1
|
Huttunen J, Adla SK, Markowicz-Piasecka M, Huttunen KM. Increased/Targeted Brain (Pro)Drug Delivery via Utilization of Solute Carriers (SLCs). Pharmaceutics 2022; 14:pharmaceutics14061234. [PMID: 35745806 PMCID: PMC9228667 DOI: 10.3390/pharmaceutics14061234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane transporters have a crucial role in compounds’ brain drug delivery. They allow not only the penetration of a wide variety of different compounds to cross the endothelial cells of the blood–brain barrier (BBB), but also the accumulation of them into the brain parenchymal cells. Solute carriers (SLCs), with nearly 500 family members, are the largest group of membrane transporters. Unfortunately, not all SLCs are fully characterized and used in rational drug design. However, if the structural features for transporter interactions (binding and translocation) are known, a prodrug approach can be utilized to temporarily change the pharmacokinetics and brain delivery properties of almost any compound. In this review, main transporter subtypes that are participating in brain drug disposition or have been used to improve brain drug delivery across the BBB via the prodrug approach, are introduced. Moreover, the ability of selected transporters to be utilized in intrabrain drug delivery is discussed. Thus, this comprehensive review will give insights into the methods, such as computational drug design, that should be utilized more effectively to understand the detailed transport mechanisms. Moreover, factors, such as transporter expression modulation pathways in diseases that should be taken into account in rational (pro)drug development, are considered to achieve successful clinical applications in the future.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Magdalena Markowicz-Piasecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Correspondence:
| |
Collapse
|
2
|
Markowicz-Piasecka M, Markiewicz A, Darłak P, Sikora J, Adla SK, Bagina S, Huttunen KM. Current Chemical, Biological, and Physiological Views in the Development of Successful Brain-Targeted Pharmaceutics. Neurotherapeutics 2022; 19:942-976. [PMID: 35391662 PMCID: PMC9294128 DOI: 10.1007/s13311-022-01228-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
One of the greatest challenges with successful pharmaceutical treatments of central nervous system (CNS) diseases is the delivery of drugs into their target sites with appropriate concentrations. For example, the physically tight blood-brain barrier (BBB) effectively blocks compounds from penetrating into the brain, also by the action of metabolizing enzymes and efflux transport mechanisms. However, many endogenous compounds, including both smaller compounds and macromolecules, like amino acids, sugars, vitamins, nucleosides, hormones, steroids, and electrolytes, have their peculiar internalization routes across the BBB. These delivery mechanisms, namely carrier-mediated transport and receptor-mediated transcytosis have been utilized to some extent in brain-targeted drug development. The incomplete knowledge of the BBB and the smaller than a desirable number of chemical tools have hindered the development of successful brain-targeted pharmaceutics. This review discusses the recent advancements achieved in the field from the point of medicinal chemistry view and discusses how brain drug delivery can be improved in the future.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Agata Markiewicz
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Patrycja Darłak
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Medical University of Lodz, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Sreelatha Bagina
- Charles River Discovery Research Services Finland Oy, Neulaniementie 4, 70210 Kuopio, Finland
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| |
Collapse
|
3
|
Wang HX, Li WP, Zhang MM, Xie MS, Qu GR, Guo HM. Synthesis of chiral pyrimidine-substituted diester D-A cyclopropanes via asymmetric cyclopropanation of phenyliodonium ylides. Chem Commun (Camb) 2020; 56:11649-11652. [PMID: 33000801 DOI: 10.1039/d0cc04536e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A highly enantioselective cyclopropanation to synthesize pyrimidine-substituted diester D-A cyclopropanes is reported. Various N1-vinylpyrimidines react well with phenyliodonium ylides, delivering chiral cyclopropanes in up to 97% yield with up to 99% ee. Through simple [3+2] annulation with benzaldehyde or ethyl glyoxylate, different chiral pyrimidine nucleoside analogues with a sugar ring could be obtained.
Collapse
Affiliation(s)
- Hai-Xia Wang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Wen-Peng Li
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Mi-Mi Zhang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Ming-Sheng Xie
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Gui-Rong Qu
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Hai-Ming Guo
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China.
| |
Collapse
|
4
|
Correia C, Xavier CPR, Duarte D, Ferreira A, Moreira S, Vasconcelos MH, Vale N. Development of potent CPP6-gemcitabine conjugates against human prostate cancer cell line (PC-3). RSC Med Chem 2020; 11:268-273. [PMID: 33479633 DOI: 10.1039/c9md00489k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/31/2019] [Indexed: 01/19/2023] Open
Abstract
Gemcitabine (dFdC) is a nucleoside analogue used in the treatment of various cancers, being a standard treatment for advanced pancreatic cancer. The effect of gemcitabine is severely compromised due to its rapid plasma degradation, systemic toxicity and drug resistance, which restricts its therapeutic efficacy. Our main goal was to develop new active conjugates of dFdC with novel cell-penetrating hexapeptides (CPP6) to facilitate intracellular delivery of this drug. All new peptides were prepared by solid phase peptide synthesis (SPPS), purified and characterized by HPLC and LC-MS. Cell-penetrating peptides (CPP) contain a considerably high ratio of positively charged amino acids, imparting them with cationic character. Tumor cells are characterized by an increased anionic nature of their membrane surface, a property that could be used by CPP to target these cells. The BxPC-3, MCF-7 and PC-3 cancer cell lines were used to evaluate the in vitro cytotoxicity of conjugates and the results showed that conjugating dFdC with CPP6 significantly enhanced cell growth inhibitory activity on PC-3 cells, with IC50 between 14 and 15 nM. These new conjugates have potential to become new therapeutic tools for cancer therapy.
Collapse
Affiliation(s)
- Cristiana Correia
- Laboratory of Pharmacology , Department of Drug Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal . .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Cristina P R Xavier
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Diana Duarte
- Laboratory of Pharmacology , Department of Drug Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal . .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - Abigail Ferreira
- Laboratory of Pharmacology , Department of Drug Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal . .,LAQV/REQUIMTE , Department of Chemical Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal
| | - Sara Moreira
- Laboratory of Pharmacology , Department of Drug Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal . .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal
| | - M Helena Vasconcelos
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal.,Laboratory of Microbiology , Department of Biological Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira, 228 , 4050-313 Porto , Portugal
| | - Nuno Vale
- Laboratory of Pharmacology , Department of Drug Sciences , Faculty of Pharmacy , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal . .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) , Rua Júlio Amaral de Carvalho , 45 , 4200-135 Porto , Portugal.,Instituto de Investigação e Inovação em Saúde (i3S) , University of Porto , Rua Alfredo Allen, 208 , 4200-135 Porto , Portugal.,Department of Molecular Pathology and Immunology , Abel Salazar Biomedical Sciences Institute (ICBAS) , University of Porto , Rua de Jorge Viterbo Ferreira 228 , 4050-313 Porto , Portugal
| |
Collapse
|
5
|
Wang HX, Li WP, Xia C, Xie MS, Qu GR, Guo HM. Enantioselective synthesis of chiral carbocyclic pyrimidine nucleosides via asymmetric cyclopropanation. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
6
|
Vaskó B, Juhász V, Tóth B, Kurunczi A, Fekete Z, Krisjanis Zolnerciks J, Kis E, Magnan R, Bidon-Chanal Badia A, Pastor-Anglada M, Hazai E, Bikadi Z, Fülöp F, Krajcsi P. Inhibitor selectivity of CNTs and ENTs. Xenobiotica 2018; 49:840-851. [PMID: 30022699 DOI: 10.1080/00498254.2018.1501832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The concentrative nucleoside transporters (CNT; solute carrier family 28 (SLC28)) and the equilibrative nucleoside transporters (ENT; solute carrier family 29 (SLC29)) are important therapeutic targets but may also mediate toxicity or adverse events. To explore the relative role of the base and the monosaccharide moiety in inhibitor selectivity we selected compounds that either harbor an arabinose moiety or a cytosine moiety, as these groups had several commercially available drug members. The screening data showed that more compounds harboring a cytosine moiety displayed potent interactions with the CNTs than compounds harboring the arabinose moiety. In contrast, ENTs showed a preference for compounds with an arabinose moiety. The correlation between CNT1 and CNT3 was good as five of six compounds displayed IC50 values within the threefold threshold and one displayed a borderline 4-fold difference. For CNT1 and CNT2 as well as for CNT2 and CNT3 only two of six IC50 values correlated and one displayed a borderline 4-fold difference. Interestingly, of the six compounds that potently interacted with both ENT1 and ENT2 only nelarabine displayed selectivity. Our data show differences between inhibitor selectivities of CNTs and ENTs as well as differences within the CNT family members.
