1
|
Chen J, Yao Y, Mao X, Chen Y, Ni F. Liver-targeted delivery based on prodrug: passive and active approaches. J Drug Target 2024; 32:1155-1168. [PMID: 39072411 DOI: 10.1080/1061186x.2024.2386416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The liver, a central organ in human metabolism, is often the primary target for drugs. However, conditions such as viral hepatitis, cirrhosis, non-alcoholic fatty liver disease (NAFLD), and hepatocellular carcinoma (HCC) present substantial health challenges worldwide. Existing treatments, which suffer from the non-specific distribution of drugs, frequently fail to achieve desired efficacy and safety, risking unnecessary liver harm and systemic side effects. PURPOSE The aim of this review is to synthesise the latest progress in the design of liver-targeted prodrugs, with a focus on passive and active targeting strategies, providing new insights into the development of liver-targeted therapeutic approaches. METHODS This study conducted an extensive literature search through databases like Google Scholar, PubMed, Web of Science, and China National Knowledge Infrastructure (CNKI), systematically collecting and selecting recent research on liver-targeted prodrugs. The focus was on targeting mechanisms, including the Enhanced Permeability and Retention (EPR) effect, the unique microenvironment of liver cancer, and active targeting through specific transporters and receptors. RESULTS Active targeting strategies achieve precise drug delivery by binding specific ligands to liver surface receptors. Passive targeting takes advantage of the EPR effect and tumour characteristics to enrich drugs in liver tumours. The review details successful cases of using small molecule ligands, peptides, antibodies and nanoparticles as drug carriers. CONCLUSION Liver-targeted prodrug strategies show great potential in enhancing the efficacy of drug treatment and reducing side effects for liver diseases. Future research should balance the advantages and limitations of both targeting strategies, focusing on optimising drug design and targeting efficiency, especially for clinical application. In-depth research on liver-specific receptors and the development of innovative targeting molecules are crucial for advancing the field of liver-targeted prodrugs.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingrui Yao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoran Mao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Martinez-Guerrero LJ, Zhang X, Wright SH, Cherrington NJ. Characterization of Human Organic Anion Transporter 4 (hOAT4) and Mouse Oat5 (mOat5) As Functional Orthologs for Renal Anion Uptake and Efflux Transport. J Pharmacol Exp Ther 2024; 391:378-386. [PMID: 38627096 PMCID: PMC11585314 DOI: 10.1124/jpet.123.001979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/09/2024] [Indexed: 11/21/2024] Open
Abstract
Organic anions (OAs) are compounds including drugs or toxicants that are negatively charged at physiologic pH and are typically transported by organic anion transporters (OATs). Human OAT4 (SLC22A11) is expressed in the apical membrane of renal proximal tubules. Although there is no rodent ortholog of hOAT4, rodents express Oat5 (Slc22a19), an anion exchanger that is also localized to the apical membrane of renal proximal tubule cells. The purpose of this study was to determine the functional similarity between mouse Oat5 and human OAT4. Chinese hamster ovary (CHO) cells expressing SLC22A11 or Slc22a19 were used to assess the transport characteristics of radiolabeled ochratoxin (OTA). We determined the kinetics of OTA transport; the resulting Michaelis constant (Kt) and maximal rate of mediated substrate transport (Jmax) values were very similar for both hOAT4 and mOat5: Kt 3.9 and 7.2 μM, respectively, and Jmax 4.4 and 3.9 pmol/cm2, respectively. For the profile of OTA inhibition by OAs, IC50 values were determined for several clinically important drugs and toxicants. The resulting IC50 values ranged from 9 μM for indomethacin to ∼600 μM for the diuretic hydrochlorothiazide. We measured the efflux of OTA from preloaded cells; both hOAT4 and mOat5 supported the efflux of OTA. These data support the hypothesis that OAT4 and Oat5 are functional orthologs and share selectivity for OTA both for reabsorption and secretion. SIGNIFICANCE STATEMENT: This study compares the selectivity profile between human organic anion transporter (OAT4) and mouse Oat5. Our data revealed a similar selectivity profile for ochratoxin A (OTA) reabsorption and secretion by these two transporters, thereby supporting the hypothesis that hOAT4 and mOat5, although not genetic orthologs, behave as functional orthologs for both uptake and efflux. These data will be instrumental in selecting an appropriate animal model when studying the renal disposition of anionic drugs and toxicants.
Collapse
Affiliation(s)
- Lucy J Martinez-Guerrero
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Xiaohong Zhang
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology (L.J.M.-G., X.Z., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona
| |
Collapse
|
3
|
Fattah S, Shinde AB, Baes M, Allegaert K, Augustijns P, Annaert P. Ontogeny of Hepatic Organic Cation Transporter-1 in Rat and Human. Drug Metab Dispos 2024; 52:1253-1261. [PMID: 39209551 DOI: 10.1124/dmd.124.001766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
The organic cation transporter (OCT)-1 mediates hepatic uptake of cationic endogenous compounds and xenobiotics. To date, limited information exists on how Oct1/OCT1 functionally develops with age in rat and human livers and how this would affect the pharmacokinetics of OCT substrates in children or juvenile animals. The functional ontogeny of rOct/hOCT was profiled in suspended rat (2-57 days old) and human hepatocytes (pediatric liver tissue donors: age 2-12 months) by determining uptake clearance of 4-[4-(dimethylamino)styryl]-N-methylpyridinium iodide (ASP+) as a known rOct/hOCT probe substrate. mRNA expression was determined in rat liver tissue corresponding to rat ages used in the functional studies, while hOCT1 mRNA expressions were determined in the same hepatocyte batches as those used for uptake studies. Maturation of rOct/hOCT activity and expression were evaluated by comparing values obtained at the various ages to the adult values. Relative to adult values (at 8 weeks), ASP+ uptake clearance in suspended rat hepatocytes aged 0, 1, 2, 3, 4, 5, and 6 weeks reached 26%, 29%, 33%, 37%, 72%, 63%, and 71%, respectively. Hepatic Oct1 mRNA expression was consistent with Oct activity (correlation coefficient of 0.92). In human hepatocytes, OCT1 activity was age dependent and also correlated with mRNA levels (correlation coefficient of 0.88). These data show that Oct1/OCT1 activities and expression mature gradually in rat/human liver, thereby mirroring the expression pattern of organic anion transporting polypeptide in rat. These high-resolution transporter ontogeny profiles will allow for more accurate prediction of the pharmacokinetics of OCT1/Oct1 substrates in pediatric populations and juvenile animals. SIGNIFICANCE STATEMENT: Organic cation transporter-1 (OCT1) represents a major drug uptake transporter in human liver. This study provides high-resolution data regarding the age-dependent function of OCT1 in the liver, based on in vitro experiments with rat and human hepatocytes obtained from donors between birth and adulthood. These ontogeny profiles will inform improved age-specific physiologically based pharmacokinetic models for OCT1 drug substrates in neonates, infants, children, and adults.
Collapse
Affiliation(s)
- Sarinj Fattah
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| | - Abhijit Babaji Shinde
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| | - Myriam Baes
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| | - Karel Allegaert
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| | - Patrick Augustijns
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| | - Pieter Annaert
- Drug Delivery and Disposition (S.F., Pa.A., Pi.A.), Laboratory of Cell Metabolism (A.B.S., M.B.), Clinical Pharmacology and Pharmacotherapy (K.A.), Department of Pharmaceutical and Pharmacological Sciences, and Department of Development and Regeneration (K.A.), KU Leuven, Leuven, Belgium; and Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands (K.A.)
| |
Collapse
|
4
|
Zeng X, Liao Y, Qiao X, Liang K, Luo Q, Deng M, Liu Y, Zhang W, Hong X, Xiao Y. Novel NIR-II fluorescent probes for biliary atresia imaging. Acta Pharm Sin B 2023; 13:4578-4590. [PMID: 37969732 PMCID: PMC10638547 DOI: 10.1016/j.apsb.2023.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/26/2023] [Accepted: 06/27/2023] [Indexed: 11/17/2023] Open
Abstract
Biliary atresia is a rare infant disease that predisposes patients to liver transplantation and death if not treated in time. However, early diagnosis is challenging because the clinical manifestations and laboratory tests of biliary atresia overlap with other cholestatic diseases. Therefore, it is very important to develop a simple, safe and reliable method for the early diagnosis of biliary atresia. Herein, a novel NIR-II fluorescence probe, HZL2, with high quantum yield, excellent biocompatibility, low cytotoxicity and rapid excretion through the liver and gallbladder was developed based on the oil/water partition coefficient and permeability. A simple fecal sample after injection of HZL2 can be used to efficiently identify the success of the mouse model of biliary atresia for the first time, allowing for an early diagnosis of the disease. This study not only developed a simple and safe method for the early diagnosis of biliary atresia with great potential in clinical translation but also provides a research tool for the development of pathogenesis and therapeutic medicines for biliary atresia.
Collapse
Affiliation(s)
- Xiaodong Zeng
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqin Liao
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xue Qiao
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, School of Ecology and Environment, Tibet University, Lhasa 850000, China
| | - Ke Liang
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
| | - Qiusi Luo
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Mingbo Deng
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Yishen Liu
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Weijing Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- School of Pharmacy, Yantai University, Yantai 264005, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Key Laboratory of Biodiversity and Environment on the Qinghai-Tibetan Plateau, Ministry of Education, School of Ecology and Environment, Tibet University, Lhasa 850000, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
| | - Yuling Xiao
- State Key Laboratory of Virology, Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| |
Collapse
|
5
|
Zhang X, Wright SH. Transport Turnover Rates for Human OCT2 and MATE1 Expressed in Chinese Hamster Ovary Cells. Int J Mol Sci 2022; 23:ijms23031472. [PMID: 35163393 PMCID: PMC8836179 DOI: 10.3390/ijms23031472] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
MATE1 (multidrug and toxin extruder 1) and OCT2 (organic cation transporter 2) play critical roles in organic cation excretion by the human kidney. The transporter turnover rate (TOR) is relevant to understanding both their transport mechanisms and interpreting the in vitro-in vivo extrapolation (IVIVE) required for physiologically-based pharmacokinetic (PBPK) modeling. Here, we use a quantitative western blot method to determine TORs for MATE1 and OCT2 proteins expressed in CHO cells. MATE1 and OCT2, each with a C-terminal V-5 epitope tag, were cell surface biotinylated and the amount of cell surface MATE1 and OCT2 protein was quantified by western analysis, using standard curves for the V5 epitope. Cell surface MATE1 and OCT2 protein represented 25% and 24%, respectively, of the total expression of these proteins in CHO cells. The number of cell surface transporters was ~55 fmol cm-2 for MATE1 and ~510 fmol cm-2 for OCT2. Dividing these values into the different Jmax values for transport of MPP, metformin, and atenolol mediated by MATE1 and OCT2 resulted in calculated TOR values (±SE, n = 4) of 84.0 ± 22.0 s-1 and 2.9 ± 0.6 s-1; metformin, 461.0 ± 121.0 s-1 and 12.6 ± 2.4 s-1; atenolol, 118.0 ± 31.0 s-1, respectively. These values are consistent with the TOR values determined for a variety of exchangers (NHEs), cotransporters (SGLTs, Lac permease), and uniporters (GLUTs, ENTs).
