101
|
Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021; 10:antiox10050701. [PMID: 33946704 PMCID: PMC8146591 DOI: 10.3390/antiox10050701] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
In humans, the glutathione S-transferases (GST) protein family is composed of seven members that present remarkable structural similarity and some degree of overlapping functionalities. GST proteins are crucial antioxidant enzymes that regulate stress-induced signaling pathways. Interestingly, overactive GST proteins are a frequent feature of many human cancers. Recent evidence has revealed that the biology of most GST proteins is complex and multifaceted and that these proteins actively participate in tumorigenic processes such as cell survival, cell proliferation, and drug resistance. Structural and pharmacological studies have identified various GST inhibitors, and these molecules have progressed to clinical trials for the treatment of cancer and other diseases. In this review, we discuss recent findings in GST protein biology and their roles in cancer development, their contribution in chemoresistance, and the development of GST inhibitors for cancer treatment.
Collapse
|
102
|
Song Y, Ma P, Gao Y, Xiao P, Xu L, Liu H. A Bibliometrics Analysis of Metformin Development From 1980 to 2019. Front Pharmacol 2021; 12:645810. [PMID: 33995056 PMCID: PMC8113770 DOI: 10.3389/fphar.2021.645810] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin, the first-line oral blood glucose-lowering agent to manage type 2 diabetes, has gained growing popularity on both clinical application and basic research since early 1980s. A thorough and systematic knowledge map of metformin is pertinent to evaluate the research frontier and determine knowledge gaps. To this end, 20, 526 publications were analyzed by bibliometrics and data visualization to demonstrate the current global research status, potential hotspots, and perspectives on future research directions. In addition, the metformin development along the historical line was illustrated over the last 40 years. In sum, this study provides a comprehensive analysis that delineates the evolution of the historical milestones of metformin development, and we discuss the future research directions based on objective data analysis from a wide spectrum of metformin research areas.
Collapse
Affiliation(s)
- Yanjun Song
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Pei Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yu Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Peigen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lijia Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Haibo Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
103
|
Xu L, Wang X, Chen Y, Soong L, Chen Y, Cai J, Liang Y, Sun J. Metformin Modulates T Cell Function and Alleviates Liver Injury Through Bioenergetic Regulation in Viral Hepatitis. Front Immunol 2021; 12:638575. [PMID: 33968030 PMCID: PMC8097169 DOI: 10.3389/fimmu.2021.638575] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Metformin is not only the first-line medication for the treatment of type 2 diabetes, but it is also effective as an anti-inflammatory, anti-oxidative and anti-tumor agent. However, the effect of metformin during viral hepatitis remains elusive. Using an adenovirus (Ad)-induced viral hepatitis mouse model, we found that metformin treatment significantly attenuated liver injury, with reduced serum aspartate transaminase (AST) and alanine transaminase (ALT) levels and liver histological changes, presumably via decreased effector T cell responses. We then demonstrated that metformin reduced mTORC1 activity in T cells from infected mice, as evidenced by decreased phosphorylation of ribosome protein S6 (p-S6). The inhibitory effects on the mTORC1 signaling by metformin was dependent on the tuberous sclerosis complex 1 (TSC1). Mechanistically, metformin treatment modulated the phosphorylation of dynamin-related protein 1 (Drp-1) and mitochondrial fission 1 protein (FIS1), resulting in increased mass in effector T cells. Moreover, metformin treatment promoted mitochondrial superoxide production, which can inhibit excessive T cell activation in viral hepatitis. Together, our results revealed a protective role and therapeutic potential of metformin against liver injury in acute viral hepatitis via modulating effector T cell activation via regulating the mTORC1 pathway and mitochondrial functions.
Collapse
Affiliation(s)
- Lanman Xu
- Department of Infectious Diseases and Liver Diseases, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo, China.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Xiaofang Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Chen
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, United States
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, China
| | - Jiyang Cai
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, United States
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
104
|
Janda M, Robledo KP, Gebski V, Armes JE, Alizart M, Cummings M, Chen C, Leung Y, Sykes P, McNally O, Oehler MK, Walker G, Garrett A, Tang A, Land R, Nicklin JL, Chetty N, Perrin LC, Hoet G, Sowden K, Eva L, Tristram A, Obermair A. Complete pathological response following levonorgestrel intrauterine device in clinically stage 1 endometrial adenocarcinoma: Results of a randomized clinical trial. Gynecol Oncol 2021; 161:143-151. [PMID: 33762086 DOI: 10.1016/j.ygyno.2021.01.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/24/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE Intrauterine levonorgestrel (LNG-IUD) is used to treat patients with endometrial adenocarcinoma (EAC) and endometrial hyperplasia with atypia (EHA) but limited evidence is available on its effectiveness. The study determined the extent to which LNG-IUD with or without metformin (M) or weight loss (WL) achieves a pathological complete response (pCR) in patients with EAC or EHA. PATIENTS AND METHODS This phase II randomized controlled clinical trial enrolled patients with histologically confirmed, clinically stage 1 FIGO grade 1 EAC or EHA; a body mass index > 30 kg/m2; a depth of myometrial invasion of less than 50% on MRI; a serum CA125 ≤ 30 U/mL. All patients received LNG-IUD and were randomized to observation (OBS), M (500 mg orally twice daily), or WL (pooled analysis). The primary outcome measure was the proportion of patients developing a pCR (defined as absence of any evidence of EAC or EHA) after 6 months. RESULTS From December 2012 to October 2019, 165 patients were enrolled and 154 completed the 6-months follow up. Women had a mean age of 53 years, and a mean BMI of 48 kg/m2. Ninety-six patients were diagnosed with EAC (58%) and 69 patients with EHA (42%). Thirty-five participants were randomized to OBS, 36 to WL and 47 to M (10 patients were withdrawn). After 6 months the rate of pCR was 61% (95% CI 42% to 77%) for OBS, 67% (95% CI 48% to 82%) for WL and 57% (95% CI 41% to 72%) for M. Across the three treatment groups, the pCR was 82% and 43% for EHA and EAC, respectively. CONCLUSION Complete response rates at 6 months were encouraging for patients with EAC and EHA across the three groups. TRIAL REGISTRATION U.S. National Library of Medicine, NCT01686126.
Collapse
Affiliation(s)
- Monika Janda
- Centre for Health Services Research, The University of Queensland, QLD, Australia
| | - Kristy P Robledo
- University of Sydney NHMRC Clinical Trials Centre, Sydney, NSW, Australia
| | - Val Gebski
- University of Sydney NHMRC Clinical Trials Centre, Sydney, NSW, Australia
| | - Jane E Armes
- Sunshine Coast University Hospital Laboratory, Birtinya, QLD, Australia
| | | | - Margaret Cummings
- University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia; Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Yee Leung
- Division of Obstetrics and Gynaecology, The University of Western Australia, WA, Australia
| | - Peter Sykes
- Christchurch Women's Hospital, Canterbury District Health Board, Christchurch, New Zealand; University of Otago, Christchurch, New Zealand
| | - Orla McNally
- Department of Oncology and Dysplasia, Royal Women's Hospital, Melbourne, VIC, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Andrea Garrett
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Amy Tang
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Russell Land
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - James L Nicklin
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Naven Chetty
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Mater Health Services, Brisbane, Australia
| | - Lewis C Perrin
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Mater Health Services, Brisbane, Australia
| | - Greet Hoet
- The Townsville Hospital, Townsville, QLD, Australia
| | | | - Lois Eva
- National Women's Health, Auckland City Hospital, Auckland, New Zealand
| | | | - Andreas Obermair
- Queensland Centre for Gynaecological Cancer Research, The University of Queensland, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia.
| |
Collapse
|
105
|
Hu D, Xie F, Xiao Y, Lu C, Zhong J, Huang D, Chen J, Wei J, Jiang Y, Zhong T. Metformin: A Potential Candidate for Targeting Aging Mechanisms. Aging Dis 2021; 12:480-493. [PMID: 33815878 PMCID: PMC7990352 DOI: 10.14336/ad.2020.0702] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Aging is a universal phenomenon in all biological organisms, defined by the loss of reproductive capacity and a progressive decline in fitness. In humans, aging is further associated with an increased incidence of disease conditions. The current aging population has become a primary public burden of the 21st century. Therefore, to delay the aging process and maintain fitness in the aging population, the discovery of novel anti-aging drugs remains an urgent need. In recent years, metformin, a widely used hypoglycemic drug, has attracted growing attention in the field of anti-aging research. Reportedly, numerous studies have indicated that metformin regulates aging-related pathways, possibly delaying the aging process by modulating these pathways. The elucidation of these anti-aging effects may provide insights into the age-retarding potential of metformin. The present review focuses on the predominant molecular mechanisms associated with aging, as well as the anti-aging effects of metformin.
Collapse
Affiliation(s)
- Die Hu
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,2Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,2Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yongwei Xiao
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,2Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Chen Lu
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Jianing Zhong
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,3Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,4Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jie Chen
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,2Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jifu Wei
- 4Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yu Jiang
- 5Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tianyu Zhong
- 1The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China.,2Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.,4Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
106
|
Stang K, Alite F, Adams W, Altoos B, Small C, Melian E, Emami B, Harkenrider M. Impact of Concurrent Coincident Use of Metformin During Lung Stereotactic Body Radiation Therapy. Cureus 2021; 13:e14157. [PMID: 33927955 PMCID: PMC8076758 DOI: 10.7759/cureus.14157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 03/28/2021] [Indexed: 01/26/2023] Open
Abstract
Introduction Recent data suggest synergy of chemoradiotherapy and metformin in locally-advanced non-small cell lung cancer (NSCLC). It remains unclear if similar synergy exists with stereotactic lung body radiation therapy (SBRT) and metformin. We analyzed the role of metformin on progression-free survival (PFS) and toxicity in the setting of lung SBRT. Methods We identified 31 patients on metformin-treated with SBRT for early-stage NSCLC. Eighty-nine similarly treated patients were chosen as controls. Kaplan-Meier method was used to estimate cumulative PFS probabilities. Results Median follow-up was 30.7 months. Forty-two patients had diabetes, 31 (74%) of which were taking metformin concurrent with SBRT. Median PFS for metformin-users vs. metformin non-users was 36.4 months vs 48.9 months, respectively (p = 0.29). Among diabetic patients, median PFS for metformin users was 36.4 months and was unobserved for non-users (p= 0.40). On univariable analysis, male sex (p = 0.03) and tumor size (p = 0.01) were associated with the risk of progression or death; use of metformin was not significant (p = 0.34). There was no difference in grade ≥2 radiation pneumonitis between metformin users vs non-users (p = 0.51) Conclusion In this retrospective sample of lung SBRT patients, we did not detect a meaningful effect of concurrent metformin use on PFS. Since SBRT and conventional RT may have different cell kill mechanisms, the previously described beneficial effects of metformin may not apply in a hypofractionated setting. These results should be validated in an independent dataset, and we await the results of ongoing clinical trials.
Collapse
Affiliation(s)
- Kyle Stang
- Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, USA
| | - Fiori Alite
- Radiation Oncology, Geisinger Cancer Institute, Danville, USA
| | - William Adams
- Public Health Sciences, Loyola University Chicago, Chicago, USA
| | - Basel Altoos
- Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, USA
| | - Christina Small
- Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, USA
| | - Edward Melian
- Radiation Oncology, Loyola University Medical Center, Maywood, USA
| | - Bahman Emami
- Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, USA
| | - Matthew Harkenrider
- Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, USA
| |
Collapse
|
107
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
108
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
109
|
Multifaceted Mechanisms of Action of Metformin Which Have Been Unraveled One after Another in the Long History. Int J Mol Sci 2021; 22:ijms22052596. [PMID: 33807522 PMCID: PMC7962041 DOI: 10.3390/ijms22052596] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 01/08/2023] Open
Abstract
While there are various kinds of drugs for type 2 diabetes mellitus at present, in this review article, we focus on metformin which is an insulin sensitizer and is often used as a first-choice drug worldwide. Metformin mainly activates adenosine monophosphate-activated protein kinase (AMPK) in the liver which leads to suppression of fatty acid synthesis and gluconeogenesis. Metformin activates AMPK in skeletal muscle as well, which increases translocation of glucose transporter 4 to the cell membrane and thereby increases glucose uptake. Further, metformin suppresses glucagon signaling in the liver by suppressing adenylate cyclase which leads to suppression of gluconeogenesis. In addition, metformin reduces autophagy failure observed in pancreatic β-cells under diabetic conditions. Furthermore, it is known that metformin alters the gut microbiome and facilitates the transport of glucose from the circulation into excrement. It is also known that metformin reduces food intake and lowers body weight by increasing circulating levels of the peptide hormone growth/differentiation factor 15 (GDF15). Furthermore, much attention has been drawn to the fact that the frequency of various cancers is lower in subjects taking metformin. Metformin suppresses the mechanistic target of rapamycin (mTOR) by activating AMPK in pre-neoplastic cells, which leads to suppression of cell growth and an increase in apoptosis in pre-neoplastic cells. It has been shown recently that metformin consumption potentially influences the mortality in patients with type 2 diabetes mellitus and coronavirus infectious disease (COVID-19). Taken together, metformin is an old drug, but multifaceted mechanisms of action of metformin have been unraveled one after another in its long history.
