351
|
Phase I dose escalation study of temsirolimus in combination with metformin in patients with advanced/refractory cancers. Cancer Chemother Pharmacol 2016; 77:973-7. [PMID: 27014780 DOI: 10.1007/s00280-016-3009-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/10/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE Mammalian target of rapamycin (mTOR) inhibitors like temsirolimus may result in undesirable AKT upregulation. Metformin inhibits mTOR through different mechanisms and may enhance temsirolimus's antitumor activity. We conducted an open-label phase I dose escalation trial of this drug combination in patients with advanced/refractory cancers. METHODS Temsirolimus, 25 mg weekly, was combined with an escalating daily dose of metformin (level 1: 500; level 2: 1000; level 3: 1500; level 4: 2000 mg) by utilizing a standard 3 + 3 trial design. Treatment was administered in 28-day cycles following initial 2-week metformin titration during the first cycle. RESULTS Twenty-one patients (median age, 56 years) with sarcoma (n = 8), colorectal (n = 3), endometrial (n = 4), uterine carcinosarcoma (n = 2), ovarian (n = 2), and other (n = 2) cancers were enrolled. Patients had received median of four prior systemic treatments. Two dose-limiting toxicities were observed (grade 3 mucositis, grade 3 renal failure); both patients continued treatment after dose modification. Fifty-six percent patients had stable disease as best response; clinical benefit rate was 22 %. Patients continued treatment for median of 11 weeks. CONCLUSIONS Combination temsirolimus/metformin was well tolerated with modestly promising effectiveness among this heavily pretreated patient cohort. We recommend a dose of temsirolimus 25 mg weekly and metformin 2000 mg daily for phase II study.
Collapse
|
352
|
Heidari F, Abbas Zade S, Mir Hosseini SH, Ghadian A. Metformin for the Prevention of Bladder Cancer Recurrence: Is it Effective? Nephrourol Mon 2016; 8:e30261. [PMID: 27570750 PMCID: PMC4983155 DOI: 10.5812/numonthly.30261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 12/27/2015] [Accepted: 02/24/2016] [Indexed: 12/30/2022] Open
Abstract
Background Many methods have been used for preventing and reducing recurrences of bladder cancers. In recent years, some investigators have examined the use of metformin for this purpose. First lines of evidence have shown that metformin inhibits cancer cell growth and prevents cancer occurrence in patients with type 2 diabetes. Objectives This study is designed to assess metformin usage in the prevention of bladder cancer recurrence after the trans-urethral resection of a bladder tumor (TUR-T). Patients and Methods In the present study, metformin was administered in the treatment of 32 patients with a history of bladder cancer, and their results were compared with those of 33 patients with bladder cancer recurrence (placebo group). Patients in the metformin group received 1000 mg metformin (2 tablets 500 mg) for 1 year. Frequency of tumor recurrence was calculated and compared with the placebo group. Results There was no statistical difference between the 2 groups with respect to the recurrence rate (P > 0.05). Although the recurrence interval was longer for the metformin group, this increase was not statistical significant (P > 0.05). Furthermore, tumor recurrence had no correlation with sex or the grade of the tumors. Conclusions According to our findings, it seems that metformin has no considerable inhibitory effect on the recurrence rate of bladder cancer, but that it can delay tumor recurrence.
Collapse
Affiliation(s)
- Fatemeh Heidari
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Shahin Abbas Zade
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Seyed Hassan Mir Hosseini
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Alireza Ghadian
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Alireza Ghadian, Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel: +98-2181262073, E-mail:
| |
Collapse
|
353
|
Song Y, Wu Y, Su X, Zhu Y, Liu L, Pan Y, Zhu B, Yang L, Gao L, Li M. Activation of AMPK inhibits PDGF-induced pulmonary arterial smooth muscle cells proliferation and its potential mechanisms. Pharmacol Res 2016; 107:117-124. [PMID: 26993101 DOI: 10.1016/j.phrs.2016.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 02/19/2016] [Accepted: 03/11/2016] [Indexed: 12/30/2022]
Abstract
The aims of the present study were to examine signaling mechanisms for PDGF-induced pulmonary arterial smooth muscle cells (PASMC) proliferation and to determine the effect of AMPK activation on PDGF-induced PASMC proliferation and its underlying mechanisms. PDGF activated PI3K/Akt/mTOR signaling pathway, and this in turn up-regulated Skp2 and consequently reduced p27 leading to PASMC proliferation. Prior incubation of PASMC with metformin induced a dramatic AMPK activation and significantly blocked PDGF-induced cell proliferation. PASMC lacking AMPKα2 were resistant to the inhibitory effect of metformin on PDGF-induced cell proliferation. Metformin did not affect Akt activation but blocked mTOR phosphorylation in response to PDGF; these were accompanied by the reversion of Skp2 up-regulation and p27 reduction. Our study suggests that the activation of AMPK negatively regulates mTOR activity to suppress PASMC proliferation and therefore has a potential value in the prevention and treatment of pulmonary hypertension by negatively modulating pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Yang Song
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Yuanyuan Wu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Xiaofan Su
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Bo Zhu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Medical College, Xian Jiaotong University, Xian, Shaanxi 710061, PR China.
| |
Collapse
|
354
|
Kim J, Lee J, Jang SY, Kim C, Choi Y, Kim A. Anticancer effect of metformin on estrogen receptor-positive and tamoxifen-resistant breast cancer cell lines. Oncol Rep 2016; 35:2553-60. [PMID: 26986571 PMCID: PMC4811404 DOI: 10.3892/or.2016.4675] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/26/2016] [Indexed: 12/21/2022] Open
Abstract
Acquisition of tamoxifen resistance (TR) during anti-estrogenic therapy using tamoxifen is a major obstacle in the treatment of estrogen receptor (ER)-positive breast cancer. As a biguanide derivative, metformin is commonly used to treat type II diabetes. It has recently emerged as a potential anticancer agent. The objective of the present study was to investigate the anticancer activity of metformin in relation to ERα expression and its signaling pathway in ERα-positive MCF-7 and MDA-MB-361 breast cancer cells as well as TR MCF-7 breast cancer cells. Metformin inhibited both protein and mRNA levels of ERα in the presence or absence of estrogen (E2) in the MCF-7, TR MCF-7 and MDA-MB-361 cells. Metformin repressed E2-inducible estrogen response element (ERE) luciferase activity, protein levels and mRNA levels of E2/ERα-regulated genes [including c-Myc, cyclin D1, progesterone receptor (PR) and pS2] to a greater degree than tamoxifen, resulting in inhibition of cell proliferation of MCF-7, TR MCF-7 and MDA-MB-361 cells. Collectively, our results suggest that one of the anticancer mechanisms of metformin could be attributable to the repression of expression and transcriptional activity of ERα. Metformin may be a good therapeutic agent for treating ERα-positive breast cancer by inhibiting the expression and function of ERα. In addition, metformin may be useful to treat tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- Jinkyoung Kim
- Department of Pathology, Korea University Guro Hospital, Guro-gu, Seoul 08308, Republic of Korea
| | - Jiyun Lee
- Department of Pathology, College of Medicine, Korea University, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Soon Young Jang
- Department of Pathology, College of Medicine, Korea University, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Chungyeul Kim
- Department of Pathology, Korea University Guro Hospital, Guro-gu, Seoul 08308, Republic of Korea
| | - Yoojin Choi
- Department of Pathology, Korea University Guro Hospital, Guro-gu, Seoul 08308, Republic of Korea
| | - Aeree Kim
- Department of Pathology, Korea University Guro Hospital, Guro-gu, Seoul 08308, Republic of Korea
| |
Collapse
|
355
|
Sacco F, Silvestri A, Posca D, Pirrò S, Gherardini PF, Castagnoli L, Mann M, Cesareni G. Deep Proteomics of Breast Cancer Cells Reveals that Metformin Rewires Signaling Networks Away from a Pro-growth State. Cell Syst 2016; 2:159-71. [PMID: 27135362 DOI: 10.1016/j.cels.2016.02.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/02/2015] [Accepted: 02/01/2016] [Indexed: 12/25/2022]
Abstract
Metformin is the most frequently prescribed drug for type 2 diabetes. In addition to its hypoglycemic effects, metformin also lowers cancer incidence. This anti-cancer activity is incompletely understood. Here, we profiled the metformin-dependent changes in the proteome and phosphoproteome of breast cancer cells using high-resolution mass spectrometry. In total, we quantified changes of 7,875 proteins and 15,813 phosphosites after metformin changes. To interpret these datasets, we developed a generally applicable strategy that overlays metformin-dependent changes in the proteome and phosphoproteome onto a literature-derived network. This approach suggested that metformin treatment makes cancer cells more sensitive to apoptotic stimuli and less sensitive to pro-growth stimuli. These hypotheses were tested in vivo; as a proof-of-principle, we demonstrated that metformin inhibits the p70S6K-rpS6 axis in a PP2A-phosphatase dependent manner. In conclusion, analysis of deep proteomics reveals both detailed and global mechanisms that contribute to the anti-cancer activity of metformin.
Collapse
Affiliation(s)
- Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Daniela Posca
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Pirrò
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Matthias Mann
- Department Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany.
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
356
|
Novelle MG, Ali A, Diéguez C, Bernier M, de Cabo R. Metformin: A Hopeful Promise in Aging Research. Cold Spring Harb Perspect Med 2016; 6:a025932. [PMID: 26931809 DOI: 10.1101/cshperspect.a025932] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Even though the inevitable process of aging by itself cannot be considered a disease, it is directly linked to life span and is the driving force behind all age-related diseases. It is an undisputable fact that age-associated diseases are among the leading causes of death in the world, primarily in industrialized countries. During the last several years, an intensive search of antiaging treatments has led to the discovery of a variety of drugs that promote health span and/or life extension. The biguanide compound metformin is widely used for treating people with type 2 diabetes and appears to show protection against cancer, inflammation, and age-related pathologies. Here, we summarize the recent developments about metformin use in translational aging research and discuss its role as a potential geroprotector.
Collapse
Affiliation(s)
- Marta G Novelle
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224 Research Center of Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Ahmed Ali
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Carlos Diéguez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland 21224
| |
Collapse
|
357
|
Al-Wahab Z, Mert I, Tebbe C, Chhina J, Hijaz M, Morris RT, Ali-Fehmi R, Giri S, Munkarah AR, Rattan R. Metformin prevents aggressive ovarian cancer growth driven by high-energy diet: similarity with calorie restriction. Oncotarget 2016; 6:10908-23. [PMID: 25895126 PMCID: PMC4484428 DOI: 10.18632/oncotarget.3434] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/23/2015] [Indexed: 12/11/2022] Open
Abstract
Caloric restriction (CR) was recently demonstrated by us to restrict ovarian cancer growth in vivo. CR resulted in activation of energy regulating enzymes adenosine monophosphate activated kinase (AMPK) and sirtuin 1 (SIRT1) followed by downstream inhibition of Akt-mTOR. In the present study, we investigated the effects of metformin on ovarian cancer growth in mice fed a high energy diet (HED) and regular diet (RD) and compared them to those seen with CR in an immunocompetent isogeneic mouse model of ovarian cancer. Mice either on RD or HED diet bearing ovarian tumors were treated with 200 mg/kg metformin in drinking water. Metformin treatment in RD and HED mice resulted in a significant reduction in tumor burden in the peritoneum, liver, kidney, spleen and bowel accompanied by decreased levels of growth factors (IGF-1, insulin and leptin), inflammatory cytokines (MCP-1, IL-6) and VEGF in plasma and ascitic fluid, akin to the CR diet mice. Metformin resulted in activation of AMPK and SIRT1 and inhibition of pAkt and pmTOR, similar to CR. Thus metformin can closely mimic CR's tumor suppressing effects by inducing similar metabolic changes, providing further evidence of its potential not only as a therapeutic drug but also as a preventive agent.