Collapse
Affiliation(s)
| | | | - Beáta Tóth
- b SOLVO Biotechnology , Budaörs , Hungary
| | | | | | | | - Emese Kis
- a SOLVO Biotechnology , Szeged , Hungary
| | | | - Axel Bidon-Chanal Badia
- c Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia i Ciències de l'Alimentació and Institute of Biomedicine (IBUB), Campus de l'Alimentació de Torribera , Universitat de Barcelona , Santa Coloma de Gramenet , Spain
| | - Marçal Pastor-Anglada
- d Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia and Institute of Biomedicine (IBUB) , Universitat de Barcelona , Barcelona , Spain.,e Oncology Program , National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBER EHD), Instituto de Salud Carlos III , Madrid , Spain
| | | | | | - Ferenc Fülöp
- g Institute of Pharmaceutical Chemistry, University of Szeged , Szeged , Hungary
| | - Peter Krajcsi
- a SOLVO Biotechnology , Szeged , Hungary.,h Department of Morphology and Physiology, Faculty of Health Sciences , Semmelweis University , Budapest , Hungary.,i Faculty of Information Technology and Bionics , Pázmány Péter Catholic University , Budapest , Hungary
| |
Collapse
|
7
|
Sekhar GN, Georgian AR, Sanderson L, Vizcay-Barrena G, Brown RC, Muresan P, Fleck RA, Thomas SA. Organic cation transporter 1 (OCT1) is involved in pentamidine transport at the human and mouse blood-brain barrier (BBB). PLoS One 2017; 12:e0173474. [PMID: 28362799 PMCID: PMC5376088 DOI: 10.1371/journal.pone.0173474] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 02/21/2017] [Indexed: 02/02/2023] Open
Abstract
Pentamidine is an effective trypanocidal drug used against stage 1 Human African Trypanosomiasis (HAT). At the blood-brain barrier (BBB), it accumulates inside the endothelial cells but has limited entry into the brain. This study examined transporters involved in pentamidine transport at the human and mouse BBB using hCMEC/D3 and bEnd.3 cell lines, respectively. Results revealed that both cell lines expressed the organic cation transporters (OCT1, OCT2 and OCT3), however, P-gp was only expressed in hCMEC/D3 cells. Polarised expression of OCT1 was also observed. Functional assays found that ATP depletion significantly increased [3H]pentamidine accumulation in hCMEC/D3 cells (***p<0.001) but not in bEnd.3 cells. Incubation with unlabelled pentamidine significantly decreased accumulation in hCMEC/D3 and bEnd.3 cells after 120 minutes (***p<0.001). Treating both cell lines with haloperidol and amantadine also decreased [3H]pentamidine accumulation significantly (***p<0.001 and **p<0.01 respectively). However, prazosin treatment decreased [3H]pentamidine accumulation only in hCMEC/D3 cells (*p<0.05), and not bEnd.3 cells. Furthermore, the presence of OCTN, MATE, PMAT, ENT or CNT inhibitors/substrates had no significant effect on the accumulation of [3H]pentamidine in both cell lines. From the data, we conclude that pentamidine interacts with multiple transporters, is taken into brain endothelial cells by OCT1 transporter and is extruded into the blood by ATP-dependent mechanisms. These interactions along with the predominant presence of OCT1 in the luminal membrane of the BBB contribute to the limited entry of pentamidine into the brain. This information is of key importance to the development of pentamidine based combination therapies which could be used to treat CNS stage HAT by improving CNS delivery, efficacy against trypanosomes and safety profile of pentamidine.
Collapse
Affiliation(s)
- Gayathri N. Sekhar
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
| | - Ana R. Georgian
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
| | - Lisa Sanderson
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
| | - Gema Vizcay-Barrena
- King’s College London, Centre for Ultrastructural Imaging, King’s College London, London Bridge United Kingdom
| | - Rachel C. Brown
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
| | - Paula Muresan
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
| | - Roland A. Fleck
- King’s College London, Centre for Ultrastructural Imaging, King’s College London, London Bridge United Kingdom
| | - Sarah A. Thomas
- King’s College London, Institute of Pharmaceutical Science, Waterloo, London United Kingdom
- * E-mail:
| |
Collapse
|
8
|
Bird NTE, Elmasry M, Jones R, Psarelli E, Dodd J, Malik H, Greenhalf W, Kitteringham N, Ghaneh P, Neoptolemos JP, Palmer D. Immunohistochemical hENT1 expression as a prognostic biomarker in patients with resected pancreatic ductal adenocarcinoma undergoing adjuvant gemcitabine-based chemotherapy. Br J Surg 2017; 104:328-336. [PMID: 28199010 DOI: 10.1002/bjs.10482] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/12/2016] [Accepted: 12/08/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Human equilibrative nucleoside transporters (hENTs) are transmembranous proteins that facilitate the uptake of nucleosides and nucleoside analogues, such as gemcitabine, into the cell. The abundance of hENT1 transporters in resected pancreatic ductal adenocarcinoma (PDAC) might make hENT1 a potential biomarker of response to adjuvant chemotherapy. The aim of this study was to see whether hENT1 expression, as determined by immunohistochemistry, was a suitable predictive marker for subsequent treatment with gemcitabine-based adjuvant chemotherapy. METHODS A systematic review was performed, searching databases from January 1997 to January 2016. Articles pertaining to hENT1 immunohistochemical analysis in resected PDAC specimens from patients who subsequently underwent adjuvant gemcitabine-based chemotherapy were identified. Eligible studies were required to contain survival data, reporting specifically overall survival (OS) and disease-free survival (DFS) with associated hazard ratios (HRs) stratified by hENT1 status. RESULTS Of 42 articles reviewed, eight were suitable for review, with seven selected for quantitative meta-analysis. The total number of patients included in the meta-analysis was 770 (405 hENT1-negative, 365 hENT1-positive). Immunohistochemically detected hENT1 expression was significantly associated with both prolonged DFS (HR 0·58, 95 per cent c.i. 0·42 to 0·79) and OS (HR 0·52, 0·38 to 0·72) in patients receiving adjuvant gemcitabine but not those having fluoropyrimidine-based adjuvant therapy. CONCLUSION Expression of hENT1 is a suitable prognostic biomarker in patients undergoing adjuvant gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- N T E Bird
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - M Elmasry
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - R Jones
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - E Psarelli
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - J Dodd
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - H Malik
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - W Greenhalf
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - N Kitteringham
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - P Ghaneh
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - J P Neoptolemos
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - D Palmer
- Liverpool University Pharmacology Unit, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| |
Collapse
|
9
|
Ciccolini J, Serdjebi C, Le Thi Thu H, Lacarelle B, Milano G, Fanciullino R. Nucleoside analogs: ready to enter the era of precision medicine? Expert Opin Drug Metab Toxicol 2016; 12:865-77. [DOI: 10.1080/17425255.2016.1192128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Joseph Ciccolini
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Cindy Serdjebi
- Assistance Publique Hôpitaux de Marseille. Multidisciplinary Oncology & Therapeutic Innovations dpt, Aix Marseille University, Marseille, France
| | - Hau Le Thi Thu
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Bruno Lacarelle
- SMARTc Unit, Inserm S_911 CRO2 Aix-Marseille University, Marseille, France
| | - Gerard Milano
- Oncopharmacology Unit, Centre Antoine Lacassagne, Nice, France
| | | |
Collapse
|
10
|
Tatani K, Hiratochi M, Kikuchi N, Kuramochi Y, Watanabe S, Yamauchi Y, Itoh F, Isaji M, Shuto S. Identification of Adenine and Benzimidazole Nucleosides as Potent Human Concentrative Nucleoside Transporter 2 Inhibitors: Potential Treatment for Hyperuricemia and Gout. J Med Chem 2016; 59:3719-31. [DOI: 10.1021/acs.jmedchem.5b01884] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Kazuya Tatani
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Masahiro Hiratochi
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Norihiko Kikuchi
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Yu Kuramochi
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Shinjiro Watanabe
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Yuji Yamauchi
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Fumiaki Itoh
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | - Masayuki Isaji
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Hotakakashiwabara, Azumino, Nagano 399-8304, Japan
| | | |
Collapse
|
11
|
Wang W, Wang J, Li Z, Zhu M, Zhang Z, Wang Y, Jing H. Promoter hypermethylation of PTPL1, PTPN6, DAPK, p16 and 5-azacitidine inhibits growth in DLBCL. Oncol Rep 2015; 35:139-46. [PMID: 26498513 DOI: 10.3892/or.2015.4347] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/18/2015] [Indexed: 11/05/2022] Open
Abstract
Aberrant hypermethylation of CpG islands of tumor suppressor is one of the mechanisms for epigenetic loss of gene function. In the present study, the methylation status of the promoter regions of protein tyrosine phosphatase (PTPN) 6, DAPK, and p16 were studied using methylation-specific polymerase chain reaction (MSP) in 26 diffuse large B cell lymphoma (DLBCL) lymphomas. In OCI-LY1 cell line, gene methylation status, expression of PTPL1 and its reactivation by DNA demethylation was determined by PCR and on the protein level by western blotting. ELISA-like reaction was used to detect global DNA methylation measurement. Induction of apoptosis by 5-azacitidine was analyzed by Annexin V/PI staining and flow cytometry. Our results show that hypermethylation of the PTPN6 gene promoter region was found in 15.4% (4/26), the DAPK gene promoter region in 30.8% (8/26), the p16 gene promoter region in 7.7% (2/26). Notably, we identified that PTPL1 was hypermethylated and transcriptionally silenced in OCI-LY1 cell line. The expression of PTPL1 was re-inducible by 5-azacytidine. 5-azacytidine also inhibits the proliferation and decreases the global methylation level of the OCI-LY1 cell line. We can conclude from our study that a higher prevalence of methylation of PTPL1, PTPN6, DAPK and p16 occur in DLBCL. Our data also highlights 5-azacytidine as a potential therapeutic candidate for DLBCL. Further studies are required to substantiate the role of methylation of PTPL1, PTPN6, DAPK and p16 as a marker in diffuse large B cell lymphoma.