Collapse
|
110
|
Rooney J, Ryan N, Liu J, Houtman R, van Beuningen R, Hsieh JH, Chang G, Chen S, Christopher Corton J. A Gene Expression Biomarker Identifies Chemical Modulators of Estrogen Receptor α in an MCF-7 Microarray Compendium. Chem Res Toxicol 2021; 34:313-329. [PMID: 33405908 PMCID: PMC10683854 DOI: 10.1021/acs.chemrestox.0c00243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Identification of chemicals that affect hormone-regulated systems will help to predict endocrine disruption. In our previous study, a 46 gene biomarker was found to be an accurate predictor of estrogen receptor (ER) α modulation in chemically treated MCF-7 cells. Here, potential ERα modulators were identified using the biomarker by screening a microarray compendium consisting of ∼1600 gene expression comparisons representing exposure to ∼1200 chemicals. A total of ∼170 chemicals were identified as potential ERα modulators. In the Connectivity Map 2.0 collection, 75 and 39 chemicals were predicted to activate or suppress ERα, and they included 12 and six known ERα agonists and antagonists/selective ERα modulators, respectively. Nineteen and eight of the total number were also identified as active in an ERα transactivation assay carried out in an MCF-7-derived cell line used to screen the Tox21 10K chemical library in agonist or antagonist modes, respectively. Chemicals predicted to modulate ERα in MCF-7 cells were examined further using global and targeted gene expression in wild-type and ERα-null cells, transactivation assays, and cell-free ERα coregulator interaction assays. Environmental chemicals classified as weak and very weak agonists were confirmed to activate ERα including apigenin, kaempferol, and oxybenzone. Novel activators included digoxin, nabumetone, ivermectin, and six progestins. Novel suppressors included emetine, mifepristone, niclosamide, and proscillaridin. Our strategy will be useful to identify environmentally relevant ERα modulators in future high-throughput transcriptomic screens.
Collapse
Affiliation(s)
- John Rooney
- Center for Computational Toxicology and Exposure, US-EPA, Research Triangle Park, NC 27711
- Present address: Integrated Lab Services, Research Triangle Park, NC
| | - Natalia Ryan
- Center for Computational Toxicology and Exposure, US-EPA, Research Triangle Park, NC 27711
- Present address: Bayer Crop Science, Research Triangle Park, NC
| | - Jie Liu
- Center for Computational Toxicology and Exposure, US-EPA, Research Triangle Park, NC 27711
| | - René Houtman
- PamGene International B.V., Den Bosch, The Netherlands
- Present address: Precision Medicine Lab, Oss, The Netherlands
| | | | - Jui-Hua Hsieh
- Kelly Government Solutions, Research Triangle Park, North Carolina
| | - Gregory Chang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte,California 91010
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte,California 91010
| | - J. Christopher Corton
- Center for Computational Toxicology and Exposure, US-EPA, Research Triangle Park, NC 27711
| |
Collapse
|
111
|
Amin S, Mallick AA, Edwards H, Cortina-Borja M, Laugharne M, Likeman M, O'Callaghan FJ. The metformin in tuberous sclerosis (MiTS) study: A randomised double-blind placebo-controlled trial. EClinicalMedicine 2021; 32:100715. [PMID: 33681737 PMCID: PMC7910694 DOI: 10.1016/j.eclinm.2020.100715] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tuberous Sclerosis Complex (TSC) is a genetic disorder characterised by the development of benign tumours secondary to loss of inhibitory regulation of the mTOR (mechanistic Target of Rapamycin) intracellular growth pathway. Metformin inhibits the mTOR pathway. We investigated whether metformin would reduce growth of hamartomas associated with tuberous sclerosis complex. METHODS In this multicentre randomized, double-blind, placebo-controlled trial, patients with a clinical diagnosis of tuberous sclerosis, aged over 10 years and with at least one renal angiomyolipoma of greater than 1 cm in diameter were enrolled. Participants were randomly allocated (1:1) by a secure website to receive metformin or placebo for 12 months. The primary outcome was percentage volume change of renal angiomyolipomas (AML) at 12 months compared to baseline. Secondary outcomes were percentage change at 12 months from baseline in volume of cerebral Subependymal Giant Cell Astrocytomas (SEGA); appearance of facial and ungual hamartomas; frequency of epileptic seizures; and adaptive behaviour. The trial is registered with The International Standard Randomised Controlled Trial Number (ISRCTN), number 92545532, and the European Union Drug Regulating Authorities Clinical Trials (EUDRACT), number 2011-001319-30. FINDINGS Between 1 November 2012 and 30 September 2015 72 patients were screened and 55 were randomly assigned to metformin (28) or placebo (27). Four participants withdrew between randomisation and starting treatment. All 51 patients who started therapy completed the trial and were assessed for outcome at 12 months. The median percentage change in angiomyolipoma (AML) volume was +7.6% (IQR -1.8% to +42.6%) for the placebo group and +8.9% (IQR 1.3% to 19.5%) for the metformin group (p = 0.28). Twenty-seven patients had SEGAs: 13 received placebo and 14 metformin. The median percentage change in SEGA volume was +3.0% (IQR -22.8% to +27.7%) for the placebo group and - 20.8% (IQR - 47.1% to - 5.0%) for the metformin group (p = 0.03). Twenty-one patients were assessed for seizure frequency: 9 received placebo and 12 received metformin. In the metformin group, a mean reduction of 43.7% from baseline in seizures was observed and in the placebo group a 3.1% mean reduction was observed, with a difference in response of 40.6% (95% CI -3.1% to +84.2%, p = 0.03). There were no significant differences between metformin and placebo groups for the other secondary outcomes. There were no deaths. Three serious adverse events (SAEs) occurred during the trial (all patients on metformin). INTERPRETATION Metformin did not reduce AML volume. Metformin did reduce SEGA volume and seizure frequency compared with placebo. There may be a role for metformin in slowing or reversing growth of some life-threatening hamartomas in TSC and for reducing seizure frequency. Further study is justified. FUNDING This study was funded by the National Institute for Health and Research (NIHR) through the The Research for Patient Benefit Programme (RfPB).
Collapse
Affiliation(s)
- Sam Amin
- Clinical Neurosciences Section, Room 41, 4th Floor PUW South, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
- Children's Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath BA1 3NG, United Kingdom
- Department of Paediatric Neurology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol BS3 8AE, United Kingdom
| | - Andrew A Mallick
- Department of Paediatric Neurology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol BS3 8AE, United Kingdom
| | - Hannah Edwards
- Population Health Sciences, Bristol Medical School, University of Bristol, United Kingdom
| | - Mario Cortina-Borja
- Population, Policy and Practice Teaching and Research Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Matthew Laugharne
- Radiology Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath BA1 3NG, United Kingdom
| | - Marcus Likeman
- Department of Paediatric Radiology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol BS3 8AE, United Kingdom
| | - Finbar J.K. O'Callaghan
- Clinical Neurosciences Section, Room 41, 4th Floor PUW South, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
- Department of Paediatric Neurology, Brain Directorate, Great Ormond Street Hospital NHS Trust, London WC1N 3JH, United Kingdom
- Children's Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath BA1 3NG, United Kingdom
- Corresponding author at: Clinical Neurosciences Section, Room 41, 4th Floor PUW South, Institute of Child Health, University College London, London WC1N 1EH, United Kingdom. f.o'
| |
Collapse
|
112
|
Pimentel I, Chen BE, Lohmann AE, Ennis M, Ligibel J, Shepherd L, Hershman DL, Whelan T, Stambolic V, Mayer I, Hobday T, Lemieux J, Thompson A, Rastogi P, Gelmon K, Rea D, Rabaglio M, Ellard S, Mates M, Bedard P, Pitre L, Vandenberg T, Dowling RJO, Parulekar W, Goodwin PJ. The Effect of Metformin vs Placebo on Sex Hormones in Canadian Cancer Trials Group MA.32. J Natl Cancer Inst 2021; 113:192-198. [PMID: 33527137 PMCID: PMC7850529 DOI: 10.1093/jnci/djaa082] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/08/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metformin has been associated with lower breast cancer (BC) risk and improved outcomes in observational studies. Multiple biologic mechanisms have been proposed, including a recent report of altered sex hormones. We evaluated the effect of metformin on sex hormones in MA.32, a phase III trial of nondiabetic BC subjects who were randomly assigned to metformin or placebo. METHODS We studied the subgroup of postmenopausal hormone receptor-negative BC subjects not receiving endocrine treatment who provided fasting blood at baseline and at 6 months after being randomly assigned. Sex hormone-binding globulin, bioavailable testosterone, and estradiol levels were assayed using electrochemiluminescence immunoassay. Change from baseline to 6 months between study arms was compared using Wilcoxon sum rank tests and regression models. RESULTS 312 women were eligible (141 metformin vs 171 placebo); the majority of subjects in each arm had T1/2, N0, HER2-negative BC and had received (neo)adjuvant chemotherapy. Mean age was 58.1 (SD=6.9) vs 57.5 (SD=7.9) years, mean body mass index (BMI) was 27.3 (SD=5.5) vs 28.9 (SD=6.4) kg/m2 for metformin vs placebo, respectively. Median estradiol decreased between baseline and 6 months on metformin vs placebo (-5.7 vs 0 pmol/L; P < .001) in univariable analysis and after controlling for baseline BMI and BMI change (P < .001). There was no change in sex hormone-binding globulin or bioavailable testosterone. CONCLUSION Metformin lowered estradiol levels, independent of BMI. This observation suggests a new metformin effect that has potential relevance to estrogen sensitive cancers.
Collapse
Affiliation(s)
- Isabel Pimentel
- Vall d`Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bingshu E Chen
- Canadian Cancer Trials Group, Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | | | | | | | - Lois Shepherd
- Canadian Cancer Trials Group, Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | - Dawn L Hershman
- Herbert Irving Cancer Center, Columbia University, New York, NY, USA
| | - Timothy Whelan
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | - Vuk Stambolic
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - Ingrid Mayer
- Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | - Julie Lemieux
- CHA-Hopital Du St-Sacrement, Hopital Enfant Jesus Site, Quebec City, Canada
| | | | - Priya Rastogi
- National Surgical Adjuvant Breast and Bowel Project, Pittsburgh, PA, USA
| | - Karen Gelmon
- BCCA–Vancouver Cancer Centre, Vancouver, BC, Canada
| | - Daniel Rea
- Institute of Cancer Research, Clinical Trials and Statistics Unit, Sutton, UK
| | | | - Susan Ellard
- BCCA-Cancer Centre for the Southern Interior, Kelowna, BC, Canada
| | - Mihaela Mates
- Cancer Centre of Southeastern Ontario, Kingston, ON, Canada
| | - Philippe Bedard
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | | | | | - Ryan J O Dowling
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - Wendy Parulekar
- Canadian Cancer Trials Group , Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | - Pamela J Goodwin
- Lunenfeld Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
113
|
Levy CF, Presswala LS, Slomovic A, Stiefel J, Schulman-Rosenbaum R. Multidisciplinary management of endocrinopathies and treatment-related toxicities in patients with Bloom syndrome and cancer. Pediatr Blood Cancer 2021; 68:e28815. [PMID: 33226170 PMCID: PMC9171660 DOI: 10.1002/pbc.28815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 11/07/2022]
Abstract
The treatment of malignancy in cancer predisposition syndromes that also confer exquisite sensitivity to standard chemotherapy and radiation regimens remains a challenge. Bloom syndrome is one such disorder that is caused by a defect in DNA repair, predisposing to the development of early-onset age-related medical conditions and malignancies. We report on two patients with Bloom syndrome who responded well to chemotherapy despite significant alterations to standard protocols necessitated by hypersensitivity. Both patients experienced severe toxicities and exacerbation of endocrine comorbidities during chemotherapy. A multidisciplinary team of oncologists and endocrinologists is best suited to care for this patient population.