Collapse
Affiliation(s)
| | - Ismail Mert
- Wayne State University, Detroit, MI, USA.,Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Calvin Tebbe
- Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Jasdeep Chhina
- Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | - Miriana Hijaz
- Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA
| | | | - Rouba Ali-Fehmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Josephine Cancer Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Adnan R Munkarah
- Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA.,Josephine Cancer Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Ramandeep Rattan
- Division of Gynecologic Oncology, Department of Women's Health, Henry Ford Hospital, Detroit, MI, USA.,Josephine Cancer Institute, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
358
|
Liu Z, Yokoyama NN, Blair CA, Li X, Avizonis D, Wu XR, Uchio E, Youssef R, McClelland M, Pollak M, Zi X. High Sensitivity of an Ha-RAS Transgenic Model of Superficial Bladder Cancer to Metformin Is Associated with ∼240-Fold Higher Drug Concentration in Urine than Serum. Mol Cancer Ther 2016; 15:430-8. [PMID: 26921394 DOI: 10.1158/1535-7163.mct-15-0714-t] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/16/2015] [Indexed: 02/07/2023]
Abstract
While pharmacoepidemiologic and laboratory studies have supported the hypothesis that the antidiabetic drug metformin may be useful in treating or preventing cancer, there is limited evidence to suggest which specific cancer sites may be particularly sensitive. Sensitivity likely is determined both by features of tumor pathophysiology and by pharmacokinetic factors. We used UPII-mutant Ha-ras transgenic mice that develop hyperplasia and low-grade, papillary urothelial cell carcinoma to determine whether metformin has activity in a model of superficial bladder cancer. Metformin significantly improved survival, reduced urinary tract obstruction, reduced bladder weight (a surrogate for tumor volume), and led to clear activation of AMP α kinase and inhibition of mTOR signaling in neoplastic tissue. We investigated the basis of the unusual sensitivity of this model to metformin, and observed that following oral dosing, urothelium is exposed to drug concentrations via the urine that are approximately 240-fold higher than those in the circulation. In addition, we observed that bladder cancer cell lines (RT4, UMUC-3, and J82) with homozygous deletion of either TSC1 or PTEN are more sensitive to metformin than those (TEU2, TCCSUP, and HT1376) with wild-type TSC1 and PTEN genes. Our findings provide a strong rationale for clinical trials of oral metformin in treatment of superficial bladder cancer.
Collapse
Affiliation(s)
- Zhongbo Liu
- Department of Urology, University of California, Irvine, Orange, California
| | - Noriko N Yokoyama
- Department of Urology, University of California, Irvine, Orange, California
| | | | - Xuesen Li
- Department of Urology, University of California, Irvine, Orange, California
| | - Daina Avizonis
- Metabolomics Core Facility, Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine and Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, New York
| | - Edward Uchio
- Department of Urology, University of California, Irvine, Orange, California
| | - Ramy Youssef
- Department of Urology, University of California, Irvine, Orange, California
| | - Michael McClelland
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Orange, California
| | - Michael Pollak
- Department of Oncology and Segal Cancer Center, McGill University, Montreal, Quebec, Canada
| | - Xiaolin Zi
- Department of Urology, University of California, Irvine, Orange, California. Department of Pharmacology, University of California, Irvine, Orange, California. Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, California.
| |
Collapse
|
359
|
Chong RW, Vasudevan V, Zuber J, Solomon SS. Metformin Has a Positive Therapeutic Effect on Prostate Cancer in Patients With Type 2 Diabetes Mellitus. Am J Med Sci 2016; 351:416-9. [PMID: 27079349 DOI: 10.1016/j.amjms.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Prostate cancer and type 2 diabetes mellitus (DM2) are both common diseases found in the elderly male population. The diabetic drug, metformin, has been shown to have antineoplastic properties and demonstrated better treatment outcomes when used as adjuvant therapy in patients with breast cancer. The hormonally-sensitive cancer analogous to breast cancer in men is prostate cancer. We investigated improved survival, lower risks of recurrences, and lower, more stable levels of prostate-specific antigen (PSA) in patients with DM2 along with prostate cancer on metformin. METHODS Patients with prostate cancer along with DM2 who remained on metformin were compared with controls who were not on metformin matched by age, weight, race and Gleason score cancer staging. The endpoints of our study included final PSA values, number of recurrences, metastases and number living for each group. RESULTS There were significantly fewer deaths (23% versus 10%), fewer recurrences (15% versus 8%), fewer metastases (5% versus 0%) and fewer secondary cancers (17% versus 6%) in the metformin group (P < 0.004). The final PSA value was lower in the metformin-treated group with a result approaching significance (P = 0.067). The primary treatments for prostate cancer (ie, surgery, radiation and androgen depletion) were found to be comparable in both the groups. CONCLUSIONS Our retrospective study shows that adjuvant metformin therapy leads to a better prognosis in prostate cancer. Not only are PSA levels controlled for several years but also there are significantly fewer cancer recurrences in metformin-treated patients. Overall, these results are promising and should be followed up with a prospective study to assess long-term survival.
Collapse
Affiliation(s)
- R William Chong
- Division of Endocrinology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Vijaya Vasudevan
- Division of Endocrinology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Medicine (Endocrinology) and Research Service, Veterans Affairs Medical Center, Memphis, Tennessee
| | - Jeffrey Zuber
- Department of Preventive Medicine, College of Medicine, Veterans Affairs Medical Center, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Solomon S Solomon
- Division of Endocrinology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee; Department of Medicine (Endocrinology) and Research Service, Veterans Affairs Medical Center, Memphis, Tennessee.
| |
Collapse
|
360
|
Bost F, Decoux-Poullot AG, Tanti JF, Clavel S. Energy disruptors: rising stars in anticancer therapy? Oncogenesis 2016; 5:e188. [PMID: 26779810 PMCID: PMC4728676 DOI: 10.1038/oncsis.2015.46] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 02/06/2023] Open
Abstract
The metabolic features of tumor cells diverge from those of normal cells. Otto Warburg was the first to observe that cancer cells dramatically increase their glucose consumption to generate ATP. He also claimed that cancer cells do not have functional mitochondria or oxidative phosphorylation (OXPHOS) but simply rely on glycolysis to provide ATP to the cell, even in the presence of oxygen (aerobic glycolysis). Several studies have revisited this observation and demonstrated that most cancer cells contain metabolically efficient mitochondria. Indeed, to sustain high proliferation rates, cancer cells require functional mitochondria to provide ATP and intermediate metabolites, such as citrate and cofactors, for anabolic reactions. This difference in metabolism between normal and tumors cells causes the latter to be more sensitive to agents that can disrupt energy homeostasis. In this review, we focus on energy disruptors, such as biguanides, 2-deoxyglucose and 5-aminoimidazole-4-carboxamide ribonucleotide, that interfere with the main metabolic pathways of the cells, OXPHOS, glycolysis and glutamine metabolism. We discuss the preclinical data and the mechanisms of action of these disruptors at the cellular and molecular levels. Finally, we consider whether these drugs can reasonably contribute to the antitumoral therapeutic arsenal in the future.
Collapse
Affiliation(s)
- F Bost
- INSERM, C3M, U1065, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France.,Univ. Nice Sophia Antipolis, C3M, U1065, Nice, France
| | - A-G Decoux-Poullot
- INSERM, C3M, U1065, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France.,Univ. Nice Sophia Antipolis, C3M, U1065, Nice, France
| | - J F Tanti
- INSERM, C3M, U1065, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France.,Univ. Nice Sophia Antipolis, C3M, U1065, Nice, France
| | - S Clavel
- INSERM, C3M, U1065, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France.,Univ. Nice Sophia Antipolis, C3M, U1065, Nice, France
| |
Collapse
|
361
|
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
|
362
|
Topcul M, Cetin I. Effects of metformin on cell kinetic parameters of MCF-7 breast cancer cells in vitro. Asian Pac J Cancer Prev 2016; 16:2351-4. [PMID: 25824763 DOI: 10.7314/apjcp.2015.16.6.2351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this study, the antiproliferative effects of the metformin was evaluated on MCF-7 Cells (human breast adenocarcinoma cell line). For this purpose cell kinetic parameters including cell proliferation assay, mitotic index and labelling index analysis were used. 30 μM, 65 μM and 130 μM Metformin doses were applied to cells for 24, 48 and 72 hours. The results showed that there was a significant decrease in cell proliferation, mitotic index and labelling index for all experimental groups (p<0.05) for all applications.
Collapse
Affiliation(s)
- Mehmet Topcul
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey E-mail :
| | | |
Collapse
|
363
|
Cai H, Zhang Y, Han TK, Everett RS, Thakker DR. Cation-selective transporters are critical to the AMPK-mediated antiproliferative effects of metformin in human breast cancer cells. Int J Cancer 2016; 138:2281-92. [DOI: 10.1002/ijc.29965] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 12/03/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Hao Cai
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy; the University of North Carolina at Chapel Hill; Chapel Hill NC
| | - Yunhui Zhang
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy; the University of North Carolina at Chapel Hill; Chapel Hill NC
- Research Center for Drug Metabolism, Jilin University; Changchun People's Republic of China
| | - Tianxiang Kevin Han
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy; the University of North Carolina at Chapel Hill; Chapel Hill NC
| | - Ruth S. Everett
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy; the University of North Carolina at Chapel Hill; Chapel Hill NC
| | - Dhiren R. Thakker
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy; the University of North Carolina at Chapel Hill; Chapel Hill NC
| |
Collapse
|
364
|
De Souza A, Khawaja KI, Masud F, Saif MW. Metformin and pancreatic cancer: Is there a role? Cancer Chemother Pharmacol 2016; 77:235-42. [PMID: 26740120 DOI: 10.1007/s00280-015-2948-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related deaths in the USA, with a 5-year survival rate of 6 %. Anti-hyperglycemic treatments for type 2 diabetes mellitus that induce hyperinsulinemia (i.e., sulfonylureas) are thought to increase cancer risk, whereas treatments that lower insulin resistance and hyperinsulinemia (i.e., metformin) are considered cancer prevention strategies. Metformin is a cornerstone in the treatment of diabetes mellitus type 2. Retrospective studies have shown a survival benefit in diabetic patients with many solid tumors including pancreatic cancer that have been treated with metformin compared with patients treated with insulin or sulfonylureas. Metformin influences various cellular pathways, including activation of the LKB1/AMPK pathway, inhibition of cell division, promotion of apoptosis and autophagy, down-regulation of circulating insulin, and activation of the immune system. Ongoing research is redefining our understanding about how metformin modulates the molecular pathways implicated in pancreatic cancer. The authors review the topic critically and also give their opinion. Further studies investigating the effect of metformin in combination with chemotherapy, targeted agents, or radiation therapy are undergoing. In addition, the role of metabolic and other biomarkers is needed.
