1
|
Chen M, Li Y, Liu Y, Jia B, Liu X, Ma T. Carbonized polymer dots derived from metformin and L-arginine for tumor cell membrane- and mitochondria-dual targeting therapy. NANOSCALE 2023; 15:17922-17935. [PMID: 37902070 DOI: 10.1039/d3nr04145j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Metformin has demonstrated antitumor potential in clinical studies; however, achieving optimal antitumor effects requires administering an extremely safe medication dose. To enhance the efficacy and reduce dosage requirements, we propose the creation of large-molecule drugs through the combination of small-molecule drugs. In this study, we developed novel polymer dots, referred to as MA-dots, with sizes of approximately 5 nm, featuring dual targeting capabilities for tumor cell membranes and mitochondria. MA-dots were synthesized using metformin and L-arginine via a rapid microwave-assisted method. Notably, the resulting MA-dots (with a half maximal inhibitory concentration (IC50) of 93.60 μg mL-1) exhibited more than a 12-fold increase in antitumor activity compared to the raw metformin material (IC50 = 1159.00 μg mL-1) over a 24-hour period. In addition, our MA-dots outperformed most metformin-derived nanodrugs in terms of antitumor efficacy. Furthermore, oral gavage treatment with MA-dots led to the suppression of A549 (lung cancer cell lines) tumor growth in vivo. Mechanistic investigations revealed that MA-dots bound to the large neutral amino acid transporter 1 (LAT1) proteins, which are overexpressed in malignant tumor cell membranes. Moreover, these MA-dots accumulated within the mitochondria, leading to increased production of reactive oxygen species (ROS), mitochondrial damage, and disruption of energy metabolism by modulating the 5'-adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway in tumor cells. This cascade of events triggers cell-cycle arrest and apoptosis. In summary, this study presented a rapid method for fabricating a novel nanoderivative, MA-dots, capable of both tumor targeting and exerting tumor-suppressive effects.
Collapse
Affiliation(s)
- Manling Chen
- Institute of Clean Energy Chemistry, Key Laboratory for Green Synthesis and Preparative Chemistry of Advanced Materials, College of Chemistry, Liaoning University, Shenyang 110036, Liaoning, P. R. China.
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, Liaoning, P. R. China
| | - Yangcheng Liu
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, Liaoning, P. R. China
| | - Baohua Jia
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia.
| | - Xue Liu
- Institute of Clean Energy Chemistry, Key Laboratory for Green Synthesis and Preparative Chemistry of Advanced Materials, College of Chemistry, Liaoning University, Shenyang 110036, Liaoning, P. R. China.
| | - Tianyi Ma
- School of Science, STEM College, RMIT University, Melbourne, VIC 3000, Australia.
| |
Collapse
|
2
|
Alhourani A, Førde JL, Nasrollahzadeh M, Eichacker LA, Herfindal L, Hagland HR. Graphene-based phenformin carriers for cancer cell treatment: a comparative study between oxidized and pegylated pristine graphene in human cells and zebrafish. NANOSCALE ADVANCES 2022; 4:1668-1680. [PMID: 36134366 PMCID: PMC9417205 DOI: 10.1039/d1na00778e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/27/2022] [Indexed: 06/16/2023]
Abstract
Graphene is an attractive choice for the development of an effective drug carrier in cancer treatment due to its high adsorption area and pH-responsive drug affinity. In combination with the highly potent metabolic drug phenformin, increased doses could be efficiently delivered to cancer cells. This study compares the use of graphene oxide (GO) and polyethylene glycol stabilized (PEGylated) pristine graphene nanosheets (PGNSs) for drug delivery applications with phenformin. The cytotoxicity and mitotoxicity of the graphene-based systems were assessed in human cells and zebrafish larvae. Targeted drug release from GO and PGNSs was evaluated at different pH levels known to arise in proliferating tumor microenvironments. PGNSs were less cytotoxic and mitotoxic than GO, and showed an increased release of phenformin at lower pH in cells, compared to GO. In addition, the systemic phenformin effect was mitigated in zebrafish larvae when bound to GO and PGNSs compared to free phenformin, as measured by flavin metabolic lifetime imaging. These results pave the way for improved phenformin-based cancer therapy using graphene nano-sheets, where PGNSs were superior to GO.
Collapse
Affiliation(s)
- Abdelnour Alhourani
- Department of Chemistry, Biosciences and Environmental Engineering, University of Stavanger Stavanger Norway
| | - Jan-Lukas Førde
- Centre for Pharmacy, Department of Clinical Science, University of Bergen Bergen Norway
- Department of Internal Medicine, Haukeland University Hospital Bergen Norway
| | - Mojdeh Nasrollahzadeh
- Department of Chemistry, Biosciences and Environmental Engineering, University of Stavanger Stavanger Norway
| | - Lutz Andreas Eichacker
- Department of Chemistry, Biosciences and Environmental Engineering, University of Stavanger Stavanger Norway
| | - Lars Herfindal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen Bergen Norway
| | - Hanne Røland Hagland
- Department of Chemistry, Biosciences and Environmental Engineering, University of Stavanger Stavanger Norway
| |
Collapse
|
3
|
Alhourani A, Førde JL, Eichacker LA, Herfindal L, Hagland HR. Improved pH-Responsive Release of Phenformin from Low-Defect Graphene Compared to Graphene Oxide. ACS OMEGA 2021; 6:24619-24629. [PMID: 34604644 PMCID: PMC8482513 DOI: 10.1021/acsomega.1c03283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Graphene-based drug carriers provide a promising addition to current cancer drug delivery options. Increased accessibility of high-quality graphene made by plasma-enhanced chemical vapor deposition (PE-CVD) makes it an attractive material to revisit in comparison to the widely studied graphene oxide (GO) in drug delivery. Here, we show the potential of repurposing the metabolic drug phenformin for cancer treatment in terms of stability, binding, and pH-responsive release. Using covalent attachment of poly(ethylene glycol) (PEG) onto pristine (PE-CVD) graphene, we show that PEG stabilized graphene nanosheets (PGNS) are stable in aqueous solutions and exhibit higher binding affinity toward phenformin than GO. Moreover, we experimentally demonstrate an improved drug release from PGNS than GO at pH levels lower than physiological conditions, yet comparable to that found in tumor microenvironments.