Collapse
Affiliation(s)
| | | | - Alana Slomovic
- Department of Pediatrics, Cohen Children’s Medical Center
| | - Jessica Stiefel
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center
| | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Department of Medicine, Long Island Jewish Medical Center, Associate Professor, Zucker School of Medicine at Hofstra/Northwell
| |
Collapse
|
114
|
Zhang K, Bai P, Dai H, Deng Z. Metformin and risk of cancer among patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Prim Care Diabetes 2021; 15:52-58. [PMID: 32605879 DOI: 10.1016/j.pcd.2020.06.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
AIM We carried out this meta-analysis on all published studies to estimate the overall cancer risk of the use of metformin in T2DM patients. METHODS We searched the PubMed, Embase and CNKI databases for all articles within a range of published years from 2007 to 2019 on the association between the use of metformin and cancer risk in T2DM patients. The odds ratio (OR) corresponding to the 95% confidence interval (95% CI) was used to assess the association using a random-effect meta-analysis. RESULTS Finally, 67 studies met the inclusion criteria for this study, with 10,695,875 T2DM patients and 145,108 cancer cases. Overall, For T2DM patients of ever vs. never metformin users, there was statistical evidence of significantly decreased cancer risk was found to be associated with ever metformin users (OR=0.70, 95% CI=0.65-0.76). Considering T2DM may be a specific and independent risk factor for various forms of cancer, due to its particular metabolic characteristics of glucose intolerance and hyperinsulinemia, we performed a comparison to estimate the effects of metformin on cancer risk with other anti-diabetes medications (ADMs), our results found significantly decreased cancer risk to be associated with the use of metformin (OR=0.80, 95% CI=0.73-0.87). CONCLUSION Our meta-analysis indicated that metformin may be a independent protective factor for cancer risk in T2DM patients.
Collapse
Affiliation(s)
- Kui Zhang
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Peng Bai
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Hao Dai
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhenhua Deng
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.
| |
Collapse
|
115
|
Targeting Cancer Metabolism and Current Anti-Cancer Drugs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:15-48. [PMID: 33725343 DOI: 10.1007/978-3-030-55035-6_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Several studies have exploited the metabolic hallmarks that distinguish between normal and cancer cells, aiming at identifying specific targets of anti-cancer drugs. It has become apparent that metabolic flexibility allows cancer cells to survive during high anabolic demand or the depletion of nutrients and oxygen. Cancers can reprogram their metabolism to the microenvironments by increasing aerobic glycolysis to maximize ATP production, increasing glutaminolysis and anabolic pathways to support bioenergetic and biosynthetic demand during rapid proliferation. The increased key regulatory enzymes that support the relevant pathways allow us to design small molecules which can specifically block activities of these enzymes, preventing growth and metastasis of tumors. In this review, we discuss metabolic adaptation in cancers and highlight the crucial metabolic enzymes involved, specifically those involved in aerobic glycolysis, glutaminolysis, de novo fatty acid synthesis, and bioenergetic pathways. Furthermore, we also review the success and the pitfalls of the current anti-cancer drugs which have been applied in pre-clinical and clinical studies.
Collapse
|
116
|
Mestareehi A, Zhang X, Seyoum B, Msallaty Z, Mallisho A, Burghardt KJ, Kowluru A, Yi Z. Metformin Increases Protein Phosphatase 2A Activity in Primary Human Skeletal Muscle Cells Derived from Lean Healthy Participants. J Diabetes Res 2021; 2021:9979234. [PMID: 34368369 PMCID: PMC8342103 DOI: 10.1155/2021/9979234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate if PP2A plays a role in metformin-induced insulin sensitivity improvement in human skeletal muscle cells. Participants. Eight lean insulin-sensitive nondiabetic participants (4 females and 4 males; age: 21.0 ± 1.0 years; BMI: 22.0 ± 0.7 kg/m2; 2-hour OGTT: 97.0 ± 6.0 mg/dl; HbA1c: 5.3 ± 0.1%; fasting plasma glucose: 87.0 ± 2.0 mg/dl; M value; 11.0 ± 1.0 mg/kgBW/min). DESIGN A hyperinsulinemic-euglycemic clamp was performed to assess insulin sensitivity in human subjects, and skeletal muscle biopsy samples were obtained. Primary human skeletal muscle cells (shown to retain metabolic characteristics of donors) were cultured from these muscle biopsies that included 8 lean insulin-sensitive participants. Cultured cells were expanded, differentiated into myotubes, and treated with 50 μM metformin for 24 hours before harvesting. PP2Ac activity was measured by a phosphatase activity assay kit (Millipore) according to the manufacturer's protocol. RESULTS The results indicated that metformin significantly increased the activity of PP2A in the myotubes for all 8 lean insulin-sensitive nondiabetic participants, and the average fold increase is 1.54 ± 0.11 (P < 0.001). CONCLUSIONS These results provided the first evidence that metformin can activate PP2A in human skeletal muscle cells derived from lean healthy insulin-sensitive participants and may help to understand metformin's action in skeletal muscle in humans.
Collapse
Affiliation(s)
- Aktham Mestareehi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Berhane Seyoum
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Zaher Msallaty
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Abdullah Mallisho
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Kyle Jon Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, USA
| | - Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Program for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
117
|
Choi M, Han J, Yang BR, Jang MJ, Kim M, Lee DW, Kim TY, Im SA, Lee HB, Moon HG, Han W, Noh DY, Lee KH. Association of Insulin, Metformin, and Statin with Mortality in Breast Cancer Patients. Cancer Res Treat 2021; 53:65-76. [PMID: 32972040 PMCID: PMC7812023 DOI: 10.4143/crt.2020.430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/22/2020] [Indexed: 12/29/2022] Open
Abstract
PURPOSE This study investigated the association of insulin, metformin, and statin use with survival and whether the association was modified by the hormone receptor status of the tumor in patients with breast cancer. MATERIALS AND METHODS We studied 7,452 patients who had undergone surgery for breast cancer at Seoul National University Hospital from 2008 to 2015 using the nationwide claims database. Exposure was defined as a recorded prescription of each drug within 12 months before the diagnosis of breast cancer. RESULTS Patients with prior insulin or statin use were more likely to be older than 50 years at diagnosis and had a higher comorbidity index than those without it (p < 0.01 for both). The hazard ratio (HR) for death with insulin use was 5.7 (p < 0.01), and the effect was attenuated with both insulin and metformin exposure with an HR of 1.2 (p=0.60). In the subgroup analyses, a heightened risk of death with insulin was further prominent with an HR of 17.9 (p < 0.01) and was offset by co-administration of metformin with an HR of 1.3 (p=0.67) in patients with estrogen receptor (ER)-negative breast cancer. Statin use was associated with increased overall mortality only in patients with ER-positive breast cancer with HR for death of 1.5 (p=0.05). CONCLUSION Insulin or statin use before the diagnosis of breast cancer was associated with an increase in all-cause mortality. Subsequent analyses suggested that metformin or statin use may have been protective in patients with ER-negative disease, which warrants further studies.
Collapse
Affiliation(s)
- Mihong Choi
- Department of Internal Medicine, The Catholic University of Korea Incheon St. Mary’s Hospital, Incheon,
Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Jiyeon Han
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul,
Korea
| | - Bo Ram Yang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul,
Korea
| | - Myoung-jin Jang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul,
Korea
| | - Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
- Cancer Research Institute, Seoul National University, Seoul,
Korea
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
| | - Tae-Yong Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
- Cancer Research Institute, Seoul National University, Seoul,
Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
- Cancer Research Institute, Seoul National University, Seoul,
Korea
| | - Han-Byoel Lee
- Department of Surgery, Seoul National University Hospital, Seoul,
Korea
| | - Hyeong-Gon Moon
- Department of Surgery, Seoul National University Hospital, Seoul,
Korea
| | - Wonshik Han
- Department of Surgery, Seoul National University Hospital, Seoul,
Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University Hospital, Seoul,
Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul,
Korea
- Cancer Research Institute, Seoul National University, Seoul,
Korea
| |
Collapse
|
118
|
BRAF Controls the Effects of Metformin on Neuroblast Cell Divisions in C. elegans. Int J Mol Sci 2020; 22:ijms22010178. [PMID: 33375360 PMCID: PMC7795703 DOI: 10.3390/ijms22010178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Metformin has demonstrated substantial potential for use in cancer treatments. Liver kinase B (LKB)-AMP-activated protein kinase (AMPK) and mTOR are reported to be the main targets of metformin in relation to its ability to prevent cancer cell proliferation. However, the role of metformin in the control of neoplastic cancer cell growth is possibly independent of LKB-AMPK and mTOR. Using C. elegans as a model, we found that the neuronal Q-cell divisions in L1-arrested worms were suppressed following metformin treatment in AMPK-deficient mutants, suggesting that the mechanism by which metformin suppresses these cell divisions is independent of AMPK. Our results showed that the mTOR pathway indeed played a role in controlling germ cell proliferation, but it was not involved in the neuronal Q-cell divisions occurring in L1-arrested worms. We found that the neuronal Q-cells divisions were held at G1/S cell stage by metformin in vivo. Additionally, we demonstrated that metformin could reduce the phosphorylation activity of BRAF and block the BRAF-MAPK oncogenesis pathway to regulate neuronal Q-cell divisions during L1 arrest. This work discloses a new mechanism by which metformin treatment acts to promote neuronal cancer prevention, and these results will help promote the study of the anticancer mechanisms underlying metformin treatments.
Collapse
|
119
|
Cairns J, Ly RC, Niu N, Kalari KR, Carlson EE, Wang L. CDC25B partners with PP2A to induce AMPK activation and tumor suppression in triple negative breast cancer. NAR Cancer 2020; 2:zcaa039. [PMID: 33385163 PMCID: PMC7751685 DOI: 10.1093/narcan/zcaa039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/28/2022] Open
Abstract
Cell division cycle 25 (CDC25) dual specificity phosphatases positively regulate the cell cycle by activating cyclin-dependent kinase/cyclin complexes. Here, we demonstrate that in addition to its role in cell cycle regulation, CDC25B functions as a regulator of protein phosphatase 2A (PP2A), a major cellular Ser/Thr phosphatase, through its direct interaction with PP2A catalytic subunit. Importantly, CDC25B alters the regulation of AMP-activated protein kinase signaling (AMPK) by PP2A, increasing AMPK activity by inhibiting PP2A to dephosphorylate AMPK. CDC25B depletion leads to metformin resistance by inhibiting metformin-induced AMPK activation. Furthermore, dual inhibition of CDC25B and PP2A further inhibits growth of 3D organoids isolated from patient derived xenograft model of breast cancer compared to CDC25B inhibition alone. Our study identifies CDC25B as a regulator of PP2A, and uncovers a mechanism of controlling the activity of a key energy metabolism marker, AMPK.
Collapse
Affiliation(s)
- Junmei Cairns
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Reynold C Ly
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nifang Niu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Krishna R Kalari
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Erin E Carlson
- Division of Biostatistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- To whom correspondence should be addressed. Tel: +1 507 284 5264; Fax: +1 507 284 4455;
| |
Collapse
|
120
|
Ribosome 18S m 6A Methyltransferase METTL5 Promotes Translation Initiation and Breast Cancer Cell Growth. Cell Rep 2020; 33:108544. [PMID: 33357433 DOI: 10.1016/j.celrep.2020.108544] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/03/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
N6 methylation at adenosine 1832 (m6A1832) of mammalian 18S rRNA, occupying a critical position within the decoding center, is modified by a conserved methyltransferase, METTL5. Here, we find that METTL5 shows strong substrate preference toward the 18S A1832 motif but not the other reported m6A motifs. Comparison with a yeast ribosome structural model unmodified at this site indicates that the modification may facilitate mRNA binding by inducing conformation changes in the mammalian ribosomal decoding center. METTL5 promotes p70-S6K activation and proper translation initiation, and the loss of METTL5 significantly reduces the abundance of polysome. METTL5 expression is elevated in breast cancer patient samples and is required for growth of several breast cancer cell lines. We further find that Caenorhabditis elegans lacking the homolog metl-5 develop phenotypes known to be associated with impaired translation. Altogether, our findings uncover critical and conserved roles of METTL5 in the regulation of translation.