Collapse
Affiliation(s)
- Wenming Wang
- Department of Hematology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Jing Wang
- Department of Hematology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Mingxia Zhu
- Department of Hematology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Yanfang Wang
- Department of Hematology, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Hongmei Jing
- Department of Hematology, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
12
|
Arimany-Nardi C, Errasti-Murugarren E, Minuesa G, Martinez-Picado J, Gorboulev V, Koepsell H, Pastor-Anglada M. Nucleoside transporters and human organic cation transporter 1 determine the cellular handling of DNA-methyltransferase inhibitors. Br J Pharmacol 2015; 171:3868-80. [PMID: 24780098 DOI: 10.1111/bph.12748] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 04/02/2014] [Accepted: 04/15/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhibitors of DNA methyltransferases (DNMTs), such as azacytidine, decitabine and zebularine, are used for the epigenetic treatment of cancer. Their action may depend upon their translocation across the plasma membrane. The aim of this study was to identify transporter proteins contributing to DNMT inhibitor action. EXPERIMENTAL APPROACH Drug interactions with selected hCNT and hENT proteins were studied in transiently transfected HeLa and MDCK cells. Interaction with human organic cation transporters (hOCTs) was assessed in transiently transfected HeLa cells and Xenopus laevis oocytes. KEY RESULTS Zebularine uptake was mediated by hCNT1, hCNT3 and hENT2. Decitabine interacted with but was not translocated by any nucleoside transporter (NT) type. hCNT expression at the apical domain of MDCK cells promoted net vectorial flux of zebularine. Neither hOCT1 nor hOCT2 transported decitabine, but both were involved in the efflux of zebularine, suggesting these proteins act as efflux transporters. hOCT1 polymorphic variants, known to alter function, decreased zebularine efflux. CONCLUSIONS AND IMPLICATIONS This study highlights the influence of human NTs and hOCTs on the pharmacokinetics and pharmacodynamics of selected DNMT inhibitors. As hOCTs may also behave as efflux transporters, they could contribute either to chemoresistance or to chemosensitivity, depending upon the nature of the drug or combination of drugs being used in cancer therapy.
Collapse
Affiliation(s)
- C Arimany-Nardi
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona (IBUB) & National Biomedical Research Institute on Liver and Gastrointestinal Diseaes (CIBERehd), Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
13
|
Tatani K, Hiratochi M, Nonaka Y, Isaji M, Shuto S. Identification of 8-aminoadenosine derivatives as a new class of human concentrative nucleoside transporter 2 inhibitors. ACS Med Chem Lett 2015; 6:244-8. [PMID: 25815140 DOI: 10.1021/ml500343r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 01/28/2015] [Indexed: 11/30/2022] Open
Abstract
Purine-rich foods have long been suspected as a major cause of hyperuricemia. We hypothesized that inhibition of human concentrative nucleoside transporter 2 (hCNT2) would suppress increases in serum urate levels derived from dietary purines. To test this hypothesis, the development of potent hCNT2 inhibitors was required. By modifying adenosine, an hCNT2 substrate, we successfully identified 8-aminoadenosine derivatives as a new class of hCNT2 inhibitors. Compound 12 moderately inhibited hCNT2 (IC50 = 52 ± 3.8 μM), and subsequent structure-activity relationship studies led to the discovery of compound 48 (IC50 = 0.64 ± 0.19 μM). Here we describe significant findings about structural requirements of 8-aminoadenosine derivatives for exhibiting potent hCNT2 inhibitory activity.
Collapse
Affiliation(s)
- Kazuya Tatani
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Kashiwabara,
Hotaka, Azumino, Nagano 399-8304, Japan
- Faculty
of Pharmaceutical Science and §Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| | - Masahiro Hiratochi
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Kashiwabara,
Hotaka, Azumino, Nagano 399-8304, Japan
| | - Yoshinori Nonaka
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Kashiwabara,
Hotaka, Azumino, Nagano 399-8304, Japan
| | - Masayuki Isaji
- Central
Research Laboratories, Kissei Pharmaceutical Co., Ltd., 4365-1, Kashiwabara,
Hotaka, Azumino, Nagano 399-8304, Japan
| | - Satoshi Shuto
- Faculty
of Pharmaceutical Science and §Center for Research and Education on Drug Discovery, Hokkaido University, Kita-12, Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| |
Collapse
|
14
|
Choi JS, Maity A, Gray T, Berdis AJ. A metal-containing nucleoside that possesses both therapeutic and diagnostic activity against cancer. J Biol Chem 2015; 290:9714-26. [PMID: 25713072 DOI: 10.1074/jbc.m114.620294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Indexed: 12/29/2022] Open
Abstract
Nucleoside transport is an essential process that helps maintain the hyperproliferative state of most cancer cells. As such, it represents an important target for developing diagnostic and therapeutic agents that can effectively detect and treat cancer, respectively. This report describes the development of a metal-containing nucleoside designated Ir(III)-PPY nucleoside that displays both therapeutic and diagnostic properties against the human epidermal carcinoma cell line KB3-1. The cytotoxic effects of Ir(III)-PPY nucleoside are both time- and dose-dependent. Flow cytometry analyses validate that the nucleoside analog causes apoptosis by blocking cell cycle progression at G2/M. Fluorescent microscopy studies show rapid accumulation in the cytoplasm within 4 h. However, more significant accumulation is observed in the nucleus and mitochondria after 24 h. This localization is consistent with the ability of the metal-containing nucleoside to influence cell cycle progression at G2/M. Mitochondrial depletion is also observed after longer incubations (Δt ∼48 h), and this effect may produce additional cytotoxic effects. siRNA knockdown experiments demonstrate that the nucleoside transporter, hENT1, plays a key role in the cellular entry of Ir(III)-PPY nucleoside. Collectively, these data provide evidence for the development of a metal-containing nucleoside that functions as a combined therapeutic and diagnostic agent against cancer.
Collapse
Affiliation(s)
- Jung-Suk Choi
- From the Department of Chemistry and the Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio 44115 and
| | - Ayan Maity
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Thomas Gray
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106
| | - Anthony J Berdis
- From the Department of Chemistry and the Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio 44115 and
| |
Collapse
|
15
|
Thomas S, Dimelow RJ. Prediction of Phosphoglycoprotein (
P‐gp
)‐Mediated Disposition in Early Drug Discovery. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/9783527673261.ch15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
16
|
|
17
|
Plačková P, Hřebabecký H, Šála M, Nencka R, Elbert T, Mertlíková-Kaiserová H. Transport mechanisms of a novel antileukemic and antiviral compound 9-norbornyl-6-chloropurine. J Enzyme Inhib Med Chem 2014; 30:57-62. [DOI: 10.3109/14756366.2013.879576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
18
|
Ekins S, Polli JE, Swaan PW, Wright SH. Computational modeling to accelerate the identification of substrates and inhibitors for transporters that affect drug disposition. Clin Pharmacol Ther 2012; 92:661-5. [PMID: 23010651 DOI: 10.1038/clpt.2012.164] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- S Ekins
- Collaborations in Chemistry, Fuquay Varina, North Carolina, USA.
