1
|
Ebner J, Schmoellerl J, Piontek M, Manhart G, Troester S, Carter BZ, Neubauer H, Moriggl R, Szakács G, Zuber J, Köcher T, Andreeff M, Sperr WR, Valent P, Grebien F. ABCC1 and glutathione metabolism limit the efficacy of BCL-2 inhibitors in acute myeloid leukemia. Nat Commun 2023; 14:5709. [PMID: 37726279 PMCID: PMC10509209 DOI: 10.1038/s41467-023-41229-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/25/2023] [Indexed: 09/21/2023] Open
Abstract
The BCL-2 inhibitor Venetoclax is a promising agent for the treatment of acute myeloid leukemia (AML). However, many patients are refractory to Venetoclax, and resistance develops quickly. ATP-binding cassette (ABC) transporters mediate chemotherapy resistance but their role in modulating the activity of targeted small-molecule inhibitors is unclear. Using CRISPR/Cas9 screening, we find that loss of ABCC1 strongly increases the sensitivity of AML cells to Venetoclax. Genetic and pharmacologic ABCC1 inactivation potentiates the anti-leukemic effects of BCL-2 inhibitors and efficiently re-sensitizes Venetoclax-resistant leukemia cells. Conversely, ABCC1 overexpression induces resistance to BCL-2 inhibitors by reducing intracellular drug levels, and high ABCC1 levels predicts poor response to Venetoclax therapy in patients. Consistent with ABCC1-specific export of glutathionylated substrates, inhibition of glutathione metabolism increases the potency of BCL-2 inhibitors. These results identify ABCC1 and glutathione metabolism as mechanisms limiting efficacy of BCL-2 inhibitors, which may pave the way to development of more effective therapies.
Collapse
Affiliation(s)
- Jessica Ebner
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Johannes Schmoellerl
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Martin Piontek
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Selina Troester
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heidi Neubauer
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Richard Moriggl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Gergely Szakács
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
- Institute of Enzymology, Research Centre of Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | - Thomas Köcher
- Vienna BioCenter Core Facilities, Vienna BioCenter, Vienna, Austria
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
| |
Collapse
|
2
|
New Advances in the Research of Resistance to Neoadjuvant Chemotherapy in Breast Cancer. Int J Mol Sci 2021; 22:ijms22179644. [PMID: 34502549 PMCID: PMC8431789 DOI: 10.3390/ijms22179644] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer has an extremely high incidence in women, and its morbidity and mortality rank first among female tumors. With the increasing development of medicine today, the clinical application of neoadjuvant chemotherapy has brought new hope to the treatment of breast cancer. Although the efficacy of neoadjuvant chemotherapy has been confirmed, drug resistance is one of the main reasons for its treatment failure, contributing to the difficulty in the treatment of breast cancer. This article focuses on multiple mechanisms of action and expounds a series of recent research advances that mediate drug resistance in breast cancer cells. Drug metabolizing enzymes can mediate a catalytic reaction to inactivate chemotherapeutic drugs and develop drug resistance. The drug efflux system can reduce the drug concentration in breast cancer cells. The combination of glutathione detoxification system and platinum drugs can cause breast cancer cells to be insensitive to drugs. Changes in drug targets have led to poorer efficacy of HER2 receptor inhibitors. Moreover, autophagy, epithelial–mesenchymal transition, and tumor microenvironment can all contribute to the development of resistance in breast cancer cells. Based on the relevant research on the existing drug resistance mechanism, the current treatment plan for reversing the resistance of breast cancer to neoadjuvant chemotherapy is explored, and the potential drug targets are analyzed, aiming to provide a new idea and strategy to reverse the resistance of neoadjuvant chemotherapy drugs in breast cancer.
Collapse
|
3
|
Malla JA, Umesh RM, Yousf S, Mane S, Sharma S, Lahiri M, Talukdar P. A Glutathione Activatable Ion Channel Induces Apoptosis in Cancer Cells by Depleting Intracellular Glutathione Levels. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202000961] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Javid Ahmad Malla
- Department of Chemistry Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Rintu M. Umesh
- Department of Biology Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Saleem Yousf
- Department of Chemistry Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Shrunal Mane
- Department of Biology Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Shilpy Sharma
- Department of Biotechnology Savitribai Phule Pune University (Formerly University of Pune) Pune Maharashtra 411007 India
| | - Mayurika Lahiri
- Department of Biology Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| | - Pinaki Talukdar
- Department of Chemistry Indian Institute of Science Education and Research Pune Dr. Homi Bhabha Road, Pashan Pune 411008 Maharashtra India
| |
Collapse
|
4
|
Malla JA, Umesh RM, Yousf S, Mane S, Sharma S, Lahiri M, Talukdar P. A Glutathione Activatable Ion Channel Induces Apoptosis in Cancer Cells by Depleting Intracellular Glutathione Levels. Angew Chem Int Ed Engl 2020; 59:7944-7952. [PMID: 32048775 DOI: 10.1002/anie.202000961] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/11/2020] [Indexed: 01/30/2023]
Abstract
Cancer cells use elevated glutathione (GSH) levels as an inner line of defense to evade apoptosis and develop drug resistance. In this study, we describe a novel 2,4-nitrobenzenesulfonyl (DNS) protected 2-hydroxyisophthalamide system that exploits GSH for its activation into free 2-hydroxyisophthalamide forming supramolecular M+ /Cl- channels. Better permeation of the DNS protected compound into MCF-7 cells compared to the free 2-hydroxyisophthalamide and GSH-activatable ion transport resulted in higher cytotoxicity, which was associated with increased oxidative stress that further reduced the intracellular GSH levels and altered mitochondrial membrane permeability leading to the induction of the intrinsic apoptosis pathway. The GSH-activatable transport-mediated cell death was further validated in rat insulinoma cells (INS-1E); wherein the intracellular GSH levels showed a direct correlation to the resulting cytotoxicity. Lastly, the active compound was found to restrict the growth and proliferation of 3D spheroids of MCF-7 cells with efficiency similar to that of the anticancer drug doxorubicin.
Collapse
Affiliation(s)
- Javid Ahmad Malla
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Rintu M Umesh
- Department of Biology, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Saleem Yousf
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Shrunal Mane
- Department of Biology, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Pune, Maharashtra, 411007, India
| | - Mayurika Lahiri
- Department of Biology, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| |
Collapse
|
5
|
Silbermann K, Stefan SM, Elshawadfy R, Namasivayam V, Wiese M. Identification of Thienopyrimidine Scaffold as an Inhibitor of the ABC Transport Protein ABCC1 (MRP1) and Related Transporters Using a Combined Virtual Screening Approach. J Med Chem 2019; 62:4383-4400. [PMID: 30925062 DOI: 10.1021/acs.jmedchem.8b01821] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A virtual screening protocol with combination of similarity search and pharmacophore modeling was applied to virtually screen a large compound library to gain new scaffolds regarding ABCC1 inhibition. Biological investigation of promising candidates revealed four compounds as ABCC1 inhibitors, three of them with scaffolds not associated with ABCC1 inhibition until now. The best hit molecule-a thienopyrimidine-was a moderately potent, competitive inhibitor of the ABCC1-mediated transport of calcein AM which also sensitized ABCC1-overexpressing cells toward daunorubicin. Further evaluation showed that it was a moderately potent, competitive inhibitor of the ABCB1-mediated transport of calcein AM, and noncompetitive inhibitor of the ABCG2-mediated pheophorbide A transport. In addition, the thienopyrimidine could also sensitize ABCB1- as well as ABCG2-overexpressing cells toward daunorubicin and SN-38, respectively, in concentration ranges that qualified it as one of the ten best triple ABCC1/ABCB1/ABCG2 inhibitors in the literature. Besides, three more new multitarget inhibitors were identified by this virtual screening approach.
Collapse
Affiliation(s)
- Katja Silbermann
- Pharmaceutical Chemistry II, Pharmaceutical Institute , Rheinische Friedrich-Wilhelms-University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Sven Marcel Stefan
- Pharmaceutical Chemistry II, Pharmaceutical Institute , Rheinische Friedrich-Wilhelms-University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Randa Elshawadfy
- Pharmaceutical Chemistry II, Pharmaceutical Institute , Rheinische Friedrich-Wilhelms-University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Vigneshwaran Namasivayam
- Pharmaceutical Chemistry II, Pharmaceutical Institute , Rheinische Friedrich-Wilhelms-University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| | - Michael Wiese
- Pharmaceutical Chemistry II, Pharmaceutical Institute , Rheinische Friedrich-Wilhelms-University of Bonn , An der Immenburg 4 , 53121 Bonn , Germany
| |
Collapse
|
6
|
Stefan SM, Wiese M. Small-molecule inhibitors of multidrug resistance-associated protein 1 and related processes: A historic approach and recent advances. Med Res Rev 2018; 39:176-264. [DOI: 10.1002/med.21510] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sven Marcel Stefan
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| | - Michael Wiese
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| |
Collapse
|
7
|
Vert A, Castro J, Ribó M, Vilanova M, Benito A. Transcriptional profiling of NCI/ADR-RES cells unveils a complex network of signaling pathways and molecular mechanisms of drug resistance. Onco Targets Ther 2018; 11:221-237. [PMID: 29379303 PMCID: PMC5757493 DOI: 10.2147/ott.s154378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Ovarian cancer has the highest mortality rate among all the gynecological cancers. This is mostly due to the resistance of ovarian cancer to current chemotherapy regimens. Therefore, it is of crucial importance to identify the molecular mechanisms associated with chemoresistance. Methods NCI/ADR-RES is a multidrug-resistant cell line that is a model for the study of drug resistance in ovarian cancer. We carried out a microarray-derived transcriptional profiling analysis of NCI/ADR-RES to identify differentially expressed genes relative to its parental OVCAR-8. Results Gene-expression profiling has allowed the identification of genes and pathways that may be important for the development of drug resistance in ovarian cancer. The NCI/ADR-RES cell line has differential expression of genes involved in drug extrusion, inactivation, and efficacy, as well as genes involved in the architectural and functional reorganization of the extracellular matrix. These genes are controlled through different signaling pathways, including MAPK–Akt, Wnt, and Notch. Conclusion Our findings highlight the importance of using orthogonal therapies that target completely independent pathways to overcome mechanisms of resistance to both classical chemotherapeutic agents and molecularly targeted drugs.
