51
|
Hoseini Shafa M, Jalal R, Kosari N, Rahmani F. Efficacy of metformin in mediating cellular uptake and inducing apoptosis activity of doxorubicin. Regul Toxicol Pharmacol 2018; 99:200-212. [PMID: 30266241 DOI: 10.1016/j.yrtph.2018.09.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 08/11/2018] [Accepted: 09/23/2018] [Indexed: 01/08/2023]
Abstract
The clinical use of doxorubicin (DOX) is limited due to its systemic side effects and drug resistance. Recent evidence suggests that metformin prevents and controls certain but not all types of cancer. The beneficial use of metformin in combination with some chemotherapeutic agents has been reported. The aim of this study is to investigate the influence of metformin on DOX-induced effects in human prostate DU145 cancer cells and clarify its molecular mechanisms. For this purpose, DU145 cells were treated with DOX or metformin, either alone or in combination with each other. The proliferation of DU145 cells was inhibited by DOX-alone and metformin-alone treatment in a time and dose-dependent manner. Metformin could enhance the cytotoxicity of DOX by increasing DOX cellular uptake and cell cycle arrest at G1/S checkpoint which is associated with the enhancement of p21 protein expression. Moreover, metformin could elevate DOX-induced apoptosis in DU145 cells in a concentration-dependent manner and DOX-induced caspase-3 activity. These findings suggest that the combined treatment of metformin with DOX potentiates the anticancer efficacy of DOX in DU145 cells via inhibiting ABCB1 function, cell cycle arrest at G1/S transition and apoptosis induction.
Collapse
Affiliation(s)
- Maryam Hoseini Shafa
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Razieh Jalal
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Negin Kosari
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
52
|
Effects of metformin on the PI3K/AKT/FOXO1 pathway in anaplastic thyroid Cancer cell lines. ACTA ACUST UNITED AC 2018; 26:93-103. [PMID: 30242671 PMCID: PMC6279666 DOI: 10.1007/s40199-018-0208-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/18/2018] [Indexed: 01/07/2023]
Abstract
Background The PI3K/AKT/FOXO signaling pathway plays an important role in the survival, proliferation and apoptosis of tumor cells. The aim of the present study was to explore whether metformin could affect insulin-promoting cell growth by regulation of this pathway. Material and methods Anaplastic thyroid cancer cells were treated with 0–60 mM metformin for 24, 48 and 72 h. Cell viability, morphology, apoptosis and migration were investigated by MTT assay, microscopy observation, AnexinV-PI and the wound healing assay, respectively. Expression levels of PI3K, AKT and FOXO1 were detected by RT-qPCR, and proteins phosphorylated levels were determined by ELISA. Results Metformin decreased cell viability and migration in a significant time-and dose-dependent manner, and induced apoptosis and morphological changes in the cells. RT-qPCR results showed that expression levels of PI3K, AKT and FOXO1 was inhibited by metformin (P < 0.05). However, there was no significant change in the expression level of AKT following metformin treatment for C643 cell line (P > 0.05). ELISA results showed that metformin treatment had no significant effects on the phosphorylated levels of PI3K, AKT and FOXO1 (P > 0.05). Conclusuion The downregulation of FOXO1 was intensified by metformin, but no increase in cell viability was observed following FOXO1 downregulation by metformin. However, the exact molecular mechanism of metformin on inhibition of the PI3K/AKT pathway and subsequent decrease in cell viability remains unclear and further studies are required for its clarification. Electronic supplementary material The online version of this article (10.1007/s40199-018-0208-2) contains supplementary material, which is available to authorized users.
Collapse
|
53
|
Anastasi E, Filardi T, Tartaglione S, Lenzi A, Angeloni A, Morano S. Linking type 2 diabetes and gynecological cancer: an introductory overview. Clin Chem Lab Med 2018; 56:1413-1425. [PMID: 29427549 DOI: 10.1515/cclm-2017-0982] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/03/2018] [Indexed: 01/03/2025]
Abstract
Type 2 diabetes (T2D) is a chronic disease with a growing prevalence and a leading cause of death in many countries. Several epidemiological studies observed an association between T2D and increased risk of many types of cancer, such as gynecologic neoplasms (endometrial, cervical, ovarian and vulvar cancer). Insulin resistance, chronic inflammation and high free ovarian steroid hormones are considered the possible mechanisms behind this complex relationship. A higher risk of endometrial cancer was observed in T2D, even though this association largely attenuated after adjusting for obesity. A clear relationship between the incidence of cervical cancer (CC) and T2D has still not be determined; however T2D might have an impact on prognosis in patients with CC. To date, studies on the association between T2D and ovarian cancer (OC) are limited. The effect of pre-existing diabetes on cancer-specific mortality has been evaluated in several studies, with less clear results. Other epidemiological and experimental studies focused on the potential role of diabetes medications, mainly metformin, in cancer development in women. The correct understanding of the link between T2D and gynecologic cancer risk and mortality is currently imperative to possibly modify screening and diagnostic-therapeutic protocols in the future.
Collapse
Affiliation(s)
- Emanuela Anastasi
- Department of Molecular Medicine, University "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy, Phone: +39 064472347, Fax: +39 064478381
| | - Tiziana Filardi
- Department of Experimental Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Sara Tartaglione
- Department of Molecular Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - Susanna Morano
- Department of Experimental Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| |
Collapse
|
54
|
Rindone GM, Gorga A, Regueira M, Pellizzari EH, Cigorraga SB, Galardo MN, Meroni SB, Riera MF. Metformin counteracts the effects of FSH on rat Sertoli cell proliferation. Reproduction 2018; 156:93-101. [DOI: 10.1530/rep-18-0233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/21/2018] [Indexed: 12/16/2022]
Abstract
Metformin (MET) is one of the most widely used anti-hyperglycemic agents for treating patients with type 2 diabetes and it has started to be used in pediatric population at ages when Sertoli cells are still proliferating. It is well known that follicle-stimulating hormone (FSH) is the major Sertoli cell mitogen. The aim of the study is to investigate a possible effect of MET, which has been shown to have anti-proliferative properties, on FSH regulation of postnatal Sertoli cell proliferation and on the molecular mechanisms involved in this regulation. The present study was performed in eight-day-old rat Sertoli cell cultures. The results obtained show that MET in the presence of FSH increases phosphorylated acetyl-CoA carboxylase and decreases phosphorylated p70S6K levels. Moreover, we show that MET decreases FSH-stimulated Sertoli cell proliferation, and this decrease is accompanied by a reduction in FSH-stimulated Ccnd1 and Ccnd2 expression and an increase in cell cycle inhibitor p21Cip expression. Altogether, these results suggest that MET can, at least in part, counteract the effect of FSH on postnatal Sertoli cell proliferation.
Collapse
|
55
|
Prognosis of ovarian cancer in women with type 2 diabetes using metformin and other forms of antidiabetic medication or statins: a retrospective cohort study. BMC Cancer 2018; 18:767. [PMID: 30055585 PMCID: PMC6064082 DOI: 10.1186/s12885-018-4676-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 12/15/2022] Open
Abstract
Background Ovarian cancer is one of the most lethal cancers and women with type 2 diabetes (T2D) have even poorer survival from it. We assessed the prognosis of ovarian cancer in women with type 2 diabetes treated with metformin, other forms of antidiabetic medication, or statins. Methods Study cohort consisted of women with T2D diagnosed with ovarian cancer in Finland 1998–2011. They were identified from a nationwide diabetes database (FinDM), being linked to several national registers. Patients were grouped according to their medication in the three years preceding ovarian cancer diagnosis. The Aalen–Johansen estimator was used to describe cumulative mortality from ovarian cancer and from other causes in different medication groups. Mortality rates were analysed by Cox models, and adjusted hazard ratios (HR) with 95% confidence intervals (95% CIs) were estimated in relation to the use of different forms of medication. Main outcome measures were death from ovarian cancer and death from other causes. Results During the accrual period 421 newly diagnosed ovarian cancers were identified in the FinDM database. No evidence was found for any differences in mortality from ovarian cancer or other causes between different antidiabetic medication groups. Pre-diagnostic use of statins was observed to be associated with decreased mortality from ovarian cancer compared with no such use (HR 0.72, 95% CI 0.56–0.93). Conclusions Our findings are inconclusive as regards the association between metformin and ovarian cancer survival. However, some evidence was found for improved prognosis of ovarian cancer with pre-diagnostic statin use, requiring cautious interpretation, though. Electronic supplementary material The online version of this article (10.1186/s12885-018-4676-z) contains supplementary material, which is available to authorized users.
Collapse
|
56
|
Liu Y, Feng Y, Liu H, Wu J, Tang Y, Wang Q. Real-time assessment of platinum sensitivity of primary culture from a patient with ovarian cancer with extensive metastasis and the platinum sensitivity enhancing effect by metformin. Oncol Lett 2018; 16:4253-4262. [PMID: 30250536 PMCID: PMC6144930 DOI: 10.3892/ol.2018.9223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/14/2018] [Indexed: 01/30/2023] Open
Abstract
The aim of the present study was to perform a rapid evaluation of the efficiency of commonly used platinum-based chemotherapy regimens for patients with ovarian cancer with extensive metastases using an in vitro method combined with culturing primary cells and real-time monitoring, and to further explore the enhanced effect of metformin on susceptibility of ovarian cancer cells to platinum-based chemotherapy. The primary omental metastatic (OM) cells were isolated from the omentum metastasis of a surgical patient with stage IIIc ovarian carcinoma. Drug sensitivity was evaluated using the xCELLigence system, and screening of the most effective platinum chemotherapy was performed through analysis of cell susceptibility to cisplatin, carboplatin, nedaplatin and paclitaxel or docetaxel alone or in combination. At the same time, this system was used to determine whether metformin was able to increase the sensitivity of cancer cells to platinum chemotherapy. The results revealed that nedaplatin exhibited the most marked cytotoxic effect on the OM cells, followed by those of carboplatin and cisplatin. The addition of docetaxel enhanced the cytotoxic effect, and the combination of platinum and paclitaxel also enhanced the effect. Metformin rapidly increased the sensitivity of cells to platinum-based chemotherapy, and this effect was dose-dependent. The sensitivity of OM cells to different platinum-based regimens was varied. The effect of metformin on chemotherapeutic sensitization of cancer cells is clear in vitro, and the real-time cell analyzer assay has the potential to assist in determining individualized drug regimens for patients with metastatic ovarian cancer.