Collapse
Affiliation(s)
- Andre De Souza
- Section of GI Cancers and Experimental Therapeutics, Department of Hematology and Oncology, Tufts University School of Medicine and Tufts Cancer Center, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Khadija Irfan Khawaja
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faisal Masud
- King Edward Medical University, Lahore, Pakistan
| | - Muhammad Wasif Saif
- Section of GI Cancers and Experimental Therapeutics, Department of Hematology and Oncology, Tufts University School of Medicine and Tufts Cancer Center, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA.
| |
Collapse
|
365
|
ur Rasool R, Rah B, Amin H, Nayak D, Chakraborty S, Rawoof A, Mintoo MJ, Yousuf K, Mukherjee D, Kumar LD, Mondhe DM, Goswami A. Dual modulation of Ras-Mnk and PI3K-AKT-mTOR pathways: A Novel c-FLIP inhibitory mechanism of 3-AWA mediated translational attenuation through dephosphorylation of eIF4E. Sci Rep 2016; 6:18800. [PMID: 26728896 PMCID: PMC4700468 DOI: 10.1038/srep18800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/24/2015] [Indexed: 12/19/2022] Open
Abstract
The eukaryotic translation initiation factor 4E (eIF4E) is considered as a key survival protein involved in cell cycle progression, transformation and apoptosis resistance. Herein, we demonstrate that medicinal plant derivative 3-AWA (from Withaferin A) suppressed the proliferation and metastasis of CaP cells through abrogation of eIF4E activation and expression via c-FLIP dependent mechanism. This translational attenuation prevents the de novo synthesis of major players of metastatic cascades viz. c-FLIP, c-Myc and cyclin D1. Moreover, the suppression of c-FLIP due to inhibition of translation initiation complex by 3-AWA enhanced FAS trafficking, BID and caspase 8 cleavage. Further ectopically restored c-Myc and GFP-HRas mediated activation of eIF4E was reduced by 3-AWA in transformed NIH3T3 cells. Detailed underlying mechanisms revealed that 3-AWA inhibited Ras-Mnk and PI3-AKT-mTOR, two major pathways through which eIF4E converges upon eIF4F hub. In addition to in vitro studies, we confirmed that 3-AWA efficiently suppressed tumor growth and metastasis in different mouse models. Given that 3-AWA inhibits c-FLIP through abrogation of translation initiation by co-targeting mTOR and Mnk-eIF4E, it (3-AWA) can be exploited as a lead pharmacophore for promising anti-cancer therapeutic development.
Collapse
Affiliation(s)
- Reyaz ur Rasool
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Bilal Rah
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Hina Amin
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Debasis Nayak
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Souneek Chakraborty
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Abdul Rawoof
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad, AP-50007, India
| | - Mubashir Javed Mintoo
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Khalid Yousuf
- Natural Product Chemistry, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Debaraj Mukherjee
- Natural Product Chemistry, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Lekha Dinesh Kumar
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad, AP-50007, India
| | - Dilip Manikaro Mondhe
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Anindya Goswami
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| |
Collapse
|
366
|
Lauretta R, Lanzolla G, Vici P, Mariani L, Moretti C, Appetecchia M. Insulin-Sensitizers, Polycystic Ovary Syndrome and Gynaecological Cancer Risk. Int J Endocrinol 2016; 2016:8671762. [PMID: 27725832 PMCID: PMC5048026 DOI: 10.1155/2016/8671762] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 12/28/2022] Open
Abstract
Preclinical, early phase clinical trials and epidemiological evidence support the potential role of insulin-sensitizers in cancer prevention and treatment. Insulin-sensitizers improve the metabolic and hormonal profile in PCOS patients and may also act as anticancer agents, especially in cancers associated with hyperinsulinemia and oestrogen dependent cancers. Several lines of evidence support the protection against cancer exerted by dietary inositol, in particular inositol hexaphosphate. Metformin, thiazolidinediones, and myoinositol postreceptor signaling may exhibit direct inhibitory effects on cancer cell growth. AMPK, the main molecular target of metformin, is emerging as a target for cancer prevention and treatment. PCOS may be correlated to an increased risk for developing ovarian and endometrial cancer (up to threefold). Several studies have demonstrated an increase in mortality rate from ovarian cancer among overweight/obese PCOS women compared with normal weight women. Long-term use of metformin has been associated with lower rates of ovarian cancer. Considering the evidence supporting a higher risk of gynaecological cancer in PCOS women, we discuss the potential use of insulin-sensitizers as a potential tool for chemoprevention, hypothesizing a possible rationale through which insulin-sensitizers may inhibit tumourigenesis.
Collapse
Affiliation(s)
- Rosa Lauretta
- Unit of Endocrinology, Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Lanzolla
- Unit of Endocrinology, Department of Systems' Medicine, University of Rome Tor Vergata, Section of Reproductive Endocrinology, Fatebenefratelli Hospital “San Giovanni Calibita” Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Luciano Mariani
- Department of Gynaecologic Oncology, HPV-Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Costanzo Moretti
- Unit of Endocrinology, Department of Systems' Medicine, University of Rome Tor Vergata, Section of Reproductive Endocrinology, Fatebenefratelli Hospital “San Giovanni Calibita” Rome, Italy
| | - Marialuisa Appetecchia
- Unit of Endocrinology, Regina Elena National Cancer Institute, Rome, Italy
- *Marialuisa Appetecchia:
| |
Collapse
|
367
|
Obesity and cancer, a case for insulin signaling. Cell Death Dis 2015; 6:e2037. [PMID: 26720346 PMCID: PMC4720912 DOI: 10.1038/cddis.2015.381] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 02/08/2023]
Abstract
Obesity is a worldwide epidemic, with the number of overweight and obese individuals climbing from just over 500 million in 2008 to 1.9 billion in 2014. Type 2 diabetes (T2D), cardiovascular disease and non-alcoholic fatty liver disease have long been associated with the obese state, whereas cancer is quickly emerging as another pathological consequence of this disease. Globally, at least 2.8 million people die each year from being overweight or obese. It is estimated that by 2020 being overweight or obese will surpass the health burden of tobacco consumption. Increase in the body mass index (BMI) in overweight (BMI>25 kg/m2) and obese (BMI>30 kg/m2) individuals is a result of adipose tissue (AT) expansion, which can lead to fat comprising >50% of the body weight in the morbidly obese. Extensive research over the last several years has painted a very complex picture of AT biology. One clear link between AT expansion and etiology of diseases like T2D and cancer is the development of insulin resistance (IR) and hyperinsulinemia. This review focuses on defining the link between obesity, IR and cancer.
Collapse
|
368
|
Dasgupta B, Chhipa RR. Evolving Lessons on the Complex Role of AMPK in Normal Physiology and Cancer. Trends Pharmacol Sci 2015; 37:192-206. [PMID: 26711141 DOI: 10.1016/j.tips.2015.11.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 02/08/2023]
Abstract
AMP kinase (AMPK) is an evolutionarily conserved enzyme required for adaptive responses to various physiological and pathological conditions. AMPK executes numerous cellular functions, some of which are often perceived at odds with each other. While AMPK is essential for embryonic growth and development, its full impact in adult tissues is revealed under stressful situations that organisms face in the real world. Conflicting reports about its cellular functions, particularly in cancer, are intriguing and a growing number of AMPK activators are being developed to treat human diseases such as cancer and diabetes. Whether these drugs will have only context-specific benefits or detrimental effects in the treatment of human cancer will be a subject of intense research. Here we review the current state of AMPK research with an emphasis on cancer and discuss the yet unresolved context-dependent functions of AMPK in human cancer.
Collapse
Affiliation(s)
- Biplab Dasgupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Rishi Raj Chhipa
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
369
|
Griss T, Vincent EE, Egnatchik R, Chen J, Ma EH, Faubert B, Viollet B, DeBerardinis RJ, Jones RG. Metformin Antagonizes Cancer Cell Proliferation by Suppressing Mitochondrial-Dependent Biosynthesis. PLoS Biol 2015; 13:e1002309. [PMID: 26625127 PMCID: PMC4666657 DOI: 10.1371/journal.pbio.1002309] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/27/2015] [Indexed: 12/17/2022] Open
Abstract
Metformin is a biguanide widely prescribed to treat Type II diabetes that has gained interest as an antineoplastic agent. Recent work suggests that metformin directly antagonizes cancer cell growth through its actions on complex I of the mitochondrial electron transport chain (ETC). However, the mechanisms by which metformin arrests cancer cell proliferation remain poorly defined. Here we demonstrate that the metabolic checkpoint kinases AMP-activated protein kinase (AMPK) and LKB1 are not required for the antiproliferative effects of metformin. Rather, metformin inhibits cancer cell proliferation by suppressing mitochondrial-dependent biosynthetic activity. We show that in vitro metformin decreases the flow of glucose- and glutamine-derived metabolic intermediates into the Tricarboxylic Acid (TCA) cycle, leading to reduced citrate production and de novo lipid biosynthesis. Tumor cells lacking functional mitochondria maintain lipid biosynthesis in the presence of metformin via glutamine-dependent reductive carboxylation, and display reduced sensitivity to metformin-induced proliferative arrest. Our data indicate that metformin inhibits cancer cell proliferation by suppressing the production of mitochondrial-dependent metabolic intermediates required for cell growth, and that metabolic adaptations that bypass mitochondrial-dependent biosynthesis may provide a mechanism of tumor cell resistance to biguanide activity. How does the antidiabetic drug metformin inhibit cancer? This metabolomic study shows that metformin blocks tumor cell proliferation independently of the classic metabolic checkpoints by suppressing mitochondrial-dependent biosynthesis. Cancer is a disease characterized by unregulated proliferation of transformed cells. To meet the increased biosynthetic demands of proliferation, biosynthetic building blocks required for cellular growth must be generated in large quantities. As cancer cells increase their anabolic metabolism to promote cell growth, there is significant interest in targeting these processes for cancer therapy. Metformin is a drug prescribed to treat Type II diabetes that has gained interest as an anti-tumor agent due to its suppressive effects on cancer cell proliferation. However, how metformin works to slow cancer cell growth has remained poorly understood. Here we show that metformin arrests cancer cell proliferation by starving mitochondria of the necessary metabolic intermediates required for anabolic metabolism in tumor cells. This results in reduced proliferation in part due to decreased synthesis of lipids used for membrane biosynthesis. We also show that some cancer cells use alternative metabolic pathways to synthesize lipids independently of mitochondrial metabolism, and that these cells are resistant to the antigrowth effects of metformin. Better understanding of mechanisms of metformin resistance will be crucial for metformin to be used as an effective anticancer agent.
Collapse
Affiliation(s)
- Takla Griss
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Emma E. Vincent
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Robert Egnatchik
- Children’s Medical Center Research Institute, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- McDermott Center for Human Growth and Development, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Harold C. Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Jocelyn Chen
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Eric H. Ma
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Brandon Faubert
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Benoit Viollet
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR 8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- McDermott Center for Human Growth and Development, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Harold C. Simmons Comprehensive Cancer Center, University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Russell G. Jones
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
370
|
Current topics on inhibitors of respiratory complex I. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1857:884-91. [PMID: 26625959 DOI: 10.1016/j.bbabio.2015.11.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/26/2015] [Accepted: 11/17/2015] [Indexed: 12/12/2022]
Abstract
There are a variety of chemicals which regulate the functions of bacterial and mitochondrial complex I. Some of them, such as rotenone and piericidin A, have been indispensable molecular tools in mechanistic studies on complex I. A large amount of experimental data characterizing the actions of complex I inhibitors has been accumulated so far. Recent X-ray crystallographic structural models of entire complex I may be helpful to carefully interpret this data. We herein focused on recent hot topics on complex I inhibitors and the subjects closely connected to these inhibitors, which may provide useful information not only on the structural and functional aspects of complex I, but also on drug design targeting this enzyme. This article is part of a Special Issue entitled Respiratory complex I, edited by Volker Zickermann and Ulrich Brandt.
