-isomerase 7 (HSD3B7) and sterol 12α-hydroxylase (CYP8B1), enzymes involved in the classic pathway of bile acid synthesis, was higher in fatty liver cows than in healthy cows. Compared with healthy cows, the hepatic mRNA abundance of alternative bile acid synthesis pathway-related genes sterol 27-hydroxylase (CYP27A1) and oxysterol 7α-hydroxylase (CYP7B1) did not differ in cows with fatty liver. The protein and mRNA abundance of bile acid transporter bile salt efflux pump (BSEP) were lower in the liver of dairy cow with fatty liver. Compared with healthy cows, the hepatic mRNA abundance of bile acid transporters solute carrier family 51 subunit α (SLC51A), ATP binding cassette subfamily C member 1 (ABCC1) and 3 (ABCC3) was greater in cows with fatty liver, whereas the solute carrier family 51 subunit β (SLC51B) did not differ. The expression of genes involved in bile acid uptake, including solute carrier family 10 member 1 (NTCP), solute carrier organic anion transporter family member 1A2 (SLCO1A2) and 2B1 (SLCO2B1) was upregulated in dairy cows with fatty liver. Furthermore, the hepatic protein and mRNA abundance of bile acid metabolism regulators farnesoid X receptor (FXR) and small heterodimer partner (SHP) were lower in cows with fatty liver than in healthy cows. Overall, these data suggest that inhibition of FXR signaling pathway may lead to the increased bile acid synthesis and uptake and decreased secretion of bile acids from hepatocytes to the bile, which elevates hepatic bile acids content in dairy cows with fatty liver. As the hepatotoxicity of bile acids has been demonstrated on nonruminant hepatocytes, it is likely that the liver injury is induced by increased hepatic bile acids content in dairy cows with fatty liver.
Collapse
Affiliation(s)
- Xiliang Du
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Mingchao Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071001, Hebei, China
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Lingxue Ju
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuting Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenwen Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuxiang Song
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lin Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Majigsuren Zolzaya
- Institute of Veterinary Medicine, Mongolian Mongolian University of Life Sciences (MULS)
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhiyuan Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guowen Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
6
|
Loos NHC, Ferreira Martins ML, Rijmers J, de Jong D, Lebre MC, Tibben M, Beijnen JH, Schinkel AH. Interplay of Ritonavir-Boosted Oral Cabazitaxel with the Organic Anion-Transporting Polypeptide (OATP) Uptake Transporters and Carboxylesterase 1 in Mice. Mol Pharm 2024; 21:1952-1964. [PMID: 38423793 DOI: 10.1021/acs.molpharmaceut.3c01205] [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] [Indexed: 03/02/2024]
Abstract
Intravenously administered chemotherapeutic cabazitaxel is used for palliative treatment of prostate cancer. An oral formulation would be more patient-friendly and reduce the need for hospitalization. We therefore study determinants of the oral pharmacokinetics of cabazitaxel in a ritonavir-boosted setting, which reduces the CYP3A-mediated first-pass metabolism of cabazitaxel. We here assessed the role of organic anion-transporting polypeptides (OATPs) in the disposition of orally boosted cabazitaxel and its active metabolites, using the Oatp1a/b-knockout and the OATP1B1/1B3-transgenic mice. These transporters may substantially affect plasma clearance and hepatic and intestinal drug disposition. The pharmacokinetics of cabazitaxel and DM2 were not significantly affected by Oatp1a/b and OATP1B1/1B3 activity. In contrast, the plasma AUC0-120 min of DM1 in Oatp1a/b-/- was 1.9-fold (p < 0.05) higher than that in wild-type mice, and that of docetaxel was 2.4-fold (p < 0.05) higher. We further observed impaired hepatic uptake and intestinal disposition for DM1 and docetaxel in the Oatp-ablated strains. None of these parameters showed rescue by the OATP1B1 or -1B3 transporters in the humanized mouse strains, suggesting a minimal role of OATP1B1/1B3. Ritonavir itself was also a potent substrate for mOatp1a/b, showing a 2.9-fold (p < 0.0001) increased plasma AUC0-120 min and 3.5-fold (p < 0.0001) decreased liver-to-plasma ratio in Oatp1a/b-/- compared to those in wild-type mice. Furthermore, we observed the tight binding of cabazitaxel and its active metabolites, including docetaxel, to plasma carboxylesterase (Ces1c) in mice, which may complicate the interpretation of pharmacokinetic and pharmacodynamic mouse studies. Collectively, these results will help to further optimize (pre)clinical research into the safety and efficacy of orally applied cabazitaxel.
Collapse
Affiliation(s)
- Nancy H C Loos
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | | | - Jamie Rijmers
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Daniëlle de Jong
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Matthijs Tibben
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
- Division of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
7
|
Levitt DG, Levitt MD. Development of a Pharmacokinetic Model That Accounts for the Plasma Concentrations of Conjugated and Unconjugated Bilirubin Observed in a Variety of Disease States. Clin Exp Gastroenterol 2023; 16:277-289. [PMID: 38143515 PMCID: PMC10749172 DOI: 10.2147/ceg.s438140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/13/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction For a large variety of liver pathologies, the plasma unconjugated (UB) and conjugated (CB) bilirubin concentrations appear to be coupled. For example, in alcoholic cirrhosis, UB and CB are roughly the same over a large range of total bilirubin, requiring an initial massive increase (about 40-fold) in plasma CB to reach the level of UB and then similar increases in UB and CB as the disease progresses. This coupling has been either unrecognized or ignored and this paper is the first attempt to try to explain it quantitatively in terms of known hepatic cell metabolic and membrane transport properties. Methods A simplified pharmacokinetic model is developed and applied to a variety of hyperbilirubinemic pathologies. A central feature of the model is based on the recent observation that double knockout of the rat OATP1A and OATP1B hepatic transporters produces a roughly 400-fold increase in plasma CB, indicating that there is a normal rapid recycling of CB from the cell to the plasma with reuptake via OATP. We use the experimental rat Km of OATP CB transport to show that OATP uptake becomes saturated at relatively low plasma CB concentrations, decreasing uptake, and producing massive (up to 1000-fold) increases in CB in some pathologies. It is assumed that UB and CB are competing for the OATP transporter, producing the increased plasma UB that is observed in "pure" CB pathologies. Results The model accurately describes the clinically observed UB and CB for pure UB (Gilbert's, hemolytic anemia) and CB (Dubin-Johnson, Rotor syndrome, biliary atresia) pathologies as well as in cirrhosis. Conclusion This model is a preliminary, first attempt to quantitatively describe UB and CB pharmacokinetics. It is hoped that it will stimulate more detailed measurements and analysis.
Collapse
Affiliation(s)
- David G Levitt
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Michael D Levitt
- Research Service, Veterans Affairs Medical Center, Minneapolis, MN, USA
| |
Collapse
|
8
|
Miyake T, Tsutsui H, Hirabayashi M, Tachibana T. Quantitative Prediction of OATP-Mediated Disposition and Biliary Clearance Using Human Liver Chimeric Mice. J Pharmacol Exp Ther 2023; 387:135-149. [PMID: 37142442 DOI: 10.1124/jpet.123.001595] [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: 01/26/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
Drug biliary clearance (CLbile) in vivo is among the most difficult pharmacokinetic parameters to predict accurately and quantitatively because biliary excretion is influenced by metabolic enzymes, transporters, and passive diffusion across hepatocyte membranes. The purpose of this study is to demonstrate the use of Hu-FRG mice [Fah-/-/Rag2-/-/Il2rg-/- (FRG) mice transplanted with human-derived hepatocytes] to quantitatively predict human organic anion transporting polypeptide (OATP)-mediated drug disposition and CLbile To predict OATP-mediated disposition, six OATP substrates (atorvastatin, fexofenadine, glibenclamide, pitavastatin, pravastatin, and rosuvastatin) were administered intravenously to Hu-FRG and Mu-FRG mice (FRG mice transplanted with mouse hepatocytes) with or without rifampicin as an OATP inhibitor. We calculated the hepatic intrinsic clearance (CLh,int) and the change of hepatic clearance (CLh) caused by rifampicin (CLh ratio). We compared the CLh,int of humans with that of Hu-FRG mice and the CLh ratio of humans with that of Hu-FRG and Mu-FRG mice. For predicting CLbile, 20 compounds (two cassette doses of 10 compounds) were administered intravenously to gallbladder-cannulated Hu-FRG and Mu-FRG mice. We evaluated the CLbile and investigated the correlation of human CLbile with that of Hu-FRG and Mu-FRG mice. We found good correlations between humans and Hu-FRG mice in CLh,int (100% within threefold) and CLh ratio (R2 = 0.94). Moreover, we observed a much better relationship between humans and Hu-FRG mice in CLbile (75% within threefold). Our results suggest that OATP-mediated disposition and CLbile can be predicted using Hu-FRG mice, making them a useful in vivo drug discovery tool for quantitatively predicting human liver disposition. SIGNIFICANCE STATEMENT: OATP-mediated disposition and biliary clearance of drugs are likely quantitatively predictable using Hu-FRG mice. The findings can enable the selection of better drug candidates and the development of more effective strategies for managing OATP-mediated DDIs in clinical studies.
Collapse
Affiliation(s)
- Taiji Miyake
- Pharmaceutical Science Department, Translational Research Division (T.M., T.T.) and Discovery Biologics Department, Research Division (H.T.), Chugai Pharmaceutical Co., Ltd., Shizuoka, Gotemba, Japan and Chugai Research Institute for Medical Science Inc., Shizuoka, Gotemba, Japan (M.H.)
| | - Haruka Tsutsui
- Pharmaceutical Science Department, Translational Research Division (T.M., T.T.) and Discovery Biologics Department, Research Division (H.T.), Chugai Pharmaceutical Co., Ltd., Shizuoka, Gotemba, Japan and Chugai Research Institute for Medical Science Inc., Shizuoka, Gotemba, Japan (M.H.)
| | - Manabu Hirabayashi
- Pharmaceutical Science Department, Translational Research Division (T.M., T.T.) and Discovery Biologics Department, Research Division (H.T.), Chugai Pharmaceutical Co., Ltd., Shizuoka, Gotemba, Japan and Chugai Research Institute for Medical Science Inc., Shizuoka, Gotemba, Japan (M.H.)
| | - Tatsuhiko Tachibana
- Pharmaceutical Science Department, Translational Research Division (T.M., T.T.) and Discovery Biologics Department, Research Division (H.T.), Chugai Pharmaceutical Co., Ltd., Shizuoka, Gotemba, Japan and Chugai Research Institute for Medical Science Inc., Shizuoka, Gotemba, Japan (M.H.)
| |
Collapse
|
9
|
Oude Elferink R, Van De Graaf SFJ. Bile Salts by the Back Road. Cell Mol Gastroenterol Hepatol 2023:S2352-345X(23)00068-1. [PMID: 37244292 PMCID: PMC10394266 DOI: 10.1016/j.jcmgh.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/29/2023]
Affiliation(s)
- Ronald Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Research Institute AGEM, University of Amsterdam, Amsterdam, the Netherlands.