Collapse
|
121
|
Baeza-Flores GDC, Guzmán-Priego CG, Parra-Flores LI, Murbartián J, Torres-López JE, Granados-Soto V. Metformin: A Prospective Alternative for the Treatment of Chronic Pain. Front Pharmacol 2020; 11:558474. [PMID: 33178015 PMCID: PMC7538784 DOI: 10.3389/fphar.2020.558474] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Metformin (biguanide) is a drug widely used for the treatment of type 2 diabetes. This drug has been used for 60 years as a highly effective antihyperglycemic agent. The search for the mechanism of action of metformin has produced an enormous amount of research to explain its effects on gluconeogenesis, protein metabolism, fatty acid oxidation, oxidative stress, glucose uptake, autophagy and pain, among others. It was only up the end of the 1990s and beginning of this century that some of its mechanisms were revealed. Metformin induces its beneficial effects in diabetes through the activation of a master switch kinase named AMP-activated protein kinase (AMPK). Two upstream kinases account for the physiological activation of AMPK: liver kinase B1 and calcium/calmodulin-dependent protein kinase kinase 2. Once activated, AMPK inhibits the mechanistic target of rapamycin complex 1 (mTORC1), which in turn avoids the phosphorylation of p70 ribosomal protein S6 kinase 1 and phosphatidylinositol 3-kinase/protein kinase B signaling pathways and reduces cap-dependent translation initiation. Since metformin is a disease-modifying drug in type 2 diabetes, which reduces the mTORC1 signaling to induce its effects on neuronal plasticity, it was proposed that these mechanisms could also explain the antinociceptive effect of this drug in several models of chronic pain. These studies have highlighted the efficacy of this drug in chronic pain, such as that from neuropathy, insulin resistance, diabetic neuropathy, and fibromyalgia-type pain. Mounting evidence indicates that chronic pain may induce anxiety, depression and cognitive impairment in rodents and humans. Interestingly, metformin is able to reverse some of these consequences of pathological pain in rodents. The purpose of this review was to analyze the current evidence about the effects of metformin in chronic pain and three of its comorbidities (anxiety, depression and cognitive impairment).
Collapse
Affiliation(s)
- Guadalupe Del Carmen Baeza-Flores
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Crystell Guadalupe Guzmán-Priego
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Leonor Ivonne Parra-Flores
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Jorge Elías Torres-López
- Laboratorio de Mecanismos de Dolor, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico.,Departamento de Anestesiología, Hospital Regional de Alta Especialidad "Dr. Juan Graham Casasús", Villahermosa, Mexico
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| |
Collapse
|
122
|
Ahn HK, Lee YH, Koo KC. Current Status and Application of Metformin for Prostate Cancer: A Comprehensive Review. Int J Mol Sci 2020; 21:ijms21228540. [PMID: 33198356 PMCID: PMC7698147 DOI: 10.3390/ijms21228540] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Metformin, an oral biguanide used for first-line treatment of type 2 diabetes mellitus, has attracted attention for its anti-proliferative and anti-cancer effects in several solid tumors, including prostate cancer (PCa). Liver kinase B1 (LKB1) and adenosine monophosphate-activated protein kinase (AMPK) activation, inhibition of the mammalian target of rapamycin (mTOR) activity and protein synthesis, induction of apoptosis and autophagy by p53 and p21, and decreased blood insulin level have been suggested as direct anti-cancer mechanisms of metformin. Research has shown that PCa development and progression are associated with metabolic syndrome and its components. Therefore, reduction in the risk of PCa and improvement in survival in metformin users may be the results of the direct anti-cancer mechanisms of the drug or the secondary effects from improvement of metabolic syndrome. In contrast, some research has suggested that there is no association between metformin use and PCa incidence or survival. In this comprehensive review, we summarize updated evidence on the relationship between metformin use and oncological effects in patients with PCa. We also highlight ongoing clinical trials evaluating metformin as an adjuvant therapy in novel drug combinations in various disease settings.
Collapse
|
123
|
Peralta S, Pinto M, Arguello T, Garcia S, Diaz F, Moraes CT. Metformin delays neurological symptom onset in a mouse model of neuronal complex I deficiency. JCI Insight 2020; 5:141183. [PMID: 33148885 PMCID: PMC7710273 DOI: 10.1172/jci.insight.141183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
Complex I (also known as NADH-ubiquinone oxidoreductase) deficiency is the most frequent mitochondrial disorder present in childhood. NADH-ubiquinone oxidoreductase iron-sulfur protein 3 (NDUFS3) is a catalytic subunit of the mitochondrial complex I; NDUFS3 is conserved from bacteria and essential for complex I function. Mutations affecting complex I, including in the Ndufs3 gene, cause fatal neurodegenerative diseases, such as Leigh syndrome. No treatment is available for these conditions. We developed and performed a detailed molecular characterization of a neuron-specific Ndufs3 conditional KO mouse model. We showed that deletion of Ndufs3 in forebrain neurons reduced complex I activity, altered brain energy metabolism, and increased locomotor activity with impaired motor coordination, balance, and stereotyped behavior. Metabolomics analyses showed an increase of glycolysis intermediates, suggesting an adaptive response to the complex I defect. Administration of metformin to these mice delayed the onset of the neurological symptoms but not of neuronal loss. This improvement was likely related to enhancement of glucose uptake and utilization, which are known effects of metformin in the brain. Despite reports that metformin inhibits complex I activity, our findings did not show worsening a complex I defect nor increases in lactic acid, suggesting that metformin should be further evaluated for use in patients with mitochondrial encephalopathies. Metformin delays onset of mitochondrial encephalopathy in a CNS model of mitochondrial oxidative phosphorylation defect.
Collapse
Affiliation(s)
| | | | | | | | | | - Carlos T Moraes
- Department of Neurology and.,Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
124
|
Kim K, Yang WH, Jung YS, Cha JH. A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity. BMB Rep 2020. [PMID: 32731915 PMCID: PMC7607149 DOI: 10.5483/bmbrep.2020.53.10.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
T-cell-based cancer immunotherapies, such as immune checkpoint blockers (ICBs) and chimeric antigen receptor (CAR)-T-cells, have significant anti-tumor effects against certain types of cancer, providing a new paradigm for cancer treatment. However, the activity of tumor infiltrating T-cells (TILs) can be effectively neutralized in the tumor microenvironment (TME) of most solid tumors, rich in various immunosuppressive factors and cells. Therefore, to improve the clinical outcomes of established T-cell-based immunotherapy, adjuvants that can comprehensively relieve multiple immunosuppressive mechanisms of TME are needed. In this regard, recent studies have revealed that metformin has several beneficial effects on anti-tumor immunity. In this mini-review, we understand the immunosuppressive properties of TME and how metformin comprehensively enhances anti-tumor immunity. Finally, we will discuss this old friend’s potential as an adjuvant for cancer immunotherapy.
Collapse
Affiliation(s)
- KyeongJin Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Korea
| | - Wen-Hao Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Jong-ho Cha
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Korea
| |
Collapse
|
125
|
Cancer Metabolism: Phenotype, Signaling and Therapeutic Targets. Cells 2020; 9:cells9102308. [PMID: 33081387 PMCID: PMC7602974 DOI: 10.3390/cells9102308] [Citation(s) in RCA: 283] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant metabolism is a major hallmark of cancer. Abnormal cancer metabolism, such as aerobic glycolysis and increased anabolic pathways, has important roles in tumorigenesis, metastasis, drug resistance, and cancer stem cells. Well-known oncogenic signaling pathways, such as phosphoinositide 3-kinase (PI3K)/AKT, Myc, and Hippo pathway, mediate metabolic gene expression and increase metabolic enzyme activities. Vice versa, deregulated metabolic pathways contribute to defects in cellular signal transduction pathways, which in turn provide energy, building blocks, and redox potentials for unrestrained cancer cell proliferation. Studies and clinical trials are being performed that focus on the inhibition of metabolic enzymes by small molecules or dietary interventions (e.g., fasting, calorie restriction, and intermittent fasting). Similar to genetic heterogeneity, the metabolic phenotypes of cancers are highly heterogeneous. This heterogeneity results from diverse cues in the tumor microenvironment and genetic mutations. Hence, overcoming metabolic plasticity is an important goal of modern cancer therapeutics. This review highlights recent findings on the metabolic phenotypes of cancer and elucidates the interactions between signal transduction pathways and metabolic pathways. We also provide novel rationales for designing the next-generation cancer metabolism drugs.
Collapse
|
126
|
Ozdemir Kutbay N, Biray Avci C, Sarer Yurekli B, Caliskan Kurt C, Shademan B, Gunduz C, Erdogan M. Effects of metformin and pioglitazone combination on apoptosis and AMPK/mTOR signaling pathway in human anaplastic thyroid cancer cells. J Biochem Mol Toxicol 2020; 34:e22547. [PMID: 32589349 DOI: 10.1002/jbt.22547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/28/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
Anaplastic cancer constitutes 1% of thyroid cancers, and it is one of the most aggressive cancers. Treatment options are external radiation therapy and/or chemotherapy. The success rate with these treatment modalities is not satisfactory. We aimed to evaluate the effects of metformin (MET) and pioglitazone (PIO) combination on apoptosis and AMP-activated protein kinase/mammalian target of rapamycin (mTOR) signaling pathway in human anaplastic thyroid cancer cells. In this study, we evaluated the effects of MET and PIO individually and the combination of the two drugs on the cellular lines SW1736 and C643 ATC. Genes contained in the mTOR signaling pathway were examined using human mTOR Signalization RT2 Profiler PCR Array. In C643 and SW1736 cell lines, IC50 doses of MET and PIO were found out as 17.69 mM, 11.64 mM, 27.12 µM, and 23.17 µM. Also, the combination of MET and PIO was determined as an additive according to isobologram analyses. We have found the downregulation of the expression levels of oncogenic genes: AKT3, CHUK, CDC42, EIF4E, HIF1A, IKBKB, ILK, MTOR, PIK3CA, PIK3CG, PLD1, PRKCA, and RICTOR genes, in the MET and PIO combination-treated cells. In addition, expression levels of tumor suppressor genes, DDIT4, DDIT4L, EIF4EBP1, EIF4EBP2, FKBP1A, FKBP8, GSK3B, MYO1C, PTEN, ULK1, and ULK2, were found to have increased significantly. The MET + PIO combination was first applied to thyroid cancer cells, and significant reductions in the level of oncogenic genes were detected. The decreases, particularly, in AKT3, DEPTOR, EIF4E, ILK, MTOR, PIK3C, and PRKCA expressions indicate that progression can be prevented in thyroid cancer cells and these genes could be selected as therapeutic targets.
