1
|
Lv N, Huang C, Huang H, Dong Z, Chen X, Lu C, Zhang Y. Overexpression of Glutathione S-Transferases in Human Diseases: Drug Targets and Therapeutic Implications. Antioxidants (Basel) 2023; 12:1970. [PMID: 38001822 PMCID: PMC10668987 DOI: 10.3390/antiox12111970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione S-transferases (GSTs) are a major class of phase II metabolic enzymes. Besides their essential role in detoxification, GSTs also exert diverse biological activities in the occurrence and development of various diseases. In the past few decades, much research interest has been paid to exploring the mechanisms of GST overexpression in tumor drug resistance. Correspondingly, many GST inhibitors have been developed and applied, solely or in combination with chemotherapeutic drugs, for the treatment of multi-drug resistant tumors. Moreover, novel roles of GSTs in other diseases, such as pulmonary fibrosis and neurodegenerative diseases, have been recognized in recent years, although the exact regulatory mechanisms remain to be elucidated. This review, firstly summarizes the roles of GSTs and their overexpression in the above-mentioned diseases with emphasis on the modulation of cell signaling pathways and protein functions. Secondly, specific GST inhibitors currently in pre-clinical development and in clinical stages are inventoried. Lastly, applications of GST inhibitors in targeting cell signaling pathways and intracellular biological processes are discussed, and the potential for disease treatment is prospected. Taken together, this review is expected to provide new insights into the interconnection between GST overexpression and human diseases, which may assist future drug discovery targeting GSTs.
Collapse
Affiliation(s)
- Ning Lv
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chunyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Haoyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Zhiqiang Dong
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
| | - Xijing Chen
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chengcan Lu
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
- Jiangning Clinical Medical College, Jiangsu University, Nanjing 211100, China
| | - Yongjie Zhang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| |
Collapse
|
2
|
Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: Beyond pharmacological inhibition. Drug Resist Updat 2021; 59:100795. [PMID: 34983733 DOI: 10.1016/j.drup.2021.100795] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/30/2022]
Abstract
Resistance to chemotherapy remains one of the most significant obstacles to successful cancer treatment. While inhibiting drug efflux mediated by ATP-binding cassette (ABC) transporters is a seemingly attractive and logical approach to combat multidrug resistance (MDR), small molecule inhibition of ABC transporters has so far failed to confer clinical benefit, despite considerable efforts by medicinal chemists, biologists, and clinicians. The long-sought treatment to eradicate cancers displaying ABC transporter overexpression may therefore lie within alternative targeting strategies. When aberrantly expressed, the ABC transporter multidrug resistance-associated protein 1 (MRP1, ABCC1) confers MDR, but can also shift cellular redox balance, leaving the cell vulnerable to select agents. Here, we explore the physiological roles of MRP1, the rational for targeting this transporter in cancer, the development of small molecule MRP1 inhibitors, and the most recent developments in alternative therapeutic approaches for targeting cancers with MRP1 overexpression. We discuss approaches that extend beyond simple MRP1 inhibition by exploiting the collateral sensitivity to glutathione depletion and ferroptosis, the rationale for targeting the shared transcriptional regulators of both MRP1 and glutathione biosynthesis, advances in gene silencing, and new molecules that modulate transporter activity to the detriment of the cancer cell. These strategies illustrate promising new approaches to address multidrug resistant disease that extend beyond the simple reversal of MDR and offer exciting routes for further research.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Verma H, Singh Bahia M, Choudhary S, Kumar Singh P, Silakari O. Drug metabolizing enzymes-associated chemo resistance and strategies to overcome it. Drug Metab Rev 2019; 51:196-223. [DOI: 10.1080/03602532.2019.1632886] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Himanshu Verma
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shalki Choudhary
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Pankaj Kumar Singh
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- MolecularModelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
4
|
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
|
5
|
Drug metabolizing enzymes and their inhibitors' role in cancer resistance. Biomed Pharmacother 2018; 105:53-65. [PMID: 29843045 DOI: 10.1016/j.biopha.2018.05.117] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 12/15/2022] Open
Abstract
Despite continuous research on chemotherapeutic agents, different mechanisms of resistance have become a major pitfall in cancer chemotherapy. Although, exhaustive efforts are being made by several researchers to target resistance against chemotherapeutic agents, there is another class of resistance mechanism which is almost carrying on unattended. This class of resistance includes pharmacokinetics resistance such as efflux by ABC transporters and drug metabolizing enzymes. ABC transporters are the membrane bound proteins which are responsible for the movement of substrates through the cell membrane. Drug metabolizing enzymes are an integral part of phase-II metabolism that helps in the detoxification of exogenous, endogenous and xenobiotics substrates. These include uridine diphospho-glucuronosyltransferases (UGTs), glutathione-S-transferases (GSTs), dihydropyrimidine dehydrogenases (DPDs) and thiopurine methyltransferases (TPMTs). These enzymes may affect the role of drugs in both positive as well negative manner, depending upon the type of tissue and cells present and when present in tumors, can result in drug resistance. However, the underlying mechanism of resistance by drug metabolizing enzymes is still not clear. Here, we have tried to cover various aspects of these enzymes in relation to anticancer drugs.
