1
|
Özvegy-Laczka C, Ungvári O, Bakos É. Fluorescence-based methods for studying activity and drug-drug interactions of hepatic solute carrier and ATP binding cassette proteins involved in ADME-Tox. Biochem Pharmacol 2023; 209:115448. [PMID: 36758706 DOI: 10.1016/j.bcp.2023.115448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
In humans, approximately 70% of drugs are eliminated through the liver. This process is governed by the concerted action of membrane transporters and metabolic enzymes. Transporters mediating hepatocellular uptake of drugs belong to the SLC (Solute carrier) superfamily of transporters. Drug efflux either toward the portal vein or into the bile is mainly mediated by active transporters of the ABC (ATP Binding Cassette) family. Alteration in the function and/or expression of liver transporters due to mutations, disease conditions, or co-administration of drugs or food components can result in altered pharmacokinetics. On the other hand, drugs or food components interacting with liver transporters may also interfere with liver function (e.g., bile acid homeostasis) and may even cause liver toxicity. Accordingly, certain transporters of the liver should be investigated already at an early stage of drug development. Most frequently radioactive probes are applied in these drug-transporter interaction tests. However, fluorescent probes are cost-effective and sensitive alternatives to radioligands, and are gaining wider application in drug-transporter interaction tests. In our review, we summarize our current understanding about hepatocyte ABC and SLC transporters affected by drug interactions. We provide an update of the available fluorescent and fluorogenic/activable probes applicable in in vitro or in vivo testing of these ABC and SLC transporters, including near-infrared transporter probes especially suitable for in vivo imaging. Furthermore, our review gives a comprehensive overview of the available fluorescence-based methods, not directly relying on the transport of the probe, suitable for the investigation of hepatic ABC or SLC-type drug transporters.
Collapse
Affiliation(s)
- Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary.
| | - Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary; Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary
| |
Collapse
|
2
|
Magdy T, Jouni M, Kuo H, Weddle CJ, Lyra–Leite D, Fonoudi H, Romero–Tejeda M, Gharib M, Javed H, Fajardo G, Ross CJD, Carleton BC, Bernstein D, Burridge PW. Identification of Drug Transporter Genomic Variants and Inhibitors That Protect Against Doxorubicin-Induced Cardiotoxicity. Circulation 2022; 145:279-294. [PMID: 34874743 PMCID: PMC8792344 DOI: 10.1161/circulationaha.121.055801] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Multiple pharmacogenomic studies have identified the synonymous genomic variant rs7853758 (G > A, L461L) and the intronic variant rs885004 in SLC28A3 (solute carrier family 28 member 3) as statistically associated with a lower incidence of anthracycline-induced cardiotoxicity. However, the true causal variant(s), the cardioprotective mechanism of this locus, the role of SLC28A3 and other solute carrier (SLC) transporters in anthracycline-induced cardiotoxicity, and the suitability of SLC transporters as targets for cardioprotective drugs has not been investigated. METHODS Six well-phenotyped, doxorubicin-treated pediatric patients from the original association study cohort were recruited again, and human induced pluripotent stem cell-derived cardiomyocytes were generated. Patient-specific doxorubicin-induced cardiotoxicity (DIC) was then characterized using assays of cell viability, activated caspase 3/7, and doxorubicin uptake. The role of SLC28A3 in DIC was then queried using overexpression and knockout of SLC28A3 in isogenic human-induced pluripotent stem cell-derived cardiomyocytes using a CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated protein 9). Fine-mapping of the SLC28A3 locus was then completed after SLC28A3 resequencing and an extended in silico haplotype and functional analysis. Genome editing of the potential causal variant was done using cytosine base editor. SLC28A3-AS1 overexpression was done using a lentiviral plasmid-based transduction and was validated using stranded RNA-sequencing after ribosomal RNA depletion. Drug screening was done using the Prestwick Chemical Library (n = 1200), followed by in vivo validation in mice. The effect of desipramine on doxorubicin cytotoxicity was also investigated in 8 cancer cell lines. RESULTS Here, using the most commonly used anthracycline, doxorubicin, we demonstrate that patient-derived cardiomyocytes recapitulate the cardioprotective effect of the SLC28A3 locus and that SLC28A3 expression influences the severity of DIC. Using Nanopore-based fine-mapping and base editing, we identify a novel cardioprotective single nucleotide polymorphism, rs11140490, in the SLC28A3 locus; its effect is exerted via regulation of an antisense long noncoding RNA (SLC28A3-AS1) that overlaps with SLC28A3. Using high-throughput drug screening in patient-derived cardiomyocytes and whole organism validation in mice, we identify the SLC competitive inhibitor desipramine as protective against DIC. CONCLUSIONS This work demonstrates the power of the human induced pluripotent stem cell model to take a single nucleotide polymorphism from a statistical association through to drug discovery, providing human cell-tested data for clinical trials to attenuate DIC.
Collapse
Affiliation(s)
- Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hui–Hsuan Kuo
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Carly J. Weddle
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Davi Lyra–Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Marisol Romero–Tejeda
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hoor Javed
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Giovanni Fajardo
- Department of Pediatrics (Division of Cardiology), Stanford University School of Medicine, Stanford, CA
| | - Colin J. D. Ross
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce C. Carleton
- British Columbia Children’s Hospital Research Institute, Vancouver, British Columbia, Canada.,Division of Translational Therapeutics Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Pharmaceutical Outcomes Programme, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Daniel Bernstein
- Department of Pediatrics (Division of Cardiology), Stanford University School of Medicine, Stanford, CA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
3
|
Jala A, Ponneganti S, Vishnubhatla DS, Bhuvanam G, Mekala PR, Varghese B, Radhakrishnanand P, Adela R, Murty US, Borkar RM. Transporter-mediated drug-drug interactions: advancement in models, analytical tools, and regulatory perspective. Drug Metab Rev 2021; 53:285-320. [PMID: 33980079 DOI: 10.1080/03602532.2021.1928687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023]
Abstract
Drug-drug interactions mediated by transporters are a serious clinical concern hence a tremendous amount of work has been done on the characterization of the transporter-mediated proteins in humans and animals. The underlying mechanism for the transporter-mediated drug-drug interaction is the induction or inhibition of the transporter which is involved in the cellular uptake and efflux of drugs. Transporter of the brain, liver, kidney, and intestine are major determinants that alter the absorption, distribution, metabolism, excretion profile of drugs, and considerably influence the pharmacokinetic profile of drugs. As a consequence, transporter proteins may affect the therapeutic activity and safety of drugs. However, mounting evidence suggests that many drugs change the activity and/or expression of the transporter protein. Accordingly, evaluation of drug interaction during the drug development process is an integral part of risk assessment and regulatory requirements. Therefore, this review will highlight the clinical significance of the transporter, their role in disease, possible cause underlying the drug-drug interactions using analytical tools, and update on the regulatory requirement. The recent in-silico approaches which emphasize the advancement in the discovery of drug-drug interactions are also highlighted in this review. Besides, we discuss several endogenous biomarkers that have shown to act as substrates for many transporters, which could be potent determinants to find the drug-drug interactions mediated by transporters. Transporter-mediated drug-drug interactions are taken into consideration in the drug approval process therefore we also provided the extrapolated decision trees from in-vitro to in-vivo, which may trigger the follow-up to clinical studies.