| | - Stan F J Van De Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, Research Institute AGEM, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Li Y, Jin Y, Taheri H, Schmidt KT, Gibson AA, Buck SAJ, Eisenmann ED, Mathijssen RHJ, Figg WD, Baker SD, Sparreboom A, Hu S. A Metabolomics Approach for Predicting OATP1B-Type Transporter-Mediated Drug–Drug Interaction Liabilities. Pharmaceutics 2022; 14:pharmaceutics14091933. [PMID: 36145680 PMCID: PMC9502272 DOI: 10.3390/pharmaceutics14091933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, various endogenous compounds have been proposed as putative biomarkers for the hepatic uptake transporters OATP1B1 and OATP1B3 that have the potential to predict transporter-mediated drug–drug interactions (DDIs). However, these compounds have often been identified from top–down strategies and have not been fully utilized as a substitute for traditional DDI studies. In an attempt to eliminate observer bias in biomarker selection, we applied a bottom–up, untargeted metabolomics screening approach in mice and found that plasma levels of the conjugated bile acid chenodeoxycholate-24-glucuronide (CDCA-24G) are particularly sensitive to deletion of the orthologous murine transporter Oatp1b2 (31-fold increase vs. wild type) or the entire Oatp1a/1b(−/−)cluster (83-fold increased), whereas the humanized transgenic overexpression of hepatic OATP1B1 or OATP1B3 resulted in the partial restoration of transport function. Validation studies with the OATP1B1/OATP1B3 inhibitors rifampin and paclitaxel in vitro as well as in mice and human subjects confirmed that CDCA-24G is a sensitive and rapid response biomarker to dose-dependent transporter inhibition. Collectively, our study confirmed the ability of CDCA-24G to serve as a sensitive and selective endogenous biomarker of OATP1B-type transport function and suggests a template for the future development of biomarkers for other clinically important xenobiotic transporters.
Collapse
Affiliation(s)
- Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Hanieh Taheri
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Keith T. Schmidt
- Clinical Pharmacology Program, Office of the Clinical Director, National Cancer Institute, Bethesda, ML 20892, USA
| | - Alice A. Gibson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Stefan A. J. Buck
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Eric D. Eisenmann
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - William D. Figg
- Clinical Pharmacology Program, Office of the Clinical Director, National Cancer Institute, Bethesda, ML 20892, USA
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Division of Outcomes and Translational Sciences, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-685-8028
| |
Collapse
|
11
|
Morita T, Akiyoshi T, Tsuchitani T, Kataoka H, Araki N, Yajima K, Katayama K, Imaoka A, Ohtani H. Inhibitory Effects of Cranberry Juice and Its Components on Intestinal OATP1A2 and OATP2B1: Identification of Avicularin as a Novel Inhibitor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3310-3320. [PMID: 35230114 DOI: 10.1021/acs.jafc.2c00065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Organic anion-transporting polypeptide (OATP) 1A2 and OATP2B1 mediate the intestinal absorption of drugs. This study aimed to identify fruit juices or fruit juice components that inhibit OATPs and assess the risk of associated food-drug interactions. Inhibitory potency was assessed by examining the uptake of [3H]estrone 3-sulfate and [3H]fexofenadine into HEK293 cells expressing OATP1A2 or OATP2B1. In vivo experiments were conducted using mice to evaluate the effects of cranberry juice on the pharmacokinetics of orally administered fexofenadine. Of eight examined fruit juices, cranberry juice inhibited the functions of both OATPs most potently. Avicularin, a component of cranberry juice, was identified as a novel OATP inhibitor. It exhibited IC50 values of 9.0 and 37 μM for the inhibition of estrone 3-sulfate uptake mediated by OATP1A2 and OATP2B1, respectively. A pharmacokinetic experiment revealed that fexofenadine exposure was significantly reduced (by 50%) by cranberry juice. Cranberry juice may cause drug interactions with OATP substrates.
Collapse
Affiliation(s)
- Tokio Morita
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Takeshi Akiyoshi
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Toshiaki Tsuchitani
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Hiroki Kataoka
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Naoya Araki
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Kodai Yajima
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Kazuhiro Katayama
- School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi, Chiba 274-8555, Japan
| | - Ayuko Imaoka
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Hisakazu Ohtani
- Graduate School of Pharmaceutical Sciences, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| |
Collapse
|
12
|
F. Martins ML, Heydari P, Li W, Martínez-Chávez A, Venekamp N, Lebre MC, Lucas L, Beijnen JH, Schinkel AH. Drug Transporters ABCB1 (P-gp) and OATP, but not Drug-Metabolizing Enzyme CYP3A4, Affect the Pharmacokinetics of the Psychoactive Alkaloid Ibogaine and its Metabolites. Front Pharmacol 2022; 13:855000. [PMID: 35308219 PMCID: PMC8931498 DOI: 10.3389/fphar.2022.855000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The psychedelic alkaloid ibogaine is increasingly used as an oral treatment for substance use disorders, despite being unlicensed in most countries and having reported adverse events. Using wild-type and genetically modified mice, we investigated the impact of mouse (m)Abcb1a/1b and Abcg2 drug efflux transporters, human and mouse OATP drug uptake transporters, and the CYP3A drug-metabolizing complex on the pharmacokinetics of ibogaine and its main metabolites. Following oral ibogaine administration (10 mg/kg) to mice, we observed a rapid and extensive conversion of ibogaine to noribogaine (active metabolite) and noribogaine glucuronide. Mouse Abcb1a/1b, in combination with mAbcg2, modestly restricted the systemic exposure (plasma AUC) and peak plasma concentration (Cmax) of ibogaine. Accordingly, we found a ∼2-fold decrease in the relative recovery of ibogaine in the small intestine with fecal content in the absence of both transporters compared to the wild-type situation. Ibogaine presented good intrinsic brain penetration even in wild-type mice (brain-to-plasma ratio of 3.4). However, this was further increased by 1.5-fold in Abcb1a/1b;Abcg2−/− mice, but not in Abcg2−/− mice, revealing a stronger effect of mAbcb1a/1b in restricting ibogaine brain penetration. The studied human OATP transporters showed no major impact on ibogaine plasma and tissue disposition, but the mOatp1a/1b proteins modestly affected the plasma exposure of ibogaine metabolites and the tissue disposition of noribogaine glucuronide. No considerable role of mouse Cyp3a knockout or transgenic human CYP3A4 overexpression was observed in the pharmacokinetics of ibogaine and its metabolites. In summary, ABCB1, in combination with ABCG2, limits the oral availability of ibogaine, possibly by mediating its hepatobiliary and/or direct intestinal excretion. Moreover, ABCB1 restricts ibogaine brain penetration. Variation in ABCB1/ABCG2 activity due to genetic variation and/or pharmacologic inhibition might therefore affect ibogaine exposure in patients, but only to a limited extent. The insignificant impact of human CYP3A4 and OATP1B1/1B3 transporters may be clinically advantageous for ibogaine and noribogaine use, as it decreases the risks of undesirable drug interactions or interindividual variation related to CYP3A4 and/or OATP activity.
Collapse
Affiliation(s)
| | - Paniz Heydari
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Wenlong Li
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alejandra Martínez-Chávez
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Nikkie Venekamp
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria C. Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Luc Lucas
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jos H. Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alfred H. Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- *Correspondence: Alfred H. Schinkel,
| |
Collapse
|
13
|
Martínez-Chávez A, Broeders J, Lebre MC, Tibben MT, Rosing H, Beijnen JH, Schinkel AH. The role of drug efflux and uptake transporters ABCB1 (P-gp), ABCG2 (BCRP) and OATP1A/1B and of CYP3A4 in the pharmacokinetics of the CDK inhibitor milciclib. Eur J Pharm Sci 2021; 159:105740. [PMID: 33524505 DOI: 10.1016/j.ejps.2021.105740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/06/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
The promising anticancer drug milciclib potently inhibits cyclin-dependent kinase (CDK) 2 and tropomyosin receptor kinase (TRK) A, and is currently in phase II clinical studies. To characterize factors affecting milciclib pharmacokinetics, we investigated whether milciclib is a substrate of the multidrug efflux and uptake transporters ABCB1 (P-gp), ABCG2 (BCRP), and OATP1A/1B, and the drug-metabolizing enzyme CYP3A, using genetically-modified mouse models and Madin-Darby Canine Kidney (MDCK-II) cells. In vitro, milciclib was transported by mAbcg2, and this was inhibited by the ABCG2 inhibitor Ko143. Upon oral administration of milciclib, its plasma exposure in Abcb1a/1b-/-, Abcg2-/-, and Abcb1a/1b;Abcg2-/- mice was similar to that found in wild-type mice. Milciclib showed good brain penetration even in wild-type mice (brain-to-plasma ratio of 1.2), but this was further increased by 5.2-fold when both Abcb1 and Abcg2 were ablated, and to a lesser extent in single Abcb1- or Abcg2-deficient mice. Oatp1a/1b deficiency had only a minor impact on the milciclib plasma AUC0-24h and Cmax. The milciclib AUC0-8h increased 1.9-fold in Cyp3a-/- mice but decreased only 1.3-fold upon overexpression of human CYP3A4. Thus, ABCB1 and ABCG2 cooperatively limit milciclib brain penetration. The low impact of OATP1 and CYP3A could be clinically favorable for milciclib, reducing the risks of unintended drug-drug interactions or interindividual variation in CYP3A4 activity.
Collapse
Affiliation(s)
- Alejandra Martínez-Chávez
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jelle Broeders
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Matthijs T Tibben
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Wang YK, Lee WP, Wang YW, Huang YH, Hou MC, Chang YL, Lan KH. Precipitating factors causing hyperbilirubinemia during chronic hepatitis C treatment with paritaprevir/ritonavir/ombitasvir and dasabuvir. J Chin Med Assoc 2020; 83:1071-1078. [PMID: 33273269 DOI: 10.1097/jcma.0000000000000429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Hepatic decompensation is a fatal on-treatment side effect during chronic hepatitis C treatment with paritaprevir/ritonavir/ombitasvir and dasabuvir (PrOD). Prompt bilirubin testing can reveal hepatic failure in susceptible patients, and clinical parameters precipitating early elevation of bilirubin can warn clinicians to avoid PrOD prescription. METHODS This retrospective study included 169 Hepatitis C virus (HCV)-genotype 1b patients who underwent a 12-week course of PrOD with or without ribavirin. Laboratory data underwent χ analysis with Fisher's exact test to determine the precipitating factors causing hyperbilirubinemia in patients who had received 1 week of treatment. RESULTS Sustained viral response was achieved in 164 patients (97.0%). Total bilirubin was ≥2 mg/dL (21.3%) in 36 patients after 1 week of treatment. Pretreatment white blood cell (WBC) <4500/µL and platelet <100,000/µL correlated with total bilirubin ≥2 mg/dL (relative risk [RR]: 21.64, 95% CI: 5.23-89.64, p < 0.001) after 1 week of treatment. Pretreatment platelet ≥100 000/µL and WBC <4500/µL correlated with direct bilirubin ≥0.45 mg/dL (RR: 6.56, 95% CI: 1.42-30.38, p = 0.016) and indirect bilirubin ≥0.6 mg/dL (RR: 4.77, 95% CI: 1.03-22.15, p = 0.046). Pretreatment platelet <100,000/µL with F3/F4 fibrosis correlated with first week total bilirubin ≥2 mg/dL (RR: 3.57, 95% CI: 1.35-9.09, p = 0.010). CONCLUSION PrOD is an effective antiviral regimen for HCV genotype 1b patients. Total bilirubin ≥2 mg/dL after 1 week of treatment serves as an early warning of irreversible progression toward hepatic decompensation, and the current study provides a guide by which to monitor chronic hepatitis C patients undergoing PrOD treatment.