Collapse
|
6
|
Rodman SN, Spence JM, Ronnfeldt TJ, Zhu Y, Solst SR, O'Neill RA, Allen BG, Guan X, Spitz DR, Fath MA. Enhancement of Radiation Response in Breast Cancer Stem Cells by Inhibition of Thioredoxin- and Glutathione-Dependent Metabolism. Radiat Res 2016; 186:385-395. [PMID: 27643875 DOI: 10.1667/rr14463.1] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The goal of this study was to determine if depletion of glutathione (GSH) and inhibition of thioredoxin (Trx) reductase (TrxR) activity could enhance radiation responses in human breast cancer stem cells by a mechanism involving thiol-dependent oxidative stress. The following were used to inhibit GSH and Trx metabolism: buthionine sulfoximine (BSO), a GSH synthesis inhibitor; sulfasalazine (SSZ), an inhibitor of xc- cysteine/glutamate antiporter; auranofin (Au), a thioredoxin reductase inhibitor; or 2-AAPA, a GSH-reductase inhibitor. Clonogenic survival, Matrigel assays, flow cytometry cancer stem cell assays (CD44+CD24-ESA+ or ALDH1) and human tumor xenograft models were used to determine the antitumor activity of drug and radiation combinations. Combined inhibition of GSH and Trx metabolism enhanced cancer cell clonogenic killing and radiation responses in human breast and pancreatic cancer cells via a mechanism that could be inhibited by N-acetylcysteine (NAC). Au, BSO and radiation also significantly decreased breast cancer cell migration and invasion in a thiol-dependent manner that could be inhibited by NAC. In addition, pretreating cells with Au sensitized breast cancer stem cell populations to radiation in vitro as determined by CD44+CD24-ESA+ or ALDH1. Combined administration of Au and BSO, given prior to irradiation, significantly increased the survival of mice with human breast cancer xenografts, and decreased the number of ALDH1+ cancer stem cells. These results indicate that combined inhibition of GSH- and Trx-dependent thiol metabolism using pharmacologically relevant agents can enhance responses of human breast cancer stem cells to radiation both in vitro and in vivo.