Collapse
Affiliation(s)
- Anna Vert
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Jessica Castro
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Marc Ribó
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Maria Vilanova
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| | - Antoni Benito
- Protein Engineering Laboratory, Department of Biology, Faculty of Sciences, Universitat de Girona.,Biomedical Research Institute of Girona (IDIBGi), Girona, Spain
| |
Collapse
|
8
|
Zhang L, Lu J, Qiu L. Synergistic effects of A-B-C-type amphiphilic copolymer on reversal of drug resistance in MCF-7/ADR breast carcinoma. Int J Nanomedicine 2016; 11:5205-5220. [PMID: 27785023 PMCID: PMC5066852 DOI: 10.2147/ijn.s115956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
P-glycoprotein (P-gp) overexpression has become the most common cause of occurrence of multidrug resistance in clinical settings. We aimed to construct a micellar polymer carrier to sensitize drug-resistant tumors to doxorubicin (DOX). This A-B-C-type amphiphilic copolymer was prepared by the sequential linkage of β-cyclodextrin, hydrophobic poly(d,l-lactide), and hydrophilic poly(ethylene glycol). Upon incubation of the DOX-loaded micelles with DOX-resistant human breast carcinoma MCF-7/ADR cells, significantly enhanced cytotoxicity and apoptosis were achieved. A series of studies on the action mechanism showed that the polymer components such as β-cyclodextrin, hydrophobic poly(d,l-lactide) segment, and poly(ethylene glycol) coordinatively contributed to the improved intracellular ATP depletion and ATPase activity, increased intracellular uptake of P-gp substrates via competitive binding to P-gp, and decreased P-gp expression in MCF-7/ADR cells. More interestingly, a similar phenomenon was observed in the zebrafish xenograft model, resulting in ~64% inhibition of MCF-7/ADR tumor growth. These results implied that the polymeric micelles displayed great potentials as P-gp modulators to reverse DOX resistance in MCF-7/ADR breast carcinoma.
Collapse
Affiliation(s)
- Lu Zhang
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou
- Drug Clinical Trial Office, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jiafei Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People’s Republic of China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou
| |
Collapse
|
9
|
Wang F, Liu X, Liu C, Liu Z, Sun L. Effects of antibiotic antitumor drugs on nucleotide levels in cultured tumor cells: an exploratory method to distinguish the mechanisms of antitumor drug action based on targeted metabolomics. Acta Pharm Sin B 2015; 5:223-30. [PMID: 26579450 PMCID: PMC4629260 DOI: 10.1016/j.apsb.2015.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/19/2014] [Accepted: 03/14/2015] [Indexed: 12/29/2022] Open
Abstract
Nucleotide pools in mammalian cells change due to the influence of antitumor drugs, which may help in evaluating the drug effect and understanding the mechanism of drug action. In this study, an ion-pair RP-HPLC method was used for a simple, sensitive and simultaneous determination of the levels of 12 nucleotides in mammalian cells treated with antibiotic antitumor drugs (daunorubicin, epirubicin and dactinomycin D). Through the use of this targeted metabolomics approach to find potential biomarkers, UTP and ATP were verified to be the most appropriate biomarkers. Moreover, a holistic statistical approach was put forward to develop a model which could distinguish 4 categories of drugs with different mechanisms of action. This model can be further validated by evaluating drugs with different mechanisms of action. This targeted metabolomics study may provide a novel approach to predict the mechanism of action of antitumor drugs.
Collapse
Key Words
- ADP, adenosine diphosphate
- AMP, adenosine monophosphate
- ANOVA, analysis of variance
- ATP, adenosine triphosphate
- AUC, area under the curve
- Antibiotic anticancer drugs
- CDP, cytidine diphosphate
- CTP, cytidine triphosphate
- DMEM, Dulbecco׳s modified eagle׳s cell culture media
- DMSO, dimethyl sulfoxide
- DNA, deoxyribonucleic acid
- EC, energy charge
- EDTA, ethylene diamine tetra-acetic acid
- FCS, fetal calf serum
- GDP, guanosine diphosphate
- GMP, guanosine monophosphate
- GTP, guanosine triphosphate
- HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
- Ion-pair HPLC
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Mechanisms of antitumor drug action
- Nucleotides
- PBS, phosphate buffered saline
- PCA, principal component analysis
- Potential biomarkers
- Principal component analysis
- RNA, ribonucleic acid
- ROC, receiver operating characteristic
- RPMI-1640, Roswell Park Memorial Institute-1640
- TBAHS, tetrabutylammonium hydrogen sulfate
- TCA, trichloroacetic acid
- Targeted metabolomics analysis
- Tumor cells
- UDP, uridine diphosphate
- UTP, uridine triphosphate
- dATP, deoxyadenosine triphosphate
- dCDP, deoxycytidine diphosphate
- dCTP, deoxycytidine triphosphate
- dGMP, deoxyribonucleic monophosphate
- dGTP, deoxyguanosine triphosphate
- dUDP, deoxyuridine diphpsphate
- dUTP, deoxyuridine triphosphate
Collapse
Affiliation(s)
- Fang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xi Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Cuichai Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zheng Liu
- School of Life Science and Bio-pharmaceutics Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding author. Tel.: +86 24 23986365; fax: +86 24 23986259.
| |
Collapse
|
10
|
Leonel C, Gelaleti GB, Jardim BV, Moschetta MG, Regiani VR, Oliveira JG, Zuccari DAPC. Expression of glutathione, glutathione peroxidase and glutathione S-transferase pi in canine mammary tumors. BMC Vet Res 2014; 10:49. [PMID: 24565113 PMCID: PMC3975948 DOI: 10.1186/1746-6148-10-49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 02/15/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Glutathione (GSH) is one of the most important agents of the antioxidant defense system of the cell because, in conjunction with the enzymes glutathione peroxidase (GSH-Px) and glutathione S transferase pi (GSTpi), it plays a central role in the detoxification and biotransformation of chemotherapeutic drugs. This study evaluated the expression of GSH and the GSH-Px and GSTpi enzymes by immunohistochemistry in 30 canine mammary tumors, relating the clinicopathological parameters, clinical outcome and survival of the bitches. In an in vitro study, the expression of the genes glutamate cysteine ligase (GCLC) and glutathione synthetase (GSS) that synthesize GSH and GSH-Px gene were verified by qPCR and subjected to treatment with doxorubicin, to check the resistance of cancer cells to chemotherapy. RESULTS The immunohistochemical expression of GSH, GSH-Px and GSTpi was compared with the clinical and pathological characteristics and the clinical outcome in the bitches, including metastasis and death.The results showed that high immunoexpression of GSH was correlated to the absence of tumor ulceration and was present in dogs without metastasis (P < 0.05). There was significant correlation of survival with the increase of GSH (P < 0.05). The expression of the GSH-Px and GSTpi enzymes showed no statistically significant correlation with the analyzed variables (p > 0.05). The analysis of the relative expression of genes responsible for the synthesis of GSH (GCLC and GSS) and GSH-Px by quantitative PCR was done with cultured cells of 10 tumor fragments from dogs with mammary tumors.The culture cells showed a decrease in GCLC and GSS expression when compared with no treated cells (P < 0.05). High GSH immunoexpression was associated with better clinical outcomes. CONCLUSION Therefore, high expression of the GSH seems to play an important role in the clinical outcome of patients with mammary tumors and suggest its use as prognostic marker. The in vitro doxorubicin treatment significantly reduces the expression of GCLC and GSS genes so we can consider them to be candidates for predictive markers of therapeutic response in mammary cancer.
Collapse
Affiliation(s)
- Camila Leonel
- Graduate Program in Genetics, Universidade Estadual Paulista – UNESP/IBILCE, São José do Rio Preto, SP, Brazil
| | - Gabriela B Gelaleti
- Graduate Program in Genetics, Universidade Estadual Paulista – UNESP/IBILCE, São José do Rio Preto, SP, Brazil
| | - Bruna V Jardim
- Graduate Program in Genetics, Universidade Estadual Paulista – UNESP/IBILCE, São José do Rio Preto, SP, Brazil
| | - Marina G Moschetta
- Graduate Program in Health Sciencies, Laboratory of Molecular Reserach in Cancer (LIMC), Departament of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, FAMERP, São José do Rio Preto, SP, Brazil
| | - Vitor R Regiani
- Graduate Program in Health Sciencies, Research Unit Genetics and Molecular Biology (UPGEM), Faculdade de Medicina de São José do Rio Preto, FAMERP, São José do Rio Preto, SP, Brazil
| | | | - Debora APC Zuccari
- Departament of Molecular Biology, Faculdade de Medicina de São José do Rio Preto, FAMERP, São José do Rio Preto, SP, Brazil
| |
Collapse
|
11
|
Akbas HS, Timur M, Ozben T. Concurrent use of antioxidants in cancer therapy: an update. Expert Rev Clin Immunol 2014; 2:931-9. [DOI: 10.1586/1744666x.2.6.931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Palakurthi S, Yellepeddi VK, Vangara KK. Recent trends in cancer drug resistance reversal strategies using nanoparticles. Expert Opin Drug Deliv 2013; 9:287-301. [PMID: 22339554 DOI: 10.1517/17425247.2012.665365] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Resistance to chemotherapy is a major obstacle in the successful amelioration of tumors in many cancer patients. Resistance is either intrinsic or acquired, involving mechanisms such as genetic aberrations, decreased influx and increased efflux of drugs. Strategies for the reversal of resistance involve the alteration of enzymes responsible for drug resistance, the modulation of proteins regulating apoptosis mechanisms and improving the uptake of drugs using nanotechnology. Novel strides in the reversal of drug resistance are emerging, involving the use of nanotechnology, targeting stem cells, etc. AREAS COVERED This paper reviews the most recent cancer drug reversal strategies involving nanotechnology for targeting cancer cells and cancer stem cells (CSCs), for enhanced uptake of micro- and macromolecular inhibitors. EXPERT OPINION Nanotechnology used in conjunction with existing therapies, such as gene therapy and P-glycoprotein inhibition, has been shown to improve the reversal of drug resistance; the mechanisms involved in this include specific targeting of drugs and nucleotide therapeutics, enhanced cellular uptake of drugs and improved bioavailability of drugs with poor physicochemical characteristics. Important strategies in the reversal of drug resistance include: a multifunctional nanoparticulate system housing a targeting moiety; therapeutics to kill resistant cancer cells and CSCs; cytotoxic drugs and a tumor microenvironment stimuli-responsive element, to release the encapsulated therapeutics.