| | | | | | | |
Collapse
|
19
|
Minuesa G, Huber-Ruano I, Pastor-Anglada M, Koepsell H, Clotet B, Martinez-Picado J. Drug uptake transporters in antiretroviral therapy. Pharmacol Ther 2011; 132:268-79. [DOI: 10.1016/j.pharmthera.2011.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/30/2011] [Indexed: 01/11/2023]
|
20
|
Cano-Soldado P, Pastor-Anglada M. Transporters that translocate nucleosides and structural similar drugs: structural requirements for substrate recognition. Med Res Rev 2011; 32:428-57. [DOI: 10.1002/med.20221] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Pedro Cano-Soldado
- Departament de Bioquímica i Biologia Molecular; Institut de Biomedicina de la Universitat de Barcelona (IBUB); Universitat de Barcelona and CIBER EHD; Barcelona Spain
| | - Marçal Pastor-Anglada
- Departament de Bioquímica i Biologia Molecular; Institut de Biomedicina de la Universitat de Barcelona (IBUB); Universitat de Barcelona and CIBER EHD; Barcelona Spain
| |
Collapse
|
21
|
|
22
|
Niitani M, Nishida K, Okuda H, Nagai K, Fujimoto S, Nagasawa K. Transport characteristics of mouse concentrative nucleoside transporter 1. Int J Pharm 2010; 388:168-74. [DOI: 10.1016/j.ijpharm.2009.12.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 12/18/2009] [Accepted: 12/24/2009] [Indexed: 10/20/2022]
|
23
|
Interaction of benzopyranone derivatives and related compounds with human concentrative nucleoside transporters 1, 2 and 3 heterologously expressed in porcine PK15 nucleoside transporter deficient cells. Structure–activity relationships and determinants of transporter affinity and selectivity. Biochem Pharmacol 2010; 79:307-20. [DOI: 10.1016/j.bcp.2009.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 08/26/2009] [Accepted: 08/28/2009] [Indexed: 11/23/2022]
|
24
|
Abstract
The prominent role for the drug efflux pump ABCB1 (P-glycoprotein) in mediating resistance to chemotherapy was first suggested in 1976 and sparked an incredible drive to restore the efficacy of anticancer drugs. Achieving this goal seemed inevitable in 1982 when a series of calcium channel blockers were demonstrated to restore the efficacy of chemotherapy agents. A large number of other compounds have since been demonstrated to restore chemotherapeutic sensitivity in cancer cells or tissues. Where do we stand almost three decades since the first reports of ABCB1 inhibition? Unfortunately, in the aftermath of extensive fundamental and clinical research efforts the situation remains gloomy. Only a small handful of compounds have reached late stage clinical trials and none are in routine clinical usage to circumvent chemoresistance. Why has the translation process been so ineffective? One factor is the multifactorial nature of drug resistance inherent to cancer tissues; ABCB1 is not the sole factor. However, expression of ABCB1 remains a significant negative prognostic indicator and is closely associated with poor response to chemotherapy in many cancer types. The main difficulties with restoration of sensitivity to chemotherapy reside with poor properties of the ABCB1 inhibitors: (1) low selectivity to ABCB1, (2) poor potency to inhibit ABCB1, (3) inherent toxicity and/or (4) adverse pharmacokinetic interactions with anticancer drugs. Despite these difficulties, there is a clear requirement for effective inhibitors and to date the strategies for generating such compounds have involved serendipity or simple chemical syntheses. This chapter outlines more sophisticated approaches making use of bioinformatics, combinatorial chemistry and structure informed drug design. Generating a new arsenal of potent and selective ABCB1 inhibitors offers the promise of restoring the efficacy of a key weapon in cancer treatment--chemotherapy.
Collapse
|
25
|
Govindarajan R, Leung GPH, Zhou M, Tse CM, Wang J, Unadkat JD. Facilitated mitochondrial import of antiviral and anticancer nucleoside drugs by human equilibrative nucleoside transporter-3. Am J Physiol Gastrointest Liver Physiol 2009; 296:G910-22. [PMID: 19164483 PMCID: PMC2670673 DOI: 10.1152/ajpgi.90672.2008] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human equilibrative nucleoside transporter-3 (hENT3) was recently reported as a pH-dependent, intracellular (lysosomal) transporter capable of transporting anti-human immunodeficiency virus (HIV) dideoxynucleosides (ddNs). Because most anti-HIV ddNs (e.g., zidovudine, AZT) exhibit clinical mitochondrial toxicity, we investigated whether hENT3 facilitates transport of anti-HIV ddNs into the mitochondria. Cellular fractionation and immunofluorescence microscopy studies in several human cell lines identified a substantial presence of hENT3 in the mitochondria, with additional presence at the cell surface of two placental cell lines (JAR, JEG3). Mitochondrial or cell surface hENT3 expression was confirmed in human hepatocytes and placental tissues, respectively. Unlike endogenous hENT3, yellow fluorescent protein (YFP)-tagged hENT3 was partially directed to the lysosomes. Xenopus oocytes expressing NH2-terminal-deleted hENT3 (expressed at the cell surface) showed pH-dependent interaction with several classes of nucleosides (anti-HIV ddNs, gemcitabine, fialuridine, ribavirin) that produce mitochondrial toxicity. Transport studies in hENT3 gene-silenced JAR cells showed significant reduction in mitochondrial transport of nucleosides and nucleoside drugs. Our data suggest that cellular localization of hENT3 is cell type dependent and the native transporter is substantially expressed in mitochondria and/or cell surface. hENT3-mediated mitochondrial transport may play an important role in mediating clinically observed mitochondrial toxicity of nucleoside drugs. In addition, our finding that hENT3 is a mitochondrial transporter is consistent with the recent finding that mutations in the hENT3 gene cause an autosomal recessive disorder in humans called the H syndrome.
Collapse
Affiliation(s)
- Rajgopal Govindarajan
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - George P. H. Leung
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mingyan Zhou
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chung-Ming Tse
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington; and Division of Gastroenterology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Varatharajan L, Thomas SA. The transport of anti-HIV drugs across blood-CNS interfaces: summary of current knowledge and recommendations for further research. Antiviral Res 2009; 82:A99-109. [PMID: 19176219 PMCID: PMC2678986 DOI: 10.1016/j.antiviral.2008.12.013] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 12/14/2008] [Accepted: 12/21/2008] [Indexed: 02/07/2023]
Abstract
The advent of highly active antiretroviral therapy (HAART), which constitutes HIV protease inhibitors, nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors and nucleotide reverse transcriptase inhibitors, has dramatically reduced the morbidity and mortality associated with human immunodeficiency virus (HIV) infection in resource-rich countries. However, this disease still kills several million people each year. Though the reason for therapeutic failure is multi-factorial, an important concern is the treatment and control of HIV within the central nervous system (CNS). Due to the restricted entry of anti-HIV drugs, the brain is thought to form a viral sanctuary site. This not only results in virological resistance, but also is often associated with the development of complications such as HIV-associated dementia. The CNS delivery of anti-HIV drugs is limited by the blood–brain and blood–CSF interfaces due to a combination of restricted paracellular movement, powerful metabolic enzymes and numerous transporters including members of the ATP binding cassette (ABC) and solute carrier (SLC) superfamilies. A better appreciation of the transporters present at the brain barriers will prove a valuable milestone in understanding the limited brain penetration of anti-HIV drugs in HIV and also aid the development of new anti-HIV drugs and drug combinations, with enhanced efficacy in the CNS. This review aims to summarise current knowledge on the transport of anti-HIV drugs across the blood–brain barrier and the choroid plexus, as well as provide recommendations for future research.