Collapse
Affiliation(s)
- Samuel N Rodman
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Jacquelyn M Spence
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Tyler J Ronnfeldt
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Yueming Zhu
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240.,b Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611; and
| | - Shane R Solst
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Rebecca A O'Neill
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Bryan G Allen
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Xiangming Guan
- c Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota 57007
| | - Douglas R Spitz
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| | - Melissa A Fath
- a Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52240
| |
Collapse
|
7
|
Webster JM, Morton CA, Johnson BF, Yang H, Rishel MJ, Lee BD, Miao Q, Pabba C, Yapp DT, Schaffer P. Functional imaging of oxidative stress with a novel PET imaging agent, 18F-5-fluoro-L-aminosuberic acid. J Nucl Med 2014; 55:657-64. [PMID: 24578242 DOI: 10.2967/jnumed.113.126664] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Glutathione is the predominant endogenous cellular antioxidant, playing a critical role in the cellular defensive response to oxidative stress by neutralizing free radicals and reactive oxygen species. With cysteine as the rate-limiting substrate in glutathione biosynthesis, the cystine/glutamate transporter (system xc(-)) represents a potentially attractive PET biomarker to enable in vivo quantification of xc(-) activity in response to oxidative stress associated with disease. We have developed a system xc(-) substrate that incorporates characteristics of both natural substrates, L-cystine and L-glutamate (L-Glu). L-aminosuberic acid (L-ASu) has been identified as a more efficient system xc(-) substrate than L-Glu, leading to an assessment of a series of anionic amino acids as prospective PET tracers. Herein, we report the synthesis and in vitro and in vivo validation of a lead candidate, (18)F-5-fluoro-aminosuberic acid ((18)F-FASu), as a PET tracer for functional imaging of a cellular response to oxidative stress with remarkable tumor uptake and retention. METHODS (18)F-FASu was identified as a potential PET tracer based on an in vitro screening of compounds similar to L-cystine and L-Glu. Affinity toward system xc(-) was determined via in vitro uptake and inhibition studies using oxidative stress-induced EL4 and SKOV-3 cells. In vivo biodistribution and PET imaging studies were performed in mice bearing xenograft tumors (EL4 and SKOV-3). RESULTS In vitro assay results determined that L-ASu inhibited system xc(-) as well as or better than L-Glu. The direct comparison of uptake of tritiated compounds demonstrated more efficient system xc(-) uptake of L-ASu than L-Glu. Radiosynthesis of (18)F-FASu allowed the validation of uptake for the fluorine-bearing derivative in vitro. Evaluation in vivo demonstrated primarily renal clearance and uptake of approximately 8 percentage injected dose per gram in SKOV-3 tumors, with tumor-to-blood and tumor-to-muscle ratios of approximately 12 and approximately 28, respectively. (18)F-FASu uptake was approximately 5 times greater than (18)F-FDG uptake in SKOV-3 tumors. Dynamic PET imaging demonstrated uptake in EL4 tumor xenografts of approximately 6 percentage injected dose per gram and good tumor retention for at least 2 h after injection. CONCLUSION (18)F-FASu is a potentially useful metabolic tracer for PET imaging of a functional cellular response to oxidative stress. (18)F-FASu may provide more sensitive detection than (18)F-FDG in certain tumors.
Collapse
Affiliation(s)
- Jack M Webster
- Diagnostics and Biomedical Technologies, GE Global Research, Niskayuna, New York
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Augustine CK, Jung SH, Sohn I, Yoo JS, Yoshimoto Y, Olson JA, Friedman HS, Ali-Osman F, Tyler DS. Gene expression signatures as a guide to treatment strategies for in-transit metastatic melanoma. Mol Cancer Ther 2010; 9:779-90. [PMID: 20371714 DOI: 10.1158/1535-7163.mct-09-0764] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In-transit metastatic melanoma, which typically presents as multifocal lesions, provides a unique setting to evaluate the utility of gene signatures for defining optimal regional therapeutic strategies and assessing the efficacy of treatment. The goal of this study was to determine whether a single multifocal lesion is representative of residual tumor burden in terms of gene expression signatures predictive of response to therapy. Using microarray-based gene expression profiling, we examined 55 in-transit melanoma lesions across 29 patients with multifocal disease. Principal component analysis, unsupervised hierarchical clustering, one-way ANOVA, binary regression analysis, and gene signatures predictive of oncogenic pathway activation were used to compare patterns of gene expression across all multifocal lesions from a patient. Patterns of gene expression were highly similar (P < 0.006; average r = 0.979) across pretreatment lesions from a single patient compared with the significantly different patterns observed across patients (P < 0.05). The findings presented in this study show that individual melanoma tumor nodules in patients with multifocal disease harbor similar patterns of gene expression and a single lesion can be used to predict response to chemotherapy, evaluate the activation status of oncogenic signaling pathways, and characterize other aspects of the biology of an individual patient's disease. These results will facilitate the use of gene expression profiling in melanoma regional therapy clinical trials to not only select optimal regional chemotherapeutic agents but to also allow for a more rational identification of candidates for specific targeted therapies and evaluation of their therapeutic efficacy. Mol Cancer Ther; 9(4); 779-90. (c)2010 AACR.