Collapse
Affiliation(s)
- Yi-Kai Wang
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Pharmacy, National Yang-Ming University Hospital, Yilan, Taiwan, ROC
| | - Wei-Ping Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ying-Wen Wang
- Healthcare Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Yuh-Lih Chang
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Keng-Hsin Lan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
15
|
Bile Acids: A Communication Channel in the Gut-Brain Axis. Neuromolecular Med 2020; 23:99-117. [PMID: 33085065 DOI: 10.1007/s12017-020-08625-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Bile acids are signalling hormones involved in the regulation of several metabolic pathways. The ability of bile acids to bind and signal through their receptors is modulated by the gut microbiome, since the microbiome contributes to the regulation and synthesis of bile acids as well to their physiochemical properties. From the gut, bacteria have been shown to send signals to the central nervous system via their metabolites, thus affecting the behaviour and brain function of the host organism. In the last years it has become increasingly evident that bile acids affect brain function, during normal physiological and pathological conditions. Although bile acids may be synthesized locally in the brain, the majority of brain bile acids are taken up from the systemic circulation. Since the composition of the brain bile acid pool may be regulated by the action of intestinal bacteria, it is possible that bile acids function as a communication bridge between the gut microbiome and the brain. However, little is known about the molecular mechanisms and the physiological roles of bile acids in the central nervous system. The possibility that bile acids may be a direct link between the intestinal microbiome and the brain is also an understudied subject. Here we review the influence of gut bacteria on the bile acid pool composition and properties, as well as striking evidence showing the role of bile acids as neuroactive molecules.
Collapse
|
16
|
Li H, Rabearivony A, Zhang W, Chen S, An X, Liu C. Chronopharmacology of simvastatin on hyperlipidaemia in high-fat diet-fed obese mice. J Cell Mol Med 2020; 24:11024-11029. [PMID: 32767644 PMCID: PMC7521315 DOI: 10.1111/jcmm.15709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/27/2020] [Accepted: 07/14/2020] [Indexed: 01/16/2023] Open
Abstract
The chronopharmacology refers to the utilization of physiological circadian rhythms to optimize the administration time of drugs, thus increasing their efficacy and safety, or reducing adverse effects. Simvastatin is one of the most widely prescribed drugs for the treatment of hypercholesterolaemia, hyperlipidemia and coronary artery disease. There are conflicting statements regarding the timing of simvastatin administration, and convincing experimental evidence remains unavailable. Thus, we aimed to examine whether different administration times would influence the efficacy of simvastatin. High‐fat diet‐fed mice were treated with simvastatin at zeitgeber time 1 (ZT1) or ZT13, respectively, for nine weeks. Simvastatin showed robust anti‐hypercholesterolaemia and anti‐hyperlipidemia effects on these obese mice, regardless of administration time. However, simvastatin administrated at ZT13, compared to ZT1, was more functional for decreasing serum levels of total cholesterol, triglycerides, non‐esterified free fatty acids and LDL cholesterol, as well as improving liver pathological characteristics. In terms of possible mechanisms, we found that simvastatin did not alter the expression of hepatic circadian clock gene in vivo, although it failed to change the period, phase and amplitude of oscillation patterns in Per2::Luc U2OS and Bmal1::Luc U2OS cells in vitro. In contrast, simvastatin regulated the expression of Hmgcr, Mdr1 and Slco2b1 in a circadian manner, which potentially contributed to the chronopharmacological function of the drug. Taken together, we provide solid evidence to suggest that different administration times affect the lipid‐lowering effects of simvastatin.
Collapse
Affiliation(s)
- Huan Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, China
| | - Anjara Rabearivony
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, China
| | - Siyu Chen
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofei An
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chang Liu
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
17
|
Wang Y, Sparidans RW, Li W, Lebre MC, Beijnen JH, Schinkel AH. OATP1A/1B, CYP3A, ABCB1, and ABCG2 limit oral availability of the NTRK inhibitor larotrectinib, while ABCB1 and ABCG2 also restrict its brain accumulation. Br J Pharmacol 2020; 177:3060-3074. [PMID: 32087611 DOI: 10.1111/bph.15034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/14/2020] [Accepted: 02/14/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Larotrectinib is a FDA-approved oral small-molecule inhibitor for treatment of neurotrophic tropomyosin receptor kinase fusion-positive cancer. We here investigated the functions of the multidrug efflux transporters ABCB1 and ABCG2, the SLCO1A/1B (OATP1A/1B) uptake transporters, and the multispecific drug-metabolizing enzyme CYP3A in larotrectinib pharmacokinetic behaviour. EXPERIMENTAL APPROACH In vitro, transepithelial drug transport and uptake assays were performed. In vivo, larotrectinib (10 mg·kg-1 ) was administered orally to relevant genetically modified mouse models. Cell medium, plasma samples, and organ homogenates were measured by a sensitive and specific LC-MS/MS larotrectinib assay. KEY RESULTS In vitro, larotrectinib was avidly transported by human (h) ABCB1 and mouse (m) Abcg2 efficiently by hABCG2 and modestly by hOATP1A2. In vivo, both mAbcb1a/1b and mAbcg2 markedly limited larotrectinib oral availability and brain and testis accumulation (by 2.1-fold, 10.4-fold, and 2.7-fold, respectively), with mAbcb1a/1b playing a more prominent role. mOatp1a/1b also restricted larotrectinib oral availability (by 3.8-fold) and overall tissue exposure, apparently by mediating substantial uptake into the liver, thus likely facilitating hepatobiliary excretion. Additionally, larotrectinib is an excellent substrate of CYP3A, which restricts the oral availability of larotrectinib and hence its tissue exposure. CONCLUSIONS AND IMPLICATIONS ABCG2 and especially ABCB1 limit the oral availability and brain and testis penetration of larotrectinib, while OATP1A/1B transporters restrict its systemic exposure by mediating hepatic uptake, thus allowing hepatobiliary excretion. CYP3A-mediated metabolism can strongly limit larotrectinib oral availability and hence its tissue concentrations. These insights may be useful in the further clinical development of larotrectinib.
Collapse
Affiliation(s)
- Yaogeng Wang
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rolf W Sparidans
- Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Wenlong Li
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maria C Lebre
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, The Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alfred H Schinkel
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Hulin A, Stocco J, Bouattour M. Clinical Pharmacokinetics and Pharmacodynamics of Transarterial Chemoembolization and Targeted Therapies in Hepatocellular Carcinoma. Clin Pharmacokinet 2019; 58:983-1014. [PMID: 31093928 DOI: 10.1007/s40262-019-00740-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The management of hepatocellular carcinoma (HCC) is based on a multidisciplinary decision tree. Treatment includes loco-regional therapy, mainly transarterial chemoembolization, for intermediate-stage HCC and systemic therapy with oral tyrosine kinase inhibitors (TKIs) for advanced HCC. Transarterial chemoembolization involves hepatic intra-arterial infusion with either conventional procedure or drug-eluting-beads. The aim of the loco-regional procedure is to deliver treatment as close as possible to the tumor both to embolize the tumor area and to enhance efficacy and minimize systemic toxicity of the anticancer drug. Pharmacokinetic studies applied to transarterial chemoembolization are rare and pharmacodynamic studies even rarer. However, all available studies lead to the same conclusions: use of the transarterial route lowers systemic exposure to the cytotoxic drug and leads to much higher tumor drug concentrations than does a similar dose via the intravenous route. However, reproducibility of the procedure remains a major problem, and no consensus exists regarding the choice of anticancer drug and its dosage. Systemic therapy with TKIs is based on sorafenib and lenvatinib as first-line treatment and regorafenib and cabozantinib as second-line treatment. Clinical use of TKIs is challenging because of their complex pharmacokinetics, with high liver metabolism yielding both active metabolites and their common toxicities. Changes in liver function over time with the progression of HCC adds further complexity to the use of TKIs. The challenges posed by TKIs and the HCC disease process means monitoring of TKIs is required to improve clinical management. To date, only partial data supporting sorafenib monitoring is available. Results from further pharmacokinetic/pharmacodynamic studies of these four TKIs are eagerly awaited and are expected to permit such monitoring and the development of consensus guidelines.
Collapse
Affiliation(s)
- Anne Hulin
- APHP, Laboratory of Pharmacology, GH Henri Mondor, EA7375, University Paris Est Creteil, 94010, Creteil, France
| | - Jeanick Stocco
- APHP, HUPNVS, Department of Clinical Pharmacy and Pharmacology, Beaujon University Hospital, 92110, Clichy, France
| | - Mohamed Bouattour
- APHP, HUPNVS, Department of Digestive Oncology, Beaujon University Hospital, 92110, Clichy, France.
| |
Collapse
|
19
|
Schulte RR, Ho RH. Organic Anion Transporting Polypeptides: Emerging Roles in Cancer Pharmacology. Mol Pharmacol 2019; 95:490-506. [PMID: 30782852 PMCID: PMC6442320 DOI: 10.1124/mol.118.114314] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/09/2019] [Indexed: 12/13/2022] Open
Abstract
The organic anion transporting polypeptides (OATPs) are a superfamily of drug transporters involved in the uptake and disposition of a wide array of structurally divergent endogenous and exogenous substrates, including steroid hormones, bile acids, and commonly used drugs, such as anti-infectives, antihypertensives, and cholesterol lowering agents. In the past decade, OATPs, primarily OATP1A2, OATP1B1, and OATP1B3, have emerged as potential mediators of chemotherapy disposition, including drugs such as methotrexate, doxorubicin, paclitaxel, docetaxel, irinotecan and its important metabolite 7-ethyl-10-hydroxycamptothecin, and certain tyrosine kinase inhibitors. Furthermore, OATP family members are polymorphic and numerous studies have shown OATP variants to have differential uptake, disposition, and/or pharmacokinetics of numerous drug substrates with important implications for interindividual differences in efficacy and toxicity. Additionally, certain OATPs have been found to be overexpressed in a variety of human solid tumors, including breast, liver, colon, pancreatic, and ovarian cancers, suggesting potential roles for OATPs in tumor development and progression and as novel targets for cancer therapy. This review focuses on the emerging roles for selected OATPs in cancer pharmacology, including preclinical and clinical studies suggesting roles in chemotherapy disposition, the pharmacogenetics of OATPs in cancer therapy, and OATP overexpression in various tumor tissues with implications for OATPs as therapeutic targets.