Collapse
Affiliation(s)
- Yingzhao Liu
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yan Feng
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hongmei Liu
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jianyong Wu
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yong Tang
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Urology Department, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530199, P.R. China
| | - Qi Wang
- Research Department, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China.,Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
57
|
Hu J, Chen JB, Cui Y, Zhu YW, Ren WB, Zhou X, Liu LF, Chen HQ, Zu XB. Association of metformin intake with bladder cancer risk and oncologic outcomes in type 2 diabetes mellitus patients: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e11596. [PMID: 30045293 PMCID: PMC6078654 DOI: 10.1097/md.0000000000011596] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recent clinical trials indicated that metformin intake might play a protective role in the incidence and oncologic outcomes of various cancers. However, its protective effect on bladder cancer remains uncertain. METHODS We performed a meta-analysis to investigate the association between metformin intake and bladder cancer risk as well as oncologic outcomes in diabetes mellitus (DM) patients. A comprehensive literature search was performed using PubMed, Embase, and the Cochrane Central Search Library in December 2017. Hazard ratio (HR) with 95% confidence interval (CI) was pooled. RESULTS A total of 9 retrospective cohort studies with 1,270,179 patients were included. A meta-analysis revealed that metformin intake was associated with an increased recurrence-free survival (HR = 0.55, 95% confidence interval [CI] = 0.35-0.88; P = .01; I = 64%), improved progression-free survival (HR = 0.70, 95% CI = 0.51-0.96; P = .03; I = 33%), and prolonged cancer-specific survival (HR = 0.57, 95% CI = 0.40-0.81; P = .002; I = 0%). However, results demonstrated that metformin intake was not associated with a decreased incidence of bladder cancer (HR = 0.82, 95% CI = 0.61-1.09; P = .17; I = 85%) or an increased overall survival in bladder cancer patients (HR = 0.83, 95% CI = 0.47-1.44; P = .50; I = 64%). CONCLUSION The present meta-analysis indicated that metformin intake could improve the prognosis of bladder cancer patients. Further prospective cohort studies and mechanistic studies are still required to determine the precise role of metformin in the initiation and progression of bladder cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xu Zhou
- Department of Urology
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | | | | | | |
Collapse
|
58
|
Al Hassan M, Fakhoury I, El Masri Z, Ghazale N, Dennaoui R, El Atat O, Kanaan A, El-Sibai M. Metformin Treatment Inhibits Motility and Invasion of Glioblastoma Cancer Cells. Anal Cell Pathol (Amst) 2018; 2018:5917470. [PMID: 30046513 PMCID: PMC6038689 DOI: 10.1155/2018/5917470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/07/2018] [Accepted: 04/18/2018] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and deadliest cancers of the central nervous system (CNS). GBMs high ability to infiltrate healthy brain tissues makes it difficult to remove surgically and account for its fatal outcomes. To improve the chances of survival, it is critical to screen for GBM-targeted anticancer agents with anti-invasive and antimigratory potential. Metformin, a commonly used drug for the treatment of diabetes, has recently emerged as a promising anticancer molecule. This prompted us, to investigate the anticancer potential of metformin against GBMs, specifically its effects on cell motility and invasion. The results show a significant decrease in the survival of SF268 cancer cells in response to treatment with metformin. Furthermore, metformin's efficiency in inhibiting 2D cell motility and cell invasion in addition to increasing cellular adhesion was also demonstrated in SF268 and U87 cells. Finally, AKT inactivation by downregulation of the phosphorylation level upon metformin treatment was also evidenced. In conclusion, this study provides insights into the anti-invasive antimetastatic potential of metformin as well as its underlying mechanism of action.
Collapse
Affiliation(s)
- Marwa Al Hassan
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Isabelle Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Zeinab El Masri
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Noura Ghazale
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Rayane Dennaoui
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Oula El Atat
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, El-Kurah, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
59
|
Songthaveesin C, Sa-Nongdej W, Limboonreung T, Chongthammakun S. Combination of metformin and 9-cis retinoic acid increases apoptosis in C6 glioma stem-like cells. Heliyon 2018; 4:e00638. [PMID: 29872770 PMCID: PMC5986546 DOI: 10.1016/j.heliyon.2018.e00638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/09/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma (GBM) is the most commonly diagnosed type of brain cancer and the leading cause of brain cancer-related death. GBM contains a subpopulation of tumor-propagating glioblastoma stem-like cells that are thought to drive cancer progression and recurrence. Although several clinical trials are ongoing to explore new chemotherapeutic agents to treat GBM, the use of metformin (Met), a first-line drug for type 2 diabetes mellitus, in cancer remains controversial. Here, we show that combining Met with 9-cis retinoic acid (9-cis RA) reduced the proliferation rate of C6-GSCs (glioblastoma stem-like cells) in vitro. The results of flow cytometric analysis showed that treatment with 9-cis RA for 24 h induced 4.5% early and 38.0% late apoptosis in C6-GSCs. Twenty-four hours of Met treatment induced 23.6% early and 33.5% late apoptosis in C6-GSCs. Combination of Met and 9-cis RA treatment significantly increased both early and late apoptosis to 30.4% and 55.4%, respectively. The present findings suggest that not only 9-cis RA but also Met has the potential to induce early and late apoptotic GSCs death by affecting the functional cytoplasmic and nuclear organelles. At the protein level, there was increased cleaved caspase-3 but decreased procaspase-3 expression in Met-, 9-cis RA- and Met+9-cis RA-treated C6 GSCs, as detected by western blotting. The ratio of cleaved caspase-3/procaspase-3 was 1.6 times higher in Met+9-cis RA-treated groups compared to control. Ultimately, a combination of Met and 9-cis RA might be a possible therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Chanchai Songthaveesin
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Wanna Sa-Nongdej
- School of Nursing, Ramathibodi Hospital, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Tanapol Limboonreung
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Sukumal Chongthammakun
- Department of Anatomy and Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
60
|
Garrido MP, Vera C, Vega M, Quest AFG, Romero C. Metformin prevents nerve growth factor-dependent proliferative and proangiogenic effects in epithelial ovarian cancer cells and endothelial cells. Ther Adv Med Oncol 2018; 10:1758835918770984. [PMID: 29774060 PMCID: PMC5949935 DOI: 10.1177/1758835918770984] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/02/2018] [Indexed: 02/05/2023] Open
Abstract
Background Epithelial ovarian cancer (EOC) is characterized by exacerbated angiogenesis regulated by proangiogenic and growth factors. Nerve growth factor (NGF) is overexpressed in EOC where it promotes proliferation as well as survival and is considered a proangiogenic factor. Metformin, a drug commonly used in the treatment of diabetes, is attributed to antineoplastic effects, but the underlying mechanisms remain unknown. Given that current therapies yield modest results in EOC patients, the aim of this study was to determine the effects of metformin on NGF-enhanced proliferation of EOC cells and the angiogenic potential of endothelial cells. Methods A2780 (EOC), HOSE (human ovarian surface epithelial) and EA.hy926 (endothelial) cells were treated with NGF and metformin. Cell viability, cell proliferation and cell cycle were evaluated in all three cell lines, and the angiogenic potential in endothelial EA.hy926 cells. Results NGF enhanced cell proliferation in A2780, HOSE and EA.hy926 cells (p < 0.05), while metformin treatment decreased cell proliferation in A2780 and EA.hy926 cells (p < 0.05). Moreover, the NGF-enhanced angiogenic score in EA.hy926 cells was prevented by metformin (p < 0.05). Conclusions Given that NGF plays a significant role in EOC progression, our current findings suggest that metformin holds considerable promise as an adjuvant treatment in ovarian cancer.
Collapse
Affiliation(s)
- Maritza P Garrido
- Laboratory of Endocrinology and Reproductive Biology, Hospital Clínico Universidad de Chile, Santiago, Chile Obstetrics and Gynecology Department, Medicine School, Universidad de Chile, Santiago, Chile
| | - Carolina Vera
- Laboratory of Endocrinology and Reproductive Biology, Hospital Clínico Universidad de Chile, Santiago, Chile Obstetrics and Gynecology Department, Medicine School, Universidad de Chile, Santiago, Chile
| | - Margarita Vega
- Laboratory of Endocrinology and Reproductive Biology, Hospital Clínico Universidad de Chile, Santiago, Chile Obstetrics and Gynecology Department, Medicine School, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Facultad de Medicina, Universidad de Chile, Santiago, Chile Laboratorio de Comunicaciones Celulares, Centro de Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC) Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad De Medicina, Universidad de Chile, Santiago, Chile Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Carmen Romero
- Hospital Clínico Universidad de Chile, Santos Dumont 999, Santiago 8380456, Chile; Laboratory of Endocrinology and Reproductive Biology, Hospital Clínico Universidad de Chile, Santiago, Chile; Obstetrics and Gynecology Department, Medicine School, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| |
Collapse
|
61
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Synergistic cytotoxicity of the dipeptidyl peptidase-IV inhibitor gemigliptin with metformin in thyroid carcinoma cells. Endocrine 2018; 59:383-394. [PMID: 29285650 DOI: 10.1007/s12020-017-1503-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/12/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE The influence of the dipeptidyl peptidase-IV inhibitor, gemigliptin alone or in combination with metformin on survival, proliferation, and migration of thyroid carcinoma cells was investigated. METHODS SW1736 and TPC-1 human thyroid carcinoma cells were used. RESULTS Gemigliptin and metformin caused cell death in a dose-dependent manner. In cells treated with both gemigliptin and metformin, compared with metformin alone, all of the combination index values were lower than 1.0, suggesting synergistic cytotoxicity of two agents. Cell viability, the percentage of viable cells, ATP levels, and mitochondrial membrane potential decreased; however, cytotoxic activity, and the protein levels of cleaved PARP, phospho-Akt and phospho-AMP-activated protein kinase (AMPK) increased. Administration of wortmannin, but not compound C, further decreased cell viability, and further increased cytotoxic activity. Moreover, compared with control, cell proliferation and migration as well as the protein levels of p53, p21, vascular cell adhesion molecule-1 (VCAM-1), and phospho-extracellular signal-regulated kinase (ERK) 1/2 decreased. The decrement of matrix metalloproteinase-2 and matrix metalloproteinase-9 protein levels was cell specific. CONCLUSIONS Our results demonstrate that gemigliptin induces cytotoxic activity, and has a synergistic activity with metformin in inducing cytotoxicity via regulation of Akt and AMPK in thyroid carcinoma cells. Furthermore, gemigliptin augments the inhibitory effect of metformin on proliferation and migration through involvement of matrix metalloproteinase-2, matrix metalloproteinase-9, p53, p21, VCAM-1, and ERK in thyroid carcinoma cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jun Goo Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Chul Sik Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sung-Hee Ihm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Moon Gi Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hyung Joon Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Seong Jin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea.
| |
Collapse
|
62
|
Gonçalves GA. p27 kip1 as a key regulator of endometriosis. Eur J Obstet Gynecol Reprod Biol 2017; 221:1-4. [PMID: 29216564 DOI: 10.1016/j.ejogrb.2017.11.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 11/05/2017] [Accepted: 11/30/2017] [Indexed: 01/04/2023]
Abstract
p27kip1 as a key regulator of endometriosis Gonçalves GA p27kip1 is a cyclin-dependent kinase (CDK) inhibitor whose specific late G1 destruction allows progression of the cell across the G1/S boundary. There is a direct relationship between low level of p27 and rapid proliferation occurring in several benign states and in many malignances. In the glandular cells of the normal endometrium, the level of p27kip1 is exceedingly low during the proliferative phase, whereas it is markedly increased during the secretory phase. The expression of p27kip1 in endometriosis is very low but has been found to increase following treatment with progesterone. However, estrogen exposure is considered as a major risk factor in developing endometrial cancer. Endometriosis endometrial cells cultures have also lower levels of p27kip1 compared to heath endometrial cells cultures and restore the cell cycle balance when transduced with an adenoviral vector carring the p27kip1 coding gene (Adp27EGFP). More uniform and rigorous studies are required to confirm these and additional markers utility in a diagnostic and possible treatment panel. As a major clinical priority is to determine which lesions can be treated medically and which require surgical intervention, focusing future studies on markers that distinguish response to hormone therapy or are involved in hormone regulation, will be important future considerations. The goal of this highlight review is to provide a broad overview of the advancements in studies about endometriosis mainly correlating the cytokine p27kip1 expression with the diagnostic and disease treatment.