Collapse
Affiliation(s)
- Nilufer Ozdemir Kutbay
- Department of Endocrinology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Banu Sarer Yurekli
- Department of Endocrinology, Ege University Medical School, Izmir, Turkey
| | | | - Behrouz Shademan
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Ege University Medical School, Izmir, Turkey
| | - Mehmet Erdogan
- Department of Endocrinology, Ege University Medical School, Izmir, Turkey
| |
Collapse
|
127
|
Xue L, Chen F, Yue F, Camacho L, Kothapalli S, Wei G, Huang S, Mo Q, Ma F, Li Y, Jiralerspong S. Metformin and an insulin/IGF-1 receptor inhibitor are synergistic in blocking growth of triple-negative breast cancer. Breast Cancer Res Treat 2020; 185:73-84. [PMID: 32940848 DOI: 10.1007/s10549-020-05927-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with poor survival outcomes. Metformin has been shown to have antitumor effects by lowering serum levels of the mitogen insulin and having pleiotropic effects on cancer cell signaling pathways. BMS-754807 is a potent and reversible inhibitor of both insulin-like growth factor 1 receptor (IGF-1R) and insulin receptor (IR). Both drugs have been reported to have some efficacy in TNBC. However, it is unclear whether the combination of the two drugs is more effective than single drug treatment in TNBC. METHODS We treated a panel of TNBC cell lines with metformin and BMS-754807 alone and in combination and tested cell viability using MTS assays. We used the CompuSyn software to analyze for additivity, synergism, or antagonism. We also examined the molecular mechanism by performing reverse phase protein assay (RPPA) to detect the candidate pathways altered by single drugs and the drug combination and used Western blotting to verify and expand the findings. RESULTS The combination of metformin and BMS-754807 showed synergy in 11 out of 13 TNBC cell lines tested (85%). RPPA analysis detected significant alterations by the drug combination of multiple proteins known to regulate cell cycle and tumor growth. In particular, the drug combination significantly increased levels of total and phosphorylated forms of the cell cycle inhibitor p27Kip1 and decreased the level of the p27Kip1 E3 ligase SCFSkp2. CONCLUSIONS We conclude that the combination of metformin and BMS-754807 is more effective than either drug alone in inhibiting cell proliferation in the majority of TNBC cell lines, and that one important mechanism may be suppression of SCFSkp2 and subsequent stabilization of the cell cycle inhibitor p27Kip1. This combination treatment may represent an effective targeted therapy for a significant subset of TNBC cases and should be further evaluated.
Collapse
Affiliation(s)
- Lei Xue
- Laboratory for Comparative Genomics and Bioinformatics, College of Life Science, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210046, Jiangsu, China.,Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Fengju Chen
- Dan L Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Fei Yue
- Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Laura Camacho
- Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Sushma Kothapalli
- Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Guanyun Wei
- Laboratory for Comparative Genomics and Bioinformatics, College of Life Science, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210046, Jiangsu, China
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.,Dan L Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Qianxing Mo
- Dan L Duncan Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics, College of Life Science, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210046, Jiangsu, China
| | - Yi Li
- Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - Sao Jiralerspong
- Lester & Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA. .,Section of Breast Medical Oncology, Division of Hematology and Oncology, University of Arizona Cancer Center, 1515 N. Campbell Ave, Tucson, AZ, 85724, USA.
| |
Collapse
|
128
|
Van Nostrand JL, Hellberg K, Luo EC, Van Nostrand EL, Dayn A, Yu J, Shokhirev MN, Dayn Y, Yeo GW, Shaw RJ. AMPK regulation of Raptor and TSC2 mediate metformin effects on transcriptional control of anabolism and inflammation. Genes Dev 2020; 34:1330-1344. [PMID: 32912901 PMCID: PMC7528705 DOI: 10.1101/gad.339895.120] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
Abstract
Here, Van Nostrand et al. investigated the mechanisms of action of the biguanide drug metformin by using a new RaptorAA mouse model, in which AMPK phospho-serine sites Ser722 and Ser792 of RAPTOR were mutated to alanine. The hepatic transcriptional response in mice on a high-fat diet treated with metformin was largely ablated by AMPK deficiency under the conditions examined, indicating the essential role of this kinase and its targets in metformin action in vivo. Despite being the frontline therapy for type 2 diabetes, the mechanisms of action of the biguanide drug metformin are still being discovered. In particular, the detailed molecular interplays between the AMPK and the mTORC1 pathway in the hepatic benefits of metformin are still ill defined. Metformin-dependent activation of AMPK classically inhibits mTORC1 via TSC/RHEB, but several lines of evidence suggest additional mechanisms at play in metformin inhibition of mTORC1. Here we investigated the role of direct AMPK-mediated serine phosphorylation of RAPTOR in a new RaptorAA mouse model, in which AMPK phospho-serine sites Ser722 and Ser792 of RAPTOR were mutated to alanine. Metformin treatment of primary hepatocytes and intact murine liver requires AMPK regulation of both RAPTOR and TSC2 to fully inhibit mTORC1, and this regulation is critical for both the translational and transcriptional response to metformin. Transcriptionally, AMPK and mTORC1 were both important for regulation of anabolic metabolism and inflammatory programs triggered by metformin treatment. The hepatic transcriptional response in mice on high-fat diet treated with metformin was largely ablated by AMPK deficiency under the conditions examined, indicating the essential role of this kinase and its targets in metformin action in vivo.
Collapse
Affiliation(s)
- Jeanine L Van Nostrand
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Kristina Hellberg
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - En-Ching Luo
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92037, USA
| | - Eric L Van Nostrand
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92037, USA
| | - Alina Dayn
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Jingting Yu
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Yelena Dayn
- Transgenic Core Facility, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92037, USA
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
129
|
Hosio M, Urpilainen E, Hautakoski A, Marttila M, Arffman M, Sund R, Ahtikoski A, Puistola U, Karihtala P, Jukkola A, Läärä E. Survival after breast cancer in women with type 2 diabetes using antidiabetic medication and statins: a retrospective cohort study. Acta Oncol 2020; 59:1110-1117. [PMID: 32478629 DOI: 10.1080/0284186x.2020.1769858] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: We assessed survival of breast cancer in women with type 2 diabetes (T2D) treated with metformin, other types of antidiabetic medication (ADM) and statins.Materials and Methods: The study cohort consisted of women with T2D and diagnosed with breast cancer in Finland in 1998─2011. Mortality rates from breast cancer and other causes were analysed by Cox models, and adjusted hazard ratios (HRs) with 95% confidence intervals (Cls) were estimated in relation to the use of different types of medication.Results: The final cohort consisted of 3,533 women. No clear evidence was found for breast cancer mortality being different in metformin users (HR 0.86, 95% Cl 0.63-1.17), but their other-cause mortality appeared to be lower (HR 0.73, 95% Cl 0.55-0.97) in comparison with women using other types of oral ADM. Other-cause mortality was higher among insulin users (HR 1.45, 95% Cl 1.16-1.80) compared with users of other oral ADMs, other than metformin. Prediagnostic statin use was observed to be associated with decreased mortality from both breast cancer (HR 0.76, 95% Cl 0.63-0.92) and other causes (HR 0.75, 95% Cl 0.64-0.87).Conclusions: We did not find any association between ADM use and disease-specific mortality among women with T2D diagnosed with breast cancer. However, interestingly, prediagnostic statin use was observed to predict reduced mortality from breast cancer and other causes. We hypothesise that treating treatment practices of T2D or hypercholesterolaemia of breast cancer patients might affect overall prognosis of women diagnosed with breast cancer and T2D.
Collapse
Affiliation(s)
- Mayu Hosio
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Elina Urpilainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Ari Hautakoski
- Research Unit of Mathematical Sciences, University of Oulu, Oulu, Finland
| | | | - Martti Arffman
- Service System Research Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Reijo Sund
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anne Ahtikoski
- Cancer and Translational Medicine Research Unit, Department of Pathology, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Oncology, University of Helsinki and Helsinki University Comprehensive Cancer Center, Helsinki, Finland
| | - Arja Jukkola
- Department of Oncology and Radiotherapy, Cancer Centre Tampere, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Esa Läärä
- Research Unit of Mathematical Sciences, University of Oulu, Oulu, Finland
| |
Collapse
|
130
|
Endometrial Cancer as a Metabolic Disease with Dysregulated PI3K Signaling: Shedding Light on Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21176073. [PMID: 32842547 PMCID: PMC7504460 DOI: 10.3390/ijms21176073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies of the female reproductive organs. The most characteristic feature of EC is the frequent association with metabolic disorders. However, the components of these disorders that are involved in carcinogenesis remain unclear. Accumulating epidemiological studies have clearly revealed that hyperinsulinemia, which accompanies these disorders, plays central roles in the development of EC via the insulin-phosphoinositide 3 kinase (PI3K) signaling pathway as a metabolic driver. Recent comprehensive genomic analyses showed that over 90% of ECs have genomic alterations in this pathway, resulting in enhanced insulin signaling and production of optimal tumor microenvironments (TMEs). Targeting PI3K signaling is therefore an attractive treatment strategy. Several clinical trials for recurrent or advanced ECs have been attempted using PI3K-serine/threonine kinase (AKT) inhibitors. However, these agents exhibited far lower efficacy than expected, possibly due to activation of alternative pathways that compensate for the PIK3-AKT pathway and allow tumor growth, or due to adaptive mechanisms including the insulin feedback pathway that limits the efficacy of agents. Overcoming these responses with careful management of insulin levels is key to successful treatment. Further interest in specific TMEs via the insulin PI3K-pathway in obese women will provide insight into not only novel therapeutic strategies but also preventive strategies against EC.
Collapse
|
131
|
Wen KC, Sung PL, Wu ATH, Chou PC, Lin JH, Huang CYF, Yeung SCJ, Lee MH. Neoadjuvant metformin added to conventional chemotherapy synergizes anti-proliferative effects in ovarian cancer. J Ovarian Res 2020; 13:95. [PMID: 32825834 PMCID: PMC7442990 DOI: 10.1186/s13048-020-00703-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022] Open
Abstract
Background Ovarian cancer is the leading cause of cancer-related death among women. Complete cytoreductive surgery followed by platinum-taxene chemotherapy has been the gold standard for a long time. Various compounds have been assessed in an attempt to combine them with conventional chemotherapy to improve survival rates or even overcome chemoresistance. Many studies have shown that an antidiabetic drug, metformin, has cytotoxic activity in different cancer models. However, the synergism of metformin as a neoadjuvant formula plus chemotherapy in clinical trials and basic studies remains unclear for ovarian cancer. Methods We applied two clinical databases to survey metformin use and ovarian cancer survival rate. The Cancer Genome Atlas dataset, an L1000 microarray with Gene Set Enrichment Analysis (GSEA) analysis, Western blot analysis and an animal model were used to study the activity of the AKT/mTOR pathway in response to the synergistic effects of neoadjuvant metformin combined with chemotherapy. Results We found that ovarian cancer patients treated with metformin had significantly longer overall survival than patients treated without metformin. The protein profile induced by low- concentration metformin in ovarian cancer predominantly involved the AKT/mTOR pathway. In combination with chemotherapy, the neoadjuvant metformin protocol showed beneficial synergistic effects in vitro and in vivo. Conclusions This study shows that neoadjuvant metformin at clinically relevant dosages is efficacious in treating ovarian cancer, and the results can be used to guide clinical trials.
Collapse
Affiliation(s)
- Kuo-Chang Wen
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Pi-Lin Sung
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Department of Obstetrics and Gynecology, Huei-Sheng Clinic, New Taipei City, 23561, Taiwan
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ping-Chieh Chou
- Department of Molecular and Cellular Oncology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Jun-Hung Lin
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, 11221, Taiwan
| | - Sai-Ching J Yeung
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mong-Hong Lee
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, 26 Yuancun Erheng Rd, Guangzhou, 510655, P.R. China.
| |
Collapse
|
132
|
Chang WH, Lai AG. An integrative pan-cancer investigation reveals common genetic and transcriptional alterations of AMPK pathway genes as important predictors of clinical outcomes across major cancer types. BMC Cancer 2020; 20:773. [PMID: 32807122 PMCID: PMC7433212 DOI: 10.1186/s12885-020-07286-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The AMP-activated protein kinase (AMPK) is an evolutionarily conserved regulator of cellular energy homeostasis. As a nexus for transducing metabolic signals, AMPK cooperates with other energy-sensing pathways to modulate cellular responses to metabolic stressors. With metabolic reprogramming being a hallmark of cancer, the utility of agents targeting AMPK has received continued scrutiny and results have demonstrated conflicting effects of AMPK activation in tumorigenesis. Harnessing multi-omics datasets from human tumors, we seek to evaluate the seemingly pleiotropic, tissue-specific dependencies of AMPK signaling dysregulation. METHODS We interrogated copy number variation and differential transcript expression of 92 AMPK pathway genes across 21 diverse cancers involving over 18,000 patients. Cox proportional hazards regression and receiver operating characteristic analyses were used to evaluate the prognostic significance of AMPK dysregulation on patient outcomes. RESULTS A total of 24 and seven AMPK pathway genes were identified as having loss- or gain-of-function features. These genes exhibited tissue-type dependencies, where survival outcomes in glioma patients were most influenced by AMPK inactivation. Cox regression and log-rank tests revealed that the 24-AMPK-gene set could successfully stratify patients into high- and low-risk groups in glioma, sarcoma, breast and stomach cancers. The 24-AMPK-gene set could not only discriminate tumor from non-tumor samples, as confirmed by multidimensional scaling analyses, but is also independent of tumor, node and metastasis staging. AMPK inactivation is accompanied by the activation of multiple oncogenic pathways associated with cell adhesion, calcium signaling and extracellular matrix organization. Anomalous AMPK signaling converged on similar groups of transcriptional targets where a common set of transcription factors were identified to regulate these targets. We also demonstrated crosstalk between pro-catabolic AMPK signaling and two pro-anabolic pathways, mammalian target of rapamycin and peroxisome proliferator-activated receptors, where they act synergistically to influence tumor progression significantly. CONCLUSION Genetic and transcriptional aberrations in AMPK signaling have tissue-dependent pro- or anti-tumor impacts. Pan-cancer investigations on molecular changes of this pathway could uncover novel therapeutic targets and support risk stratification of patients in prospective trials.