Collapse
|
27
|
Endres CJ, Moss AM, Ke B, Govindarajan R, Choi DS, Messing RO, Unadkat JD. The role of the equilibrative nucleoside transporter 1 (ENT1) in transport and metabolism of ribavirin by human and wild-type or Ent1-/- mouse erythrocytes. J Pharmacol Exp Ther 2009; 329:387-98. [PMID: 19164463 DOI: 10.1124/jpet.108.145854] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The polar nucleoside drug ribavirin is front-line treatment for chronic hepatitis C virus infection. The human equilibrative nucleoside transporter (ENT) 1 transports ribavirin into erythrocytes where it is phosphorylated. These phosphorylated metabolites accumulate in the erythrocytes and produce dose-limiting hemolytic anemia. Here, we examined the in vitro and ex vivo transport and metabolism of ribavirin by erythrocytes isolated from humans and Ent1-null mice. Ribavirin (2.4 microM) uptake was significantly higher (1044 +/- 255 amol/microg/10 s) into erythrocytes from Ent1(+/+) mice compared with that from Ent1(-/-) mice (76.48 +/- 11.20 amol/microg/10 s). Our results showed a saturable (K(m) of 382 +/- 75.1 microM) transport of [(3)H]ribavirin into erythrocytes from Ent1(+/+) mice. We found that ribavirin concentration rapidly (within 60 s) reached equilibrium in erythrocytes using a time course of [(3)H]ribavirin transport (2.5 microM) and metabolism in mouse and human erythrocytes for 8 h. However, total radioactivity of ribavirin was predominantly attributed to the phosphorylated metabolites ribavirin monophosphate and ribavirin triphosphate. Our findings allow us to estimate ribavirin transport, diffusion, and metabolic clearance and to predict in vivo accumulation of ribavirin phosphates in erythrocytes of both mice and humans. Our modeling of ribavirin in erythrocytes on long-term administration of ribavirin suggests that the accumulation of ribavirin inside the cells is dependent on ENT1/Ent1 transport and the rates of intracellular phosphorylation and the degradation of the phosphorylated metabolites. We predict that Ent1(+/+) and Ent1(-/-) mice will serve as excellent models to investigate the contribution of Ent1 to the pharmacokinetics and toxicity of ribavirin in vivo.
Collapse
|
28
|
Pastor-Anglada M, Cano-Soldado P, Errasti-Murugarren E, Casado FJ. SLC28 genes and concentrative nucleoside transporter (CNT) proteins. Xenobiotica 2008; 38:972-94. [PMID: 18668436 DOI: 10.1080/00498250802069096] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The human concentrative nucleoside transporter (hCNT) protein family has three members, hCNT1, 2, and 3, encoded by SLC28A1, A2, and A3 genes, respectively. hCNT1 and hCNT2 translocate pyrimidine- and purine-nucleosides, respectively, by a sodium-dependent mechanism, whereas hCNT3 shows broad substrate selectivity and the unique ability of translocating nucleosides both in a sodium- and a proton-coupled manner. hCNT proteins are also responsible for the uptake of most nucleoside-derived antiviral and anticancer drugs. Thus, hCNTs are key pharmacological targets. This review focuses on several crucial aspects of hCNT biology and pharmacology: protein structure-function, structural determinants for transportability, pharmacogenetics of hCNT-encoding genes, role of hCNT proteins in nucleoside-based therapeutics, and finally hCNT physiology.
Collapse
Affiliation(s)
- M Pastor-Anglada
- Facultat de Biologia, Departament de Bioquimica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain.
| | | | | | | |
Collapse
|
29
|
Carter NS, Yates P, Arendt CS, Boitz JM, Ullman B. Purine and pyrimidine metabolism in Leishmania. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 625:141-54. [PMID: 18365665 DOI: 10.1007/978-0-387-77570-8_12] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Purines and pyrimidines are indispensable to all life, performing many vital functions for cells: ATP serves as the universal currency of cellular energy, cAMP and cGMP are key second messenger molecules, purine and pyrimidine nucleotides are precursors for activated forms of both carbohydrates and lipids, nucleotide derivatives of vitamins are essential cofactors in metabolic processes, and nucleoside triphosphates are the immediate precursors for DNA and RNA synthesis. Unlike their mammalian and insect hosts, Leishmania lack the metabolic machinery to make purine nucleotides de novo and must rely on their host for preformed purines. The obligatory nature of purine salvage offers, therefore, a plethora of potential targets for drug targeting, and the pathway has consequently been the focus of considerable scientific investigation. In contrast, Leishmania are prototrophic for pyrimidines and also express a small complement of pyrimidine salvage enzymes. Because the pyrimidine nucleotide biosynthetic pathways of Leishmania and humans are similar, pyrimidine metabolism in Leishmania has generally been considered less amenable to therapeutic manipulation than the purine salvage pathway. However, evidence garnered from a variety of parasitic protozoa suggests that the selective inhibition of pyrimidine biosynthetic enzymes offers a rational therapeutic paradigm. In this chapter, we present an overview of the purine and pyrimidine pathways in Leishmania, make comparisons to the equivalent pathways in their mammalian host, and explore how these pathways might be amenable to selective therapeutic targeting.
Collapse
Affiliation(s)
- Nicola S Carter
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, Oregon 97239-3098, USA
| | | | | | | | | |
Collapse
|
30
|
Govindarajan R, Endres CJ, Whittington D, LeCluyse E, Pastor-Anglada M, Tse CM, Unadkat JD. Expression and hepatobiliary transport characteristics of the concentrative and equilibrative nucleoside transporters in sandwich-cultured human hepatocytes. Am J Physiol Gastrointest Liver Physiol 2008; 295:G570-80. [PMID: 18635603 PMCID: PMC2536788 DOI: 10.1152/ajpgi.00542.2007] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We previously reported that both the concentrative (hCNT) and equilibrative (hENT) nucleoside transporters are expressed in the human liver (21). Here we report a study that investigated the expression of these transporters (transcripts and proteins) and their role in the hepatobiliary transport of nucleosides/nucleoside drugs using sandwich-cultured human hepatocytes. In the hepatic tissue, the rank order of the mRNA expression of the transporters was hCNT1 approximately hENT1>hENT2 approximately hCNT2>hCNT3. In sandwich-cultured hepatocytes, the mRNA expression of hCNT2 and hENT2 was comparable to that in hepatic tissue, whereas the expression of corresponding transporters in the two-dimensional hepatocyte cultures was lower. Colocalization studies demonstrated predominant localization of these transporters at the sinusoidal membrane and of hENT1, hCNT1, and hCNT2 at the canalicular membrane. In the sandwich-cultured hepatocytes, ENTs were the major contributors to the transport of thymidine (hENT1, 63%; hENT2, 23%) or guanosine (hENT1, 53%; hENT2, 24%) into the hepatocytes followed by hCNT1 (10%) for thymidine or hCNT2 (23%) for guanosine. Although ribavirin was predominately transported (89%) into the hepatocytes by hENT1, fialuridine (FIAU) was transported by both hENT1 (30%) and hCNTs (61%). The extensively metabolized natural nucleosides were not effluxed into the bile, whereas significant biliary-efflux was observed of FIAU (19%), ribavirin (30%), and formycin B (35%). We conclude that the hepatic activity of hENT1 and hCNT1/2 transporters will determine the in vivo hepatic distribution and therefore the efficacy and/or toxicity of nucleoside drugs used to treat hepatic diseases.
Collapse
Affiliation(s)
- Rajgopal Govindarajan
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher J. Endres
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dale Whittington
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward LeCluyse
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marçal Pastor-Anglada
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chung-Ming Tse
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jashvant D. Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington; CellzDirect, Pittsboro, North Carolina; Department of Biochemistry and Molecular Biology, Institute of Biomedicine (IBUB) University of Barcelona and Centro de Investigación Biomédica en Red en el Área temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain; and Department of Medicine, Division of Gastroenterology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Chishu T, Sai Y, Nishimura T, Sato K, Kose N, Nakashima E. Potential of various drugs to inhibit nucleoside uptake in rat syncytiotrophoblast cell line, TR-TBT 18d-1. Placenta 2008; 29:461-7. [PMID: 18329095 DOI: 10.1016/j.placenta.2008.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 01/17/2008] [Accepted: 01/22/2008] [Indexed: 10/22/2022]
Abstract
The placenta requires nucleosides as nutrients for fetal growth, so it is important to examine potential interactions between placental transports of nucleosides and drugs to ensure the safety of pharmacotherapy during pregnancy. The purposes of this study are to clarify the uptake mechanisms of nucleosides from the maternal side of the syncytiotrophoblast and to investigate the inhibitory effect of various drugs on nucleoside uptake, using the rat syncytiotrophoblast cell line TR-TBT 18d-1, which shows syncytial-like morphology and functional expression of several transporters. Initial uptake of [(3)H]uridine or [(3)H]adenosine from the apical side of TR-TBT 18d-1 was markedly reduced by an excess of the respective unlabelled compound, and was slightly reduced by replacement of Na(+) with N-methyl-d-glucamine, indicating that both uptakes were Na(+)-independent. [(3)H]Uridine and [(3)H]adenosine uptakes in the absence of Na(+) were significantly and concentration-dependently inhibited by both 0.1 microM and 100 microM nitrobenzylthioinosine, suggesting the involvement of equilibrative nucleoside transporters (ENTs, SLC29). Kinetic analysis of adenosine uptake yielded a K(m) value of approximately 17 microM. These results are consistent with the reported uptake characteristics of uridine and adenosine by ENT1 and ENT2. The uptakes were significantly reduced by high concentrations of several nucleoside drugs, including cytarabine, vidarabine, zidovudine, mizoribine, caffeine and amitriptyline, but the effects were small within the therapeutic concentration ranges. In summary, our results suggest that ENTs are involved in apical uptake of uridine and adenosine in the syncytiotrophoblast. However, therapeutic concentrations of the drugs tested in this study might have little influence on maternal-to-fetal nucleoside transfer.