Collapse
|
371
|
Jeong YK, Kim MS, Lee JY, Kim EH, Ha H. Metformin Radiosensitizes p53-Deficient Colorectal Cancer Cells through Induction of G2/M Arrest and Inhibition of DNA Repair Proteins. PLoS One 2015; 10:e0143596. [PMID: 26599019 PMCID: PMC4657889 DOI: 10.1371/journal.pone.0143596] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 11/06/2015] [Indexed: 12/17/2022] Open
Abstract
The present study addressed whether the combination of metformin and ionizing radiation (IR) would show enhanced antitumor effects in radioresistant p53-deficient colorectal cancer cells, focusing on repair pathways for IR-induced DNA damage. Metformin caused a higher reduction in clonogenic survival as well as greater radiosensitization and inhibition of tumor growth of p53-/- than of p53+/+ colorectal cancer cells and xenografts. Metformin combined with IR induced accumulation of tumor cells in the G2/M phase and delayed the repair of IR-induced DNA damage. In addition, this combination significantly decreased levels of p53-related homologous recombination (HR) repair compared with IR alone, especially in p53-/- colorectal cancer cells and tumors. In conclusion, metformin enhanced radiosensitivity by inducing G2/M arrest and reducing the expression of DNA repair proteins even in radioresistant HCT116 p53-/- colorectal cancer cells and tumors. Our study provides a scientific rationale for the clinical use of metformin as a radiosensitizer in patients with p53-deficient colorectal tumors, which are often resistant to radiotherapy.
Collapse
Affiliation(s)
- Youn Kyoung Jeong
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
- * E-mail: (HH); (MSK)
| | - Ji Young Lee
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Eun Ho Kim
- Division of Heavy Ion Clinical Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
- * E-mail: (HH); (MSK)
| |
Collapse
|
372
|
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
|
373
|
Kim J, Lee J, Kim C, Choi J, Kim A. Anti-cancer effect of metformin by suppressing signaling pathway of HER2 and HER3 in tamoxifen-resistant breast cancer cells. Tumour Biol 2015; 37:5811-9. [PMID: 26581908 DOI: 10.1007/s13277-015-4440-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
Development of new therapeutic strategies is becoming increasingly important to overcome tamoxifen resistance. Recently, much interest has been focused on anti-tumor effects of metformin commonly used to treat type II diabetes. Increased protein expression and signaling of epidermal growth factor receptor (EGFR) family is a possible mechanism involved in tamoxifen resistance. Since HER2/HER3 heterodimers are able to induce strong downstream signaling and activate various biological responses such as cellular proliferation and growth, we investigated the anti-cancer effect of metformin by inhibition of signaling pathway via downregulation of HER2 and HER3 using tamoxifen-resistant MCF-7 (TR MCF-7) cells. Compared to MCF-7 cells, TR MCF-7 cells showed increased expression of EGFR, HER2, and HER3, and metformin inhibited the expression of these proteins in a dose- and time-dependent manner. Metformin inhibited activation of HER2 (Tyr1248)/HER3 (Tyr1289)/Akt (Ser473) as well as cell proliferation and colony formation by estrogenic promotion in MCF-7 and TR MCF-7 cells. Known as a HER3 ligand, heregulin (HRG)-β1-induced phosphorylation of HER2, HER3 and Akt, and protein interaction of HER2/HER3 and colony formation were inhibited by metformin in both cells. Consistent with the results in the two cell lines, we identified that metformin inhibited HER2/HER3/Akt signaling axis activated by HRG-β1 using the HER2 and HER3-overexpressing breast cancer cell line SK-BR-3. Lastly, lapatinib-induced HER3 upregulation was significantly inhibited by treatment of metformin in HER3 siRNA-transfected TR MCF-7 cells. These data suggest that metformin might overcome tamoxifen resistance through the inhibition of expression and signaling of receptor tyrosine kinase HER2 and HER3.
Collapse
MESH Headings
- Adenocarcinoma/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents, Hormonal/pharmacology
- Breast Neoplasms/pathology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Induction/drug effects
- ErbB Receptors/biosynthesis
- Estradiol/pharmacology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, erbB-1
- Genes, erbB-2
- Humans
- Lapatinib
- MCF-7 Cells
- Metformin/pharmacology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neuregulin-1/antagonists & inhibitors
- Neuregulin-1/physiology
- Quinazolines/antagonists & inhibitors
- Quinazolines/pharmacology
- RNA Interference
- RNA, Small Interfering/genetics
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/biosynthesis
- Receptor, ErbB-3/genetics
- Signal Transduction/drug effects
- Tamoxifen/pharmacology
- Tumor Stem Cell Assay
Collapse
Affiliation(s)
- Jinkyoung Kim
- Department of Pathology, Korea University Guro Hospital, #148 Gurodong-ro, Guro-gu, Seoul, 152-703, Korea
| | - Jiyun Lee
- Department of Pathology, College of Medicine, Korea University, #73 Inchon-ro, Seongbuk-gu, Seoul, 136-705, Korea
| | - Chungyeul Kim
- Department of Pathology, Korea University Guro Hospital, #148 Gurodong-ro, Guro-gu, Seoul, 152-703, Korea
| | - Jinhyuk Choi
- Department of Pathology, Korea University Guro Hospital, #148 Gurodong-ro, Guro-gu, Seoul, 152-703, Korea
| | - Aeree Kim
- Department of Pathology, Korea University Guro Hospital, #148 Gurodong-ro, Guro-gu, Seoul, 152-703, Korea.
| |
Collapse
|
374
|
Isaacsson Velho PH, Castro G, Chung CH. Novel Targeted Agents in Head and Neck Squamous Cell Carcinoma. Hematol Oncol Clin North Am 2015; 29:993-1009. [PMID: 26568544 DOI: 10.1016/j.hoc.2015.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Based on currently available genomic data, most head and neck squamous cell carcinoma have few targetable aberrations and immediate clinical translation is challenging. However, potential therapeutic agents listed in this article need to be thoroughly evaluated because there are compelling scientific rationales supporting their development. Concerted effort is required to identify better predictive biomarkers of clinical benefit and improve the therapeutic index. Clinicians need to better understand resistance mechanisms, generate novel hypotheses for appropriate combination regimens and dosing schedules, develop more accurate model systems, and conduct innovative clinical trials.
Collapse
Affiliation(s)
- Pedro H Isaacsson Velho
- Department of Clinical Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Gilberto Castro
- Department of Clinical Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Christine H Chung
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Johns Hopkins Medical Institutions, 1550 Orleans Street CRB-2 Room 546, Baltimore, MD 21287-0014, USA; Department of Otolaryngology-Head and Neck Surgery, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Johns Hopkins Medical Institutions, 1550 Orleans Street CRB-2 Room 546, Baltimore, MD 21287-0014, USA.
| |
Collapse
|
375
|
Marinello PC, da Silva TNX, Panis C, Neves AF, Machado KL, Borges FH, Guarnier FA, Bernardes SS, de-Freitas-Junior JCM, Morgado-Díaz JA, Luiz RC, Cecchini R, Cecchini AL. Mechanism of metformin action in MCF-7 and MDA-MB-231 human breast cancer cells involves oxidative stress generation, DNA damage, and transforming growth factor β1 induction. Tumour Biol 2015; 37:5337-46. [DOI: 10.1007/s13277-015-4395-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/05/2015] [Indexed: 11/27/2022] Open
|
376
|
Lin HC, Stein JD, Nan B, Childers D, Newman-Casey PA, Thompson DA, Richards JE. Association of Geroprotective Effects of Metformin and Risk of Open-Angle Glaucoma in Persons With Diabetes Mellitus. JAMA Ophthalmol 2015; 133:915-23. [PMID: 26022641 DOI: 10.1001/jamaophthalmol.2015.1440] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IMPORTANCE Caloric restriction mimetic drugs have geroprotective effects that delay or reduce risks for a variety of age-associated systemic diseases, suggesting that such drugs might also have the potential to reduce risks of blinding ophthalmologic conditions for which age is a major risk factor. OBJECTIVE To determine whether the caloric restriction mimetic drug metformin hydrochloride is associated with reduced risk of open-angle glaucoma (OAG) in persons with diabetes mellitus. DESIGN, SETTING, AND PATIENTS Retrospective cohort study of patients aged 40 years or older with diabetes mellitus and no preexisting record of OAG in a large US managed care network from January 1, 2001, through December 31, 2010. EXPOSURES Quantity of metformin and other prescribed diabetes medications as captured from outpatient pharmacy records. MAIN OUTCOMES AND MEASURES Risk of developing OAG. RESULTS Of 150 016 patients with diabetes mellitus, 5893 (3.9%) developed OAG. After adjusting for confounding factors, those prescribed the highest quartile of metformin hydrochloride (>1110 g in 2 years) had a 25% reduced OAG risk relative to those who took no metformin (hazard ratio = 0.75; 95% CI, 0.59-0.95; P = .02). Every 1-g increase in metformin hydrochloride use was associated with a 0.16% reduction in OAG risk (adjusted hazard ratio = 0.99984; 95% CI, 0.99969-0.99999; P = .04), which predicts that taking a standard dose of 2 g of metformin hydrochloride per day for 2 years would result in a 20.8% reduction in risk of OAG. After accounting for potential confounders, including metformin and diabetic medications, the risk of developing OAG was increased by 8% (hazard ratio = 1.08; 95% CI, 1.03-1.13; P = .003) for each unit of increase in glycated hemoglobin level. CONCLUSIONS AND RELEVANCE Metformin use is associated with reduction in risk of developing OAG, and risk is reduced even when accounting for glycemic control in the form of glycated hemoglobin level. Other diabetes medications did not confer a similar OAG risk reduction. This study suggests that metformin may be affecting OAG risk on multiple levels, some involving improved glycemic control and some involving mechanisms outside glycemic control such as neurogenesis, inflammatory systems, or longevity pathways targeted by caloric restriction mimetic drugs. If confirmed by prospective clinical trials, these findings could lead to novel treatments for this sight-threatening disease.