Collapse
Affiliation(s)
- Aishwarya Jala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Srikanth Ponneganti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Devi Swetha Vishnubhatla
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Gayathri Bhuvanam
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Prithvi Raju Mekala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Bincy Varghese
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Pullapanthula Radhakrishnanand
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | | | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| |
Collapse
|
4
|
Chen Y, Xue F, Xia G, Zhao Z, Chen C, Li Y, Zhang Y. Transepithelial transport mechanisms of 7,8-dihydroxyflavone, a small molecular TrkB receptor agonist, in human intestinal Caco-2 cells. Food Funct 2019; 10:5215-5227. [PMID: 31384856 DOI: 10.1039/c9fo01007f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
7,8-Dihydroxyflavone (7,8-DHF), as a high-affinity TrkB receptor agonist, has been extensively explored in many human disorders involving brain-derived neurotrophic factor (BDNF) such as Alzheimer's disease, Parkinson's disease, depression, and obesity. However, to date, the transepithelial transport mechanisms of 7,8-DHF in the intestines remain unclear. The aim of our work was to quantify and to characterize in vitro transport of naturally occurring 7,8-DHF distinguished by its physicochemical and pharmacological properties. We discussed the transport mechanisms of 7,8-DHF using the Caco-2 cell model to determine the bi-directional permeability with different environmental factors (time, concentration, pH, metabolic inhibitors etc.). The influx and efflux characteristics of 7,8-DHF were also clarified. 7,8-DHF was poorly transported across Caco-2 cell monolayers by mainly passive diffusion via a transcellular pathway and not a paracellular pathway. The transport of 7,8-DHF was time and concentration-dependent in both the apical (AP) to basolateral (BL) side and the reverse direction. Interestingly, decreasing the pH from 7.4 to 6.0 markedly enhanced 7,8-DHF transport. It is noteworthy that 7,8-DHF transport was strongly inhibited by metabolic inhibitors and was highly dependent on temperature. The efflux ratio (ER) values at different concentrations were all above 1.5, indicating the existence of the efflux transporter. We found that breast cancer resistance protein (BCRP) was not involved in 7,8-DHF secretion and that the transport mechanism of 7,8-DHF was passive transport with an active efflux mediated by P-glycoprotein (P-gp) and multidrug resistance associated proteins (MRPs), particularly MRP 2. Moreover, the use of various influx transporter inhibitors in Caco-2 cells showed that organic cation transporters (OCTs) and organic anion-transporting polypeptides (OATPs) participated in 7,8-DHF transport. Taken together, the elucidated transport characteristics of 7,8-DHF provide useful information for designing novel and efficient delivery systems and avoiding food-food or food-drug interactions.
Collapse
Affiliation(s)
- Yufeng Chen
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment; Zhejiang University, Hangzhou 310058, China.
| | | | | | | | | | | | | |
Collapse
|
5
|
Nie Y, Yang J, Liu S, Sun R, Chen H, Long N, Jiang R, Gui C. Genetic polymorphisms of human hepatic OATPs: functional consequences and effect on drug pharmacokinetics. Xenobiotica 2019; 50:297-317. [DOI: 10.1080/00498254.2019.1629043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yingmin Nie
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jingjie Yang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shuai Liu
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Ruiqi Sun
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Huihui Chen
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Nan Long
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Rui Jiang
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chunshan Gui
- Department of Pharmaceutical Analysis, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
6
|
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
|
7
|
Crowe A, Miller J, Yue W. Genotyping of the OATP1B1 c. 521 T>C Polymorphism from the Formalin-Fixed Paraffin-Embedded (FFPE) Tissue Specimens: An Optimized Protocol. Bio Protoc 2019; 9:e3343. [DOI: 10.21769/bioprotoc.3343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
8
|
Bolger GT, Licollari A, Tan A, Greil R, Vcelar B, Greil-Ressler S, Weiss L, Schönlieb C, Magnes T, Radl B, Majeed M, Sordillo PP. Pharmacokinetics of liposomal curcumin (Lipocurc™) infusion: effect of co-medication in cancer patients and comparison with healthy individuals. Cancer Chemother Pharmacol 2018; 83:265-275. [PMID: 30430227 DOI: 10.1007/s00280-018-3730-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE Investigation of the impact of co-medication on the plasma levels of curcumin and tetrahydrocurcumin (THC) in cancer patients and a comparison of the pharmacokinetics of curcumin and plasma levels of THC between cancer patients and healthy individuals following intravenous infusion of Lipocurc™ (liposomal curcumin). METHODS Correlation analysis was used to determine the impact of co-medication on infusion rate normalized plasma levels of curcumin and THC in cancer patients and to compare the plasma levels of curcumin and THC at different infusion rates between cancer patients and healthy individuals. In vitro hepatocyte and red blood cell distribution experiments were conducted with Lipocurc™ to support clinical findings. Plasma concentration time data were analyzed by the non-compartmental method to determine and compare the pharmacokinetic parameters of curcumin in cancer patients and healthy individuals. RESULTS Of 44 co-medications studied, three medications targeting the renin-angiotensin system, Lisinopril, Ramipril, and Valsartan elevated plasma levels of curcumin and THC in three cancer patients infused with Lipocurc™. Cell distribution experiments indicated that the disposition of curcumin in red blood cells may be a target for elevation of the plasma levels of curcumin. Plasma levels of curcumin in cancer patients increased to a greater extent with increased infusion rate compared to healthy individuals. Upon termination of infusion, the elimination phase for curcumin was shorter with a shorter terminal half-life and smaller volume of distribution for curcumin in cancer patients compared to healthy individuals. CONCLUSION Either co-medications or health status, or both, can impact the pharmacokinetics of curcumin infusion (as Lipocurc™) in cancer patients.
Collapse
Affiliation(s)
- Gordon T Bolger
- Nucro-Technics, 2000 Ellesmere Road, Unit 16, Scarborough, ON, M1H 2W4, Canada.
| | - Albert Licollari
- Nucro-Technics, 2000 Ellesmere Road, Unit 16, Scarborough, ON, M1H 2W4, Canada
| | - Amin Tan
- Nucro-Technics, 2000 Ellesmere Road, Unit 16, Scarborough, ON, M1H 2W4, Canada
| | - Richard Greil
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Brigitta Vcelar
- Polymun Scientific Immunbiologische Forschung GmbH, Klosterneuburg, Austria
| | - Sigrun Greil-Ressler
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
| | - Lukas Weiss
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
| | - Charlotte Schönlieb
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
| | - Teresa Magnes
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
| | - Bianca Radl
- III rd Medical Department, Paracelsus Medical University Salzburg, Salzburg, Austria
- Salzburg Cancer Research Institute-Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Salzburg, Austria
| | | | - Peter P Sordillo
- SignPath Pharma, Inc, 51 East 82 Street, #1A, New York, NY, 10028, USA.