Collapse
Affiliation(s)
- Abdelnour Alhourani
- Department
of Chemistry, Biosciences and Environmental Technology, University of Stavanger, 4021 Stavanger, Norway
| | - Jan-Lukas Førde
- Centre
for Pharmacy, Department of Clinical Science, University of Bergen, 5007 Bergen, Norway
- Department
of Internal Medicine, Haukeland University
Hospital, 5021 Bergen, Norway
| | - Lutz Andreas Eichacker
- Department
of Chemistry, Biosciences and Environmental Technology, University of Stavanger, 4021 Stavanger, Norway
| | - Lars Herfindal
- Centre
for Pharmacy, Department of Clinical Science, University of Bergen, 5007 Bergen, Norway
| | - Hanne Røland Hagland
- Department
of Chemistry, Biosciences and Environmental Technology, University of Stavanger, 4021 Stavanger, Norway
| |
Collapse
|
4
|
Singh AR, Gu JJ, Zhang Q, Torka P, Sundaram S, Mavis C, Hernandez-Ilizaliturri FJ. Metformin sensitizes therapeutic agents and improves outcome in pre-clinical and clinical diffuse large B-cell lymphoma. Cancer Metab 2020; 8:10. [PMID: 32647571 PMCID: PMC7336499 DOI: 10.1186/s40170-020-00213-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The treatment of diffuse large B-cell lymphoma (DLBCL) is limited by the development of resistance to therapy, and there is a need to develop novel therapeutic strategies for relapsed and refractory aggressive lymphoma. Metformin is an oral agent for type 2 diabetes that has been shown to decrease cancer risk and lower mortality in other types of cancer. METHODS We performed a retrospective analysis of the RPCCC database looking at patients with DLBCL treated with front-line chemotherapy. We also performed pre-clinical studies looking at the effect of metformin on cell viability, cell number, Ki67, ATP production, apoptosis, ROS production, mitochondrial membrane potential, cell cycle, effect with chemotherapeutic agents, and rituximab. Finally, we studied mouse models to see the anti-tumor effect of metformin. RESULTS Among diabetic patients, metformin use was associated with improved progression-free survival (PFS) and overall survival (OS) compared to diabetic patients not on metformin. Our pre-clinical studies showed metformin is itself capable of anti-tumor effects and causes cell cycle arrest in the G1 phase. Metformin induces apoptosis, ROS production, and increased mitochondrial membrane permeability. Metformin exhibited additive/synergistic effects when combined with traditional chemotherapy or rituximab in vitro. In vivo, metformin in combination with rituximab showed improved survival compared with rituximab monotherapy. CONCLUSIONS Our retrospective analysis showed that metformin with front-line chemotherapy in diabetic patients resulted in improved PFS and OS. Our pre-clinical studies demonstrate metformin has potential to re-sensitize resistant lymphoma to the chemo-immunotherapy and allow us to develop a hypothesis as to its activity in DLBCL.
Collapse
Affiliation(s)
| | - Juan J. Gu
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
- Department Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Qunling Zhang
- Department of Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Pallawi Torka
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Suchitra Sundaram
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Cory Mavis
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
- Department Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Francisco J. Hernandez-Ilizaliturri
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
- Department Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| |
Collapse
|
5
|
Ghiasi B, Sarokhani D, Najafi F, Motedayen M, Dehkordi AH. The Relationship Between Prostate Cancer and Metformin Consumption: A Systematic Review and Meta-analysis Study. Curr Pharm Des 2020; 25:1021-1029. [PMID: 30767734 DOI: 10.2174/1381612825666190215123759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/11/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Prostate cancer is the most common malignant cancer in men worldwide and after lung cancer, it is the second leading cause of cancer mortality in men. The purpose of this study was to investigate the relationship between prostate cancer and metformin consumption in men. METHODS The current study is a systematic and meta-analysis review based on the PRISMA statement. To access the studies of domestic and foreign databases, Iran Medex, SID, Magiran, Iran Doc, Medlib, ProQuest, Science Direct, PubMed, Scopus, Web of Science and the Google Scholar search engine were searched during the 2009- 2018 period for related keywords. In order to evaluate the heterogeneity of the studies, Q test and I2 indicator were used. The data were analyzed using the STATA 15.1 software. RESULTS In 11 studies with a sample size of 877058, the odds ratio of metformin consumption for reducing prostate cancer was estimated at 0.89 (95%CI: 0.67-1.17). Meta-regression also showed there was no significant relationship between the odds ratio and the publication year of the study. However, there was a significant relationship between the odds ratio and the number of research samples. CONCLUSION Using metformin in men reduces the risk of prostate cancer but it is not statistically significant.
Collapse
Affiliation(s)
- Bahareh Ghiasi
- Department of Nephrology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Diana Sarokhani
- Research Center for Environmental Determinants of Health (RCEDH), School of Public Health, Kermanshah Uninversity of Medical Sciences, Kermanshah, Iran
| | - Farid Najafi
- Research Center for Environmental Determinants of Health (RCEDH), School of Public Health, Kermanshah Uninversity of Medical Sciences, Kermanshah, Iran
| | - Morteza Motedayen
- Cardiology Department, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Hasanpour Dehkordi
- Department of Medical-Surgical, Faculty of Nursing and Midwifery, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
6
|
Benjamin D, Colombi M, Hindupur SK, Betz C, Lane HA, El-Shemerly MYM, Lu M, Quagliata L, Terracciano L, Moes S, Sharpe T, Wodnar-Filipowicz A, Moroni C, Hall MN. Syrosingopine sensitizes cancer cells to killing by metformin. SCIENCE ADVANCES 2016; 2:e1601756. [PMID: 28028542 PMCID: PMC5182053 DOI: 10.1126/sciadv.1601756] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/20/2016] [Indexed: 05/04/2023]
Abstract
We report that the anticancer activity of the widely used diabetic drug metformin is strongly potentiated by syrosingopine. Synthetic lethality elicited by combining the two drugs is synergistic and specific to transformed cells. This effect is unrelated to syrosingopine's known role as an inhibitor of the vesicular monoamine transporters. Syrosingopine binds to the glycolytic enzyme α-enolase in vitro, and the expression of the γ-enolase isoform correlates with nonresponsiveness to the drug combination. Syrosingopine sensitized cancer cells to metformin and its more potent derivative phenformin far below the individual toxic threshold of each compound. Thus, combining syrosingopine and codrugs is a promising therapeutic strategy for clinical application for the treatment of cancer.