Collapse
Affiliation(s)
- Hsien-Chang Lin
- Department of Applied Health Science, Indiana University, Bloomington
| | - Joshua D Stein
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor3Institute for Health Care Policy and Innovation, University of Michigan, Ann Arbor
| | - Bin Nan
- Department of Biostatistics, University of Michigan, Ann Arbor
| | - David Childers
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor5Center for Statistical Consultation and Research, University of Michigan, Ann Arbor
| | - Paula Anne Newman-Casey
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor3Institute for Health Care Policy and Innovation, University of Michigan, Ann Arbor
| | - Debra A Thompson
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor6Department of Biological Chemistry, University of Michigan, Ann Arbor
| | | |
Collapse
|
377
|
EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition. BMC Cancer 2015; 15:855. [PMID: 26542945 PMCID: PMC4636066 DOI: 10.1186/s12885-015-1845-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/23/2015] [Indexed: 01/04/2023] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, is one of the major factors regulating cancer cells metabolism and is considered a promising target for treating cancer. The prototypical NAMPT inhibitor FK866 effectively lowers NAD+ levels in cancer cells, reducing the activity of NAD+-dependent enzymes, lowering intracellular ATP, and promoting cell death. Results We show that FK866 induces a translational arrest in leukemia cells through inhibition of MTOR/4EBP1 signaling and of the initiation factors EIF4E and EIF2A. Specifically, treatment with FK866 is shown to induce 5′AMP-activated protein kinase (AMPK) activation, which, together with EIF2A phosphorylation, is responsible for the inhibition of protein synthesis. Notably, such an effect was also observed in patients’ derived primary leukemia cells including T-cell Acute Lymphoblastic Leukemia. Jurkat cells in which AMPK or LKB1 expression was silenced or in which a non-phosphorylatable EIF2A mutant was ectopically expressed showed enhanced sensitivity to the NAMPT inhibitor, confirming a key role for the LKB1-AMPK-EIF2A axis in cell fate determination in response to energetic stress via NAD+ depletion. Conclusions We identified EIF2A phosphorylation as a novel early molecular event occurring in response to NAMPT inhibition and mediating protein synthesis arrest. In addition, our data suggest that tumors exhibiting an impaired LBK1- AMPK- EIF2A response may be especially susceptible to NAMPT inhibitors and thus become an elective indication for this type of agents. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1845-1) contains supplementary material, which is available to authorized users.
Collapse
|
378
|
Winters B, Plymate S, Zeliadt SB, Holt S, Zhang X, Hu E, Lin DW, Morrissey C, Wooldridge B, Gore JL, Porter MP, Wright JL. Metformin effects on biochemical recurrence and metabolic signaling in the prostate. Prostate 2015. [PMID: 26201966 PMCID: PMC4578998 DOI: 10.1002/pros.23049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Metformin has received considerable attention as a potential anti-cancer agent. Animal and in-vitro prostate cancer (PCa) models have demonstrated decreased tumor growth with metformin, however the precise mechanisms are unknown. We examine the effects of metformin on PCa biochemical recurrence (BCR) in a large clinical database followed by evaluating metabolic signaling changes in a cohort of men undergoing prostate needle biopsy (PNB). METHODS Men treated for localized PCa were identified in a comprehensive clinical database between 2001 and 2010. Cox regression was performed to determine association with BCR relative to metformin use. We next identified a separate case-control cohort of men undergoing prostate needle biopsy (PNB) stratified by metformin use. Differences in mean IHC scores were compared with linear regression for phosphorylated IR, IGF-IR, AKT, and AMPK. RESULTS One thousand seven hundred and thirty four men were evaluated for BCR with mean follow up of 41 months (range 1-121 months). "Ever" metformin use was not associated with BCR (HR 1.12, 0.77-1.65), however men reporting both pre/post-treatment metformin use had a 45% reduction in BCR (HR = 0.55 (0.31-0.96)). For the tissue-based study, 48 metformin users and 42 controls underwent PNB. Significantly greater staining in phosphorylated nuclear (p-IR, p-AKT) and cytoplasmic (p-IR, p-IGF-1R) insulin signaling proteins were seen in patients with PCa detected compared to those with negative PNB (P-values all <0.006). When stratified by metformin use, IGF-1R remained significantly elevated (P = 0.01) in men with PCa detected whereas p-AMPK (P = 0.05) was elevated only in those without PCa. CONCLUSION Metformin use is associated with reduced BCR after treatment of localized PCa when considering pre-diagnostic and cumulative dosing. In men with cancer detected on PNB, insulin signaling markers were significantly elevated compared to negative PNB patients. The finding of IGF-1R elevation in positive PNBs versus p-AMPK elevation in negative PNBs suggests altered metabolic pathway activation precipitated by metformin use.
Collapse
Affiliation(s)
- Brian Winters
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Stephen Plymate
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Health Services Research & Development and GRECC, VA Puget Sound Health Care System, Seattle, WA
| | - Steven B Zeliadt
- Health Services Research & Development and GRECC, VA Puget Sound Health Care System, Seattle, WA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sarah Holt
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Xiaotun Zhang
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Elaine Hu
- Health Services Research & Development and GRECC, VA Puget Sound Health Care System, Seattle, WA
| | - Daniel W. Lin
- Department of Urology, University of Washington School of Medicine, Seattle, WA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Bryan Wooldridge
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - John L Gore
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Michael P Porter
- Department of Urology, University of Washington School of Medicine, Seattle, WA
| | - Jonathan L Wright
- Department of Urology, University of Washington School of Medicine, Seattle, WA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
379
|
Metformin: A Novel Biological Modifier of Tumor Response to Radiation Therapy. Int J Radiat Oncol Biol Phys 2015; 93:454-64. [PMID: 26383681 DOI: 10.1016/j.ijrobp.2015.06.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 01/03/2023]
|
380
|
Coperchini F, Leporati P, Rotondi M, Chiovato L. Expanding the therapeutic spectrum of metformin: from diabetes to cancer. J Endocrinol Invest 2015; 38:1047-55. [PMID: 26233338 DOI: 10.1007/s40618-015-0370-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/23/2015] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Metformin, an oral hypoglycemic agent, was introduced in the clinical practice for the treatment of type 2 diabetes mellitus more than a half-century ago. Over the years, several studies demonstrated that diabetic patients treated with metformin have a lower incidence of cancer, raising the hypothesis that the spectrum of clinical applications of the drug could be expanded also to cancer therapy. Following these initial findings, a large number of studies were performed aimed at elucidating the effects of metformin on different types of tumor, at explaining its direct and indirect anti-cancer mechanisms and at identifying the molecular pathways targeted by the drug. Several clinical trials were also performed aimed at evaluating the potential anti-cancer effect of metformin among diabetic and non-diabetic patients affected by different types of cancer. While the results of several clinical studies are encouraging, a considerable number of other investigations do not support a role of metformin as an anti-cancer agent, and highlight variables possibly accounting for discrepancies. AIM We hereby review the results of in vitro and in vivo studies addressing the issue of the anti-cancer effects of metformin. CONCLUSIONS If in vitro data appear solid, the results provided by in vivo studies are somehow controversial. In this view, larger studies are needed to fully elucidate the role of metformin on cancer development and progression, as well as the specific clinical settings in which metformin could become an anti-cancer drug.
Collapse
Affiliation(s)
- F Coperchini
- Chair of Endocrinology, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Fondazione Salvatore Maugeri I.R.C.C.S., University of Pavia, Via Maugeri 10, 27100, Pavia, Italy
| | - P Leporati
- Chair of Endocrinology, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Fondazione Salvatore Maugeri I.R.C.C.S., University of Pavia, Via Maugeri 10, 27100, Pavia, Italy
| | - M Rotondi
- Chair of Endocrinology, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Fondazione Salvatore Maugeri I.R.C.C.S., University of Pavia, Via Maugeri 10, 27100, Pavia, Italy
| | - L Chiovato
- Chair of Endocrinology, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Fondazione Salvatore Maugeri I.R.C.C.S., University of Pavia, Via Maugeri 10, 27100, Pavia, Italy.
| |
Collapse
|
381
|
Shafaee A, Dastyar DZ, Islamian JP, Hatamian M. Inhibition of tumor energy pathways for targeted esophagus cancer therapy. Metabolism 2015; 64:1193-8. [PMID: 26271140 DOI: 10.1016/j.metabol.2015.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/18/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022]
Abstract
Interest in targeting cancer metabolism has been renewed in recent years with the discovery that many cancer related pathways have a profound effect on metabolism and that many tumors become dependent on specific metabolic processes. Accelerated glucose uptake during anaerobic glycolysis and loss of regulation between glycolytic metabolism and respiration, are the major metabolic changes found in malignant cells. The non-metabolizable glucose analog, 2-deoxy-D-glucose inhibits glucose synthesis and adenosine triphosphate production. The adenosine monophosphate-activated protein kinase (AMPK) is a key sensor of cellular energy and AMPK is a potential target for cancer prevention and/or treatment. Metformin is an activator of AMPK which inhibits protein synthesis and gluconeogenesis during cellular stress. This article reviews the status of clinical and laboratory researches exploring targeted therapies via metabolic pathways for treatment of esophageal cancer.
Collapse
Affiliation(s)
- Abbas Shafaee
- Department of Radiology, School of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Zarei Dastyar
- Department of Medical Radiation Science, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalil Pirayesh Islamian
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Milad Hatamian
- Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
382
|
Metformin synergistically sensitizes FLT3-ITD-positive acute myeloid leukemia to sorafenib by promoting mTOR-mediated apoptosis and autophagy. Leuk Res 2015; 39:1421-7. [PMID: 26505133 DOI: 10.1016/j.leukres.2015.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/11/2015] [Accepted: 09/13/2015] [Indexed: 02/05/2023]
Abstract
Mutations of Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD), accounting for approximately 30% of patients with acute myeloid leukemia (AML), results in poor therapeutic efficacy and short survival. Sorafenib, an oral multikinase inhibitor, can inhibit FLT3 and improve clinical outcome of FLT3 mutated leukemia. Our current studies have shown that, the antidiabetic drug metformin also exerts anti-leukemic effect by activating p-AMPK and synergistically sensitizes FLT3 mutated AML to sorafenib. Both agents suppress cell proliferation in a dose-dependent manner and induce apoptosis via cell cycle arrest, but does not obviously modulate autophagy marker, light chain 3 (LC3). Mechanistically, in the presence of metformin, the anticancer potential of sorafenib, accompanying with increased LC3 levels, is found to be synergistically enhanced with the remarkably reduced protein expression of the mTOR/p70S6K/4EBP1 pathway, while not appreciably altering cell cycle. Overall, these results show metformin in aid of sorafenib may represent a promising and attractive strategy for the treatment of FLT3-ITD mutated AML.
Collapse
|
383
|
Wang FZ, Xie ZS, Xing L, Zhang BF, Zhang JL, Cui PF, Qiao JB, Shi K, Cho CS, Cho MH, Xu X, Li P, Jiang HL. Biocompatible polymeric nanocomplexes as an intracellular stimuli-sensitive prodrug for type-2 diabetes combination therapy. Biomaterials 2015; 73:149-59. [PMID: 26409000 DOI: 10.1016/j.biomaterials.2015.09.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022]
Abstract
Combination therapy is usually considered as a promising strategy owing to its advantages such as reduced doses, minimized side effects and improved therapeutic efficiency in a variety of diseases including diabetes. Here we synthesized a new highly intracellular stimuli-sensitive chitosan-graft-metformin (CS-MET) prodrug by imine reaction between oxidative chitosan and metformin for type 2 diabetes (T2D) therapy. Hypothetically, CS-MET functions dually as an anti-diabetes prodrug as well as a gene delivery vector without superfluous materials. CS-MET formed nanocomplexes with therapeutic gene through electrostatic interactions and entered cells by Organic Cation Transporter (OCT)-independent endocytosis. The incorporation of metformin into chitosan has been found to increase endosomal escape via the proton sponge effect. When vector carrying a short-hairpin RNA (shRNA) silencing sterol regulatory element-binding protein (SREBP), a major transcription factor involved in de novo lipogenisis, it reduced the SREBP mRNA and proteins efficiently. Furthermore, by intraperitoneal injection, CS-MET/shSREBP nanocomplexes effectively knocked down SREBP in livers of western-type diet (WD)-induced obese C57BL/6J mice, markedly reversed insulin resistance and alleviated the fatty liver phenotype without obvious toxic effects. Thus we were able to show that the intracellular stimuli-sensitive CS-MET prodrug renders a potential platform to increase the anti-diabetes activity with synergistic enhancement of gene therapy.