Collapse
Affiliation(s)
- Srinath Palakurthi
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA.
| | | | | |
Collapse
|
13
|
(99m)Tc(N)-DBODC(5), a potential radiolabeled probe for SPECT of multidrug resistance: in vitro study. J Biol Inorg Chem 2013; 18:523-38. [PMID: 23543234 DOI: 10.1007/s00775-013-0997-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 03/10/2013] [Indexed: 12/13/2022]
Abstract
[(99m)Tc(N)(DBODC)(PNP5)](+) [DBODC is bis(N-ethoxyethyl)dithiocarbamato; PNP5 is bis(dimethoxypropylphosphinoethyl)ethoxyethylamine], abbreviated as (99m)Tc(N)-DBODC(5), is a lipophilic cationic mixed compound investigated as a myocardial imaging agent. The findings that this tracer accumulates in mitochondrial structures through a mechanism mediated by the negative mitochondrial membrane potential and that the rapid efflux of (99m)Tc(N)-DBODC(5) from nontarget tissues seems to be associated with the multidrug resistance (MDR) P-glycoprotein (P-gp) transport function open up the possibility to extend its clinical applications to tumor imaging and noninvasive MDR studies. The rate of uptake at 4 and 37 °C of (99m)Tc(N)-DBODC(5) was evaluated in vitro in selected human cancer cell lines and in the corresponding sublines before and after P-gp and/or MDR-associated protein (MRP) modulator/inhibitor treatment using (99m)Tc-sestamibi as a reference. The results indicated that (1) the uptake of both (99m)Tc(N)-DBODC(5) and (99m)Tc-sestamibi is correlated to metabolic activity of the cells and (2) the cellular accumulation is connected to the level of P-gp/MRP expression; in fact, an enhancement of uptake in resistant cells was observed after treatment with opportune MDR inhibitor/modulator, indicating that the selective blockade of P-gp/MRP prevented efflux of the tracers. This study provides a preliminary indication of the applicability of (99m)Tc(N)-DBODC(5) in tumor imaging and in detecting P-gp/MRP-mediated drug resistance in human cancer. In addition, the possibility to control the hydrophobicity and pharmacological activity of this heterocomplex through the variation of the substituents on the ligands backbone without affecting the P2S2 coordinating sphere makes (99m)Tc(N)-DBODC(5) a suitable scaffold for the preparation of a molecular probe for single photon emission computed tomography of MDR.
Collapse
|
14
|
Xue T, Zhang Y, Zhang L, Yao L, Hu X, Xu LX. Proteomic Analysis of Two Metabolic Proteins with Potential to Translocate to Plasma Membrane Associated with Tumor Metastasis Development and Drug Targets. J Proteome Res 2013; 12:1754-63. [DOI: 10.1021/pr301100r] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ting Xue
- School of Biomedical Engineering
and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- School of Biomedical Engineering
and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Luofu Zhang
- School
of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yao
- School of Biomedical Engineering
and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofang Hu
- School of Biomedical Engineering
and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lisa X. Xu
- School of Biomedical Engineering
and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Tai DEJ, Jin WS, Wu CS, Si HW, Cao XD, Guo AJ, Chang JC. Changes in intracellular redox status influence multidrug resistance in gastric adenocarcinoma cells. Exp Ther Med 2012; 4:291-296. [PMID: 23139717 DOI: 10.3892/etm.2012.591] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 05/15/2012] [Indexed: 12/27/2022] Open
Abstract
Multidrug resistance (MDR) to chemotherapeutic agents is a major obstacle for the treatment of various types of cancers. The exact mechanism of MDR has not yet been fully clarified, although it has been frequently associated with the variation of intracellular redox status. The levels of intracellular glutathione (GSH) are considered to play a vital role in the regulation of the intracellular redox status. In our study, we investigated the effects of buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis, and NAC, a cysteine source for GSH synthesis, on sensitive gastric adenocarcinoma cells (SGC7901) and cisplatin-resistant SGC7901/DDP cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The two cell lines were pretreated with various non-toxic concentrations of BSO for 24 h and combined with fluorouracil (5-FU) or mitomycin (MMC) in the presence or absence of NAC before culturing further. After various treatments, the IC(50) values of MMC and 5-FU were calculated and intracellular GSH levels were measured using the glutathione reductase/5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) recycling assay without anticancer drug stimulation under the same microenvironments. The study demonstrated that BSO increased the sensitivity of the cells to chemotherapeutics while NAC exhibited the reverse effect, particularly in drug-resistant cells. It is, therefore, possible that changes in intracellular GSH levels affect the chemosensitivity of the resistant cells to a greater extent than that of their parent cells. This study indicates that variation in the intracellular redox status may be closely correlated with MDR and may provide a valuable basic strategy for anticancer therapy.
Collapse
Affiliation(s)
- DE-Jun Tai
- Department of General Surgery, The First Affiliated Hospital
| | | | | | | | | | | | | |
Collapse
|
16
|
Cai BL, Xu XF, Fu SM, Shen LL, Zhang J, Guan SM, Wu JZ. Nuclear translocation of MRP1 contributes to multidrug resistance of mucoepidermoid carcinoma. Oral Oncol 2011; 47:1134-40. [PMID: 21903448 DOI: 10.1016/j.oraloncology.2011.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 08/10/2011] [Accepted: 08/10/2011] [Indexed: 10/17/2022]
Abstract
Multidrug resistance-related protein 1 (MRP1 or ABCC1), a membrane-bound energy-dependent efflux transporter, is overexpressed in several kinds of multidrug-resistant cell lines and related to multidrug-resistance (MDR) of various cancers. In this study, we investigated whether MRP1 was involved in the chemoresistance of mucoepidermoid carcinoma (MEC). We demonstrated that down-regulation of MRP1 in MC3/5FU, a drug-resistant MEC cell line, by RNA interference increased the drug sensitivity of the cells to 5-fluorouracil, doxorubicin, pharmorubicin, bleomycin-A5, cis-platinum and taxol. However, no significant quantitative difference of MRP1 mRNA and protein expression was found between MC3/5FU cells and its parental cell line (MC3) as determined by RT-PCR and Western blot. Interestingly, MRP1 was translocated from the cytoplasmic membrane of the MC3 cells to the nuclei of MC3/5FU cells as revealed by indirect immunofluorescence staining. Furthermore, MRP1 down-regulation mainly decreased the nuclear expression of MRP1 rather than the cytoplasmic membrane expression. Our results suggested that MRP1 was involved in the chemoresistance of MEC and MRP1 may confer drug-resistance by a mechanism associated with its nuclear translocation.
Collapse
Affiliation(s)
- Bo-Lei Cai
- Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, 145 Chang Le Xi Road, Xi'an 710032, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Polyphosphazene nanoparticles for cytoplasmic release of doxorubicin with improved cytotoxicity against Dox-resistant tumor cells. J Colloid Interface Sci 2010; 355:374-82. [PMID: 21220138 DOI: 10.1016/j.jcis.2010.12.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 11/30/2010] [Accepted: 12/01/2010] [Indexed: 11/23/2022]
Abstract
This study involved the construction of self-assembled nanoparticles from novel pH-sensitive amphiphilic polyphosphazenes. These nanoparticles provide fast pH-responsive drug release and have the capability to disturb endosomal membranes. The polymers were prepared by linking N,N-diisopropylethylenediamine (DPA) onto a backbone of PEGylated polyphosphazene. In vitro cell viability measurements demonstrated the superior efficacy of these pH-responsive nanoparticles over free doxorubicin (Dox): the IC50 was over 60 times lower than that of free Dox against a Dox-resistant cell line. Using flow cytometry and confocal microscopy, the further investigation of the intracellular distribution of Dox and fluorescent probes provided evidence that, upon internalization by cells through endocytic pathways, the pH-sensitive polymer would disrupt membranes of endosomal compartments, releasing the cargo drugs into the cytoplasm in a burst-like manner. This resulted in reduced likelihood of drug efflux via exocytosis, and reversal of the drug resistance of the tumor cells. Generally, the pH-responsive nanoparticles designed in this study have achieved their potential as a drug delivery system for tumor therapy applications.
Collapse
|
18
|
Chintala S, Tóth K, Yin MB, Bhattacharya A, Smith SB, Ola MS, Cao S, Durrani FA, Zinia TR, Dean R, Slocum HK, Rustum YM. Downregulation of cystine transporter xc by irinotecan in human head and neck cancer FaDu xenografts. Chemotherapy 2010; 56:223-33. [PMID: 20551639 DOI: 10.1159/000316334] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 03/09/2010] [Indexed: 01/21/2023]
Abstract
BACKGROUND The purpose of this study was: (1) to document the critical requirement of cystine for growth of human tumor cells in vitro, and (2) to determine the effect of the anticancer agent irinotecan on the cystine transporter x(c)(-) in head and neck FaDu xenografts. METHODS Cell growth was measured by sulforhodamine B assay. xCT protein, glutathione (GSH) and DNA damage were determined using Western blot, spectrophotometry, and immunohistochemistry, respectively. RESULTS Depletion of cystine from the medium inhibited tumor cell growth. Treatment of FaDu tumor with a therapeutic dose of irinotecan resulted in depression of xCT protein levels, leading to tumor growth retardation and downregulation of GSH with increased reactive oxygen species (ROS). The accumulation of ROS correlated with increased DNA damage as evidenced by increased H2AX. CONCLUSION Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan.