Collapse
Affiliation(s)
- Wai Hoong Chang
- Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK
| | - Alvina G Lai
- Institute of Health Informatics, University College London, 222 Euston Road, London, NW1 2DA, UK.
| |
Collapse
|
133
|
Zhu Y, Wang T, Wu J, Huang O, Zhu L, He J, Li Y, Chen W, Chen X, Shen K. Biomarkers of Insulin and the Insulin-Like Growth Factor Axis in Relation to Breast Cancer Risk in Chinese Women. Onco Targets Ther 2020; 13:8027-8036. [PMID: 32848423 PMCID: PMC7429223 DOI: 10.2147/ott.s258357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The interplay between biomarkers of insulin and the insulin-like growth factor (IGF) axis in the context of breast cancer risk is unclear. METHODS We measured the concentrations of insulin, C-peptide, IGF1, and IGF binding protein 3 (IGFBP3) and calculated the homeostasis model assessment of insulin resistance (HOMA-IR) index and the IGF1/IGFBP3 ratio among 2536 patients with breast cancer and 2528 patients with benign breast disease recruited from Ruijin Hospital, Shanghai, China, between 2012 and 2017. RESULTS Multivariable-adjusted odds ratios (ORs) for breast cancer associated with the highest quartiles versus the lowest quartiles of insulin and IGF factors were 1.45 (95% CI, 1.20-1.75) for insulin, 1.32 (1.08-1.60) for C-peptide, 1.53 (1.26-1.85) for HOMA-IR, and 1.27 (1.05-1.53) for IGF1; these associations did not differ substantially across stratifications of age, body mass index, age at menarche, or menopausal status (all P for interaction >0.05). In the joint analysis, the highest quartile of IGF1 was associated with the greatest risk of breast cancer in the highest quartiles of insulin (OR, 1.77; 95% CI, 1.29-2.44), C-peptide (1.60; 1.17-2.20), and HOMA-IR (1.90; 1.38-2.62), compared with the risks associated with the combination of the lowest quartiles of IGF1 and each insulin factor. In stratification analysis, the positive association between IGF1 and breast cancer was stronger in the highest quartiles of insulin (P[interaction] = 0.29), C-peptide (P[interaction] = 0.020), and HOMA-IR (P[interaction] = 0.075). CONCLUSION Our findings indicate effect modifications of insulin, C-peptide, and insulin resistance on the relationship between IGF1 and breast cancer risk in Chinese women.
Collapse
Affiliation(s)
- Yifei Zhu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Tiange Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jiayi Wu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Ou Huang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Li Zhu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jianrong He
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yafen Li
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Weiguo Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaosong Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
134
|
Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020; 12:247. [PMID: 32848716 PMCID: PMC7426063 DOI: 10.3389/fnagi.2020.00247] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
The adult central nervous system (CNS) contains resident stem cells within specific niches that maintain a self-renewal and proliferative capacity to generate new neurons, astrocytes, and oligodendrocytes throughout adulthood. Physiological aging is associated with a progressive loss of function and a decline in the self-renewal and regenerative capacities of CNS stem cells. Also, the biggest risk factor for neurodegenerative diseases is age, and current in vivo and in vitro models of neurodegenerative diseases rarely consider this. Therefore, combining both aging research and appropriate interrogation of animal disease models towards the understanding of the disease and age-related stem cell failure is imperative to the discovery of new therapies. This review article will highlight the main intrinsic and extrinsic regulators of neural stem cell (NSC) aging and discuss how these factors impact normal homeostatic functions within the adult brain. We will consider established in vivo animal and in vitro human disease model systems, and then discuss the current and future trajectories of novel senotherapeutics that target aging NSCs to ameliorate brain disease.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
135
|
Linkeviciute-Ulinskiene D, Patasius A, Kincius M, Zabuliene L, Smailyte G. Preexisting diabetes, metformin use and long-term survival in patients with prostate cancer. Scand J Urol 2020; 54:401-407. [PMID: 32748714 DOI: 10.1080/21681805.2020.1798502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To assess prostate cancer-specific and overall survival in prostate cancer patients with or without preexisting type 2 diabetes mellitus (T2DM) with regards to metformin use. METHODS Patients diagnosed with prostate cancer in the Lithuanian population between 2001 and 2005 were identified through the Lithuanian Cancer Registry and followed until 2016, date of death, loss to follow-up or whichever came first. Information regarding the diagnosis of T2DM and antihyperglycemic medications were obtained from the National Health Insurance Fund database. Prostate cancer-specific and overall survival outcomes were analysed using univariate and multivariate Cox proportional hazard models. RESULTS Out of 6689 men included, 254 (3.8%) had preexisting T2DM. There were 4807 deaths during follow-up, including 2084 from prostate cancer. No differences were found in prostate cancer-specific survival between men with or without T2DM. The risk of overall mortality was higher (HR = 1.24, 95% CI = 1.07-1.43) in diabetic men. Univariate analysis showed cancer stage at diagnosis and age to be significant predictors of survival. After adjustment for age and stage at diagnosis, there was no difference in prostate-specific survival between non-diabetic patients compared to metformin users or metformin non-users. However, overall survival was lower in T2DM patients, with a higher mortality risk for metformin non-users (HR = 1.63, 95% CI = 1.27-2.10). Prostate cancer-specific mortality risk was insignificantly lower in diabetic men on metformin (HR = 0.74, 95% CI = 0.54-1.02). CONCLUSION There was no difference in long-term prostate cancer-specific survival in patients with or without T2DM. Overall survival was lower in T2DM patients not treated with metformin.
Collapse
Affiliation(s)
- Donata Linkeviciute-Ulinskiene
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ausvydas Patasius
- Laboratory of Cancer Epidemiology, National Cancer Institute, Vilnius, Lithuania.,Department of Public Health, Institute of Health Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Marius Kincius
- Department of Oncourology, National Cancer Institute, Vilnius, Lithuania
| | - Lina Zabuliene
- Clinic of Rheumatology, Orthopaedics, Traumatology and Reconstructive Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Giedre Smailyte
- Laboratory of Cancer Epidemiology, National Cancer Institute, Vilnius, Lithuania.,Department of Public Health, Institute of Health Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
136
|
Ma M, Ma C, Li P, Ma C, Ping F, Li W, Xu L, Zhang H, Sun Q, Li Y. Low glucose enhanced metformin's inhibitory effect on pancreatic cancer cells by suppressing glycolysis and inducing energy stress via up-regulation of miR-210-5p. Cell Cycle 2020; 19:2168-2181. [PMID: 32718270 PMCID: PMC7513847 DOI: 10.1080/15384101.2020.1796036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To explore mechanisms underlying the discrepancy in anti-tumor effects of metformin on pancreatic cancer cells PANC-1 under different glucose conditions. We cultured PANC-1 cells in 25 mM and 5 mM glucose media, then treated with or without metformin. It showed that metformin significantly inhibited proliferation and viability, induced apoptosis of PANC-1 cells, which was more pronounced in low-glucose than in high-glucose group. Metformin up-regulated the expression of miR-210-5p in low glucose, but not in high glucose. miR-210-5p mimic inhibited the viability of PANC-1 cells and further enhanced the inhibitory effect of metformin. miR-210-5p down-regulated the expression of PFKFB2, a predicted target gene of miR-210-5p, reduced the activity of PFK1 and LDH. Metformin significantly inhibited the expression of phosphorylation-PFKFB2(p-PFKFB2) in the low-glucose group and inhibited the LDH activity both in the low and high glucose groups, thus inhibiting anaerobic glycolysis and inducing energy stress. Cells in the high glucose group could make a compensatory adaptation to the energy stress induced by metformin through increasing glucose consumption. However, due to the limited glucose supply and high dependence on anaerobic glycolysis of cells in the low glucose group, they couldn’t make effective adaptive compensation. Therefore, cells in the low-glucose group were more vulnerable to the toxicity of metformin. In conclusion, the enhanced inhibitory effect of metformin on PANC-1 cells cultured in low glucose may be due to the up-regulation of the expression of miR-210-5p, then inhibiting anaerobic glycolytic flux and inducing energy stress via repressing the expression of p-PFKFB2 and activity of LDH. Abbreviations PC: pancreatic cancer; DM: diabetes mellitus; PFKFB2: 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase2; PFK1: phosphofructokinases; LDH: lactate dehydrogenase; F-2,6-BP: fructose 2,6-bisphosphate
Collapse
Affiliation(s)
- Minglei Ma
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Chifa Ma
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical, Sciences and Peking Union Medical College , Beijing, China.,Diabetes Research Center of Chinese Academy of Medical Sciences , Beijing 100050, China
| | - Chunxiao Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical, Sciences and Peking Union Medical College , Beijing, China.,Diabetes Research Center of Chinese Academy of Medical Sciences , Beijing 100050, China
| | - Fan Ping
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Wei Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Lingling Xu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Huabing Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Qi Sun
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| | - Yuxiu Li
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College , Beijing 100730,China
| |
Collapse
|
137
|
Oxidative stress and TGF-β1 induction by metformin in MCF-7 and MDA-MB-231 human breast cancer cells are accompanied with the downregulation of genes related to cell proliferation, invasion and metastasis. Pathol Res Pract 2020; 216:153135. [PMID: 32853957 DOI: 10.1016/j.prp.2020.153135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022]
Abstract
High doses of metformin induces oxidative stress (OS) and transforming growth factor β1 (TGF-β1) in breast cancer cells, which was associated with increased cancer stem cell population, local invasion, liver metastasis and treatment resistance. Considering the impact of TGF- β1 and OS in breast cancer and the interrelation between these two pathways, the objective of this work was to investigate the effects of consecutive metformin treatments, at a non-cytotoxic dosage, in TGF- β1 targets in MCF-7 and MDA-MB-231 cells. Cells were exposed to 6 μM of metformin for seven consecutive passages. Samples were collected to immunocytochemistry (evaluation of p53, Nf-кB, NRF2 and TGF-β1), biochemical (determination of lipoperoxidation, total thiols and nitric oxide/peroxynitrite levels) and molecular biology analyzes (microarray and Real-time quantitative array PCR). Microarray analysis confirmed alterations in genes related to OS and TGF-β1. Treatment interfered in several TGF-β1 target-genes. Metformin upregulated genes involved in OS generation and apoptosis, and downregulated genes associated with metastasis and epithelial mesenchymal transition in MCF-7 cells. In MDA-MB-231 cells, metformin downregulated genes involved with cell invasion, viability and proliferation. The results shows that even a non-cytotoxic dosage of metformin can promote a less aggressive profile of gene expression in breast cancer cells.
