101
|
Laskar J, Bhattacharjee K, Sengupta M, Choudhury Y. Anti-Diabetic Drugs: Cure or Risk Factors for Cancer? Pathol Oncol Res 2018. [DOI: 10.1007/s12253-018-0402-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
102
|
Nam SY. Obesity-Related Digestive Diseases and Their Pathophysiology. Gut Liver 2018; 11:323-334. [PMID: 27890867 PMCID: PMC5417774 DOI: 10.5009/gnl15557] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 12/25/2015] [Indexed: 12/13/2022] Open
Abstract
Obesity is a growing medical and public health problem worldwide. Many digestive diseases are related to obesity. In this article, the current state of our knowledge of obesity-related digestive diseases, their pathogenesis, and the medical and metabolic consequences of weight reduction are discussed. Obesity-related digestive diseases include gastroesophageal reflux disease, Barrett’s esophagus, esophageal cancer, colon polyp and cancer, nonalcoholic fatty liver disease, hepatitis C-related disease, hepatocellular carcinoma, gallstone, cholangiocarcinoma, and pancreatic cancer. Although obesity-related esophageal diseases are associated with altered mechanical and humoral factors, other obesity-related digestive diseases seem to be associated with obesity-induced altered circulating levels of adipocytokines and insulin resistance. The relationship between functional gastrointestinal disease and obesity has been debated. This review provides a comprehensive evaluation of the obesity-related digestive diseases, including pathophysiology, obesity-related risk, and medical and metabolic effects of weight reduction in obese subjects.
Collapse
Affiliation(s)
- Su Youn Nam
- Department of Gastroenterology, Gastric Cancer Center, Kyungpook National University Medical Center, Daegu, Korea
| |
Collapse
|
103
|
Hu S, Ouyang Q, Cheng Q, Wang J, Feng F, Qiao L, Gan W, Shi Y, Wu D, Jiang X. Phenformin inhibits cell proliferation and induces cell apoptosis and autophagy in cholangiocarcinoma. Mol Med Rep 2018; 17:6028-6032. [PMID: 29436644 DOI: 10.3892/mmr.2018.8573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 10/10/2017] [Indexed: 11/06/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignant tumor and the prognosis of patients with advanced stage disease remains poor. Therefore, the identification of novel treatment agents for CCA is required. In the present study, the biological effects of the diabetes therapeutic agent, phenformin, in CCA cell lines was investigated. Cell Counting Kit‑8 cell viability, cellular clone formation and subcutaneous tumor formation assays were performed, which revealed that phenformin inhibited CCA cell proliferation and growth both in vitro and in vivo. In addition, phenformin induced CCA cell apoptosis and autophagy. Phenformin partly activated the liver kinase B1 (LKB1)/5' AMP‑activated protein kinase signaling pathway to exert its biological effects on CCA cell lines, as demonstrated by knockdown of LKB1, which reversed these effects. In conclusion, the present study demonstrated the biological effects of phenformin in CCA and suggested that phenformin may be a potential novel agent for CCA treatment.
Collapse
Affiliation(s)
- Shuyang Hu
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Qing Ouyang
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Qingbao Cheng
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Jinghan Wang
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Feiling Feng
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Liang Qiao
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Wei Gan
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Yang Shi
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Demin Wu
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| | - Xiaoqing Jiang
- First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
104
|
Dogan Turacli I, Umudum H, Pampal A, Candar T, Kavasoglu L, Sari Y. Do MCF7 cells cope with metformin treatment under energetic stress in low glucose conditions? Mol Biol Rep 2018; 45:195-201. [PMID: 29397517 DOI: 10.1007/s11033-018-4152-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/30/2018] [Indexed: 12/22/2022]
Abstract
There is a growing body of evidence about metformin being effective in cancer therapy. Despite controversies about the ways of its effectiveness, several ongoing clinical trials are evaluating the drug when used as an adjuvant or a neo-adjuvant agent. We aimed to investigate metformin's effects on proliferation, metastasis, and hormone receptor expressions in breast cancer cell line MCF-7 incubated in two different glucose conditions. MCF-7 cells were incubated in high or low glucose media and treated with various doses of metformin. The cell viability was studied using MTT test. The Ki-67, estrogen and progesterone receptor expression were evaluated by ICC and galectin-3 expression was evaluated by ELISA or spectrophotometrically. The cell viability following consecutive metformin doses in either glucose condition for 24 and 48 h represented a significant decrease when compared to control. The proliferation detected in low glucose medium following metformin at doses < 20 mM was found significantly decreased when compared to high glucose medium at 48 h. In terms of galectin-3 levels, the increase in high glucose medium treated with metformin and the decrease in low glucose medium were found statistically significant when compared to control. Progesterone receptor staining demonstrated a significant increase in low glucose medium. Our findings represent better outcomes for cancer lines incubated in low glucose medium treated with metformin in terms of viability, receptor expression and metastatic activity, and highlight the potential benefit of metformin especially in restraining the cancer cell's ability to cope energetic stress in low glucose conditions.