| |
Collapse
|
9
|
Qi X, Wagenaar E, Xu W, Huang K, Schinkel AH. Ochratoxin A transport by the human breast cancer resistance protein (BCRP), multidrug resistance protein 2 (MRP2), and organic anion-transporting polypeptides 1A2, 1B1 and 2B1. Toxicol Appl Pharmacol 2017; 329:18-25. [PMID: 28532671 DOI: 10.1016/j.taap.2017.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 01/12/2023]
Abstract
Ochratoxin A (OTA) is a fungal secondary metabolite that can contaminate various foods. OTA has several toxic effects like nephrotoxicity, hepatotoxicity, and neurotoxicity in different animal species, but its mechanisms of toxicity are still unclear. How OTA accumulates in kidney, liver, and brain is as yet unknown, but transmembrane transport proteins are likely involved. We studied transport of OTA in vitro, using polarized MDCKII cells transduced with cDNAs of the efflux transporters mouse (m)Bcrp, human (h)BCRP, mMrp2, or hMRP2, and HEK293 cells overexpressing cDNAs of the human uptake transporters OATP1A2, OATP1B1, OATP1B3, or OATP2B1 at pH7.4 and 6.4. MDCKII-mBcrp cells were more resistant to OTA toxicity than MDCKII parental and hBCRP-transduced cells. Transepithelial transport experiments showed some apically directed transport by MDCKII-mBcrp cells at pH7.4, whereas both mBcrp and hBCRP clearly transported OTA at pH6.4. There was modest transport of OTA by mMrp2 and hMRP2 only at pH6.4. OATP1A2 and OATP2B1 mediated uptake of OTA both at pH7.4 and 6.4, but OATP1B1 only at pH7.4. There was no detectable transport of OTA by OATP1B3. Our data indicate that human BCRP and MRP2 can mediate elimination of OTA from cells, thus reducing OTA toxicity. On the other hand, human OATP1A2, OATP1B1, and OATP2B1 can mediate cellular uptake of OTA, which could aggravate OTA toxicity.
Collapse
Affiliation(s)
- Xiaozhe Qi
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Els Wagenaar
- Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| | - Alfred H Schinkel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Zaïr ZM, Singer DR. Influx transporter variants as predictors of cancer chemotherapy-induced toxicity: systematic review and meta-analysis. Pharmacogenomics 2016; 17:1189-1205. [PMID: 27380948 DOI: 10.2217/pgs-2015-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM Chemotherapeutic agents have been shown to increase lung patient survival, however their use may be limited by their serious adverse effects. We aimed to assess int impact of pharmacogenetic variation of influx transporters on inter-individual patient variation in adverse drug reactions. PATIENTS & METHODS We conducted a meta-analysis and systemic review and identified 16 publications, totaling 1510 patients, to be eligible for review. RESULTS Meta-analysis showed east-Asian patients expressing SLCO1B1 521T>C or 1118G>A to have a two- to fourfold increased risk of irinotecan-induced neutropenia but not diarrhea. American patients, expressing SLC19A1 IVS2(4935) G>A, were further associated with pemetrexed/gemcitabine-induced grade 3+ leukopenia. CONCLUSION Future studies should look to robust validation of SLCO1B1 and SLC19A1 as prognostic markers in the management of lung cancer patients.
Collapse
Affiliation(s)
| | - Donald Rj Singer
- Yale University School of Medicine, New Haven, CT, USA.,Fellowship of Postgraduate Medicine 11 Chandos Street, London, UK
| |
Collapse
|
11
|
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
|
12
|
Rubinchik-Stern M, Shmuel M, Bar J, Eyal S, Kovo M. Maternal-fetal transfer of indocyanine green across the perfused human placenta. Reprod Toxicol 2016; 62:100-5. [PMID: 27132189 DOI: 10.1016/j.reprotox.2016.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 04/04/2016] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
Abstract
Indocyanine green (ICG) is an FDA-approved near-infrared imaging probe, given also to pregnant women. We aimed to characterize ICG's transplacental transfer using the ex-vivo perfusion model. Placentas were obtained from caesarean deliveries. Cotyledons were cannulated and dually perfused. ICG, 9.6μg/mL and antipyrine (50μg/mL) were added to the maternal circulation in the absence (n=4) or the presence of the organic anion transporting polypeptide (OATPs) inhibitor rifampin (10μg/mL; n=5) or the P-glycoprotein inhibitor valspodar (2μg/mL; n=3). ICG's maternal-to-fetal transfer was evaluated over 180min. The cumulative percent of ICG in the fetal reservoir was minor. When ICG transfer was normalized to that of antipyrine, it was lower in the presence of rifampin (a 41% decrease; p<0.05). Valspodar did not appear to modify the kinetics of ICG. ICG's transplacental transfer is minimal and is probably OATP-mediated. The placenta is an effective protective barrier to ICG's distribution into the fetus.
Collapse
Affiliation(s)
- Miriam Rubinchik-Stern
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| | - Miriam Shmuel
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| | - Jacob Bar
- Department of Obstetrics and Gynecology, Edith Wolfson Medical Center, Holon, Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel.
| | - Michal Kovo
- Department of Obstetrics and Gynecology, Edith Wolfson Medical Center, Holon, Affiliated with Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
13
|
MacLeod AK, McLaughlin LA, Henderson CJ, Wolf CR. Activation status of the pregnane X receptor influences vemurafenib availability in humanized mouse models. Cancer Res 2015; 75:4573-81. [PMID: 26363009 PMCID: PMC4634205 DOI: 10.1158/0008-5472.can-15-1454] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/04/2015] [Indexed: 12/20/2022]
Abstract
Vemurafenib is a revolutionary treatment for melanoma, but the magnitude of therapeutic response is highly variable, and the rapid acquisition of resistance is frequent. Here, we examine how vemurafenib disposition, particularly through cytochrome P450-mediated oxidation pathways, could potentially influence these outcomes using a panel of knockout and transgenic humanized mouse models. We identified CYP3A4 as the major enzyme involved in the metabolism of vemurafenib in in vitro assays with human liver microsomes. However, mice expressing human CYP3A4 did not process vemurafenib to a greater extent than CYP3A4-null animals, suggesting that other pregnane X receptor (PXR)-regulated pathways may contribute more significantly to vemurafenib metabolism in vivo. Activation of PXR, but not of the closely related constitutive androstane receptor, profoundly reduced circulating levels of vemurafenib in humanized mice. This effect was independent of CYP3A4 and was negated by cotreatment with the drug efflux transporter inhibitor elacridar. Finally, vemurafenib strongly induced PXR activity in vitro, but only weakly induced PXR in vivo. Taken together, our findings demonstrate that vemurafenib is unlikely to exhibit a clinically significant interaction with CYP3A4, but that modulation of bioavailability through PXR-mediated regulation of drug transporters (e.g., by other drugs) has the potential to markedly influence systemic exposure and thereby therapeutic outcomes.