Collapse
Affiliation(s)
- T Chishu
- Department of Pharmaceutics, Kyoritsu University of Pharmacy, 1-5-30 Shiba-koen, Minato-ku, Tokyo 105-8512, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Compensatory effects of the human nucleoside transporters on the response to nucleoside-derived drugs in breast cancer MCF7 cells. Biochem Pharmacol 2008; 75:639-48. [DOI: 10.1016/j.bcp.2007.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 10/02/2007] [Accepted: 10/05/2007] [Indexed: 11/24/2022]
|
33
|
Zhu Z, Buolamwini JK. Constrained NBMPR analogue synthesis, pharmacophore mapping and 3D-QSAR modeling of equilibrative nucleoside transporter 1 (ENT1) inhibitory activity. Bioorg Med Chem 2008; 16:3848-65. [PMID: 18289860 DOI: 10.1016/j.bmc.2008.01.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 01/20/2008] [Accepted: 01/23/2008] [Indexed: 10/22/2022]
Abstract
Conformationally constrained analogue synthesis was undertaken to aid in pharmacophore mapping and 3D-QSAR analysis of nitrobenzylmercaptopurine riboside (NBMPR) congeners as equilibriative nucleoside transporter 1 (ENT1) inhibitors. In our previous study [J. Med. Chem. 2003, 46, 831-837], novel regioisomeric nitro-1,2,3,4-tetrahydroisoquinoline conformationally constrained analogues of NBMPR were synthesized and evaluated as ENT1 ligands. 7-NO(2)-1,2,3,4-Tetrahydroisoquino-2-yl purine riboside was identified as the analogue with the nitro group in the best orientation at the NBMPR binding site of ENT1. In the present study, further conformational constraining was introduced by synthesizing 5'-O,8-cyclo derivatives. The flow cytometrically determined binding affinities indicated that the additional 5'-O,8-cyclo constraining was unfavorable for binding to the ENT1 transporter. The structure-activity relationship (SAR) acquired was applied to pharmacophore mapping using the PHASE program. The best pharmacophore hypothesis obtained embodied an anti-conformation with three hydrogen-bond acceptors, one hydrophobic center, and two aromatic rings involving the 3'-OH, 4'-oxygen, the NO(2) group, the benzyl phenyl and the imidazole and pyrimidine portions of the purine ring, respectively. A PHASE 3D-QSAR model derived with this pharmacophore yielded an r(2) of 0.916 for four (4) PLS components, and an excellent external test set predictive r(2) of 0.78 for 39 compounds. This pharmacophore was used for molecular alignment in a comparative molecular field analysis (CoMFA) 3D-QSAR study that also afforded a predictive model with external test set validation predictive r(2) of 0.73. Thus, although limited, this study suggests that the bioactive conformation for NBMPR at the ENT1 transporter could be anti. The study has also suggested an ENT1 inhibitory pharmacophore, and established a predictive CoMFA 3D-QSAR model that might be useful for novel ENT1 inhibitor discovery and optimization.
Collapse
Affiliation(s)
- Zhengxiang Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, 847 Monroe Avenue, Suite 327, Memphis, TN 38163, USA
| | | |
Collapse
|
34
|
Leung GPH, Tse CM. The role of mitochondrial and plasma membrane nucleoside transporters in drug toxicity. Expert Opin Drug Metab Toxicol 2008; 3:705-18. [PMID: 17916056 DOI: 10.1517/17425255.3.5.705] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many anticancer and antiviral drugs are nucleoside analogues, which interfere with nucleotide metabolism and DNA replication to produce pharmacological effects. Clinical efficacy and toxicity of nucleoside drugs are closely associated with nucleoside transporters because they mediate the transport of nucleoside drugs across biological membranes. Two families of human nucleoside transporters (equilibrative nucleoside transporters and concentrative nucleoside transporters) have been extensively studied for several decades. They are widely distributed, from the plasma membrane to membranes of organelles such as mitochondria, and the distribution differs in different tissues. In addition, they have different specificities to nucleoside drugs. The characteristics of equilibrative and concentrative nucleoside transporters affect the therapeutic outcomes achieved with anticancer and antiviral nucleoside drugs. In this review, an overview of the role of mitochondrial and plasma membrane nucleoside transporters in nucleoside drug toxicity is provided. Rational design and therapeutic application of nucleoside analogues are also discussed.
Collapse
Affiliation(s)
- George P H Leung
- The University of Hong Kong, Department of Pharmacology, Hong Kong.
| | | |
Collapse
|
35
|
Minuesa G, Purcet S, Erkizia I, Molina-Arcas M, Bofill M, Izquierdo-Useros N, Casado FJ, Clotet B, Pastor-Anglada M, Martinez-Picado J. Expression and Functionality of Anti-Human Immunodeficiency Virus and Anticancer Drug Uptake Transporters in Immune Cells. J Pharmacol Exp Ther 2007; 324:558-67. [DOI: 10.1124/jpet.107.131482] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
36
|
Zhang J, Visser F, King KM, Baldwin SA, Young JD, Cass CE. The role of nucleoside transporters in cancer chemotherapy with nucleoside drugs. Cancer Metastasis Rev 2007; 26:85-110. [PMID: 17345146 DOI: 10.1007/s10555-007-9044-4] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nucleoside analogs are important components of treatment regimens for various malignancies. Nucleoside-specific membrane transporters mediate plasma membrane permeation of physiologic nucleosides and most nucleoside analogs, for which the initial event is cellular conversion of nucleosides to active agents. Understanding of the roles of nucleoside transporters in nucleoside drug toxicity and resistance will provide opportunities for potentiating anticancer efficacy and avoiding resistance. Because transportability is a possible determinant of toxicity and resistance of many nucleoside analogs, nucleoside transporter abundance might be a prognostic marker to assess drug resistance. Elucidation of the structural determinants of nucleoside analogs for interaction with transporter proteins as well as the structural features of transporter proteins required for permeant interaction and translocation will lead to "transportability guidelines" for the rational design and therapeutic application of nucleoside analogs as anticancer drugs. It should eventually be possible to develop clinical assays that predict sensitivity and/or resistance to nucleoside anti-cancer drugs and thus to identify those patient populations that will most likely benefit from optimal nucleoside analog treatments. This review discusses recent results from structure/function studies of human nucleoside transporters, the role of nucleoside transport processes in the cytotoxicity and resistance of several anticancer nucleoside analogs and strategies to improve the nucleoside transporter-related anticancer effects of nucleoside analogs.
Collapse
Affiliation(s)
- Jing Zhang
- Membrane Protein Research Group, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | |
Collapse
|
37
|
Kaler G, Truong DM, Khandelwal A, Nagle M, Eraly SA, Swaan PW, Nigam SK. Structural variation governs substrate specificity for organic anion transporter (OAT) homologs. Potential remote sensing by OAT family members. J Biol Chem 2007; 282:23841-53. [PMID: 17553798 PMCID: PMC3812435 DOI: 10.1074/jbc.m703467200] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Organic anion transporters (OATs, SLC22) interact with a remarkably diverse array of endogenous and exogenous organic anions. However, little is known about the structural features that determine their substrate selectivity. We examined the substrate binding preferences and transport function of olfactory organic anion transporter, Oat6, in comparison with the more broadly expressed transporter, Oat1 (first identified as NKT). In analyzing interactions of both transporters with over 40 structurally diverse organic anions, we find a correlation between organic anion potency (pKi) and hydrophobicity (logP) suggesting a hydrophobicity-driven association with transporter-binding sites, which appears particularly prominent for Oat6. On the other hand, organic anion binding selectivity between Oat6 and Oat1 is influenced by the anion mass and net charge. Smaller mono-anions manifest greater potency for Oat6 and di-anions for Oat1. Comparative molecular field analysis confirms these mechanistic insights and provides a model for predicting new OAT substrates. By comparative molecular field analysis, both hydrophobic and charged interactions contribute to Oat1 binding, although it is predominantly the former that contributes to Oat6 binding. Together, the data suggest that, although the three-dimensional structures of these two transporters may be very similar, the binding pockets exhibit crucial differences. Furthermore, for six radiolabeled substrates, we assessed transport efficacy (Vmax) for Oat6 and Oat1. Binding potency and transport efficacy had little correlation, suggesting that different molecular interactions are involved in substrate binding to the transporter and translocation across the membrane. Substrate specificity for a particular transporter may enable design of drugs for targeting to specific tissues (e.g. olfactory mucosa). We also discuss how these data suggest a possible mechanism for remote sensing between OATs in different tissue compartments (e.g. kidney, olfactory mucosa) via organic anions.