Collapse
|
9
|
Dourado DFAR, Fernandes PA, Mannervik B, Ramos MJ. Glutathione Transferase A1-1: Catalytic Importance of Arginine 15. J Phys Chem B 2010; 114:1690-7. [DOI: 10.1021/jp908251z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daniel F. A. R. Dourado
- REQUIMTE/Departamento de Química Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre, 687, 4169-007 Porto, Portugal, and Department of Biochemistry and Organic Chemistry, Uppsala University, BMC Box 576, SE-75123 Uppsala, Sweden
| | - Pedro Alexandrino Fernandes
- REQUIMTE/Departamento de Química Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre, 687, 4169-007 Porto, Portugal, and Department of Biochemistry and Organic Chemistry, Uppsala University, BMC Box 576, SE-75123 Uppsala, Sweden
| | - Bengt Mannervik
- REQUIMTE/Departamento de Química Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre, 687, 4169-007 Porto, Portugal, and Department of Biochemistry and Organic Chemistry, Uppsala University, BMC Box 576, SE-75123 Uppsala, Sweden
| | - Maria João Ramos
- REQUIMTE/Departamento de Química Faculdade de Ciências, Universidade do Porto Rua do Campo Alegre, 687, 4169-007 Porto, Portugal, and Department of Biochemistry and Organic Chemistry, Uppsala University, BMC Box 576, SE-75123 Uppsala, Sweden
| |
Collapse
|
10
|
Augustine CK, Yoo JS, Potti A, Yoshimoto Y, Zipfel PA, Friedman HS, Nevins JR, Ali-Osman F, Tyler DS. Genomic and molecular profiling predicts response to temozolomide in melanoma. Clin Cancer Res 2009; 15:502-10. [PMID: 19147755 DOI: 10.1158/1078-0432.ccr-08-1916] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite objective response rates of only approximately 13%, temozolomide remains one of the most effective single chemotherapy agents against metastatic melanoma, second only to dacarbazine, the current standard of care for systemic treatment of melanoma. The goal of this study was to identify molecular and/or genetic markers that correlate with, and could be used to predict, response to temozolomide-based treatment regimens and that reflect the intrinsic properties of a patient's tumor. EXPERIMENTAL DESIGN Using a panel of 26 human melanoma-derived cell lines, we determined in vitro temozolomide sensitivity, O(6)-methylguanine-DNA methyltransferase (MGMT) activity, MGMT protein expression and promoter methylation status, and mismatch repair proficiency, as well as the expression profile of 38,000 genes using an oligonucleotide-based microarray platform. RESULTS The results showed a broad spectrum of temozolomide sensitivity across the panel of cell lines, with IC(50) values ranging from 100 micromol/L to 1 mmol/L. There was a significant correlation between measured temozolomide sensitivity and a gene expression signature-derived prediction of temozolomide sensitivity (P < 0.005). Notably, MGMT alone showed a significant correlation with temozolomide sensitivity (MGMT activity, P < 0.0001; MGMT expression, P <or= 0.0001). The promoter methylation status of the MGMT gene, however, was not consistent with MGMT gene expression or temozolomide sensitivity. CONCLUSIONS These results show that melanoma resistance to temozolomide is conferred predominantly by MGMT activity and suggest that MGMT expression could potentially be a useful tool for predicting the response of melanoma patients to temozolomide therapy.