Collapse
Affiliation(s)
- Sreenivasulu Chintala
- Department of Cancer Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xu W, Xi B, Wu J, An H, Zhu J, Abassi Y, Feinstein SC, Gaylord M, Geng B, Yan H, Fan W, Sui M, Wang X, Xu X. Natural product derivative Bis(4-fluorobenzyl)trisulfide inhibits tumor growth by modification of beta-tubulin at Cys 12 and suppression of microtubule dynamics. Mol Cancer Ther 2010; 8:3318-30. [PMID: 19996274 DOI: 10.1158/1535-7163.mct-09-0548] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bis(4-fluorobenzyl)trisulfide (BFBTS) is a synthetic molecule derived from a bioactive natural product, dibenzyltrisulfide, found in a subtropical shrub, Petiveria allieacea. BFBTS has potent anticancer activities to a broad spectrum of tumor cell lines with IC50 values from high nanomolar to low micromolar and showed equal anticancer potency between tumor cell lines overexpressing multidrug-resistant gene, MDR1 (MCF7/adr line and KBv200 line), and their parental MCF7 line and KB lines. BFBTS inhibited microtubule polymerization dynamics in MCF7 cells, at a low nanomolar concentration of 54 nmol/L, while disrupting microtubule filaments in cells at low micromolar concentration of 1 micromol/L. Tumor cells treated with BFBTS were arrested at G2-M phase, conceivably resulting from BFBTS-mediated antimicrotubule activities. Mass spectrometry studies revealed that BFBTS bound and modified beta-tubulin at residue Cys12, forming beta-tubulin-SS-fluorobenzyl. The binding site differs from known antimicrotubule agents, suggesting that BFBTS functions as a novel antimicrotubule agent. BFBTS at a dose of 25 mg/kg inhibited tumor growth with relative tumor growth rates of 19.91%, 18.5%, and 23.42% in A549 lung cancer, Bcap-37 breast cancer, and SKOV3 ovarian cancer xenografts, respectively. Notably, BFBTS was more potent against MDR1-overexpressing MCF7/adr breast cancer xenografts with a relative tumor growth rate of 12.3% than paclitaxel with a rate of 43.0%. BFBTS displays a novel antimicrotubule agent with potentials for cancer therapeutics.
Collapse
Affiliation(s)
- Wanhong Xu
- Hangzhou High Throughput Drug Screening Center, ACEA Biosciences, Inc., 6779 Mesa Ridge Road, Suite 100, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem Pharmacol 2009; 79:817-24. [PMID: 19883630 DOI: 10.1016/j.bcp.2009.10.017] [Citation(s) in RCA: 263] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 12/19/2022]
Abstract
Multidrug resistance-associated protein (MRP-1/ABCC1) transports a wide range of therapeutic agents and may play a critical role in the development of multidrug resistance (MDR) in tumor cells. However, the regulation of MRP-1 remains controversial. To explore whether miRNAs are involved in the regulation of MRP-1 expression and modulate the sensitivity of tumor cells to chemotherapeutic agents, we analyzed miRNA expression levels in VP-16-resistant MDR cell line, MCF-7/VP, in comparison with its parent cell line, MCF-7, using a miRNA microarray. MCF-7/VP overexpressed MRP-1 mRNA and protein not MDR-1 and BCRP. miR-326 was downregulated in MCF-7/VP compared to MCF-7. Additionally, miR-326 was downregulated in a panel of advanced breast cancer tissues and consistent reversely with expression levels of MRP-1. Furthermore, the elevated levels of miR-326 in the mimics-transfected VP-16-resistant cell line, MCF-7/VP, downregulated MRP-1 expression and sensitized these cells to VP-16 and doxorubicin. These findings demonstrate for the first time the involvement of miRNAs in multidrug resistance mediated by MRP-1 and suggest that miR-326 may be an efficient agent for preventing and reversing MDR in tumor cells.
Collapse
|
21
|
Lewis-Wambi JS, Swaby R, Kim H, Jordan VC. Potential of l-buthionine sulfoximine to enhance the apoptotic action of estradiol to reverse acquired antihormonal resistance in metastatic breast cancer. J Steroid Biochem Mol Biol 2009; 114:33-9. [PMID: 19167492 PMCID: PMC2869080 DOI: 10.1016/j.jsbmb.2008.12.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 12/31/2008] [Indexed: 11/26/2022]
Abstract
L-Buthionine sulfoximine (BSO) is a potent inhibitor of glutathione biosynthesis and studies have shown that it is capable of enhancing the apoptotic effects of several chemotherapeutic agents. Previous studies have shown that long-term antihormonal therapy leads to acquired drug resistance and that estrogen, which is normally a survival signal, is a potent apoptotic agent in these resistant cells. Interestingly, we have developed an antihormone-resistant breast cancer cell line, MCF-7:2A, which is resistant to estrogen-induced apoptosis but has elevated levels of glutathione. In the present study, we examined whether BSO is capable of sensitizing antihormone-resistant MCF-7:2A cells to estrogen-induced apoptosis. Our results showed that treatment of MCF-7:2A cells with 1nM E2 plus 100muM BSO combination for 1 week reduced the growth of these cells by almost 80-90% whereas the individual treatments had no significant effect on growth. TUNEL and 4',6-diamidino-2-phenylindole (DAPI) staining showed that the inhibitory effect of the combination treatment was due to apoptosis. Our data indicates that glutathione participates in retarding apoptosis in antihormone-resistant human breast cancer cells and that depletion of this molecule by BSO may be critical in predisposing resistant cells to estrogen-induced apoptosis.
Collapse
Affiliation(s)
- Joan S Lewis-Wambi
- Department of Medical Sciences, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Ramona Swaby
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Helen Kim
- Department of Medical Sciences, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - V. Craig Jordan
- Department of Medical Sciences, Fox Chase Cancer Center, Philadelphia, PA, United States
- Corresponding author: V. Craig Jordan, Department of Medical Sciences, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111. Phone: 215-728-7410; Fax: 215-728-7034.
| |
Collapse
|
22
|
Meijerman I, Beijnen JH, Schellens JH. Combined action and regulation of phase II enzymes and multidrug resistance proteins in multidrug resistance in cancer. Cancer Treat Rev 2008; 34:505-20. [DOI: 10.1016/j.ctrv.2008.03.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 02/11/2008] [Accepted: 03/01/2008] [Indexed: 01/16/2023]
|
23
|
Richardson DR, Lok HC. The nitric oxide–iron interplay in mammalian cells: Transport and storage of dinitrosyl iron complexes. Biochim Biophys Acta Gen Subj 2008; 1780:638-51. [DOI: 10.1016/j.bbagen.2007.12.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/03/2007] [Accepted: 12/18/2007] [Indexed: 02/05/2023]
|
24
|
Chang XB. A molecular understanding of ATP-dependent solute transport by multidrug resistance-associated protein MRP1. Cancer Metastasis Rev 2007; 26:15-37. [PMID: 17295059 DOI: 10.1007/s10555-007-9041-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over a million new cases of cancers are diagnosed each year in the United States and over half of these patients die from these devastating diseases. Thus, cancers cause a major public health problem in the United States and worldwide. Chemotherapy remains the principal mode to treat many metastatic cancers. However, occurrence of cellular multidrug resistance (MDR) prevents efficient killing of cancer cells, leading to chemotherapeutic treatment failure. Numerous mechanisms of MDR exist in cancer cells, such as intrinsic or acquired MDR. Overexpression of ATP-binding cassette (ABC) drug transporters, such as P-glycoprotein (P-gp or ABCB1), breast cancer resistance protein (BCRP or ABCG2) and/or multidrug resistance-associated protein (MRP1 or ABCC1), confers an acquired MDR due to their capabilities of transporting a broad range of chemically diverse anticancer drugs. In addition to their roles in MDR, there is substantial evidence suggesting that these drug transporters have functions in tissue defense. Basically, these drug transporters are expressed in tissues important for absorption, such as in lung and gut, and for metabolism and elimination, such as in liver and kidney. In addition, these drug transporters play an important role in maintaining the barrier function of many tissues including blood-brain barrier, blood-cerebral spinal fluid barrier, blood-testis barrier and the maternal-fetal barrier. Thus, these ATP-dependent drug transporters play an important role in the absorption, disposition and elimination of the structurally diverse array of the endobiotics and xenobiotics. In this review, the molecular mechanism of ATP-dependent solute transport by MRP1 will be addressed.
Collapse
Affiliation(s)
- Xiu-bao Chang
- Mayo Clinic College of Medicine, Scottsdale, AZ 85259, USA.
| |
Collapse
|
25
|
Narang VS, Pauletti GM, Gout PW, Buckley DJ, Buckley AR. Sulfasalazine-induced reduction of glutathione levels in breast cancer cells: enhancement of growth-inhibitory activity of Doxorubicin. Chemotherapy 2007; 53:210-7. [PMID: 17356269 DOI: 10.1159/000100812] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 02/28/2006] [Indexed: 11/19/2022]
Abstract
BACKGROUND We previously showed that the anti-inflammatory drug, sulfasalazine (salicylazosulfapyridine, SASP), can arrest proliferation of MCF-7 and MDA-MB-231 mammary cancer cells by inhibiting uptake of cystine via the x(c-) cystine/glutamate antiporter. Here we examined SASP with regard to reduction of cellular glutathione (GSH) levels and drug efficacy-enhancing ability. METHODS GSH levels were measured spectrophotometrically. Cellular drug retention was determined with 3H-labeled methotrexate, and drug efficacy with a colony formation assay. RESULTS Incubation of the mammary cancer cells with SASP (0.3-0.5 mM) led to reduction of their GSH content in a time- and concentration-dependent manner. Similar to MK-571, a multidrug resistance-associated protein inhibitor, SASP increased intracellular accumulation of methotrexate. Preincubation of cells with SASP (0.3 mM) significantly enhanced the potency of the anticancer agent doxorubicin (2.5 nM). CONCLUSIONS SASP-induced reduction of cellular GSH levels can lead to growth arrest of mammary cancer cells and enhancement of anticancer drug efficacy.