Collapse
|
6
|
Meher N, Seo K, Wang S, Bidkar AP, Fogarty M, Dhrona S, Huang X, Tang R, Blaha C, Evans MJ, Raleigh DR, Jun YW, VanBrocklin HF, Desai TA, Wilson DM, Ozawa T, Flavell RR. Synthesis and Preliminary Biological Assessment of Carborane-Loaded Theranostic Nanoparticles to Target Prostate-Specific Membrane Antigen. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54739-54752. [PMID: 34752058 DOI: 10.1021/acsami.1c16383] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Boron neutron capture therapy (BNCT) is an encouraging therapeutic modality for cancer treatment. Prostate-specific membrane antigen (PSMA) is a cell membrane protein that is abundantly overexpressed in prostate cancer and can be targeted with radioligand therapies to stimulate clinical responses in patients. In principle, a spatially targeted neutron beam together with specifically targeted PSMA ligands could enable prostate cancer-targeted BNCT. Thus, we developed and tested PSMA-targeted poly(lactide-co-glycolide)-block-poly(ethylene glycol) (PLGA-b-PEG) nanoparticles (NPs) loaded with carborane and tethered to the radiometal chelator deferoxamine B (DFB) for simultaneous positron emission tomography (PET) imaging and selective delivery of boron to prostate cancer. Monomeric PLGA-b-PEGs were covalently functionalized with either DFB or the PSMA ligand ACUPA. Different nanoparticle formulations were generated by nanoemulsification of the corresponding unmodified and DFB- or ACUPA-modified monomers in varying percent fractions. The nanoparticles were efficiently labeled with 89Zr and were subjected to in vitro and in vivo evaluation. The optimized DFB(25)ACUPA(75) NPs exhibited strong in vitro binding to PSMA in direct binding and competition radioligand binding assays in PSMA(+) PC3-Pip cells. [89Zr]DFB(25) NPs and [89Zr]DFB(25)ACUPA(75) NPs were injected to mice with bilateral PSMA(-) PC3-Flu and PSMA(+) PC3-Pip dual xenografts. The NPs demonstrated twofold superior accumulation in PC3-Pip tumors to that of PC3-Flu tumors with a tumor/blood ratio of 25; however, no substantial effect of the ACUPA ligands was detected. Moreover, fast release of carborane from the NPs was observed, resulting in a low boron delivery to tumors in vivo. In summary, these data demonstrate the synthesis, characterization, and initial biological assessment of PSMA-targeted, carborane-loaded PLGA-b-PEG nanoparticles and establish the foundation for future efforts to enable their best use in vivo.
Collapse
Affiliation(s)
- Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
| | - Kyounghee Seo
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
| | - Miko Fogarty
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
| | - Xiao Huang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
| | - Ryan Tang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
| | - Charles Blaha
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158-2517, United States
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143, United States
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California 94143, United States
| | - Young-Wook Jun
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158-2517, United States
- Department of Otolaryngology, University of California, San Francisco, San Francisco, California 94158, United States
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158, United States
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
| | - Tomoko Ozawa
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94143, United States
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California 94143-0981, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158-2517, United States
| |
Collapse
|
7
|
Martinez-Guerrero L, Zhang X, Zorn KM, Ekins S, Wright SH. Cationic Compounds with SARS-CoV-2 Antiviral Activity and Their Interaction with Organic Cation Transporter/Multidrug and Toxin Extruder Secretory Transporters. J Pharmacol Exp Ther 2021; 379:96-107. [PMID: 34253645 PMCID: PMC9006906 DOI: 10.1124/jpet.121.000619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
In the wake of the COVID-19 pandemic, drug repurposing has been highlighted for rapid introduction of therapeutics. Proposed drugs with activity against SARS-CoV-2 include compounds with positive charges at physiologic pH, making them potential targets for the organic cation secretory transporters of kidney and liver, i.e., the basolateral organic cation transporters, OCT1 and OCT2; and the apical multidrug and toxin extruders, MATE1 and MATE2-K. We selected several compounds proposed to have in vitro activity against SARS-CoV-2 (chloroquine, hydroxychloroquine, quinacrine, tilorone, pyronaridine, cetylpyridinium, and miramistin) to test their interaction with OCT and MATE transporters. We used Bayesian machine learning models to generate predictions for each molecule with each transporter and also experimentally determined IC50 values for each compound against labeled substrate transport into CHO cells that stably expressed OCT2, MATE1, or MATE2-K using three structurally distinct substrates (atenolol, metformin and 1-methyl-4-phenylpyridinium) to assess the impact of substrate structure on inhibitory efficacy. For the OCTs substrate identity influenced IC50 values, although the effect was larger and more systematic for OCT2. In contrast, inhibition of MATE1-mediated transport was largely insensitive to substrate identity. Unlike MATE1, inhibition of MATE2-K was influenced, albeit modestly, by substrate identity. Maximum unbound plasma concentration/IC50 ratios were used to identify potential clinical DDI recommendations; all the compounds interacted with the OCT/MATE secretory pathway, most with sufficient avidity to represent potential DDI issues for secretion of cationic drugs. This should be considered when proposing cationic agents as repurposed antivirals. SIGNIFICANCE STATEMENT: Drugs proposed as potential COVID-19 therapeutics based on in vitro activity data against SARS-CoV-2 include compounds with positive charges at physiological pH, making them potential interactors with the OCT/MATE renal secretory pathway. We tested seven such molecules as inhibitors of OCT1/2 and MATE1/2-K. All the compounds blocked transport activity regardless of substrate used to monitor activity. Suggesting that plasma concentrations achieved by normal clinical application of the test agents could be expected to influence the pharmacokinetics of selected cationic drugs.
Collapse
Affiliation(s)
- Lucy Martinez-Guerrero
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Xiaohong Zhang
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Kimberley M Zorn
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Sean Ekins
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| |
Collapse
|
8
|
Gyawali A, Hyeon SJ, Ryu H, Kang YS. The Alteration of L-Carnitine Transport and Pretreatment Effect under Glutamate Cytotoxicity on Motor Neuron-Like NSC-34 Lines. Pharmaceutics 2021; 13:pharmaceutics13040551. [PMID: 33919926 PMCID: PMC8070968 DOI: 10.3390/pharmaceutics13040551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/04/2022] Open
Abstract
L-Carnitine (LC) is essential for transporting fatty acids to the mitochondria for β-oxidation. This study was performed to examine the alteration of the LC transport system in wild type (WT, NSC-34/hSOD1WT) and mutant type (MT, NSC-34/hSOD1G93A) amyotrophic lateral sclerosis (ALS) models. The uptake of [3H]L-carnitine was dependent on time, temperature, concentration, sodium, pH, and energy in both cell lines. The Michaelis–Menten constant (Km) value as well as maximum transport velocity (Vmax) indicated that the MT cell lines showed the higher affinity and lower capacity transport system, compared to that of the WT cell lines. Additionally, LC uptake was inhibited by organic cationic compounds but unaffected by organic anions. OCTN1/slc22a4 and OCTN2/slc22a5 siRNA transfection study revealed both transporters are involved in LC transport in NSC-34 cell lines. Additionally, slc22a4 and slc22a5 was significantly decreased in mouse MT models compared with that in ALS WT littermate models in the immune-reactivity study. [3H]L-Carnitine uptake and mRNA expression pattern showed the pretreatment of LC and acetyl L-carnitine (ALC) attenuated glutamate induced neurotoxicity in NSC-34 cell lines. These findings indicate that LC and ALC supplementation can prevent the neurotoxicity and neuro-inflammation induced by glutamate in motor neurons.
Collapse
Affiliation(s)
- Asmita Gyawali
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Seung Jae Hyeon
- Laboratory for Brain Gene Regulation and Epigenetics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.J.H.); (H.R.)
| | - Hoon Ryu
- Laboratory for Brain Gene Regulation and Epigenetics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (S.J.H.); (H.R.)
| | - Young-Sook Kang
- College of Pharmacy and Drug Information Research Institute, Sookmyung Women’s University, Seoul 04310, Korea;
- Correspondence: ; Tel.: +82-2-710-9562; Fax: +82-2-710-9871
| |
Collapse
|
9
|
Extrahepatic Drug Transporters in Liver Failure: Focus on Kidney and Gastrointestinal Tract. Int J Mol Sci 2020; 21:ijms21165737. [PMID: 32785140 PMCID: PMC7461118 DOI: 10.3390/ijms21165737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging information suggests that liver pathological states may affect the expression and function of membrane transporters in the gastrointestinal tract and the kidney. Altered status of the transporters could affect drug as well as endogenous compounds handling with subsequent clinical consequences. It seems that changes in intestinal and kidney transporter functions provide the compensatory activity of eliminating endogenous compounds (e.g., bile acids) generated and accumulated due to liver dysfunction. A literature search was conducted on the Ovid and PubMed databases to select relevant in vitro, animal and human studies that have reported expression, protein abundance and function of the gastrointestinal and kidney operating ABC (ATP-binding cassette) transporters and SLC (solute carriers) carriers. The accumulated data suggest that liver failure-associated transporter alterations in the gastrointestinal tract and kidney may affect drug pharmacokinetics. The altered status of drug transporters in those organs in liver dysfunction conditions may provide compensatory activity in handling endogenous compounds, affecting local drug actions as well as drug pharmacokinetics.
Collapse
|
10
|
Chen Z, Peng H, Zhang C. Advances in kidney-targeted drug delivery systems. Int J Pharm 2020; 587:119679. [PMID: 32717283 DOI: 10.1016/j.ijpharm.2020.119679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/28/2020] [Accepted: 07/18/2020] [Indexed: 12/19/2022]
Abstract
The management and treatment of kidney diseases currently have caused a huge global burden. Although the application of nanotechnology for the therapy of kidney diseases is still at an early stages, it has profound potential of development. More and more nano-based drug delivery systems provide novel solutions for the treatment of kidney diseases. This article summarizes the physiological and anatomical properties of the kidney and the biological and physicochemical characters of drug delivery systems, which affects the ability of drug to target the kidney, and highlights the prospects, opportunities, and challenges of nanotechnology in the therapy of kidney diseases.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China.
| | - Changmei Zhang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China.
| |
Collapse
|
11
|
Chatuphonprasert W, Nawaratt N, Jarukamjorn K. Reused palm oil from frying pork or potato induced expression of cytochrome P450s and the SLCO1B1 transporter in HepG2 cells. J Food Biochem 2020; 44:e13178. [PMID: 32160325 DOI: 10.1111/jfbc.13178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/29/2020] [Accepted: 02/19/2020] [Indexed: 11/27/2022]
Abstract
Deep frying degrades the oil and generates harmful products. This study evaluated effects of reused palm oil (from frying pork or potato) on expression of cytochrome P450s (CYPs), the transporter (SLCO1B1), and lipid metabolism regulators; proliferator-activated receptors (PPAR) and sterol regulatory element binding protein (SREBP). Human hepatic carcinoma cell line (HepG2) cells were incubated with oleic acid (OA), new palm oil, or reused palm oils for 24 hr. Fatty acid accumulation was examined by Nile red staining. Total RNA was extracted, followed by RT/qPCR of the target genes. Fatty acid accumulation was significantly different between the new and the reused oils. Expression of CYP1A2, CYP2C19, CYP2E1, CYP3A4, CYP4A11, and SLCO1B1 was induced by reused oils. Expression of PPAR-α was strongly increased in all treatments while SREBP-1a and SREBP-1c were suppressed. Modification of CYPs, PPAR-α, and SLCO1B1 by palm oil might increase the risk of fatty acid accumulation with associated oxidative stress. Therefore, consumption of palm oil or reused oil should be limited. PRACTICAL APPLICATIONS: Deep frying degrades the oil and generates harmful products. This study evaluated effects of reused palm oil (from frying pork or potato) on expression of cytochrome P450s (CYPs), the transporter (SLCO1B1), and lipid metabolism regulators; PPAR and SREBP in HepG2 cells. Both of the reused oils-induced profiles of all CYP and SLCO1B1, but the new oil upregulated CYP2E1, CYP3A4, and CYP4A11. PPAR-α was induced while SREBP-1a and SREBP-1c were suppressed by all treatments. Inductions of CYPs with suppression of SREBP-1a and SREBP-1c might contribute to an increased risk of fatty acid accumulation. These findings revealed the impacts of reused palm oil on metabolism via CYPs which related to oxidative stress for further study. Hence, consumption of palm oil or reused cooking oil should be of concern.