Collapse
Affiliation(s)
- A Kenneth MacLeod
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, United Kingdom
| | - Lesley A McLaughlin
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, United Kingdom
| | - Colin J Henderson
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, United Kingdom
| | - C Roland Wolf
- Division of Cancer, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, United Kingdom.
| |
Collapse
|
14
|
Bishara A, Meir M, Portnoy E, Shmuel M, Eyal S. Near Infrared Imaging of Indocyanine Green Distribution in Pregnant Mice and Effects of Concomitant Medications. Mol Pharm 2015; 12:3351-7. [DOI: 10.1021/acs.molpharmaceut.5b00374] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ameer Bishara
- Institute
for Drug Research,
School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Meir
- Institute
for Drug Research,
School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Emma Portnoy
- Institute
for Drug Research,
School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miri Shmuel
- Institute
for Drug Research,
School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sara Eyal
- Institute
for Drug Research,
School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
15
|
Da Silva CG, Honeywell RJ, Dekker H, Peters GJ. Physicochemical properties of novel protein kinase inhibitors in relation to their substrate specificity for drug transporters. Expert Opin Drug Metab Toxicol 2015; 11:703-717. [PMID: 25633410 DOI: 10.1517/17425255.2015.1006626] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Small molecule tyrosine and serine-threonine kinase inhibitors (TKIs and STKIs) are emerging drugs that interfere with downstream signaling pathways involved in cancer proliferation, invasion, metastasis and angiogenesis. The understanding of their pharmacokinetics, the identification of their transporters and the modulating activity exerted on transporters is pivotal to predict therapy efficacy and to avoid unwarranted drug treatment combinations. AREAS COVERED Experimental or in silico data were collected and summarized on TKIs and STKIs physico-chemical properties, which influence their transport, metabolism and efficacy, and TKIs and STKIs as influx transporter substrates and inhibitors. In addition, the uptake by tumor cell influx transporters and some factors in the tumor microenvironment affecting the uptake of TKIs and STKIs by cancer cells are briefly covered. EXPERT OPINION Membrane transporters play an important role in the pharmacokinetics and hence the efficacy of anticancer drugs, including TKIs and STKIs. These drugs are substrates and inhibitors of various transporters. Drug resistance may be bypassed not only by identifying the proper transporter but also by selective combinations, which may either downregulate or increase transporter activity. However, care has to be taken because this profile might be disease, drug and patient specific.
Collapse
Affiliation(s)
- Candido G Da Silva
- VU University Medical Center, Department of Medical Oncology , PO Box 7057, 1007 MB Amsterdam , The Netherlands
| | | | | | | |
Collapse
|
16
|
Bai RY, Staedtke V, Wanjiku T, Rudek MA, Joshi A, Gallia GL, Riggins GJ. Brain Penetration and Efficacy of Different Mebendazole Polymorphs in a Mouse Brain Tumor Model. Clin Cancer Res 2015; 21:3462-3470. [PMID: 25862759 DOI: 10.1158/1078-0432.ccr-14-2681] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/27/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Mebendazole (MBZ), first used as an antiparasitic drug, shows preclinical efficacy in models of glioblastoma and medulloblastoma. Three different mebendazole polymorphs (A, B, and C) exist, and a detailed assessment of the brain penetration, pharmacokinetics, and antitumor properties of each individual mebendazole polymorph is necessary to improve mebendazole-based brain cancer therapy. EXPERIMENTAL DESIGN AND RESULTS In this study, various marketed and custom-formulated mebendazole tablets were analyzed for their polymorph content by IR spectroscopy and subsequently tested in an orthotopic GL261 mouse glioma model for efficacy and tolerability. The pharmacokinetics and brain concentration of mebendazole polymorphs and two main metabolites were analyzed by LC/MS. We found that polymorph B and C both increased survival in a GL261 glioma model, as B exhibited greater toxicity. Polymorph A showed no benefit. Polymorph B and C both reached concentrations in the brain that exceeded the IC₅₀ in GL261 cells 29-fold. In addition, polymorph C demonstrated an AUC₀₋₂₄h brain-to-plasma (B/P) ratio of 0.82, whereas B showed higher plasma AUC and lower B/P ratio. In contrast, polymorph A presented markedly lower levels in the plasma and brain. Furthermore, the combination with elacridar was able to significantly improve the efficacy of polymorph C in GL261 glioma and D425 medulloblastoma models in mice. CONCLUSIONS Among mebendazole polymorphs, C reaches therapeutically effective concentrations in the brain tissue and tumor with fewer side effects, and is the better choice for brain cancer therapy. Its efficacy can be further enhanced by combination with elacridar.
Collapse
Affiliation(s)
- Ren-Yuan Bai
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Verena Staedtke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teresia Wanjiku
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michelle A Rudek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Avadhut Joshi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gary L Gallia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gregory J Riggins
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Lee CA, O'Connor MA, Ritchie TK, Galetin A, Cook JA, Ragueneau-Majlessi I, Ellens H, Feng B, Taub ME, Paine MF, Polli JW, Ware JA, Zamek-Gliszczynski MJ. Breast cancer resistance protein (ABCG2) in clinical pharmacokinetics and drug interactions: practical recommendations for clinical victim and perpetrator drug-drug interaction study design. Drug Metab Dispos 2015; 43:490-509. [PMID: 25587128 DOI: 10.1124/dmd.114.062174] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Breast cancer resistance protein (BCRP; ABCG2) limits intestinal absorption of low-permeability substrate drugs and mediates biliary excretion of drugs and metabolites. Based on clinical evidence of BCRP-mediated drug-drug interactions (DDIs) and the c.421C>A functional polymorphism affecting drug efficacy and safety, both the US Food and Drug Administration and European Medicines Agency recommend preclinical evaluation and, when appropriate, clinical assessment of BCRP-mediated DDIs. Although many BCRP substrates and inhibitors have been identified in vitro, clinical translation has been confounded by overlap with other transporters and metabolic enzymes. Regulatory recommendations for BCRP-mediated clinical DDI studies are challenging, as consensus is lacking on the choice of the most robust and specific human BCRP substrates and inhibitors and optimal study design. This review proposes a path forward based on a comprehensive analysis of available data. Oral sulfasalazine (1000 mg, immediate-release tablet) is the best available clinical substrate for intestinal BCRP, oral rosuvastatin (20 mg) for both intestinal and hepatic BCRP, and intravenous rosuvastatin (4 mg) for hepatic BCRP. Oral curcumin (2000 mg) and lapatinib (250 mg) are the best available clinical BCRP inhibitors. To interrogate the worst-case clinical BCRP DDI scenario, study subjects harboring the BCRP c.421C/C reference genotype are recommended. In addition, if sulfasalazine is selected as the substrate, subjects having the rapid acetylator phenotype are recommended. In the case of rosuvastatin, subjects with the organic anion-transporting polypeptide 1B1 c.521T/T genotype are recommended, together with monitoring of rosuvastatin's cholesterol-lowering effect at baseline and DDI phase. A proof-of-concept clinical study is being planned by a collaborative consortium to evaluate the proposed BCRP DDI study design.
