1
|
Bhati FK, Bhat MK. An anti-neoplastic tale of metformin through its transport. Life Sci 2024; 357:123060. [PMID: 39278619 DOI: 10.1016/j.lfs.2024.123060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Metformin is an attractive candidate drug among all the repurposed drugs for cancer. Extensive preclinical and clinical research has evaluated its efficacy in cancer therapy, revealing a mixed outcome in clinical settings. To fully exploit metformin's therapeutic potential, understanding cellular factors relevant to its transport and accumulation in cancer cells needs to be understood. This review highlights the relevance of metformin transporter status towards its anti-cancer potential. Metformin transporters are regulated at pre-transcriptional, transcriptional, and post-translational levels. Moreover, the tumour microenvironment can also influence metformin accumulation in cancer cells. Also, Metformin treatment can regulate its transporters by altering global DNA methylation, protein acetylation, and transcription factors. Importantly, metformin transporters not only influence chemotherapeutic drug toxicity but are also associated with the prognosis and survival of individuals having cancer. Strategic decisions based on the expression and regulation of metformin transporters holds promise for its therapeutic implications and relevance.
Collapse
Affiliation(s)
- Firoz Khan Bhati
- Biotechnology Research and Innovation Council - National Centre for Cell Science (BRIC- NCCS), Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411 007, India
| | - Manoj Kumar Bhat
- Biotechnology Research and Innovation Council - National Centre for Cell Science (BRIC- NCCS), Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411 007, India.
| |
Collapse
|
2
|
Nojima I, Wada J. Metformin and Its Immune-Mediated Effects in Various Diseases. Int J Mol Sci 2023; 24:ijms24010755. [PMID: 36614197 PMCID: PMC9821749 DOI: 10.3390/ijms24010755] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Metformin has been a long-standing prescribed drug for treatment of type 2 diabetes (T2D) and its beneficial effects on virus infection, autoimmune diseases, aging and cancers are also recognized. Metformin modulates the differentiation and activation of various immune-mediated cells such as CD4+ and CD+8 T cells. The activation of adenosine 5'-monophosphate-activated protein kinase (AMPK) and mammalian target of rapamycin complex 1 (mTORC1) pathway may be involved in this process. Recent studies using Extracellular Flux Analyzer demonstrated that metformin alters the activities of glycolysis, oxidative phosphorylation (OXPHOS), lipid oxidation, and glutaminolysis, which tightly link to the modulation of cytokine production in CD4+ and CD+8 T cells in various disease states, such as virus infection, autoimmune diseases, aging and cancers.
Collapse
Affiliation(s)
| | - Jun Wada
- Correspondence: ; Tel.: +81-86-235-7232; FAX: +81-86-222-5214
| |
Collapse
|
3
|
Angenoorth TJF, Maier J, Stankovic S, Bhat S, Sucic S, Freissmuth M, Sitte HH, Yang JW. Rescue of Misfolded Organic Cation Transporter 3 Variants. Cells 2022; 12:39. [PMID: 36611832 PMCID: PMC9818475 DOI: 10.3390/cells12010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Organic cation transporters (OCTs) are membrane proteins that take up monoamines, cationic drugs and xenobiotics. We previously reported novel missense mutations of organic cation transporter 3 (OCT3, SLC22A3), some with drastically impacted transport capabilities compared to wildtype. For some variants, this was due to ER retention and subsequent degradation of the misfolded transporter. For other transporter families, it was previously shown that treatment of misfolded variants with pharmacological and chemical chaperones could restore transport function to a certain degree. To investigate two potentially ER-bound, misfolded variants (D340G and R348W), we employed confocal and biochemical analyses. In addition, radiotracer uptake assays were conducted to assess whether pre-treatment with chaperones could restore transporter function. We show that pre-treatment of cells with the chemical chaperone 4-PBA (4-phenyl butyric acid) leads to increased membrane expression of misfolded variants and is associated with increased transport capacity of D340G (8-fold) and R348W (1.5 times) compared to untreated variants. We herein present proof of principle that folding-deficient SLC22 transporter variants, in particular those of OCT3, are amenable to rescue by chaperones. These findings need to be extended to other SLC22 members with corroborated disease associations.
Collapse
Affiliation(s)
- Thomas J. F. Angenoorth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Julian Maier
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Stevan Stankovic
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Shreyas Bhat
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
- Department of Physics, Université de Montréal, 1375 Avenue Thérèse-Lavoie-Roux, Montréal, QC H3T 1J4, Canada
- Department of Pharmacology and Physiology, Université de Montréal, 2960 Chemin de la Tour, Montréal, QC H3T 1J4, Canada
| | - Sonja Sucic
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Harald H. Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090 Vienna, Austria
| |
Collapse
|
4
|
Gomes Candido Reis D, Levy D, Lage LADPC, Culler HF, Rocha V, Bydlowski SP, Nogueira Zerbini MC, Pereira J. New genetic prognostic biomarkers in primary central nervous system lymphoma (PCNSL). Brain Behav 2021; 11:e02061. [PMID: 33591648 PMCID: PMC8035458 DOI: 10.1002/brb3.2061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/16/2020] [Accepted: 01/17/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND PCNSL is a rare extranodal NHL with poor prognosis. Tumorigenesis has been associated with hyperactivation of BCR downstream and NFkB pathways. We studied the prognosis of the relative expression profile of target genes of NFkB pathway (MYC, BCL2), the essential transcriptional regulator in hematopoiesis LMO2, the checkpoint regulation pathway MGMT, the transcription factor POU2F1, the immune checkpoint gene PDCD1, and the proto-oncogene and transcriptional repressor gene BCL6 and its proteins in PCNSL. METHODS This study is a retrospective cohort study; 35 immunocompetent PCNSL-DLBCL patients had their gene expression (RT-qPCR) normalized to internal control gene GUSB. RESULTS Median patient age was 62 years, median OS was 42.6 months (95% CI: 26.6-58.6), PFS was 41 months (95% CI: 19.7-62.4), and DFS was 59.2 months (95% CI 31.9-86.6). A moderate correlation was found between the gene/protein expressions of MYC (kappa = 0.596, p = .022) and of BCL2 (kappa = 0.426, p = .042). Relative gene expression of MYC ≥ 0.201 (HR 6.117; p = .003) was associated with worse 5-year OS. Relative gene expression of MYC ≥ 0.201 (HR 3.96; p = .016) and MGMT ≥ 0.335 (HR 3.749; p = .056) was associated with worse PFS. Age > 60 years and IELSG score moderate/high were also associated with worse prognosis. CONCLUSIONS Overexpression of MYC and overexpression of MGMT were prognostic markers associated with unfavorable clinical outcomes in PCNSL.
Collapse
Affiliation(s)
- Diego Gomes Candido Reis
- Department of Hematology, Hemotherapy and Cell Therapy, Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Débora Levy
- Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Luís Alberto de Pádua Covas Lage
- Department of Hematology, Hemotherapy and Cell Therapy, Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil.,Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| | - Hebert Fabrício Culler
- Department of Hematology, Hemotherapy and Cell Therapy, Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil
| | - Vanderson Rocha
- Department of Hematology, Hemotherapy and Cell Therapy, Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil.,Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil.,Fundação Pró-Sangue Hemocentro de São Paulo, São Paulo, Brazil.,Churchill Hospital, Oxford University, Oxford, UK
| | - Sérgio Paulo Bydlowski
- Laboratory of Immunology (LIM19), Heart Institute (InCor), Faculdade de Medicina, Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Juliana Pereira
- Department of Hematology, Hemotherapy and Cell Therapy, Laboratory of Medical Investigation in Pathogenesis and Directed Therapy in Onco-Immuno-Hematology (LIM-31), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, SP, Brazil.,Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
| |
Collapse
|
5
|
Zhou S, Zeng S, Shu Y. Drug-Drug Interactions at Organic Cation Transporter 1. Front Pharmacol 2021; 12:628705. [PMID: 33679412 PMCID: PMC7925875 DOI: 10.3389/fphar.2021.628705] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
The interaction between drugs and various transporters is one of the decisive factors that affect the pharmacokinetics and pharmacodynamics of drugs. The organic cation transporter 1 (OCT1) is a member of the Solute Carrier 22A (SLC22A) family that plays a vital role in the membrane transport of organic cations including endogenous substances and xenobiotics. This article mainly discusses the drug-drug interactions (DDIs) mediated by OCT1 and their clinical significance.