Collapse
Affiliation(s)
| | - Nawaratt Nawaratt
- Research Group for Pharmaceutical Activities of Natural Products using Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products using Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
12
|
Anderson JT, Huang KM, Lustberg MB, Sparreboom A, Hu S. Solute Carrier Transportome in Chemotherapy-Induced Adverse Drug Reactions. Rev Physiol Biochem Pharmacol 2020; 183:177-215. [PMID: 32761456 DOI: 10.1007/112_2020_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Members of the solute carrier (SLC) family of transporters are responsible for the cellular influx of a broad range of endogenous compounds and xenobiotics. These proteins are highly expressed in the gastrointestinal tract and eliminating organs such as the liver and kidney, and are considered to be of particular importance in governing drug absorption and elimination. Many of the same transporters are also expressed in a wide variety of organs targeted by clinically important anticancer drugs, directly affect cellular sensitivity to these agents, and indirectly influence treatment-related side effects. Furthermore, targeted intervention strategies involving the use of transport inhibitors have been recently developed, and have provided promising lead candidates for combinatorial therapies associated with decreased toxicity. Gaining a better understanding of the complex interplay between transporter-mediated on-target and off-target drug disposition will help guide the further development of these novel treatment strategies to prevent drug accumulation in toxicity-associated organs, and improve the safety of currently available treatment modalities. In this report, we provide an update on this rapidly emerging field with particular emphasis on anticancer drugs belonging to the classes of taxanes, platinum derivatives, nucleoside analogs, and anthracyclines.
Collapse
Affiliation(s)
- Jason T Anderson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Maryam B Lustberg
- Department of Medical Oncology, The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
13
|
Baidya ATK, Ghosh K, Amin SA, Adhikari N, Nirmal J, Jha T, Gayen S. In silico modelling, identification of crucial molecular fingerprints, and prediction of new possible substrates of human organic cationic transporters 1 and 2. NEW J CHEM 2020. [DOI: 10.1039/c9nj05825g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cation membrane transporters are crucial to regulate movement of foreign molecules within the body. The present study found out structural fingerprints within molecules to be recognized as substrate/non-substrate against these transporters.
Collapse
Affiliation(s)
- Anurag T. K. Baidya
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr H. S. Gour University
- Sagar
- India
| | - Kalyan Ghosh
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr H. S. Gour University
- Sagar
- India
| | - Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
| | - Nilanjan Adhikari
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
| | - Jayabalan Nirmal
- Translational Pharmaceutics Laboratory
- Department of Pharmacy
- BITS-Pilani
- Hyderabad Campus
- Hyderabad 500078
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
| | - Shovanlal Gayen
- Laboratory of Drug Design and Discovery
- Department of Pharmaceutical Sciences
- Dr H. S. Gour University
- Sagar
- India
| |
Collapse
|
14
|
Wright SH. Molecular and cellular physiology of organic cation transporter 2. Am J Physiol Renal Physiol 2019; 317:F1669-F1679. [PMID: 31682169 DOI: 10.1152/ajprenal.00422.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Organic cation transporters play a critical role in mediating the distribution of cationic pharmaceuticals. Indeed, organic cation transporter (OCT)2 is the initial step in the renal secretion of organic cations and consequently plays a defining role in establishing the pharmacokinetics of many cationic drugs. Although a hallmark of OCTs is their broad selectivity, this characteristic also makes them targets for unwanted, adverse drug-drug interactions (DDIs), making them a focus for efforts to develop models of ligand interaction that could predict and preempt these adverse interactions. This review discusses the molecular characteristics of these transporters as well as the evidence that established the OCTs as key players in the distribution of organic cations. However, the primary focus is the present understanding of the complexity of ligand interaction with OCTs, particularly OCT2, including evidence for the presence of multiple ligand-binding sites and the influence of substrate structure on the affinity of the transporter for inhibitory ligands. This leads to a discussion of the complexities associated with the development of protocols for assessing the inhibitory potential of new molecular entities to perpetrate unwanted DDIs, the criteria that should be considered in the interpretation of the results of such protocols, and the challenges associated with development of models capable of predicting unwanted DDIs.
Collapse
Affiliation(s)
- Stephen H Wright
- Department of Physiology, University of Arizona, Tucson, Arizona
| |
Collapse
|
15
|
Sandoval PJ, Morales M, Secomb TW, Wright SH. Kinetic basis of metformin-MPP interactions with organic cation transporter OCT2. Am J Physiol Renal Physiol 2019; 317:F720-F734. [PMID: 31313952 DOI: 10.1152/ajprenal.00152.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Organic cation transporter 2 (OCT2) clears the blood of cationic drugs. Efforts to understand OCT2 selectivity as a means to predict the potential of new molecular entities (NMEs) to produce unwanted drug-drug interactions typically assess the influence of the NMEs on inhibition of transport. However, the identity of the substrate used to assess transport activity can influence the quantitative profile of inhibition. Metformin and 1-methyl-4-phenylpyridinium (MPP), in particular, display markedly different inhibitory profiles, with IC50 values for inhibition of MPP transport often being more than fivefold greater than IC50 values for the inhibition of metformin transport by the same compound, suggesting that interaction of metformin and MPP with OCT2 cannot be restricted to competition for a single binding site. Here, we determined the kinetic basis for the mutual inhibitory interaction of metformin and MPP with OCT2 expressed in Chinese hamster ovary cells. Although metformin did produce simple competitive inhibition of MPP transport, MPP was a mixed-type inhibitor of metformin transport, decreasing the maximum rate of mediated substrate transport and increasing the apparent Michaelis constant (Ktapp) for OCT2-mediated metformin transport. Furthermore, whereas the IC50 value for metformin's inhibition of MPP transport did not differ from the Ktapp value for metformin transport, the IC50 value for MPP's inhibition of metformin transport was less than its Ktapp value for transport. The simplest model to account for these observations required the influence of a distinct inhibitory site for MPP that, when occupied, decreases the translocation of substrate. These observations underscore the complexity of ligand interaction with OCT2 and argue for use of multiple substrates to obtain the needed kinetic assessment of NME interactions with OCT2.
Collapse
Affiliation(s)
| | - Mark Morales
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- Department of Physiology, University of Arizona, Tucson, Arizona
| |
Collapse
|
16
|
Zhu S, Tian R, Antaris AL, Chen X, Dai H. Near-Infrared-II Molecular Dyes for Cancer Imaging and Surgery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900321. [PMID: 31025403 PMCID: PMC6555689 DOI: 10.1002/adma.201900321] [Citation(s) in RCA: 507] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/03/2019] [Indexed: 05/05/2023]
Abstract
Fluorescence bioimaging affords a vital tool for both researchers and surgeons to molecularly target a variety of biological tissues and processes. This review focuses on summarizing organic dyes emitting at a biological transparency window termed the near-infrared-II (NIR-II) window, where minimal light interaction with the surrounding tissues allows photons to travel nearly unperturbed throughout the body. NIR-II fluorescence imaging overcomes the penetration/contrast bottleneck of imaging in the visible region, making it a remarkable modality for early diagnosis of cancer and highly sensitive tumor surgery. Due to their convenient bioconjugation with peptides/antibodies, NIR-II molecular dyes are desirable candidates for targeted cancer imaging, significantly overcoming the autofluorescence/scattering issues for deep tissue molecular imaging. To promote the clinical translation of NIR-II bioimaging, advancements in the high-performance small molecule-derived probes are critically important. Here, molecules with clinical potential for NIR-II imaging are discussed, summarizing the synthesis and chemical structures of NIR-II dyes, chemical and optical properties of NIR-II dyes, bioconjugation and biological behavior of NIR-II dyes, whole body imaging with NIR-II dyes for cancer detection and surgery, as well as NIR-II fluorescence microscopy imaging. A key perspective on the direction of NIR-II molecular dyes for cancer imaging and surgery is also discussed.
Collapse
Affiliation(s)
- Shoujun Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | | | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Hongjie Dai
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
17
|
Modeling Gadoxetate Liver Uptake and Efflux Using Dynamic Contrast-Enhanced Magnetic Resonance Imaging Enables Preclinical Quantification of Transporter Drug-Drug Interactions. Invest Radiol 2019; 53:563-570. [PMID: 29771727 DOI: 10.1097/rli.0000000000000480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The aim of this study was to model the in vivo transporter-mediated uptake and efflux of the hepatobiliary contrast agent gadoxetate in the liver. The efficacy of the proposed technique was assessed for its ability to provide quantitative insights into drug-drug interactions (DDIs), using rifampicin as inhibitor. MATERIALS AND METHODS Three groups of C57 mice were scanned twice with a dynamic gadoxetate-enhanced magnetic resonance imaging protocol, using a 3-dimensional spoiled gradient-echo sequence for approximately 72 minutes. Before the second magnetic resonance imaging session, 2 of the groups received a rifampicin dose of 20 (n = 7) or 40 (n = 7) mg/kg, respectively. Data from regions of interest in the liver were analyzed using 2 simplifications of a 2-compartment uptake and efflux model to provide estimates for the gadoxetate uptake rate (ki) into the hepatocytes and its efflux rate (kef) into the bile. Both models were assessed for goodness-of-fit in the group without rifampicin (n = 9), and the appropriate model was selected for assessing the ability to monitor DDIs in vivo. RESULTS Seven of 9 mice from the group without rifampicin were assessed for model implementation and reproducibility. A simple 3 parameter model (ki, kef, and extracellular space, vecs) adequately described the observed liver concentration time series with mean ki = 0.47 ± 0.11 min and mean kef = 0.039 ± 0.016 min. Visually, the area under the liver concentration time profile was reduced for the groups receiving rifampicin. Furthermore, tracer kinetic modeling demonstrated a significant dose-dependent decrease in the uptake (5.9- and 17.3-fold decrease for 20 mg/kg and 40 mg/kg, respectively) and efflux rates (2.2- and 7.9-fold decrease) compared with the first scan for each group. CONCLUSIONS This study presents the first in vivo implementation of a 2-compartment uptake and efflux model to monitor DDIs at the transporter-protein level, using the clinically relevant organic anion transporting polypeptide inhibitor rifampicin. The technique has the potential to be a novel alternative to other methods, allowing real-time changes in transporter DDIs to be measured directly in vivo.
Collapse
|
18
|
Dai Q, Geng H, Yu Q, Hao J, Cui J. Polyphenol-Based Particles for Theranostics. Theranostics 2019; 9:3170-3190. [PMID: 31244948 PMCID: PMC6567970 DOI: 10.7150/thno.31847] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/06/2019] [Indexed: 02/06/2023] Open
Abstract
Polyphenols, due to their high biocompatibility and wide occurrence in nature, have attracted increasing attention in the engineering of functional materials ranging from films, particles, to bulk hydrogels. Colloidal particles, such as nanogels, hollow capsules, mesoporous particles and core-shell structures, have been fabricated from polyphenols or their derivatives with a series of polymeric or biomolecular compounds through various covalent and non-covalent interactions. These particles can be designed with specific properties or functionalities, including multi-responsiveness, radical scavenging capabilities, and targeting abilities. Moreover, a range of cargos (e.g., imaging agents, anticancer drugs, therapeutic peptides or proteins, and nucleic acid fragments) can be incorporated into these particles. These cargo-loaded carriers have shown their advantages in the diagnosis and treatment of diseases, especially of cancer. In this review, we summarize the assembly of polyphenol-based particles, including polydopamine (PDA) particles, metal-phenolic network (MPN)-based particles, and polymer-phenol particles, and their potential biomedical applications in various diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Qiong Dai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Huimin Geng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Qun Yu
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
19
|
Organic anion transporting polypeptide 2B1 – More than a glass-full of drug interactions. Pharmacol Ther 2019; 196:204-215. [DOI: 10.1016/j.pharmthera.2018.12.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
A high-risk papillomavirus 18 E7 affibody-enabled in vivo imaging and targeted therapy of cervical cancer. Appl Microbiol Biotechnol 2019; 103:3049-3059. [PMID: 30770966 DOI: 10.1007/s00253-019-09655-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/14/2019] [Accepted: 01/16/2019] [Indexed: 10/27/2022]
Abstract
High-risk papillomavirus (HPV) is one of the major reasons for cervical cancer, causing most lethal gynecologic malignancies worldwide. For cervical cancer progression, oncogene E7 plays vital roles and is used as one of the major targets for cervical tumor diagnosis and treatment. In the clinic, successful treatment of cervical cancer relies on diagnosing the disease at an early stage, where a late-stage diagnosis usually led to treatment failure. In this work, we designed and purified an HPV18 E7 oncogene targeting affibody, named as ZHPV18E7, for in vitro and in vivo imaging and targeted treatment of cervical cancer. In vitro, ZHPV18E7 showed a specific targeting effect against an HPV18 positive cell line; as a contrast, the affibody did not target the HPV18 negative cell line. In vivo, we tested the bio-distribution of the affibody in mice bearing cervical cancer. The whole animal imaging analysis indicated the affibody-targeted tumor tissue specifically with 10 min after injection, and the affibody reached the highest level at tumor tissues 45 min after injection. At the 24th hour after injection, the affibody still maintained a certain level in tumor tissues compared to other organs. To test the therapeutic effect of this affibody, we modified the affibody (i.e., ZHPV18E7) with a clinically used anti-cancer agent (i.e., Pseudomonas exotoxin). In a mice cervical cancer model, ZHPV18E7 was able to deliver Pseudomonas exotoxin to tumor tissues effectively, showing great potential for cancer treatment. This study indicated that ZHPV18E7 could be employed for in vitro imaging and targeted treatment of cervical cancer. Beyond the chemotherapeutic agent used in this work, the affibody could be extended for carrying other therapeutic agents for cervical cancer treatment.