Collapse
Affiliation(s)
- Rachael R Schulte
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Richard H Ho
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
20
|
Fu Q, Chen M, Anderson JT, Sun X, Hu S, Sparreboom A, Baker SD. Interaction Between Sex and Organic Anion-Transporting Polypeptide 1b2 on the Pharmacokinetics of Regorafenib and Its Metabolites Regorafenib-N-Oxide and Regorafenib-Glucuronide in Mice. Clin Transl Sci 2019; 12:400-407. [PMID: 30955241 PMCID: PMC6662550 DOI: 10.1111/cts.12630] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/02/2019] [Indexed: 12/27/2022] Open
Abstract
Regorafenib, a multikinase inhibitor used in the treatment of various solid tumors, undergoes extensive uridine 5′‐diphosphate glucuronosyltransferase (Ugt)1a9‐mediated glucuronidation to form regorafenib‐N‐β‐glucuronide (M7; RG), but the contribution of hepatic uptake transporters, such as organic anion‐transporting polypeptide (Oatp)1b2, to the pharmacokinetics of regorafenib remains poorly understood. Using NONMEM‐based, population‐based, parent‐metabolite modeling, we found that Oatp1b2 and sex strongly impact the systemic exposure to RG in mice receiving oral regorafenib. Metabolic studies revealed that the liver microsomal expression of cytochrome P450 (Cyp)3a11 is twofold lower in female mice, whereas Ugt1a9 levels and function are not sex dependent. This finding is consistent with the metabolism of regorafenib occurring via two competing pathways, and the lack of Oatp1b2 results in decreased clearance of RG. The described model provides mechanistic insights into the in vivo disposition of regorafenib.
Collapse
Affiliation(s)
- Qiang Fu
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Mingqing Chen
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jason T Anderson
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Xinxin Sun
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmaceutical Chemistry and Comprehensive Cancer Center, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Chen L, Yang H, Yu C, Yuan M, Li H. High hepatic exposure of furanocoumarins in Radix Angelica dahuricae is associated with transporter mediated active uptake. JOURNAL OF ETHNOPHARMACOLOGY 2018; 212:74-85. [PMID: 29055720 DOI: 10.1016/j.jep.2017.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Angelica dahuricae (RAD), the roots of Angelica dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav, is a well-known traditional Chinese medicine (TCM) and has been used for centuries to treat headaches, toothaches, nose congestion, abscesses, furunculoses, and acne. This herb is also one of frequently reported TCMs showing the herb-drug interaction potential. Furanocoumarins are main bioactive components of RAD. AIM OF THE STUDY This study is designed to characterize the tissue distribution profiles of furanocoumarins after oral administration of RAD extract in rats and to explore the mechanism underlying the high hepatic exposure of the major furanocoumarins. MATERIALS AND METHODS The tissue distribution of nine furanocoumarins was determined in rats after an oral dose of 0.46g/kg RAD extract using high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Unbound fractions (ƒu) of major furanocoumarins, including imperatorin (IM), isoimperatorin (IIM), bergapten (BER) and oxypeucedanin hydrate (OXYH), were measured in rat plasma and selected tissue homogenates (liver, kidney, lung and brain) with Rapid Equilibrium Dialysis (RED) method. The temperature dependent hepatic uptake of IM, IIM, BER and OXYH were evaluated in suspended rat primary hepatocytes at 4°C or 37°C by the oil-spin method. The uptake kinetics was conducted in the cells over a wide concentration range. The furanocoumarins were co-incubated with a panel of transporter inhibitors to investigate the involvement of uptake transporters in the hepatic uptake. The transcellular transport characteristics of IM, IIM, BER and OXYH were further assessed using Caco-2 cell monolayer model. RESULTS IM, IIM, BER and OXYH were found to be the major bioactive furanocoumarins in rat plasma and tissues, representing more than 90% exposure for all the detected furanocoumarins. The most concentrative organ of major furanocoumarins was the liver, with liver-to-plasma exposure ratio (Kp,AUC) of 5.1, 6.5 and 4.7 for IM, IIM and BER, and 2.3 for OXYH, respectively. IM, IIM and BER also showed higher concentrations in the kidney with Kp above 2.2. The higher protein binding of the furanocoumarins partially contributed to their higher tissue exposure. In suspended rat primary hepatocyte, the hepatic uptake of IM, IIM, BER and OXYH was temperature-dependent, with considerably higher uptake at 37°C than at 4°C. Uptake kinetics indicated that the hepatic uptake of IM, IIM, BER and OXYH involved both active transport and passive diffusion processes. For IM, IIM and BER, the contribution of the active transport was greater than the passive process, with the CLactive/CLuptake > 72%. Ritonavir (RTN) and cyclosporine A (CsA), the known inhibitors of organic anion transporting polypeptide (Oatp) significantly inhibited the hepatic uptake of IM and BER, while the inhibitor of the organic anion transporters (Oat) probenecid (PBC) remarkably reduced IIM uptake. In the Caco-2 cell model, the furanocoumarins were highly permeable in the apical to basolateral direction without notable active efflux. CONCLUSION The furanocoumarins rapidly and widely distributed into various tissues after oral dose of the RAD extract. IM, IIM, BER and OXYH were the major components detected in both plasma and tissues. Liver was the most distributed tissue of the total and free furanocoumarins. Non-specific protein binding contributed partially to the higher tissue exposures of these bioactive components. The Oatp and Oat mediated active uptake played the primal role in the high hepatic exposure of the furanocoumarins.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Haiying Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Chenchen Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Mei Yuan
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Hua Li
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| |
Collapse
|
22
|
Functional shift with maintained regenerative potential following portal vein ligation. Sci Rep 2017; 7:18065. [PMID: 29273725 PMCID: PMC5741735 DOI: 10.1038/s41598-017-18309-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/08/2017] [Indexed: 02/08/2023] Open
Abstract
Selective portal vein ligation (PVL) allows the two-stage surgical resection of primarily unresectable liver tumours by generating the atrophy and hypertrophy of portally ligated (LL) and non-ligated lobes (NLL), respectively. To evaluate critically important underlying functional alterations, present study characterised in vitro and vivo liver function in male Wistar rats (n = 106; 210-250 g) before, and 24/48/72/168/336 h after PVL. Lobe weights and volumes by magnetic resonance imaging confirmed the atrophy-hypertrophy complex. Proper expression and localization of key liver transporters (Ntcp, Bsep) and tight junction protein ZO-1 in isolated hepatocytes demonstrated constantly present viable and well-polarised cells in both lobes. In vitro taurocholate and bilirubin transport, as well as in vivo immunohistochemical Ntcp and Mrp2 expressions were bilaterally temporarily diminished, whereas LL and NLL structural acinar changes were divergent. In vivo bile and bilirubin-glucuronide excretion mirrored macroscopic changes, whereas serum bilirubin levels remained unaffected. In vivo functional imaging (indocyanine-green clearance test; 99mTc-mebrofenin hepatobiliary scintigraphy; confocal laser endomicroscopy) indicated transitionally reduced global liver uptake and -excretion. While LL functional involution was permanent, NLL uptake and excretory functions recovered excessively. Following PVL, functioning cells remain even in LL. Despite extensive bilateral morpho-functional changes, NLL functional increment restores temporary declined transport functions, emphasising liver functional assessment.
Collapse
|
23
|
Kim M, Deacon P, Tirona RG, Kim RB, Pin CL, Meyer zu Schwabedissen HE, Wang R, Schwarz UI. Characterization of OATP1B3 and OATP2B1 transporter expression in the islet of the adult human pancreas. Histochem Cell Biol 2017; 148:345-357. [DOI: 10.1007/s00418-017-1580-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
|
24
|
Paritaprevir and Ritonavir Liver Concentrations in Rats as Assessed by Different Liver Sampling Techniques. Antimicrob Agents Chemother 2017; 61:AAC.02283-16. [PMID: 28264852 DOI: 10.1128/aac.02283-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/12/2017] [Indexed: 12/28/2022] Open
Abstract
The liver is crucial to pharmacology, yet substantial knowledge gaps exist in the understanding of its basic pharmacologic processes. An improved understanding for humans requires reliable and reproducible liver sampling methods. We compared liver concentrations of paritaprevir and ritonavir in rats by using samples collected by fine-needle aspiration (FNA), core needle biopsy (CNB), and surgical resection. Thirteen Sprague-Dawley rats were evaluated, nine of which received paritaprevir/ritonavir at 30/20 mg/kg of body weight by oral gavage daily for 4 or 5 days. Drug concentrations were measured using liquid chromatography-tandem mass spectrometry on samples collected via FNA (21G needle) with 1, 3, or 5 passes (FNA1, FNA3, and FNA5); via CNB (16G needle); and via surgical resection. Drug concentrations in plasma were also assessed. Analyses included noncompartmental pharmacokinetic analysis and use of Bland-Altman techniques. All liver tissue samples had higher paritaprevir and ritonavir concentrations than those in plasma. Resected samples, considered the benchmark measure, resulted in estimations of the highest values for the pharmacokinetic parameters of exposure (maximum concentration of drug in serum [Cmax] and area under the concentration-time curve from 0 to 24 h [AUC0-24]) for paritaprevir and ritonavir. Bland-Altman analyses showed that the best agreement occurred between tissue resection and CNB, with 15% bias, followed by FNA3 and FNA5, with 18% bias, and FNA1 and FNA3, with a 22% bias for paritaprevir. Paritaprevir and ritonavir are highly concentrated in rat liver. Further research is needed to validate FNA sampling for humans, with the possible derivation and application of correction factors for drug concentration measurements.
Collapse
|
25
|
Wang X, Zheng L, Wu J, Tang B, Zhang M, Zhu D, Lin X. Constitutive androstane receptor activation promotes bilirubin clearance in a murine model of alcoholic liver disease. Mol Med Rep 2017; 15:3459-3466. [PMID: 28393244 PMCID: PMC5436297 DOI: 10.3892/mmr.2017.6435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 11/29/2016] [Indexed: 12/20/2022] Open
Abstract
Increased plasma levels of bilirubin have been reported in rat models and patients with alcoholic liver disease (ALD). The constitutive androstane receptor (CAR) is a known xenobiotic receptor, which induces the detoxification and transport of bilirubin. In the present study, the bilirubin transport regulatory mechanisms, and the role of CAR activation in hepatic and extrahepatic bilirubin clearance were investigated in a murine model of ALD. The mice were fed a Lieber-DeCarli ethanol diet or an isocaloric control diet for 4 weeks, followed by the administration of CAR agonists, 1,4-bis-[2-(3,5-dichlorpyridyloxy)]benzene (TCPOBOP) and phenobarbital (PB), and their vehicles to examine the effect of the pharmacological activation of CAR on serum levels of bilirubin and on the bilirubin clearance pathway in ALD by serological survey, western blotting and reverse transcription-quantitative polymerase chain reaction. The results showed that chronic ethanol ingestion impaired the nuclear translocation of CAR, which was accompanied by elevated serum levels of bilirubin, suppression of the expression of hepatic and renal organic anion transporting polypeptide (OATP) 1A1 and hepatic multidrug resistance-associated protein 2 (MRP2), and induction of the expression of UDP-glucuronosyltransferase (UGT) 1A1. The activation of CAR by TCPOBOP and PB resulted in downregulation of the serum levels of bilirubin followed by selective upregulation of the expression levels of OATP1A1, OATP1A4, UGT1A1 and MRP2 in ALD. These results revealed the bilirubin transport regulatory mechanisms and highlighted the importance of CAR in modulating the bilirubin clearance pathway in the ALD mouse model.