Collapse
Affiliation(s)
- Vishal S Narang
- College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | | | | |
Collapse
|
26
|
Watts RN, Hawkins C, Ponka P, Richardson DR. Nitrogen monoxide (NO)-mediated iron release from cells is linked to NO-induced glutathione efflux via multidrug resistance-associated protein 1. Proc Natl Acad Sci U S A 2006; 103:7670-5. [PMID: 16679408 PMCID: PMC1472503 DOI: 10.1073/pnas.0602515103] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nitrogen monoxide (NO) plays a role in the cytotoxic mechanisms of activated macrophages against tumor cells by inducing iron (Fe) release. We have shown that NO-mediated Fe efflux from cells required glutathione (GSH), and we have hypothesized that a GS-Fe-NO complex was released. Hence, we studied the role of the GSH-conjugate transporter multidrug resistance-associated protein 1 (MRP1) in NO-mediated Fe efflux. MCF7-VP cells overexpressing MRP1 exhibited a 3- to 4-fold increase in NO-mediated 59Fe and GSH efflux compared with WT cells (MCF7-WT) over 4 h. Similar results were found for other MRP1-overexpressing cell types but not those expressing another drug efflux pump, P-glycoprotein. NO-mediated 59Fe and GSH efflux were temperature- and energy-dependent and were significantly decreased by the GSH-depleting agent and MRP1 transport inhibitor L-buthionine-[S,R]-sulfoximine. Other MRP1 inhibitors, MK571, probenecid, and difloxacin, significantly inhibited NO-mediated 59Fe release. EPR spectroscopy demonstrated the dinitrosyl-dithiol-Fe complex (DNIC) peak in NO-treated cells was increased by MRP1 inhibitors, indicating inhibited DNIC transport from cells. The extent of DNIC accumulation correlated with the ability of MRP1 inhibitors to prevent NO-mediated 59Fe efflux. MCF7-VP cells were more sensitive than MCF7-WT cells to growth inhibition by effects of NO, which was potentiated by L-buthionine-[S,R]-sulfoximine. These data indicate the importance of GSH in NO-mediated inhibition of proliferation. Collectively, NO stimulates Fe and GSH efflux from cells via MRP1. Active transport of NO by MRP1 overcomes diffusion that is inefficient and nontargeted, which has broad ramifications for understanding NO biology.
Collapse
Affiliation(s)
- Ralph N. Watts
- *Children's Cancer Institute Australia for Medical Research, Sydney 2031, Australia
| | - Clare Hawkins
- The Heart Research Institute, Sydney 2050, Australia; and
| | - Prem Ponka
- Lady Davis Institute, Jewish General Hospital and Department of Physiology, McGill University, Montreal, QC, Canada H3T 1E2
- To whom correspondence may be addressed. E-mail:
or
| | - Des R. Richardson
- *Children's Cancer Institute Australia for Medical Research, Sydney 2031, Australia
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
27
|
Gariboldi MB, Terni F, Ravizza R, Meschini S, Marra M, Condello M, Arancia G, Monti E. The nitroxide Tempol modulates anthracycline resistance in breast cancer cells. Free Radic Biol Med 2006; 40:1409-18. [PMID: 16631531 DOI: 10.1016/j.freeradbiomed.2005.12.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Revised: 12/09/2005] [Accepted: 12/12/2005] [Indexed: 01/19/2023]
Abstract
The occurrence of multidrug resistance (MDR) is the major obstacle to successful anthracycline-based cancer chemotherapy. In the present study, we assessed the effects of Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl, TPL), a piperidine nitroxide with growth-inhibitory properties in tumor cell lines, on a number of molecular mechanisms involved in the resistance of human breast adenocarcinoma cell lines to doxorubicin (DOX). Cytotoxicity studies in MCF-7 wildtype and their MDR variant MCF-7 Adr(R) cells showed a synergistic effect between TPL and DOX when exposure to TPL preceded or was simultaneous with DOX treatment in MCF-7 Adr(R) cells. This effect of TPL seems to be due in part to its ability to increase peroxide levels and to deplete cellular glutathione pools. In addition, TPL increased DOX accumulation in MCF-7 Adr(R) cells by interfering with P-glycoprotein-mediated DOX efflux, as evidenced using a specific antibody that recognizes the active form of the protein. TPL was also found to affect the expression levels of proteins involved in response to drug treatment (e.g., p53, bcl2, bax, p21). Taken together, our results indicate that TPL is a potential new agent that may improve the clinical effect of DOX in tumors exhibiting a MDR phenotype.
Collapse
|
28
|
Liu Z, Stevenson GD, Barrett HH, Furenlid LR, Wilson DW, Kastis GA, Bettan M, Woolfenden JM. Imaging recognition of inhibition of multidrug resistance in human breast cancer xenografts using 99mTc-labeled sestamibi and tetrofosmin. Nucl Med Biol 2006; 32:573-83. [PMID: 16026704 PMCID: PMC1402231 DOI: 10.1016/j.nucmedbio.2005.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 04/22/2005] [Accepted: 04/25/2005] [Indexed: 12/28/2022]
Abstract
BACKGROUND (99m)Tc-sestamibi (MIBI) and (99m)Tc-tetrofosmin (TF) are avid transport substrates recognized by the multidrug resistance (MDR) P-glycoprotein (Pgp). This study was designed to compare the properties of MIBI and TF in assessing the inhibition of Pgp by PSC833 in severe combined immunodeficient mice bearing MCF7 human breast tumors using SPECT imaging. METHODS Animals with drug-sensitive (MCF/WT) and drug-resistant (MCF7/AdrR) tumors were treated by PSC833 and by carrier vehicle 1 h before imaging, respectively. Dynamic images were acquired for 30 min after intravenous injection of MIBI/TF using a SPECT system, FastSPECT. The biodistribution of MIBI and TF was determined at the end of the imaging session. RESULTS MCF7/WT in the absence and presence of PSC833 could be visualized by MIBI and TF imaging within 5 min and remained detectable for 30 min postinjection. MCF7/AdrR could be visualized only 2-5 min without PSC833 treatment but could be detected for 30 min with PSC833, very similar to MCF7/WT. MCF7/AdrR without PSC833 showed significantly greater radioactive washout than MCF7/WT and MCF7/AdrR with PSC833 treatment. PSC833 increased the accumulation (%ID/g) in MCF7/AdrR 3.0-fold (1.62+/-0.15 vs. 0.55+/-0.05, P<.05) for TF and 1.9-fold (1.21+/-0.04 vs. 0.64+/-0.05, P<.05) for MIBI but did not affect MCF7/WT. CONCLUSIONS The feasibility of MIBI and TF for assessment of MDR expression and inhibition was demonstrated in mice through FastSPECT imaging. The results indicate that TF may be at least comparable with MIBI in recognizing Pgp expression and modulation.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adenocarcinoma/diagnostic imaging
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Animals
- Blotting, Western
- Breast Neoplasms/diagnostic imaging
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Cyclosporins
- Doxorubicin/therapeutic use
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Female
- Humans
- Metabolic Clearance Rate
- Mice
- Mice, SCID
- Organ Specificity
- Organophosphorus Compounds/pharmacokinetics
- Organotechnetium Compounds/pharmacokinetics
- Radiopharmaceuticals/pharmacokinetics
- Technetium Tc 99m Sestamibi/pharmacokinetics
- Tissue Distribution
- Tomography, Emission-Computed, Single-Photon/methods
- Transplantation, Heterologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Zhonglin Liu
- Department of Radiology, The University of Arizona, Tucson, AZ 85724-5067, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Karmakova T, Feofanov A, Pankratov A, Kazachkina N, Nazarova A, Yakubovskaya R, Lebedeva V, Ruziyev R, Mironov A, Maurizot JC, Vigny P. Tissue distribution and in vivo photosensitizing activity of 13,15-[N-(3-hydroxypropyl)]cycloimide chlorin p6 and 13,15-(N-methoxy)cycloimide chlorin p6 methyl ester. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2006; 82:28-36. [PMID: 16236520 DOI: 10.1016/j.jphotobiol.2005.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Revised: 08/08/2005] [Accepted: 08/16/2005] [Indexed: 11/21/2022]
Abstract
Photosensitizers 13,15-[N-(3-hydroxypropyl)]cycloimide chlorin p6 (HPC) and 13,15-(N-methoxy)cycloimide chlorin p6 methyl ester (MMC) absorb at 711 nm and possess high photoinduced cytotoxicity in vitro. Here we report, that photodynamic therapy with HPC and MMC provide considerable antitumor effect in mice bearing subcutaneous P338 lymphoma. The highest antitumor effect was achieved at a dose of 4 micromol/kg when 1.5 h delay between dye injection and light irradiation (drug-light interval) was used. According to the confocal spectral imaging studies of tissue sections this drug-light interval corresponds to a maximum of tumor accumulation of MMC and HPC (tumor to skin accumulation ratio is 8-10). Short (15 min) drug-light interval can be used for efficient vasculature-targeted photodynamic therapy with HPC at a dose of 1 micromol/kg, whereas MMC is ineffective at the short drug-light interval. Relationships between the features of tissue distribution and efficacy of photodynamic therapy at different drug-light intervals are discussed for HPC and MMC.