Collapse
|
21
|
Tian R, Ma H, Yang Q, Wan H, Zhu S, Chandra S, Sun H, Kiesewetter DO, Niu G, Liang Y, Chen X. Rational design of a super-contrast NIR-II fluorophore affords high-performance NIR-II molecular imaging guided microsurgery. Chem Sci 2019; 10:326-332. [PMID: 30713641 PMCID: PMC6333232 DOI: 10.1039/c8sc03751e] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/08/2018] [Indexed: 12/23/2022] Open
Abstract
In vivo molecular imaging in the "transparent" near-infrared II (NIR-II) window has demonstrated impressive benefits in reaching millimeter penetration depths with high specificity and imaging quality. Previous NIR-II molecular imaging generally relied on high hepatic uptake fluorophores with an unclear mechanism and antibody-derived conjugates, suffering from inevitable nonspecific retention in the main organs/skin with a relatively low signal-to-background ratio. It is still challenging to synthesize a NIR-II fluorophore with both high quantum yield and minimal liver-retention feature. Herein, we identified the structural design and excretion mechanism of novel NIR-II fluorophores for NIR-II molecular imaging with an extremely clean background. With the optimized renally excreted fluorophore-peptide conjugates, superior NIR-II targeting imaging was accompanied by the improved signal-to-background ratio during tumor detection with reducing off-target tissue exposure. An unprecedented NIR-II imaging-guided microsurgery was achieved using such an imaging platform, which provides us with a great preclinical example to accelerate the potential clinical translation of NIR-II imaging.
Collapse
Affiliation(s)
- Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| | - Huilong Ma
- Department of Materials Science & Engineering , Shenzhen Key Laboratory of Printed Organic Electronics , South University of Science & Technology of China , Shenzhen 518055 , China .
| | - Qinglai Yang
- Department of Materials Science & Engineering , Shenzhen Key Laboratory of Printed Organic Electronics , South University of Science & Technology of China , Shenzhen 518055 , China .
- Research Center for Advanced Materials and Biotechnology , Research Institute of Tsinghua University in Shenzhen , Shenzhen 518057 , China
| | - Hao Wan
- Department of Chemistry , Stanford University , Stanford , CA 94305 , USA
| | - Shoujun Zhu
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| | - Swati Chandra
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| | - Haitao Sun
- State Key Laboratory of Precision Spectroscopy , School of Physics and Materials Science , East China Normal University , Shanghai 200062 , China
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| | - Yongye Liang
- Department of Materials Science & Engineering , Shenzhen Key Laboratory of Printed Organic Electronics , South University of Science & Technology of China , Shenzhen 518055 , China .
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine , National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , USA . ;
| |
Collapse
|
22
|
Comprehensive Substrate Characterization of 22 Antituberculosis Drugs for Multiple Solute Carrier (SLC) Uptake Transporters In Vitro. Antimicrob Agents Chemother 2018; 62:AAC.00512-18. [PMID: 30012768 DOI: 10.1128/aac.00512-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/08/2018] [Indexed: 12/30/2022] Open
Abstract
The substrate potentials of antituberculosis drugs on solute carrier (SLC) transporters are not well characterized to date, despite a well-established understanding of their drug dispositions and pharmacokinetics. In this study, we investigated comprehensively the substrate potentials of the 22 currently available antituberculosis drugs for SLC family transporter-mediated uptake, using Xenopus laevis oocytes and stably transfected HEK-293 cells in vitro The result suggested that ethambutol, isoniazid, amoxicillin, and prothionamide act as novel substrates for the SLC transporters. In addition, in the presence of representative transporter inhibitors, the uptake of the antituberculosis drugs was markedly decreased compared with the uptake in the absence of inhibitor, suggesting involvement of the corresponding transporters. A cellular uptake study was performed, and the Km values of ethambutol were found to be 526.1 ± 15.6, 212.0 ± 20.1, 336.8 ± 20.1, and 455.0 ± 28 μM for organic cation transporter 1 (OCT1), OCT2, OCTN1, and OCTN2, respectively. Similarly, the Km of prothionamide was 805.8 ± 23.4 μM for OCT1, while the Km values of isoniazid and amoxicillin for organic anion transporter 3 (OAT3) were 233.7 ± 14.1 and 161.4 ± 10.6 μM, respectively. The estimated in vivo drug-drug interaction indexes from in vitro transporter inhibition kinetics for verapamil, probenecid, and ibuprofen against ethambutol, prothionamide, isoniazid, and amoxicillin were found to show potential for clinical drug interactions. In conclusion, this is the first study that demonstrated 22 antituberculosis drug interactions with transporters. This study will be helpful for mechanistic understanding of the disposition, drug-drug interactions, and pharmacokinetics of these antituberculosis drugs.
Collapse
|
23
|
Zhu S, Yung BC, Chandra S, Niu G, Antaris AL, Chen X. Near-Infrared-II (NIR-II) Bioimaging via Off-Peak NIR-I Fluorescence Emission. Theranostics 2018; 8:4141-4151. [PMID: 30128042 PMCID: PMC6096392 DOI: 10.7150/thno.27995] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
Significantly reduced photon scattering and minimal tissue autofluorescence levels in the second biological transparency window (NIR-II; 1000-1700 nm) facilitate higher resolution in vivo biological imaging compared to tradition NIR fluorophores (~700-900 nm). However, the existing palette of NIR-II fluorescent agents including semiconducting inorganic nanomaterials and recently introduced small-molecule organic dyes face significant technical and regulatory hurdles prior to clinical translation. Fortunately, recent spectroscopic characterization of NIR-I dyes (e.g., indocyanine green (ICG), IRDye800CW and IR-12N3) revealed long non-negligible emission tails reaching past 1500 nm. Repurposing the most widely used NIR dye in medicine, in addition to those in the midst of clinical trials creates an accelerated pathway for NIR-II clinical translation. This review focuses on the significant advantage of imaging past 1000 nm with NIR-I fluorophores from both a basic and clinical viewpoint. We further discuss optimizing NIR-I dyes around their NIR-II/shortwave infrared (SWIR) emission, NIR-II emission tail characteristics and prospects of NIR-II imaging with clinically available and commercially available dyes.
Collapse
Affiliation(s)
- Shoujun Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Bryant C. Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Swati Chandra
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| | | | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), 35A Convent Dr, Bethesda, Maryland 20892, United States
| |
Collapse
|
24
|
Zhu S, Hu Z, Tian R, Yung BC, Yang Q, Zhao S, Kiesewetter DO, Niu G, Sun H, Antaris AL, Chen X. Repurposing Cyanine NIR-I Dyes Accelerates Clinical Translation of Near-Infrared-II (NIR-II) Bioimaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802546. [PMID: 29985542 DOI: 10.1002/adma.201802546] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/30/2018] [Indexed: 05/24/2023]
Abstract
The significantly reduced tissue autofluorescence and scattering in the NIR-II region (1000-1700 nm) opens many exciting avenues for detailed investigation of biological processes in vivo. However, the existing NIR-II fluorescent agents, including many molecular dyes and inorganic nanomaterials, are primarily focused on complicated synthesis routes and unknown immunogenic responses with limited potential for clinical translation. Herein, the >1000 nm tail emission of conventional biocompatible NIR cyanine dyes with emission peaks at 700-900 nm is systematically investigated, and a type of bright dye for NIR-II imaging with high potential for accelerating clinical translation is identified. The asymmetry of the π domain in the S1 state of NIR cyanine dyes is proven to result in a twisted intramolecular charge-transfer process and NIR-II emission, establishing a general rule to guide future NIR-I/II fluorophore synthesis. The screened NIR dyes are identified to possess a bright emission tail in the NIR-II region along with high quantum yield, high molar-extinction coefficient, rapid fecal excretion, and functional groups amenable for bioconjugation. As a result, NIR cyanine dyes can be used for NIR-II imaging to afford superior contrast and real-time imaging of several biological models, facilitating the translation of NIR-II bioimaging to clinical theranostic applications.
Collapse
Affiliation(s)
- Shoujun Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Zhubin Hu
- State Key Laboratory of Precision Spectroscopy, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | - Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Bryant C Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | | | - Su Zhao
- Nirmidas Biotech, Palo Alto, CA, 94303, USA
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Haitao Sun
- State Key Laboratory of Precision Spectroscopy, School of Physics and Materials Science, East China Normal University, Shanghai, 200062, China
| | | | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| |
Collapse
|
25
|
Sandoval PJ, Zorn KM, Clark AM, Ekins S, Wright SH. Assessment of Substrate-Dependent Ligand Interactions at the Organic Cation Transporter OCT2 Using Six Model Substrates. Mol Pharmacol 2018; 94:1057-1068. [PMID: 29884691 DOI: 10.1124/mol.117.111443] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/29/2018] [Indexed: 01/08/2023] Open
Abstract
Organic cation transporter (OCT) 2 mediates the entry step for organic cation secretion by renal proximal tubule cells and is a site of unwanted drug-drug interactions (DDIs). But reliance on decision tree-based predictions of DDIs at OCT2 that depend on IC50 values can be suspect because they can be influenced by choice of transported substrate; for example, IC50 values for the inhibition of metformin versus MPP transport can vary by 5- to 10-fold. However, it is not clear whether the substrate dependence of a ligand interaction is common among OCT2 substrates. To address this question, we screened the inhibitory effectiveness of 20 µM concentrations of several hundred compounds against OCT2-mediated uptake of six structurally distinct substrates: MPP, metformin, N,N,N-trimethyl-2-[methyl(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino]ethanaminium (NBD-MTMA), TEA, cimetidine, and 4-4-dimethylaminostyryl-N-methylpyridinium (ASP). Of these, MPP transport was least sensitive to inhibition. IC50 values for 20 structurally diverse compounds confirmed this profile, with IC50 values for MPP averaging 6-fold larger than those for the other substrates. Bayesian machine-learning models of ligand-induced inhibition displayed generally good statistics after cross-validation and external testing. Applying our ASP model to a previously published large-scale screening study for inhibition of OCT2-mediated ASP transport resulted in comparable statistics, with approximately 75% of "active" inhibitors predicted correctly. The differential sensitivity of MPP transport to inhibition suggests that multiple ligands can interact simultaneously with OCT2 and supports the recommendation that MPP not be used as a test substrate for OCT2 screening. Instead, metformin appears to be a comparatively representative OCT2 substrate for both in vitro and in vivo (clinical) use.