Collapse
Affiliation(s)
- G A Gonçalves
- Faculdades Integradas Padre Albino (FIPA), Rua dos Estudantes, 225, Cep - 15.809 -144, Catanduva, São Paulo, Brazil.
| |
Collapse
|
63
|
Forcato S, Novi DRBDS, Costa NO, Borges LI, Góes MLMD, Ceravolo GS, Gerardin DCC. In utero and lactational exposure to metformin induces reproductive alterations in male rat offspring. Reprod Toxicol 2017; 74:48-58. [DOI: 10.1016/j.reprotox.2017.08.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023]
|
64
|
Doherty JA, Jensen A, Kelemen LE, Pearce CL, Poole E, Schildkraut JM, Terry KL, Tworoger SS, Webb PM, Wentzensen N. Current Gaps in Ovarian Cancer Epidemiology: The Need for New Population-Based Research. J Natl Cancer Inst 2017; 109:3847624. [PMID: 29117355 DOI: 10.1093/jnci/djx144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/19/2017] [Indexed: 12/25/2022] Open
Abstract
With recent ovarian cancer screening studies showing no clinically significant mortality benefit, preventing this disease, identifying high-risk populations, and extending survival remain priorities. However, several challenges are impeding progress in ovarian cancer research. With most studies capturing exposure information from 10 or more years ago, evaluation of how changing patterns of exposures, such as new oral contraceptive formulations and increased intrauterine device use, might influence ovarian cancer risk and survival is difficult. Risk factors for ovarian cancer should be evaluated in the context of tumor histotypes, which have unique molecular features and cells of origin; this is a task that requires large collaborative studies to achieve meaningful sample sizes. Importantly, identification of novel modifiable risk factors, in addition to those currently known to reduce risk (eg, childbearing, tubal ligation, oral contraceptive use), is needed; this is not feasibly implemented at a population level. In this Commentary, we describe important gaps in knowledge and propose new approaches to advance epidemiologic research to improve ovarian cancer prevention and survival, including updated classification of tumors, collection of data on changing and novel exposures, longer follow-up on existing studies, evaluation of diverse populations, development of better risk prediction models, and collaborating prospectively with consortia to develop protocols for new studies that will allow seamless integration for future pooled analyses.
Collapse
Affiliation(s)
| | - Jennifer A Doherty
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Allan Jensen
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Linda E Kelemen
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Celeste L Pearce
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Elizabeth Poole
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Joellen M Schildkraut
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Kathryn L Terry
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Shelley S Tworoger
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Penelope M Webb
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| | - Nicolas Wentzensen
- Affiliations of authors: Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT (JAD); Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark (AJ); Department of Public Health Sciences and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC (LEK); Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI (CLP); Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA (CLP); Channing Division of Network Medicine (EP, SST) and Obstetrics and Gynecology Epidemiology Center (KLT), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA (EP, SST, KLT); Department of Public Health Sciences, University of Virginia, Charlottesville, CA (JMS); Population Health Department, QIMR Berghofer Medical Research Institute, Herston, Australia (PMW); Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD (NW)
| |
Collapse
|
65
|
Gong J, Kelekar G, Shen J, Shen J, Kaur S, Mita M. The expanding role of metformin in cancer: an update on antitumor mechanisms and clinical development. Target Oncol 2017; 11:447-67. [PMID: 26864078 DOI: 10.1007/s11523-016-0423-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metformin has been used for nearly a century to treat type 2 diabetes mellitus. Epidemiologic studies first identified the association between metformin and reduced risk of several cancers. The anticancer mechanisms of metformin involve both indirect or insulin-dependent pathways and direct or insulin-independent pathways. Preclinical studies have demonstrated metformin's broad anticancer activity across a spectrum of malignancies. Prospective clinical trials involving metformin in the chemoprevention and treatment of cancer now number in the hundreds. We provide an update on the anticancer mechanisms of metformin and review the results thus far available from prospective clinical trials investigating metformin's efficacy in cancer.
Collapse
Affiliation(s)
- Jun Gong
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gauri Kelekar
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - James Shen
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John Shen
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sukhpreet Kaur
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Monica Mita
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Experimental Therapeutics Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, SCCT Mezzanine MS 35, Los Angeles, CA, 90048, USA.
| |
Collapse
|
66
|
Sereni MI, Baldelli E, Gambara G, Ravaggi A, Hodge KA, Alberts DS, Guillen-Rodriguez JM, Dong T, Memo M, Odicino F, Angioli R, Liotta LA, Pecorelli SL, Petricoin EF, Pierobon M. Kinase-driven metabolic signalling as a predictor of response to carboplatin-paclitaxel adjuvant treatment in advanced ovarian cancers. Br J Cancer 2017; 117:494-502. [PMID: 28664915 PMCID: PMC5558684 DOI: 10.1038/bjc.2017.195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/19/2017] [Accepted: 06/01/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The biological mechanisms underlying early- and advanced-stage epithelial ovarian cancers (EOCs) are still poorly understood. This study explored kinase-driven metabolic signalling in early and advanced EOCs, and its role in tumour progression and response to carboplatin-paclitaxel treatment. METHODS Tumour epithelia were isolated from two independent sets of primary EOC (n=72 and 30 for the discovery and the validation sets, respectively) via laser capture microdissection. Reverse phase protein microarrays were used to broadly profile the kinase-driven metabolic signalling of EOC with particular emphasis on the LBK1-AMPK and AKT-mTOR axes. Signalling activation was compared between early and advanced lesions, and carboplatin-paclitaxel-sensitive and -resistant tumours. RESULTS Advanced EOCs were characterised by a heterogeneous kinase-driven metabolic signature and decreased phosphorylation of the AMPK-AKT-mTOR axis compared to early EOC (P<0.05 for AMPKα T172, AMPKα1 S485, AMPKβ1 S108, AKT S473 and T308, mTOR S2448, p70S6 S371, 4EBP1 S65, GSK-3 α/β S21/9, FOXO1 T24/FOXO3 T32, and FOXO1 S256). Advanced tumours with low relative activation of the metabolic signature and increased FOXO1 T24/FOXO3 T32 phosphorylation (P=0.041) were associated with carboplatin-paclitaxel resistance. CONCLUSIONS If validated in a larger cohort of patients, the decreased AMPK-AKT-mTOR activation and phosphorylation of FOXO1 T24/FOXO3 T32 may help identify carboplatin-paclitaxel-resistant EOC patients.
Collapse
Affiliation(s)
- Maria Isabella Sereni
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
- Department of Obstetrics and Gynecology, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Roma, Italy
| | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - Guido Gambara
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - Antonella Ravaggi
- Division of Gynecologic Oncology, ‘Angelo Nocivelli’ Institute of Molecular Medicine, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - K Alex Hodge
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - David S Alberts
- The University of Arizona Cancer Center, 3838N Campbell Ave, Tucson, AZ 85719, USA
| | | | - Ting Dong
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Franco Odicino
- Division of Gynecologic Oncology, ‘Angelo Nocivelli’ Institute of Molecular Medicine, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Roberto Angioli
- Department of Obstetrics and Gynecology, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Roma, Italy
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - Sergio L Pecorelli
- Division of Gynecologic Oncology, ‘Angelo Nocivelli’ Institute of Molecular Medicine, University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, Manassas, VA 20110, USA
| |
Collapse
|
67
|
Wang SB, Lei KJ, Liu JP, Jia YM. Continuous use of metformin can improve survival in type 2 diabetic patients with ovarian cancer: A retrospective study. Medicine (Baltimore) 2017; 96:e7605. [PMID: 28723808 PMCID: PMC5521948 DOI: 10.1097/md.0000000000007605] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Evidence indicates that type 2 diabetes may stimulate the initiation and progression of several types of cancer. Metformin, a drug most commonly used to treat type 2 diabetes, may inhibit cancer cell growth and reduce the risk of cancer. However, evidence of the antitumor effects of metformin on ovarian cancer is still limited.In this study, we retrospectively examined the effects of metformin on ovarian cancer patients with diabetes at our institution.We identified 568 consecutive patients who were newly diagnosed with ovarian cancer and treated between January 2011 and March 2014. Patients with International Federation of Gynecology and Obstetrics (FIGO) stage I to IV epithelial ovarian, fallopian, or peritoneal cancer were included. Patients with type 1 diabetes, incomplete records (including medication records) and any other cancer before their ovarian cancer diagnosis, as well as those diagnosed with diabetes more than 6 months after their ovarian cancer diagnosis, were excluded. Out of 568 patients, 48 (8.5%) patients with type 2 diabetes continuously used metformin, 34 (5.9%) patients with type 2 diabetes did not take metformin, 22 (3.9%) patients with type 2 diabetes discontinued metformin, and 464 (81.7%) ovarian cancer patients were nondiabetic controls. Longer progression-free survival (PFS) and overall survival (OS) were observed in ovarian cancer patients with diabetes who were taking metformin than in diabetic patients not taking metformin, diabetic patients who discontinued metformin, and nondiabetic ovarian cancer patients (P = .001). After adjusting for possible confounders, metformin use was associated with a lower risk for disease relapse [hazard ratio (HR) = 0.34; 95% confidence interval (CI): 0.27-0.67; P < .01] and disease-related death (HR = 0.29; 95% CI: 0.13-0.58, P = .03) among ovarian cancer patients with diabetes.Metformin use may decrease the risk for disease recurrence and death in patients with ovarian cancer, but the drug treatment must be continuous.
Collapse
Affiliation(s)
| | | | - Jia-Pei Liu
- Laboratory Medicine, the Second People's Hospital of Yibin City, Yibin, Sichuan, China
| | | |
Collapse
|
68
|
Mohamed Suhaimi NA, Phyo WM, Yap HY, Choy SHY, Wei X, Choudhury Y, Tan WJ, Tan LAPY, Foo RSY, Tan SHS, Tiang Z, Wong CF, Koh PK, Tan MH. Metformin Inhibits Cellular Proliferation and Bioenergetics in Colorectal Cancer Patient-Derived Xenografts. Mol Cancer Ther 2017; 16:2035-2044. [PMID: 28533437 DOI: 10.1158/1535-7163.mct-16-0793] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 02/06/2023]
Abstract
There is increasing preclinical evidence suggesting that metformin, an antidiabetic drug, has anticancer properties against various malignancies, including colorectal cancer. However, the majority of evidence, which was derived from cancer cell lines and xenografts, was likely to overestimate the benefit of metformin because these models are inadequate and require supraphysiologic levels of metformin. Here, we generated patient-derived xenograft (PDX) lines from 2 colorectal cancer patients to assess the properties of metformin and 5-fluorouracil (5-FU), the first-line drug treatment for colorectal cancer. Metformin (150 mg/kg) as a single agent inhibits the growth of both PDX tumors by at least 50% (P < 0.05) when administered orally for 24 days. In one of the PDX models, metformin given concurrently with 5-FU (25 mg/kg) leads to an 85% (P = 0.054) growth inhibition. Ex vivo culture of organoids generated from PDX demonstrates that metformin inhibits growth by executing metabolic changes to decrease oxygen consumption and activating AMPK-mediated pathways. In addition, we also performed genetic characterizations of serial PDX samples with corresponding parental tissues from patients using next-generation sequencing (NGS). Our pilot NGS study demonstrates that PDX represents a useful platform for analysis in cancer research because it demonstrates high fidelity with parental tumor. Furthermore, NGS analysis of PDX may be useful to determine genetic identifiers of drug response. This is the first preclinical study using PDX and PDX-derived organoids to investigate the efficacy of metformin in colorectal cancer. Mol Cancer Ther; 16(9); 2035-44. ©2017 AACR.