Collapse
Affiliation(s)
| | - Haldun Umudum
- Department of Pathology, Ufuk University, Ankara, Turkey
| | - Arzu Pampal
- Department of Pediatrics Surgery, Ufuk University, Ankara, Turkey
| | - Tuba Candar
- Department of Medical Biochemistry, Ufuk University, Ankara, Turkey
| | | | - Yaren Sari
- Faculty of Medicine, Ufuk University, Ankara, Turkey
| |
Collapse
|
105
|
Najafi M, Cheki M, Rezapoor S, Geraily G, Motevaseli E, Carnovale C, Clementi E, Shirazi A. Metformin: Prevention of genomic instability and cancer: A review. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 827:1-8. [PMID: 29502733 DOI: 10.1016/j.mrgentox.2018.01.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/28/2017] [Accepted: 01/15/2018] [Indexed: 12/21/2022]
Abstract
The diabetes drug metformin can mitigate the genotoxic effects of cytotoxic agents and has been proposed to prevent or even cure certain cancers. Metformin reduces DNA damage by mechanisms that are only incompletely understood. Metformin scavenges free radicals, including reactive oxygen species and nitric oxide, which are produced by genotoxicants such as ionizing or non-ionizing radiation, heavy metals, and chemotherapeutic agents. The drug may also increase the activities of antioxidant enzymes and inhibit NADPH oxidase, cyclooxygenase-2, and inducible nitric oxide synthase, thereby limiting macrophage recruitment and inflammatory responses. Metformin stimulates the DNA damage response (DDR) in the homologous end-joining, homologous recombination, and nucleotide excision repair pathways. This review focuses on the protective properties of metformin against genomic instability.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohsen Cheki
- Department of Radiologic Technology, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazale Geraily
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences L. Sacco, Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, Università di Milano, Milan, Italy
| | - Emilio Clementi
- Scientific Institute, IRCCS E. Medea, Bosisio Parini, Lecco, Italy; Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, L. Sacco University Hospital, Università di Milano, Milan, Italy
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
106
|
Zi F, Zi H, Li Y, He J, Shi Q, Cai Z. Metformin and cancer: An existing drug for cancer prevention and therapy. Oncol Lett 2018; 15:683-690. [PMID: 29422962 PMCID: PMC5772929 DOI: 10.3892/ol.2017.7412] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 09/22/2017] [Indexed: 12/17/2022] Open
Abstract
Metformin is a standard clinical drug used to treat type 2 diabetes mellitus (T2DM) and polycystic ovary syndrome. Recently, epidemiological studies and meta-analyses have revealed that patients with T2DM have a lower incidence of tumor development than healthy controls and that patients diagnosed with cancer have a lower risk of mortality when treated with metformin, demonstrating an association between metformin and tumorigenesis. In vivo and in vitro studies have revealed that metformin has a direct antitumor effect, which may depress tumor proliferation and induce the apoptosis, autophagy and cell cycle arrest of tumor cells. The mechanism underpinning the antitumor effect of metformin has not been well established. Studies have demonstrated that reducing insulin and insulin-like growth factor levels in the peripheral blood circulation may lead to the inhibition of phosphoinositide 3-kinase/Akt/mechanistic target of rapamycin (mTOR) signaling or activation of AMP-activated protein kinase, which inhibits mTOR signaling, a process that may be associated with the antitumor effect of metformin. The present review primarily focuses on the recent progress in understanding the function of metformin in tumor development.