Collapse
Affiliation(s)
- Philip J Sandoval
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (P.J.S., S.H.W.); Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.); and Molecular Materials Informatics, Inc., Montreal, Quebec, Canada (A.M.C.)
| | - Kimberley M Zorn
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (P.J.S., S.H.W.); Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.); and Molecular Materials Informatics, Inc., Montreal, Quebec, Canada (A.M.C.)
| | - Alex M Clark
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (P.J.S., S.H.W.); Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.); and Molecular Materials Informatics, Inc., Montreal, Quebec, Canada (A.M.C.)
| | - Sean Ekins
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (P.J.S., S.H.W.); Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.); and Molecular Materials Informatics, Inc., Montreal, Quebec, Canada (A.M.C.)
| | - Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (P.J.S., S.H.W.); Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.); and Molecular Materials Informatics, Inc., Montreal, Quebec, Canada (A.M.C.)
| |
Collapse
|
26
|
Hu S, Leblanc AF, Gibson AA, Hong KW, Kim JY, Janke LJ, Li L, Vasilyeva A, Finkelstein DB, Sprowl JA, Sweet DH, Schlatter E, Ciarimboli G, Schellens J, Baker SD, Pabla N, Sparreboom A. Identification of OAT1/OAT3 as Contributors to Cisplatin Toxicity. Clin Transl Sci 2017; 10:412-420. [PMID: 28689374 PMCID: PMC5593168 DOI: 10.1111/cts.12480] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/12/2017] [Indexed: 12/17/2022] Open
Abstract
Cisplatin is among the most widely used anticancer drugs and known to cause a dose‐limiting nephrotoxicity, which is partially dependent on the renal uptake carrier OCT2. We here report a previously unrecognized, OCT2‐independent pathway of cisplatin‐induced renal injury that is mediated by the organic anion transporters OAT1 and OAT3. Using transporter‐deficient mouse models, we found that this mechanism regulates renal uptake of a mercapturic acid metabolite of cisplatin that acts as a precursor of a potent nephrotoxin. The function of these two transport systems can be simultaneously inhibited by the tyrosine kinase inhibitor nilotinib through noncompetitive mechanisms, without compromising the anticancer properties of cisplatin. Collectively, our findings reveal a novel pathway that explains the fundamental basis of cisplatin‐induced nephrotoxicity, with potential implications for its therapeutic management.
Collapse
Affiliation(s)
- S Hu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - A F Leblanc
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - A A Gibson
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - K W Hong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - J Y Kim
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - L J Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - L Li
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - A Vasilyeva
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - D B Finkelstein
- Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - J A Sprowl
- Department of Pharmaceutical, Social and Administrative Sciences, School of Pharmacy, D'Youville College, Buffalo, New York, USA
| | - D H Sweet
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - E Schlatter
- Medical Clinic D, Experimental Nephrology, Münster Medical Faculty, Münster, Germany
| | - G Ciarimboli
- Medical Clinic D, Experimental Nephrology, Münster Medical Faculty, Münster, Germany
| | - Jhm Schellens
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S D Baker
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - N Pabla
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| | - A Sparreboom
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
27
|
Severance AC, Sandoval PJ, Wright SH. Correlation between Apparent Substrate Affinity and OCT2 Transport Turnover. J Pharmacol Exp Ther 2017; 362:405-412. [PMID: 28615288 DOI: 10.1124/jpet.117.242552] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 12/30/2022] Open
Abstract
Organic cation (OC) transporter 2 (OCT2) mediates the first step in the renal secretion of many cationic drugs: basolateral uptake from blood into proximal tubule cells. The impact of this process on the pharmacokinetics of drug clearance as estimated using a physiologically-based pharmacokinetic approach relies on an accurate understanding of the kinetics of transport because the ratio of the maximal rate of transport to the Michaelis constant (i.e., Jmax/ Kt) provides an estimate of the intrinsic clearance (Clint) used in in vitro-in vivo extrapolation of experimentally determined transport data. Although the multispecificity of renal OC secretion, including that of the OCT2 transporter, is widely acknowledged, the possible relationship between relative affinity of the transporter for its diverse substrates and the maximal rates of their transport has received little attention. In this study, we determined the Jmax and apparent Michaelis constant (Ktapp) values for six structurally distinct OCT2 substrates and found a strong correlation between Jmax and Ktapp; high-affinity substrates [Ktapp values <50 µM, including 1-methyl-4-phenylpyridinium, or 1-methyl-4-phenylpyridinium (MPP), and cimetidine] displayed systematically lower Jmax values (<50 pmol cm-2 min-1) than did low-affinity substrates (Ktapp >200 µM, including choline and metformin). Similarly, preloading OCT2-expressing cells with low-affinity substrates resulted in systematically larger trans-stimulated rates of MPP uptake than did preloading with high-affinity substrates. The data are quantitatively consistent with the hypothesis that dissociation of bound substrate from the transporter is rate limiting in establishing maximal rates of OCT2-mediated transport. This systematic relationship may provide a means to estimate Clint for drugs for which transport data are lacking.
Collapse
Affiliation(s)
| | - Philip J Sandoval
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
28
|
Evaluation of para-Aminosalicylic Acid as a Substrate of Multiple Solute Carrier Uptake Transporters and Possible Drug Interactions with Nonsteroidal Anti-inflammatory Drugs In Vitro. Antimicrob Agents Chemother 2017; 61:AAC.02392-16. [PMID: 28223391 DOI: 10.1128/aac.02392-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/15/2017] [Indexed: 11/20/2022] Open
Abstract
para-Aminosalicylic acid (PAS) is a second-line antituberculosis drug that has been used to treat multidrug-resistant and extensively drug-resistant tuberculosis for more than 60 years. Renal secretion and glomerular filtration are the major pathways for the elimination of PAS. We comprehensively studied PAS transport by using cell lines that overexpressed various transporters and found that PAS acts as a novel substrate of an organic anionic polypeptide (OATP1B1), organic cationic transporters (OCT1 and OCT2), and organic anion transporters (OAT1 and OAT3) but is not a substrate of any ATP-binding cassette (ABC) transporters. Net PAS uptake was measured, and the transport affinities (Km values) for OATP1B1, OCT1, OCT2, OAT1, and OAT3 were found to be 50.0, 20.3, 28.7, 78.1, and 100.1 μM, respectively. The net uptake rates suggested that renal OAT1 and OAT3 play relatively major roles in PAS elimination. The representative inhibitors rifampin for OATP1B1, probenecid for OAT1 and OAT3, and verapamil for OCT1 and OCT2 greatly inhibited PAS uptake, suggesting that PAS is dependent on multiple transporters for uptake. We also evaluated nonsteroidal anti-inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs), and metformin for the inhibition of PAS uptake via these transporters. Half-maximal (50%) inhibitory concentrations (IC50s) were kinetically determined and used to predict the drug-drug interactions (DDIs) affecting these transporters' activity toward PAS. We found that rifampin, probenecid, ibuprofen, naproxen, cimetidine, and quinidine each exhibited a significant potential for in vivo DDIs with PAS. In this study, PAS was found to be a novel substrate of several transporters, and drugs that inhibit these transporters can reduce PAS elimination.
Collapse
|
29
|
Cellular Models and In Vitro Assays for the Screening of modulators of P-gp, MRP1 and BCRP. Molecules 2017; 22:molecules22040600. [PMID: 28397762 PMCID: PMC6153761 DOI: 10.3390/molecules22040600] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/24/2017] [Accepted: 03/28/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are highly expressed in tumor cells, as well as in organs involved in absorption and secretion processes, mediating the ATP-dependent efflux of compounds, both endogenous substances and xenobiotics, including drugs. Their expression and activity levels are modulated by the presence of inhibitors, inducers and/or activators. In vitro, ex vivo and in vivo studies with both known and newly synthesized P-glycoprotein (P-gp) inducers and/or activators have shown the usefulness of these transport mechanisms in reducing the systemic exposure and specific tissue access of potentially harmful compounds. This article focuses on the main ABC transporters involved in multidrug resistance [P-gp, multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP)] expressed in tissues of toxicological relevance, such as the blood-brain barrier, cardiovascular system, liver, kidney and intestine. Moreover, it provides a review of the available cellular models, in vitro and ex vivo assays for the screening and selection of safe and specific inducers and activators of these membrane transporters. The available cellular models and in vitro assays have been proposed as high throughput and low-cost alternatives to excessive animal testing, allowing the evaluation of a large number of compounds.
Collapse
|
30
|
Fattah S, Shinde AB, Matic M, Baes M, van Schaik RHN, Allegaert K, Parmentier C, Richert L, Augustijns P, Annaert P. Inter-Subject Variability in OCT1 Activity in 27 Batches of Cryopreserved Human Hepatocytes and Association with OCT1 mRNA Expression and Genotype. Pharm Res 2017; 34:1309-1319. [PMID: 28364304 DOI: 10.1007/s11095-017-2148-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/17/2017] [Indexed: 01/11/2023]
Abstract
PURPOSE OCT1/3 (Organic Cation Transporter-1 and -3; SLC22A1/3) are transmembrane proteins localized at the basolateral membrane of hepatocytes. They mediate the uptake of cationic endogenous compounds and/or xenobiotics. The present study was set up to verify whether the previously observed variability in OCT activity in hepatocytes may be explained by inter-individual differences in OCT1/3 mRNA levels or OCT1 genotype. METHODS Twenty-seven batches of cryopreserved human hepatocytes (male and female, age 24-88 y) were characterized for OCT activity, normalized OCT1/3 mRNA expression, and OCT1 genetic mutation. ASP+ (4-[4-(dimethylamino)styryl]-N-methylpyridinium iodide) was used as probe substrate. RESULTS ASP+ uptake ranged between 75 ± 61 and 2531 ± 202 pmol/(min × million cells). The relative OCT1 and OCT3 mRNA expression ranged between 0.007-0.46 and 0.0002-0.005, respectively. The presence of one or two nonfunctional SLC22A1 alleles was observed in 13 batches and these exhibited significant (p = 0.04) association with OCT1 and OCT3 mRNA expression. However, direct association between genotype and OCT activity could not be established. CONCLUSION mRNA levels and genotype of OCT only partially explain inter-individual variability in OCT-mediated transport. Our findings illustrate the necessity of in vitro transporter activity profiling for better understanding of inter-individual drug disposition behavior.
Collapse
Affiliation(s)
- Sarinj Fattah
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N II Herestraat 49 Box 921, 3000, Leuven, Belgium
| | - Abhijit Babaji Shinde
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Maja Matic
- Department Clinical Chemistry, Erasmus University Medical Centre, Rotterdam, Netherlands.,Intensive Care and Department of Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Ron H N van Schaik
- Department Clinical Chemistry, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Karel Allegaert
- Intensive Care and Department of Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Lysiane Richert
- KaLy-Cell, Plobsheim, France.,Université de Franche-Comté, 4267, Besançon, EA, France
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N II Herestraat 49 Box 921, 3000, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Campus Gasthuisberg O&N II Herestraat 49 Box 921, 3000, Leuven, Belgium.
| |
Collapse
|
31
|
Parvez MM, Kaisar N, Shin HJ, Jung JA, Shin JG. Inhibitory Interaction Potential of 22 Antituberculosis Drugs on Organic Anion and Cation Transporters of the SLC22A Family. Antimicrob Agents Chemother 2016; 60:6558-6567. [PMID: 27550354 PMCID: PMC5075059 DOI: 10.1128/aac.01151-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/12/2016] [Indexed: 11/20/2022] Open
Abstract
Twenty-two currently marketed antituberculosis drugs were comprehensively evaluated for their inhibitory effect on organic anionic transporter (OAT)- and organic cation transporter (OCT)-mediated uptake using stably transfected HEK293 cells in vitro We observed moderate to strong inhibitory effects on OAT1- and OAT3-mediated para-aminohippurate (PAH) uptake and OCT1- and OCT2-mediated N-methyl-4-phenylpylidinium acetate (MPP+) uptake. Ciprofloxacin, linezolid, para-aminosalicylic acid (PAS), and rifampin were observed to have strong inhibitory effects, with the concentrations for a 50% inhibitory effect (IC50s) being 35.1, 31.1, 37.6, and 48.1 μM, respectively, for OAT1 and >100, 21.9, 24.6, and 30.2 μM, respectively, for OAT3. Similarly, pyrazinamide, rifabutin, and levofloxacin were observed to have inhibitory effects, with IC50 values being 36.5, 42.7, and 30.3 μM, respectively, for OCT1 and with the IC50 value for PAS being 94.2 μM for OCT2. In addition, we used zidovudine and metformin as clinically prescribed substrates of OATs and OCTs, respectively, and zidovudine and metformin uptake was also strongly inhibited by the antituberculosis drugs. Among the tested drugs, the highest drug-drug interaction (DDI) indexes were found for PAS, which were 9.3 to 13.9 for OAT1 and 12.0 to 17.7 for OAT3, and linezolid, which were 1.18 to 2.15 for OAT1 and 1.7 to 3.01 for OAT3. Similarly, the DDI indexes of pyrazinamide and levofloxacin were 0.57 and 0.30, respectively, for OCT1, and the DDI index of PAS was 3.8 for OCT2, suggesting a stronger possibility (DDI index value cutoff, >0.1) of in vivo DDIs. This is the first comprehensive report of the inhibitory potential of anti-TB drugs on OAT- and OCT-mediated uptake of prototype and clinically prescribed substrate drugs in vitro, providing an ability to predict DDIs between anti-TB drugs and other coprescribed drugs in clinical studies in vivo.