Collapse
Affiliation(s)
| | - Wai Min Phyo
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Hao Yun Yap
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | | | - Xiaona Wei
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Yukti Choudhury
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Wai Jin Tan
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | | | - Roger Sik Yin Foo
- Genome Institute of Singapore, Singapore.,Cardiovascular Research Institute, National University Health Systems, Singapore
| | | | | | | | | | - Min-Han Tan
- Institute of Bioengineering and Nanotechnology Singapore, Singapore. .,Concord Cancer Hospital Singapore, Singapore.,National Cancer Centre Singapore, Singapore
| |
Collapse
|
69
|
EL-Arabey AA. New insight for metformin against bladder cancer. Genes Environ 2017; 39:13. [PMID: 28373897 PMCID: PMC5376285 DOI: 10.1186/s41021-017-0074-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/03/2017] [Indexed: 01/26/2023] Open
Abstract
International Agency for Research on Cancer (IARC) estimated that bladder cancer is the ninth most common cancer in the world, with 430,000 new cases and 165,000 deaths in 2012. Bladder cancer represents the fourth most common cancer in men and ninth most common cancer in women. It is the second most prevalent cancer in men 60 years of age or older in United States. Looking further down, continuing advancements in cancer research could potentially offer more choices for clinician and patient with longer survival and better quality of life. Although, bladder cancer represents an ideal tumor model to test and apply cancer prevention strategies; there are limited studies about application of metformin in the management of bladder cancer. Here, I will shed light on the proposed mechanisms of anti-carcinogenic effects of metformin and cohort of these mechanisms with the novel application of metformin as therapy of bladder cancer.
Collapse
Affiliation(s)
- Amr Ahmed EL-Arabey
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
- CAS-TWAS Fellowship at University of Science and Technology of China (USTC), Hefei, 23027 China
| |
Collapse
|
70
|
p27kip1 overexpression regulates IL-1β in the microenvironment of stem cells and eutopic endometriosis co-cultures. Cytokine 2017; 89:229-234. [DOI: 10.1016/j.cyto.2015.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 12/03/2015] [Accepted: 12/22/2015] [Indexed: 12/11/2022]
|
71
|
Polycystic ovary syndrome and risk of endometrial, ovarian, and breast cancer: a systematic review. FERTILITY RESEARCH AND PRACTICE 2016; 2:14. [PMID: 28620541 PMCID: PMC5424400 DOI: 10.1186/s40738-016-0029-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/25/2016] [Indexed: 02/07/2023]
Abstract
Background Polycystic ovary syndrome (PCOS) is a complex endocrine disorder with an estimated prevalence of 4–21% in reproductive aged women. The altered metabolic and hormonal environment among women with PCOS may increase their risk of some types of cancer. Methods We performed a comprehensive review of the literature using numerous search terms for all studies examining the associations between polycystic ovary syndrome and related characteristics and cancer published in English through October 2016. This review summarizes the epidemiological findings on the associations between PCOS and endometrial, ovarian, and breast cancers and discusses the methodological issues, complexities, and underlying mechanisms of these associations. Results We identified 11 individual studies and 3 meta-analyses on the associations between PCOS and endometrial cancer, 8 studies and 1 meta-analysis for ovarian cancer, and 10 studies and 1 meta-analysis for breast cancer. Multiple studies reported that women with PCOS were at a higher risk for endometrial cancer; however, many did not take into account body mass index (BMI), a strong and well-established risk factor for endometrial cancer. The association with ovarian cancer was less clear, but a potentially increased risk of the borderline serous subtype was reported by two studies. No consistent association between PCOS risk and breast cancer was observed. Conclusion The associations between PCOS and endometrial, ovarian, and breast cancer are complex, with the need to consider many methodological issues in future analyses. Larger well-designed studies, or pooled analyses, may help clarify these complex associations.
Collapse
|
72
|
PPAR Gamma in Neuroblastoma: The Translational Perspectives of Hypoglycemic Drugs. PPAR Res 2016; 2016:3038164. [PMID: 27799938 PMCID: PMC5069360 DOI: 10.1155/2016/3038164] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma (NB) is the most common and aggressive pediatric cancer, characterized by a remarkable phenotypic diversity and high malignancy. The heterogeneous clinical behavior, ranging from spontaneous remission to fatal metastatic disease, is attributable to NB biology and genetics. Despite major advances in therapies, NB is still associated with a high morbidity and mortality. Thus, novel diagnostic, prognostic, and therapeutic approaches are required, mainly to improve treatment outcomes of high-risk NB patients. Among neuroepithelial cancers, NB is the most studied tumor as far as PPAR ligands are concerned. PPAR ligands are endowed with antitumoral effects, mainly acting on cancer stem cells, and constitute a possible add-on therapy to antiblastic drugs, in particular for NB with unfavourable prognosis. While discussing clinical background, this review will provide a synopsis of the major studies about PPAR expression in NB, focusing on the potential beneficial effects of hypoglycemic drugs, thiazolidinediones and metformin, to reduce the occurrence of relapses as well as tumor regrowth in NB patients.
Collapse
|
73
|
Sun R, Ma X, Cai X, Pan X, Liu D. The effect and mechanism of action of metformin on in vitro FaDu cell proliferation. J Int Med Res 2016; 44:1049-1054. [PMID: 27688683 PMCID: PMC5536548 DOI: 10.1177/0300060516642645] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective To investigate the effect and mechanism of action of metformin on proliferation of a human hypopharyngeal carcinoma cell line (FaDu). Methods FaDu cells were treated with metformin (25–125 mmol/l). Cell proliferation was evaluated via CCK-8 assay. Real-time quantitative reverse transcription–polymerase chain reaction was used to evaluate microRNA (miR)-21-5p and PDCD4 (programmed cell death 4) expression. PDCD4 protein was quantified by Western blot. Results Metformin significantly inhibited FaDu cell proliferation in a dose- (25–100 mmol/l) and time-dependent manner (12 h–36 h), significantly downregulated miR-21-5p, and upregulated PDCD4 mRNA and protein expression. Conclusions Metformin significantly inhibited FaDu cell proliferation, possibly via downregulation of miR-21-5p and upregulation of PDCD4.
Collapse
Affiliation(s)
- Ruijie Sun
- 1 Department of Otorhinolaryngology, Qilu Hospital of Shandong University, China
| | - Xiaojie Ma
- 2 Department of Otorhinolaryngology, Qilu Hospital (Qingdao) of Shandong University, China
| | - Xiaolan Cai
- 2 Department of Otorhinolaryngology, Qilu Hospital (Qingdao) of Shandong University, China
| | - Xinliang Pan
- 1 Department of Otorhinolaryngology, Qilu Hospital of Shandong University, China.,2 Department of Otorhinolaryngology, Qilu Hospital (Qingdao) of Shandong University, China
| | - Dayu Liu
- 1 Department of Otorhinolaryngology, Qilu Hospital of Shandong University, China.,2 Department of Otorhinolaryngology, Qilu Hospital (Qingdao) of Shandong University, China
| |
Collapse
|
74
|
Patel S, Singh N, Kumar L. Evaluation of Effects of Metformin in Primary Ovarian Cancer Cells. Asian Pac J Cancer Prev 2016; 16:6973-9. [PMID: 26514477 DOI: 10.7314/apjcp.2015.16.16.6973] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is the third most common cause of cancer in Indian women. Despite an initial 70-80% response rate, most patients relapse within 1-2 years and develop chemoresistance. Hence, identification or repositioning of drugs to resensitise ovarian cancer cells to existing chemotherapy is needed. Traditionally immortalized cell lines have been used in research, but these may contain genetic aberrations and chromosomal abnormalities serving as poor indicators of normal cell phenotype and progression of early-stage disease. The use of primary cells, maintained for only short periods of time in vitro, may serve as the best representative for studying in vivo conditions of the tissues from which they are derived. In this study we have attempted to evaluate the effect of metformin (an antidiabetic drug) in primary ovarian cancer cells because of its promising effect in other solid tumours. MATERIALS AND METHODS Primary cultures of epithelial ovarian cancer cells established from ascitic fluid of untreated ovarian cancer patients were used. The cells were treated with metformin at doses standardized by MTT assay and its ability to induce apoptosis was studied. The cells were analysed for apoptosis and apoptosis related proteins by flow cytometry and western blotting respectively. RESULTS Metformin induced apoptosis in ovarian cancer cells, provoking cell cycle arrest in the G0/G1 and S phase. It induced apoptosis in ovarian cancer cells by, down-regulating Bcl-2 and up-regulating Bax expression. CONCLUSIONS Metformin was able to induce apoptosis in primary ovarian cancer cells by modulating the expression of Bcl-2 family proteins. These data are relevant to ongoing translational research efforts exploring the chemotherapeutic potential of metformin.
Collapse
Affiliation(s)
- Seema Patel
- Department of Biochemistry, AIIMS, New Delhi, India E-mail : ,
| | | | | |
Collapse
|
75
|
Litchfield LM, Mukherjee A, Eckert MA, Johnson A, Mills KA, Pan S, Shridhar V, Lengyel E, Romero IL. Hyperglycemia-induced metabolic compensation inhibits metformin sensitivity in ovarian cancer. Oncotarget 2016; 6:23548-60. [PMID: 26172303 PMCID: PMC4695136 DOI: 10.18632/oncotarget.4556] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022] Open
Abstract
Increasing interest in repurposing the diabetic medication metformin for cancer treatment has raised important questions about the translation of promising preclinical findings to therapeutic efficacy, especially in non-diabetic patients. A significant limitation of the findings to date is the use of supraphysiologic metformin doses and hyperglycemic conditions in vitro. Our goals were to determine the impact of hyperglycemia on metformin response and to address the applicability of metformin as a cancer therapeutic in non-diabetic patients. In normoglycemic conditions, lower concentrations of metformin were required to inhibit cell viability, while metformin treatment in hyperglycemic conditions resulted in increased glucose uptake and glycolytic flux, contributing to cell survival. Mechanistically, maintenance of c-Myc expression under conditions of hyperglycemia or via gene amplification facilitated metabolic escape from the effects of metformin. In vivo, treatment of an ovarian cancer mouse model with metformin resulted in greater tumor weight reduction in normoglycemic vs. hyperglycemic mice, with increased c-Myc expression observed in metformin-treated hyperglycemic mice. These findings indicate that hyperglycemia inhibits the anti-cancer effects of metformin in vitro and in vivo. Furthermore, our results suggest that metformin may elicit stronger responses in normoglycemic vs. hyperglycemic patients, highlighting the need for prospective clinical testing in patients without diabetes.