Collapse
Affiliation(s)
- Don Benjamin
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Marco Colombi
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | - Charles Betz
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Heidi A. Lane
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | | | - Min Lu
- Institute for Medical Microbiology, University of Basel, 4003 Basel, Switzerland
| | - Luca Quagliata
- Molecular Pathology, University Hospital Basel, 4003 Basel, Switzerland
| | - Luigi Terracciano
- Molecular Pathology, University Hospital Basel, 4003 Basel, Switzerland
| | - Suzette Moes
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Timothy Sharpe
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | | | | | - Michael N. Hall
- Biozentrum, University of Basel, 4056 Basel, Switzerland
- Corresponding author.
| |
Collapse
|
7
|
Kannarkatt J, Alkharabsheh O, Tokala H, Dimitrov NV. Metformin and Angiogenesis in Cancer - Revisited. Oncology 2016; 91:179-184. [DOI: 10.1159/000448175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/27/2016] [Indexed: 11/19/2022]
|
8
|
Othman EM, Oli RG, Arias-Loza PA, Kreissl MC, Stopper H. Metformin Protects Kidney Cells From Insulin-Mediated Genotoxicity In Vitro and in Male Zucker Diabetic Fatty Rats. Endocrinology 2016; 157:548-59. [PMID: 26636185 DOI: 10.1210/en.2015-1572] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hyperinsulinemia is thought to enhance cancer risk. A possible mechanism is induction of oxidative stress and DNA damage by insulin, Here, the effect of a combination of metformin with insulin was investigated in vitro and in vivo. The rationales for this were the reported antioxidative properties of metformin and the aim to gain further insights into the mechanisms responsible for protecting the genome from insulin-mediated oxidative stress and damage. The comet assay, a micronucleus frequency test, and a mammalian gene mutation assay were used to evaluate the DNA damage produced by insulin alone or in combination with metformin. For analysis of antioxidant activity, oxidative stress, and mitochondrial disturbances, the cell-free ferric reducing antioxidant power assay, the superoxide-sensitive dye dihydroethidium, and the mitochondrial membrane potential-sensitive dye 5,5',6,6'tetrachloro-1,1',3,3'-tetraethylbenzimidazol-carbocyanine iodide were applied. Accumulation of p53 and pAKT were analyzed. As an in vivo model, hyperinsulinemic Zucker diabetic fatty rats, additionally exposed to insulin during a hyperinsulinemic-euglycemic clamp, were treated with metformin. In the rat kidney samples, dihydroethidium staining, p53 and pAKT analysis, and quantification of the oxidized DNA base 8-oxo-7,8-dihydro-2'-deoxyguanosine were performed. Metformin did not show intrinsic antioxidant activity in the cell-free assay, but protected cultured cells from insulin-mediated oxidative stress, DNA damage, and mutation. Treatment of the rats with metformin protected their kidneys from oxidative stress and genomic damage induced by hyperinsulinemia. Metformin may protect patients from genomic damage induced by elevated insulin levels. This may support efforts to reduce the elevated cancer risk that is associated with hyperinsulinemia.
Collapse
Affiliation(s)
- Eman Maher Othman
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - R G Oli
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Paula-Anahi Arias-Loza
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Michael C Kreissl
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| |
Collapse
|
9
|
Westhaus A, Blumrich EM, Dringen R. The Antidiabetic Drug Metformin Stimulates Glycolytic Lactate Production in Cultured Primary Rat Astrocytes. Neurochem Res 2015; 42:294-305. [PMID: 26433380 DOI: 10.1007/s11064-015-1733-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 12/11/2022]
Abstract
Metformin is the most frequently used drug for the treatment of type 2 diabetes in humans. However, only little is known about effects of metformin on brain metabolism. To investigate potential metabolic consequences of an exposure of brain cells to metformin, we incubated rat astrocyte-rich primary cultures with this compound. Metformin in concentrations of up to 30 mM did not acutely compromise the viability of astrocytes, but caused a time- and concentration-dependent increase in cellular glucose consumption and lactate production. For acute incubations in the hour range, the presence of 10 mM metformin doubled the glycolytic flux, while already 1 mM metformin doubled glycolytic flux during incubation for 24 h. In addition to metformin, also other guanidino compounds increased astrocytic lactate production. After 4 h of incubation, half-maximal stimulation of glycolysis was observed for metformin, guanidine and phenformin at concentrations of around 3 mM, 3 mM and 30 µM, respectively. The acute stimulation of glycolytic lactate production by metformin was persistent after removal of extracellular metformin and was also observed, if glucose was absent from the incubation medium or replaced by other hexoses. The metformin-induced stimulation of glycolytic flux was not prevented by compound C, an inhibitor of AMP-dependent protein kinase, nor was it additive to the stimulation of glycolytic flux caused by respiratory chain inhibitors. These data demonstrate that the antidiabetic drug metformin has the potential to strongly activate glycolytic lactate production in brain astrocytes.
Collapse
Affiliation(s)
- Adrian Westhaus
- Centre for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany
| | - Eva Maria Blumrich
- Centre for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany.,Centre for Environmental Research and Sustainable Technology, Leobener Strasse, 28359, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany. .,Centre for Environmental Research and Sustainable Technology, Leobener Strasse, 28359, Bremen, Germany.
| |
Collapse
|
10
|
Shafaee A, Dastyar DZ, Islamian JP, Hatamian M. Inhibition of tumor energy pathways for targeted esophagus cancer therapy. Metabolism 2015; 64:1193-8. [PMID: 26271140 DOI: 10.1016/j.metabol.2015.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/18/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022]
Abstract
Interest in targeting cancer metabolism has been renewed in recent years with the discovery that many cancer related pathways have a profound effect on metabolism and that many tumors become dependent on specific metabolic processes. Accelerated glucose uptake during anaerobic glycolysis and loss of regulation between glycolytic metabolism and respiration, are the major metabolic changes found in malignant cells. The non-metabolizable glucose analog, 2-deoxy-D-glucose inhibits glucose synthesis and adenosine triphosphate production. The adenosine monophosphate-activated protein kinase (AMPK) is a key sensor of cellular energy and AMPK is a potential target for cancer prevention and/or treatment. Metformin is an activator of AMPK which inhibits protein synthesis and gluconeogenesis during cellular stress. This article reviews the status of clinical and laboratory researches exploring targeted therapies via metabolic pathways for treatment of esophageal cancer.