Collapse
Affiliation(s)
- Feng-Zhen Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Shen Xie
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Bing-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Liang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Peng-Fei Cui
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jian-Bin Qiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Kun Shi
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou 221009, China
| | - Chong-Su Cho
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 151-921, South Korea
| | - Myung-Haing Cho
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Department of Nano Fusion Technology, Graduate School of Convergence Science and Technology, Seoul National University, Suwon 443-270, South Korea; Graduate Group of Tumor Biology, Seoul National University, Seoul 151-742, South Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 443-270, South Korea
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacognosy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
384
|
Dokla EME, Fang CS, Lai PT, Kulp SK, Serya RAT, Ismail NSM, Abouzid KAM, Chen CS. Development of Potent Adenosine Monophosphate Activated Protein Kinase (AMPK) Activators. ChemMedChem 2015; 10:1915-23. [PMID: 26350292 DOI: 10.1002/cmdc.201500371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Indexed: 01/28/2023]
Abstract
Previously, we reported the identification of a thiazolidinedione-based adenosine monophosphate activated protein kinase (AMPK) activator, compound 1 (N-[4-({3-[(1-methylcyclohexyl)methyl]-2,4-dioxothiazolidin-5-ylidene}methyl)phenyl]-4-nitro-3-(trifluoromethyl)benzenesulfonamide), which provided a proof of concept to delineate the intricate role of AMPK in regulating oncogenic signaling pathways associated with cell proliferation and epithelial-mesenchymal transition (EMT) in cancer cells. In this study, we used 1 as a scaffold to conduct lead optimization, which generated a series of derivatives. Analysis of the antiproliferative and AMPK-activating activities of individual derivatives revealed a distinct structure-activity relationship and identified 59 (N-(3-nitrophenyl)-N'-{4-[(3-{[3,5-bis(trifluoromethyl)phenyl]methyl}-2,4-dioxothiazolidin-5-ylidene)methyl]phenyl}urea) as the optimal agent. Relative to 1, compound 59 exhibits multifold higher potency in upregulating AMPK phosphorylation in various cell lines irrespective of their liver kinase B1 (LKB1) functional status, accompanied by parallel changes in the phosphorylation/expression levels of p70S6K, Akt, Foxo3a, and EMT-associated markers. Consistent with its predicted activity against tumors with activated Akt status, orally administered 59 was efficacious in suppressing the growth of phosphatase and tensin homologue (PTEN)-null PC-3 xenograft tumors in nude mice. Together, these findings suggest that 59 has clinical value in therapeutic strategies for PTEN-negative cancer and warrants continued investigation in this regard.
Collapse
Affiliation(s)
- Eman M E Dokla
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Room 336, Parks Hall, 500 West 12th Ave., Columbus, OH, 43210, USA.,Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, POB 11566, Abbassia, Cairo, Egypt
| | - Chun-Sheng Fang
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Room 336, Parks Hall, 500 West 12th Ave., Columbus, OH, 43210, USA
| | - Po-Ting Lai
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Room 336, Parks Hall, 500 West 12th Ave., Columbus, OH, 43210, USA
| | - Samuel K Kulp
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Room 336, Parks Hall, 500 West 12th Ave., Columbus, OH, 43210, USA
| | - Rabah A T Serya
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, POB 11566, Abbassia, Cairo, Egypt
| | - Nasser S M Ismail
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, POB 11566, Abbassia, Cairo, Egypt
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, POB 11566, Abbassia, Cairo, Egypt.
| | - Ching-Shih Chen
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, The Ohio State University, Room 336, Parks Hall, 500 West 12th Ave., Columbus, OH, 43210, USA. .,Institute of Biological Chemistry, Academia Sinica, 128 Academia Road Sec. 2, Nankang, Taipei, Taiwan.
| |
Collapse
|
385
|
Hsu CC, Wu LC, Hsia CY, Yin PH, Chi CW, Yeh TS, Lee HC. Energy metabolism determines the sensitivity of human hepatocellular carcinoma cells to mitochondrial inhibitors and biguanide drugs. Oncol Rep 2015; 34:1620-1628. [PMID: 26133123 DOI: 10.3892/or.2015.4092] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/15/2015] [Indexed: 11/05/2022] Open
Abstract
Human hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide particularly in Asia. Deregulation of cellular energetics was recently included as one of the cancer hallmarks. Compounds that target the mitochondria in cancer cells were proposed to have therapeutic potential. Biguanide drugs which inhibit mitochondrial complex I and repress mTOR signaling are clinically used to treat type 2 diabetes mellitus patients (T2DM) and were recently found to reduce the risk of HCC in T2DM patients. However, whether alteration of energy metabolism is involved in regulating the sensitivity of HCC to biguanide drugs is still unclear. In the present study, we treated four HCC cell lines with mitochondrial inhibitors (rotenone and oligomycin) and biguanide drugs (metformin and phenformin), and found that the HCC cells which had a higher mitochondrial respiration rate were more sensitive to these treatments; whereas the HCC cells which exhibited higher glycolysis were more resistant. When glucose was replaced by galactose in the medium, the altered energy metabolism from glycolysis to mitochondrial respiration in the HCC cells enhanced the cellular sensitivity to mitochondrial inhibitors and biguanides. The energy metabolism change enhanced AMP-activated protein kinase (AMPK) activation, mTOR repression and downregulation of cyclin D1 and Mcl-1 in response to the mitochondrial inhibitors and biguanides. In conclusion, our results suggest that increased mitochondrial oxidative metabolism upregulates the sensitivity of HCC to biguanide drugs. Enhancing the mitochondrial oxidative metabolism in combination with biguanide drugs may be a therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Chia-Chi Hsu
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Ling-Chia Wu
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Cheng-Yuan Hsia
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Pen-Hui Yin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan, R.O.C
| | - Chin-Wen Chi
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Tien-Shun Yeh
- Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan, R.O.C
| |
Collapse
|
386
|
Cooper AC, Fleming IN, Phyu SM, Smith TAD. Changes in [18F]Fluoro-2-deoxy-D-glucose incorporation induced by doxorubicin and anti-HER antibodies by breast cancer cells modulated by co-treatment with metformin and its effects on intracellular signalling. J Cancer Res Clin Oncol 2015; 141:1523-32. [PMID: 25579456 DOI: 10.1007/s00432-015-1909-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/04/2015] [Indexed: 11/28/2022]
Abstract
PURPOSES Metformin, currently undergoing clinical trials as an adjuvant for the treatment of breast cancer, modulates the activity of key intracellular signalling molecules which affect 2-[(18)F]Fluoro-2-deoxy-D-glucose ([(18)F]FDG) incorporation. Here, we investigate the effect of drugs used in the treatment of breast cancer combined with metformin on [(18)F]FDG incorporation in HER2- or HER1-overexpressing breast cancer cells to determine whether or not metformin may obscure changes in [(18)F]FDG incorporation induced by clinically utilised anticancer drugs in the treatment of breast cancer. METHODS Three breast cancer cell lines expressing HER2 and one HER2 negative but HER1 positive were exposed to metformin, doxorubicin and trastuzumab or cetuximab. Cytotoxicity was measured by the MTT assay. Expression of active (phospho-) AMPK, PKB (Akt) and ERK was determined by Western blotting. [(18)F]FDG incorporation by cells exposed to drug combinations with metformin was determined. Glucose transport was assessed by measuring the initial rate of uptake of [(3)H]O-methyl-D-glucose ([(3)H]OMG). Phosphorylation of [(18)F]FDG was determined in intact cells after exposure to [(18)F]FDG. RESULTS Phospho-AMPK was increased by metformin in all cell lines whilst phospho-Akt and phospho-ERK expressions were decreased in two. Metformin treatment increased [(18)F]FDG incorporation in all cell lines, and treatment with anti-HER antibodies or doxorubicin only produced minor modulations in the increase induced by metformin alone. Glucose transport was increased in BT474 cells and decreased in SKBr3 and MDA-MB-468 cells after treatment with metformin. The fraction of phosphorylated [(18)F]FDG was increased in metformin-treated cells compared with controls, suggesting that hexokinase efficiency was increased by metformin. CONCLUSION This is the first study to show that increased [(18)F]FDG incorporation by breast cancer cells induced by metformin overwhelms the effect of doxorubicin and anti-HER treatments on [(18)F]FDG incorporation. Metformin-induced increased [(18)F]FDG incorporation was consistently associated with enhanced [(18)F]FDG phosphorylation.
Collapse
Affiliation(s)
- Alasdair C Cooper
- Biomedical Physics Building, School of Medicine and Dentistry, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, Scotland, UK,
| | | | | | | |
Collapse
|
387
|
Yue W, Zheng X, Lin Y, Yang CS, Xu Q, Carpizo D, Huang H, DiPaola RS, Tan XL. Metformin combined with aspirin significantly inhibit pancreatic cancer cell growth in vitro and in vivo by suppressing anti-apoptotic proteins Mcl-1 and Bcl-2. Oncotarget 2015; 6:21208-24. [PMID: 26056043 PMCID: PMC4673260 DOI: 10.18632/oncotarget.4126] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/02/2015] [Indexed: 12/16/2022] Open
Abstract
Metformin and aspirin have been studied extensively as cancer preventive or therapeutic agents. However, the effects of their combination on pancreatic cancer cells have not been investigated. Herein, we evaluated the effects of metformin and aspirin, alone or in combination, on cell viability, migration, and apoptosis as well as the molecular changes in mTOR, STAT3 and apoptotic signaling pathways in PANC-1 and BxPC3 cells. Metformin and aspirin, at relatively low concentrations, demonstrated synergistically inhibitory effects on cell viability. Compared to the untreated control or individual drug, the combination of metformin and aspirin significantly inhibited cell migration and colony formation of both PANC-1 and BxPC-3 cells. Metformin combined with aspirin significantly inhibited the phosphorylation of mTOR and STAT3, and induced apoptosis as measured by caspase-3 and PARP cleavage. Remarkably, metformin combined with aspirin significantly downregulated the anti-apoptotic proteins Mcl-1 and Bcl-2, and upregulated the pro-apoptotic proteins Bim and Puma, as well as interrupted their interactions. The downregulation of Mcl-1 and Bcl-2 was independent of AMPK or STAT3 pathway but partially through mTOR signaling and proteasome degradation. In a PANC-1 xenograft mouse model, we demonstrated that the combination of metformin and aspirin significantly inhibited tumor growth and downregulated the protein expression of Mcl-1 and Bcl-2 in tumors. Taken together, the combination of metformin and aspirin significantly inhibited pancreatic cancer cell growth in vitro and in vivo by regulating the pro- and anti-apoptotic Bcl-2 family members, supporting the continued investigation of this two drug combination as chemopreventive or chemotherapeutic agents for pancreatic cancer.