Collapse
Affiliation(s)
- Christina K Augustine
- Department of Surgery, and Duke Institute for Genome Sciences and Policy, Duke University Medical Center and Durham VA Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Dourado D, Fernandes P, Mannervik B, Ramos M. Glutathione Transferase: New Model for Glutathione Activation. Chemistry 2008; 14:9591-8. [DOI: 10.1002/chem.200800946] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
12
|
Lo M, Wang YZ, Gout PW. The x(c)- cystine/glutamate antiporter: a potential target for therapy of cancer and other diseases. J Cell Physiol 2008; 215:593-602. [PMID: 18181196 DOI: 10.1002/jcp.21366] [Citation(s) in RCA: 323] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The x(c) (-) cystine/glutamate antiporter is a major plasma membrane transporter for the cellular uptake of cystine in exchange for intracellular glutamate. Its main functions in the body are mediation of cellular cystine uptake for synthesis of glutathione essential for cellular protection from oxidative stress and maintenance of a cystine:cysteine redox balance in the extracellular compartment. In the past decade it has become evident that the x(c) (-) transporter plays an important role in various aspects of cancer, including: (i) growth and progression of cancers that have a critical growth requirement for extracellular cystine/cysteine, (ii) glutathione-based drug resistance, (iii) excitotoxicity due to excessive release of glutamate, and (iv) uptake of herpesvirus 8, a causative agent of Kaposi's sarcoma. The x(c) (-) transporter also plays a role in certain CNS and eye diseases. This review focuses on the expression and function of the x(c) (-) transporter in cells and tissues with particular emphasis on its role in disease pathogenesis. The potential use of x(c) (-) inhibitors (e.g., sulfasalazine) for arresting tumor growth and/or sensitizing cancers is discussed.
Collapse
Affiliation(s)
- Maisie Lo
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
13
|
Olivier S, Robe P, Bours V. Can NF-κB be a target for novel and efficient anti-cancer agents? Biochem Pharmacol 2006; 72:1054-68. [PMID: 16973133 DOI: 10.1016/j.bcp.2006.07.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2006] [Revised: 07/20/2006] [Accepted: 07/24/2006] [Indexed: 12/23/2022]
Abstract
Since the discovery of the NF-kappaB transcription factor in 1986 and the cloning of the genes coding for NF-kappaB and IkappaB proteins, many studies demonstrated that this transcription factor can, in most cases, protect transformed cells from apoptosis and therefore participate in the onset or progression of many human cancers. Molecular studies demonstrated that ancient widely used drugs, known for their chemopreventive or therapeutic activities against human cancers, inhibit NF-kappaB, usually among other biological effects. It is therefore considered that the anti-cancer activities of NSAIDs (non-steroidal anti-inflammatory drugs) or glucocorticoids are probably partially related to the inhibition of NF-kappaB and new clinical trials are being initiated with old compounds such as sulfasalazine. In parallel, many companies have developed novel agents acting on the NF-kappaB pathway: some of these agents are supposed to be NF-kappaB specific (i.e. IKK inhibitors) while others have wide-range biological activities (i.e. proteasome inhibitors). Today, the most significant clinical data have been obtained with bortezomib, a proteasome inhibitor, for the treatment of multiple myeloma. This review discusses the preclinical and clinical data obtained with these various drugs and their putative future developments.
Collapse
Affiliation(s)
- Sabine Olivier
- Department of Rheumatology, Centre for Biomedical Integrative Genoproteomics, University of Liège, CHU B35, Sart-Tilman, 4000 Liège, Belgium
| | | | | |
Collapse
|
14
|
Mathew N, Kalyanasundaram M, Balaraman K. GlutathioneS-transferase (GST) inhibitors. Expert Opin Ther Pat 2006. [DOI: 10.1517/13543776.16.4.431] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
15
|
Procopio A, Alcaro S, Cundari S, De Nino A, Ortuso F, Sacchetta P, Pennelli A, Sindona G. Molecular Modeling, Synthesis, and Preliminary Biological Evaluation of Glutathione-S-Transferase Inhibitors as Potential Therapeutic Agents. J Med Chem 2005; 48:6084-9. [PMID: 16162009 DOI: 10.1021/jm0504609] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Changes in the GSH/GST system have been found to correlate with resistance to anticancer alkylating agents, presumably through accelerated detoxification of these drugs since some GSTs have been shown to catalyze the conjugation of GSH to specific antineoplastic agents. GSH-alkyl derivatives were designed by molecular modeling, synthesized, and tested as inhibitors of human GST-Pi.