Collapse
Affiliation(s)
- Lacey M Litchfield
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Abir Mukherjee
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Mark A Eckert
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Alyssa Johnson
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Kathryn A Mills
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Shawn Pan
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Viji Shridhar
- Department of Laboratory Medicine and Experimental Pathology, Mayo Clinic Cancer Center, Rochester, Minnesota, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| | - Iris L Romero
- Department of Obstetrics and Gynecology, Gordon Center for Integrative Science, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
76
|
Patel S, Singh N, Kumar L. Anticancer role of antidiabetic drug Metformin in ovarian cancer cells. INTERNATIONAL JOURNAL OF CANCER THERAPY AND ONCOLOGY 2016. [DOI: 10.14319/ijcto.42.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
77
|
Soo JSS, Ng CH, Tan SH, Malik RA, Teh YC, Tan BS, Ho GF, See MH, Taib NAM, Yip CH, Chung FFL, Hii LW, Teo SH, Leong CO. Metformin synergizes 5-fluorouracil, epirubicin, and cyclophosphamide (FEC) combination therapy through impairing intracellular ATP production and DNA repair in breast cancer stem cells. Apoptosis 2016; 20:1373-87. [PMID: 26276035 DOI: 10.1007/s10495-015-1158-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metformin, an AMPK activator, has been reported to improve pathological response to chemotherapy in diabetic breast cancer patients. To date, its mechanism of action in cancer, especially in cancer stem cells (CSCs) have not been fully elucidated. In this study, we demonstrated that metformin, but not other AMPK activators (e.g. AICAR and A-769662), synergizes 5-fluouracil, epirubicin, and cyclophosphamide (FEC) combination chemotherapy in non-stem breast cancer cells and breast cancer stem cells. We show that this occurs through an AMPK-dependent mechanism in parental breast cancer cell lines. In contrast, the synergistic effects of metformin and FEC occurred in an AMPK-independent mechanism in breast CSCs. Further analyses revealed that metformin accelerated glucose consumption and lactate production more severely in the breast CSCs but the production of intracellular ATP was severely hampered, leading to a severe energy crisis and impairs the ability of CSCs to repair FEC-induced DNA damage. Indeed, addition of extracellular ATP completely abrogated the synergistic effects of metformin on FEC sensitivity in breast CSCs. In conclusion, our results suggest that metformin synergizes FEC sensitivity through distinct mechanism in parental breast cancer cell lines and CSCs, thus providing further evidence for the clinical relevance of metformin for the treatment of cancers.
Collapse
Affiliation(s)
- Jaslyn Sian-Siu Soo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, 1 Jalan SS12/1A, 47500, Subang Jaya, Selangor, Malaysia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Metformin use and gynecological cancers: A novel treatment option emerging from drug repositioning. Crit Rev Oncol Hematol 2016; 105:73-83. [PMID: 27378194 DOI: 10.1016/j.critrevonc.2016.06.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 04/19/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Metformin exerts antitumor effects mainly through AMP-activated protein kinase [AMPK] activation and phosphatidylinositol 3-kinase [PI3K]-Akt-mammalian target of rapamycin [mTOR] inhibition. This drug leads to activation of the cellular energy-sensing liver kinase B1 [LKB1]/AMPK pathway. LKB1 is implicated as a tumor suppressor gene in molecular pathogenesis of different malignancies. AMPK is a serine/threonine protein kinase that acts as an ultra-sensitive cellular energy sensor maintaining the energy balance within the cell. AMPK activation inhibits mRNA translation and proliferation in cancer cells via down-regulation of PI3K/Akt/mTOR pathway. Moreover, metformin decreases the production of insulin, insulin-like growth factor, inflammatory cytokines and vascular endothelial growth factor, and therefore it exerts anti-mitotic, anti-inflammatory and anti-angiogenetic effects. Recent in vitro and experimental data suggest that metformin electively targets cancer stem cells, and acts together with chemotherapy to block tumor growth in different cancers. Several epidemiological studies and meta-analysis have shown that metformin use is associated with decreased cancer risk and/or reduced cancer mortality for different malignancies. The present review analyzes the recent biological and clinical data suggesting a possible growth-static effect of metformin also in gynecological cancers. The large majority of available clinical data on the anti-cancer potential of metformin are based on observational studies. Therefore long-term phase II-III clinical trials are strongly warranted to further investigate metformin activity in gynecological cancers.
Collapse
|
79
|
Preclinical evaluation of olaparib and metformin combination in BRCA1 wildtype ovarian cancer. Gynecol Oncol 2016; 142:323-31. [PMID: 27282964 DOI: 10.1016/j.ygyno.2016.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/02/2016] [Accepted: 06/05/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVES BRCA mutated ovarian cancers show increased responsiveness to PARP inhibitors. PARP inhibitors target DNA repair and provide a second hit to BRCA mutated tumors, resulting in "synthetic lethality". We investigated a combination of metformin and olaparib to provide "synthetic lethality" in BRCA intact ovarian cancer cells. METHODS Ovarian cancer cell lines (UWB1.289, UWB1.289.BRCA, SKOV3, OVCAR5, A2780 and C200) were treated with a combination of metformin and olaparib. Cell viability was assessed by MTT and colony formation assays. Flow cytometry was used to detect cell cycle events. In vivo studies were performed in SKOV3 or A2780 xenografts in nude mice. Animals were treated with single agent, metformin or olaparib or combination. Molecular downstream effects were examined by immunohistochemistry. RESULTS Compared to single drug treatment, combination of olaparib and metformin resulted in significant reduction of cell proliferation and colony formation (p<0.001) in ovarian cancer cells. This treatment was associated with a significant S-phase cell cycle arrest (p<0.05). Combination of olaparib and metformin significantly inhibited SKOV3 and A2780 ovarian tumor xenografts which were accompanied with decreased Ki-index (p<0.001). Metformin did not affect DNA damage signaling, while olaparib induced adenosine monophosphate activated kinase activation; that was further potentiated with metformin combination in vivo. CONCLUSION Combining PARP inhibitors with metformin enhances its anti-proliferative activity in BRCA mutant ovarian cancer cells. Furthermore, the combination showed significant activity in BRCA intact cancer cells in vitro and in vivo. This is a promising treatment regimen for women with epithelial ovarian cancer irrespective of BRCA status.
Collapse
|
80
|
Ciccone MA, Maoz A, Casabar JK, Machida H, Mabuchi S, Matsuo K. Clinical outcome of treatment with serine-threonine kinase inhibitors in recurrent epithelial ovarian cancer: a systematic review of literature. Expert Opin Investig Drugs 2016; 25:781-96. [PMID: 27101098 DOI: 10.1080/13543784.2016.1181748] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION While serine-threonine kinases (STK) are attractive therapeutic targets in epithelial ovarian cancer, clinical outcomes of STK inhibitors in the management of recurrent disease have not been completely described. AREAS COVERED A systematic literature review of published clinical studies on STK inhibitors targeting mTOR, MAPK, and aurora kinase pathways in recurrent epithelial ovarian cancer was conducted, revealing 18 clinical trials (497 patients). Pooled analyses were performed to assess treatment response, survival time, and adverse events. Median progression-free survival was 3.4 months in STK inhibitor-based therapy, and the average response rate and clinical benefit rate were 13% and 67%, respectively. Among regimens comprised of only STK inhibitors (11 trials, 299 patients), median progression-free time was 2.7 months, response rate was 10%, and clinical benefit rate was 64%. Compared to single STK inhibitor monotherapy (52.5%), clinical benefit rates significantly improved when STK inhibitors were combined with a cytotoxic agent (71.4%), other class biological agent (74.2%), or an additional STK inhibitor (95.0%) (all, P ≤ 0.002). EXPERT OPINION STK inhibitor-based therapy showed modest activity for recurrent epithelial ovarian cancer with reasonable clinical benefit rates, suggesting its potential utility for maintaining disease stability if supported by future studies. Efficacy appears greatly improved in appropriately selected patient populations, especially those with low-grade serous ovarian carcinoma, platinum-sensitive disease, cancers with somatic RAS or BRAF mutations, and when used in a combination regimen with a cytotoxic or biological agent.
Collapse
Affiliation(s)
- Marcia A Ciccone
- a Division of Gynecologic Oncology, Department of Obstetrics and Gynecology , University of Southern California , Los Angeles , CA , USA
| | - Asaf Maoz
- b Norris Comprehensive Cancer Center , University of Southern California , Los Angeles , CA , USA
| | - Jennifer K Casabar
- a Division of Gynecologic Oncology, Department of Obstetrics and Gynecology , University of Southern California , Los Angeles , CA , USA
| | - Hiroko Machida
- a Division of Gynecologic Oncology, Department of Obstetrics and Gynecology , University of Southern California , Los Angeles , CA , USA
| | - Seiji Mabuchi
- c Department of Obstetrics and Gynecology , Osaka University Graduate School of Medicine , Osaka , Japan
| | - Koji Matsuo
- a Division of Gynecologic Oncology, Department of Obstetrics and Gynecology , University of Southern California , Los Angeles , CA , USA.,b Norris Comprehensive Cancer Center , University of Southern California , Los Angeles , CA , USA
| |
Collapse
|
81
|
Sośnicki S, Kapral M, Węglarz L. Molecular targets of metformin antitumor action. Pharmacol Rep 2016; 68:918-25. [PMID: 27362768 DOI: 10.1016/j.pharep.2016.04.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/29/2016] [Accepted: 04/29/2016] [Indexed: 12/28/2022]
Abstract
Epidemiological studies have shown that metformin, a first line therapeutic agent for diabetes mellitus, reduced the risk of developing various malignancies. Several preclinical studies established some possible mechanisms of its anticancer effects. The primary effect of metformin action is a decrease in cell energy status, which activates AMP-activated kinase (AMPK), a cellular metabolic sensor. This event is followed by a decrease in serum concentrations of insulin and insulin growth factor I (IGF-I), the potent mitogens for cancer cells. In addition to the indirect mode of action, metformin may exhibit direct inhibitory effect on cancer cells by targeting mammalian target of rapamycin (mTOR) signaling and anabolic processes. This review gathers information on mechanisms of metformin antitumor activity, with special attention given to the impact of this antidiabetic drug on insulin/PI3K/mTOR and AMPK signaling. Furthermore, the factors required for this novel activity of metformin are discussed.