Collapse
Affiliation(s)
- M Masud Parvez
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Nazia Kaisar
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Ho Jung Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
| | - Jin Ah Jung
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, Republic of Korea
- Department of Clinical Pharmacology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| |
Collapse
|
32
|
Mandal A, Agrahari V, Khurana V, Pal D, Mitra AK. Transporter effects on cell permeability in drug delivery. Expert Opin Drug Deliv 2016; 14:385-401. [PMID: 27449574 DOI: 10.1080/17425247.2016.1214565] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The role of drug transporters as one of the determinants of cellular drug permeability has become increasingly evident. Despite the lipophilicity of a drug molecule as rate-limiting factor for passive diffusion across biological membranes, carrier-mediated and active transport have gained attention over the years. A better understanding of the effects and roles of these influx transporters towards transmembrane permeability of a drug molecule need to be delineated for drug development and delivery. Areas covered: This review focuses on findings relative to role of transporters in drug absorption and bioavailability. Particularly the areas demanding further research have been emphasized. This review will also highlight various transporters expressed on vital organs and their effects on drug pharmacokinetics. Expert opinion: Significant efforts have been devoted to understand the role of transporters, their iterative interplay with metabolizing enzymes through molecular enzymology, binding and structure-activity relationship studies. A few assays such as parallel artificial membrane permeation assay (PAMPA) have been developed to analyze drug transport across phospholipid membranes. Although large web-accessible databases on tissue selective expression profiles at transcriptomic as well as proteomic are available, there is a need to collocate the scattered literature on the role of transporters in drug development and delivery.
Collapse
Affiliation(s)
- Abhirup Mandal
- a Division of Pharmaceutical Sciences, School of Pharmacy , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Vibhuti Agrahari
- a Division of Pharmaceutical Sciences, School of Pharmacy , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Varun Khurana
- b R&D , INSYS Therapeutics Inc , Chandler , AZ , USA
| | - Dhananjay Pal
- a Division of Pharmaceutical Sciences, School of Pharmacy , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Ashim K Mitra
- c UMKC School of Pharmacy, Division of Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| |
Collapse
|
33
|
Ezzeddine B, Raoudha JM, Mohamed D, Abdelfattah EF. Therapeutic Efficacy ofHammada scopariaExtract Against Ethanol Induced Renal Dysfunction in Wistar Rats. J Food Biochem 2016. [DOI: 10.1111/jfbc.12307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Bourogaa Ezzeddine
- Laboratoire d'Ecophysiologie Animale; Laboratoire d'Ecophysiologie Animale, Faculté des Sciences de Sfax, PO Box 1171, Sfax Tunisie PO Box 1171 Sfax Tunisie
- Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax; PO Box 1171 Sfax Tunisie
| | - Jarraya-Mezghani Raoudha
- Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax; PO Box 1171 Sfax Tunisie
| | - Damak Mohamed
- Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax; PO Box 1171 Sfax Tunisie
| | - El Feki Abdelfattah
- Laboratoire d'Ecophysiologie Animale; Laboratoire d'Ecophysiologie Animale, Faculté des Sciences de Sfax, PO Box 1171, Sfax Tunisie PO Box 1171 Sfax Tunisie
| |
Collapse
|
34
|
Martínez-Guerrero LJ, Morales M, Ekins S, Wright SH. Lack of Influence of Substrate on Ligand Interaction with the Human Multidrug and Toxin Extruder, MATE1. Mol Pharmacol 2016; 90:254-64. [PMID: 27418674 DOI: 10.1124/mol.116.105056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/30/2016] [Indexed: 12/14/2022] Open
Abstract
Multidrug and toxin extruder (MATE) 1 plays a central role in mediating renal secretion of organic cations, a structurally diverse collection of compounds that includes ∼40% of prescribed drugs. Because inhibition of transport activity of other multidrug transporters, including the organic cation transporter (OCT) 2, is influenced by the structure of the transported substrate, the present study screened over 400 drugs as inhibitors of the MATE1-mediated transport of four structurally distinct organic cation substrates: the commonly used drugs: 1) metformin and 2) cimetidine; and two prototypic cationic substrates, 3) 1-methyl-4-phenylpyridinium (MPP), and 4) the novel fluorescent probe, N,N,N-trimethyl-2-[methyl(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino]ethanaminium iodide. Transport was measured in Chinese hamster ovary cells that stably expressed the human ortholog of MATE1. Comparison of the resulting inhibition profiles revealed no systematic influence of substrate structure on inhibitory efficacy. Similarly, IC50 values for 26 structurally diverse compounds revealed no significant influence of substrate structure on the kinetic interaction of inhibitor with MATE1. The IC50 data were used to generate three-dimensional quantitative pharmacophores that identified hydrophobic regions, H-bond acceptor sites, and an ionizable (cationic) feature as key determinants for ligand binding to MATE1. In summary, in contrast to the behavior observed with some other multidrug transporters, including OCT2, the results suggest that substrate identity exerts comparatively little influence on ligand interaction with MATE1.
Collapse
Affiliation(s)
- Lucy J Martínez-Guerrero
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.J.M.-G., M.M., S.H.W.); and Collaborations in Chemistry, Fuquay-Varina, North Carolina (S.E.)
| | - Mark Morales
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.J.M.-G., M.M., S.H.W.); and Collaborations in Chemistry, Fuquay-Varina, North Carolina (S.E.)
| | - Sean Ekins
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.J.M.-G., M.M., S.H.W.); and Collaborations in Chemistry, Fuquay-Varina, North Carolina (S.E.)
| | - Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.J.M.-G., M.M., S.H.W.); and Collaborations in Chemistry, Fuquay-Varina, North Carolina (S.E.)
| |
Collapse
|
35
|
Wagner DJ, Hu T, Wang J. Polyspecific organic cation transporters and their impact on drug intracellular levels and pharmacodynamics. Pharmacol Res 2016; 111:237-246. [PMID: 27317943 DOI: 10.1016/j.phrs.2016.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/02/2016] [Indexed: 01/11/2023]
Abstract
Most drugs are intended to act on molecular targets residing within a specific tissue or cell type. Therefore, the drug concentration within the target tissue or cells is most relevant to its pharmacological effect. Increasing evidences suggest that drug transporters not only play a significant role in governing systemic drug levels, but are also an important gate keeper for intra-tissue and intracellular drug concentrations. This review focuses on polyspecific organic cation transporters, which include the organic cation transporters 1-3 (OCT1-3), the multidrug and toxin extrusion proteins 1-2 (MATE1-2) and the plasma membrane monoamine transporter (PMAT). Following an overview of the tissue distribution, transport mechanisms, and functional characteristics of these transporters, we highlight the studies demonstrating the ability of locally expressed OCTs to impact intracellular drug concentrations and directly influence their pharmacological and toxicological activities. Specifically, OCT1-mediated metformin access to its site of action in the liver is impacted by genetic polymorphisms and chemical inhibition of OCT1. The impact of renal OCT2 and MATE1/2-K in cisplatin intrarenal accumulation and nephrotoxicity is reviewed. New data demonstrating the role of OCT3 in salivary drug accumulation and secretion is discussed. Whenever possible, the pharmacodynamic response and toxicological effects is presented and discussed in light of intra-tissue and intracellular drug exposure. Current challenges, knowledge gaps, and future research directions are discussed. Understanding the impact of transporters on intra-tissue and intracellular drug concentrations has important implications for rational-based optimization of drug efficacy and safety.
Collapse
Affiliation(s)
- David J Wagner
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| | - Tao Hu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States.
| |
Collapse
|
36
|
Characterization of 22 Antituberculosis Drugs for Inhibitory Interaction Potential on Organic Anionic Transporter Polypeptide (OATP)-Mediated Uptake. Antimicrob Agents Chemother 2016; 60:3096-105. [PMID: 26976869 DOI: 10.1128/aac.02765-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/06/2016] [Indexed: 12/21/2022] Open
Abstract
We investigated the inhibitory interaction potential of 22 currently marketed antituberculosis (TB) drugs on organic anion-transporting polypeptide 1B1 (OATP1B1)-, OATP2B1-, and OATP1B3-mediated uptake using in vitro Xenopus oocytes and HEK cells. Rifabutin, ethambutol, amoxicillin, linezolid, p-amino salicylic acid, and rifapentine exhibited mild to moderate inhibitory effects on OATP-mediated uptake of estrone-3 sulfate, estradiol 17β-d-glucuronide, and rosuvastatin. The 50% inhibitory concentration (IC50) values of rifabutin, amoxicillin, ethambutol, p-amino salicylic acid, and linezolid were 35.4, 36.2, 57.6, 72.6, and 65.9 μM, respectively, for uptake mediated by organic anionic transporter polypeptide 1B1 (OATP1B1) and 28.8, 28.9, 53.9, 31.5, and 61.0 μM, respectively, for uptake mediated by organic anionic transporter polypeptide 1B3 (OATP1B3). Streptomycin and linezolid showed greater inhibition of organic anionic transporter polypeptide 2B1 (OATP2B1)-mediated uptake, with IC50 values of 33.2 and 35.6 μM, respectively, along with mild inhibition of other drugs. Furthermore, rifabutin, amoxicillin, and rifapentine significantly inhibited OATP1B1-mediated rosuvastatin uptake, with IC50 values of 12.3, 13.0, and 11.0 μM, respectively, which showed a similar profile to estrone-3 sulfate uptake. The calculated R values ([I]u inlet,max/Ki, where [I]u inlet,max represents the maximum estimated inhibitor concentration inlet to the liver and Ki is the inhibition constant) as the drug-drug interaction (DDI) indexes of PAS, ethambutol, and amoxicillin were 26.1, 6.5, and 4.3 for OATP1B1 and 52.0, 8.0, and 4.6 for OATP1B3, and those for streptomycin, amikacin, and linezolid were 5.0, 4.2, and 4.4 for OATP2B1, respectively, suggesting a higher possibility of in vivo DDIs. This study is the first comprehensive report to show the novel inhibitory potential of 22 marketed anti-TB drugs on OATP-mediated uptake, providing evidence for future in vivo clinical DDI studies.
Collapse
|
37
|
Martínez-Guerrero LJ, Evans KK, Dantzler WH, Wright SH. The multidrug transporter MATE1 sequesters OCs within an intracellular compartment that has no influence on OC secretion in renal proximal tubules. Am J Physiol Renal Physiol 2016; 310:F57-67. [PMID: 26538438 DOI: 10.1152/ajprenal.00318.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/29/2015] [Indexed: 01/21/2023] Open
Abstract
Secretion of organic cations (OCs) across renal proximal tubules (RPTs) involves basolateral OC transporter (OCT)2-mediated uptake from the blood followed by apical multidrug and toxin extruder (MATE)1/2-mediated efflux into the tubule filtrate. Whereas OCT2 supports electrogenic OC uniport, MATE is an OC/H(+) exchanger. As assessed by epifluorescence microscopy, cultured Chinese hamster ovary (CHO) cells that stably expressed human MATE1 accumulated the fluorescent OC N,N,N-trimethyl-2-[methyl(7-nitrobenzo[c][l,2,5]oxadiazol-4-yl)amino]ethanaminium (NBD-MTMA) in the cytoplasm and in a smaller, punctate compartment; accumulation in human OCT2-expressing cells was largely restricted to the cytoplasm. A second intracellular compartment was also evident in the multicompartmental kinetics of efflux of the prototypic OC [(3)H]1-methyl-4-phenylpyridinium (MPP) from MATE1-expressing CHO cells. Punctate accumulation of NBD-MTMA was markedly reduced by coexposure of MATE1-expressing cells with 5 μM bafilomycin (BAF), an inhibitor of V-type H(+)-ATPase, and accumulation of [(3)H]MPP and [(3)H]NBD-MTMA was reduced by >30% by coexposure with 5 μM BAF. BAF had no effect on the initial rate of MATE1-mediated uptake of NBD-MTMA, suggesting that the influence of BAF was a secondary effect involving inhibition of V-type H(+)-ATPase. The accumulation of [(3)H]MPP by isolated single nonperfused rabbit RPTs was also reduced >30% by coexposure to 5 μM BAF, suggesting that the native expression in RPTs of MATE protein within endosomes can increase steady-state OC accumulation. However, the rate of [(3)H]MPP secretion by isolated single perfused rabbit RPTs was not affected by 5 μM BAF, suggesting that vesicles loaded with OCs(+) are not likely to recycle into the apical plasma membrane at a rate sufficient to provide a parallel pathway for OC secretion.