Collapse
Affiliation(s)
- Antonio Procopio
- Dipartimento di Scienze Farmaco-Biologiche, Università della Magna Graecia, Complesso Ninì Barbieri, 88021 Roccelletta di Borgia (Cz), Italy.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Li M, Ittmann MM, Rowley DR, Knowlton AA, Vaid AK, Epner DE. Glutathione S-transferase pi is upregulated in the stromal compartment of hormone independent prostate cancer. Prostate 2003; 56:98-105. [PMID: 12746833 DOI: 10.1002/pros.10249] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Glutathione S-transferase (GST) pi is a detoxifying enzyme abundant in normal prostate basal cells but only rarely expressed in prostate cancer cells. The current studies are the first to focus on GST pi in the stromal compartment of prostate tumors. METHODS We employed immunohistochemical, immunofluorescence, and Western blot analysis to measure GST pi expression and subcellular localization in 21 primary and metastatic tumors from patients with hormone independent prostate cancer, as well as seven lymph node metastases and six prostatectomy specimens. RESULTS GST pi was detectable in stromal cells in 17 of the 21 hormone independent prostate tumors. GST pi tissue distribution in hormone independent tumors coincided with vimentin staining, suggesting that GST pi is expressed by reactive fibroblasts and/or myofibroblasts. CONCLUSIONS The current results suggest that prostate cancer cells induce an injury response in the stroma during progression to hormone independence, which results in GST pi expression. Stromal GST pi may contribute to chemoresistence of advanced prostate cancer.
Collapse
Affiliation(s)
- Ming Li
- Department of Medicine, Houston, Texas
| | | | | | | | | | | |
Collapse
|
17
|
Jones RB. Clinical pharmacology of melphalan and its implications for clinical resistance to anticancer agents. Cancer Treat Res 2003; 112:305-22. [PMID: 12481722 DOI: 10.1007/978-1-4615-1173-1_15] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Roy B Jones
- Bone Marrow Transplant Program, University of Colorado Health Science Center, Denver, Colorado, USA
| |
Collapse
|
18
|
Harbottle A, Daly AK, Atherton K, Campbell FC. Role of glutathione S-transferase P1, P-glycoprotein and multidrug resistance-associated protein 1 in acquired doxorubicin resistance. Int J Cancer 2001; 92:777-83. [PMID: 11351295 DOI: 10.1002/ijc.1283] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
While P-glycoprotein (Pgp) and multidrug resistance-associated protein 1 (MRP1) are known to be important in acquired doxorubicin resistance, the role of glutathione S-transferases (GST) remains unclear. Our study assessed roles of these 3 factors in a human drug-sensitive carcinoma cell line (HEp2), a subclone made resistant by prolonged incubation in doxorubicin (HEp2A), and HEp2 cells stably transfected with human GSTP1. Drug-resistant HEp2A cells showed greater total GST activity, GSTP class enzyme expression, Pgp expression, MRP1 transcript expression, drug efflux and at least 13-fold greater resistance to doxorubicin than the parent HEp2 cell line. GSTM class enzyme expression was similar in both cell types, while GSTA class enzymes were not detected. In the resistant HEp2A cells, cytotoxicity was markedly enhanced by the Pgp/MRP inhibitor verapamil at low doxorubicin concentrations. The GST inhibitor curcumin also enhanced cytotoxicity in HEp2A cells when the Pgp/MRP efflux barrier had been reversed by verapamil or overcome by high doxorubicin concentrations. In addition, curcumin had a chemosensitising effect at low doxorubicin concentrations in HEp2 cells. Stable transfection of HEp2 cells with human GSTP1 increases doxorubicin resistance 3-fold over control cells. Our study indicates involvement of GSTP enzymes as well as efflux mechanisms in the acquired doxorubicin-resistance phenotype.
Collapse
Affiliation(s)
- A Harbottle
- Department of Surgical Sciences, The Medical School, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | | | | | | |
Collapse
|
19
|
Oakley AJ, Lo Bello M, Nuccetelli M, Mazzetti AP, Parker MW. The ligandin (non-substrate) binding site of human Pi class glutathione transferase is located in the electrophile binding site (H-site). J Mol Biol 1999; 291:913-26. [PMID: 10452896 DOI: 10.1006/jmbi.1999.3029] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutathione S -transferases (GSTs) play a pivotal role in the detoxification of foreign chemicals and toxic metabolites. They were originally termed ligandins because of their ability to bind large molecules (molecular masses >400 Da), possibly for storage and transport roles. The location of the ligandin site in mammalian GSTs is still uncertain despite numerous studies in recent years. Here we show by X-ray crystallography that the ligandin binding site in human pi class GST P1-1 occupies part of one of the substrate binding sites. This work has been extended to the determination of a number of enzyme complex crystal structures which show that very large ligands are readily accommodated into this substrate binding site and in all, but one case, causes no significant movement of protein side-chains. Some of these molecules make use of a hitherto undescribed binding site located in a surface pocket of the enzyme. This site is conserved in most, but not all, classes of GSTs suggesting it may play an important functional role.