Collapse
Affiliation(s)
- Tatyana Karmakova
- Hertsen Moscow Oncological Institute, 2nd Botkinskiy pr., 3, Moscow 125284, Russia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Stehfest E, Torky A, Glahn F, Foth H. Non-destructive micromethod for MRP1 functional assay in human lung tumor cells. Arch Toxicol 2005; 80:125-33. [PMID: 16374597 DOI: 10.1007/s00204-005-0017-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Accepted: 07/13/2005] [Indexed: 12/20/2022]
Abstract
Defense against toxic endo- and xenobiotics is a major concern of all living species and ABC transporters play a vital role in this defense system. Multidrug resistance associated proteins 1 (MRP1) is a cellular detoxifying factor supposed to transport a wide range of compounds across cell membranes either as GSH conjugates or as co-transport accompanying glutathione transposition. The cellular localization of MRP1 is a determining factor whether the transport function can take place. In this study we have undertaken experiments on the transport activity of MRP1 in cultured human lung tumor cells in order to check whether MRP1 is expressed as a functionally active protein. For this purpose we have adapted a quantitative fluorescence imaging assay to conditions where a small number of attached cells should be repeatedly measured by a non-destructive method. In cultured A549, H358 and H322 cells MRP1 is located in the cell membrane as observed by immunocytochemistry. Efflux of 5,6-carboxy-2'-7'-dichloro-fluorescein (CDF) from lung cells was sensitive toward the MRP1 inhibitor MK571 while verapamil had no effect. On the other hand, efflux of Rhodamin 123, a Pgp-glycoprotein substrate, from lung cells reacted to inhibition by verapamil, while MK571 had no effect. Modulation of glutathion content of lung cells by N-acetyl cystein and buthionine sulfoximine shifted CDF efflux toward higher or lower rates, respectively. These experiments confirm that MRP1 function can be followed in the attached cells in vitro under non-toxic concentrations of the substrates without the need to harvest and destroy the cells.
Collapse
Affiliation(s)
- Ekkehard Stehfest
- Institute of Environmental Toxicology, University of Halle, Franzosenweg 1a, 06097 Halle/Saale, Germany
| | | | | | | |
Collapse
|
31
|
Zhou XF, Coburn RA, Morris ME. Effects of New 4-Aryl-1,4-Dihydropyridines and 4-Arylpyridines on Drug Efflux Mediated by Multidrug Resistance-Associated Protein 1. J Pharm Sci 2005; 94:2256-65. [PMID: 16136554 DOI: 10.1002/jps.20406] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The purpose of this study is to evaluate the effects of newly synthesized 4-aryl-1,4-dihydropyridine and pyridines on drug efflux mediated by multidrug resistance-associated protein 1 (MRP1, ABCC1). These compounds were designed to maximize inhibition of P-glycoprotein and minimize calcium channel binding activity, based on structure modifications of niguldipine. A [3H]vinblastine accumulation study was conducted in human small cell lung cancer H69AR (overexpressing MRP1) and wild type H69 cells. Five out of 16 dihydropyridines and 6 out of 9 pyridines were found to significantly increase the intracellular accumulation of vinblastine in resistant H69AR cells (p<0.01) at a concentration of 2.5 microM. Daunomycin accumulation studies, determined using a flow cytometric assay, were also performed in H69AR and human pancreatic adenocarcinoma Panc-1 cells and the results were highly correlated with those obtained from the [3H]vinblastine accumulation studies. Four compounds, which significantly increased vinblastine accumulation, were tested for their effect on daunomycin cytotoxicity in H69AR cells and found to significantly decrease the IC50 of daunomycin, confirming the accumulation study results. Compounds were also tested for their effect on intracellular glutathione (GSH) concentrations, a cosubstrate for MRP1-mediated efflux in H69AR and Panc-1 cells. No significant changes in the intracellular GSH level were observed in H69AR cells after treatment with these test compounds. However, following a 2-hr and 24-hr incubation with a dihydropyridine compound, Im, and its pyridine derivative IIm, there was a small (approximately 20%) but statistically significant decrease in intracellular GSH in Panc-1 cells. Our results indicate that some dihydropyridine and pyridine compounds in our series could inhibit MRP1-mediated transport and that GSH modulation plays a minor, if any, role in this effect.
Collapse
Affiliation(s)
- Xiao-Fei Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, New York 14260-1200, USA
| | | | | |
Collapse
|
32
|
Wang J, Hu Z, Ye Z. Effect of trastuzumab in combination with IFN alpha-2b on HER2 and- MRP1 of ACHN. Curr Med Sci 2005; 25:326-8. [PMID: 16201286 DOI: 10.1007/bf02828157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Indexed: 11/29/2022]
Abstract
To study the effect of Trastuzumab in combination with IFN alpha-2b on HER2 and MRP1 of ACHN in vitro, ACHN cell line of RCC was cultured by employing cell culture. The tetrazolium-based colorimetric assay was used to evaluate the growth-inhibiting effect of Trastuzumab with IFN alpha-2b. SP method was utilized to determine the expression of HER2 and MRP1 of the cells. Our results showed that Trastuzumab had inhibitory effect on the growth of renal tumor cells and reversing effect on the multi-drug-resistance (MDR) in RCC in a time- and dose-dependent manner. Treated with Trastuzumab with or without IFN alpha-2b, the expression of HER2 and MRP1 genes of RCC was decreased significantly (P<0.05). It was concluded that Trastuzumab with IFN alpha-2b could inhibit the proliferation of RCC and the expression of HER2 and MRP1 of ACHN and to some extent, reverse the MDR of the tumor cells.
Collapse
Affiliation(s)
- Jiajing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | |
Collapse
|
33
|
Villa AM, Doglia SM. Mitochondria in tumor cells studied by laser scanning confocal microscopy. JOURNAL OF BIOMEDICAL OPTICS 2004; 9:385-394. [PMID: 15065906 DOI: 10.1117/1.1646414] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We present here a confocal fluorescence microscopy study of mitochondria in sensitive and resistant carcinoma cells by using two potentiometric probes of mitochondria, rhodamine 123 (R123) and dimethylaminostyryl-methylpyridiniumiodine. We have found that active mitochondria in sensitive MCF-7 and multidrug resistant MCF-7/DX carcinoma cells are very different in localization and morphology. In sensitive cells active mitochondria are found in the perinuclear region, whereas in the multidrug resistance (MDR) subline they are confined to the cell periphery. Interestingly, the MDR revertant verapamil has been found to restore in MCF-7/DX cells the same pattern of active mitochondria seen in sensitive cells. We have also studied R123 in human lung carcinoma A549 cells, which display a low responsivity to doxorubicin, and overexpress the lung resistance-related protein. In addition to perinuclear mitochondria, peripheral mitochondria with weaker fluorescence can be seen in this cell line. Interestingly, in the two examined carcinoma lines we have been able to recognize by image analysis a common new star-lobed morphology. Our results indicate that in resistant carcinoma cells two populations of mitochondria coexist with different localization, morphology, and activity.
Collapse
Affiliation(s)
- Anna Maria Villa
- INFM-UdR Milano Bicocca and Dipartimento di Biotecnologie e Bioscienze, Universita degli Studi di Milano Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
| | | |
Collapse
|
34
|
Gong Y, Duvvuri M, Krise JP. Separate Roles for the Golgi Apparatus and Lysosomes in the Sequestration of Drugs in the Multidrug-resistant Human Leukemic Cell Line HL-60. J Biol Chem 2003; 278:50234-9. [PMID: 14522995 DOI: 10.1074/jbc.m306606200] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sequestration of drugs away from cellular target sites into cytoplasmic organelles of multidrug-resistant (MDR) cancer cells has been recently shown to be a cause for ineffective drug therapy. This process is poorly understood despite the fact that it has been observed in a large number of MDR cancer cell lines. Analysis of drug sequestration in these cells has traditionally been done using fluorescent anthracycline antibiotics (i.e. daunorubicin, doxorubicin). This narrow selection of substrates has resulted in a limited understanding of sequestration mechanisms and the intracellular compartments that are involved. To better characterize this phenotype, we chose to examine the sequestration of molecules having different acid/base properties in the MDR HL-60 human leukemic cell line. Here we show that weakly basic drug daunorubicin is sequestered into lysosomes according to a pH partitioning type mechanism, whereas sulforhodamime 101, a zwitterionic molecule, is sequestered into the Golgi apparatus through a drug transporter-mediated process. Quantitative intracellular pH measurements reveal that the lysosome-tocytosol pH gradient is expanded in the MDR line. Moreover, the MDR cells overexpress the multidrug resistance-related protein (MRP1), which is localized to the Golgi apparatus. These results demonstrate, for the first time, that two distinct mechanisms for intracellular compartmentalization are operational in a single MDR cell line.
Collapse
Affiliation(s)
- Yuping Gong
- Division of Drug Delivery and Disposition, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | |
Collapse
|
35
|
Abstract
Drug resistance, intrinsic or acquired, is a problem for all chemotherapeutic agents. In this review, we examine numerous strategies that have been tested or proposed to reverse drug resistance. Included among these strategies are approaches targeting the apoptosis pathway. Although the process of apoptosis is complex, it provides several potential sites for therapeutic intervention. A variety of targets and approaches are being pursued, including the suppression of proteins inhibiting apoptosis using antisense oligonucleotides (ASOs), and small molecules targeted at proteins that modulate apoptosis. An alternate strategy is based on numerous studies that have documented methylation of critical regions in the genome in human cancers. Consequently, efforts have been directed at re-expressing genes, including genes that affect drug sensitivity, using 5-azacytidine and 2'-deoxy-5-azacytidine (DAC, decitabine) as demethylating agents. While this strategy may be effective as a single modality, success will most likely be achieved if it is used to modulate gene expression in combination with other modalities such as chemotherapy. At a more basic level, attempts have been made to modulate glutathione (GSH) levels. Owing to its reactivity and high intracellular concentrations, GSH has been implicated in resistance to several chemotherapeutic agents. Several approaches designed to deplete intracellular GSH levels have been pursued including the use of buthionine-(S,R)-sulfoxime (BSO), a potent and specific inhibitor of gamma-glutamyl cysteine synthetase (gamma-GCS), the rate-limiting step in the synthesis of GSH, a hammerhead ribozyme against gamma-GCS mRNA to downregulate specifically its levels and targeting cJun expression to reduce GSH levels. Alternate strategies have targeted p53. The frequent occurrence of p53 mutations in human cancer has led to the development of numerous approaches to restore wild-type (wt) p53. The goals of these interventions are to either revert the malignant phenotype or enhance drug sensitivity. The approach most extensively investigated has utilized one of several viral vectors. An alternate approach, the use of small molecules to restore wt function to mutant p53, remains an option. Finally, the conceptually simplest mechanism of resistance is one that reduces intracellular drug accumulation. Such reduction can be effected by a variety of drug efflux pumps, of which the most widely studied is P-glycoprotein (Pgp). The first strategy utilized to inhibit Pgp function relied on the identification of non-chemotherapeutic agents as competitors. Other approaches have included the use of hammerhead ribozymes against the MDR-1 gene and MDR-1-targeted ASOs. Although modulation of drug resistance has not yet been proven to be an effective clinical tool, we have learned an enormous amount about drug resistance. Should we succeed, these pioneering basic and clinical studies will have paved the road for future developments.