Collapse
Affiliation(s)
- Stanisław Sośnicki
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland, Department of Biochemistry, Sosnowiec, Poland.
| | - Małgorzata Kapral
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland, Department of Biochemistry, Sosnowiec, Poland.
| | - Ludmiła Węglarz
- School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland, Department of Biochemistry, Sosnowiec, Poland.
| |
Collapse
|
82
|
Targeting of free fatty acid receptor 1 in EOC: A novel strategy to restrict the adipocyte-EOC dependence. Gynecol Oncol 2016; 141:72-9. [DOI: 10.1016/j.ygyno.2016.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/15/2016] [Accepted: 02/21/2016] [Indexed: 12/14/2022]
|
83
|
Sun XJ, Zhang P, Li HH, Jiang ZW, Jiang CC, Liu H. Cisplatin combined with metformin inhibits migration and invasion of human nasopharyngeal carcinoma cells by regulating E-cadherin and MMP-9. Asian Pac J Cancer Prev 2016; 15:4019-23. [PMID: 24935589 DOI: 10.7314/apjcp.2014.15.9.4019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Metformin has been shown to be useful in reducing insulin resistance by restoring sensitivity. Recent evidence suggests that metformin might also possess anti-tumour activity. This study aimed to investigate the effects of cisplatin combined with metformin on the proliferation, invasion and migration of HNE1/DDP human nasopharyngeal carcinoma (NPC) cells, and to provide a new target for treating metastasis. The MTT assay was used to assess viability of HNE1/DDP cells after exposure to different concentrations of 2, 5-diaminopyrimidine-4, 6-diol (DDP; 2, 4, 8, 16, and 32 μmol·L(-1)), metformin (5, 10, 15, 20, and 25 μmol·L(-1)), and 4 μmol·L(-1) of DDP combined with metformin. Wound healing and transwell migration assays were performed to assess cell migration and invasion, and expression of E-cadherin and MMP-9 was detected using Western blotting. MTT assay results showed that DDP could inhibit the proliferation of HNE1/DDP cells in a time- and concentration-dependent manner, with an IC50 of 32.0 μmol·L(-1) at 24 h (P < 0.05), whereas low concentrations of DDP had almost no inhibitory effects on cell invasion and migration. DDP combined with metformin significantly inhibited cell invasion and migration. In addition, genes related to migration and invasion, such as those of E-cadherin and MMP-9, showed differential expression in the NPC cell line HNE1/DDP. In the present study, with an increasing concentration of metformin, the expression of MMP-9 was downregulated whereas that of E-cadherin was significantly upregulated. Taken together, our results show that cisplatin combined with metformin has effects on proliferation, invasion, and migration of human NPC cells.
Collapse
Affiliation(s)
- Xiao-Jin Sun
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China E-mail :
| | | | | | | | | | | |
Collapse
|
84
|
Abdel Malek MAY, Jagannathan S, Malek E, Sayed DM, Elgammal SA, Abd El-Azeem HG, Thabet NM, Driscoll JJ. Molecular chaperone GRP78 enhances aggresome delivery to autophagosomes to promote drug resistance in multiple myeloma. Oncotarget 2016; 6:3098-110. [PMID: 25605012 PMCID: PMC4413640 DOI: 10.18632/oncotarget.3075] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/17/2014] [Indexed: 11/25/2022] Open
Abstract
Despite the clinical benefit of the proteasome inhibitor bortezomib, multiple myeloma (MM) patients invariably relapse through poorly defined mechanisms. Myeloma cells inevitably develop chemoresistance that leads to disease relapse and patient-related deaths. Studies in tumor cell lines and biopsies obtained from patients refractory to therapy have revealed that myeloma cells adapt to stress by inducing expression of glucose-regulated protein 78 (GRP78), an endoplasmic reticulum (ER) chaperone with anti-apoptotic properties. Treatment of myeloma cells with bortezomib increased GRP78 levels and activated GRP78-dependent autophagy. Expression profiling indicated that GRP78-encoding HSPA5 was significantly upregulated in bortezomib-resistant cells. Co-treatment with the anti-diabetic agent metformin suppressed GRP78 and enhanced the anti-proliferative effect of bortezomib. Bortezomib treatment led to GRP78 co-localization with proteotoxic protein aggregates, known as aggresomes. Pharmacologic suppression, genetic ablation or mutational inactivation of GRP78 followed by bortezomib treatment led to the accumulation of aggresomes but impaired autophagy and enhanced anti-myeloma effect of bortezomib. GRP78 was co-immunoprecipitated with the KDEL receptor, an ER quality control regulator that binds proteins bearing the KDEL motif to mediate their retrieval from the Golgi complex back to the ER. Taken together, we demonstrate that inhibition of GRP78 functional activity disrupts autophagy and enhances the anti-myeloma effect of bortezomib.
Collapse
Affiliation(s)
- Mohamed A Y Abdel Malek
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sajjeev Jagannathan
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ehsan Malek
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douaa M Sayed
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Sahar A Elgammal
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hanan G Abd El-Azeem
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nabila M Thabet
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - James J Driscoll
- The Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Hematology and Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,University of Cincinnati Cancer Institute, Cincinnati, OH, USA
| |
Collapse
|
85
|
Hijaz M, Das S, Mert I, Gupta A, Al-Wahab Z, Tebbe C, Dar S, Chhina J, Giri S, Munkarah A, Seal S, Rattan R. Folic acid tagged nanoceria as a novel therapeutic agent in ovarian cancer. BMC Cancer 2016; 16:220. [PMID: 26979107 PMCID: PMC4791781 DOI: 10.1186/s12885-016-2206-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 02/20/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nanomedicine is a very promising field and nanomedical drugs have recently been used as therapeutic agents against cancer. In a previous study, we showed that Nanoceria (NCe), nanoparticles of cerium oxide, significantly inhibited production of reactive oxygen species, cell migration and invasion of ovarian cancer cells in vitro, without affecting cell proliferation and significantly reduced tumor growth in an ovarian cancer xenograft nude model. Increased expression of folate receptor-α, an isoform of membrane-bound folate receptors, has been described in ovarian cancer. To enable NCe to specifically target ovarian cancer cells, we conjugated nanoceria to folic acid (NCe-FA). Our aim was to investigate the pre-clinical efficacy of NCe-FA alone and in combination with Cisplatin. METHODS Ovarian cancer cell lines were treated with NCe or NCe-FA. Cell viability was assessed by MTT and colony forming units. In vivo studies were carried in A2780 generated mouse xenografts treated with 0.1 mg/Kg NCe, 0.1 mg/Kg; NCe-FA and cisplatinum, 4 mg/Kg by intra-peritoneal injections. Tumor weights and burden scores were determined. Immunohistochemistry and toxicity assays were used to evaluate treatment effects. RESULTS We show that folic acid conjugation of NCe increased the cellular NCe internalization and inhibited cell proliferation. Mice treated with NCe-FA had a lower tumor burden compared to NCe, without any vital organ toxicity. Combination of NCe-FA with cisplatinum decreased the tumor burden more significantly. Moreover, NCe-FA was also effective in reducing proliferation and angiogenesis in the xenograft mouse model. CONCLUSION Thus, specific targeting of ovarian cancer cells by NCe-FA holds great potential as an effective therapeutic alone or in combination with standard chemotherapy.
Collapse
Affiliation(s)
- Miriana Hijaz
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Soumen Das
- Advanced Materials Processing and Analysis Center, Nanoscience and Technology Center, Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Ismail Mert
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA.,Wayne State University, Detroit, MI, USA
| | - Ankur Gupta
- Advanced Materials Processing and Analysis Center, Nanoscience and Technology Center, Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Zaid Al-Wahab
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Calvin Tebbe
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Sajad Dar
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Jasdeep Chhina
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Josephine Ford Cancer Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Adnan Munkarah
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA.,Josephine Ford Cancer Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Nanoscience and Technology Center, Materials Science and Engineering, University of Central Florida, Orlando, FL, USA.,College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA. .,Josephine Ford Cancer Institute, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
86
|
Novel approach to target cancer stem cells for therapy. Med Hypotheses 2016; 88:83-5. [DOI: 10.1016/j.mehy.2015.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 01/15/2023]
|
87
|
Scherbakov AM, Sorokin DV, Tatarskiy VV, Prokhorov NS, Semina SE, Berstein LM, Krasil'nikov MA. The phenomenon of acquired resistance to metformin in breast cancer cells: The interaction of growth pathways and estrogen receptor signaling. IUBMB Life 2016; 68:281-92. [PMID: 26892736 DOI: 10.1002/iub.1481] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 01/10/2016] [Indexed: 01/02/2023]
Abstract
Metformin, a biguanide antidiabetic drug, is used to decrease hyperglycemia in patients with type 2 diabetes. Recently, the epidemiological studies revealed the potential of metformin as an anti-tumor drug for several types of cancer, including breast cancer. Anti-tumor metformin action was found to be mediated, at least in part, via activation of adenosine monophosphate-activated protein kinase (AMPK)-intracellular energy sensor, which inhibits the mammalian target of rapamycin (mTOR) and some other signaling pathways. Nevertheless, some patients can be non-sensitive or resistant to metformin action. Here we analyzed the mechanism of the formation of metformin-resistant phenotype in breast cancer cells and its role in estrogen receptor (ER) regulation. The experiments were performed on the ER-positive MCF-7 breast cancer cells and metformin-resistant MCF-7 subline (MCF-7/M) developed due to long-term metformin treatment. The transcriptional activity of NF-κB and ER was measured by the luciferase reporter gene analysis. The protein expression was determined by immunoblotting (Snail1, (phospho)AMPK, (phospho)IκBα, (phospho)mTOR, cyclin D1, (phospho)Akt and ERα) and immunohistochemical analysis (E-cadherin). We have found that: 1) metformin treatment of MCF-7 cells is accompanied with the stimulation of AMPK and inhibition of growth-related proteins including IκBα, NF-κB, cyclin D1 and ERα; 2) long-term metformin treatment lead to the appearance and progression of cross-resistance to metformin and tamoxifen; the resistant cells are characterized with the unaffected AMPK activity, but the irreversible ER suppression and constitutive activation of Akt/Snail1 signaling; 3) Akt/Snail1 signaling is involved into progression of metformin resistance. The results presented may be considered as the first evidence of the progression of cross-resistance to metformin and tamoxifen in breast cancer cells. Importantly, the acquired resistance to both drugs is based on the constitutive activation of Akt/Snail1/E-cadherin signaling that opens new perspectives to overcome the metformin/tamoxifen resistance of breast cancer.