Collapse
Affiliation(s)
- Abbas Shafaee
- Department of Radiology, School of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Zarei Dastyar
- Department of Medical Radiation Science, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalil Pirayesh Islamian
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Milad Hatamian
- Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
11
|
Metformin and erlotinib synergize to inhibit basal breast cancer. Oncotarget 2015; 5:10503-17. [PMID: 25361177 PMCID: PMC4279389 DOI: 10.18632/oncotarget.2391] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/21/2014] [Indexed: 12/16/2022] Open
Abstract
Basal-like breast cancers (BBCs) are enriched for increased EGFR expression and decreased expression of PTEN. We found that treatment with metformin and erlotinib synergistically induced apoptosis in a subset of BBC cell lines. The drug combination led to enhanced reduction of EGFR, AKT, S6 and 4EBP1 phosphorylation, as well as prevented colony formation and inhibited mammosphere outgrowth. Our data with other compounds suggested that biguanides combined with EGFR inhibitors have the potential to outperform other targeted drug combinations and could be employed in other breast cancer subtypes, as well as other tumor types, with activated EGFR and PI3K signaling. Analysis of BBC cell line alterations led to the hypothesis that loss of PTEN sensitized cells to the drug combination which was confirmed using isogenic cell line models with and without PTEN expression. Combined metformin and erlotinib led to partial regression of PTEN-null and EGFR-amplified xenografted MDA-MB-468 BBC tumors with evidence of significant apoptosis, reduction of EGFR and AKT signaling, and lack of altered plasma insulin levels. Combined treatment also inhibited xenografted PTEN null HCC-70 BBC cells. Measurement of trough plasma drug levels in xenografted mice and a separately performed pharmacokinetics modeling study support possible clinical translation.
Collapse
|
12
|
Matyszewski A, Czarnecka A, Kawecki M, Korzeń P, Safir IJ, Kukwa W, Szczylik C. Impaired glucose metabolism treatment and carcinogenesis. Oncol Lett 2015; 10:589-594. [PMID: 26622538 DOI: 10.3892/ol.2015.3324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/29/2015] [Indexed: 12/29/2022] Open
Abstract
Carbohydrate metabolism disorders increase the risk of carcinogenesis. Diabetes mellitus alters numerous physiological processes that may encourage cancer growth. However, treating impaired glucose homeostasis may actually promote neoplasia; maintaining proper glucose plasma concentrations reduces metabolic stresses, however, certain medications may themselves result in oncogenic effects. A number of previous studies have demonstrated that metformin reduces the cancer risk. However, the use of sulfonylurea derivatives correlates with an increased risk of developing a malignancy. Another form of treatment, insulin therapy, involves using various forms of insulin that differ in pharmacodynamics, pharmacokinetics and efficacy. Previous studies have indicated that certain insulin variants also affect the cancer risk. The results from analyses that address the safety of long-lasting insulin types raise the most concern regarding the increased risk of malignancy. Rapid development of novel diabetic medications and their widespread use carries the risk of potentially increased rates of cancer, unnoticeable in limited, randomized, controlled trials. In the present review, the results of clinical and epidemiological studies are evaluated to assess the safety of anti-hyperglycemic medications and their effect on cancer risk and outcomes.
Collapse
Affiliation(s)
- Artur Matyszewski
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland ; Department of Internal Medicine, Cardiology and Hypertensionr, L. Rydygier Province Hospital, Suwałki, Poland ; Department of Nephrology and Dialysis Center, L. Rydygier Province Hospital, Suwałki, Poland
| | - Anna Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Maciej Kawecki
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Korzeń
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Ilan J Safir
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wojciech Kukwa
- Department of Otorhinolaryngology, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
13
|
Epithelial–mesenchymal transition in human cancer: Comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther 2015; 150:33-46. [PMID: 25595324 DOI: 10.1016/j.pharmthera.2015.01.004] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/05/2015] [Indexed: 02/07/2023]
|
14
|
Welch MR, Grommes C. Retrospective analysis of the effects of steroid therapy and antidiabetic medication on survival in diabetic glioblastoma patients. CNS Oncol 2015; 2:237-46. [PMID: 25054464 DOI: 10.2217/cns.13.12] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIMS Type 2 diabetes mellitus (DM2) affects 10% of the population, but little is known about how DM2 and antidiabetic medication impact glioblastoma (GBM) patients. PATIENTS & METHODS We retrospectively reviewed GBM patients with DM2 seen at a single institution from 1998 to 2010. RESULTS Of 988 GBMs, 124 (12.6%) were affected by DM2. Thirty-four developed DM2 after steroid use and 89 had pre-existing DM2. Median overall survival among diabetic GBMs was 10 months compared with 13 months among nondiabetics. Only 15% of diabetic patients achieved sustained steroid taper. Sixty-seven (54%) were managed with a single antidiabetic medication and, within this monotherapy group, Karnofsky Performance Score, resection status, steroid dependency and metformin use were the most important predictors of survival on multivariate analysis. CONCLUSION The prevalence of DM2 among GBMs is similar to that of the general population. A more aggressive approach to steroid tapering and the choice of antidiabetic drug may improve survival within this patient population.
Collapse
Affiliation(s)
- Mary R Welch
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | |
Collapse
|
15
|
La Vecchia C, Bosetti C. Metformin: are potential benefits on cancer risk extended to cancer survival? Oncologist 2013; 18:1245-7. [PMID: 24258614 DOI: 10.1634/theoncologist.2013-0381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Carlo La Vecchia
- Department of Epidemiology, IRCCS-lstituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | |
Collapse
|
16
|
Abstract
One-carbon metabolism involving the folate and methionine cycles integrates nutritional status from amino acids, glucose and vitamins, and generates diverse outputs, such as the biosynthesis of lipids, nucleotides and proteins, the maintenance of redox status and the substrates for methylation reactions. Long considered a 'housekeeping' process, this pathway has recently been shown to have additional complexity. Genetic and functional evidence suggests that hyperactivation of this pathway is a driver of oncogenesis and establishes a link to cellular epigenetic status. Given the wealth of clinically available agents that target one-carbon metabolism, these new findings could present opportunities for translation into precision cancer medicine.