Collapse
Affiliation(s)
- Xiuyan Wang
- Department of Gastroenterology, Wenzhou People's Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Liyu Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jinming Wu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Binbin Tang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Mengqin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Debin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xianfan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
26
|
Affiliation(s)
- Eleni Kotsampasakou
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| | - Gerhard F. Ecker
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| |
Collapse
|
27
|
Durmus S, van Hoppe S, Schinkel AH. The impact of Organic Anion-Transporting Polypeptides (OATPs) on disposition and toxicity of antitumor drugs: Insights from knockout and humanized mice. Drug Resist Updat 2016; 27:72-88. [PMID: 27449599 DOI: 10.1016/j.drup.2016.06.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 05/07/2016] [Accepted: 06/17/2016] [Indexed: 12/12/2022]
Abstract
It is now widely accepted that organic anion-transporting polypeptides (OATPs), especially members of the OATP1A/1B family, can have a major impact on the disposition and elimination of a variety of endogenous molecules and drugs. Owing to their prominent expression in the sinusoidal plasma membrane of hepatocytes, OATP1B1 and OATP1B3 play key roles in the hepatic uptake and plasma clearance of a multitude of structurally diverse anti-cancer and other drugs. Here, we present a thorough assessment of the currently available OATP1A and OATP1B knockout and transgenic mouse models as key tools to study OATP functions in vivo. We discuss recent studies using these models demonstrating the importance of OATPs, primarily in the plasma and hepatic clearance of anticancer drugs such as taxanes, irinotecan/SN-38, methotrexate, doxorubicin, and platinum compounds. We further discuss recent work on OATP-mediated drug-drug interactions in these mouse models, as well as on the role of OATP1A/1B proteins in the phenomenon of hepatocyte hopping, an efficient and flexible way of liver detoxification for both endogenous and exogenous substrates. Interestingly, glucuronide conjugates of both the heme breakdown product bilirubin and the protein tyrosine kinase-targeted anticancer drug sorafenib are strongly affected by this process. The clinical relevance of variation in OATP1A/1B activity in patients has been previously revealed by the effects of polymorphic variants and drug-drug interactions on drug toxicity. The development of in vivo tools to study OATP1A/1B functions has greatly advanced our mechanistic understanding of their functional role in drug pharmacokinetics, and their implications for therapeutic efficacy and toxic side effects of anticancer and other drug treatments.
Collapse
Affiliation(s)
- Selvi Durmus
- Bilkent University, Department of Molecular Biology and Genetics, 06800 Bilkent, Ankara, Turkey
| | - Stéphanie van Hoppe
- The Netherlands Cancer Institute, Division of Molecular Oncology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Molecular Oncology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Wanek T, Halilbasic E, Visentin M, Mairinger S, Römermann K, Stieger B, Kuntner C, Müller M, Langer O, Trauner M. Influence of 24-Nor-Ursodeoxycholic Acid on Hepatic Disposition of [(18)F]Ciprofloxacin, a Positron Emission Tomography Study in Mice. J Pharm Sci 2016; 105:106-12. [PMID: 26852845 DOI: 10.1016/j.xphs.2015.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 10/28/2015] [Accepted: 11/05/2015] [Indexed: 01/10/2023]
Abstract
24-nor-ursodeoxycholic acid (norUDCA) is a novel therapeutic approach to cholestatic liver diseases. In mouse models of cholestasis, norUDCA induces basolateral multidrug resistance-associated proteins 4 (Mrp4) and 3 in hepatocytes, which provide alternative escape routes for bile acids accumulating during cholestasis but could also result in altered hepatic disposition of concomitantly administered substrate drugs. We used positron emission tomography imaging to study the influence of norUDCA on hepatic disposition of the model Mrp4 substrate [(18)F]ciprofloxacin in wild-type and Mdr2((-/-)) mice, a model of cholestasis. Animals underwent [(18)F]ciprofloxacin positron emission tomography at baseline and after norUDCA treatment. After norUDCA treatment, liver-to-blood area under the curve ratio of [(18)F]ciprofloxacin was significantly decreased compared to baseline, both in wild-type (-34.0 ± 2.1%) and Mdr2((-/-)) mice (-20.5 ± 6.0%). [(18)F]Ciprofloxacin uptake clearance from blood into liver was reduced by -17.1 ± 9.0% in wild-type and by -20.1 ± 7.3% in Mdr2((-/-)) mice. Real-time PCR analysis showed significant increases in hepatic Mrp4 and multidrug resistance-associated protein 3 mRNA after norUDCA. Transport experiments in organic anion transporting polypeptide (OATP)1B1-, OATP1B3-, and OATP2B1-transfected cells revealed weak transport of [(14)C]ciprofloxacin by OATP1B3 and OATP2B1 and no inhibition by norUDCA. In conclusion, our data suggest that changes in hepatic [(18)F]ciprofloxacin disposition in mice after norUDCA treatment were caused by induction of basolateral Mrp4 in hepatocytes.
Collapse
Affiliation(s)
- Thomas Wanek
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Emina Halilbasic
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Severin Mairinger
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Center for Systems Neuroscience, Hannover, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Claudia Kuntner
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Oliver Langer
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Austria.
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
29
|
Fujita D, Saito Y, Nakanishi T, Tamai I. Organic Anion Transporting Polypeptide (OATP)2B1 Contributes to Gastrointestinal Toxicity of Anticancer Drug SN-38, Active Metabolite of Irinotecan Hydrochloride. Drug Metab Dispos 2016; 44:1-7. [PMID: 26526067 DOI: 10.1124/dmd.115.066712] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/30/2015] [Indexed: 11/22/2022] Open
Abstract
Gastrointestinal toxicity, such as late-onset diarrhea, is a significant concern in irinotecan hydrochloride (CPT-11)-containing regimens. Prophylaxis of late-onset diarrhea has been reported with use of Japanese traditional (Kampo) medicine containing baicalin and with the antibiotic cefixime, and this has been explained in terms of inhibition of bacterial deconjugation of SN-38-glucuronide since unconjugated SN-38 (active metabolite of CPT-11) is responsible for the gastrointestinal toxicity. It is also prerequisite for SN-38 to be accumulated in intestinal tissues to exert toxicity. Based on the fact that liver-specific organic anion transporting polypeptide (OATP)1B1, a member of the same family as OATP2B1, is known to be involved in hepatic transport of SN-38, we hypothesized that intestinal transporter OATP2B1 contributes to the accumulation of SN-38 in gastrointestinal tissues, and its inhibition would help prevent associated toxicity. We found that uptake of SN-38 by OATP2B1-expressing Xenopus oocytes was significantly higher than that by control oocytes. OATP2B1-mediated uptake of SN-38 was saturable, pH dependent, and decreased in the presence of baicalin, cefixime, or fruit juices such as apple juice. In vivo gastrointestinal toxicity of SN-38 in mice caused by oral administration for consecutive 5 days was prevented by coingestion of apple juice. Thus, OATP2B1 contributes to the uptake of SN-38 by intestinal tissues, triggering gastrointestinal toxicity. So, in addition to the reported inhibition of bacterial β-glucuronidase by cefixime or baicalin, inhibition of OATP2B1 may also contribute to prevention of gastrointestinal toxicity. Apple juice may be helpful for prophylaxis of late-onset diarrhea observed in CPT-11 therapy without disturbance of the intestinal microflora.
Collapse
Affiliation(s)
- Daichi Fujita
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (D.F., Y.S., T.N., I.T.), and Department of Pharmacy, National Cancer Center Hospital, Tokyo, Japan (Y.S.)
| | - Yoshimasa Saito
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (D.F., Y.S., T.N., I.T.), and Department of Pharmacy, National Cancer Center Hospital, Tokyo, Japan (Y.S.)
| | - Takeo Nakanishi
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (D.F., Y.S., T.N., I.T.), and Department of Pharmacy, National Cancer Center Hospital, Tokyo, Japan (Y.S.)
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan (D.F., Y.S., T.N., I.T.), and Department of Pharmacy, National Cancer Center Hospital, Tokyo, Japan (Y.S.)
| |
Collapse
|
30
|
Kotsampasakou E, Brenner S, Jäger W, Ecker GF. Identification of Novel Inhibitors of Organic Anion Transporting Polypeptides 1B1 and 1B3 (OATP1B1 and OATP1B3) Using a Consensus Vote of Six Classification Models. Mol Pharm 2015; 12:4395-404. [PMID: 26469880 PMCID: PMC4674819 DOI: 10.1021/acs.molpharmaceut.5b00583] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Organic anion transporting polypeptides
1B1 and 1B3 are transporters
selectively expressed on the basolateral membrane of the hepatocyte.
Several studies reveal that they are involved in drug–drug
interactions, cancer, and hyperbilirubinemia. In this study, we developed
a set of classification models for OATP1B1 and 1B3 inhibition based
on more than 1700 carefully curated compounds from literature, which
were validated via cross-validation and by use of an external test
set. After combining several sets of descriptors and classifiers,
the 6 best models were selected according to their statistical performance
and were used for virtual screening of DrugBank. Consensus scoring
of the screened compounds resulted in the selection and purchase of
nine compounds as potential dual inhibitors and of one compound as
potential selective OATP1B3 inhibitor. Biological testing of the compounds
confirmed the validity of the models, yielding an accuracy of 90%
for OATP1B1 and 80% for OATP1B3, respectively. Moreover, at least
half of the new identified inhibitors are associated with hyperbilirubinemia
or hepatotoxicity, implying a relationship between OATP inhibition
and these severe side effects.