Collapse
Affiliation(s)
- A J Oakley
- The Ian Potter Foundation Protein Crystallography Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, 3065, Australia
| | | | | | | | | |
Collapse
|
20
|
Tew KD, Dutta S, Schultz M. Inhibitors of glutathione S-transferases as therapeutic agents. Adv Drug Deliv Rev 1997; 26:91-104. [PMID: 10837536 DOI: 10.1016/s0169-409x(97)00029-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glutathione S-transferases (GST) are a family of phase II detoxification enzymes with broad substrate specificities. They catalyze the conjugation of glutathione with many different types of xenobiotics, rendering the compound more water soluble and thus more easily eliminated. Resistance to cancer chemotherapeutic drugs, such as the alkylating agents, has been directly correlated with the overexpression of GSTs. Subsequently, a rationale has been established to utilize agents that inhibit GST in combination with alkylating agents to circumvent this resistance. Two such agents, ethacrynic acid (EA) and Terrapin 199 (TER 199), have been examined for this purpose. EA, an inhibitor of all classes of GST isozymes, has been used clinically in combination with thiotepa. More recently, TER 199, a glutathione analog-based GST inhibitor, has been modeled specifically to inhibit GST pi, an enzyme which is commonly found at high levels in human tumor biopsies. Furthermore, a therapeutic strategy has been designed to take advantage of GST pi activation of a prodrug, TER 286. Recent studies have investigated the molecular mechanisms involved in the cellular response to GST inhibitors and have employed techniques such as differential display to examine altered gene expression as well as to identify novel genes induced by these agents. Overall, this strategy may provide further insight into the action of these agents in the cell as well as prove useful in endeavors to modulate anticancer drug resistance.
Collapse
Affiliation(s)
- KD Tew
- Department of Pharmacology, Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
21
|
Kunze T. Phosphono analogues of glutathione as new inhibitors of glutathione S-transferases. Arch Pharm (Weinheim) 1996; 329:503-9. [PMID: 8997900 DOI: 10.1002/ardp.19963291106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Phosphono-analogues of glutathione containing the O = P(OR)2 moiety in place of the cysteinyl residue CH2SH 1a-1d were prepared by solution phase peptide synthesis. Benzyl, benzyloxy-carbonyl, and tert-butyl protecting groups were used to mask the individual amino acid functional groups. The formation of peptide bonds was achieved by the usual peptide synthesis via activation of carboxylic functions with cyclohexylcarbodiimide and subsequent reaction with free amino groups. The thus obtained, fully-protected peptides were each purified by normal phase column chromatography. Deprotection was accomplished by hydrogenolysis and by treatment with HBr/acetic acid yielding the desired phosphonic acid diester 1a-1d. The inhibition of the glutathione conjugation of 1-chloro-2,4-dinitrobenzene by human placental glutathione S-transferase was studied by determining the IC50 values of the new glutathione analogues. The IC50 values were 291 microM, 139 microM, 64 microM, and 21 microM for the dimethyl, diethyl, diisopropyl, and di-n-butyl esters, respectively. The results clearly show that the formal substitution of the glutathione thiol function by phosphonic acid esters leads to a new class of glutathione S-transferase inhibitors. Further investigations directed at the question of whether or not these glutathione analogues are suitable for a modulation in chemotherapy are in progress.
Collapse
Affiliation(s)
- T Kunze
- Pharmazeutisches Institut, Christian-Albrechts-Universität, Kiel, Germany
| |
Collapse
|
22
|
Affiliation(s)
- M L O'Brien
- Department of Pharmacology, University of Pennsylvania, Philadelphia 19111, USA
| | | |
Collapse
|