Collapse
Affiliation(s)
- Alexander M Scherbakov
- Laboratory of Clinical Biochemistry, Institute of Clinical Oncology, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Danila V Sorokin
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Victor V Tatarskiy
- Laboratory of Cell Death Mechanisms, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Nikolay S Prokhorov
- Laboratory of Microbial Viruses, S.N. Winogradsky Institute of Microbiology, Russian Academy of Sciences, Moscow, Russia
| | - Svetlana E Semina
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| | - Lev M Berstein
- Laboratory of Oncoendocrinology, N.N. Petrov Research Institute of Oncology, St, Petersburg, Russia
| | - Mikhail A Krasil'nikov
- Laboratory of Molecular Endocrinology, Institute of Carcinogenesis, N.N. Blokhin Cancer Research Centre, Moscow, Russia
| |
Collapse
|
88
|
Guo Q, Liu Z, Jiang L, Liu M, Ma J, Yang C, Han L, Nan K, Liang X. Metformin inhibits growth of human non-small cell lung cancer cells via liver kinase B-1-independent activation of adenosine monophosphate-activated protein kinase. Mol Med Rep 2016; 13:2590-6. [PMID: 26847819 PMCID: PMC4768996 DOI: 10.3892/mmr.2016.4830] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 01/11/2016] [Indexed: 11/16/2022] Open
Abstract
Metformin, the most widely administered oral anti-diabetic therapeutic agent, exerts its glucose-lowering effect predominantly via liver kinase B1 (LKB1)-dependent activation of adenosine monophosphate-activated protein kinase (AMPK). Accumulating evidence has demonstrated that metformin possesses potential antitumor effects. However, whether the antitumor effect of metformin is via the LKB1/AMPK signaling pathway remains to be determined. In the current study, the effects of metformin on proliferation, cell cycle progression, and apoptosis of human non-small cell lung cancer (NSCLC) H460 (LKB1-null) and H1299 (LKB1-positive) cells were assessed, and the role of LKB1/AMPK signaling in the anti-growth effects of metformin were investigated. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, cell cycle distribution and apoptosis were assessed by flow cytometry, and protein expression levels were measured by western blotting. Metformin inhibited proliferation, induced significant cell cycle arrest at the G0–G1 phase and increased apoptosis in NSCLC cells in a time- and concentration-dependent manner, regardless of the level of LKB1 protein expression. Furthermore, knockdown of LKB1 with short hairpin RNA (shRNA) did not affect the antiproliferative effect of metformin in the H1299 cells. Metformin stimulated AMPK phosphorylation and subsequently suppressed the phosphorylation of mammalian target of rapamycin and its downstream effector, 70-kDa ribosomal protein S6 kinase in the two cell lines. These effects were abrogated by silencing AMPK with small interfering RNA (siRNA). In addition, knockdown of AMPK with siRNA inhibited the effect of metformin on cell proliferation in the two cell lines. These results provide evidence that the growth inhibition of metformin in NSCLC cells is mediated by LKB1-independent activation of AMPK, indicating that metformin may be a potential therapeutic agent for the treatment of human NSCLC.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Zhiyan Liu
- Department of Respiration, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Lili Jiang
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Mengjie Liu
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Jiequn Ma
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Chengcheng Yang
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Lili Han
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Kejun Nan
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| | - Xuan Liang
- Department of Oncology, First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
89
|
Metformin Treatment Does Not Inhibit Growth of Pancreatic Cancer Patient-Derived Xenografts. PLoS One 2016; 11:e0147113. [PMID: 26760500 PMCID: PMC4711922 DOI: 10.1371/journal.pone.0147113] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/29/2015] [Indexed: 01/27/2023] Open
Abstract
There is currently tremendous interest in developing anti-cancer therapeutics targeting cell signaling pathways important for both cancer cell metabolism and growth. Several epidemiological studies have shown that diabetic patients taking metformin have a decreased incidence of pancreatic cancer. This has prompted efforts to evaluate metformin, a drug with negligible toxicity, as a therapeutic modality in pancreatic cancer. Preclinical studies in cell line xenografts and one study in patient-derived xenograft (PDX) models were promising, while recently published clinical trials showed no benefit to adding metformin to combination therapy regimens for locally advanced and metastatic pancreatic cancer. PDX models in which patient tumors are directly engrafted into immunocompromised mice have been shown to be excellent preclinical models for biomarker discovery and therapeutic development. We evaluated the response of four PDX tumor lines to metformin treatment and found that all four of our PDX lines were resistant to metformin. We found that the mechanisms of resistance may occur through lack of sustained activation of adenosine monophosphate-activated protein kinase (AMPK) or downstream reactivation of the mammalian target of rapamycin (mTOR). Moreover, combined treatment with metformin and mTOR inhibitors failed to improve responses in cell lines, which further indicates that metformin alone or in combination with mTOR inhibitors will be ineffective in patients, and that resistance to metformin may occur through multiple pathways. Further studies are required to better understand these mechanisms of resistance and inform potential combination therapies with metformin and existing or novel therapeutics.
Collapse
|
90
|
He J, Wang K, Zheng N, Qiu Y, Xie G, Su M, Jia W, Li H. Metformin suppressed the proliferation of LoVo cells and induced a time-dependent metabolic and transcriptional alteration. Sci Rep 2015; 5:17423. [PMID: 26616174 PMCID: PMC4663508 DOI: 10.1038/srep17423] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/29/2015] [Indexed: 12/24/2022] Open
Abstract
Metformin is a widely used anti-diabetic drug with potential anti-tumor activity. However, little is known about its global metabolic and transcriptional impacts on tumor cells. In current study, we performed a metabolic profiling on human-derived colon cancer LoVo cells treated by 10 mM metformin for 8, 24 and 48 h. An obvious time-dependent metabolic alteration was observed from 8 to 48 h, prior to the reduction of cell viability. A total of 47, 45 and 66 differential metabolites were identified between control and metformin-treated cells at three time points. Most of the metabolites were up-regulated at 8 h, but down-regulated at 24 and 48 h by metformin. These metabolites were mainly involved in carbohydrates, lipids, amino acids, vitamins and nucleotides metabolism pathways. Meanwhile, the transcirptomic profile revealed 134 and 3061 differentially expressed genes at 8 and 24 h by metformin. In addition to the cancer signaling pathways, expression of genes involved in cell energy metabolism pathways was significantly altered, which were further validated with genes in glucose metabolism pathway. Altogether, our current data indicate that metformin suppressed the proliferation of LoVo cells, which may be due to the modulation on cell energy metabolism at both metabolic and transcriptional levels in a time-dependent way.
Collapse
Affiliation(s)
- Jiaojiao He
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ke Wang
- Laboratory of Integrative Medicine Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Zheng
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunping Qiu
- Stable Isotope and Metabolomics Core Facility, Diabetes Center Albert Einstein College of Medicine, 1300 Morris Part Ave, Bronx, New York, 10461, USA
| | - Guoxiang Xie
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA
| | - Mingming Su
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA
| | - Wei Jia
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, 96813, USA
| | - Houkai Li
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
91
|
Metformin Induces Cell Cycle Arrest and Apoptosis in Drug-Resistant Leukemia Cells. LEUKEMIA RESEARCH AND TREATMENT 2015; 2015:516460. [PMID: 26688757 PMCID: PMC4673355 DOI: 10.1155/2015/516460] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/18/2015] [Accepted: 09/20/2015] [Indexed: 12/25/2022]
Abstract
Recent epidemiological studies indicate that the antidiabetic drug metformin has chemosensitizing and chemopreventive effects against carcinogenesis. Here, we demonstrate that metformin exerts varying degrees of antitumor activity against human leukemia cells, as reflected by differences in growth inhibition, apoptosis, and alterations to metabolic enzymes. In metformin-sensitive cells, autophagy was not induced but rather it blocked proliferation by means of arresting cells in the S and G2/M phases which was associated with the downregulation of cyclin A, cyclin B1, and cdc2, but not that of cyclin E. In 10E1-CEM cells that overexpress Bcl-2 and are drug-resistant, the effect of metformin on proliferation was more pronounced, also inducing the activation of the caspases 3/7 and hence apoptosis. In all sensitive cells, metformin decreased the Δψm and it modified the expression of enzymes involved in energy metabolism: PKCε (PKCepsilon) and PKCδ (PKCdelta). In sensitive cells, metformin altered PKCε and PKCδ expression leading to a predominance of PKCε over PKCδ which implies a more glycolytic state. The opposite occurs in the nonresponsive cells. In conclusion, we provide new insights into the activity of metformin as an antitumoral agent in leukemia cells that could be related to its capability to modulate energy metabolism.
Collapse
|
92
|
Al-Wahab Z, Tebbe C, Chhina J, Dar SA, Morris RT, Ali-Fehmi R, Giri S, Munkarah AR, Rattan R. Dietary energy balance modulates ovarian cancer progression and metastasis. Oncotarget 2015; 5:6063-75. [PMID: 25026276 PMCID: PMC4171613 DOI: 10.18632/oncotarget.2168] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A high energy balance, or caloric excess, accounts as a tumor promoting factor, while a negative energy balance via caloric restriction, has been shown to delay cancer progression. The effect of energy balance on ovarian cancer progression was investigated in an isogeneic immunocompetent mouse model of epithelial ovarian cancer kept on a regimen of regular diet, high energy diet (HED) and calorie restricted diet (CRD), prior to inoculating the animals intraperitoneally with the mouse ovarian surface epithelial ID8 cancer cells. Tumor evaluation revealed that mice group on HED displayed the most extensive tumor formation with the highest tumor score at all organ sites (diaphragm, peritoneum, bowel, liver, kidney, spleen), accompanied with increased levels of insulin, leptin, insulin growth factor-1 (IGF-1), monocyte chemoattractant protein-1 (MCP-1), VEGF and interleukin 6 (IL-6). On the other hand, the mice group on CRD exhibited the least tumor burden associated with a significant reduction in levels of insulin, IGF-1, leptin, MCP-1, VEGF and IL-6. Immunohistochemistry analysis of tumors from HED mice showed higher activation of Akt and mTOR with decreased adenosine monophosphate activated kinase (AMPK) and SIRT1 activation, while tumors from the CRD group exhibited the reverse profile. In conclusion, ovarian cancer growth and metastasis occurred more aggressively under HED conditions and was significantly curtailed under CRD. The suggested mechanism involves modulated secretion of growth factors, cytokines and altered regulation of AMPK and SIRT1 that converges on mTOR inhibition. While the role of a high energy state in ovarian cancer has not been confirnmed in the literature, the current findings support investigating the potential impact of diet modulation as adjunct to other anticancer therapies and as possible individualized treatment strategy of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Zaid Al-Wahab
- Division of Gynecology Oncology, Wayne State University, Detroit, MI
| | - Calvin Tebbe
- Department of Women's Health, Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Jasdeep Chhina
- Department of Women's Health, Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Sajad A Dar
- Department of Women's Health, Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Robert T Morris
- Division of Gynecology Oncology, Wayne State University, Detroit, MI
| | - Rouba Ali-Fehmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State Univeristy, Detroit, MI
| | | | - Adnan R Munkarah
- Department of Women's Health, Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Ramandeep Rattan
- Department of Women's Health, Obstetrics and Gynecology, Wayne State University, Detroit, MI
| |
Collapse
|
93
|
Purow B. Repurposing existing agents as adjunct therapies for glioblastoma. Neurooncol Pract 2015; 3:154-163. [PMID: 31386097 DOI: 10.1093/nop/npv041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Indexed: 12/16/2022] Open
Abstract
Numerous non-oncologic medications have been found in the last decade to have anti-cancer properties. While the focus in oncology research should clearly remain on deriving new therapeutic strategies, repurposing these existing medications may offer the potential to rapidly enhance the effectiveness of treatment for resistant cancers. Glioblastoma, the most common and lethal brain cancer, is highly resistant to standard therapies and would benefit from even minor improvements in treatment. Numerous agents already in the clinic for non-cancer applications have been found to also possess potential against cancer or specifically against glioblastoma. These include agents with activities affecting oxidative stress, the immune reponse, epigenetic modifiers, cancer cell metabolism, and angiogenesis and invasiveness. This review serves as a guide for potential ways to repurpose individual drugs alongside standard glioblastoma therapies.