Collapse
Affiliation(s)
- Tito Fojo
- Center for Cancer Research, National Cancer Institute, Building 10, Room 12-C-103, 9000 Rockville Pike, Bethesda, MA 20892, USA.
| | | |
Collapse
|
36
|
Veuger MJT, Honders MW, Spoelder HE, Willemze R, Barge RMY. Inactivation of deoxycytidine kinase and overexpression of P-glycoprotein in AraC and daunorubicin double resistant leukemic cell lines. Leuk Res 2003; 27:445-53. [PMID: 12620296 DOI: 10.1016/s0145-2126(02)00224-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
AraC resistance in vitro is explained by inactivation of dCK, while resistance to DNR is described by overexpression of multidrug efflux pumps like Pgp or MRP. Thus far, no correlation between resistance mechanisms in vitro and in patients with AML has been documented. We generated AraC and DNR double resistant cell lines to investigate resistance mechanisms of both agents. In these cell lines involvement of dCK was extensively investigated and Pgp expression and activity was determined. Our data implicate that similar resistance mechanisms like inactivation of dCK coincided by alternatively spliced dCK forms and overexpression of Pgp are induced in single-as well as in double resistant leukemic cell lines.
Collapse
Affiliation(s)
- Marjan J T Veuger
- Laboratory of Experimental Hematology, C2-R, Leiden University Medical Center, Department of Hematology, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
37
|
Zhang S, Morris ME. Effects of the flavonoids biochanin A, morin, phloretin, and silymarin on P-glycoprotein-mediated transport. J Pharmacol Exp Ther 2003; 304:1258-67. [PMID: 12604704 DOI: 10.1124/jpet.102.044412] [Citation(s) in RCA: 236] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Flavonoids are constituents of fruits, vegetables, and plant-derived beverages, as well as components in herbal-containing dietary supplements. The objective of this investigation was to characterize the effect of flavonoids on P-glycoprotein (P-gp)-mediated cellular efflux and to determine the molecular mechanism(s) of the flavonoid-drug interaction. Studies were conducted in the sensitive and multidrug resistant human breast cancer cell lines MCF-7 and MDA435/LCC6 and examined the effects of the flavonoids biochanin A, morin, phloretin, and silymarin on daunomycin (DNM) accumulation and doxorubicin cytotoxicity. The potential mechanism(s) involved in the interaction was evaluated by determining flavonoid effects on 1) P-gp ATPase activity, 2) [(3)H]azidopine photoaffinity labeling of P-gp, and 3) cellular P-gp levels. The flavonoids increased [(3)H]DNM accumulation in P-gp positive cells, but not P-gp negative cells, and these effects were both flavonoid concentration- and P-gp expression level-dependent. Biochanin A and silymarin potentiated doxorubicin cytotoxicity in P-gp positive cells. Biochanin A and phloretin stimulated, whereas morin and silymarin inhibited P-gp ATPase activity, confirming that these flavonoids interact with P-gp. Morin and silymarin significantly inhibited [(3)H]azidopine photoaffinity labeling of P-gp, suggesting a direct interaction with P-gp substrate binding. A 24-h preincubation with all flavonoids, followed by flavonoid removal, did not alter cellular P-gp level in P-gp positive cells. In conclusion, biochanin A, morin, phloretin, and silymarin all inhibited P-gp-mediated cellular efflux and the mechanism of the interaction involved, at least in part, a direct interaction. The findings of this study indicate a potential for significant flavonoid-drug interactions with P-gp substrates.
Collapse
Affiliation(s)
- Shuzhong Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, New York, USA
| | | |
Collapse
|
38
|
Mercatante DR, Mohler JL, Kole R. Cellular response to an antisense-mediated shift of Bcl-x pre-mRNA splicing and antineoplastic agents. J Biol Chem 2002; 277:49374-82. [PMID: 12381725 DOI: 10.1074/jbc.m209236200] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Overexpression of Bcl-xL, an anti-apoptotic member of the Bcl-2 family, negatively correlates with the sensitivity of various cancers to chemotherapeutic agents. We show here that high levels of expression of Bcl-xL promoted apoptosis of cells treated with an antisense oligonucleotide (5'Bcl-x AS) that shifts the splicing pattern of Bcl-x pre-mRNA from the anti-apoptotic variant, Bcl-xL, to the pro-apoptotic variant, Bcl-xS. This surprising finding illustrates the advantage of antisense-induced modulation of alternative splicing versus down-regulation of targeted genes. It also suggests a specificity of the oligonucleotide effects since non-cancerous cells with low levels of Bcl-xL should resist the treatment. 5'Bcl-x AS sensitized cells to several antineoplastic agents and radiation and was effective in promoting apoptosis of MCF-7/ADR cells, a breast cancer cell line resistant to doxorubicin via overexpression of the mdr1 gene. Efficacy of 5'Bcl-x AS combined with chemotherapeutic agents in the PC3 prostate cancer cell line may be translated to clinical prostate cancer since recurrent prostate cancer tissue samples expressed higher levels of Bcl-xL than benign prostate tissue. Treatment with 5'Bcl-x AS may enhance the efficacy of standard anti-cancer regimens and should be explored, especially in recurrent prostate cancer.
Collapse
Affiliation(s)
- Danielle R Mercatante
- UNC Lineberger Comprehensive Cancer Center and Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599-7295, USA
| | | | | |
Collapse
|
39
|
Jin J, Huang M, Wei HL, Liu GT. Mechanism of 5-fluorouracil required resistance in human hepatocellular carcinoma cell line Bel 7402. World J Gastroenterol 2002; 8:1029-34. [PMID: 12439919 PMCID: PMC4656374 DOI: 10.3748/wjg.v8.i6.1029] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the resistance mechanism of 5-fluorouracil (5-FU) in Bel7402/5-FU cells which was established in our lab by in vitro continuous stepwise exposure of human hepatocellular carcinoma (HCC) cell line Bel7402 to 5-FU.
METHODS: The expression of multidrug resistance-associated protein (MRP) and thymidylate synthase (TS) in Bel7402 cells was detected by immonocytochemistry. The fluorescein (FLU) accumulation, an index of MRP functional activity, was determined by flow cytometry. The distribution of FLU was observed by confocal laser scanning microscope. The spectrofluorometry was used to show the intracelluar content of glutathione (GSH). Cell growth inhibition was determined by MTT assay. The activity of glutathione S-transferases (GSTs) was determined by spectrophotometry.
RESULTS: A higher expression of MRP in the Bel7402/5-FU cells was observed by using monoclonal mouse anti-MRP antibody, MRPr-1, in comparison with Bel7402 cells. Bel7402/5-FU cells also showed a significant decrease of FLU accumulation. FLU mainly accumulated in the nucleus with a high nuclear/cytoplasmic ratio in Bel7402 cells, whereas there was no difference of FLU accumulation between the nucleus and cytoplasm in Bel7402/5-FU cells. The intracellular GSH content in Bel7402/5-FU cells was almost 3 folds higher than that in Bel7402 cells. Addition of D, L-buthione-S, R-sulfoximine (BSO) dose-dependently reduced the GSH content in Bel7402/ 5-FU cells, however, only a weak enhancement on the cytotoxicity of 5-FU and doxorubicin (Dox) to Bel7402/5-FU cells was observed. Bel7402/5-FU cells also exhibited 29.1% higher total GSTs activity than Bel7402 cells. Immunocytochemical staining by using anti-TS monoclonal antibody TS 106 showed that the level of TS in Bel7402/5-FU cells elevated markedly as compared with Bel7402 cells.
CONCLUSION: The continuous exposure of Bel7402 cells to 5-FU led to overexpression of TS and MRP, as well as increased intracellular GSH content and total GST activity.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmacology, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
40
|
Liem AA, Chamberlain MP, Wolf CR, Thompson AM. The role of signal transduction in cancer treatment and drug resistance. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2002; 28:679-84. [PMID: 12359208 DOI: 10.1053/ejso.2002.1303] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Drug resistance in the treatment of cancer still remains a major clinical challenge, in part due to an insufficient understanding of the pathways by which these drugs interact with the mechanisms underlying cellular behaviour and cancer pathogenesis. Signal transduction involves cell differentiation, proliferation and cell death with alterations in these mechanisms being involved in the pathogenesis of cancer. It has been postulated that such pathways could be linked to anti-cancer drug resistance. Recently, novel approaches to overcome anti-cancer drug resistance through manipulation of signal transduction pathways, have been introduced in clinical trials. In this article we present a review of the current understanding in the field of signal transduction and the existing evidence for its role in drug resistance. We also discuss its clinical relevance with regard to overcoming drug resistance.
Collapse
Affiliation(s)
- A A Liem
- Department of Surgery and Molecular Oncology, University of Dundee, UK
| | | | | | | |
Collapse
|
41
|
Yang JY, Luo HY, Lin QY, Liu ZM, Yan LN, Lin P, Zhang J, Lei S. Subcellular daunorubicin distribution and its relation to multidrug resistance phenotype in drug-resistant cell line SMMC-7721/R. World J Gastroenterol 2002; 8:644-9. [PMID: 12174371 PMCID: PMC4656313 DOI: 10.3748/wjg.v8.i4.644] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the correlation between subcellular daunorubicin distribution and the multidrug resistance phenotype in drug-resistant cell line SMMC-7721/R.