Collapse
|
138
|
Chung WS, Le PH, Kuo CJ, Chen TH, Kuo CF, Chiou MJ, Chou WC, Yeh TS, Hsu JT. Impact of Metformin Use on Survival in Patients with Gastric Cancer and Diabetes Mellitus Following Gastrectomy. Cancers (Basel) 2020; 12:2013. [PMID: 32717852 PMCID: PMC7465508 DOI: 10.3390/cancers12082013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
Studies have shown the anticancer effects of metformin in vitro. However, whether metformin can prevent cancer recurrence or prolong survival in patients with gastric cancer (GC) and diabetes mellitus (DM) post-gastrectomy remains unknown. We evaluated the beneficial effects of metformin in patients with GC and DM post-gastrectomy. We recruited 2400 patients with GC (1749 without DM, 651 with DM) who underwent surgery between 1997 and 2010. Patients with DM were stratified into metformin (group 1) and non-metformin (group 2) users. Their clinicopathological data were recorded prospectively, and demographics, recurrence-free survival (RFS), and cancer-specific survival (CSS) were compared. Tumour recurrence risk and cause of death were analysed between groups 1 and 2 among patients with DM stratified by tumour stage. We also compared RFS and overall survival among patients with and without DM. Tumour recurrence occurred in 201 patients with GC: 57 (25%) in group 1 and 144 (37%) in group 2. After adjusting for confounders, metformin significantly prolonged CSS (hazard ratio (HR) = 0.54, 95% confidence interval (CI) = 0.38-0.77) in patients with stage I-III GC and DM. In subgroup analysis, metformin users with stage III GC and DM had significantly prolonged CSS compared to non-metformin users (HR = 0.45, 95% CI = 0.30-0.68), with an insignificant difference in patients with stage I-II GC. Adjusted HRs for RFS and CSS were significantly lower in patients with stage I-III GC and DM than those in patients without DM (0.67 (95% CI = 0.54-0.92) and 0.62 (95% CI = 0.50-0.77), respectively), with an insignificant difference in patients with stage I GC. Metformin significantly reduces tumour recurrence risk and improves CSS in patients with stage III GC and DM post-gastrectomy. Further prospective studies may confirm the efficacy of metformin as an adjunctive treatment for advanced GC postoperatively.
Collapse
Affiliation(s)
- Wai-Shan Chung
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (W.-S.C.); (T.-S.Y.)
| | - Po-Hsien Le
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Chiang-Jung Kuo
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Tsung-Hsing Chen
- Department of Gastroenterology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (P.-H.L.); (C.-J.K.); (T.-H.C.)
| | - Chang-Fu Kuo
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan;
| | - Meng-Jiun Chiou
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan;
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan;
| | - Ta-Sen Yeh
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (W.-S.C.); (T.-S.Y.)
| | - Jun-Te Hsu
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (W.-S.C.); (T.-S.Y.)
| |
Collapse
|
139
|
Eslami SS, Jafari D, Montazeri H, Sadeghizadeh M, Tarighi P. Combination of Curcumin and Metformin Inhibits Cell Growth and Induces Apoptosis without Affecting the Cell Cycle in LNCaP Prostate Cancer Cell Line. Nutr Cancer 2020; 73:1026-1039. [PMID: 32657143 DOI: 10.1080/01635581.2020.1783327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Side effects and chemotherapy resistance, demand new therapeutics with minimal side effects. Here, we investigated the combined effect of curcumin and metformin on the LNCaP prostate cancer cell line. LNCaP cells were treated with curcumin, metformin, and their combination at different concentrations. Cell viability was assessed by MTT assay and expression of Bax, Bcl-2, mTOR, hTERT, PUMA, p53 and p21 genes was analyzed by real-time PCR. Apoptosis and cell cycle were assessed by flow cytometry. Our results revealed that the viability of cells treated with curcumin, metformin, and their combination was significantly (P < 0.05) reduced with increasing the concentration and prolonging the treatment time. Meanwhile, the combination showed a synergistic effect within 48 h. In the curcumin treated group, the expression of Bcl-2 and hTERT genes diminished. In the metformin treated group, the expression of Bax and PUMA genes was enhanced while the expression of Bcl-2, hTERT, mTOR, and p53 genes declined. Although all treatments induced apoptosis, the combination of curcumin and metformin showed the maximum level of apoptosis, cytotoxicity, and expression of Bax gene. The combination of curcumin and metformin showed synergistic effects within 48 h. This combination could be a potential therapeutic candidate for prostate cancer to be further investigated.
Collapse
Affiliation(s)
- Seyed Sadegh Eslami
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Montazeri
- School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parastoo Tarighi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
140
|
Kulkarni AS, Gubbi S, Barzilai N. Benefits of Metformin in Attenuating the Hallmarks of Aging. Cell Metab 2020; 32:15-30. [PMID: 32333835 PMCID: PMC7347426 DOI: 10.1016/j.cmet.2020.04.001] [Citation(s) in RCA: 439] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/04/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Biological aging involves an interplay of conserved and targetable molecular mechanisms, summarized as the hallmarks of aging. Metformin, a biguanide that combats age-related disorders and improves health span, is the first drug to be tested for its age-targeting effects in the large clinical trial-TAME (targeting aging by metformin). This review focuses on metformin's mechanisms in attenuating hallmarks of aging and their interconnectivity, by improving nutrient sensing, enhancing autophagy and intercellular communication, protecting against macromolecular damage, delaying stem cell aging, modulating mitochondrial function, regulating transcription, and lowering telomere attrition and senescence. These characteristics make metformin an attractive gerotherapeutic to translate to human trials.
Collapse
Affiliation(s)
- Ameya S Kulkarni
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA; Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, New York, NY, USA.
| | - Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA; Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, New York, NY, USA.
| |
Collapse
|
141
|
Gadag S, Sinha S, Nayak Y, Garg S, Nayak UY. Combination Therapy and Nanoparticulate Systems: Smart Approaches for the Effective Treatment of Breast Cancer. Pharmaceutics 2020; 12:E524. [PMID: 32521684 PMCID: PMC7355786 DOI: 10.3390/pharmaceutics12060524] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer has become one of the biggest concerns for oncologists in the past few decades because of its unpredictable etiopathology and nonavailability of personalized translational medicine. The number of women getting affected by breast cancer has increased dramatically, owing to lifestyle and environmental changes. Besides, the development of multidrug resistance has become a challenge in the therapeutic management of breast cancer. Studies reveal that the use of monotherapy is not effective in the management of breast cancer due to high toxicity and the development of resistance. Combination therapies, such as radiation therapy with adjuvant therapy, endocrine therapy with chemotherapy, and targeted therapy with immunotherapy, are found to be effective. Thus, multimodal and combination treatments, along with nanomedicine, have emerged as a promising strategy with minimum side effects and drug resistance. In this review, we emphasize the multimodal approaches and recent advancements in breast cancer treatment modalities, giving importance to the current data on clinical trials. The novel treatment approach by targeted therapy, according to type, such as luminal, HER2 positive, and triple-negative breast cancer, are discussed. Further, passive and active targeting technologies, including nanoparticles, bioconjugate systems, stimuli-responsive, and nucleic acid delivery systems, including siRNA and aptamer, are explained. The recent research exploring the role of nanomedicine in combination therapy and the possible use of artificial intelligence in breast cancer therapy is also discussed herein. The complexity and dynamism of disease changes require the constant upgrading of knowledge, and innovation is essential for future drug development for treating breast cancer.
Collapse
Affiliation(s)
- Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Shristi Sinha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Sanjay Garg
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Usha Y. Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| |
Collapse
|
142
|
Stewart JM, Lamy R, Wu F, Keenan JD. Relationship between Oral Metformin Use and Age-Related Macular Degeneration. Ophthalmol Retina 2020; 4:1118-1119. [PMID: 32525055 DOI: 10.1016/j.oret.2020.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Jay M Stewart
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California.
| | - Ricardo Lamy
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| | - Frances Wu
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| | - Jeremy D Keenan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Francis I. Proctor Foundation for Research in Ophthalmology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
143
|
Manipulation of Metabolic Pathways and Its Consequences for Anti-Tumor Immunity: A Clinical Perspective. Int J Mol Sci 2020; 21:ijms21114030. [PMID: 32512898 PMCID: PMC7312891 DOI: 10.3390/ijms21114030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
In the relatively short history of anti-tumor treatment, numerous medications have been developed against a variety of targets. Intriguingly, although many anti-tumor strategies have failed in their clinical trials, metformin, an anti-diabetic medication, demonstrated anti-tumor effects in observational studies and even showed its synergistic potential with immune checkpoint inhibitors (ICIs) in subsequent clinical studies. Looking back from bedside-to-bench, it may not be surprising that the anti-tumor effect of metformin derives largely from its ability to rewire aberrant metabolic pathways within the tumor microenvironment. As one of the most promising breakthroughs in oncology, ICIs were also found to exert their immune-stimulatory effects at least partly via rewiring metabolic pathways. These findings underscore the importance of correcting metabolic pathways to achieve sufficient anti-tumor immunity. Herein, we start by introducing the tumor microenvironment, and then we review the implications of metabolic syndrome and treatments for targeting metabolic pathways in anti-tumor therapies. We further summarize the close associations of certain aberrant metabolic pathways with impaired anti-tumor immunity and introduce the therapeutic effects of targeting these routes. Lastly, we go through the metabolic effects of ICIs and conclude an overall direction to manipulate metabolic pathways in favor of anti-tumor responses.
Collapse
|
144
|
Srivastava SP, Goodwin JE. Cancer Biology and Prevention in Diabetes. Cells 2020; 9:cells9061380. [PMID: 32498358 PMCID: PMC7349292 DOI: 10.3390/cells9061380] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
The available evidence suggests a complex relationship between diabetes and cancer. Epidemiological data suggest a positive correlation, however, in certain types of cancer, a more complex picture emerges, such as in some site-specific cancers being specific to type I diabetes but not to type II diabetes. Reports share common and differential mechanisms which affect the relationship between diabetes and cancer. We discuss the use of antidiabetic drugs in a wide range of cancer therapy and cancer therapeutics in the development of hyperglycemia, especially antineoplastic drugs which often induce hyperglycemia by targeting insulin/IGF-1 signaling. Similarly, dipeptidyl peptidase 4 (DPP-4), a well-known target in type II diabetes mellitus, has differential effects on cancer types. Past studies suggest a protective role of DPP-4 inhibitors, but recent studies show that DPP-4 inhibition induces cancer metastasis. Moreover, molecular pathological mechanisms of cancer in diabetes are currently largely unclear. The cancer-causing mechanisms in diabetes have been shown to be complex, including excessive ROS-formation, destruction of essential biomolecules, chronic inflammation, and impaired healing phenomena, collectively leading to carcinogenesis in diabetic conditions. Diabetes-associated epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) contribute to cancer-associated fibroblast (CAF) formation in tumors, allowing the epithelium and endothelium to enable tumor cell extravasation. In this review, we discuss the risk of cancer associated with anti-diabetic therapies, including DPP-4 inhibitors and SGLT2 inhibitors, and the role of catechol-o-methyltransferase (COMT), AMPK, and cell-specific glucocorticoid receptors in cancer biology. We explore possible mechanistic links between diabetes and cancer biology and discuss new therapeutic approaches.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, Yale University, New Haven, CT 06520-8064, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520-8066, USA
- Correspondence: (S.P.S.); (J.E.G.)
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, Yale University, New Haven, CT 06520-8064, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520-8066, USA
- Correspondence: (S.P.S.); (J.E.G.)
| |
Collapse
|
145
|
Bever KM, Borazanci EH, Thompson EA, Durham JN, Pinero K, Jameson GS, Vrana A, Liu M, Wilt C, Wu AA, Fu W, Wang H, Yin Y, Leal JP, Jesus-Acosta AD, Zheng L, Laheru DA, Von Hoff DD, Jaffee EM, Powell JD, Le DT. An exploratory study of metformin with or without rapamycin as maintenance therapy after induction chemotherapy in patients with metastatic pancreatic adenocarcinoma. Oncotarget 2020; 11:1929-1941. [PMID: 32523648 PMCID: PMC7260120 DOI: 10.18632/oncotarget.27586] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Purpose: Metformin combined with the mTOR inhibitor rapamycin showed potential synergistic anti-tumor activity in preclinical studies in pancreatic ductal adenocarcinoma (PDA). This phase 1b study (NCT02048384) was conducted to evaluate the feasibility and activity of metformin +/– rapamycin in the maintenance setting for unselected patients with metastatic PDA (mPDA) treated with chemotherapy. Materials and Methods: Eligible patients with stable or responding mPDA after ≥ 6 months on chemotherapy were randomized 1:1 to metformin alone (Arm A) or with rapamycin (Arm B), stratified by prior treatment with FOLFIRINOX. Fluorodeoxyglucose (FDG) PET scans and peripheral blood mononuclear cells were obtained for exploratory analyses. Results: 22 subjects (11 per arm) received treatment per protocol. Median PFS/OS were 3.5 and 13.2 months respectively, with 2 year OS rate of 37%; there were no differences between arms. No responses were observed by RECIST; however, decreases in FDG avidity and/or CA19-9 were observed in several long-term survivors. Treatment related adverse events of Grade ≥ 3 occurred in 0% vs 27% of patients in Arm A vs B and were asymptomatic hematologic or electrolyte abnormalities that were not clinically significant. Improved survival was associated with low baseline neutrophil: lymphocyte ratio, baseline lack of assessable disease by PET, and greater expansion of dendritic cells following treatment. Conclusions: Metformin +/– rapamycin maintenance for mPDA was well-tolerated and several patients achieved stable disease associated with exceptionally long survival. Further prospective studies are needed to clarify the role of these agents in the maintenance setting and to enhance patient selection for such approaches.