Collapse
Affiliation(s)
- Eleni Kotsampasakou
- Department of Pharmaceutical Chemistry, University of Vienna , Althanstrasse 14, 1090 Vienna, Austria
| | - Stefan Brenner
- Department of Pharmaceutical Chemistry, University of Vienna , Althanstrasse 14, 1090 Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Chemistry, University of Vienna , Althanstrasse 14, 1090 Vienna, Austria
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna , Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
31
|
Abstract
Many of the compounds taken up by the liver are organic anions that circulate tightly bound to protein carriers such as albumin. The fenestrated sinusoidal endothelium of the liver permits these compounds to have access to hepatocytes. Studies to characterize hepatic uptake of organic anions through kinetic analyses, suggested that it was carrier-mediated. Attempts to identify specific transporters by biochemical approaches were largely unsuccessful and were replaced by studies that utilized expression cloning. These studies led to identification of the organic anion transport proteins (oatps), a family of 12 transmembrane domain glycoproteins that have broad and often overlapping substrate specificities. The oatps mediate Na(+)-independent organic anion uptake. Other studies identified a seven transmembrane domain glycoprotein, Na(+)/taurocholate transporting protein (ntcp) as mediating Na(+)-dependent uptake of bile acids as well as other organic anions. Although mutations or deficiencies of specific members of the oatp family have been associated with transport abnormalities, there have been no such reports for ntcp, and its physiologic role remains to be determined, although expression of ntcp in vitro recapitulates the characteristics of Na(+)-dependent bile acid transport that is seen in vivo. Both ntcp and oatps traffic between the cell surface and intracellular vesicular pools. These vesicles move through the cell on microtubules, using the microtubule based motors dynein and kinesins. Factors that regulate this motility are under study and may provide a unique mechanism that can alter the plasma membrane content of these transporters and consequently their accessibility to circulating ligands.
Collapse
Affiliation(s)
- Allan W Wolkoff
- The Herman Lopata Chair in Liver Disease Research, Professor of Medicine and Anatomy and Structural Biology, Associate Chair of Medicine for Research, Chief, Division of Gastroenterology and Liver Diseases, Director, Marion Bessin Liver Research Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| |
Collapse
|
32
|
Lee HH, Leake BF, Teft W, Tirona RG, Kim RB, Ho RH. Contribution of hepatic organic anion-transporting polypeptides to docetaxel uptake and clearance. Mol Cancer Ther 2015; 14:994-1003. [PMID: 25695959 DOI: 10.1158/1535-7163.mct-14-0547] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 02/10/2015] [Indexed: 11/16/2022]
Abstract
The antimicrotubular agent docetaxel is a widely used chemotherapeutic drug for the treatment of multiple solid tumors and is predominantly dependent on hepatic disposition. In this study, we evaluated drug uptake transporters capable of transporting radiolabeled docetaxel. By screening an array of drug uptake transporters in HeLa cells using a recombinant vaccinia-based method, five organic anion-transporting polypeptides (OATP) capable of docetaxel uptake were identified: OATP1A2, OATP1B1, OATP1B3, OATP1C1, and Oatp1b2. Kinetic analysis of docetaxel transport revealed similar kinetic parameters among hepatic OATP1B/1b transporters. An assessment of polymorphisms (SNPs) in SLCO1B1 and SLCO1B3 revealed that a number of OATP1B1 and OATP1B3 variants were associated with impaired docetaxel transport. A Transwell-based vectorial transport assay using MDCKII stable cells showed that docetaxel was transported significantly into the apical compartment of double-transfected (MDCKII-OATP1B1/MDR1 and MDCKII-OATP1B3/MDR1) cells compared with single-transfected (MDCKII-OATP1B1 and MDCKII-OATP1B3) cells (P < 0.05) or control (MDCKII-Co) cells (P < 0.001). In vivo docetaxel transport studies in Slco1b2(-/-) mice showed approximately >5.5-fold higher plasma concentrations (P < 0.01) and approximately 3-fold decreased liver-to-plasma ratio (P < 0.05) of docetaxel compared with wild-type (WT) mice. The plasma clearance of docetaxel in Slco1b2(-/-) mice was 83% lower than WT mice (P < 0.05). In conclusion, this study demonstrates the important roles of OATP1B transporters to the hepatic disposition and clearance of docetaxel, and supporting roles of these transporters for docetaxel pharmacokinetics.
Collapse
Affiliation(s)
- Hannah H Lee
- Division of Hematology and Oncology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Brenda F Leake
- Division of Hematology and Oncology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wendy Teft
- Division of Clinical Pharmacology, Schulich School of Medicine and Dentistry, Western University/University of Western Ontario, London, Ontario, Canada
| | - Rommel G Tirona
- Division of Clinical Pharmacology, Schulich School of Medicine and Dentistry, Western University/University of Western Ontario, London, Ontario, Canada
| | - Richard B Kim
- Division of Clinical Pharmacology, Schulich School of Medicine and Dentistry, Western University/University of Western Ontario, London, Ontario, Canada
| | - Richard H Ho
- Division of Hematology and Oncology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
33
|
He JL, Zhou ZW, Yin JJ, He CQ, Zhou SF, Yu Y. Schisandra chinensis regulates drug metabolizing enzymes and drug transporters via activation of Nrf2-mediated signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 9:127-46. [PMID: 25552902 PMCID: PMC4277124 DOI: 10.2147/dddt.s68501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug metabolizing enzymes (DMEs) and drug transporters are regulated via epigenetic, transcriptional, posttranscriptional, and translational and posttranslational modifications. Phase I and II DMEs and drug transporters play an important role in the disposition and detoxification of a large number of endogenous and exogenous compounds. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a critical regulator of a variety of important cytoprotective genes that are involved in disposition and detoxification of xenobiotics. Schisandra chinensis (SC) is a commonly used traditional Chinese herbal medicine that has been primarily used to protect the liver because of its potent antioxidative and anti-inflammatory activities. SC can modulate some DMEs and drug transporters, but the underlying mechanisms are unclear. In this study, we aimed to explore the role of Nrf2 in the regulatory effect of SC extract (SCE) on selected DMEs and drug transporters in human hepatocellular liver carcinoma cell line (HepG2) cells. The results showed that SCE, schisandrin A, and schisandrin B significantly increased the expression of NAD(P)H: Nicotinamide Adenine Dinucleotide Phosphate-oxidase or:quinone oxidoreductase 1, heme oxygenase-1, glutamate–cysteine ligase, and glutathione S-transferase A4 at both transcriptional and posttranscriptional levels. Incubation of HepG2 cells with SCE resulted in a significant increase in the intracellular level of glutathione and total glutathione S-transferase content. SCE significantly elevated the messenger ribonucleic acid and protein levels of P-glycoprotein and multidrug resistance-associated protein 2 and 4, whereas the expression of organic anion transporting peptide 1A2 and 1B1 was significantly downregulated by SCE. Knockdown of Nrf2 by small interfering ribonucleic acid attenuated the regulatory effect of SCE on these DMEs and drug transporters. SCE significantly upregulated Nrf2 and promoted the translocation of Nrf2 from cytoplasm to the nuclei. Additionally, SCE significantly suppressed the expression of cytosolic Kelch-like ECH-associated protein 1 (the repressor of Nrf2) and remarkably increased Nrf2 stability in HepG2 cells. Taken together, our findings suggest that the hepatoprotective effects of SCE may be partially ascribed to the modulation of DMEs and drug transporters via Nrf2-mediated signaling pathway. SCE may alter the pharmacokinetics of other coadministered drugs that are substrates of these DMEs and transporters and thus cause unfavorable herb–drug interactions.
Collapse
Affiliation(s)
- Jin-Lian He
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guiyang Medical University, Guiyang, Guizhou, People's Republic of China
| | - Juan-Juan Yin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Chang-Qiang He
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guiyang Medical University, Guiyang, Guizhou, People's Republic of China
| | - Yang Yu
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
34
|
Abstract
Potential drug-drug interactions mediated by the ATP-binding cassette (ABC) transporter and solute carrier (SLC) transporter families are of clinical and regulatory concern. However, the endogenous functions of these drug transporters are not well understood. Discussed here is evidence for the roles of ABC and SLC transporters in the handling of diverse substrates, including metabolites, antioxidants, signalling molecules, hormones, nutrients and neurotransmitters. It is suggested that these transporters may be part of a larger system of remote communication ('remote sensing and signalling') between cells, organs, body fluid compartments and perhaps even separate organisms. This broader view may help to clarify disease mechanisms, drug-metabolite interactions and drug effects relevant to diabetes, chronic kidney disease, metabolic syndrome, hypertension, gout, liver disease, neuropsychiatric disorders, inflammatory syndromes and organ injury, as well as prenatal and postnatal development.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0693, USA
| |
Collapse
|
35
|
Safety and pharmacokinetics of the CIME combination of drugs and their metabolites after a single oral dosing in healthy volunteers. Eur J Drug Metab Pharmacokinet 2014; 41:125-38. [DOI: 10.1007/s13318-014-0239-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 11/20/2014] [Indexed: 01/07/2023]
|
36
|
Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res 2014; 56:1085-99. [PMID: 25210150 DOI: 10.1194/jlr.r054114] [Citation(s) in RCA: 401] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
In addition to their classical roles as detergents to aid in the process of digestion, bile acids have been identified as important signaling molecules that function through various nuclear and G protein-coupled receptors to regulate a myriad of cellular and molecular functions across both metabolic and nonmetabolic pathways. Signaling via these pathways will vary depending on the tissue and the concentration and chemical structure of the bile acid species. Important determinants of the size and composition of the bile acid pool are their efficient enterohepatic recirculation, their host and microbial metabolism, and the homeostatic feedback mechanisms connecting hepatocytes, enterocytes, and the luminal microbiota. This review focuses on the mammalian intestine, discussing the physiology of bile acid transport, the metabolism of bile acids in the gut, and new developments in our understanding of how intestinal metabolism, particularly by the gut microbiota, affects bile acid signaling.
Collapse
Affiliation(s)
- Paul A Dawson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| | - Saul J Karpen
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Emory University, Atlanta, GA 30322
| |
Collapse
|
37
|
Levitt DG, Levitt MD. Quantitative assessment of the multiple processes responsible for bilirubin homeostasis in health and disease. Clin Exp Gastroenterol 2014; 7:307-28. [PMID: 25214800 PMCID: PMC4159128 DOI: 10.2147/ceg.s64283] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Serum bilirubin measurements are commonly obtained for the evaluation of ill patients and to screen for liver disease in routine physical exams. An enormous research effort has identified the multiple mechanisms involved in the production and metabolism of conjugated (CB) and unconjugated bilirubin (UB). While the qualitative effects of these mechanisms are well understood, their expected quantitative influence on serum bilirubin homeostasis has received less attention. In this review, each of the steps involved in bilirubin production, metabolism, hepatic cell uptake, and excretion is quantitatively examined. We then attempt to predict the expected effect of normal and defective function on serum UB and CB levels in health and disease states including hemolysis, extra- and intrahepatic cholestasis, hepatocellular diseases (eg, cirrhosis, hepatitis), and various congenital defects in bilirubin conjugation and secretion (eg, Gilbert's, Dubin-Johnson, Crigler-Najjar, Rotor syndromes). Novel aspects of this review include: 1) quantitative estimates of the free and total UB and CB in the plasma, hepatocyte, and bile; 2) detailed discussion of the important implications of the recently recognized role of the hepatic OATP transporters in the maintenance of CB homeostasis; 3) discussion of the differences between the standard diazo assay versus chromatographic measurement of CB and UB; 4) pharmacokinetic implications of the extremely high-affinity albumin binding of UB; 5) role of the enterohepatic circulation in physiologic jaundice of newborn and fasting hyperbilirubinemia; and 6) insights concerning the clinical interpretation of bilirubin measurements.