Collapse
Affiliation(s)
- Jason W Locasale
- Field of Biochemistry and Molecular Cell Biology, Cornell University, Ithaca New York 14850, USA.
| |
Collapse
|
17
|
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma is a devastating disease, with an overall 5-year survival rate of only 3% to 5%. As the current therapies offer very limited survival benefits, novel therapeutic strategies are urgently required to treat this disease. Here, we determined whether metformin administration inhibits the growth of PANC-1 and MiaPaCa-2 tumor xenografts in vivo. METHODS Different xenograft models, including orthotopic implantation, were used to determine whether intraperitoneal or oral administration of metformin inhibits the growth of pancreatic cancer in vivo. RESULTS We demonstrate that metformin given once daily intraperitoneally at various doses (50-250 mg/kg) to nude mice inhibited the growth of PANC-1 xenografts in a dose-dependent manner. A significant effect of metformin was obtained at 50 mg/kg and maximal effect at 200 mg/kg. Metformin administration also caused a significant reduction in the phosphorylation of ribosomal S6 protein and ERK in these xenografts. Metformin also inhibited the growth of pancreatic cancer xenografts when administered orally (2.5 mg/mL) either before or after tumor implantation. Importantly, oral administration of metformin also inhibited the growth of MiaPaCa-2 tumors xenografted orthotopically. CONCLUSIONS The studies presented here provide further evidence indicating that metformin offers a potential novel approach for pancreatic ductal adenocarcinoma prevention and therapy.
Collapse
|
18
|
Beck E, Scheen AJ. Quels bénéfices antitumoraux attendre de la metformine ? ANNALES D'ENDOCRINOLOGIE 2013; 74:137-47. [DOI: 10.1016/j.ando.2013.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
19
|
Soares HP, Ni Y, Kisfalvi K, Sinnett-Smith J, Rozengurt E. Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells. PLoS One 2013; 8:e57289. [PMID: 23437362 PMCID: PMC3578870 DOI: 10.1371/journal.pone.0057289] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 01/20/2013] [Indexed: 11/18/2022] Open
Abstract
The mTOR pathway is aberrantly stimulated in many cancer cells, including pancreatic ductal adenocarcinoma (PDAC), and thus it is a potential target for therapy. However, the mTORC1/S6K axis also mediates negative feedback loops that attenuate signaling via insulin/IGF receptor and other tyrosine kinase receptors. Suppression of these feed-back loops unleashes over-activation of upstream pathways that potentially counterbalance the antiproliferative effects of mTOR inhibitors. Here, we demonstrate that treatment of PANC-1 or MiaPaCa-2 pancreatic cancer cells with either rapamycin or active-site mTOR inhibitors suppressed S6K and S6 phosphorylation induced by insulin and the GPCR agonist neurotensin. Rapamycin caused a striking increase in Akt phosphorylation at Ser(473) while the active-site inhibitors of mTOR (KU63794 and PP242) completely abrogated Akt phosphorylation at this site. Conversely, active-site inhibitors of mTOR cause a marked increase in ERK activation whereas rapamycin did not have any stimulatory effect on ERK activation. The results imply that first and second generation of mTOR inhibitors promote over-activation of different pro-oncogenic pathways in PDAC cells, suggesting that suppression of feed-back loops should be a major consideration in the use of these inhibitors for PDAC therapy. In contrast, metformin abolished mTORC1 activation without over-stimulating Akt phosphorylation on Ser(473) and prevented mitogen-stimulated ERK activation in PDAC cells. Metformin induced a more pronounced inhibition of proliferation than either KU63794 or rapamycin while, the active-site mTOR inhibitor was more effective than rapamycin. Thus, the effects of metformin on Akt and ERK activation are strikingly different from allosteric or active-site mTOR inhibitors in PDAC cells, though all these agents potently inhibited the mTORC1/S6K axis.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/pharmacology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Feedback, Physiological/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Indoles/pharmacology
- Insulin/pharmacology
- Metformin/pharmacology
- Morpholines/pharmacology
- Neurotensin/pharmacology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Phosphorylation/drug effects
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Purines/pharmacology
- Pyrimidines/pharmacology
- Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors
- Ribosomal Protein S6 Kinases, 70-kDa/genetics
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- Heloisa P. Soares
- Division of Digestive Diseases, Department of Medicine; CURE: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, United States of America
- Division of Hematology-Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Yang Ni
- Division of Digestive Diseases, Department of Medicine; CURE: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, United States of America
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Krisztina Kisfalvi
- Division of Digestive Diseases, Department of Medicine; CURE: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, United States of America
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine; CURE: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine; CURE: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
20
|
Mitocans, Mitochondria-Targeting Anticancer Drugs. ACTA ACUST UNITED AC 2012. [DOI: 10.1201/b12308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
21
|
Tseng CH. Diabetes, insulin use, and non-Hodgkin lymphoma mortality in Taiwan. Metabolism 2012; 61:1003-9. [PMID: 22237115 DOI: 10.1016/j.metabol.2011.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 02/08/2023]
Abstract
The objective was to evaluate non-Hodgkin lymphoma (NHL) mortality trends and mortality rate ratios between diabetes patients and the general population, and to study NHL risk factors among diabetes patients in Taiwan. A cohort of 80 397 patients with type 2 diabetes mellitus older than 45 years was recruited in 1995-1998 and followed up until 2006. Age-standardized NHL mortality in 1995-2006 was calculated. Non-Hodgkin lymphoma risk factors in diabetes patients were evaluated using Cox regression. Age-standardized NHL mortality trend was steady. Eighty-two male and 69 female diabetes patients died of NHL (crude mortality rates, 35.1 and 23.0 per 100 000 person-years, respectively; corresponding overall mortality rate ratios comparing diabetes patients to the general population, 2.06 and 2.14). The mortality rate ratios were 1.47, 2.33, and 2.78 for men aged at least 65, 55 to 64, and 45 to 54 years, respectively; the corresponding ratios for women were 1.48, 2.22, and 2.79. Age and male sex were significant risk factors, whereas insulin use, diabetes duration, smoking, body mass index, and area of residence were not. Diabetes duration became a significant factor after excluding patients who died of NHL within 5 years of diabetes diagnosis. Patients with diabetes have a higher risk of mortality from NHL, but insulin use is not associated with NHL mortality. Future studies are needed to fully elucidate any association between increased mortality rate ratio and younger age as well as the lack of association between NHL and insulin use demonstrated herein.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
22
|
Abstract
Excess total and cardiovascular morbidity and mortality remain very high among those with type 2 diabetes versus those without diabetes. Clinical trials to lower blood glucose have been disappointing probably because the participants were too late in the natural history of diabetes and already had extensive vascular disease. Insulin resistance measured simply by elevated fasting blood insulin is an early marker of β-cell stress and peripheral insulin resistance. Metformin will prevent development of diabetes among patients with impaired fasting glucose but only for the short term. Metformin reduces risk of coronary heart disease. The drug is safe, low cost, and may also prevent cancer. The combination of diet and exercise followed by metformin in the early phase of "insulin resistance" may reduce or delay both atherosclerosis and arteriosclerosis complications associated with diabetes. Preventive therapy must begin much earlier than before clinical diagnosis of diabetes and aim to initially lower blood insulin levels or insulin resistance.