Collapse
Affiliation(s)
- L J Martínez-Guerrero
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - K K Evans
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - W H Dantzler
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - S H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
38
|
Tuncel AT, Ruppert T, Wang BT, Okun JG, Kölker S, Morath MA, Sauer SW. Maleic Acid--but Not Structurally Related Methylmalonic Acid--Interrupts Energy Metabolism by Impaired Calcium Homeostasis. PLoS One 2015; 10:e0128770. [PMID: 26086473 PMCID: PMC4473014 DOI: 10.1371/journal.pone.0128770] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 04/30/2015] [Indexed: 12/26/2022] Open
Abstract
Maleic acid (MA) has been shown to induce Fanconi syndrome via disturbance of renal energy homeostasis, though the underlying pathomechanism is still under debate. Our study aimed to examine the pathomechanism underlying maleic acid-induced nephrotoxicity. Methylmalonic acid (MMA) is structurally similar to MA and accumulates in patients affected with methymalonic aciduria, a defect in the degradation of branched-chain amino acids, odd-chain fatty acids and cholesterol, which is associated with the development of tubulointerstitial nephritis resulting in chronic renal failure. We therefore used MMA application as a control experiment in our study and stressed hPTECs with MA and MMA to further validate the specificity of our findings. MMA did not show any toxic effects on proximal tubule cells, whereas maleic acid induced concentration-dependent and time-dependent cell death shown by increased lactate dehydrogenase release as well as ethidium homodimer and calcein acetoxymethyl ester staining. The toxic effect of MA was blocked by administration of single amino acids, in particular L-alanine and L-glutamate. MA application further resulted in severe impairment of cellular energy homeostasis on the level of glycolysis, respiratory chain, and citric acid cycle resulting in ATP depletion. As underlying mechanism we could identify disturbance of calcium homeostasis. MA toxicity was critically dependent on calcium levels in culture medium and blocked by the extra- and intracellular calcium chelators EGTA and BAPTA-AM respectively. Moreover, MA-induced cell death was associated with activation of calcium-dependent calpain proteases. In summary, our study shows a comprehensive pathomechanistic concept for MA-induced dysfunction and damage of human proximal tubule cells.
Collapse
Affiliation(s)
- Ali Tunç Tuncel
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
- * E-mail:
| | - Thorsten Ruppert
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Bei-Tzu Wang
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Jürgen Günther Okun
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Stefan Kölker
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Marina Alexandra Morath
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| | - Sven Wolfgang Sauer
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children’s Hospital, Heidelberg, Germany
| |
Collapse
|
39
|
Nicolaï J, De Bruyn T, Van Veldhoven PP, Keemink J, Augustijns P, Annaert P. Verapamil hepatic clearance in four preclinical rat models: towards activity-based scaling. Biopharm Drug Dispos 2015; 36:462-80. [PMID: 25963583 DOI: 10.1002/bdd.1959] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/09/2015] [Accepted: 04/20/2015] [Indexed: 12/22/2022]
Abstract
The current study was designed to cross-validate rat liver microsomes (RLM), suspended rat hepatocytes (SRH) and the isolated perfused rat liver (IPRL) model against in vivo pharmacokinetic data, using verapamil as a model drug. Michaelis-Menten constants (Km), for the metabolic disappearance kinetics of verapamil in RLM and SRH (freshly isolated and cryopreserved), were determined and corrected for non-specific binding. The 'unbound' Km determined with RLM (2.8 µM) was divided by the 'unbound' Km determined with fresh and cryopreserved SRH (3.9 µM and 2.1 µM, respectively) to calculate the ratio of intracellular to extracellular unbound concentration (Kpu,u). Kpu,u was significantly different between freshly isolated (0.71) and cryopreserved (1.31) SRH, but intracellular capacity for verapamil metabolism was maintained after cryopreservation (200 vs. 191 µl/min/million cells). Direct comparison of intrinsic clearance values (Clint) in RLM versus SRH, yielded an activity-based scaling factor (SF) of 0.28-0.30 mg microsomal protein/million cells (MPPMC). Merging the IPRL-derived Clint with the MPPMC and SRH data, resulted in scaling factors for MPPGL (80 and 43 mg microsomal protein/g liver) and HPGL (269 and 153 million cells/g liver), respectively. Likewise, the hepatic blood flow (61 ml/min/kg b.wt) was calculated using IPRL Clint and the in vivo Cl. The scaling factors determined here are consistent with previously reported CYP450-content based scaling factors. Overall, the results show that integrated interpretation of data obtained with multiple preclinical tools (i.e. RLM, SRH, IPRL) can contribute to more reliable estimates for scaling factors and ultimately to improved in vivo clearance predictions based on in vitro experimentation.
Collapse
Affiliation(s)
- J Nicolaï
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, O&N2, Leuven, Belgium
| | - T De Bruyn
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, O&N2, Leuven, Belgium
| | - P P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions, KU Leuven Department of Cellular and Molecular Medicine, O&N1, Leuven, Belgium
| | - J Keemink
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, O&N2, Leuven, Belgium
| | - P Augustijns
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, O&N2, Leuven, Belgium
| | - P Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, O&N2, Leuven, Belgium
| |
Collapse
|
40
|
(125)I-Labelled 2-Iodoestrone-3-sulfate: synthesis, characterization and OATP mediated transport studies in hormone dependent and independent breast cancer cells. Nucl Med Biol 2014; 42:274-82. [PMID: 25542669 DOI: 10.1016/j.nucmedbio.2014.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Organic Anion Transporting Polypeptides (OATP) are a family of membrane associated transporters that facilitate estrone-3-sulphate (E3S) uptake by hormone dependent, post-menopausal breast cancers. We have established E3S as a potential ligand for targeting hormone dependent breast cancer cells, and in this study sought to prepare and investigate radioiodinated E3S as a tool to study the OATP system. METHODS 2- and 4-Iodoestrone-3-sulfates were prepared from estrone via aromatic iodination followed by a rapid and high yielding sulfation procedure. The resulting isomers were separated by preparative HPLC and verified by (1)H NMR and analytical HPLC. Transport studies of 2- and 4-[(125)I]-E3S were conducted in hormone dependent (i.e. MCF-7) and hormone independent (i.e. MDA-MB-231) breast cancer cells in the presence or absence of the specific transport inhibitor, bromosulfophthalein (BSP). Cellular localization of OATP1A2, OATP2B1, OATP3A1 and OATP4A1 were determined by immunofluorescence analysis using anti-Na(+)/K(+) ATPase-α (1:100 dilution) and DAPI as plasma membrane and nuclear markers, respectively. RESULTS Significantly (p<0.01) higher total accumulation of 2-[(125)I]-E3S was observed in hormone dependent MCF-7 as compared to hormone independent MDA-MB-231 breast cancer cells. In contrast 4-[(125)I]-E3S did not show cellular accumulation in either case. The efficiency of 2-[(125)I]-E3S transport (expressed as a ratio of Vmax/Km) was 2.4 times greater in the MCF-7 as compared to the MDA-MB-231 breast cancer cells. OATP1A2, OATP3A1 and OATP4A1 expression was localized in plasma membranes of MCF-7 and MDA-MB-231 cells confirming the functional role of these transporters in radioiodinated E3S cellular uptake. CONCLUSION An efficient method for the preparation of 2- and 4-[(125)I]-E3S was developed and where the former demonstrated potential as an in vitro probe for the OATP system. The new E3S probe can be used to study the OATP system and as a platform to create radiopharmaceuticals for imaging breast cancer.
Collapse
|
41
|
Abstract
The properties of intestinal folate absorption were documented decades ago. However, it was only recently that the proton-coupled folate transporter (PCFT) was identified and its critical role in folate transport across the apical brush-border membrane of the proximal small intestine established by the loss-of-function mutations identified in the PCFT gene in subjects with hereditary folate malabsorption and, more recently, by the Pcft-null mouse. This article reviews the current understanding of the properties of PCFT-mediated transport and how they differ from those of the reduced folate carrier. Other processes that contribute to the transport of folates across the enterocyte, along with the contribution of the enterohepatic circulation, are considered. Important unresolved issues are addressed, including the mechanism of intestinal folate absorption in the absence of PCFT and regulation of PCFT gene expression. The impact of a variety of ions, organic molecules, and drugs on PCFT-mediated folate transport is described.
Collapse
Affiliation(s)
- Michele Visentin
- Departments of Molecular Pharmacology and Medicine, Albert Einstein College of Medicine, Bronx, New York 10461; , , ,
| | | | | | | |
Collapse
|
42
|
Gotovdorj T, Lee E, Lim Y, Cha EJ, Kwon D, Hong E, Kim Y, Oh MY. 2,3,7,8-Tetrachlorodibenzo-p-dioxin induced cell-specific drug transporters with acquired cisplatin resistance in cisplatin sensitive cancer cells. J Korean Med Sci 2014; 29:1188-98. [PMID: 25246735 PMCID: PMC4168170 DOI: 10.3346/jkms.2014.29.9.1188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/05/2014] [Indexed: 12/23/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can induce drug transporter genes such as the ATP-binding cassette G member 2 (ABCG2), which contributes to multidrug resistance. We investigated the effect of TCDD pretreatment on drug transporters induction from cancer cells of various origins. Cell viabilities after treatment of cisplatin were measured to evaluate acquiring cisplatin resistance by TCDD. Acquring cisplatin resistance was found only in cisplatin senstivie cancer cells including gastric SNU601, colon LS180, brain CRT-MG and lymphoma Jurkat cells which showed a significant increase in cell viability after combined treatment with TCDD and cisplatin. High increase of ABCG2 gene expression was found in SNU601 and LS180 cells with a mild increase in the expression of the ABCC3, ABCC5,and SLC29A2 genes in SNU601 cells, and of major vault protein (MVP) in LS180 cells. The AhR inhibitor kaempferol suppressed the upregulation of ABCG2 expression and reversed the TCDD-induced increase in cell viability in LS180 cells. However, in CRT-MG cells, other transporter genes including ABCC1, ABCC5, ABCA3, ABCA2, ABCB4, ABCG1, and SLC29A1 were up-regulated. These findings suggested the acquiring cisplatin resistance by TCDD associated with cancer cell-type-specific induction of drug transporters.
Collapse
Affiliation(s)
- Tuvshinjargal Gotovdorj
- Molecular, Cellular and Developmental Biology, Division of Biomedical Science, Graduate School, Korea University, Seoul, Korea
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Eunil Lee
- Molecular, Cellular and Developmental Biology, Division of Biomedical Science, Graduate School, Korea University, Seoul, Korea
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, Korea
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
- Graduate School of Public Health, Korea University, Seoul, Korea
| | - Yongchul Lim
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Eun Jeong Cha
- Department of Preventive Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Daeho Kwon
- Department of Microbiology, College of Medicine, Kwandong University, Gangneung, Korea
| | - Eunyoung Hong
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
| | - YunJeong Kim
- Graduate School of Public Health, Korea University, Seoul, Korea
| | - Min-Yeong Oh
- Graduate School of Public Health, Korea University, Seoul, Korea
| |
Collapse
|
43
|
Leavens TL, Tell LA, Kissell LW, Smith GW, Smith DJ, Wagner SA, Shelver WL, Wu H, Baynes RE, Riviere JE. Development of a physiologically based pharmacokinetic model for flunixin in cattle (Bos taurus). Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2014; 31:1506-21. [PMID: 25082521 DOI: 10.1080/19440049.2014.938363] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Frequent violation of flunixin residues in tissues from cattle has been attributed to non-compliance with the USFDA-approved route of administration and withdrawal time. However, the effect of administration route and physiological differences among animals on tissue depletion has not been determined. The objective of this work was to develop a physiologically based pharmacokinetic (PBPK) model to predict plasma, liver and milk concentrations of flunixin in cattle following intravenous (i.v.), intramuscular (i.m.) or subcutaneous (s.c.) administration for use as a tool to determine factors that may affect the withdrawal time. The PBPK model included blood flow-limited distribution in all tissues and elimination in the liver, kidney and milk. Regeneration of parent flunixin due to enterohepatic recirculation and hydrolysis of conjugated metabolites was incorporated in the liver compartment. Values for physiological parameters were obtained from the literature, and partition coefficients for all tissues but liver and kidney were derived empirically. Liver and kidney partition coefficients and elimination parameters were estimated for 14 pharmacokinetic studies (including five crossover studies) from the literature or government sources in which flunixin was administered i.v., i.m. or s.c. Model simulations compared well with data for the matrices following all routes of administration. Influential model parameters included those that may be age or disease-dependent, such as clearance and rate of milk production. Based on the model, route of administration would not affect the estimated days to reach the tolerance concentration (0.125 mg kg(-1)) in the liver of treated cattle. The majority of USDA-reported violative residues in liver were below the upper uncertainty predictions based on estimated parameters, which suggests the need to consider variability due to disease and age in establishing withdrawal intervals for drugs used in food animals. The model predicted that extravascular routes of administration prolonged flunixin concentrations in milk, which could result in violative milk residues in treated cattle.