Collapse
Affiliation(s)
- Katherine M Bever
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA.,Co-first authors
| | - Erkut H Borazanci
- Virginia Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Molecular Medicine Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.,Co-first authors
| | - Elizabeth A Thompson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - Jennifer N Durham
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - Kimberly Pinero
- Virginia Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA
| | - Gayle S Jameson
- Virginia Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Molecular Medicine Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Amber Vrana
- Virginia Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA
| | - Meizheng Liu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - Cara Wilt
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Annie A Wu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Wei Fu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Hao Wang
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai City, China
| | - Jeffrey P Leal
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ana De Jesus-Acosta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Daniel A Laheru
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Daniel D Von Hoff
- Virginia Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Molecular Medicine Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| | - Jonathan D Powell
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA
| | - Dung T Le
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, USA.,The Skip Viragh Center for Pancreas Cancer at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
146
|
Boussios S, Mikropoulos C, Samartzis E, Karihtala P, Moschetta M, Sheriff M, Karathanasi A, Sadauskaite A, Rassy E, Pavlidis N. Wise Management of Ovarian Cancer: On the Cutting Edge. J Pers Med 2020; 10:E41. [PMID: 32455595 PMCID: PMC7354604 DOI: 10.3390/jpm10020041] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer mortality among women. Two-thirds of patients present at advanced stage at diagnosis, and the estimated 5 year survival rate is 20-40%. This heterogeneous group of malignancies has distinguishable etiology and molecular biology. Initially, single-gene sequencing was performed to identify germline DNA variations associated with EOC. However, hereditary EOC syndrome can be explained by germline pathogenic variants (gPVs) in several genes. In this regard, next-generation sequencing (NGS) changed clinical diagnostic testing, allowing assessment of multiple genes simultaneously in a faster and cheaper manner than sequential single gene analysis. As we move into the era of personalized medicine, there is evidence that poly (ADP-ribose) polymerase (PARP) inhibitors exploit homologous recombination (HR) deficiency, especially in breast cancer gene 1 and 2 (BRCA1/2) mutation carriers. Furthermore, extensive preclinical data supported the development of aurora kinase (AURK) inhibitors in specific tumor types, including EOC. Their efficacy may be optimized in combination with chemotherapeutic or other molecular agents. The efficacy of metformin in ovarian cancer prevention is under investigation. Certain mutations, such as ARID1A mutations, and alterations in the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway, which are specific in ovarian clear cell carcinoma (OCCC) and endometrioid ovarian carcinoma (EnOC), may offer additional therapeutic targets in these clinical entities. Malignant ovarian germ cell tumors (MOGCTs) are rare and randomized trials are extremely challenging for the improvement of the existing management and development of novel strategies. This review attempts to offer an overview of the main aspects of ovarian cancer, catapulted from the molecular mechanisms to therapeutic considerations.
Collapse
Affiliation(s)
- Stergios Boussios
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
| | - Christos Mikropoulos
- St Luke’s Cancer Center, Royal Surrey County Hospital, Egerton Rd, Guildford GU2 7XX, UK;
| | - Eleftherios Samartzis
- Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland;
| | - Peeter Karihtala
- Department of Oncology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, P.O. Box 100, FI-00029 Helsinki, Finland;
| | - Michele Moschetta
- Cambridge University Hospitals NHS Foundation Trust, Hills Rd, Cambridge CB2 0QQ, UK;
| | - Matin Sheriff
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Afroditi Karathanasi
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Agne Sadauskaite
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, 94805 Villejuif, France;
- Department of Hematology-Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut 166830, Lebanon
| | - Nicholas Pavlidis
- Medical School, University of Ioannina, Stavros Niarchou Avenue, 45110 Ioannina, Greece;
| |
Collapse
|
147
|
Metformin: A Possible Option in Cancer Chemotherapy. Anal Cell Pathol (Amst) 2020; 2020:7180923. [PMID: 32399389 PMCID: PMC7201450 DOI: 10.1155/2020/7180923] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/20/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin has been used for a long time as an antidiabetic medication for type 2 diabetes. It is used either as a monotherapy or in combination with other antidiabetic medications. The drug came into prominence in diabetes and other conditions with cardiovascular risk after the landmark study of 1995 by the United Kingdom Prospective Diabetes Study which emphasized its importance. However, the drug has been used in experimental trials in various aspects of medicine and pharmacology such as in reproductive medicine, cancer chemotherapy, metabolic diseases, and neurodegenerative diseases. It has been in use in the treatment of polycystic ovarian disease and obesity and is being considered in type 1 diabetes. This study seeks to evaluate the relevance of metformin in cancer management. Different mechanisms have been proposed for its antitumor action which involves the following: (a) the activation of adenosine monophosphate kinase, (b) modulation of adenosine A1 receptor (ADORA), (c) reduction in insulin/insulin growth factors, and (d) the role of metformin in the inhibition of endogenous reactive oxygen species (ROS); and its resultant damage to deoxyribonucleic acid (DNA) molecule is another paramount antitumor mechanism.
Collapse
|
148
|
Biondo LA, Teixeira AAS, de O. S. Ferreira KC, Neto JCR. Pharmacological Strategies for Insulin Sensitivity in Obesity and Cancer: Thiazolidinediones and Metformin. Curr Pharm Des 2020; 26:932-945. [PMID: 31969093 DOI: 10.2174/1381612826666200122124116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/21/2019] [Indexed: 12/19/2022]
Abstract
Background:
Chronic diseases, such as obesity and cancer, have high prevalence rates. Both diseases
have hyperinsulinemia, hyperglycemia, high levels of IGF-1 and inflammatory cytokines in common. Therefore,
these can be considered triggers for cancer development and growth. In addition, low-grade inflammation that
modulates the activation of immune cells, cellular metabolism, and production of cytokines and chemokines are
common in obesity, cancer, and insulin resistance. Pharmacological strategies are necessary when a change in
lifestyle does not improve glycemic homeostasis. In this regard, thiazolidinediones (TZD) possess multiple molecular
targets and regulate PPARγ in obesity and cancer related to insulin resistance, while metformin acts
through the AMPK pathway.
Objective:
The aim of this study was to review TZD and metformin as pharmacological treatments for insulin
resistance associated with obesity and cancer.
Conclusions:
Thiazolidinediones restored adiponectin secretion and leptin sensitivity, reduced lipid droplets in
hepatocytes and orexigen peptides in the hypothalamus. In cancer cells, TZD reduced proliferation, production of
reactive oxygen species, and inflammation by acting through the mTOR and NFκB pathways. Metformin has
similar effects, though these are AMPK-dependent. In addition, both drugs can be efficient against certain side
effects caused by chemotherapy.
Collapse
Affiliation(s)
- Luana A. Biondo
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Alexandre A. S. Teixeira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Karen C. de O. S. Ferreira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jose C. R. Neto
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
149
|
Hampsch RA, Wells JD, Traphagen NA, McCleery CF, Fields JL, Shee K, Dillon LM, Pooler DB, Lewis LD, Demidenko E, Huang YH, Marotti JD, Goen AE, Kinlaw WB, Miller TW. AMPK Activation by Metformin Promotes Survival of Dormant ER + Breast Cancer Cells. Clin Cancer Res 2020; 26:3707-3719. [PMID: 32321715 DOI: 10.1158/1078-0432.ccr-20-0269] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Despite adjuvant endocrine therapy for patients with estrogen receptor alpha (ER)-positive breast cancer, dormant residual disease can persist for years and eventually cause tumor recurrence. We sought to deduce mechanisms underlying the persistence of dormant cancer cells to identify therapeutic strategies. EXPERIMENTAL DESIGN Mimicking the aromatase inhibitor-induced depletion of estrogen levels used to treat patients, we developed preclinical models of dormancy in ER+ breast cancer induced by estrogen withdrawal in mice. We analyzed tumor xenografts and cultured cancer cells for molecular and cellular responses to estrogen withdrawal and drug treatments. Publicly available clinical breast tumor gene expression datasets were analyzed for responses to neoadjuvant endocrine therapy. RESULTS Dormant breast cancer cells exhibited upregulated 5' adenosine monophosphate-activated protein kinase (AMPK) levels and activity, and upregulated fatty acid oxidation. While the antidiabetes AMPK-activating drug metformin slowed the estrogen-driven growth of cells and tumors, metformin promoted the persistence of estrogen-deprived cells and tumors through increased mitochondrial respiration driven by fatty acid oxidation. Pharmacologic or genetic inhibition of AMPK or fatty acid oxidation promoted clearance of dormant residual disease, while dietary fat increased tumor cell survival. CONCLUSIONS AMPK has context-dependent effects in cancer, cautioning against the widespread use of an AMPK activator across disease settings. The development of therapeutics targeting fat metabolism is warranted in ER+ breast cancer.
Collapse
Affiliation(s)
- Riley A Hampsch
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jason D Wells
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Nicole A Traphagen
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Charlotte F McCleery
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jennifer L Fields
- Department of Microbiology & Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Kevin Shee
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Lloye M Dillon
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Darcy B Pooler
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Lionel D Lewis
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Eugene Demidenko
- Department of Community & Family Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Yina H Huang
- Department of Microbiology & Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jonathan D Marotti
- Department of Pathology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.,Department of Comprehensive Breast Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Abigail E Goen
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - William B Kinlaw
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Todd W Miller
- Department of Molecular & Systems Biology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. .,Department of Comprehensive Breast Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| |
Collapse
|
150
|
Lu M, Xing H, Cheng L, Liu H, Lang L, Yang T, Zhao X, Xu H, Ding P. A dual-functional buformin-mimicking poly(amido amine) for efficient and safe gene delivery. J Drug Target 2020; 28:923-932. [PMID: 32312081 DOI: 10.1080/1061186x.2020.1729770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Biguanides (i.e. metformin, phenformin and buformin) are antidiabetic drugs with potential antitumor effects. Herein, a polycationic polymer, N,N'-bis(cystamine)acrylamide-buformin (CBA-Bu), containing multiple biodegradable disulphide bonds and buformin-mimicking side chains was synthesised. CBA-Bu was equipped with high efficiency and safety profile for gene delivery, meanwhile exhibiting potential antitumor efficacy. As a gene vector, CBA-Bu was able to condense plasmid DNA (pDNA) into nano-sized (<200 nm), positively-charged (>30 mV) uniform polyplexes that were well resistant to heparin and DNase I. Due to the reduction responsiveness of the disulphide bonds, CBA-Bu/pDNA polyplexes could release the loaded pDNA in the presence of dithiothreitol, and induce extremely low cytotoxicity in NIH/3T3 and U87 MG cells. The transfection results showed that CBA-Bu had a cellular uptake efficiency comparable to 25 kDa PEI, while a significantly higher gene expression level. Additionally, CBA-Bu had a lower IC50 value than its non-biguanide counterpart in two cancer cell lines. Furthermore, CBA-Bu could activate AMPK and inhibit mTOR pathways in U87 MG cells, a mechanism involved in the antitumor effect of biguanides. Taken together, CBA-Bu represented an advanced gene vector combining desirable gene delivery capability with potential antitumor activity, which was promising to achieve enhanced therapeutic efficacy in antitumor gene therapy.
Collapse
Affiliation(s)
- Mei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haonan Xing
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Lin Cheng
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Lang Lang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, Maine, USA
| | - Xiaoyun Zhao
- School of life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Hui Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|