Collapse
Affiliation(s)
- Shiwei Zhou
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, MD, United States.,Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Hunan, China
| | - Sujuan Zeng
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China
| | - Yan Shu
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, MD, United States
| |
Collapse
|
6
|
Sutherland R, Meeson A, Lowes S. Solute transporters and malignancy: establishing the role of uptake transporters in breast cancer and breast cancer metastasis. Cancer Metastasis Rev 2021; 39:919-932. [PMID: 32388639 PMCID: PMC7497311 DOI: 10.1007/s10555-020-09879-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The solute carrier (SLC) superfamily encompasses a large variety of membrane-bound transporters required to transport a diverse array of substrates over biological membranes. Physiologically, they are essential for nutrient uptake, ion transport and waste removal. However, accumulating evidence suggest that up- and/or downregulation of SLCs may play a pivotal role in the pathogenesis of human malignancy. Endogenous substrates of SLCs include oestrogen and its conjugates, the handling of which may be of importance in hormone-dependent cancers. The SLCs play a significant role in the handling of therapeutic agents including anticancer drugs. Differential SLC expression in cancers may, therefore, impact on the efficacy of treatments. However, there is also a small body of evidence to suggest the dysregulated expression of some of these transporters may be linked to cancer metastasis. This review draws on the current knowledge of the roles of SLC transporters in human cancers in order to highlight the potential significance of these solute carriers in breast cancer pathogenesis and treatment. Graphical abstract ![]()
Collapse
Affiliation(s)
- Rachel Sutherland
- Biosciences Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle Upon Tyne, UK. .,Translational and Clinical Research Institute, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK.
| | - Annette Meeson
- Biosciences Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle Upon Tyne, UK
| | - Simon Lowes
- Translational and Clinical Research Institute, Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK.,Breast Screening and Assessment Unit, Queen Elizabeth Hospital, Gateshead Health NHS Foundation Trust, Gateshead, Sheriff Hill, UK
| |
Collapse
|
7
|
Substrates and Inhibitors of Organic Cation Transporters (OCTs) and Plasma Membrane Monoamine Transporter (PMAT) and Therapeutic Implications. Handb Exp Pharmacol 2021; 266:119-167. [PMID: 34495395 DOI: 10.1007/164_2021_516] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gene products of the SLC22A gene family (hOCT1, hOCT2, and hOCT3) and of the SLC29A4 gene (hPMAT or hENT4) are all polyspecific organic cation transporters. Human OCTs (including hPMAT) are expressed in peripheral tissues such as small intestine, liver, and kidney involved in the pharmacokinetics of drugs. In the human brain, all four transporters are expressed at the blood-brain barrier (BBB), hOCT2 is additionally expressed in neurons, and hOCT3 and hPMAT in glia. More than 40% of the presently used drugs are organic cations. This chapter lists and discusses all known drugs acting as substrates or inhibitors of these four organic cation transporters, independently of whether the transporter is expressed in the central nervous system (CNS) or in peripheral tissues. Of interest is their involvement in drug absorption, distribution, and excretion as well as potential OCT-associated drug-drug interactions (DDIs), with a focus on drugs that act in the CNS.
Collapse
|
8
|
Jinakote M, Ontawong A, Soodvilai S, Pimta J, Pasachan T, Chatsudthipong V, Srimaroeng C. High affinity of 4-(4-(dimethylamino)styryl)-N-methylpyridinium transport for assessing organic cation drugs in hepatocellular carcinoma cells. Fundam Clin Pharmacol 2020; 34:365-379. [PMID: 31883148 DOI: 10.1111/fcp.12531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Human organic cation transporter 1 (hOCT1) and human organic cation transporter 3 (hOCT3) are highly expressed in hepatocytes and play important roles in cationic drug absorption, distribution, and elimination. A previous study demonstrated that downregulation of hOCT1 and hOCT3 mRNA was related to hepatocellular carcinoma (HepG2) prognosis and severity. Whether these transporters expressed in HepG2 cells serve for cationic drug delivery has not been investigated. Besides radioactive transport, options for assessing hOCTs in hepatocytes are limited. This study clarified the significant roles of hOCTs in HepG2 by comparing cationic fluorescent 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+ ) with traditional [3 H]-1-methyl-4-phenylpyridinium (MPP+ ). The results showed ASP+ was preferably transported into HepG2 compared to [3 H]-MPP+ with high affinity and a high maximal transport rate. Selective transport of ASP+ mediated by hOCTs was influenced by extracellular pH, temperature, and membrane depolarization, corresponding to hOCT1 and hOCT3 expressions. Furthermore, transport of cationic drugs, metformin, and paclitaxel in HepG2 cells was blunted by OCT inhibitors, suggesting that hOCT1 and hOCT3 expressed in HepG2 cells exhibit notable impacts on cationic drug actions. The fluorescent ASP+ -based in vitro model may also provide a rapid and powerful analytical tool for further screening of cationic drug actions and interactions with hOCTs, particularly hOCT1 and hOCT3 in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Metee Jinakote
- Faculty of Oriental Medicine, Chiang Rai College, Chiang Rai, 57000, Thailand.,Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Atcharaporn Ontawong
- Unit of Excellence of Coffee, Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, 56000, Thailand
| | - Sunhapas Soodvilai
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Jeerawat Pimta
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Tipthida Pasachan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Varanuj Chatsudthipong
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
9
|
Abstract
The organic cation transporters (OCTs) OCT1, OCT2, OCT3, novel OCT (OCTN)1, OCTN2, multidrug and toxin exclusion (MATE)1, and MATE kidney-specific 2 are polyspecific transporters exhibiting broadly overlapping substrate selectivities. They transport organic cations, zwitterions, and some uncharged compounds and operate as facilitated diffusion systems and/or antiporters. OCTs are critically involved in intestinal absorption, hepatic uptake, and renal excretion of hydrophilic drugs. They modulate the distribution of endogenous compounds such as thiamine, L-carnitine, and neurotransmitters. Sites of expression and functions of OCTs have important impact on energy metabolism, pharmacokinetics, and toxicity of drugs, and on drug-drug interactions. In this work, an overview about the human OCTs is presented. Functional properties of human OCTs, including identified substrates and inhibitors of the individual transporters, are described. Sites of expression are compiled, and data on regulation of OCTs are presented. In addition, genetic variations of OCTs are listed, and data on their impact on transport, drug treatment, and diseases are reported. Moreover, recent data are summarized that indicate complex drug-drug interaction at OCTs, such as allosteric high-affinity inhibition of transport and substrate dependence of inhibitor efficacies. A hypothesis about the molecular mechanism of polyspecific substrate recognition by OCTs is presented that is based on functional studies and mutagenesis experiments in OCT1 and OCT2. This hypothesis provides a framework to imagine how observed complex drug-drug interactions at OCTs arise. Finally, preclinical in vitro tests that are performed by pharmaceutical companies to identify interaction of novel drugs with OCTs are discussed. Optimized experimental procedures are proposed that allow a gapless detection of inhibitory and transported drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Brosseau N, Ramotar D. The human organic cation transporter OCT1 and its role as a target for drug responses. Drug Metab Rev 2019; 51:389-407. [PMID: 31564168 DOI: 10.1080/03602532.2019.1670204] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The human organic cation uptake transporter OCT1, encoded by the SLC22A1 gene, is highly expressed in the liver and reported to possess a broad substrate specificity. OCT1 operates by facilitated diffusion and allows the entry of nutrients into cells. Recent findings revealed that OCT1 can mediate the uptake of drugs for treating various diseases such as cancers. The levels of OCT1 expression correlate with the responses towards many drugs and functionally defective OCT1 lead to drug resistance. It has been recently proposed that OCT1 should be amongst the crucial drug targets used for pharmacogenomic analyses. Several single nucleotide polymorphisms exist and are distributed across the entire OCT1 gene. While there are differences in the OCT1 gene polymorphisms between populations, there are at least five variants that warrant consideration in any genetic screen. To date, and despite two decades of research into OCT1 functional role, it still remains uncertain what are the define substrates for this uptake transporter, although studies from mice revealed that one of the substrates is vitamin B1. It is also unclear how OCT1 recognizes a broad array of ligands and whether this involves specific modifications and interactions with other proteins. In this review, we highlight the current findings related to OCT1 with the aim of propelling further studies on this key uptake transporter.