Collapse
Affiliation(s)
- David G Levitt
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Michael D Levitt
- Research Service, Veterans Affairs Medical Center, Minneapolis, MN, USA
| |
Collapse
|
38
|
Differential cellular expression of organic anion transporting peptides OATP1A2 and OATP2B1 in the human retina and brain: implications for carrier-mediated transport of neuropeptides and neurosteriods in the CNS. Pflugers Arch 2014; 467:1481-1493. [DOI: 10.1007/s00424-014-1596-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/28/2014] [Accepted: 08/06/2014] [Indexed: 02/07/2023]
|
39
|
Iusuf D, Hendrikx JJ, van Esch A, van de Steeg E, Wagenaar E, Rosing H, Beijnen JH, Schinkel AH. Human OATP1B1, OATP1B3 and OATP1A2 can mediate thein vivouptake and clearance of docetaxel. Int J Cancer 2014; 136:225-33. [DOI: 10.1002/ijc.28970] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/28/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Dilek Iusuf
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Jeroen J.M.A. Hendrikx
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
- Department of Pharmacy & Pharmacology; Slotervaart Hospital; Amsterdam The Netherlands
| | - Anita van Esch
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Evita van de Steeg
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Els Wagenaar
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology; Slotervaart Hospital; Amsterdam The Netherlands
| | - Jos H. Beijnen
- Department of Pharmacy & Pharmacology; Slotervaart Hospital; Amsterdam The Netherlands
| | - Alfred H. Schinkel
- Division of Molecular Oncology; The Netherlands Cancer Institute; Amsterdam The Netherlands
| |
Collapse
|
40
|
Nieuweboer AJM, Hu S, Gui C, Hagenbuch B, Ghobadi Moghaddam-Helmantel IM, Gibson AA, de Bruijn P, Mathijssen RHJ, Sparreboom A. Influence of drug formulation on OATP1B-mediated transport of paclitaxel. Cancer Res 2014; 74:3137-45. [PMID: 24755470 DOI: 10.1158/0008-5472.can-13-3634] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Taxane antineoplastic agents are extensively taken up into hepatocytes by OATP1B-type transporters before metabolism and excretion. Because the biodistributional properties imposed upon these agents by different solubilizers drive clinically important pharmacodynamic endpoints, we tested the hypothesis that the in vitro and in vivo interaction of taxanes with OATP1B transporters is affected by the choice of drug delivery system. Transport of paclitaxel, docetaxel, and cabazitaxel was studied in vitro using various cell lines transfected with OATP1B1, OATP1B3, or the rodent equivalent OATP1B2. Pharmacokinetic studies were done in wild-type and OATP1B2-knockout mice in the presence or absence of polysorbate 80 (PS80) or Kolliphor EL (formerly Cremophor EL; CrEL). Paclitaxel and docetaxel, but not cabazitaxel, were transported substrates of OATP1B1, OATP1B3, and OATP1B2, and these in vitro transport processes were strongly reduced in the presence of clinically relevant concentrations of PS80 and CrEL. When paclitaxel was administered without any solubilizers, deficiency of OATP1B2 in mice was associated with a significantly decreased systemic clearance because of a liver distribution defect (P=0.000484). However, this genotype dependence of paclitaxel clearance was masked in the presence of PS80 or CrEL because of significant inhibition of OATP1B2-mediated hepatocellular uptake of the drug (P<0.05). Our findings confirm the importance of OATP1B-type transporters in the hepatic elimination of taxanes and indicate that this process can be inhibited by PS80 and CrEL. These results suggest that the likelihood of drug-drug interactions mediated by these transporters is strongly dependent on the selected taxane solubilizer.
Collapse
Affiliation(s)
- Annemieke J M Nieuweboer
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Shuiying Hu
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Chunshan Gui
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Bruno Hagenbuch
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Inge M Ghobadi Moghaddam-Helmantel
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Alice A Gibson
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Peter de Bruijn
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Ron H J Mathijssen
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| | - Alex Sparreboom
- Authors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, MissouriAuthors' Affiliations: Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee; and Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Missouri
| |
Collapse
|
41
|
Inhibition of OATP1B1 by tyrosine kinase inhibitors: in vitro-in vivo correlations. Br J Cancer 2014; 110:894-8. [PMID: 24398510 PMCID: PMC3929889 DOI: 10.1038/bjc.2013.811] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/04/2013] [Accepted: 12/06/2013] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Several tyrosine kinase inhibitors (TKIs) can decrease docetaxel clearance in patients by an unknown mechanism. We hypothesised that these interactions are mediated by the hepatic uptake transporter OATP1B1. METHODS The influence of 16 approved TKIs on transport was studied in vitro using HEK293 cells expressing OATP1B1 or its mouse equivalent Oatp1b2. Pharmacokinetic studies were performed with Oatp1b2-knockout and OATP1B1-transgenic mice. RESULTS All docetaxel-interacting TKIs, including sorafenib, were identified as potent inhibitors of OATP1B1 in vitro. Although Oatp1b2 deficiency in vivo was associated with increased docetaxel exposure, single- or multiple-dose sorafenib did not influence docetaxel pharmacokinetics. CONCLUSION These findings highlight the importance of identifying proper preclinical models for verifying and predicting TKI-chemotherapy interactions involving transporters.
Collapse
|
42
|
Higgins JW, Bao JQ, Ke AB, Manro JR, Fallon JK, Smith PC, Zamek-Gliszczynski MJ. Utility of Oatp1a/1b-knockout and OATP1B1/3-humanized mice in the study of OATP-mediated pharmacokinetics and tissue distribution: case studies with pravastatin, atorvastatin, simvastatin, and carboxydichlorofluorescein. Drug Metab Dispos 2014; 42:182-92. [PMID: 24194513 PMCID: PMC11918263 DOI: 10.1124/dmd.113.054783] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although organic anion transporting polypeptide (OATP)-mediated hepatic uptake is generally conserved between rodents and humans at a gross pharmacokinetic level, the presence of three major hepatic OATPs with broad overlap in substrate and inhibitor affinity, and absence of rodent-human orthologs preclude clinical translation of single-gene knockout/knockin findings. At present, changes in pharmacokinetics and tissue distribution of pravastatin, atorvastatin, simvastatin, and carboxydichlorofluorescein were studied in oatp1a/1b-knockout mice lacking the three major hepatic oatp isoforms, and in knockout mice with liver-specific knockin of human OATP1B1 or OATP1B3. Relative to wild-type controls, oatp1a/1b-knockout mice exhibited 1.6- to 19-fold increased intravenous and 2.1- to 115-fold increased oral drug exposure, due to 33%-75% decreased clearance, 14%-60% decreased volume of distribution, and ≤74-fold increased oral bioavailability, with the magnitude of change depending on the contribution of oatp1a/1b to pharmacokinetics. Hepatic drug distribution was 4.2- to 196-fold lower in oatp1a/1b-knockout mice; distributional attenuation was less notable in kidney, brain, cardiac, and skeletal muscle. Knockin of OATP1B1 or OATP1B3 partially restored control clearance, volume, and bioavailability values (24%-142% increase, ≤47% increase, and ≤77% decrease vs. knockout, respectively), such that knockin pharmacokinetic profiles were positioned between knockout and wild-type mice. Consistent with liver-specific humanization, only hepatic drug distribution was partially restored (1.3- to 6.5-fold increase vs. knockout). Exposure and liver distribution changes in OATP1B1-humanized versus knockout mice predicted the clinical impact of OATP1B1 on oral exposure and contribution to human hepatic uptake of statins within 1.7-fold, but only after correcting for human/humanized mouse liver relative protein expression factor (OATP1B1 = 2.2, OATP1B3 = 0.30).
Collapse
Affiliation(s)
- J William Higgins
- Drug Disposition (J.W.H., J.Q.B., A.B.K., M.J.Z.-G.) and Global Statistical Sciences (J.R.M.), Lilly Research Laboratories, Indianapolis, Indiana; and Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina (J.K.F., P.C.S., M.J.Z.-G.)
| | | | | | | | | | | | | |
Collapse
|
43
|
Direct determination of free bilirubin in serum at sub-nanomolar levels. Anal Chim Acta 2014; 809:174-82. [DOI: 10.1016/j.aca.2013.11.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/15/2013] [Accepted: 11/16/2013] [Indexed: 11/20/2022]
|
44
|
Wang WJ, Murray JW, Wolkoff AW. Oatp1a1 requires PDZK1 to traffic to the plasma membrane by selective recruitment of microtubule-based motor proteins. Drug Metab Dispos 2014; 42:62-9. [PMID: 24115750 PMCID: PMC3876792 DOI: 10.1124/dmd.113.054536] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/10/2013] [Indexed: 11/22/2022] Open
Abstract
Previous studies identified a family of organic anion transport proteins (OATPs), many of which have C-terminal PDZ binding consensus sequences. In particular, the C-terminal four amino acids of Oatp1a1, a transporter on rat and mouse hepatocytes, comprise a consensus binding site for PDZK1. In PDZK1 knockout mice and in transfected cells where PDZK1 expression was knocked down, Oatp1a1 accumulates in intracellular vesicles. The present study tests the hypothesis that Oatp1a1 traffics to and from the cell surface in vesicles along microtubules, and that PDZK1 guides recruitment of specific motors to these vesicles. Oatp1a1-containing vesicles were prepared from wild-type and PDZK1 knockout mice. As seen by immunofluorescence, kinesin-1, a microtubule plus-end directed motor, was largely associated with vesicles from wild-type mouse liver, whereas dynein, a minus-end directed motor, was largely associated with vesicles from PDZK1 knockout mouse liver. Quantification of motility on directionally marked microtubules following addition of 50 µM ATP showed that wild-type vesicles moved equally toward the plus and minus ends whereas PDZK1 knockout vesicles moved predominantly toward the minus end, consistent with net movement toward the cell interior. These studies provide a novel mechanism by which PDZK1 regulates intracellular trafficking of Oatp1a1 by recruiting specific motors to Oatp1a1-containing vesicles. In the absence of PDZK1, Oatp1a1-containing vesicles cannot recruit kinesin-1 and associate with dynein as a predominant minus-end directed motor. Whether this is a result of direct interaction of the Oatp1a1 cytoplasmic domain with dynein or with a dynein-containing protein complex remains to be established.