Collapse
Affiliation(s)
- Lewis H Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 130 North Bellefield Avenue, Pittsburgh, PA 15261, USA.
| |
Collapse
|
23
|
Berstein LM. Metformin in obesity, cancer and aging: addressing controversies. Aging (Albany NY) 2012; 4:320-9. [PMID: 22589237 PMCID: PMC3384433 DOI: 10.18632/aging.100455] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/29/2012] [Indexed: 12/25/2022]
Abstract
Metformin, an oral anti-diabetic drug, is being considered increasingly for treatment and prevention of cancer, obesity as well as for the extension of healthy lifespan. Gradually accumulating discrepancies about its effect on cancer and obesity can be explained by the shortage of randomized clinical trials, differences between control groups (reference points), gender- and age-associated effects and pharmacogenetic factors. Studies of the potential antiaging effects of antidiabetic biguanides, such as metformin, are still experimental for obvious reasons and their results are currently ambiguous. Here we discuss whether the discrepancies in different studies are merely methodological or inherently related to individual differences in responsiveness to the drug.
Collapse
Affiliation(s)
- Lev M Berstein
- Laboratory of Oncoendocrinology, N.N.Petrov Research Institute of Oncology, St. Petersburg, Russia.
| |
Collapse
|
24
|
Appleyard MVCL, Murray KE, Coates PJ, Wullschleger S, Bray SE, Kernohan NM, Fleming S, Alessi DR, Thompson AM. Phenformin as prophylaxis and therapy in breast cancer xenografts. Br J Cancer 2012; 106:1117-22. [PMID: 22361631 PMCID: PMC3304424 DOI: 10.1038/bjc.2012.56] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Observations that diabetics treated with biguanide drugs have a reduced risk of developing cancer have prompted an enthusiasm for these agents as anti-cancer therapies. We sought to determine the efficacy of the biguanide phenformin in the chemoprophylaxis and in the treatment of oestrogen receptor (ER)-positive MCF7 and receptor triple-negative MDAMB231 xenografts in immunocompromised mice. We also compared the efficacy of phenformin and metformin in the treatment of MDAMB231. METHODS Immunocompromised mice were divided into groups: (1) phenformin administered for 2 weeks prior to cell injection; (2) established tumours treated with phenformin; (3) established tumours treated with metformin (only for MDAMB231 tumours); (4) untreated controls. Post-treatment tumours, liver and spleen were harvested for further analysis. RESULTS Phenformin significantly inhibited both the development and growth of MCF7 and MDAMB231 tumours, and for MDAMB231 at greater efficacy than metformin without murine toxicity. The number of mitotic figures was significantly fewer in xenografts treated with phenformin compared with controls. Results suggested that the mechanism of action of phenformin in vivo is consistent with AMPK activation. CONCLUSION Phenformin has clinical potential as an antineoplastic agent and should be considered for clinical trials both in ER-positive and triple-negative breast cancer.
Collapse
Affiliation(s)
- M V C L Appleyard
- Centre for Oncology and Molecular Medicine, University of Dundee, Dundee DD1 9SY, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rohlena J, Dong LF, Ralph SJ, Neuzil J. Anticancer drugs targeting the mitochondrial electron transport chain. Antioxid Redox Signal 2011; 15:2951-74. [PMID: 21777145 DOI: 10.1089/ars.2011.3990] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Mitochondria are emerging as highly intriguing organelles showing promise but that are yet to be fully exploited as targets for anticancer drugs. RECENT ADVANCES A group of compounds that induce mitochondrial destabilization, thereby affecting the physiology of cancer cells, has been defined and termed 'mitocans.' Based on their mode of action of targeting in and around mitochondria, we have placed these agents into several groups including hexokinase inhibitors, compounds targeting Bcl-2 family proteins, thiol redox inhibitors, VDAC/ANT targeting drugs, electron transport chain-targeting drugs, lipophilic cations targeting the inner membrane, agents affecting the tricarboxylic acid cycle, drugs targeting mtDNA, and agents targeting other presently unknown sites. CRITICAL ISSUES Mitocans have a potential to prove highly efficient in suppressing various malignant diseases in a selective manner. They include compounds that are currently in clinical trial and offer substantial promise to become clinically applied drugs. Here we update and redefine the individual classes of mitocans, providing examples of the various members of these groups with a particular focus on agents targeting the electron transport chain, and indicate their potential application in clinical practice. FUTURE DIRECTIONS Even though reactive oxygen species induction is important for the anticancer activity of many mitocans, the precise sequence of events preceding and following this pivotal event are not yet fully clarified, and warrant further investigation. This is imperative for effective deployment of these compounds in the clinic.
Collapse
Affiliation(s)
- Jakub Rohlena
- Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | |
Collapse
|
26
|
Jung JW, Park SB, Lee SJ, Seo MS, Trosko JE, Kang KS. Metformin represses self-renewal of the human breast carcinoma stem cells via inhibition of estrogen receptor-mediated OCT4 expression. PLoS One 2011; 6:e28068. [PMID: 22132214 PMCID: PMC3223228 DOI: 10.1371/journal.pone.0028068] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 10/31/2011] [Indexed: 12/22/2022] Open
Abstract
Metformin, a Type II diabetic treatment drug, which inhibits transcription of gluconeogenesis genes, has recently been shown to lower the risk of some diabetes-related tumors, including breast cancer. Recently, “cancer stem cells” have been demonstrated to sustain the growth of tumors and are resistant to therapy. To test the hypothesis that metformin might be reducing the risk to breast cancers, the human breast carcinoma cell line, MCF-7, grown in 3-dimensional mammospheres which represent human breast cancer stem cell population, were treated with various known and suspected breast cancer chemicals with and without non-cytotoxic concentrations of metformin. Using OCT4 expression as a marker for the cancer stem cells, the number and size were measured in these cells. Results demonstrated that TCDD (100 nM) and bisphenol A (10 µM) increased the number and size of the mammospheres, as did estrogen (10 nM E2). By monitoring a cancer stem cell marker, OCT4, the stimulation by these chemicals was correlated with the increased expression of OCT4. On the other hand, metformin at 1 and 10 mM concentration dramatically reduced the size and number of mammospheres. Results also demonstrated the metformin reduced the expression of OCT4 in E2 & TCDD mammospheres but not in the bisphenol A mammospheres, suggesting different mechanisms of action of the bisphenol A on human breast carcinoma cells. In addition, these results support the use of 3-dimensional human breast cancer stem cells as a means to screen for potential human breast tumor promoters and breast chemopreventive and chemotherapeutic agents.