Collapse
Affiliation(s)
- Nicolas Brosseau
- Department of Medicine, Maisonneuve-Rosemont Hospital, Research Center, Université de Montréal, Montréal, Québec, Canada
| | - Dindial Ramotar
- Department of Medicine, Maisonneuve-Rosemont Hospital, Research Center, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
11
|
Li J, Yang Z, Tuo B. Role of OCT1 in hepatocellular carcinoma. Onco Targets Ther 2019; 12:6013-6022. [PMID: 31413596 PMCID: PMC6662865 DOI: 10.2147/ott.s212088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/10/2019] [Indexed: 01/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most commonly diagnosed cancers causing death worldwide. It is difficult to detect at an early stage and most patients with advanced HCC rarely achieve satisfying therapeutic results. Accordingly, researchers have been trying to find new biomarkers for diagnosis and new methods of treatment. OCT1, a member of solute carrier super family, is highly expressed in normal liver tissues, and predominantly transports endogenous and exogenous substances, such as metabolites, drugs and toxins to hepatocytes. Studies have demonstrated that the expression of OCT1 is related to the progression and survival of HCC patients. Furthermore, sorafenib, which is regarded as the only effective molecular targeting drug for advanced HCC, is affected by OCT1 variants. In the current review, we summarized the reports about OCT1 and HCC in order to present a comprehensive overview of the relationship between OCT1 and HCC.
Collapse
Affiliation(s)
- Jingguo Li
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi, People's Republic of China
| | - Zhengyi Yang
- Department of Gastroenterology, Bijie First People's Hospital, Bijie, Guizhou Province, People's Republic of China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
12
|
Mechanisms of Anticancer Drug Resistance in Hepatoblastoma. Cancers (Basel) 2019; 11:cancers11030407. [PMID: 30909445 PMCID: PMC6468761 DOI: 10.3390/cancers11030407] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/25/2022] Open
Abstract
The most frequent liver tumor in children is hepatoblastoma (HB), which derives from embryonic parenchymal liver cells or hepatoblasts. Hepatocellular carcinoma (HCC), which rarely affects young people, causes one fourth of deaths due to cancer in adults. In contrast, HB usually has better prognosis, but this is still poor in 20% of cases. Although more responsive to chemotherapy than HCC, the failure of pharmacological treatment used before and/or after surgical resection is an important limitation in the management of patients with HB. To advance in the implementation of personalized medicine it is important to select the best combination among available anti-HB drugs, such as platinum derivatives, anthracyclines, etoposide, tyrosine-kinase inhibitors, Vinca alkaloids, 5-fluorouracil, monoclonal antibodies, irinotecan and nitrogen mustards. This requires predicting the sensitivity to these drugs of each tumor at each time because, it should be kept in mind, that cancer chemoresistance is a dynamic process of Darwinian nature. For this goal it is necessary to improve our understanding of the mechanisms of chemoresistance involved in the refractoriness of HB against the pharmacological challenge and how they evolve during treatment. In this review we have summarized the current knowledge on the multifactorial and complex factors responsible for the lack of response of HB to chemotherapy.
Collapse
|
13
|
Al-Abdulla R, Perez-Silva L, Abete L, Romero MR, Briz O, Marin JJG. Unraveling ‘The Cancer Genome Atlas’ information on the role of SLC transporters in anticancer drug uptake. Expert Rev Clin Pharmacol 2019; 12:329-341. [DOI: 10.1080/17512433.2019.1581605] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ruba Al-Abdulla
- Experimental Hepatology and Drug Targeting (HEVEFARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Laura Perez-Silva
- Experimental Hepatology and Drug Targeting (HEVEFARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Lorena Abete
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marta R. Romero
- Experimental Hepatology and Drug Targeting (HEVEFARM), University of Salamanca, IBSAL, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEFARM), University of Salamanca, IBSAL, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Jose J. G. Marin
- Experimental Hepatology and Drug Targeting (HEVEFARM), University of Salamanca, IBSAL, Salamanca, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| |
Collapse
|
14
|
Kalimuthu K, Lubin BC, Bazylevich A, Gellerman G, Shpilberg O, Luboshits G, Firer MA. Gold nanoparticles stabilize peptide-drug-conjugates for sustained targeted drug delivery to cancer cells. J Nanobiotechnology 2018; 16:34. [PMID: 29602308 PMCID: PMC5877385 DOI: 10.1186/s12951-018-0362-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/21/2018] [Indexed: 11/12/2022] Open
Abstract
Background Peptide-drug-conjugates (PDCs) are being developed as an effective strategy to specifically deliver cytotoxic drugs to cancer cells. However one of the challenges to their successful application is the relatively low stability of peptides in the blood, liver and kidneys. Since AuNPs seem to have a longer plasma half-life than PDCs, one approach to overcoming this problem would be to conjugate the PDCs to gold nanoparticles (AuNPs), as these have demonstrated favorable physico-chemical and safety properties for drug delivery systems. We set out to test whether PEG coated-AuNPs could provide a suitable platform for the non-covalent loading of pre-formed PDCs and whether this modification would affect the bioavailability of the PDCs and their cytotoxicity toward target cancer cells. Methods Peptides specifically internalized by A20 murine lymphoma cells were isolated from a phage library displaying 7mer linear peptides. Peptide specificity was validated by flow cytometry and confocal microscopy. PDCs were synthesized containing a selected peptide (P4) and either chlorambucil (Chlor), melphalan (Melph) or bendamustine (Bend). Gold nanoparticles were sequentially coated with citrate, PEG-6000 and then PDC (PDC-PEG-AuNP). The physico-chemical properties of the coated particles were analyzed by electrophoresis, TEM, UV–VIS and FTIR. Stability of free and PDC-coated AuNP was determined. Results Biopanning of the phage library resulted in discovery of several novel peptides that internalized into A20 cells. One of these (P4) was used to synthesize PDCs containing either Chlor, Melph or Bend. All three PDCs specifically killed A20 target cells, however they had short half-lives ranging from 10.6 to 15.4 min. When coated to PEG-AuNPs, the half-lives were extended to 21.0–22.3 h. The PDC-PEG-AuNPs retained cytotoxicity towards the target cells. Moreover, whereas pre-incubation for 24 h of free PDCs almost completely abolished their cytotoxic activity, the PDC-PEG-AuNPs were still active even after 72 h pre-incubation. Conclusions Peptide-drug-conjugates hold potential for improving the target efficacy of chemotherapeutic drugs, however their short half-lives may limit their application. This hurdle can be overcome by easily conjugating them to gold nanoparticles. This conjugation also opens up the possibility of developing slow release formulations of targeted drug delivery systems containing PDCs. Electronic supplementary material The online version of this article (10.1186/s12951-018-0362-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Bat-Chen Lubin
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel.,Eastern R&D Center, Ariel, 40700, Israel
| | | | - Gary Gellerman
- Dept. Chemical Sciences, Ariel University, 40700, Ariel, Israel
| | - Ofer Shpilberg
- Tel Aviv & Pre-Med Faculty, Assuta Medical Center, Institute of Hematology, Ariel University, 40700, Ariel, Israel
| | - Galia Luboshits
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel
| | - Michael A Firer
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel. .,Ariel Center for Applied Cancer Research, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
15
|
Hucke A, Ciarimboli G. The Role of Transporters in the Toxicity of Chemotherapeutic Drugs: Focus on Transporters for Organic Cations. J Clin Pharmacol 2017; 56 Suppl 7:S157-72. [PMID: 27385173 DOI: 10.1002/jcph.706] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/11/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022]
Abstract
The introduction of chemotherapy in the treatment of cancer is one of the most important achievements of modern medicine, even allowing the cure of some lethal diseases such as testicular cancer and other malignant neoplasms. The number and type of chemotherapeutic agents available have steadily increased and have developed until the introduction of targeted tumor therapy. It is now evident that transporters play an important role for determining toxicity of chemotherapeutic drugs not only against target but also against nontarget cells. This is of special importance for intracellularly active hydrophilic drugs, which cannot freely penetrate the plasma membrane. Because many important chemotherapeutic agents are substrates of transporters for organic cations, this review discusses the known interaction of these substances with these transporters. A particular focus is given to the role of transporters for organic cations in the development of side effects of chemotherapy with platinum derivatives and in the efficacy of recently developed tyrosine kinase inhibitors to specifically target cancer cells. It is evident that specific inhibition of uptake transporters may be a possible strategy to protect against undesired side effects of platinum derivatives without compromising their antitumor efficacy. These transporters are also important for efficient targeting of tyrosine kinase inhibitors to cancer cells. However, in order to achieve the aims of protecting from undesired toxicities and improving the specificity of uptake by tumor cells, an exact knowledge of transporter expression, function, regulation under normal and pathologic conditions, and of genetically and epigenetically regulation is mandatory.