Collapse
Affiliation(s)
- Caroline A Lee
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Meeghan A O'Connor
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Tasha K Ritchie
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Aleksandra Galetin
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Jack A Cook
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Isabelle Ragueneau-Majlessi
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Harma Ellens
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Bo Feng
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Mitchell E Taub
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Mary F Paine
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Joseph W Polli
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Joseph A Ware
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| | - Maciej J Zamek-Gliszczynski
- Drug Metabolism and Pharmacokinetics, QPS LLC, Research Triangle Park, North Carolina (C.A.L.); Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (M.A.O., M.E.T.); Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, The University of Manchester, Manchester, United Kingdom (A.G.); Pharmacokinetics and Drug Metabolism (B.F.) and Clinical Pharmacology, Global Innovative Pharma Business (J.A.C.), Pfizer Inc., Groton, Connecticut; School of Pharmacy, University of Washington, Seattle, Washington (I.R.-M., T.K.R.); Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park, North Carolina (M.J.Z.-G., J.W.P.) and King of Prussia, Pennsylvania (H.E.); College of Pharmacy, Washington State University, Spokane, Washington (M.F.P.); and Clinical Pharmacology, Genentech, South San Francisco, California (J.A.W.)
| |
Collapse
|
18
|
Thakkar N, Lockhart AC, Lee W. Role of Organic Anion-Transporting Polypeptides (OATPs) in Cancer Therapy. AAPS JOURNAL 2015; 17:535-45. [PMID: 25735612 DOI: 10.1208/s12248-015-9740-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 02/11/2015] [Indexed: 12/31/2022]
Abstract
The superfamily of organic anion-transporting polypeptides (OATPs, gene symbol SLCO) includes important transporters handling a variety of endogenous and xenobiotic substrates. Currently, 11 human OATPs are known and their substrates include endogenous hormones and their conjugates, anticancer drugs, and imaging agents. The contribution of OATPs to the in vivo disposition of these substrates has been extensively investigated. An accumulating body of evidence also indicates that the expression of some OATPs may be up- or downregulated in several types of cancers, suggesting potential pathogenic roles during the development and progression of cancer. Given that the role of OATPs in handling cancer therapeutics has been already covered by several excellent reviews, this review will focus on the recent progresses on the topic, in particular the role of OATPs in the disposition of anticancer drugs, the impact of OATP genetic variations on the function of OATPs, and the OATPs differentially expressed in cancer and their potential roles in cancer development, progression, and treatment.
Collapse
Affiliation(s)
- Nilay Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
| | | | | |
Collapse
|
19
|
Li Q, Liu T, Li Y, Luo S, Zhu Q, Zhang L, Zhao T. Transport and killing mechanism of a novel camptothecin-deoxycholic acid derivate on hepatocellular carcinoma cells. J Drug Target 2014; 22:543-52. [PMID: 24725118 DOI: 10.3109/1061186x.2014.906603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Camptothecin-20(s)-O-glycine ester-[N-(3'α, 12'α-dihydroxy-24'-carbonyl-5'β-cholan)] (A2), 10-(3'α,12'α-dihydroxy-5'β-cholan-24'-carboxyl)-(20 s)-camptothecin (C2), and 10-O-(3-O-(3'α, 12'α-dihydroxy-24'-carbonyl-5'β-cholan)-propyl)-(20S)-camptothecin (D2) are novel camptothecin-deoxycholic acid analogues. MTT assays were performed to assess the anticancer activity of these compounds against hepatocellular carcinoma SMMC-7721, breast carcinoma MCF-7, and colorectal carcinoma HCT-116 cells. A2 had a high killing ability on SMMC-7721 cells selectively, but C2 and D2 did not exhibit selectivity with regard to SMMC-7721 killing. Uptake assays were performed in an effort to elucidate the transport mechanisms of A2 into SMMC-7721 cells. A2 increased the mRNA expression of OATP1B3 (an organic anion-transporting polypeptide) and uptake of A2 was inhibited by rifampin (inhibitor of OATP1B3), which indicated that the transporter-mediated transport of A2 was mediated by OATP1B3. In addition, according to the western blot and apoptosis assays, we found that A2 killed SMMC-7721 cells by inducing cell apoptosis mainly via an AIF (apoptosis-inducing factor) pathway and a caspase-dependent mitochondria apoptosis pathway.
Collapse
Affiliation(s)
- Qingyong Li
- College of Pharmaceutical Science, Zhejiang University of Technology , Hangzhou , China
| | | | | | | | | | | | | |
Collapse
|
20
|
OATP1B1-related drug-drug and drug-gene interactions as potential risk factors for cerivastatin-induced rhabdomyolysis. Pharmacogenet Genomics 2014; 23:355-64. [PMID: 23652407 DOI: 10.1097/fpc.0b013e3283620c3b] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Genetic variation in drug metabolizing enzymes and membrane transporters as well as concomitant drug therapy can modulate the beneficial and the deleterious effects of drugs. We investigated whether patients exhibiting rhabdomyolysis who were taking cerivastatin possess functional genetic variants in SLCO1B1 and whether they were on concomitant medications that inhibit OATP1B1, resulting in accumulation of cerivastatin. METHODS This study had three components: (a) resequencing the SLCO1B1 gene in 122 patients who developed rhabdomyolysis while on cerivastatin; (b) functional evaluation of the identified SLCO1B1 nonsynonymous variants and haplotypes in in-vitro HEK293/FRT cells stably transfected with pcDNA5/FRT empty vector, SLCO1B1 reference, variants, and haplotypes; and (c) in-vitro screening of 15 drugs commonly used among the rhabdomyolysis cases for inhibition of OATP1B1-mediated uptake of cerivastatin in HEK293/FRT cells stably transfected with reference SLCO1B1. RESULTS The resequencing of the SLCO1B1 gene identified 54 variants. In-vitro functional analysis of SLCO1B1 nonsynonymous variants and haplotypes showed that the V174A, R57Q, and P155T variants, a novel frameshift insertion, OATP1B1*14 and OATP1B1*15 haplotype were associated with a significant reduction (P<0.001) in cerivastatin uptake (32, 18, 72, 3.4, 2.1 and 5.7% of reference, respectively). Furthermore, clopidogrel and seven other drugs were shown to inhibit OATP1B1-mediated uptake of cerivastatin. CONCLUSION Reduced function of OATP1B1 related to genetic variation and drug-drug interactions likely contributed to cerivastatin-induced rhabdomyolysis. Although cerivastatin is no longer in clinical use, these findings may translate to related statins and other substrates of OATP1B1.
Collapse
|
21
|
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
|
22
|
Abstract
Organic anion-transporting polypeptides (OATPs) encoded by the SLCO genes constitute an important transporter superfamily that mediates transmembrane transport of various clinical drugs and endogenous nutrients. Eleven human OATPs with different transport functions are expressed in various tissues. Bile acids, steroid hormone conjugates, prostaglandins, testosterone and thyroid hormones that promote cell proliferation are typical substrates of OATPs. Many important clinical drugs have been identified as substrates of OATP1B1, OATP1B3, OATP2B1 and OATP1A2. Liver-specific OATP1B1 and OATP1B3 as well as testis-specific OATP6A1 are expressed in malignancies and can act as biomarkers for many tumours. Various studies have shown the associations of genetic polymorphisms in OATP genes with the uptake pharmacokinetics of their substrates. Because of their abundant expression in tumours and their high transport activity for many cancer drugs, OATPs should be considered as important therapeutic targets in anti-cancer drug design.