Collapse
Affiliation(s)
- Ji-Won Jung
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
| | - Sang-Bum Park
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
| | - Soo-Jin Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
| | - Min-Soo Seo
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
| | - James E. Trosko
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Pediatrics and Human Development, College of Human Medicine, Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan, United States of America
| | - Kyung-Sun Kang
- Department of Veterinary Public Health, College of Veterinary Medicine, Adult Stem Cell Research Center, Seoul National University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
27
|
Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 2011; 10:868-80. [PMID: 22037041 DOI: 10.1038/nrd3531] [Citation(s) in RCA: 759] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mammalian target of rapamycin (mTOR) is an atypical protein kinase that controls growth and metabolism in response to nutrients, growth factors and cellular energy levels, and it is frequently dysregulated in cancer and metabolic disorders. Rapamycin is an allosteric inhibitor of mTOR, and was approved as an immuno-suppressant in 1999. In recent years, interest has focused on its potential as an anticancer drug. However, the performance of rapamycin and its analogues (rapalogues) has been undistinguished despite isolated successes in subsets of cancer, suggesting that the full therapeutic potential of targeting mTOR has yet to be exploited. A new generation of ATP-competitive inhibitors that directly target the mTOR catalytic site display potent and comprehensive mTOR inhibition and are in early clinical trials.
Collapse
|
28
|
Tan BX, Yao WX, Ge J, Peng XC, Du XB, Zhang R, Yao B, Xie K, Li LH, Dong H, Gao F, Zhao F, Hou JM, Su JM, Liu JY. Prognostic influence of metformin as first-line chemotherapy for advanced nonsmall cell lung cancer in patients with type 2 diabetes. Cancer 2011; 117:5103-11. [PMID: 21523768 DOI: 10.1002/cncr.26151] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/13/2011] [Accepted: 03/02/2011] [Indexed: 02/05/2023]
Abstract
BACKGROUND It has been reported that antidiabetic drugs affect the risk of cancer and the prognosis of patients with diabetes, but few studies have demonstrated the influence of different antidiabetic agents on outcomes after anticancer therapy among patients with cancer. The objective of this study was to evaluate the influence of the antidiabetic drugs metformin and insulin on the prognosis of patients with advanced nonsmall cell lung cancer (NSCLC) plus type 2 diabetes who received first-line chemotherapy. METHODS Data on patients with NSCLC who had diabetes from 5 hospitals in China during January 2004 to March 2009 were reviewed retrospectively. Ninety-nine patients were included in the final analysis. The influence of metformin and insulin on chemotherapy response rates and survival in these patients was evaluated. RESULTS Chemotherapy with metformin (Group A) produced superior results compared with insulin (Group B) and compared with drugs other than metformin and insulin (Group C) in terms of both progression-free survival (PFS) (8.4 months vs 4.7 months vs 6.4 months, respectively; P = .002) and overall survival (OS) (20.0 months vs 13.1 months vs 13.0 months, respectively; P = .007). Although no significant differences in the response rate (RR) were observed between these 3 groups, when groups B and C (ie, the nonmetformin group) were combined, there was a tendency for better disease control in Group A than that in nonmetformin group. No significant difference in survival was observed between chemotherapy with insulin (Group B) versus other drugs (Group C). CONCLUSIONS The current data suggested that metformin may improve chemotherapy outcomes and survival for patients who have NSCLC with diabetes.
Collapse
Affiliation(s)
- Ben-Xu Tan
- Department of Medical Oncology, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Piątkiewicz P, Czech A. Glucose metabolism disorders and the risk of cancer. Arch Immunol Ther Exp (Warsz) 2011; 59:215-30. [PMID: 21448680 DOI: 10.1007/s00005-011-0119-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 11/29/2010] [Indexed: 12/13/2022]
Abstract
Diabetes and cancer are diseases which take the size of an epidemic spread across the globe. Those diseases are influenced by many factors, both genetic and environmental. Precise knowledge of the complex relationships and interactions between these two conditions is of great importance for their prevention and treatment. Many epidemiological studies have shown that certain types of cancer, especially gastrointestinal cancers (pancreas, liver, colon) and also the urinary and reproductive system cancers in women are more common in patients with diabetes or related metabolic disorders. There are also studies showing the inverse relationship between diabetes and cancer, or the lack of it, but they are less numerous and relate mainly to prostate cancer or squamous cell carcinoma of the esophagus. Epidemiological studies, however, do not say anything about the mechanisms of these dependencies. For this purpose, molecular research is needed on the metabolism of cells (including tumor cells) and on metabolic dysfunctions that arise due to changes in the cell environment taking place in the sick, as well as in the intensely treated human organism.
Collapse
Affiliation(s)
- Paweł Piątkiewicz
- Chair and Department of Internal Medicine and Diabetology, Medical University of Warsaw, Brodnowski Hospital, Kondratowicza 8, 03-242 Warsaw, Poland.