Collapse
Affiliation(s)
- Anna Hucke
- Experimental Nephrology, Medical Clinic D, Münster University Hospital, Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Medical Clinic D, Münster University Hospital, Münster, Germany
| |
Collapse
|
16
|
Ataei A, Solovyeva VV, Poorebrahim M, Blatt NL, Salafutdinov II, Şahin F, Kiyasov AP, Yalvaç ME, Rizvanov AA. A Genome-Wide Analysis of mRNA Expression in Human Tooth Germ Stem Cells Treated with Pluronic P85. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0254-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
17
|
Zaïr ZM, Singer DR. Influx transporter variants as predictors of cancer chemotherapy-induced toxicity: systematic review and meta-analysis. Pharmacogenomics 2016; 17:1189-1205. [PMID: 27380948 DOI: 10.2217/pgs-2015-0005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM Chemotherapeutic agents have been shown to increase lung patient survival, however their use may be limited by their serious adverse effects. We aimed to assess int impact of pharmacogenetic variation of influx transporters on inter-individual patient variation in adverse drug reactions. PATIENTS & METHODS We conducted a meta-analysis and systemic review and identified 16 publications, totaling 1510 patients, to be eligible for review. RESULTS Meta-analysis showed east-Asian patients expressing SLCO1B1 521T>C or 1118G>A to have a two- to fourfold increased risk of irinotecan-induced neutropenia but not diarrhea. American patients, expressing SLC19A1 IVS2(4935) G>A, were further associated with pemetrexed/gemcitabine-induced grade 3+ leukopenia. CONCLUSION Future studies should look to robust validation of SLCO1B1 and SLC19A1 as prognostic markers in the management of lung cancer patients.
Collapse
Affiliation(s)
| | - Donald Rj Singer
- Yale University School of Medicine, New Haven, CT, USA.,Fellowship of Postgraduate Medicine 11 Chandos Street, London, UK
| |
Collapse
|
18
|
Hyrsova L, Smutny T, Trejtnar F, Pavek P. Expression of organic cation transporter 1 (OCT1): unique patterns of indirect regulation by nuclear receptors and hepatospecific gene regulation. Drug Metab Rev 2016; 48:139-58. [DOI: 10.1080/03602532.2016.1188936] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lucie Hyrsova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czech Republic
| | - Tomas Smutny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czech Republic
| | - Frantisek Trejtnar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czech Republic
| | - Petr Pavek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czech Republic
| |
Collapse
|
19
|
The human organic cation transporter OCT1 mediates high affinity uptake of the anticancer drug daunorubicin. Sci Rep 2016; 6:20508. [PMID: 26861753 PMCID: PMC4748219 DOI: 10.1038/srep20508] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023] Open
Abstract
Anthracyclines such as daunorubicin are anticancer agents that are transported into cells, and exert cytotoxicity by blocking DNA metabolism. Although there is evidence for active uptake of anthracyclines into cells, the specific transporter involved in this process has not been identified. Using the high-grade serous ovarian cancer cell line TOV2223G, we show that OCT1 mediated the high affinity (Km ~ 5 μM) uptake of daunorubicin into the cells, and that micromolar amounts of choline completely abolished the drug entry. OCT1 downregulation by shRNA impaired daunorubicin uptake into the TOV2223G cells, and these cells were significantly more resistant to the drug in comparison to the control shRNA. Transfection of HEK293T cells, which accommodated the ectopic expression of OCT1, with a plasmid expressing OCT1-EYFP showed that the transporter was predominantly localized to the plasma membrane. These transfected cells exhibited an increase in the uptake of daunorubicin in comparison to control cells transfected with an empty EYFP vector. Furthermore, a variant of OCT1, OCT1-D474C-EYFP, failed to enhance daunorubicin uptake. This is the first report demonstrating that human OCT1 is involved in the high affinity transport of anthracyclines. We postulate that OCT1 defects may contribute to the resistance of cancer cells treated with anthracyclines.
Collapse
|
20
|
Le Roy B, Tixier L, Pereira B, Sauvanet P, Buc E, Pétorin C, Déchelotte P, Pezet D, Balayssac D. Assessment of the Relation between the Expression of Oxaliplatin Transporters in Colorectal Cancer and Response to FOLFOX-4 Adjuvant Chemotherapy: A Case Control Study. PLoS One 2016; 11:e0148739. [PMID: 26859833 PMCID: PMC4747515 DOI: 10.1371/journal.pone.0148739] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/22/2016] [Indexed: 12/13/2022] Open
Abstract
Background Adjuvant chemotherapy for colorectal cancer is mainly based on the combination of 5-fluorouracil, folinic acid and oxaliplatin (FOLFOX-4). The pharmacological target of oxaliplatin remains intracellular and therefore dependent on its entry into cells. The intracellular distribution of oxaliplatin is mediated by organic cation transporters 1, 2 and 3 (OCT1, 2 and 3), copper transporter 1 (CTR1) and ATPase Cu2+ transporting beta polypeptide (ATP7B) and may modulate the efficacy of oxaliplatin-based chemotherapy. The aim of this study was to perform a retrospective study to assess the relation between the expression of oxaliplatin transporters in colorectal cancer before chemotherapy and the response to FOLFOX-4 adjuvant chemotherapy in responder and non-responder patients. Methods This retrospective study was conducted at a single center (University Hospital of Clermont-Ferrand, France). The target population was patients with resectable colorectal cancer operated between 2006 and 2013. Inclusion criteria were defined for the responder patients as no cancer recurrence 3 years after the end of chemotherapy, and for the non-responder patients as cancer recurrence within 1 year. Other inclusion criteria were stages IIb–IV cancers, first-line adjuvant FOLFOX-4 chemotherapy, and the availability of resected primary tumor samples. Exclusion criteria were preoperative chemotherapy and/or radiotherapy, a targeted therapy, other anticancer drugs, cancer recurrence between the first and the third year after the end of chemotherapy and follow-up < 3 years. Immunostaining of oxaliplatin transporters (OCT1, 2, 3, CTR1 and ATP7B) and Ki-67 was assessed in tumor samples. Results Retrospectively, 31 patients have been selected according to inclusion and exclusion criteria (15 responders and 16 non-responders). Before FOLFOX-4 regimen, OCT3 expression was significantly lower in responder patients compared to non-responders (p<0.001). According to multivariate analysis, OCT3 remains an independent criterion for adjuvant FOLFOX chemotherapy response (p = 0.039). No significant relation is reported between chemotherapy response and the expression of OCT1 (p = 0.49), OCT2 (p = 0.09), CTR1 (p = 0.45), ATP7B (p = 0.94) and Ki-67 (p = 0.34) in tumors. Conclusions High expression of OCT3 could be an independent factor related to resistance to FOLFOX-4 chemotherapy.