Collapse
Affiliation(s)
- Tianyu Liu
- State Engineering Laboratory of Bio-Resources Eco-Utilization, Northeast Forestry University, Ministry of Education , Harbin , China and
| | | |
Collapse
|
23
|
Tsakalozou E, Adane ED, Kuo KL, Daily A, Moscow JA, Leggas M. The effect of breast cancer resistance protein, multidrug resistant protein 1, and organic anion-transporting polypeptide 1B3 on the antitumor efficacy of the lipophilic camptothecin 7-t-butyldimethylsilyl-10-hydroxycamptothecin (AR-67) in vitro. Drug Metab Dispos 2013; 41:1404-13. [PMID: 23620484 PMCID: PMC3684821 DOI: 10.1124/dmd.112.050021] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 04/24/2013] [Indexed: 11/22/2022] Open
Abstract
AR-67 (7-t-butyldimethylsilyl-10-hydroxycamptothecin) is a lipophilic camptothecin analog, currently under early stage clinical trials. Transporters are known to have an impact on the disposition of camptothecins and on the response to chemotherapeutics in general due to their expression in tumor tissues. Therefore, we investigated the interplay between the breast cancer resistance protein (BCRP), multidrug resistant protein 1 (MDR1), and organic anion-transporting polypeptide (OATP) 1B1/1B3 transporters and AR-67 and their impact on the toxicity profile of AR-67. Using cell lines expressing the aforementioned transporters, we showed that the lipophilic AR-67 lactone form is a substrate for efflux transporters BCRP and MDR1. Additionally, OATP1B1 and OATP1B3 facilitated the uptake of AR-67 carboxylate in SLCO1B1- and SLCO1B3-transfected cell systems compared with the mock-transfected ones. Notably, both BCRP and MDR1 conferred resistance to AR-67 lactone. Prompted by recent studies showing increased OATP1B3 expression in certain cancer types, we investigated the effect of OATP1B3 expression on cell viability after exposure to AR-67 carboxylate. OATP1B3-expressing cells had increased carboxylate uptake as compared with mock-transfected cells but were not sensitized because the intracellular amount of lactone was 50-fold higher than that of carboxylate and comparable between OATP1B3-expressing and OATP1B3-nonexpressing cells. In conclusion, BCRP- and MDR1-mediated efflux of AR-67 lactone confers resistance to AR-67, but OATP1B3-mediated uptake of the AR-67 carboxylate does not sensitize OATP1B3-expressing tumor cells.
Collapse
Affiliation(s)
- Eleftheria Tsakalozou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | | | | | | | | | | |
Collapse
|
24
|
Liu L, Unadkat JD. Interaction between HIV protease inhibitors (PIs) and hepatic transporters in sandwich cultured human hepatocytes: implication for PI-based DDIs. Biopharm Drug Dispos 2013; 34:155-64. [PMID: 23280499 DOI: 10.1002/bdd.1832] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 11/07/2022]
Abstract
Although HIV protease inhibitors (PIs) produce profound metabolic interactions through inactivation/inhibition of CYP3A enzymes, their role as victims of transporter-based drug-drug interactions (DDIs) is less well understood. Therefore, this study investigated if the PIs, nelfinavir (NFV), ritonavir (RTV), lopinavir (LPV) or amprenavir (APV) were transported into sandwich-cultured human hepatocytes (SCHH), and whether OATPs contributed to this transport. The findings showed that, except for (3) H-APV, no significant decrease in the total hepatocyte accumulation of the (3) H-PIs was detected in the presence of the corresponding unlabeled PI, indicating that the uptake of the other PIs was not mediated. Further, hepatocyte biliary efflux studies using (3) H-APV and unlabeled APV confirmed this decrease to be due to inhibition of sinusoidal influx transporter(s) and not the canalicular efflux transporters. Moreover, this sinusoidal transport of APV was not OATP-mediated. The results indicate that the hepatic uptake of NFV, RTV or LPV was primarily mediated by passive diffusion. The hepatic uptake of APV was mediated by an unidentified sinusoidal transporter(s). Therefore, NFV, RTV or LPV will not be victims of DDIs involving inhibition of hepatic influx transporters; however, the disposition of APV may be affected if its sinusoidal transport is inhibited.
Collapse
Affiliation(s)
- Li Liu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
25
|
Tumor-specific expression of organic anion-transporting polypeptides: transporters as novel targets for cancer therapy. JOURNAL OF DRUG DELIVERY 2013; 2013:863539. [PMID: 23431456 PMCID: PMC3574750 DOI: 10.1155/2013/863539] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 12/24/2012] [Indexed: 01/16/2023]
Abstract
Members of the organic anion transporter family (OATP) mediate the transmembrane uptake of clinical important drugs and hormones thereby affecting drug disposition and tissue penetration. Particularly OATP subfamily 1 is known to mediate the cellular uptake of anticancer drugs (e.g., methotrexate, derivatives of taxol and camptothecin, flavopiridol, and imatinib). Tissue-specific expression was shown for OATP1B1/OATP1B3 in liver, OATP4C1 in kidney, and OATP6A1 in testis, while other OATPs, for example, OATP4A1, are expressed in multiple cells and organs. Many different tumor entities show an altered expression of OATPs. OATP1B1/OATP1B3 are downregulated in liver tumors, but highly expressed in cancers in the gastrointestinal tract, breast, prostate, and lung. Similarly, testis-specific OATP6A1 is expressed in cancers in the lung, brain, and bladder. Due to their presence in various cancer tissues and their limited expression in normal tissues, OATP1B1, OATP1B3, and OATP6A1 could be a target for tumor immunotherapy. Otherwise, high levels of ubiquitous expressed OATP4A1 are found in colorectal cancers and their metastases. Therefore, this OATP might serve as biomarkers for these tumors. Expression of OATP is regulated by nuclear receptors, inflammatory cytokines, tissue factors, and also posttranslational modifications of the proteins. Through these processes, the distribution of the transporter in the tissue will be altered, and a shift from the plasma membrane to cytoplasmic compartments is possible. It will modify OATP uptake properties and, subsequently, change intracellular concentrations of drugs, hormones, and various other OATP substrates. Therefore, screening tumors for OATP expression before therapy should lead to an OATP-targeted therapy with higher efficacy and decreased side effects.