METHODS: The multidrug resistant cell line SMMC-7721/R, a human hepatocellular carcinoma cell line, was established. Antisense oligonucleotides (AS-ODN) were used to obtain different multidrug resistance phenotypes by inhibiting the expression of mdr1 gene and/or multidrug resistance-related protein gene (mrp) using Lipofectamine as delivery agent. Expression of mdr1 and mrp genes was evaluated by RT-PCR and Western blotting. Intracellular daunorubicin (DNR) concentration was measured by flow cytometry. Subcellular DNR distribution was analyzed by confocal laser scanning microscopy. Adriamycin (ADM) and DNR sensitivity was examined by MTT method.
RESULTS: Low level expression of mdr1 and mrp mRNAs and no expression of P-Glycoprotein (P-gp) and multidrug resistance-related protein (P190) were detected in parental sensitive cells SMMC-7721/S, but over-expression of these two genes was observed in drug-resistant cell SMMC-7721/R. The expression of mdr1 and mrp genes in SMMC-7721/R cells was down-regulated to the level in the SMMC-7721/S cells by AS-ODN. Intracellular DNR concentration in SMMC-7721/S cells was 10 times higher than that in SMMC-7721/R cells. In SMMC7721/S cells intracellular DNR distributed evenly in the nucleus and cytoplasm, while in SMMC-7721/R cells DNR distributed in a punctate pattern in the cytoplasm and was reduced in the nucleus. DNR concentration in SMMC-7721/R cells co-transfected with AS-ODNs targeting to mdr1 and mrp mRNAs recovered to 25 percent of that in SMMC7721/S cells. Intracellular DNR distribution pattern in drug-resistant cells treated by AS-ODN was similar to drug-sensitive cell, and the cells resistance index (RI) to DNR and ADM decreased at most from 88.0 and 116.0 to 4.0 and 2.3, respectively. Co-Transfection of two AS-ODNs showed a stronger synergistic effect than separate transfection.
CONCLUSIONS: P-gp and P190 are two members mediating MDR in cell line SMMC7721/R. Intracellular drug concentration increase and subcellular distribution change are two important factors in multidrug resistance (MDR) formation. The second factor, drugs transport by P-gp and P190 from cell nucleus to organell in cytoplasm, may play a more important role.
Collapse
Affiliation(s)
- Jia-Yin Yang
- Department of General surgery, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou 310003, China.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chauvier D, Kegelaer G, Morjani H, Manfait M. Reversal of multidrug resistance-associated protein-mediated daunorubicin resistance by camptothecin. J Pharm Sci 2002; 91:1765-75. [PMID: 12115804 DOI: 10.1002/jps.10162] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The multidrug-resistance (MR) status of camptothecin (CPT) was investigated in colon adenocarcinoma HT29 cells, leukemia K562, and breast carcinoma MCF7 cells expressing P-glycoprotein (Pgp) and/or MR-associated protein (MRP1). The concentration that induced 50% growth inhibition (IC(50)) against CPT was 0.14 and 0.20 microM in parental K562/WT and MCF7/WT cells, respectively. The drug resistant subline KH30 and MCF7/VP cells, which both overexpress MRP1, presented IC(50) values of 0.63 and 3.10 microM, respectively. The resulting resistance indexes were 3.80 and 12.50, respectively. However, in KH300 cells, a cell line that preferentially overexpresses Pgp, the IC(50) of CPT was 0.08 microM and thus did not exhibit resistance against CPT. In MCF7/DoX cells, preferentially overexpressing Pgp, but also a significant level of MRP1, the IC(50) of CPT was 0.64 microM and thus presented a resistance index of 3.26 against CPT. The cytotoxic effect of CPT was modulated in cells expressing MRP1 (MCF7/VP, HT29 cells) by the specific MRP1 modulators, probenecid and MK571. These results led us to consider CPT as a substrate for MRP1 and a potential modulator of MRP1 activity. To test this hypothesis, we examined the ability of nontoxic concentrations of CPT to sensitize MRP1-overexpressing cells to daunorubicin (DNR). In MCF7/VP and KH30 cells, nontoxic concentrations of CPT were able to enhance cytotoxicity of DNR and its nuclear accumulation. Sequential and simultaneous associations of CPT (100 nM) and DNR provided complete reversal of resistance, thus showing a synergistic effect in KH30 cells. However, simultaneous association (with 10 or 20 nM CPT) had an additive effect in MCF7/VP. These data suggest that CPT could be proposed as a candidate for the reversal of the MRP1 phenotype at clinically achievable concentrations.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/metabolism
- Antibiotics, Antineoplastic/pharmacology
- Antineoplastic Agents, Phytogenic/pharmacology
- Breast Neoplasms/pathology
- Camptothecin/pharmacology
- Cell Line
- Cell Nucleus/metabolism
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Daunorubicin/metabolism
- Daunorubicin/pharmacology
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia/pathology
- Microscopy, Confocal
- Reverse Transcriptase Polymerase Chain Reaction
- Tetrazolium Salts
- Thiazoles
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- David Chauvier
- Unité MéDIAN, CNRS UMR 6142, UFR de Pharmacie, IFR53, 51 rue Cognacq Jay, 51096 Reims, France
| | | | | | | |
Collapse
|
43
|
Ballinger JR. 99m
Tc‐Tetrofosmin for Functional Imaging of P‐glycoprotein Modulation In Vivo. J Clin Pharmacol 2001. [DOI: 10.1177/0091270001417007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Ballinger JR. Imaging multidrug resistance with radiolabeled substrates for P-glycoprotein and multidrug resistance protein. Cancer Biother Radiopharm 2001; 16:1-7. [PMID: 11279792 DOI: 10.1089/108497801750095907] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
45
|
Muzzammil T, Moore MJ, Hedley D, Ballinger JR. Comparison of (99m)Tc-sestamibi and doxorubicin to monitor inhibition of P-glycoprotein function. Br J Cancer 2001; 84:367-73. [PMID: 11161402 PMCID: PMC2363739 DOI: 10.1054/bjoc.2000.1621] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
P-glycoprotein (Pgp) overexpression is a well-recognized factor in resistance to chemotherapy. Doxorubicin flow cytometry is used to monitor Pgp function in haematological specimens and biopsies from other cancers, and radionuclide imaging with sestamibi has recently shown promise for non-invasive monitoring. In the present study the two methods were directly compared in single-cell suspensions of three variants of the human breast carcinoma cell line MCF7: sensitive MCF7/WT, doxorubicin-selected MCF7/AdrR, and MDR1-gene-transfected MCF7/BC19 cells with doxorubicin resistance factors of 1, 192, and 14, respectively. Accumulation of sestamibi and mean fluorescence of doxorubicin (5.5 microM) were assessed over 60 min in the presence and absence of Pgp modulators GG918 (0.01 to 0.2 microM) and PSC833 (0.05 to 2.0 microM). Accumulation curves for sestamibi and doxorubicin differed among the cell variants under control conditions, with sestamibi showing a significantly greater difference between WT and resistant cells than doxorubicin. Both GG918 and PSC833 reversed uptake deficits to WT levels for sestamibi in MCF7/BC19 cells and doxorubicin in MCF7/BC19 and MCF7/AdrR cells, but failed to show the same effect for sestamibi in MCF7/AdrR cells (approximately 30% of MCF7/WT level). Thus, both methods clearly distinguished sensitive from resistant MCF7 variants, with the radionuclide method showing greater sensitivity.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/drug effects
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Acridines/pharmacology
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Blotting, Western
- Cell Survival/drug effects
- Cyclosporins/pharmacology
- Dose-Response Relationship, Drug
- Doxorubicin/pharmacokinetics
- Doxorubicin/pharmacology
- Genes, MDR/genetics
- Genes, MDR/physiology
- Humans
- Immunohistochemistry
- Isoquinolines/pharmacology
- Radiopharmaceuticals/pharmacokinetics
- Technetium Tc 99m Sestamibi/pharmacokinetics
- Tetrahydroisoquinolines
- Time Factors
- Tumor Cells, Cultured/cytology
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
Collapse
Affiliation(s)
- T Muzzammil
- Department of Pharmaceutical Sciences, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada
| | | | | | | |
Collapse
|
46
|
Muzzammil T, Moore MJ, Ballinger JR. In vitro comparison of sestamibi, tetrofosmin, and furifosmin as agents for functional imaging of multidrug resistance in tumors. Cancer Biother Radiopharm 2000; 15:339-46. [PMID: 11041018 DOI: 10.1089/cbr.2000.15.339] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sestamibi, tetrofosmin, and furifosmin are 99mTc-labeled myocardial perfusion imaging agents which have been shown to be substrates for P-glycoprotein (Pgp), the multidrug-resistance transporter which is overexpressed in some tumors. The three tracers were directly compared in vitro in the human breast cancer cell line MCF7-WT and two multidrug-resistant variants, MCF7-BC19 (MDR1 gene transfected) and MCF7-AdrR (doxorubicin selected). Tracer accumulation over the course of 60 minutes was determined. Dose-response curves were generated for two modulators of Pgp function, GG918 and PSC833. The general shape of accumulation curves for the three tracers in MCF7-WT cells was similar, with accumulation levels being sestamibi > tetrofosmin > furifosmin. Accumulation of sestamibi and furifosmin in MCF7-BC19 cells was reduced to 10% and 21% of MCF7-WT levels, respectively, but this accumulation deficit could be completely reversed by addition of 0.1 microM GG918 or 2 microM PSC833. Accumulation of sestamibi and tetrofosmin in MCF7-AdrR cells was 1.6% and 12% of MCF7-WT levels, respectively, and could only be enhanced to 30% and 45% of MCF7-WT levels by addition of GG918 or PSC833. In contrast, furifosmin showed similar levels of accumulation in MCF7-WT and MCF7-BC19 cells, slightly lower levels in MCF7-AdrR cells, and no consistent response to Pgp modulators. These results support the continued investigation of sestamibi and tetrofosmin as agents for functional imaging of multidrug resistance in human cancer.
Collapse
Affiliation(s)
- T Muzzammil
- Department of Pharmaceutical Sciences, University of Toronto, Canada
| | | | | |
Collapse
|