Collapse
Affiliation(s)
- Bertrand Le Roy
- CHU Clermont-Ferrand, Service de chirurgie et oncologie digestive, F-63003, Clermont-Ferrand, France
| | - Lucie Tixier
- CHU Clermont-Ferrand, Service d’anatomopathologie, F-63003, Clermont-Ferrand, France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Délégation à la Recherche Clinique et à l’Innovation, F-63003, Clermont-Ferrand, France
| | - Pierre Sauvanet
- CHU Clermont-Ferrand, Service de chirurgie et oncologie digestive, F-63003, Clermont-Ferrand, France
- INSERM/UdA U1071, USC INRA 2018, M2iSH, F-63001, Clermont-Ferrand, France
| | - Emmanuel Buc
- CHU Clermont-Ferrand, Service de chirurgie et oncologie digestive, F-63003, Clermont-Ferrand, France
- INSERM/UdA U1071, USC INRA 2018, M2iSH, F-63001, Clermont-Ferrand, France
| | - Caroline Pétorin
- CHU Clermont-Ferrand, Service de chirurgie et oncologie digestive, F-63003, Clermont-Ferrand, France
| | - Pierre Déchelotte
- CHU Clermont-Ferrand, Service d’anatomopathologie, F-63003, Clermont-Ferrand, France
- Université d’Auvergne, R2D2 – EA 7281, F-63001, Clermont-Ferrand, France
| | - Denis Pezet
- CHU Clermont-Ferrand, Service de chirurgie et oncologie digestive, F-63003, Clermont-Ferrand, France
- INSERM/UdA U1071, USC INRA 2018, M2iSH, F-63001, Clermont-Ferrand, France
| | - David Balayssac
- CHU Clermont-Ferrand, Délégation à la Recherche Clinique et à l’Innovation, F-63003, Clermont-Ferrand, France
- INSERM U1107, Neuro-Dol, F-63001, Clermont-Ferrand, France
- * E-mail:
| |
Collapse
|
21
|
Matthaei J, Kuron D, Faltraco F, Knoch T, Dos Santos Pereira JN, Abu Abed M, Prukop T, Brockmöller J, Tzvetkov MV. OCT1 mediates hepatic uptake of sumatriptan and loss-of-function OCT1 polymorphisms affect sumatriptan pharmacokinetics. Clin Pharmacol Ther 2016; 99:633-41. [PMID: 26659468 DOI: 10.1002/cpt.317] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/02/2015] [Indexed: 01/14/2023]
Abstract
The low bioavailability of the anti-migraine drug sumatriptan is partially caused by first-pass hepatic metabolism. In this study, we analyzed the impact of the hepatic organic cation transporter OCT1 on sumatriptan cellular uptake, and of OCT1 polymorphisms on sumatriptan pharmacokinetics. OCT1 transported sumatriptan with high capacity and sumatriptan uptake into human hepatocytes was strongly inhibited by the OCT1 inhibitor MPP(+) . Sumatriptan uptake was not affected by the Met420del polymorphism, but was strongly reduced by Arg61Cys and Gly401Ser, and completely abolished by Gly465Arg and Cys88Arg. Plasma concentrations in humans with two deficient OCT1 alleles were 215% of those with fully active OCT1 (P = 0.0003). OCT1 also transported naratriptan, rizatriptan, and zolmitriptan, suggesting a possible impact of OCT1 polymorphisms on the pharmacokinetics of other triptans as well. In conclusion, OCT1 is a high-capacity transporter of sumatriptan and polymorphisms causing OCT1 deficiency have similar effects on sumatriptan pharmacokinetics as those observed in subjects with liver impairment.
Collapse
Affiliation(s)
- J Matthaei
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - D Kuron
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - F Faltraco
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - T Knoch
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - J N Dos Santos Pereira
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - M Abu Abed
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - T Prukop
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - J Brockmöller
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| | - M V Tzvetkov
- Institute for Clinical Pharmacology, University Medical Center, Georg-August University, Göttingen, Germany
| |
Collapse
|
22
|
Zheng J, Chan T, Zhu L, Yan X, Cao Z, Wang Y, Zhou F. The inhibitory effects of camptothecin (CPT) and its derivatives on the substrate uptakes mediated by human solute carrier transporters (SLCs). Xenobiotica 2016; 46:831-40. [DOI: 10.3109/00498254.2015.1129080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jian Zheng
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| | - Ting Chan
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| | - Ling Zhu
- Save Sight Institute, University of Sydney, Sydney, NSW, Australia, and
| | - Xiufeng Yan
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
| | - Zhisong Cao
- The CHRISTUS Stehlin Foundation for Cancer Research, Houston, TX, USA
| | - Yang Wang
- Alkali Soil Natural Environmental Science Center, Northeast Forestry University/Key Laboratory of Saline-alkali Vegetation Ecology Restoration in Oil Field, Ministry of Education, Harbin, P.R. China,
| | - Fanfan Zhou
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia,
| |
Collapse
|
23
|
Role of SLC22A1 polymorphic variants in drug disposition, therapeutic responses, and drug-drug interactions. THE PHARMACOGENOMICS JOURNAL 2015; 15:473-87. [PMID: 26526073 DOI: 10.1038/tpj.2015.78] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 08/20/2015] [Accepted: 09/08/2015] [Indexed: 02/08/2023]
Abstract
The SCL22A1 gene encodes the broad selectivity transporter hOCT1. hOCT1 is expressed in most epithelial barriers thereby contributing to drug pharmacokinetics. It is also expressed in different drug target cells, including immune system cells and others. Thus, this membrane protein might also contribute to drug pharmacodynamics. Up to 1000 hOCT1 polymorphisms have been identified so far, although only a small fraction of those have been mechanistically studied. A paradigm in the field of drug transporter pharmacogenetics is the impact of hOCT1 gene variability on metformin clinical parameters, affecting area under the concentration-time curve, Cmax and responsiveness. However, hOCT1 also mediates the translocation of a variety of drugs used as anticancer, antiviral, anti-inflammatory, antiemetic agents as well as drugs used in the treatment of neurological diseases among. This review focuses exclusively on those drugs for which some pharmacogenetic data are available, and aims at highlighting the need for further clinical research in this area.
Collapse
|
24
|
Muenzner JK, Rehm T, Biersack B, Casini A, de Graaf IAM, Worawutputtapong P, Noor A, Kempe R, Brabec V, Kasparkova J, Schobert R. Adjusting the DNA Interaction and Anticancer Activity of Pt(II) N-Heterocyclic Carbene Complexes by Steric Shielding of the Trans Leaving Group. J Med Chem 2015; 58:6283-92. [DOI: 10.1021/acs.jmedchem.5b00896] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
| | | | | | - Angela Casini
- Department
of Pharmacokinetics, Toxicology and Targeting, Research Institute
of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Inge A. M. de Graaf
- Department
of Pharmacokinetics, Toxicology and Targeting, Research Institute
of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pawida Worawutputtapong
- Department
of Pharmacokinetics, Toxicology and Targeting, Research Institute
of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | - Viktor Brabec
- Institute
of Biophysics, Academy of Sciences of the Czech Republic, CZ-61265 Brno, Czech Republic
| | - Jana Kasparkova
- Department
of Biophysics, Faculty of Science, Palacky University, 17. listopadu
12, CZ-77146 Olomouc, Czech Republic
| | | |
Collapse
|
25
|
Huber S, Huettner JP, Hacker K, Bernhardt G, König J, Buschauer A. Esters of Bendamustine Are by Far More Potent Cytotoxic Agents than the Parent Compound against Human Sarcoma and Carcinoma Cells. PLoS One 2015. [PMID: 26196503 PMCID: PMC4721923 DOI: 10.1371/journal.pone.0133743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The alkylating agent bendamustine is approved for the treatment of hematopoietic malignancies such as non-Hodgkin lymphoma, chronic lymphocytic leukemia and multiple myeloma. As preliminary data on recently disclosed bendamustine esters suggested increased cytotoxicity, we investigated representative derivatives in more detail. Especially basic esters, which are positively charged under physiological conditions, were in the crystal violet and the MTT assay up to approximately 100 times more effective than bendamustine, paralleled by a higher fraction of early apoptotic cancer cells and increased expression of p53. Analytical studies performed with bendamustine and representative esters revealed pronounced cellular accumulation of the derivatives compared to the parent compound. In particular, the pyrrolidinoethyl ester showed a high enrichment in tumor cells and inhibition of OCT1- and OCT3-mediated transport processes, suggesting organic cation transporters to be involved. However, this hypothesis was not supported by the differential expression of OCT1 (SLC22A1) and OCT3 (SLC22A3), comparing a panel of human cancer cells. Bendamustine esters proved to be considerably more potent cytotoxic agents than the parent compound against a broad panel of human cancer cell types, including hematologic and solid malignancies (e.g. malignant melanoma, colorectal carcinoma and lung cancer), which are resistant to bendamustine. Interestingly, spontaneously immortalized human keratinocytes, as a model of “normal” cells, were by far less sensitive than tumor cells against the most potent bendamustine esters.