Collapse
|
26
|
Gong IY, Kim RB. Impact of Genetic Variation in OATP Transporters to Drug Disposition and Response. Drug Metab Pharmacokinet 2013; 28:4-18. [DOI: 10.2133/dmpk.dmpk-12-rv-099] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Portnoy E, Gurina M, Magdassi S, Eyal S. Evaluation of the Near Infrared Compound Indocyanine Green as a Probe Substrate of P-Glycoprotein. Mol Pharm 2012; 9:3595-601. [DOI: 10.1021/mp300472y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Emma Portnoy
- Institute
of Drug Research, School of Pharmacy, Faculty of Medicine, and ‡Casali Institute,
Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem,
Jerusalem, Israel
| | - Marina Gurina
- Institute
of Drug Research, School of Pharmacy, Faculty of Medicine, and ‡Casali Institute,
Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem,
Jerusalem, Israel
| | - Shlomo Magdassi
- Institute
of Drug Research, School of Pharmacy, Faculty of Medicine, and ‡Casali Institute,
Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem,
Jerusalem, Israel
| | - Sara Eyal
- Institute
of Drug Research, School of Pharmacy, Faculty of Medicine, and ‡Casali Institute,
Institute of Chemistry and Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem,
Jerusalem, Israel
| |
Collapse
|
28
|
Roth M, Obaidat A, Hagenbuch B. OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J Pharmacol 2012; 165:1260-87. [PMID: 22013971 DOI: 10.1111/j.1476-5381.2011.01724.x] [Citation(s) in RCA: 574] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The human organic anion and cation transporters are classified within two SLC superfamilies. Superfamily SLCO (formerly SLC21A) consists of organic anion transporting polypeptides (OATPs), while the organic anion transporters (OATs) and the organic cation transporters (OCTs) are classified in the SLC22A superfamily. Individual members of each superfamily are expressed in essentially every epithelium throughout the body, where they play a significant role in drug absorption, distribution and elimination. Substrates of OATPs are mainly large hydrophobic organic anions, while OATs transport smaller and more hydrophilic organic anions and OCTs transport organic cations. In addition to endogenous substrates, such as steroids, hormones and neurotransmitters, numerous drugs and other xenobiotics are transported by these proteins, including statins, antivirals, antibiotics and anticancer drugs. Expression of OATPs, OATs and OCTs can be regulated at the protein or transcriptional level and appears to vary within each family by both protein and tissue type. All three superfamilies consist of 12 transmembrane domain proteins that have intracellular termini. Although no crystal structures have yet been determined, combinations of homology modelling and mutation experiments have been used to explore the mechanism of substrate recognition and transport. Several polymorphisms identified in members of these superfamilies have been shown to affect pharmacokinetics of their drug substrates, confirming the importance of these drug transporters for efficient pharmacological therapy. This review, unlike other reviews that focus on a single transporter family, briefly summarizes the current knowledge of all the functionally characterized human organic anion and cation drug uptake transporters of the SLCO and the SLC22A superfamilies.
Collapse
Affiliation(s)
- Megan Roth
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | |
Collapse
|
29
|
Karlgren M, Vildhede A, Norinder U, Wisniewski JR, Kimoto E, Lai Y, Haglund U, Artursson P. Classification of inhibitors of hepatic organic anion transporting polypeptides (OATPs): influence of protein expression on drug-drug interactions. J Med Chem 2012; 55:4740-63. [PMID: 22541068 PMCID: PMC3361267 DOI: 10.1021/jm300212s] [Citation(s) in RCA: 267] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
The hepatic organic anion transporting polypeptides (OATPs)
influence the pharmacokinetics of several drug classes and are involved
in many clinical drug–drug interactions. Predicting potential
interactions with OATPs is, therefore, of value. Here, we developed
in vitro and in silico models for identification and prediction of
specific and general inhibitors of OATP1B1, OATP1B3, and OATP2B1.
The maximal transport activity (MTA) of each OATP in human liver was
predicted from transport kinetics and protein quantification. We then
used MTA to predict the effects of a subset of inhibitors on atorvastatin
uptake in vivo. Using a data set of 225 drug-like compounds, 91 OATP
inhibitors were identified. In silico models indicated that lipophilicity
and polar surface area are key molecular features of OATP inhibition.
MTA predictions identified OATP1B1 and OATP1B3 as major determinants
of atorvastatin uptake in vivo. The relative contributions to overall
hepatic uptake varied with isoform specificities of the inhibitors.
Collapse
Affiliation(s)
- Maria Karlgren
- Department of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Ueno Y, Matsuda H, Mizutani H, Iwamoto T, Okuda M. Involvement of Specific Transport System on Uptake of Lactone Form of SN-38 in Human Intestinal Epithelial Cell Line Caco-2. Biol Pharm Bull 2012; 35:54-8. [DOI: 10.1248/bpb.35.54] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yusuke Ueno
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Hiroko Matsuda
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Hideki Mizutani
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
- College of Pharmacy, Kinjo Gakuin University
| | - Takuya Iwamoto
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| | - Masahiro Okuda
- Department of Clinical Pharmacy and Biopharmaceutics, Graduate School of Medicine, Mie University
| |
Collapse
|
31
|
Hirose K, Kozu C, Yamashita K, Maruo E, Kitamura M, Hasegawa J, Omoda K, Murakami T, Maeda Y. Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Oncol Lett 2011; 3:694-698. [PMID: 22740978 DOI: 10.3892/ol.2011.533] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/16/2011] [Indexed: 12/29/2022] Open
Abstract
In irinotecan (CPT-11)-based chemotherapy, neutropenia and diarrhea are often induced. In the present study, the clinical significance of the concentration ratios of 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronide (SN-38G) and SN-38 in the plasma in predicting CPT-11-induced neutropenia was examined. A total of 17 patients with colorectal cancer and wild-type UDP-glucuronosyltransferase (UGT)1A1 gene were enrolled and treated with CPT-11 as part of the FOLFIRI regimen [CPT-11 and fluorouracil (5-FU)]. Blood was taken exactly 15 min following a 2-h continuous infusion of CPT-11. Plasma concentrations of SN-38, SN-38G and CPT-11 were determined by a modified high-performance liquid chromatography (HPLC) method. The median, maximum and minimum values of plasma SN-38G/SN-38 ratios were 4.25, 7.09 and 1.03, respectively, indicating that UGT activities are variable among patients with the wild-type UGT1A1 gene. The plasma SN-38G/SN-38 ratios decreased with an increase in the trial numbers of chemotherapy (r=0.741, p=0.000669), suggesting that CPT-11 treatment suppresses UGT activity, and the low plasma SN-38G/SN-38 ratios resulted in the induction of greater neutropenia. However, in this analysis, 2 clearly separated regression lines were observed between plasma SN-38G/SN-38 ratios and neutropenia induction. In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. One-point determination of the plasma SN-38G/SN-38 ratio may provide indications for the prediction of CPT-11-induced neutropenia and adjustment of the optimal dose, although further studies are required.
Collapse
Affiliation(s)
- Koichi Hirose
- Department of Pharmacy, Osaka Rosai Hospital, Sakai, Osaka 591-8025
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Obaidat A, Roth M, Hagenbuch B. The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu Rev Pharmacol Toxicol 2011. [PMID: 21854228 DOI: 10.1146/annurev‐pharmtox‐010510‐100556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organic anion transporting polypeptides (OATPs) are members of the SLCO gene superfamily of proteins. The 11 human OATPs are classified into 6 families and subfamilies on the basis of their amino acid sequence similarities. OATPs are expressed in several epithelial tissues throughout the body and transport mainly amphipathic molecules with molecular weights of more than 300 kDa. Members of the OATP1 and OATP2 families are functionally the best-characterized OATPs. Among these are the multispecific OATP1A2, OATP1B1, OATP1B3, and OATP2B1. They transport various endo- and xenobiotics, including hormones and their conjugates as well as numerous drugs such as several anticancer agents. Recent reports demonstrate that some OATPs are up- or downregulated in several cancers and that OATP expression might affect cancer development. On the basis of the findings summarized in this review, we propose that OATPs could be valuable targets for anticancer therapy.