Collapse
Affiliation(s)
- Gregory Kaler
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - David M. Truong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Akash Khandelwal
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201
| | - Megha Nagle
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Satish A. Eraly
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter W. Swaan
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201
| | - Sanjay K. Nigam
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
38
|
Hosseini SH, Kohler JJ, Haase CP, Tioleco N, Stuart T, Keebaugh E, Ludaway T, Russ R, Green E, Long R, Wang L, Eriksson S, Lewis W. Targeted transgenic overexpression of mitochondrial thymidine kinase (TK2) alters mitochondrial DNA (mtDNA) and mitochondrial polypeptide abundance: transgenic TK2, mtDNA, and antiretrovirals. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:865-74. [PMID: 17322372 PMCID: PMC1864875 DOI: 10.2353/ajpath.2007.060655] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mitochondrial toxicity limits nucleoside reverse transcriptase inhibitors (NRTIs) for acquired immune deficiency syndrome. NRTI triphosphates, the active moieties, inhibit human immunodeficiency virus reverse transcriptase and eukaryotic mitochondrial DNA polymerase pol-gamma. NRTI phosphorylation seems to correlate with mitochondrial toxicity, but experimental evidence is lacking. Transgenic mice (TGs) with cardiac overexpression of thymidine kinase isoforms (mitochondrial TK2 and cytoplasmic TK1) were used to study NRTI mitochondrial toxicity. Echocardiography and nuclear magnetic resonance imaging defined cardiac performance and structure. TK gene copy and enzyme activity, mitochondrial (mt) DNA and polypeptide abundance, succinate dehydrogenase and cytochrome oxidase histochemistry, and electron microscopy correlated with transgenesis, mitochondrial structure, and biogenesis. Antiretroviral combinations simulated therapy. Untreated hTK1 or TK2 TGs exhibited normal left ventricle mass. In TK2 TGs, cardiac TK2 gene copy doubled, activity increased 300-fold, and mtDNA abundance doubled. Abundance of the 17-kd subunit of complex I, succinate dehydrogenase histochemical activity, and cristae density increased. NRTIs increased left ventricle mass 20% in TK2 TGs. TK activity increased 3 logs in hTK1 TGs, but no cardiac phenotype resulted. NRTIs abrogated functional effects of transgenically increased TK2 activity but had no effect on TK2 mtDNA abundance. Thus, NRTI mitochondrial phosphorylation by TK2 is integral to clinical NRTI mitochondrial toxicity.
Collapse
Affiliation(s)
- Seyed H Hosseini
- Department of Pathology, Emory University School of Medicine, 7117 Woodruff Memorial Building, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Owen RP, Badagnani I, Giacomini KM. Molecular Determinants of Specificity for Synthetic Nucleoside Analogs in the Concentrative Nucleoside Transporter, CNT2. J Biol Chem 2006; 281:26675-82. [PMID: 16840788 DOI: 10.1074/jbc.m513421200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the concentrative nucleoside transporter (CNT) family (SLC28) mediate the transport of naturally-occurring nucleosides, and nucleoside analog drugs across the plasma membrane of epithelial cells. Each of the three CNT family members has a distinct specificity for naturally occurring nucleosides, and residues that contribute to the specificity of each transporter have been identified. In contrast, the molecular determinants of specificity for synthetic nucleoside analogs are not known. In this study, we take advantage of the large species difference that exists between human and rat CNT2 (hCNT2 and rCNT2) in their ability to transport the nucleoside analog drug cladribine, 2CdA, (rCNT2 > > > hCNT2) to identify the critical domains and amino acid residues that contribute to the observed difference in specificity between CNT2 orthologs. Using chimeric proteins of human and rat CNT2, we determined that the C-terminal half of CNT2 contained the determinants of 2CdA selectivity. We replaced key residues in the C terminus of hCNT2 with the equivalent residue in rCNT2. One residue in the C-terminal portion of CNT2 was found to significantly contribute to 2CdA selectivity: hCNT2-S354A. This mutant caused an increase of 5-6-fold over hCNT2. The 2-chloro pharmacophore, rather than the 2'-deoxyribose was responsible for the reduced 2CdA uptake by hCNT2. Our data are consistent with a model in which an increased capability for hydrogen bonding in critical amino acids that reside in the C terminus of rCNT2 contributes to its enhanced selectivity for 2CdA.
Collapse
Affiliation(s)
- Ryan P Owen
- Department of Biopharmaceutical Sciences, The University of California, San Francisco, San Francisco, California 94158, USA
| | | | | |
Collapse
|
40
|
Balakrishnan A, Polli JE. Apical sodium dependent bile acid transporter (ASBT, SLC10A2): a potential prodrug target. Mol Pharm 2006; 3:223-30. [PMID: 16749855 PMCID: PMC2796132 DOI: 10.1021/mp060022d] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A major hurdle impeding the successful clinical development of drug candidates can be poor intestinal permeability. Low intestinal permeability may be enhanced by a prodrug approach targeting membrane transporters in the small intestine. Transporter specificity, affinity, and capacity are three factors in targeted prodrug design. The human apical sodium dependent bile acid transporter (SLC10A2) belongs to the solute carrier family (SLC) of transporters and is an important carrier protein expressed in the small intestine. In spite of its appearing to be an excellent target for prodrug design, few studies have targeted human apical sodium dependent bile acid transporter (hASBT) to improve oral bioavailability. This review discusses bile acids including their chemistry and their absorptive disposition. Additionally, hASBT-mediated prodrug targeting is discussed, including QSAR, in vitro models for hASBT assay, and the current progress in utilizing hASBT as a drug delivery target.
Collapse
Affiliation(s)
- Anand Balakrishnan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201
| | - James E. Polli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201
- Author to whom, correspondence should be addressed, James E. Polli, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, HSF2, room 623, Baltimore, MD 21201, Telephone: 410-706-8292, Fax : 410-706-5017,
| |
Collapse
|
41
|
Purcet S, Minuesa G, Molina-Arcas M, Erkizia I, Casado FJ, Clotet B, Martínez-Picado J, Pastor-Anglada M. 3′-Azido-2′,3′-Dideoxythymidine (Zidovudine) Uptake Mechanisms in T Lymphocytes. Antivir Ther 2006. [DOI: 10.1177/135965350601100603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The nucleoside reverse transcriptase inhibitors (NRTIs) make up a family of antiretroviral drugs widely used in the treatment of HIV-1 infection. Human concentrative nucleoside transporters and equilibrative nucleoside transporters, encoded by SLC28 and SLC29 gene families, respectively, are known to be involved in the uptake of a variety of nucleoside analogues used in anticancer treatment. We therefore examined whether SLC28- and SLC29-encoded proteins contribute to the entry of these NRTIs into the human leukaemic T-cell line Molt-4. Cis-inhibition experiments demonstrated that nucleoside transporters have a negligible role in antiviral drug uptake. Moreover, the previously identified 3′-azido-2′,3′-dideoxythymidine (zidovudine; AZT) carriers, organic anion transporters (organic anion transporter [hOATs], members of the SLC22 gene family) have not been detected in T cells, either functionally or at the mRNA level, thus ruling out a role for hOATs in antiviral drug uptake in these cells. Nevertheless, the data provided here argue against the hypothesis of simple diffusion across the plasma membrane as the unique mechanism of AZT uptake. Actually, this pyrimidine derivative seems to have a temperature-sensitive route of entrance, a finding that, along with the evidence that, AZT inhibits its own uptake and its transport into phytohaemagglutinin-stimulated peripheral blood mononuclear cells is upregulated, strongly support the idea that AZT uptake into T-cells is associated with a mediated and regulated, transport mechanism.