Collapse
Affiliation(s)
- Stefan Huber
- Pharmaceutical and Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Johannes Philip Huettner
- Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Kristina Hacker
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Günther Bernhardt
- Pharmaceutical and Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Buschauer
- Pharmaceutical and Medicinal Chemistry II, Institute of Pharmacy, University of Regensburg, Regensburg, Germany
- * E-mail:
| |
Collapse
|
26
|
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: 1.8] [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
|
27
|
Arimany-Nardi C, Montraveta A, Lee-Vergés E, Puente XS, Koepsell H, Campo E, Colomer D, Pastor-Anglada M. Human organic cation transporter 1 (hOCT1) as a mediator of bendamustine uptake and cytotoxicity in chronic lymphocytic leukemia (CLL) cells. THE PHARMACOGENOMICS JOURNAL 2015; 15:363-71. [PMID: 25582574 DOI: 10.1038/tpj.2014.77] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/13/2014] [Accepted: 11/05/2014] [Indexed: 01/20/2023]
Abstract
Bendamustine is used in the treatment of chronic lymphocytic leukemia (CLL). Routes for bendamustine entry into target cells are unknown. This study aimed at identifying transporter proteins implicated in bendamustine uptake. Our results showed that hOCT1 is a bendamustine transporter, as bendamustine could cis-inhibit the uptake of a canonical hOCT1 substrate, with a Ki in the micromolar range, consistent with the EC50 values of the cytotoxicity triggered by this drug in HEK293 cells expressing hOCT1. hOCT1 polymorphic variants determining impaired bendamustine-transporter interaction, consistently reduced bendamustine cytotoxicity in HEK293 cells stably expressing them. Exome genotyping of the SLC22A1 gene, encoding hOCT1, was undertaken in a cohort of 241 CLL patients. Ex vivo cytotoxicity to bendamustine was measured in a subset of cases and shown to correlate with SLC22A1 polymorphic variants. In conclusion, hOCT1 is a suitable bendamustine transporter, thereby contributing to its cytotoxic effect depending upon the hOCT1 genetic variants expressed.
Collapse
Affiliation(s)
- C Arimany-Nardi
- 1] Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona, Spain [2] Oncology Program, National Biomedical Research Institute of Liver and Gastrointestinal Diseases (CIBER ehd), Instituto de Salud Carlos III, Madrid, Spain
| | - A Montraveta
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Lee-Vergés
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - X S Puente
- Institute of Oncology, University of Oviedo, Oviedo, Spain
| | - H Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Insitute, University of Würzburg, Würzburg, Germany
| | - E Campo
- Hematopathology Unit, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - D Colomer
- 1] Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain [2] Hematopathology Unit, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - M Pastor-Anglada
- 1] Department of Biochemistry and Molecular Biology, Institute of Biomedicine, University of Barcelona, Barcelona, Spain [2] Oncology Program, National Biomedical Research Institute of Liver and Gastrointestinal Diseases (CIBER ehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Copy number variability analysis of pharmacogenes in patients with lymphoma, leukemia, hepatocellular, and lung carcinoma using The Cancer Genome Atlas data. Pharmacogenet Genomics 2015; 25:1-7. [DOI: 10.1097/fpc.0000000000000097] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Hagos Y, Hundertmark P, Shnitsar V, Marada VVVR, Wulf G, Burckhardt G. Renal human organic anion transporter 3 increases the susceptibility of lymphoma cells to bendamustine uptake. Am J Physiol Renal Physiol 2014; 308:F330-8. [PMID: 25477469 DOI: 10.1152/ajprenal.00467.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic lymphatic leukemia (CLL) is often associated with nephritic syndrome. Effective treatment of CLL by chlorambucil and bendamustine leads to the restoration of renal function. In this contribution, we sought to elucidate the impact of organic anion transporters (OATs) on the uptake of bendamustine and chlorambucil as a probable reason for the superior efficacy of bendamustine over chlorambucil in the treatment of CLL. We examined the effects of structural analogs of p-aminohippurate (PAH), melphalan, chlorambucil, and bendamustine, on OAT1-mediated [(3)H]PAH uptake and OAT3- and OAT4-mediated [(3)H]estrone sulfate (ES) uptake in stably transfected human embryonic kidney-293 cells. Melphalan had no significant inhibitory effect on any OAT, whereas chlorambucil reduced OAT1-, OAT3-, and OAT4-mediated uptake of PAH or ES down to 14.6%, 16.3%, and 66.0% of control, respectively. Bendamustine inhibited only OAT3-mediated ES uptake, which was reduced down to 14.3% of control cells, suggesting that it interacts exclusively with OAT3. The IC50 value for OAT3 was calculated to be 0.8 μM. Real-time PCR experiments demonstrated a high expression of OAT3 in lymphoma cell lines as well as primary CLL cells. OAT3-mediated accumulation of bendamustine was associated with reduced cell proliferation and an increased rate of apoptosis. We conclude that the high efficacy of bendamustine in treating CLL might be partly contributed to the expression of OAT3 in lymphoma cells and the high affinity of bendamustine for this transporter.
Collapse
Affiliation(s)
- Yohannes Hagos
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Göttingen, Germany; and
| | - Philip Hundertmark
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Göttingen, Germany; and
| | - Volodymyr Shnitsar
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Göttingen, Germany; and
| | - Venkata V V R Marada
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Göttingen, Germany; and
| | - Gerald Wulf
- Klinik für Hämatologie und Onkologie, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Gerhard Burckhardt
- Institut für Vegetative Physiologie und Pathophysiologie, Universitätsmedizin Göttingen, Göttingen, Germany; and
| |
Collapse
|
30
|
Hagos Y, Wegner W, Kuehne A, Floerl S, Marada VV, Burckhardt G, Henjakovic M. HNF4α Induced Chemosensitivity to Oxaliplatin and 5-FU Mediated by OCT1 and CNT3 in Renal Cell Carcinoma. J Pharm Sci 2014; 103:3326-34. [DOI: 10.1002/jps.24128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/11/2014] [Accepted: 07/31/2014] [Indexed: 12/16/2022]
|
31
|
Li Q, Shu Y. Role of solute carriers in response to anticancer drugs. MOLECULAR AND CELLULAR THERAPIES 2014; 2:15. [PMID: 26056583 PMCID: PMC4452062 DOI: 10.1186/2052-8426-2-15] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/14/2014] [Indexed: 12/20/2022]
Abstract
Membrane transporters play critical roles in moving a variety of anticancer drugs across cancer cell membrane, thereby determining chemotherapy efficacy and/or toxicity. The retention of anticancer drugs in cancer cells is the result of net function of efflux and influx transporters. The ATP-binding cassette (ABC) transporters are mainly the efflux transporters expressing at cancer cells, conferring the chemo-resistance in various malignant tumors, which has been well documented over the past decades. However, the function of influx transporters, in particular the solute carriers (SLC) in cancer cells, has only been recently well recognized to have significant impact on cancer therapy. The SLC transporters not only directly bring anticancer agents into cancer cells but also serve as the uptake mediators of essential nutrients for tumor growth and survival. In this review, we concentrate on the interaction of SLC transporters with anticancer drugs and nutrients, and their impact on chemo-sensitivity or -resistance of cancer cells. The differential expression patterns of SLC transporters between normal and tumor tissues may be well utilized to achieve specific delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA ; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan 410078 China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA
| |
Collapse
|
32
|
Xiao S, Liao S, Zhou Y, Jiang B, Li Y, Xue M. High expression of octamer transcription factor 1 in cervical cancer. Oncol Lett 2014; 7:1889-1894. [PMID: 24932254 PMCID: PMC4049708 DOI: 10.3892/ol.2014.2023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 03/04/2014] [Indexed: 11/18/2022] Open
Abstract
Cervical carcinoma is the second most prevalent malignancy in females worldwide. The crucial etiologic factors involved in the development of cervical carcinoma include infection with papillomavirus, and the structural or functional mutation of oncogenes and tumor suppressor genes. The abnormal change of octamer transcription factor 1 (OCT1) is associated with tumor progression and a poor patient survival rate. However, little is known regarding the effect of OCT1 in cervical cancer. In the present study, flow cytometry, western blot analysis and quantitative polymerase chain reaction (qPCR) were peformed to identify differentially expressed OCT1 in cervical cancer tissue and adjacent non-cancerous tissues. The normalized OCT1 gene expression in cervical cancer was 5.98 times higher compared with the adjacent non-cancerous tissues. Western blot analysis and flow cytometry assessed the levels of OCT1 protein. The results of these two differential techniques showed that the protein expression level of OCT1 was greater in cervical cancer tissues, which corresponded with the qPCR results. Finally, as OCT1 is a potential target gene for microRNA (miR)-1467, -1185, -4493 and -3919, their expression levels were analyzed in cervical cancer tissues and adjacent non-cancerous tissues; they were downregulated by ~45% in the cervical cancer samples. The results of the present study showed that OCT1 is highly expressed in cervical cancer tissues and indicated that OCT-1 may be significant in cervical cancer.