Collapse
Affiliation(s)
- Wen Yue
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Xi Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Yong Lin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Chung S. Yang
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, P. R. China
| | - Darren Carpizo
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Huarong Huang
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, P. R. China
- Allan H. Conney Laboratory for Anticancer Research, Guangdong University of Technology, Guangzhou, P. R. China
| | - Robert S. DiPaola
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Xiang-Lin Tan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Epidemiology, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
388
|
Silvestri A, Palumbo F, Rasi I, Posca D, Pavlidou T, Paoluzi S, Castagnoli L, Cesareni G. Metformin Induces Apoptosis and Downregulates Pyruvate Kinase M2 in Breast Cancer Cells Only When Grown in Nutrient-Poor Conditions. PLoS One 2015; 10:e0136250. [PMID: 26291325 PMCID: PMC4546379 DOI: 10.1371/journal.pone.0136250] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/03/2015] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Metformin is proposed as adjuvant therapy in cancer treatment because of its ability to limit cancer incidence by negatively modulating the PI3K/AKT/mTOR pathway. In vitro, in addition to inhibiting cancer cell proliferation, metformin can also induce apoptosis. The molecular mechanism underlying this second effect is still poorly characterized and published data are often contrasting. We investigated how nutrient availability can modulate metformin-induced apoptosis in three breast cancer cell lines. MATERIAL AND METHODS MCF7, SKBR3 and MDA-MB-231 cells were plated in MEM medium supplemented with increasing glucose concentrations or in DMEM medium and treated with 10 mM metformin. Cell viability was monitored by Trypan Blue assay and treatment effects on Akt/mTOR pathway and on apoptosis were analysed by Western Blot. Moreover, we determined the level of expression of pyruvate kinase M2 (PKM2), a well-known glycolytic enzyme expressed in cancer cells. RESULTS Our results showed that metformin can induce apoptosis in breast cancer cells when cultured at physiological glucose concentrations and that the pro-apoptotic effect was completely abolished when cells were grown in high glucose/high amino acid medium. Induction of apoptosis was found to be dependent on AMPK activation but, at least partially, independent of TORC1 inactivation. Finally, we showed that, in nutrient-poor conditions, metformin was able to modulate the intracellular glycolytic equilibrium by downregulating PKM2 expression and that this mechanism was mediated by AMPK activation. CONCLUSION We demonstrated that metformin induces breast cancer cell apoptosis and PKM2 downregulation only in nutrient-poor conditions. Not only glucose levels but also amino acid concentration can influence the observed metformin inhibitory effect on the mTOR pathway as well as its pro-apoptotic effect. These data demonstrate that the reduction of nutrient supply in tumors can increase metformin efficacy and that modulation of PKM2 expression/activity could be a promising strategy to boost metformin anti-cancer effect.
Collapse
Affiliation(s)
- Alessandra Silvestri
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
- * E-mail:
| | - Francesco Palumbo
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Ignazio Rasi
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Daniela Posca
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Theodora Pavlidou
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Serena Paoluzi
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Luisa Castagnoli
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
| | - Giovanni Cesareni
- Laboratory of Molecular Genetics, Department of Biology, Tor Vergata University, Rome, Italy
- IRCCS, Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
389
|
Jara JA, López-Muñoz R. Metformin and cancer: Between the bioenergetic disturbances and the antifolate activity. Pharmacol Res 2015; 101:102-8. [PMID: 26277279 DOI: 10.1016/j.phrs.2015.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 01/18/2023]
Abstract
For decades, metformin has been the first-line drug for the treatment of type II diabetes mellitus, and it thus is the most widely prescribed antihyperglycemic drug. Retrospective studies associate the use of metformin with a reduction in cancer incidence and cancer-related death. However, despite extensive research about the molecular effects of metformin in cancer cells, its mode of action remains controversial. In this review, we summarize the current molecular evidence in an effort to elucidate metformin's mode of action against cancer cells. Some authors describe that metformin acts directly on mitochondria, inhibiting complex I and restricting the cell's ability to cope with energetic stress. Furthermore, as the drug interrupts the tricarboxylic acid cycle, metformin-induced alteration of mitochondrial function leads to a compensatory increase in lactate and glycolytic ATP. It has also been reported that cell cycle arrest, autophagy, apoptosis and cell death induction is mediated by the activation of AMPK and Redd1 proteins, thus inhibiting the mTOR pathway. Additionally, unbiased metabolomics studies have provided strong evidence to support that metformin alters the methionine and folate cycles, with a concomitant decrease in nucleotide synthesis. Indeed, purines such as thymidine or hypoxanthine restore the proliferation of tumor cells treated with metformin in vitro. Consequently, some authors prefer to refer to metformin as an "antimetabolite drug" rather than a "mitochondrial toxin". Finally, we also review the current controversy concerning the relationship between the experimental conditions of in vitro-reported effects and the plasma concentrations achieved by chronic treatment with metformin.
Collapse
Affiliation(s)
- J A Jara
- Unidad de Farmacología y Farmacogenética, ICOD, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - R López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
390
|
Braghiroli MI, de Celis Ferrari ACR, Pfiffer TE, Alex AK, Nebuloni D, Carneiro AS, Caparelli F, Senna L, Lobo J, Hoff PM, Riechelmann RP. Phase II trial of metformin and paclitaxel for patients with gemcitabine-refractory advanced adenocarcinoma of the pancreas. Ecancermedicalscience 2015; 9:563. [PMID: 26316884 PMCID: PMC4544571 DOI: 10.3332/ecancer.2015.563] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In patients with adenocarcinoma of the pancreas, there are no standard second-line regimens. Many pre-clinical studies have shown that metformin alone or when combined with paclitaxel has antitumour effects on this tumour. We have tested here the combination of paclitaxel and metformin for patients with gemcitabine-refractory pancreatic cancer. METHODS An uncontrolled phase II trial was carried out based on a two-stage Simon's design, with metformin and paclitaxel for patients with locally advanced or metastatic pancreatic cancer whose disease had progressed during first line treatment with a gemcitabine-based regimen. The primary endpoint was the disease control rate at eight weeks as per response evaluation criteria in solid tumours (RECIST) 1.1. Patients received paclitaxel 80 mg/m(2) weekly for three weeks every 28 days and metformin 850 mg p.o. t.i.d. continuously until progression or intolerance state was reached. RESULTS Twenty patients were enrolled from July 2011 to January 2014: N = 6 (31.6%) achieved the primary endpoint, with all presenting stable disease. Median overall survival (OS) was 128 days (range 17-697) and the median progression free survival (PFS) was 44 days (range 14-210). Eight patients (40%) presented treatment-related G3-4 toxicities with the most common one being diarrhoea. CONCLUSIONS Despite the encouraging pre-clinical evidence of the antitumour activity of metformin in adenocarcinoma of the pancreas, the primary endpoint of the disease control rate was not met. Besides, the treatment combination was poorly tolerated and could not be studied further. This study highlights the importance of performing clinical trials to reassure preclinical or observational data.
Collapse
Affiliation(s)
- Maria Ignez Braghiroli
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Anezka C R de Celis Ferrari
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Tulio Eduardo Pfiffer
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Alexandra Kichfy Alex
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Daniela Nebuloni
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Allyne S Carneiro
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Fernanda Caparelli
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Luiz Senna
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Juliana Lobo
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Paulo Marcelo Hoff
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| | - Rachel P Riechelmann
- Discipline of Radiology and Oncology, Instituto do Cancer do Estado de São Paulo, Universidade de São Paulo, Brazil, Av Dr Arnaldo 251, 12º andar São Paulo, SP, 01246-000 Brazil
| |
Collapse
|
391
|
Pan Q, Yang GL, Yang JH, Lin SL, Liu N, Liu SS, Liu MY, Zhang LH, Huang YR, Shen RL, Liu Q, Gao JX, Bo JJ. Metformin can block precancerous progression to invasive tumors of bladder through inhibiting STAT3-mediated signaling pathways. J Exp Clin Cancer Res 2015; 34:77. [PMID: 26245871 PMCID: PMC4553001 DOI: 10.1186/s13046-015-0183-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/15/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metformin is the first line of oral antidiabetic drug in the biguanide class for treatment of type 2 diabetes. Increasing evidence has suggested that it is a potential anti-tumor drug. However, the mechanisms underlying inhibiting tumor development remain elusive, especially in bladder tumors. METHODS T24 and J82 cell lines were used as an in vitro model, and 24 female SD rats were used to build an N-methyl-N-nitrosourea (MNU)-induced orthotopic rat bladder cancer model. Transfection of lentivirus-based shRNA was used to construct the STAT3-KNOCKDOWN T24 cell line. After metformin treatment, the viability of bladde cancer cells was determined by CCK8. Cell cycle distribution and apoptosis were assessed by flow cytometry. The migration and invasion abilities of cells were evaluated by wound healing and transwell asssays. The inactivation of stat3 pahtway was examined by qRTPCR, western blot and Immunofluorescence. RESULTS Metformin can effectively inhibit precancerous progression to invasive cancer in an MNU-induced rat orthotopic bladder tumor model, although it could not completely suppress normal cells transforming into tumor cells. While the MNU could induce 50 % rats (4/8) to develop invasive bladder cancers, the rats co-administrated with metformin failed to develop invasive tumors but retained at precancerous or non-invasive stages, exhibiting as dysplasia, papillary tumor and/or carcinoma in situ (CIS). Accordingly, phosphorylation of signal transducer and activator of transcription 3 (STAT3), which is a well known oncogene, was significantly inhibited in the tumors of rats treated with metformin. In vitro experiments revealed that the metformin could efficiently inhibit STAT3 activation, which was associated with the cell cycle arrest, reduction of cell proliferation, migration and invasiveness, and increase in apoptotic cell death of bladder cancer cell lines. CONCLUSIONS These findings provide for the first time the evidence that metformin can block precancerous lesions progressing to invasive tumors through inhibiting the activation of STAT3 pathway, and may be used for treatment of the non-invasive bladder cancers to prevent them from progression to invasive tumors.
Collapse
Affiliation(s)
- Qi Pan
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Guo-Liang Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiang-Hua Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Shi-Long Lin
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ning Liu
- State Key Laboratory of Oncogene and Related Genes, Shanghai, China.
| | - Shan-Shan Liu
- State Key Laboratory of Oncogene and Related Genes, Shanghai, China.
| | - Meng-Yao Liu
- State Key Laboratory of Oncogene and Related Genes, Shanghai, China.
| | - Lian-Hua Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yi-Ran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ru-long Shen
- Department of Pathology, Ohio State University School of Medicine, Columbus, OH, USA.