Collapse
Affiliation(s)
- Sergi Purcet
- Regulació dels Sistemes de Transport (RST), Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Gerard Minuesa
- IrsiCaixa Foundation, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Míriam Molina-Arcas
- Regulació dels Sistemes de Transport (RST), Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Itziar Erkizia
- IrsiCaixa Foundation, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - F Javier Casado
- Regulació dels Sistemes de Transport (RST), Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Bonaventura Clotet
- IrsiCaixa Foundation, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Javier Martínez-Picado
- IrsiCaixa Foundation, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Marçal Pastor-Anglada
- Regulació dels Sistemes de Transport (RST), Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Shin HC, Kim JS, Vig BS, Song X, Drach JC, Amidon GL. Interaction of intestinal nucleoside transporter hCNT2 with amino acid ester prodrugs of floxuridine and 2-bromo-5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole. Biol Pharm Bull 2006; 29:247-52. [PMID: 16462026 DOI: 10.1248/bpb.29.247] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amino acid ester prodrugs of antiviral and anticancer nucleoside drugs were developed to improve oral bioavailability or to reduce systemic toxicity. We studied the interaction of human concentrative nucleoside transporter (hCNT2) cloned from intestine with various amino acid ester prodrugs of floxuridine (FUdR) and 5,6-dichloro-2-bromo-1-beta-D-ribofuranosylbenzimidazole (BDCRB). Na(+)-dependent uptakes of [(3)H]-inosine and [(3)H]-adenosine were measured in U251 cells transiently expressing intestinal hCNT2. FUdR significantly inhibited the uptake of both [(3)H]-inosine and [(3)H]-adenosine (60-70% of control), while its amino acid ester prodrugs including Val, Phe, Pro, Asp, and Lys esters exhibited markedly decreased inhibition potency (10-30% of control). On the other hand, BDCRB and its amino acid prodrugs markedly inhibited the uptake of both [(3)H]-inosine and [(3)H]-adenosine. Val, Phe, and Pro ester prodrugs of BDCRB showed similar inhibition capacities as parent compound BDCRB (80-90% for adenosine and 60-80% for inosine). The amino acid site of attachment (3'- and 5'-monoesters) and stereochemistry (L- and D-amino acid esters), did not significantly affect the uptake of [(3)H]-inosine and [(3)H]-adenosine. These results demonstrate that the hCNT2 favorably interacts with BDCRB and its amino acid prodrugs, compared to those of FUdR, and that neutral amino acid esters of BDCRB have a high affinity toward this transporter. Therefore, the intestinal hCNT2 may be a target transporter as a factor for modulating oral pharmacokinetics of BDCRB prodrugs.
Collapse
Affiliation(s)
- Ho-Chul Shin
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Transporter proteins facilitate the transfer of solutes across the cell membrane and have an intricate role in drug absorption, distribution and excretion. Because of their substrate promiscuity, several transporters represent viable pharmacological targets for enhancing drug absorption, preventing drug toxicity or facilitating localized tissue delivery. However, the slow emergence of high-resolution structures for these proteins has hampered the intelligent design of transporter substrates. Nonetheless, currently available functional, as well as structural, data provide an attractive scaffold for generating fusion models that merge substrate-based SARs and protein-based homology structures. The resultant models offer features that extend single modality paradigms in predictive function.
Collapse
Affiliation(s)
- Cheng Chang
- Biophysics Program, Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
44
|
Hu H, Endres CJ, Chang C, Umapathy NS, Lee EW, Fei YJ, Itagaki S, Swaan PW, Ganapathy V, Unadkat JD. Electrophysiological Characterization and Modeling of the Structure Activity Relationship of the Human Concentrative Nucleoside Transporter 3 (hCNT3). Mol Pharmacol 2006; 69:1542-53. [PMID: 16446384 DOI: 10.1124/mol.105.018945] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We characterized the electrophysiology, kinetics, and quantitative structure-activity relationship (QSAR) of the human concentrative nucleoside transporter 3 (hCNT3) expressed in Xenopus laevis oocytes by measuring substrate-induced inward currents using a two-microelectrode voltage-clamp system. At membrane potentials between -30 and -150 mV, sodium activation of gemcitabine transport was sigmoidal, with a K0.5 of 8.5+/-0.3 mM for Na+ and a Hill coefficient of 2.2+/-0.25 independent of membrane potential. We measured the Imax and K0.5 for substrate at -50 mV for the nucleoside analog drugs gemcitabine (638+/-58 nA, 59.7+/-17.5 microM), ribavirin (546+/-37 nA, 61.0+/-13.2 microM), AZT (420+/-4 nA, 310+/-9 microM), and 3-deazauridine (506+/-30 nA, 50.8+/-9.90 microM). K0.5 and Imax for substrate were dependent on membrane potential (both increasing as the membrane became more hyperpolarized) for all four drugs. hCNT3 also exhibited pre-steady-state currents. The quantitative structure-activity relationship (QSAR) was examined using comparative molecular field analysis and comparative molecular similarity indices analysis of the inward currents induced by 27 nucleoside analogs with substitutions at both the ribose and the nucleobase. Two statistically significant QSAR models identified electrostatic interaction as the major force in hCNT3 transport and attributed a critical role to the 3'-hydroxyl position of hCNT3 substrates. Steric hindrance at the 3-position and positive charge at the 5-position of the pyrimidine ring were favorable for transport. Two hCNT3 pharmacophore models revealed the minimal features required for hCNT3 transport as two hydrogen bond acceptors at 3'-OH and 5'-O and the hydrophobic center occupied by the base ring.
Collapse
Affiliation(s)
- Huankai Hu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chang C, Swaan PW. Computational approaches to modeling drug transporters. Eur J Pharm Sci 2005; 27:411-24. [PMID: 16274971 DOI: 10.1016/j.ejps.2005.09.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 09/27/2005] [Indexed: 11/15/2022]
Abstract
Computational modeling has advanced our understanding of drug absorption, tissue distribution, excretion and toxicity profiles by providing both direct and indirect knowledge of drug-transporter interactions that would otherwise be unavailable using experimental methods. Currently, two complementary approaches are available in modeling transporters: substrate-based and transporter-based methods. The transporter-based approach directly predicts the transporter's three-dimensional structure to assist in understanding the drug transport process, whereas substrate-based models infer such information by studying a group of substrates or inhibitors with measured activities. In this review, the available strategies in both transporter-based and substrate-based approaches are explained and illustrated with applications and case studies. With increasing computational power and continuously improving modeling algorithms, computational techniques can assist in further understanding transporter-substrate interactions as well as, the optimization of transporter-directed drug design.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201, USA
| | | |
Collapse
|
46
|
Zhang J, Smith KM, Tackaberry T, Visser F, Robins MJ, Nielsen LPC, Nowak I, Karpinski E, Baldwin SA, Young JD, Cass CE. Uridine binding and transportability determinants of human concentrative nucleoside transporters. Mol Pharmacol 2005; 68:830-9. [PMID: 15955867 DOI: 10.1124/mol.105.012187] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human concentrative nucleoside transporters 1, 2, and 3 (hCNT1, hCNT2, and hCNT3) exhibit different functional characteristics, and a better understanding of their permeant selectivities is critical for development of nucleoside analog drugs with optimal pharmacokinetic properties. In this study, the sensitivity of a high-throughput yeast expression system used previously for hCNT1 and hCNT3 was improved and used to characterize determinants for interaction of uridine (Urd) with hCNT2. The observed changes of binding energy between hCNT2 and different Urd analogs suggested that it interacts with C3'-OH, C5'-OH, and N3-H of Urd. The C2' and C5 regions of Urd played minor but significant roles for Urd-hCNT2 binding, possibly through Van der Waals interactions. Because the yeast assay only provided information about potential transportability, the permeant selectivities of recombinant hCNT1, hCNT2, and hCNT3 produced in Xenopus laevis oocytes were investigated using a two-electrode voltage clamp assay. hCNT1-mediated transport was sensitive to modifications of the N3, C3', and C5' positions of Urd. hCNT2 showed some tolerance for transporting Urd analogs with C2' or C5 modifications, little tolerance for N3 modifications, and no tolerance for any modifications at C3' or C5' of Urd. Although hCNT3 was sensitive to C3' modifications, it transported a broad range of variously substituted Urd analogs. The transportability profiles identified in this study, which reflected the binding profiles well, should prove useful in the development of anticancer and antiviral therapies with nucleoside drugs that are permeants of members of the hCNT protein family.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11540 University Ave., Edmonton, Alberta, Canada T6H 1Z2
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Efforts to improve oral drug bioavailability have grown in parallel with the pharmaceutical industry. As the number and chemical diversity of drugs has increased, new strategies have been required to develop orally active therapeutics. The past two decades have been characterised by an increased understanding of the causes of low bioavailability and a great deal of innovation in oral drug delivery technologies, marked by an unprecedented growth of the drug delivery industry. The advent of biotechnology and consequent proliferation of biopharmaceuticals have brought new challenges to the drug delivery field. In spite of the difficulties associated with developing oral forms of this type of therapeutics, significant progress has been made in the past few years, with some oral proteins, peptides and other macromolecules currently advancing through clinical trials. This article reviews the approaches that have been successfully applied to improve oral drug bioavailability, primarily, prodrug strategies, lead optimisation through medicinal chemistry and formulation design. Specific strategies to improve the oral bioavailability of biopharmaceuticals are also discussed.
Collapse
|