Collapse
Affiliation(s)
- Songshu Xiao
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Shan Liao
- Molecular Genetics Laboratory, Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yanhong Zhou
- Molecular Genetics Laboratory, Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Bin Jiang
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yueran Li
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Min Xue
- Department of Gynecology and Obstetrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
33
|
Meyer Zu Schwabedissen HE, Begunk R, Hussner J, Juhnke BO, Gliesche D, Böttcher K, Sternberg K, Schmitz KP, Kroemer HK. Cell-Specific Expression of Uptake Transporters—A Potential Approach for Cardiovascular Drug Delivery Devices. Mol Pharm 2014; 11:665-72. [DOI: 10.1021/mp400245g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Abstract
Metformin is the first-line treatment for type 2 diabetes. Results from several clinical studies have indicated that type 2 diabetic patients treated with metformin might have a lower cancer risk. One of the primary metabolic changes observed in malignant cell transformation is an increased catabolic glucose metabolism. In this context, once it has entered the cell through organic cation transporters, metformin decreases mitochondrial respiration chain activity and ATP production that, in turn, activates AMP-activated protein kinase, which regulates energy homeostasis. In addition, metformin reduces cellular energy availability and glucose entrapment by inhibiting hexokinase-II, which catalyses the glucose phosphorylation reaction. In this review, we discuss recent findings on molecular mechanisms that sustain the anticancer effect of metformin through regulation of glucose metabolism. In particular, we have focused on the emerging action of metformin on glycolysis in normal and cancer cells, with a drug discovery perspective.
Collapse
Affiliation(s)
- Barbara Salani
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| | - Alberto Del Rio
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| | - Cecilia Marini
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| | - Gianmario Sambuceti
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| | - Renzo Cordera
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| | - Davide Maggi
- Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy Department of Internal Medicine (DIMI)University of Genova, Viale Benedetto XV/6, 16132 Genova, ItalyIRCCS Azienda Ospedaliera Universitaria San Martino - IST Istituto Nazionale per la Ricerca sul Cancro16132 Genova, ItalyDepartment of ExperimentalDiagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum, University of Bologna, Bologna, ItalyCNR Institute of Organic Synthesis and Photoreactivity (ISOF)40129 Bologna, ItalyCNR Institute of Molecular Bioimaging and Physiology (IBFM)16132 Genova, ItalyDepartment of Health Science (DISSAL)University of Genova, 16132 Genova, Italy
| |
Collapse
|
35
|
Koepsell H. The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 2013; 34:413-35. [PMID: 23506881 DOI: 10.1016/j.mam.2012.10.010] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 08/18/2012] [Indexed: 12/14/2022]
Abstract
The SLC22 family contains 13 functionally characterized human plasma membrane proteins each with 12 predicted α-helical transmembrane domains. The family comprises organic cation transporters (OCTs), organic zwitterion/cation transporters (OCTNs), and organic anion transporters (OATs). The transporters operate as (1) uniporters which mediate facilitated diffusion (OCTs, OCTNs), (2) anion exchangers (OATs), and (3) Na(+)/zwitterion cotransporters (OCTNs). They participate in small intestinal absorption and hepatic and renal excretion of drugs, xenobiotics and endogenous compounds and perform homeostatic functions in brain and heart. Important endogeneous substrates include monoamine neurotransmitters, l-carnitine, α-ketoglutarate, cAMP, cGMP, prostaglandins, and urate. It has been shown that mutations of the SLC22 genes encoding these transporters cause specific diseases like primary systemic carnitine deficiency and idiopathic renal hypouricemia and are correlated with diseases such as Crohn's disease and gout. Drug-drug interactions at individual transporters may change pharmacokinetics and toxicities of drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, 97070 Würzburg, Germany.
| |
Collapse
|
36
|
Herraez E, Lozano E, Macias RIR, Vaquero J, Bujanda L, Banales JM, Marin JJG, Briz O. Expression of SLC22A1 variants may affect the response of hepatocellular carcinoma and cholangiocarcinoma to sorafenib. Hepatology 2013; 58:1065-1073. [PMID: 23532667 DOI: 10.1002/hep.26425] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/22/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Reduced drug uptake is an important mechanism of chemoresistance. Down-regulation of SLC22A1 encoding the organic cation transporter-1 (OCT1) may affect the response of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CGC) to sorafenib, a cationic drug. Here we investigated whether SLC22A1 variants may contribute to sorafenib chemoresistance. Complete sequencing and selective variant identification were carried out to detect single nucleotide polymorphisms (SNPs) in SLC22A1 complementary DNA (cDNA). In HCC and CGC biopsies, in addition to previously described variants, two novel alternative spliced variants and three SNPs were identified. To study their functional consequences, these variants were mimicked by directed mutagenesis and expressed in HCC (Alexander and SK-Hep-1) and CGC (TFK1) cells. The two novel described variants, R61S fs*10 and C88A fs*16, encoded truncated proteins unable to reach the plasma membrane. Both variants abolished OCT1-mediated uptake of tetraethylammonium, a typical OCT1 substrate, and were not able to induce sorafenib sensitivity. In cells expressing functional OCT1 variants, OCT1 inhibition with quinine prevented sorafenib-induced toxicity. Expression of OCT1 variants in Xenopus laevis oocytes and determination of quinine-sensitive sorafenib uptake by high-performance liquid chromatography-dual mass spectrometry confirmed that OCT1 is able to transport sorafenib and that R61S fs*10 and C88A fs*16 abolish this ability. Screening of these SNPs in 23 HCC and 15 CGC biopsies revealed that R61S fs*10 was present in both HCC (17%) and CGC (13%), whereas C88A fs*16 was only found in HCC (17%). Considering all SLC22A1 variants, at least one inactivating SNP was found in 48% HCC and 40% CGC. CONCLUSION Development of HCC and CGC is accompanied by the appearance of aberrant OCT1 variants that, together with decreased OCT1 expression, may dramatically affect the ability of sorafenib to reach active intracellular concentrations in these tumors.
Collapse
Affiliation(s)
- Elisa Herraez
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Role of the plasma membrane transporter of organic cations OCT1 and its genetic variants in modern liver pharmacology. BIOMED RESEARCH INTERNATIONAL 2013; 2013:692071. [PMID: 23984399 PMCID: PMC3747481 DOI: 10.1155/2013/692071] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/04/2013] [Indexed: 01/11/2023]
Abstract
Changes in the uptake of many drugs by the target cells may dramatically affect the pharmacological response. Thus, downregulation of SLC22A1, which encodes the organic cation transporter type 1 (OCT1), may affect the response of healthy hepatocytes and liver cancer cells to cationic drugs, such as metformin and sorafenib, respectively. Moreover, the overall picture may be modified to a considerable extent by the preexistence or the appearance during the pathogenic process of genetic variants. Some rare OCT1 variants enhance transport activity, whereas other more frequent variants impair protein maturation, plasma membrane targeting or the function of this carrier, hence reducing intracellular active drug concentrations. Here, we review current knowledge of the role of OCT1 in modern liver pharmacology, which includes the use of cationic drugs to treat several diseases, some of them of great clinical relevance such as diabetes and primary liver cancer (cholangiocarcinoma and hepatocellular carcinoma). We conclude that modern pharmacology must consider the individual evaluation of OCT1 expression/function in the healthy liver and in the target tissue, particularly if this is a tumor, in order to predict the lack of response to cationic drugs and to be able to design individualized pharmacological treatments with the highest chances of success.