Collapse
Affiliation(s)
- Benjamin Purow
- Neurology Department, University of Virginia Neuro-Oncology Division, Old Medical School Room 4881, 21 Hospital Drive, Charlottesville, VA 22908, USA (B.P.)
| |
Collapse
|
94
|
Diabetes mellitus and gynecologic cancer: molecular mechanisms, epidemiological, clinical and prognostic perspectives. Arch Gynecol Obstet 2015; 293:239-46. [PMID: 26338721 DOI: 10.1007/s00404-015-3858-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 08/18/2015] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Diabetes mellitus, the prevalence of which has increased dramatically worldwide, may put patients at a higher risk of cancer. The aim of our study is the clarification of the possible mechanisms linking diabetes mellitus and gynecological cancer and their epidemiological relationship. MATERIALS AND METHODS This is a narrative review of the current literature, following a search on MEDLINE and the Cochrane Library, from their inception until January 2012. Articles investigating gynecologic cancer (endometrial, ovarian, and breast) incidence in diabetic patients were extracted. RESULTS The strong evidence for a positive association between diabetes mellitus and the risk for cancer indicates that energy intake in excess to energy expenditure, or the sequelae thereof, is involved in gynecological carcinogenesis. This risk may be further heightened by glucose which can directly promote the production of tumor cells by functioning as a source of energy. Insulin resistance accompanied by secondary hyperinsulinemia is hypothezised to have a mitogenic effect. Steroid hormones are in addition potent regulators of the balance between cellular differentiation, proliferation, and apoptosis. Inflammatory pathways may also be implicated, as a correlation seems to exist between diabetes mellitus and breast or endometrial carcinoma pathogenesis, although an analogous correlation with ovarian carcinoma is still under investigation. Antidiabetic agents have been correlated with elevated cancer risk, while metformin seems to lower the risk. CONCLUSION Diabetes mellitus is associated with an elevation in gynecologic cancer risk. Moreover, there are many studies exploring the prognosis of patients with diabetes and gynecological cancer, the outcome and the overall survival in well-regulated patients.
Collapse
|
95
|
Tseng CH. Metformin reduces ovarian cancer risk in Taiwanese women with type 2 diabetes mellitus. Diabetes Metab Res Rev 2015; 31:619-626. [PMID: 25820555 DOI: 10.1002/dmrr.2649] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/28/2015] [Accepted: 03/11/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND Whether metformin therapy affects ovarian cancer risk in Asian patients with type 2 diabetes mellitus has not been investigated. METHODS Data analysis was performed in 2014. The reimbursement databases of Taiwanese female patients with a new diagnosis of type 2 diabetes mellitus between 1998 and 2002 (n = 479,475) were retrieved from the National Health Insurance for follow-up of ovarian cancer until the end of 2009. Metformin was treated as a time-dependent variable; and of these patients, 286,106 were never-users, and 193,369 were ever-users. A time-dependent approach was used to calculate ovarian cancer incidence and estimate hazard ratios by Cox regression for never-users (as referent group), ever-users and subgroups of metformin exposure (tertiles of cumulative duration and cumulative dose). RESULTS During follow-up, 601 metformin ever-users and 2600 never-users developed ovarian cancer, representing an incidence of 49.4 and 146.4 per 100,000 person-years, respectively. The overall fully adjusted hazard ratio (95% confidence intervals) for ever-users versus never-users was 0.658 (0.593-0.730). The fully adjusted hazard ratios for the first, second and third tertiles of cumulative duration of metformin therapy were 1.169 (1.019-1.341), 0.761 (0.644-0.898) and 0.276 (0.225-0.340), respectively (p trend < 0.01) and 1.220 (1.067-1.395), 0.610 (0.513-0.725) and 0.305 (0.248-0.374), respectively (p trend < 0.01), for a cumulative dose of metformin. In additional analyses, sulfonylureas but not the other antidiabetic drugs were associated with a reduced risk of ovarian cancer. CONCLUSIONS Metformin use is associated with a decreased risk of ovarian cancer.
Collapse
Affiliation(s)
- Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
96
|
Hu X, Jiang F, Bao Q, Qian H, Fang Q, Shao Z. Compound 13, an α1-selective small molecule activator of AMPK, potently inhibits melanoma cell proliferation. Tumour Biol 2015; 37:1071-8. [DOI: 10.1007/s13277-015-3854-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/28/2015] [Indexed: 12/25/2022] Open
|
97
|
Patel S, Kumar L, Singh N. Metformin and epithelial ovarian cancer therapeutics. Cell Oncol (Dordr) 2015; 38:365-75. [DOI: 10.1007/s13402-015-0235-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2015] [Indexed: 12/13/2022] Open
|
98
|
Mayer MJ, Klotz LH, Venkateswaran V. Metformin and prostate cancer stem cells: a novel therapeutic target. Prostate Cancer Prostatic Dis 2015. [PMID: 26215782 DOI: 10.1038/pcan.2015.35] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prostate cancer is the second most frequently diagnosed cancer in the world. Localized disease can be effectively treated with radiation therapy or radical prostatectomy. However, advanced prostate cancer is more difficult to treat and if metastatic, is incurable. There is a need for more effective therapy for advanced prostate cancer. One potential target is the cancer stem cell (CSC). CSCs have been described in several solid tumors, including prostate cancer, and contribute to therapeutic resistance and tumor recurrence. Metformin, a common oral biguanide used to treat type 2 diabetes, has been demonstrated to have anti-neoplastic effects. Specifically, metformin targets CSCs in breast cancer, pancreatic cancer, glioblastoma and colon cancer. Metformin acts directly on the mitochondria to inhibit oxidative phosphorylation and reduce mitochondrial ATP production. This forces tumor cells to compensate by increasing the rate of glycolysis. CSCs rely heavily on mitochondrial oxidative phosphorylation for energy production. The glycolytic switch results in an energy crisis in these cells. Metformin could be used to exploit this metabolic weakness in CSCs. This would increase CSC sensitivity to conventional cancer therapies, circumventing treatment resistance and enhancing treatment efficacy. This review will explore the characteristics of prostate CSCs, their role in tumor propagation and therapeutic resistance and the role of metformin as a potential prostate CSC sensitizer to current anticancer therapies.
Collapse
Affiliation(s)
- M J Mayer
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - L H Klotz
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - V Venkateswaran
- Division of Urology, Department of Surgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
99
|
FAN CONG, WANG YUNSHAN, LIU ZIMING, SUN YING, WANG XIUWEN, WEI GUANGWEI, WEI JUNMIN. Metformin exerts anticancer effects through the inhibition of the Sonic hedgehog signaling pathway in breast cancer. Int J Mol Med 2015; 36:204-14. [PMID: 25999130 PMCID: PMC4494591 DOI: 10.3892/ijmm.2015.2217] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/13/2015] [Indexed: 02/04/2023] Open
Abstract
Metformin, a widely prescribed antidiabetic drug, has previously been shown to lower the risk of certain types of cancer, including that of breast cancer, and to improve prognosis. Its anticancer effects, which are mediated by the activation of AMP-activated protein kinase (AMPK), have become notable. The Sonic hedgehog (Shh) signaling pathway is involved in changes in mammary ducts and malignant transformation. The aim of the present study was to elucidate the role of the Shh pathway in mediating the anticancer effects of metformin and the correlation between AMPK and the Shh pathway. We investigated the effectiveness of metformin in inhibiting the proliferation, migration, invasion and stemness of breast cancer cells in vitro using RNA extraction and reverse transcription‑polymerase chain reaction (RT-PCR), western blot analysis, cell proliferation assay, scratch-wound assay (cell migration assay), cell invasion assay, mammosphere culture and flow cytometry. In in vivo experiments, a tumor xenograft model was used to detect the effects of metformin on cancer cell proliferation. The results revealed that the treatment of breast cancer cells with metformin led to the inhibition of the Shh signaling pathway. Importantly, metformin inhibited recombinant human Shh (rhShh)‑induced cell migration, invasion, and stemness, and impaired cell proliferation both in vitro and in vivo. Furthermore, the small interfering RNA (siRNA)‑mediated downregulation of AMPK reversed the inhibitory effects of metformin on rhShh‑induced Gli-1 expression and stemness. Our findings identified a role of the Shh signaling pathway in the anticancer effects of metformin in breast cancer. Furthermore, we revealed that the metformin-mediated inhibition of the Shh signaling pathway may be dependent on AMPK.
Collapse
Affiliation(s)
- CONG FAN
- Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - YUNSHAN WANG
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China,International Biotechnology R&D Center, Shandong University School of Ocean, Weihai, Shandong 264209, P.R. China
| | - ZIMING LIU
- Department of Emergency Medicine, The Fifth People’s Hospital, Jinan, Shandong 250022, P.R. China
| | - YING SUN
- Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - XIUWEN WANG
- Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - GUANGWEI WEI
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - JUNMIN WEI
- Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China,Correspondence to: Professor Junmin Wei, Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong 250012, P.R. China, E-mail:
| |
Collapse
|
100
|
Piguet AC, Saran U, Simillion C, Keller I, Terracciano L, Reeves HL, Dufour JF. Regular exercise decreases liver tumors development in hepatocyte-specific PTEN-deficient mice independently of steatosis. J Hepatol 2015; 62:1296-303. [PMID: 25623824 DOI: 10.1016/j.jhep.2015.01.017] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Unhealthy lifestyles predispose people to non-alcoholic steatohepatitis (NASH), which may further result in the development of hepatocellular carcinoma (HCC). Although NASH patients benefit from physical activity, it is unknown whether regular exercise reduces the risk of developing HCC. Therefore, we studied the effect of regular exercise on the development of HCC in male hepatocyte-specific PTEN-deficient mice (AlbCrePten(flox/flox)), which develop steatohepatitis and HCC spontaneously. METHODS Mice were fed a standardized 10% fat diet and were randomly divided into exercise or sedentary groups. The exercise group ran on a motorized treadmill for 60 min/day, 5 days/week during 32 weeks. RESULTS After 32 weeks of regular exercise, 71% of exercised mice developed nodules larger than 15 mm(3)vs. 100% of mice in the sedentary group. The mean number of tumors per liver was reduced by exercise, as well as the total tumoral volume per liver. Exercise did not affect steatosis and had no effect on the non-alcoholic fatty liver disease (NAFLD) Activity Score (NAS). Exercise decreased tumor cell proliferation. Mechanistically, exercise stimulated the phosphorylation of AMPK and its substrate raptor, which decreased the kinase activity of mTOR. CONCLUSIONS These data show a beneficial effect of regular exercise on the development of HCC in an experimental model of NASH and offer a rationale for encouraging predisposed patients to increase their physical activity for the prevention of HCC.
Collapse
Affiliation(s)
- Anne-Christine Piguet
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Uttara Saran
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Cedric Simillion
- Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Irene Keller
- Department of Clinical Research, University of Berne, Berne, Switzerland
| | - Luigi Terracciano
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Helen L Reeves
- Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, UK; The Liver Group, Newcastle University, Newcastle-upon-Tyne, UK
| | - Jean-François Dufour
- Hepatology, Department of Clinical Research, University of Berne, Berne, Switzerland; University Clinic of Visceral Surgery and Medicine, Inselspital Berne, Berne, Switzerland.
| |
Collapse
|