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jian-Xin Gao
- State Key Laboratory of Oncogene and Related Genes, Shanghai, China. .,Laboratory of Tumorigenesis and Immunity, Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Juan-Jie Bo
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
392
|
Kallergi A, Chambre C, Duchemann B, Fysekidis M, Bihan H. Diabetes Mellitus and Colorectal Cancer Risk. CURRENT COLORECTAL CANCER REPORTS 2015. [DOI: 10.1007/s11888-015-0274-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
393
|
Wang J, Gao Q, Wang D, Wang Z, Hu C. Metformin inhibits growth of lung adenocarcinoma cells by inducing apoptosis via the mitochondria-mediated pathway. Oncol Lett 2015; 10:1343-1349. [PMID: 26622674 DOI: 10.3892/ol.2015.3450] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/29/2015] [Indexed: 01/30/2023] Open
Abstract
Metformin is commonly used to treat type II diabetes, although it may also reduce the risk of cancer and improve the associated prognosis. However, its mode of action in cancer remains unclear. The present study evaluated the effects of metformin on lung adenocarcinoma A549 cells and identified molecular mechanisms of metformin activity. The A549 cells were treated with metformin at different concentrations and cell viability was assayed by using an MTT assay. The cell cycle and the apoptosis rate were assayed by flow cytometry. Nude mice were transplanted with A549 cells and the tumor growth inhibition rate was detected. Once the A549 cells had been treated with 20 mM metformin for 48 h, the cell cycle was arrested in the G0/Gl phase and the apoptosis rate was 20.57±3.16%. The expression of the B-cell lymphoma (Bcl)-2 and Bcl-extra large proteins was downregulated following metformin treatment, while Bax protein expression was significantly increased. Tumor size in the high-dose metformin and cisplatin plus metformin groups was significantly smaller, and the inhibition rates were 41.3 and 72.9%, respectively, compared with the control group. These results indicated that metformin displays anticancer activity against lung adenocarcinoma by causing G1 arrest of the cell cycle and subsequent cell apoptosis through the mitochondria-dependent pathway in A549 cells. Furthermore, it was found that metformin dramatically inhibited lung adenocarcinoma tumor growth in vivo. These data suggest that metformin may become a potential cytotoxic drug in the prevention and treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Junling Wang
- Department of Medical Oncology, Binzhou Tuberculosis Control Center, Binzhou Medical College, Binzhou, Shandong 250000, P.R. China
| | - Qiuling Gao
- Department of Radiology, Binzhou People's Hospital, Binzhou, Shandong 250000, P.R. China
| | - Decui Wang
- Department of Medical Oncology, Binzhou Tuberculosis Control Center, Binzhou Medical College, Binzhou, Shandong 250000, P.R. China
| | - Zhiqiang Wang
- Department of Medical Oncology, Binzhou Tuberculosis Control Center, Binzhou Medical College, Binzhou, Shandong 250000, P.R. China
| | - Chun Hu
- Department of Oncology, RuiJin Hospital, Shanghai JiaoTong University, Shanghai 200001, P.R. China
| |
Collapse
|
394
|
Olivares O, Däbritz JHM, King A, Gottlieb E, Halsey C. Research into cancer metabolomics: Towards a clinical metamorphosis. Semin Cell Dev Biol 2015; 43:52-64. [PMID: 26365277 DOI: 10.1016/j.semcdb.2015.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 12/26/2022]
Abstract
The acknowledgement that metabolic reprogramming is a central feature of cancer has generated high expectations for major advances in both diagnosis and treatment of malignancies through addressing metabolism. These have so far only been partially fulfilled, with only a few clinical applications. However, numerous diagnostic and therapeutic compounds are currently being evaluated in either clinical trials or pre-clinical models and new discoveries of alterations in metabolic genes indicate future prognostic or other applicable relevance. Altogether, these metabolic approaches now stand alongside other available measures providing hopes for the prospects of metabolomics in the clinic. Here we present a comprehensive overview of both ongoing and emerging clinical, pre-clinical and technical strategies for exploiting unique tumour metabolic traits, highlighting the current promises and anticipations of research in the field.
Collapse
Affiliation(s)
- Orianne Olivares
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, UK
| | - J Henry M Däbritz
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, UK
| | - Ayala King
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, UK
| | - Eyal Gottlieb
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, UK.
| | - Christina Halsey
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, UK.
| |
Collapse
|
395
|
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
|
396
|
Pharmacologic screens reveal metformin that suppresses GRP78-dependent autophagy to enhance the anti-myeloma effect of bortezomib. Leukemia 2015; 29:2184-91. [PMID: 26108695 PMCID: PMC4635337 DOI: 10.1038/leu.2015.157] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 02/07/2023]
Abstract
Although the therapeutic benefit of proteasome inhibition in multiple myeloma remains unchallenged, drug resistance inevitably emerges through mechanisms that remain elusive. Bortezomib provokes unwanted protein accumulation and the endoplasmic reticulum stress to activate the unfolded protein response (UPR) and autophagy as compensatory mechanisms that restore protein homeostasis. High-throughput screens to detect pharmacologics that modulated autophagy to enhance the anti-myeloma effect of bortezomib revealed metformin, a widely used antidiabetic agent with proven efficacy and limited adverse effects. Metformin co-treatment with bortezomib suppressed induction of the critical UPR effector glucose-regulated protein 78 (GRP78) to impair autophagosome formation and enhance apoptosis. Gene expression profiling of newly diagnosed myeloma patient tumors further correlated the hyperexpression of GRP78-encoding HSPA5 with reduced clinical response to bortezomib. The effect of bortezomib was enhanced with metformin co-treatment using myeloma patient tumor cells and the chemoresistant, stem cell-like side population that may contribute to disease recurrence. The relevance of the findings was confirmed in vivo as shown by metformin co-treatment with bortezomib that delayed the growth of myeloma xenotransplants. Taken together, our results suggest that metformin suppresses GRP78, a key driver of bortezomib-induced autophagy, and support the pharmacologic repositioning of metformin to enhance the anti-myeloma benefit of bortezomib.
Collapse
|
397
|
Transcriptional remodeling in response to transfer upon carbon-limited or metformin-supplemented media in S. cerevisiae and its effect on chronological life span. Appl Microbiol Biotechnol 2015; 99:6775-89. [PMID: 26099330 DOI: 10.1007/s00253-015-6728-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/17/2015] [Accepted: 05/25/2015] [Indexed: 12/16/2022]
Abstract
One of the factors affecting chronological life span (CLS) in budding yeast is nutrient, especially carbon limitation. Aside from metabolites in the growth medium such as glucose, amino acids, and acetic acid, many pharmaceuticals have also been proven to alter CLS. Besides their impact on life span, these drugs are also prospective chemicals to treat the age-associated diseases, so the identification of their action mechanism and their potential side effects is of crucial importance. In this study, the effects of caloric restriction and metformin, a dietary mimetic pharmaceutical, on yeast CLS are compared. Saccharomyces cerevisiae cells grown in synthetic dextrose complete (SDC) up to mid-exponential phase were either treated with metformin or were subjected to glucose limitation. The impacts of these perturbations were analyzed via transcriptomics, and the common (stimulation of glucose uptake, induction of mitochondrial maintenance, and reduction of protein translation) and divergent (stimulation of aerobic respiration and reprogramming of respiratory electron transport chain (ETC)) cellular responses specific to each treatment were determined. These results revealed that both glucose limitation and metformin treatment stimulate CLS extension and involve the mitochondrial function, probably by creating an efficient mitochondria-to-nucleus signaling of either aerobic respiration or ETC signaling stimulation, respectively.
Collapse
|
398
|
Abstract
On the basis of data obtained from a prospective cohort of Chinese patients with type 2 diabetes mellitus (T2DM), we discuss cancer subphenotypes (risk factors) in patients with T2DM, which can lead to drug-cancer subphenotype interactions. These subphenotypes include HDL cholesterol levels <1.0 mmol/l, co-occurrence of LDL cholesterol levels <2.8 mmol/l and triglyceride levels <1.7 mmol/l, and co-occurrence of LDL cholesterol levels <2.8 mmol/l and albuminuria. The increased risk of cancer associated with low levels of HDL cholesterol, low LDL cholesterol levels plus low triglyceride levels, and low levels of LDL cholesterol plus albuminuria can be reduced by treatment with metformin, renin-angiotensin system (RAS) inhibitors and statins, respectively. Mechanistic studies support the hypothesis that dysregulation of the 5'-AMP-activated protein kinase pathway and crosstalk between the RAS and insulin-like growth factor 1-cholesterol pathways create a cancer-promoting milieu in patients with T2DM. These findings highlight that in Chinese individuals, multiple pathways are implicated in the link between T2DM and cancer, which can generate multiple subphenotypes as well as drug-subphenotype interactions.
Collapse
Affiliation(s)
- Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Heung M Lee
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong SAR, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
399
|
Imai A, Ichigo S, Matsunami K, Takagi H, Yasuda K. Clinical benefits of metformin in gynecologic oncology. Oncol Lett 2015; 10:577-582. [PMID: 26622536 DOI: 10.3892/ol.2015.3262] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 05/20/2015] [Indexed: 12/28/2022] Open
Abstract
Evidence has suggested that diabetes may contribute to the initiation and progression of specific types of cancer. Metformin, a biguanide, has become the preferred first-line therapy for the treatment of type 2 diabetes. Metformin is inexpensive, has a proven safety profile and is able to be safely combined with additional antidiabetic agents. In addition to the well-established antidiabetic effects of metformin, there has also been notable interest in its antitumor properties. The present review discusses the emerging role of metformin as an example of an existing drug, used worldwide in the treatment of diabetes, which has been demonstrated to exert significant in vitro and in vivo anticancer activities and has thus been investigated in clinical trials. In gynecologic oncology, metformin has been suggested to exhibit significant treatment efficacy against endometrial cancer. Three studies have demonstrated the potential therapeutic effects of metformin on the survival outcome of patients with ovarian cancer and in ovarian cancer prevention. However, this evidence was based on observational studies. Metformin has been shown to exert no statistically significant beneficial effect on cervical cancer incidence or mortality. By cancer site, the current limited insights highlight the need for clinical investigations and better-designed studies, along with evaluation of the effects of metformin on cancer at other sites.
Collapse
Affiliation(s)
- Atsushi Imai
- Department of Obstetrics and Gynecology, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan ; Institute of Endocrine-Related Cancer, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan
| | - Satoshi Ichigo
- Department of Obstetrics and Gynecology, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan
| | - Kazutoshi Matsunami
- Department of Obstetrics and Gynecology, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan
| | - Hiroshi Takagi
- Department of Obstetrics and Gynecology, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan
| | - Keigo Yasuda
- Department of Endocrinology and Internal Medicine, Matsunami General Hospital, Kasamatsu, Gifu 501-6062, Japan
| |
Collapse
|
400
|
Association between Gastroenterological Malignancy and Diabetes Mellitus and Anti-Diabetic Therapy: A Nationwide, Population-Based Cohort Study. PLoS One 2015; 10:e0125421. [PMID: 25978841 PMCID: PMC4433253 DOI: 10.1371/journal.pone.0125421] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/23/2015] [Indexed: 01/15/2023] Open
Abstract
Background The relationship between diabetes mellitus (DM) and cancer incidence has been evaluated in limited kinds of cancer. The effect of anti-diabetic therapy (ADT) on carcinogenesis among diabetic patients is also unclear. Materials and Methods Using population-based representative insurance claims data in Taiwan, 36,270 DM patients and 145,080 comparison subjects without DM were identified from claims from 2005 to 2010. The association between the top ten leading causes of cancer-related death in Taiwan and DM was evaluated. Whether ADT altered the risk of developing cancer was also investigated. Results Incidence of cancer at any site was significantly higher in patients with DM than in those without (p<0.001). The risk of carcinogenesis imparted by DM was greatest in gastroenterological malignancies (liver, pancreas, and colorectal cancer) as well as lung, breast and oral cancer (p<0.001). Among the oral types of ADT, metformin decreased the risk of lung and liver cancer, but had less effect on reducing the risk of colorectal cancer. α-glucosidase inhibitor decreased the risk of developing liver, colorectal, and breast cancer. Apart from intermediate-acting insulin, rapid-acting, long-acting, and combination insulin treatment significantly reduced the overall cancer risk among all DM patients. In subgroup analysis, long-acting insulin treatment significantly decreased the risk of lung, liver, and colorectal cancer. Conclusion Our results supported the notion that pre-existing DM increases the incidence of gastroenterological cancer. ADT, especially metformin, α-glucosidase inhibitor, and long-acting insulin treatment, may protect patients with DM against these malignancies. It is crucial that oncologists should closely collaborate with endocrinologists to provide an optimal cancer-specific therapy and diabetic treatment to patients simultaneously with cancer and DM.
Collapse
|