Collapse
Affiliation(s)
- Wen-Jun Wang
- Department of Anatomy and Structural Biology, Marion Bessin Liver Research Center, Division of Gastroenterology and Liver Diseases, Albert Einstein College of Medicine, Bronx, New York
| | | | | |
Collapse
|
45
|
Walker AL, Lancaster CS, Finkelstein D, Ware RE, Sparreboom A. Organic anion transporting polypeptide 1B transporters modulate hydroxyurea pharmacokinetics. Am J Physiol Cell Physiol 2013; 305:C1223-9. [PMID: 23986199 PMCID: PMC3882360 DOI: 10.1152/ajpcell.00232.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/19/2013] [Indexed: 12/31/2022]
Abstract
Hydroxyurea is currently the only FDA-approved drug that ameliorates the pathophysiology of sickle cell anemia. Unfortunately, substantial interpatient variability in the pharmacokinetics (PK) of hydroxyurea may result in variation of the drug's efficacy. However, little is known about mechanisms that modulate hydroxyurea PK. Recent in vitro studies identifying hydroxyurea as a substrate for organic anion transporting polypeptide (OATP1B) transporters prompted the current investigation assessing the role of OATP1B transporters in modulating hydroxyurea PK. Using wild-type and Oatp1b knockout (Oatp1b(-/-)) mice, hydroxyurea PK was analyzed in vivo by measuring [(14)C]hydroxyurea distribution in plasma, kidney, liver, urine, or the exhaled (14)CO2 metabolite. Plasma levels were significantly reduced by 20% in Oatp1b(-/-) mice compared with wild-type (area under the curve of 38.64 or 48.45 μg·h(-1)·ml(-1), respectively) after oral administration, whereas no difference was observed between groups following intravenous administration. Accumulation in the kidney was significantly decreased by twofold in Oatp1b(-/-) mice (356.9 vs. 748.1 pmol/g), which correlated with a significant decrease in urinary excretion. Hydroxyurea accumulation in the liver was also decreased (136.6 vs. 107.3 pmol/g in wild-type or Oatp1b(-/-) mice, respectively) correlating with a decrease in exhaled (14)CO2. These findings illustrate that deficiency of Oatp1b transporters alters the absorption, distribution, and elimination of hydroxyurea thus providing the first in vivo evidence that cell membrane transporters may play a significant role in modulating hydroxyurea PK. Future studies to investigate other transporters and their role in hydroxyurea disposition are warranted for understanding the sources of variation in hydroxyurea's PK.
Collapse
Affiliation(s)
- Aisha L Walker
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | | | | |
Collapse
|
46
|
Iusuf D, Ludwig M, Elbatsh A, van Esch A, van de Steeg E, Wagenaar E, van der Valk M, Lin F, van Tellingen O, Schinkel AH. OATP1A/1B transporters affect irinotecan and SN-38 pharmacokinetics and carboxylesterase expression in knockout and humanized transgenic mice. Mol Cancer Ther 2013; 13:492-503. [PMID: 24194565 DOI: 10.1158/1535-7163.mct-13-0541] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Organic anion-transporting polypeptides (OATP) mediate the hepatic uptake of many drugs, thus codetermining their clearance. Impaired hepatic clearance due to low-activity polymorphisms in human OATP1B1 may increase systemic exposure to SN-38, the active and toxic metabolite of the anticancer prodrug irinotecan. We investigated the pharmacokinetics and toxicity of irinotecan and SN-38 in Oatp1a/1b-null mice: Plasma exposure of irinotecan and SN-38 was increased 2 to 3-fold after irinotecan dosing (10 mg/kg, i.v.) compared with wild-type mice. Also, liver-to-plasma ratios were significantly reduced, suggesting impaired hepatic uptake of both compounds. After 6 daily doses of irinotecan, Oatp1a/1b-null mice suffered from increased toxicity. However, Oatp1a/1b-null mice had increased levels of carboxylesterase (Ces) enzymes, which caused higher conversion of irinotecan to SN-38 in plasma, potentially complicating pharmacokinetic analyses. Ces inhibitors blocked this increased conversion. Interestingly, liver-specific humanized OATP1B1 and OATP1B3 transgenic mice had normalized hepatic expression of Ces1 genes. While irinotecan liver-to-plasma ratios in these humanized mice were similar to those in Oatp1a/1b-null mice, SN-38 liver-to-plasma ratios returned to wild-type levels, suggesting that human OATP1B proteins mediate SN-38, but not irinotecan uptake in vivo. Upon direct administration of SN-38 (1 mg/kg, i.v.), Oatp1a/1b-null mice had increased SN-38 plasma levels, lower liver concentrations, and decreased cumulative biliary excretion of SN-38. Mouse Oatp1a/1b transporters have a role in the plasma clearance of irinotecan and SN-38, whereas human OATP1B transporters may only affect SN-38 disposition. Oatp1a/1b-null mice have increased expression and activity of Ces1 enzymes, whereas humanized mice provide a rescue of this phenotype.
Collapse
Affiliation(s)
- Dilek Iusuf
- Corresponding Author: Alfred H. Schinkel, Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sticova E, Jirsa M. New insights in bilirubin metabolism and their clinical implications. World J Gastroenterol 2013; 19:6398-6407. [PMID: 24151358 PMCID: PMC3801310 DOI: 10.3748/wjg.v19.i38.6398] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 07/18/2013] [Accepted: 08/09/2013] [Indexed: 02/06/2023] Open
Abstract
Bilirubin, a major end product of heme breakdown, is an important constituent of bile, responsible for its characteristic colour. Over recent decades, our understanding of bilirubin metabolism has expanded along with the processes of elimination of other endogenous and exogenous anionic substrates, mediated by the action of multiple transport systems at the sinusoidal and canalicular membrane of hepatocytes. Several inherited disorders characterised by impaired bilirubin conjugation (Crigler-Najjar syndrome type I and type II, Gilbert syndrome) or transport (Dubin-Johnson and Rotor syndrome) result in various degrees of hyperbilirubinemia of either the predominantly unconjugated or predominantly conjugated type. Moreover, disrupted regulation of hepatobiliary transport systems can explain jaundice in many acquired liver disorders. In this review, we discuss the recent data on liver bilirubin handling based on the discovery of the molecular basis of Rotor syndrome. The data show that a substantial fraction of bilirubin conjugates is primarily secreted by MRP3 at the sinusoidal membrane into the blood, from where they are subsequently reuptaken by sinusoidal membrane-bound organic anion transporting polypeptides OATP1B1 and OATP1B3. OATP1B proteins are also responsible for liver clearance of bilirubin conjugated in splanchnic organs, such as the intestine and kidney, and for a number of endogenous compounds, xenobiotics and drugs. Absence of one or both OATP1B proteins thus may have serious impact on toxicity of commonly used drugs cleared by this system such as statins, sartans, methotrexate or rifampicin. The liver-blood cycling of conjugated bilirubin is impaired in cholestatic and parenchymal liver diseases and this impairment most likely contributes to jaundice accompanying these disorders.
Collapse
|
48
|
Tang SC, Hendrikx JJMA, Beijnen JH, Schinkel AH. Genetically modified mouse models for oral drug absorption and disposition. Curr Opin Pharmacol 2013; 13:853-8. [PMID: 24021267 DOI: 10.1016/j.coph.2013.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/15/2013] [Accepted: 08/21/2013] [Indexed: 01/09/2023]
Abstract
Intestinal absorption is an essential step in the therapeutic use of most orally administered drugs and often mediated by enterocyte transmembrane transporters. Here we discuss several of these drug transport systems and knockout mouse models to study them. These studies showed that Multidrug resistance-associated protein 2 (Mrp2) can limit intestinal drug absorption. Organic cation transporter n1 (Octn1) and Octn2 might also facilitate intestinal drug absorption, although direct in vivo evidence is lacking. On the other hand, intestinal uptake of drugs is facilitated by the Equilibrative nucleoside transporter 1 (Ent1), Mrp3 and possibly Mrp4. No significant role in intestinal absorption for Oct1 and Oct2 or for Organic anion-transporting polypeptides (Oatp) 1a and 1b was found so far.
Collapse
Affiliation(s)
- Seng Chuan Tang
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
49
|
Lancaster CS, Sprowl JA, Walker AL, Hu S, Gibson AA, Sparreboom A. Modulation of OATP1B-type transporter function alters cellular uptake and disposition of platinum chemotherapeutics. Mol Cancer Ther 2013; 12:1537-44. [PMID: 23757163 DOI: 10.1158/1535-7163.mct-12-0926] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Expression of the human organic anion transporting polypeptides OATP1B1 and OATP1B3 has been previously believed to be restricted to hepatocytes. Here we show that the gene encoding OATP1B3, but not OATP1B1, is abundantly expressed in multiple human solid tumors that include hepatocellular, lung, and ovarian carcinomas. Surprisingly, OATP1B3 gene expression in a panel of 60 human tumor cell lines was linked with sensitivity to multiple cytotoxic agents, including the platinum anticancer drugs cisplatin, carboplatin, and oxaliplatin. In addition, overexpression of OATP1B3 in mammalian cells increased cellular accumulation of platinum agents and decreased cell survival. In mice with a targeted disruption of the ortholog transporter Oatp1b2, the liver-to-plasma ratio of cisplatin was significantly reduced compared with wild-type mice, without concurrent changes in expression profiles of other transporter genes. Our findings indicate an unexpected role for tumoral and host OATP1B-type carriers in the toxicity and disposition of platinum anticancer drugs, and may provide a foundation for understanding the extensive interindividual pharmacodynamic variability seen with these drugs in patients.
Collapse
Affiliation(s)
- Cynthia S Lancaster
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | | | | | | |
Collapse
|
50
|
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. [PMID: 23429889 DOI: 10.1124/mol.112.081927] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Organic anion-transporting polypeptides (OATPs) mediate the liver uptake and hence plasma clearance of a broad range of drugs. For rosuvastatin, a cholesterol-lowering drug and OATP1A/1B substrate, the liver represents both its main therapeutic target and its primary clearance organ. Here we studied the impact of Oatp1a/1b uptake transporters on the pharmacokinetics of rosuvastatin using wild-type and Oatp1a/1b-null mice. After oral administration (15 mg/kg), intestinal absorption of rosuvastatin was not impaired in Oatp1a/1b-null mice, but systemic exposure (area under the curve) was 8-fold higher in these mice compared with wild-type. Although liver exposure was comparable between the two mouse strains (despite the increased blood exposure), the liver-to-blood ratios were markedly decreased (>10-fold) in the absence of Oatp1a/1b transporters. After intravenous administration (5 mg/kg), systemic exposure was 3-fold higher in Oatp1a/1b-null mice than in the wild-type mice. Liver, small intestinal, and kidney exposure were slightly, but not significantly, increased in Oatp1a/1b-null mice. The biliary excretion of rosuvastatin was very fast, with 60% of the dose eliminated within 15 minutes after intravenous administration, and also not significantly altered in Oatp1a/1b-null mice. Rosuvastatin renal clearance, although still minor, was increased ∼15-fold in Oatp1a/1b-null males, suggesting a role of Oatp1a1 in the renal reabsorption of rosuvastatin. Absence of Oatp1a/1b uptake transporters increases the systemic exposure of rosuvastatin by reducing its hepatic extraction ratio. However, liver concentrations are not significantly affected, most likely due to the compensatory activity of high-capacity, low-affinity alternative uptake transporters at higher systemic rosuvastatin levels and the absence of efficient alternative rosuvastatin clearance mechanisms.
Collapse
Affiliation(s)
- Dilek Iusuf
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|