| | | |
Collapse
|
30
|
Lee MS, Hsu CC, Wahlqvist ML, Tsai HN, Chang YH, Huang YC. Type 2 diabetes increases and metformin reduces total, colorectal, liver and pancreatic cancer incidences in Taiwanese: a representative population prospective cohort study of 800,000 individuals. BMC Cancer 2011; 11:20. [PMID: 21241523 PMCID: PMC3031263 DOI: 10.1186/1471-2407-11-20] [Citation(s) in RCA: 361] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 01/18/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metformin protection against cancer risk in Orientals is uncertain. We examined the possible metformin effect on total, esophageal, gastric, colorectal (CRC), hepatocellular (HCC) and pancreatic cancers in a Taiwanese cohort. METHODS A representative sample of 800,000 was drawn from the Taiwanese National Health Insurance data of 2000. A cohort of 480,984 participants 20 years or older, diabetes-cancer-free on 1st January 2000 was formed and categorized as four groups by DM and metformin usage status. Eligible incident cancer events had to occur one year after the index date until the end of 2007. The Cox proportional-hazards model evaluated relative risk of cancer for treated DM patients with or without metformin. The covariates included age, gender, other oral anti-hyperglycemic medication, Charlson comorbidity index (CCI) score and metformin exposure dosage and duration. RESULTS With diabetes but no anti-hyperglycemic medication, cancer incidence density increased at least 2-fold for total, CRC and HCC. On metformin, total, CRC and HCC incidences decreased to near non-diabetic levels but to varying degrees depending on gender and cancer type (CRC in women, liver in men). Adjustment for other oral anti-hyperglycemic agents usage and CCI made the benefit of metformin more evident [hazard ratios (95% confidence intervals): total 0.12 (0.08-0.19), CRC 0.36 (0.13-0.98), liver 0.06 (0.02-0.16), pancreas 0.15 (0.03-0.79)]. There was a significant gender interaction with metformin in CRC which favored women. Metformin dosage for a significant decrease in cancer incidence was ≤ 500 mg/day. CONCLUSIONS Metformin can reduce the incidences of several gastroenterological cancers in treated diabetes.
Collapse
Affiliation(s)
- Meei-Shyuan Lee
- School of Public Health, National Defense Medical Center, Taipei, ROC.
| | | | | | | | | | | |
Collapse
|
31
|
Berstein LM. Modern approach to metabolic rehabilitation of cancer patients: biguanides (phenformin and metformin) and beyond. Future Oncol 2010; 6:1313-23. [PMID: 20799876 DOI: 10.2217/fon.10.87] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Comparing the experience accumulated for more than 40 years in the Laboratory of Endocrinology of Petrov Institute of Oncology (St Petersburg, Russia) with similar approaches practiced elsewhere, evidence supports the reasonability of metabolic rehabilitation of patients suffering from breast cancer or other hormone-dependent malignancies. The primary objective of such approaches is to improve treatment results by ameliorating hormonal-metabolic disturbances, including excess body fat, glucose intolerance, insulin resistance and manifestations of endocrine-genotoxic switchings, and modify tissue and cellular targets or mechanisms related or nondirectly related to the aforementioned disturbances. The relevant measures may be categorized as pharmacological (antidiabetic biguanides exemplified with metformin being most popular but not exclusive) and nonpharmacological (rational nutrition, moderate physical activity and so forth) and used separately or in different combinations.
Collapse
Affiliation(s)
- Lev M Berstein
- N.N.Petrov Research Institute of Oncology, Pesochny-2, Leningradskaja 68, St Petersburg, Russia.
| |
Collapse
|
32
|
Wang KH, Kao AP, Chang CC, Lee JN, Hou MF, Long CY, Chen HS, Tsai EM. Increasing CD44+/CD24(-) tumor stem cells, and upregulation of COX-2 and HDAC6, as major functions of HER2 in breast tumorigenesis. Mol Cancer 2010; 9:288. [PMID: 21044318 PMCID: PMC2989327 DOI: 10.1186/1476-4598-9-288] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 11/02/2010] [Indexed: 12/21/2022] Open
Abstract
Background Cancer cells are believed to arise primarily from stem cells. CD44+/CD24- have been identified as markers for human breast cancer stem cells. Although, HER2 is a well known breast cancer oncogene, the mechanisms of action of this gene are not completely understood. Previously, we have derived immortal (M13SV1), weakly tumorigenic (M13SV1R2) and highly tumorigenic (M13SV1R2N1) cell lines from a breast epithelial cell type with stem cell phenotypes after successive SV40 large T-antigen transfection, X-ray irradiation and ectopic expression of HER2/C-erbB2/neu. Recently, we found that M13SV1R2 cells became non-tumorigenic after growing in a growth factor/hormone-deprived medium (R2d cells). Results In this study, we developed M13SV1R2N1 under the same growth factor/hormone-deprived condition (R2N1d cells). This provides an opportunity to analyze HER2 effect on gene expression associated with tumorigenesis by comparative study of R2d and R2N1d cells with homogeneous genetic background except HER2 expression. The results reveal distinct characters of R2N1d cells that can be ascribed to HER2: 1) development of fast-growing tumors; 2) high frequency of CD44+/CD24- cells (~50% for R2N1d vs. ~10% for R2d); 3) enhanced expression of COX-2, HDAC6 mediated, respectively, by MAPK and PI3K/Akt pathways, and many genes associated with inflammation, metastasis, and angiogenesis. Furthermore, HER2 expression can be down regulated in non-adhering R2N1d cells. These cells showed longer latent period and lower rate of tumor development compared with adhering cells. Conclusions HER2 may induce breast cancer by increasing the frequency of tumor stem cells and upregulating the expression of COX-2 and HDAC6 that play pivotal roles in tumor progression.
Collapse
Affiliation(s)
- Kai-Hung Wang
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Rozengurt E, Sinnett-Smith J, Kisfalvi K. Crosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clin Cancer Res 2010; 16:2505-11. [PMID: 20388847 DOI: 10.1158/1078-0432.ccr-09-2229] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin/insulin-like growth factor 1(IGF-1) receptors and G protein-coupled receptors (GPCR) signaling systems are implicated in autocrine-paracrine stimulation of a variety of malignancies, including ductal adenocarcinoma of the pancreas, one of the most lethal human diseases. Novel targets for pancreatic cancer therapy are urgently needed. We identified a crosstalk between insulin/IGF-1 receptors and GPCR signaling systems in pancreatic cancer cells, leading to enhanced signaling, DNA synthesis, and proliferation. Crosstalk between these signaling systems depends on mammalian target of rapamycin (mTOR) complex 1 (mTORC1). Metformin, the most widely used drug in the treatment of type 2 diabetes, activates AMP kinase (AMPK), which negatively regulates mTORC1. Recent results show that metformin-induced activation of AMPK disrupts crosstalk between insulin/IGF-1 receptor and GPCR signaling in pancreatic cancer cells and inhibits the growth of these cells in xenograft models. Given that insulin/IGF-1 and GPCRs are implicated in other malignancies, a similar crosstalk mechanism may be operative in other cancer cell types. Recent epidemiological studies linked administration of metformin with a reduced risk of pancreatic, breast, and prostate cancer in diabetic patients. We posit that crosstalk between insulin/IGF-1 receptor and GPCR signaling is a mechanism for promoting the development of certain types of cancer and a target for the prevention and therapy of these diseases via metformin administration.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095-1786, USA.
| | | | | |
Collapse
|