Collapse
Affiliation(s)
- Teresa L Leavens
- a Center for Chemical Toxicology Research and Pharmacokinetics, Department of Population Health and Pathobiology , College of Veterinary Medicine, North Carolina State University , Raleigh , NC , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Qualitative de novo analysis of full length cDNA and quantitative analysis of gene expression for common marmoset (Callithrix jacchus) transcriptomes using parallel long-read technology and short-read sequencing. PLoS One 2014; 9:e100936. [PMID: 24977701 PMCID: PMC4076266 DOI: 10.1371/journal.pone.0100936] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/28/2014] [Indexed: 12/24/2022] Open
Abstract
The common marmoset (Callithrix jacchus) is a non-human primate that could prove useful as human pharmacokinetic and biomedical research models. The cytochromes P450 (P450s) are a superfamily of enzymes that have critical roles in drug metabolism and disposition via monooxygenation of a broad range of xenobiotics; however, information on some marmoset P450s is currently limited. Therefore, identification and quantitative analysis of tissue-specific mRNA transcripts, including those of P450s and flavin-containing monooxygenases (FMO, another monooxygenase family), need to be carried out in detail before the marmoset can be used as an animal model in drug development. De novo assembly and expression analysis of marmoset transcripts were conducted with pooled liver, intestine, kidney, and brain samples from three male and three female marmosets. After unique sequences were automatically aligned by assembling software, the mean contig length was 718 bp (with a standard deviation of 457 bp) among a total of 47,883 transcripts. Approximately 30% of the total transcripts were matched to known marmoset sequences. Gene expression in 18 marmoset P450- and 4 FMO-like genes displayed some tissue-specific patterns. Of these, the three most highly expressed in marmoset liver were P450 2D-, 2E-, and 3A-like genes. In extrahepatic tissues, including brain, gene expressions of these monooxygenases were lower than those in liver, although P450 3A4 (previously P450 3A21) in intestine and P450 4A11- and FMO1-like genes in kidney were relatively highly expressed. By means of massive parallel long-read sequencing and short-read technology applied to marmoset liver, intestine, kidney, and brain, the combined next-generation sequencing analyses reported here were able to identify novel marmoset drug-metabolizing P450 transcripts that have until now been little reported. These results provide a foundation for mechanistic studies and pave the way for the use of marmosets as model animals for drug development in the future.
Collapse
|
46
|
Wulkersdorfer B, Wanek T, Bauer M, Zeitlinger M, Müller M, Langer O. Using positron emission tomography to study transporter-mediated drug-drug interactions in tissues. Clin Pharmacol Ther 2014; 96:206-13. [PMID: 24682030 PMCID: PMC4153445 DOI: 10.1038/clpt.2014.70] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/21/2014] [Indexed: 01/08/2023]
Abstract
Drug disposition is highly regulated by membrane transporters. Some transporter-mediated drug–drug interactions (DDIs) may not manifest themselves in changes in systemic exposure but rather in changes in tissue exposure of drugs. To better assess the impact of transporter-mediated DDIs in tissues, positron emission tomography (PET)—a noninvasive imaging method—plays an increasingly important role. In this article, we provide examples of how PET can be used to assess transporter-mediated DDIs in different organs.
Collapse
Affiliation(s)
- B Wulkersdorfer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - T Wanek
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - M Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - M Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - M Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - O Langer
- 1] Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria [2] Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
47
|
Hoehne A, Behera D, Parsons WH, James ML, Shen B, Borgohain P, Bodapati D, Prabhakar A, Gambhir SS, Yeomans DC, Biswal S, Chin FT, Bois JD. A 18F-Labeled Saxitoxin Derivative for in Vivo PET-MR Imaging of Voltage-Gated Sodium Channel Expression Following Nerve Injury. J Am Chem Soc 2013; 135:18012-5. [DOI: 10.1021/ja408300e] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Aileen Hoehne
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Deepak Behera
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - William H. Parsons
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Michelle L. James
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Bin Shen
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Preeti Borgohain
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Deepika Bodapati
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Archana Prabhakar
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Sanjiv S. Gambhir
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - David C. Yeomans
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Sandip Biswal
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - Frederick T. Chin
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| | - J. Du Bois
- Department of Radiology, ‡Department of Chemistry, and §Department of
Anesthesia, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
48
|
Tang H, Shen DR, Han YH, Kong Y, Balimane P, Marino A, Gao M, Wu S, Xie D, Soars MG, O’Connell JC, Rodrigues AD, Zhang L, Cvijic ME. Development of Novel, 384-Well High-Throughput Assay Panels for Human Drug Transporters. ACTA ACUST UNITED AC 2013; 18:1072-83. [DOI: 10.1177/1087057113494807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transporter proteins are known to play a critical role in affecting the overall absorption, distribution, metabolism, and excretion characteristics of drug candidates. In addition to efflux transporters (P-gp, BCRP, MRP2, etc.) that limit absorption, there has been a renewed interest in influx transporters at the renal (OATs, OCTs) and hepatic (OATPs, BSEP, NTCP, etc.) organ level that can cause significant clinical drug-drug interactions (DDIs). Several of these transporters are also critical for hepatobiliary disposition of bilirubin and bile acid/salts, and their inhibition is directly implicated in hepatic toxicities. Regulatory agencies took action to address transporter-mediated DDI with the goal of ensuring drug safety in the clinic and on the market. To meet regulatory requirements, advanced bioassay technology and automation solutions were implemented for high-throughput transporter screening to provide structure-activity relationship within lead optimization. To enhance capacity, several functional assay formats were miniaturized to 384-well throughput including novel fluorescence-based uptake and efflux inhibition assays using high-content image analysis as well as cell-based radioactive uptake and vesicle-based efflux inhibition assays. This high-throughput capability enabled a paradigm shift from studying transporter-related issues in the development space to identifying and dialing out these concerns early on in discovery for enhanced mechanism-based efficacy while circumventing DDIs and transporter toxicities.
Collapse
Affiliation(s)
- Huaping Tang
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Ding Ren Shen
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Yong-Hae Han
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Yan Kong
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Praveen Balimane
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Anthony Marino
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Mian Gao
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Sophie Wu
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Dianlin Xie
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew G. Soars
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Jonathan C. O’Connell
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - A. David Rodrigues
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Litao Zhang
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Mary Ellen Cvijic
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
49
|
Martínez-Guerrero LJ, Wright SH. Substrate-dependent inhibition of human MATE1 by cationic ionic liquids. J Pharmacol Exp Ther 2013; 346:495-503. [PMID: 23785176 DOI: 10.1124/jpet.113.204206] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The multidrug and toxin extruders 1- and 2-K (MATE1 and MATE2-K) are expressed in the luminal membrane of renal proximal tubule cells and provide the active step in the secretion of molecules that carry a net positive charge at physiologic pH, so-called organic cations. The present study tested whether structurally distinct MATE substrates can display different quantitative profiles of inhibition when interacting with structurally distinct ligands. The tested ligands were three structurally similar cationic ionic liquids (ILs, salts in the liquid state: N-butylpyridinium, NBuPy; 1-methyl-3-butylimidazolium, Bmim; and N-butyl-N-methylpyrrolidinium, BmPy). Uptake was measured using Chinese hamster ovary cells that stably expressed MATE1 or MATE2-K. By trans-stimulation, all three ILs were transported by both MATE transporters. The three ILs also inhibited uptake of three structurally distinct MATE substrates: 1-methyl-4-phenylpyridinium (MPP), triethylmethylammonium (TEMA), and N,N,N-trimethyl-2-[methyl(7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino]ethanaminium (NBD-MTMA). MATE1 displayed a higher affinity for the pyridinium-based NBuPy (IC50 values, 2-4 µM) than for either the pyrrolidinium- (BmPy; 20-70 µM) or imidazolium-based ILs (Bmim; 15-60 µM). Inhibition of MPP, TEMA, and NBD-MTMA transport by NBuPy was competitive, with comparable Ki values against all substrates. Bmim also competitively blocked the three substrates but with Ki values that differed significantly (20 µM against MPP and 30 µM against NBD-MTMA versus 60 µM against TEMA). Together, these data indicate that renal secretion of ILs by the human kidney involves MATE transporters and suggest that the mechanism of transport inhibition is ligand-dependent, supporting the hypothesis that the binding of substrates to MATE transporters involves interaction with a binding surface with multiple binding sites.
Collapse
Affiliation(s)
- Lucy J Martínez-Guerrero
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona 85724, USA
| | | |
Collapse
|
50
|
Banerjee N, Fonge H, Mikhail A, Reilly RM, Bendayan R, Allen C. Estrone-3-sulphate, a potential novel ligand for targeting breast cancers. PLoS One 2013; 8:e64069. [PMID: 23717534 PMCID: PMC3661587 DOI: 10.1371/journal.pone.0064069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 04/11/2013] [Indexed: 11/19/2022] Open
Abstract
The current study investigates the potential of estrone-3-sulphate (E3S) as a ligand for targeting Organic Anion Transporting Polypeptides (OATP), a family of membrane associated uptake transporters, for detection and diagnosis of hormone dependent breast cancers. E3S, an OATP substrate, is a predominant source of tumour estradiol in post-menopausal patients. To assess the potential of E3S as a ligand, distribution of exogenous E3S was determined at the whole body, tumour and cellular levels in murine models of hormone-dependent (MCF-7) and independent (MDA-MB-231) breast cancers. The highest levels of tumour uptake were observed at 6 h post injection (p.i) with significant difference (p = 0.04) between the level in MCF-7 (13.9±3.1%ID/g) and MDA-MB-231 (10.4±1.1%ID/g) (%ID/g: percentage of the total injected dose per gram tissue). The highest tumour-to-blood ratios (MCF-7∶7.4±1.2; MDA-MB-231∶9.1±2.1) were observed at 48 p.i., and highest tumour-to-muscle ratios (MCF-7∶10.7±1.5; MDA-MB-231∶3.8±0.7) were observed at 6 h p.i. Analogous to total tumour uptake, ex vivo tumour cell uptake at 2 h p.i. was 6 fold higher in MCF-7 in comparison to MDA-MB-231 tumour cells. Blocking studies, conducted by pre-administration of 100-fold excess E3S, resulted in significantly lower (MCF-7: p = 0.01; MDA-MB-231: p = 0.02) tumour uptake in both xenograft models, suggesting the involvement of an active carrier-mediated process. The expression of OATP1A2 was detected in tumour sections from both xenografts, with significantly higher expression (p = 0.002) in the MCF-7 xenografts. Overall, the higher tumour uptake and tumour-to-muscle ratio, alongside the higher expression of OATP1A2, in the MCF-7 xenograft model suggests the potential of E3S to serve as a novel ligand for targeting hormone dependent breast cancers.
Collapse
Affiliation(s)
- Nilasha Banerjee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Humphrey Fonge
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Mikhail
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Raymond M. Reilly
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Christine Allen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|