Collapse
|
38
|
Lin R, Li X, Li J, Zhang L, Xu F, Chu Y, Li J. Long-term cisplatin exposure promotes methylation of the OCT1 gene in human esophageal cancer cells. Dig Dis Sci 2013; 58:694-8. [PMID: 23053895 DOI: 10.1007/s10620-012-2424-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 09/19/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cisplatin-based chemotherapy is widely used for treatment of a variety of human malignant solid and metastatic tumors, including esophageal cancer. However, the clinical effect of this drug is limited because of intrinsic and acquired resistance to it. Organic cation transporters (OCTs) are important in the cellular uptake of cisplatin. AIM Our objective was to test the hypothesis that cisplatin resistance is associated with alteration of expression of OCTs. METHODS Levels of expression of OCTs in paired esophageal cancer and adjacent non-cancerous tissues were examined by use of immunohistochemistry. RESULTS We found that OCT1 silencing impaired cisplatin-mediated apoptosis of esophageal cancer cells. The level of OCT1 mRNA in cisplatin-resistant cells was markedly reduced compared with parental cells. Promoter methylation of OCT1 was induced in cisplatin-resistant cells. CONCLUSION This study shows that long-term exposure to cisplatin promotes methylation of the OCT1 gene in human esophageal cancer cells, which in turn results in cisplatin resistance.
Collapse
Affiliation(s)
- Rui Lin
- Department of Gastroenterology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Marin JJG. Plasma membrane transporters in modern liver pharmacology. SCIENTIFICA 2012; 2012:428139. [PMID: 24278693 PMCID: PMC3820525 DOI: 10.6064/2012/428139] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 09/26/2012] [Indexed: 06/02/2023]
Abstract
The liver plays a crucial role in the detoxification of drugs used in the treatment of many diseases. The liver itself is the target for drugs aimed to modify its function or to treat infections and tumours affecting this organ. Both detoxification and pharmacological processes occurring in the liver require the uptake of the drug by hepatic cells and, in some cases, the elimination into bile. These steps have been classified as detoxification phase 0 and phase III, respectively. Since most drugs cannot cross the plasma membrane by simple diffusion, the involvement of transporters is mandatory. Several members of the superfamilies of solute carriers (SLC) and ATP-binding cassette (ABC) proteins, with a minor participation of other families of transporters, account for the uptake and efflux, respectively, of endobiotic and xenobiotic compounds across the basolateral and apical membranes of hepatocytes and cholangiocytes. These transporters are also involved in the sensitivity and refractoriness to the pharmacological treatment of liver tumours. An additional interesting aspect of the role of plasma membrane transporters in liver pharmacology regards the promiscuity of many of these carriers, which accounts for a variety of drug-drug, endogenous substances-drug and food components-drug interactions with clinical relevance.
Collapse
Affiliation(s)
- Jose J. G. Marin
- Laboratory of Experimental Hepatology and Drug Targeting (HEVEFARM), IBSAL, University of Salamanca and CIBERehd, Spain
- Department of Physiology and Pharmacology, Campus Miguel de Unamuno E.D. S09, 37007 Salamanca, Spain
| |
Collapse
|
40
|
Jouan E, Le Vee M, Denizot C, Da Violante G, Fardel O. The mitochondrial fluorescent dye rhodamine 123 is a high-affinity substrate for organic cation transporters (OCTs) 1 and 2. Fundam Clin Pharmacol 2012; 28:65-77. [DOI: 10.1111/j.1472-8206.2012.01071.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 07/11/2012] [Accepted: 07/19/2012] [Indexed: 01/10/2023]
Affiliation(s)
- Elodie Jouan
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie; 2 Avenue du Pr Léon Bernard 35043 Rennes France
| | - Marc Le Vee
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie; 2 Avenue du Pr Léon Bernard 35043 Rennes France
| | - Claire Denizot
- Technologie Servier; 25-27 rue Eugène Vignat 45000 Orléans France
| | | | - Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie; 2 Avenue du Pr Léon Bernard 35043 Rennes France
- Pôle Biologie; Centre Hospitalier Universitaire; 2 rue Henri Le Guilloux 35033 Rennes France
| |
Collapse
|
41
|
Takeuchi K, Shibata M, Kashiyama E, Umehara K. Expression levels of multidrug resistance-associated protein 4 (MRP4) in human leukemia and lymphoma cell lines, and the inhibitory effects of the MRP-specific inhibitor MK-571 on methotrexate distribution in rats. Exp Ther Med 2012. [PMID: 23181130 PMCID: PMC3503844 DOI: 10.3892/etm.2012.627] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the development of anti-blood cancer drugs, the chronic myelocytic leukemia (KU812), acute myelocytic leukemia (KG-1) and lymphoma (U937) cell lines are commonly used in preclinical pharmacology studies as human cancer xenograft models in mice. In the present study, mRNA expression levels of typical human ATP-binding cassette (ABC) transporters in these human blood cancer cell lines were analyzed by real-time polymerase chain reaction (RT-PCR). Based on the results, the expression level of multidrug resistance-associated protein 4 (MRP4) was found to be extremely high in KU812 cells compared with those of other transporters. Additionally, MRP4 expression levels were found to be relatively high in U937, KG-1 and a blood cell line derived from a healthy subject (RPMI 1788). In addition, to elucidate the contribution of MRP4 to the methotrexate (MTX) distribution in normal blood cells and tissues, [3H]MTX was intravenously (i.v.) administered to two groups of rats. Animals in one group received [3H]MTX only; the other group was concomitantly administered i.v. MK-571, a typical inhibitor of MRP transporters. No marked difference was observed between the two groups; the Kp values (tissue concentration/plasma concentration) of the concomitant group showed slightly higher values compared with those of the MTX alone group in erythrocytes (1.4 times, P<0.001), spleen (1.3 times, P<0.05) and thymus (1.2 times, P<0.05), respectively. Although in the present study we could not evaluate the direct involvement of MRP4 in blood cancer cells in which MRP4 expression was excessively high, these results suggest a possible functional role of MRP4 in blood cancer cells and albeit only slightly in normal blood cells/tissues.
Collapse
Affiliation(s)
- Kenji Takeuchi
- Department of Drug Metabolism, Drug Safety Research Center, Tokushima Research Institute, Otsuka Pharmaceutical Co., Ltd., Kawauchi-cho, Tokushima 771-0192, Japan
| | | | | | | |
Collapse
|
42
|
Xiao RP. Beta-adrenergic signaling in the heart: dual coupling of the beta2-adrenergic receptor to G(s) and G(i) proteins. SCIENCE'S STKE : SIGNAL TRANSDUCTION KNOWLEDGE ENVIRONMENT 2001; 2001:re15. [PMID: 11604549 DOI: 10.1126/stke.2001.104.re15] [Citation(s) in RCA: 131] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Beta-adrenergic receptor (AR) subtypes are archetypical members of the G protein-coupled receptor (GPCR) superfamily. Whereas both beta1AR and beta2AR stimulate the classic G(s)-adenylyl cyclase-3',5'-adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling cascade, beta2AR couples to both G(s) and G(i) proteins, activating bifurcated signaling pathways. In the heart, dual coupling of the beta2AR to G(s) and G(i) results in compartmentalization of the G(s)-stimulated cAMP signal, thus selectively affecting plasma membrane effectors (such as L-type Ca(2+) channels) and bypassing cytoplasmic target proteins (such as phospholamban and myofilament contractile proteins). More important, the beta2AR-to-G(i) branch delivers a powerful cell survival signal that counters apoptosis induced by the concurrent G(s)-mediated signal or by a wide range of assaulting factors. This survival pathway sequentially involves G(i), G(beta)(gamma), phosphoinositide 3-kinase, and Akt. Furthermore, cardiac-specific transgenic overexpression of betaAR subtypes in mice results in distinctly different phenotypes in terms of the likelihood of cardiac hypertrophy and heart failure. These findings indicate that stimulation of the two betaAR subtypes activates overlapping, but different, sets of signal transduction mechanisms, and fulfills distinct or even opposing physiological and pathophysiological roles. Because of these differences, selective activation of cardiac beta2AR may provide catecholamine-dependent inotropic support without cardiotoxic consequences, which might have beneficial effects in the failing heart.
Collapse
Affiliation(s)
- R P Xiao
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, Baltimore, MD 21224, USA.
| |
Collapse
|
43
|
Xiao RP. -Adrenergic Signaling in the Heart: Dual Coupling of the 2-Adrenergic Receptor to Gs and Gi Proteins. Sci Signal 2001. [DOI: 10.1126/scisignal.1042001re15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|