Collapse
Affiliation(s)
- Amanda Obaidat
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | | | | |
Collapse
|
33
|
Obaidat A, Roth M, Hagenbuch B. The expression and function of organic anion transporting polypeptides in normal tissues and in cancer. Annu Rev Pharmacol Toxicol 2011; 52:135-51. [PMID: 21854228 DOI: 10.1146/annurev-pharmtox-010510-100556] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Organic anion transporting polypeptides (OATPs) are members of the SLCO gene superfamily of proteins. The 11 human OATPs are classified into 6 families and subfamilies on the basis of their amino acid sequence similarities. OATPs are expressed in several epithelial tissues throughout the body and transport mainly amphipathic molecules with molecular weights of more than 300 kDa. Members of the OATP1 and OATP2 families are functionally the best-characterized OATPs. Among these are the multispecific OATP1A2, OATP1B1, OATP1B3, and OATP2B1. They transport various endo- and xenobiotics, including hormones and their conjugates as well as numerous drugs such as several anticancer agents. Recent reports demonstrate that some OATPs are up- or downregulated in several cancers and that OATP expression might affect cancer development. On the basis of the findings summarized in this review, we propose that OATPs could be valuable targets for anticancer therapy.
Collapse
Affiliation(s)
- Amanda Obaidat
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | | | | |
Collapse
|
34
|
Niemi M, Pasanen MK, Neuvonen PJ. Organic anion transporting polypeptide 1B1: a genetically polymorphic transporter of major importance for hepatic drug uptake. Pharmacol Rev 2011; 63:157-81. [PMID: 21245207 DOI: 10.1124/pr.110.002857] [Citation(s) in RCA: 481] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The importance of membrane transporters for drug pharmacokinetics has been increasingly recognized during the last decade. Organic anion transporting polypeptide 1B1 (OATP1B1) is a genetically polymorphic influx transporter expressed on the sinusoidal membrane of human hepatocytes, and it mediates the hepatic uptake of many endogenous compounds and xenobiotics. Recent studies have demonstrated that OATP1B1 plays a major, clinically important role in the hepatic uptake of many drugs. A common single-nucleotide variation (coding DNA c.521T>C, protein p.V174A, rs4149056) in the SLCO1B1 gene encoding OATP1B1 decreases the transporting activity of OATP1B1, resulting in markedly increased plasma concentrations of, for example, many statins, particularly of active simvastatin acid. The variant thereby enhances the risk of statin-induced myopathy and decreases the therapeutic indexes of statins. However, the effect of the SLCO1B1 c.521T>C variant is different on different statins. The same variant also markedly affects the pharmacokinetics of several other drugs. Furthermore, certain SLCO1B1 variants associated with an enhanced clearance of methotrexate increase the risk of gastrointestinal toxicity by methotrexate in the treatment of children with acute lymphoblastic leukemia. Certain drugs (e.g., cyclosporine) potently inhibit OATP1B1, causing clinically significant drug interactions. Thus, OATP1B1 plays a major role in the hepatic uptake of drugs, and genetic variants and drug interactions affecting OATP1B1 activity are important determinants of individual drug responses. In this article, we review the current knowledge about the expression, function, substrate characteristics, and pharmacogenetics of OATP1B1 as well as its role in drug interactions, in parts comparing with those of other hepatocyte-expressed organic anion transporting polypeptides, OATP1B3 and OATP2B1.
Collapse
Affiliation(s)
- Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, PO Box 20, Helsinki, FI-00014, Finland.
| | | | | |
Collapse
|
35
|
Fahrmayr C, Fromm MF, König J. Hepatic OATP and OCT uptake transporters: their role for drug-drug interactions and pharmacogenetic aspects. Drug Metab Rev 2010; 42:380-401. [PMID: 20100011 DOI: 10.3109/03602530903491683] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Uptake transporters in the basolateral membrane of hepatocytes are important for the hepatobiliary elimination of drugs. Further, since drug-metabolizing enzymes are located intracellularly, uptake into hepatocytes is a prerequisite for their subsequent metabolism. Therefore, alteration of uptake transporter function (e.g., by concomitantly administered drugs or due to functional consequences of genetic variations, leading to reduced transport function) may result in a change in drug pharmacokinetics. In this review, we focus on the hepatocellularly expressed members of the OATP and OCT family, their impact on transport-mediated drug-drug interactions, and on the functional consequences of variations in genes encoding these transporters.
Collapse
Affiliation(s)
- Christina Fahrmayr
- Department of Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | | | | |
Collapse
|
36
|
Adane ED, Liu Z, Xiang TX, Anderson BD, Leggas M. Factors Affecting the In Vivo Lactone Stability and Systemic Clearance of the Lipophilic Camptothecin Analogue AR-67. Pharm Res 2010; 27:1416-25. [DOI: 10.1007/s11095-010-0137-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 03/24/2010] [Indexed: 10/19/2022]
|
37
|
Kwak JO, Lee SH, Lee GS, Kim MS, Ahn YG, Lee JH, Kim SW, Kim KH, Lee MG. Selective inhibition of MDR1 (ABCB1) by HM30181 increases oral bioavailability and therapeutic efficacy of paclitaxel. Eur J Pharmacol 2009; 627:92-8. [PMID: 19903471 DOI: 10.1016/j.ejphar.2009.11.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 10/19/2009] [Accepted: 11/03/2009] [Indexed: 02/04/2023]
Abstract
Multi-drug resistance 1 (MDR1, ABCB1), also known as P-glycoprotein (P-gp), restricts intestinal uptake of many drugs, and contributes to cellular resistance to cancer chemotherapy. In this study, we examined the pharmacologic characteristics of HM30181, a newly developed MDR1 inhibitor, and tested its capacity to increase the oral bioavailability and efficacy of paclitaxel, an anti-cancer drug usually given by intravenous injection. In the ATPase assay using MDR1-enriched vesicles, HM30181 showed the highest potency (IC(50)=0.63nM) among several MDR1 inhibitors, including cycloporin A, XR9576, and GF120918, and effectively blocked transepithelial transport of paclitaxel in MDCK monolayers (IC(50)=35.4nM). The ATPase inhibitory activity of HM30181 was highly selective to MDR1. HM30181 did not inhibit MRP1 (ABCC1), MRP2 (ABCC2), and MRP3 (ABCC3), and partially inhibited BCRP (ABCG2) only at very high concentrations. Importantly, co-administration of HM30181 (10mg/kg) greatly increased oral bioavailability of paclitaxel from 3.4% to 41.3% in rats. Moreover, oral co-administration of paclitaxel and HM30181 showed a tumor-inhibitory strength equal or superior to that of intravenous paclitaxel in the xenograft model in nude mice. These results identify HM30181 as a highly selective and potent inhibitor of MDR1, which in combination with paclitaxel, may provide an orally effective anti-tumor regimen.
Collapse
Affiliation(s)
- Jin-Oh Kwak
- Department of Pharmacology, Brain Korea 21 Project for Medical Sciences, Institute of Gastroenterology, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul 120-752, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|