Collapse
Affiliation(s)
- Fuming Zi
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Huapu Zi
- Department of Oncology, Rizhao Traditional Chinese Medicine Hospital of Shandong Traditional Chinese Medicine University, Rizhao, Shandong 276800, P.R. China
| | - Yi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Qingzhi Shi
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
107
|
Ashamalla M, Youssef I, Yacoub M, Jayarangaiah A, Gupta N, Ray J, Iqbal S, Miller R, Singh J, McFarlane SI. Obesity, Diabetes and Gastrointestinal Malignancy: The role of Metformin and other Anti-diabetic Therapy. GLOBAL JOURNAL OF OBESITY, DIABETES AND METABOLIC SYNDROME 2018; 5:008-14. [PMID: 30533942 PMCID: PMC6282807 DOI: 10.17352/2455-8583.000032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The association between Diabetes and cancer has been known for decades with obesity and insulin resistance being postulated as the main underlying risk factors for both disorders. With rise of the epidemic of obesity in the USA and around the globe, there has been a rise in diabetes that is currently reaching epidemic proportions. Diabetes is known to be associated with increased risk of several types of malignancy including breast, cervical, pancreatic and colon cancer. In this review, we discuss the epidemic of obesity and its consequential epidemic of diabetes highlighting the pathophysiologic mechanisms of increased cancer in the diabetic population. We will then discuss the role of insulin therapy as well as, other antidiabetic medications, particularly metformin that has been to be associated with lower risk as well as better survival with GI malignancies based on several studies including a study that was recently published by our group.
Collapse
Affiliation(s)
- Michael Ashamalla
- Northwell Health care system, Department of Medicine, New Hyde Park, New York 11040, USA
| | - Irini Youssef
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Mena Yacoub
- Northside Hospital, St. Petersburg, Florida, 33709, USA
| | - Apoorva Jayarangaiah
- Department of Internal Medicine, Wake Forest University, Baptist Health System, Winston-Salem, N.C, USA
| | - Nikita Gupta
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Justina Ray
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Sadat Iqbal
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Regina Miller
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Joie Singh
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA
| | - Samy I McFarlane
- Department of Radiation Oncology, Department of Medicine, Division of Endocrinology, SUNY-Downstate, Brooklyn, NY 11203, USA,Corresponding author: Samy I McFarlane Distinguished Teaching Professor and associate Dean, College of Medicine, Department of Medicine, Division of Endocrinology, Internal Medicine Residency Program Director, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 50, Brooklyn, NY 11203-2098, USA, Tel: 718-270-3711; Fax 718-270-6358;
| |
Collapse
|
108
|
Biondani G, Peyron JF. Metformin, an Anti-diabetic Drug to Target Leukemia. Front Endocrinol (Lausanne) 2018; 9:446. [PMID: 30147674 PMCID: PMC6095966 DOI: 10.3389/fendo.2018.00446] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Metformin, a widely used anti-diabetic molecule, has attracted a strong interest in the last 10 years as a possible new anti-cancer molecule. Metformin acts by interfering with mitochondrial respiration, leading to an activation of the AMPK tumor-suppressive pathway to promote catabolic-energy saving reactions and block anabolic ones that are associated with abnormal cell proliferation. Metformin also acts at the organism level. In type 2 diabetes patients, metformin reduces hyperglycemia and increases insulin sensitivity by enhancing insulin-stimulated glucose uptake in muscles, liver, and adipose tissue and by reducing glucose output by the liver. Lowering insulin and insulin-like growth factor 1 (IGF-1) levels that stimulate cancer growth could be important features of metformin's mode of action. Despite continuous progress in treatments with the use of targeted therapies and now immunotherapies, acute leukemias are still of very poor prognosis for relapse patients, demonstrating an important need for new treatments deriving from the identification of their pathological supportive mechanisms. In the last decade, it has been realized that if cancer cells modify and reprogram their metabolism to feed their intense biochemical needs associated with their runaway proliferation, they develop metabolic addictions that could represent attractive targets for new therapeutic strategies that intend to starve and kill cancer cells. This Mini Review explores the anti-leukemic potential of metformin and its mode of action on leukemia metabolism.
Collapse
|
109
|
Zheng G, Shen Z, Xu A, Jiang K, Wu P, Yang X, Chen X, Shao J. Synergistic Chemopreventive and Therapeutic Effects of Co-drug UA-Met: Implication in Tumor Metastasis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:10973-10983. [PMID: 29227654 DOI: 10.1021/acs.jafc.7b04378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The anticancer properties of ursolic acid (UA) and metformin (Met) have been well demonstrated. However, whether these compounds can act synergistically to prevent and treat cancer is not known. We present in this study, the synergism between UA and Met, and that of a new codrug made of UA and Met (UA-Met) against several cancer cell lines. The combination of high concentration of UA (25, 50, 75, 100 μM) and Met (5, 10, 20, 40 mM) resulted in synergetic cytotoxicity on MDA-MB-231 and MCF-7 cells (CI < 0.8). Molecular and cellular studies showed that codrug UA-Met significantly inhibited the invasion (∼55.3 ± 2.74%) and migration (∼52.4 ± 1.57%) of TGF-β induced breast cancer MDA-MB-231 and MCF-7 cells in vitro at low concentration of 10 μM. These effects were accompanied by down-regulation of CXCR4, uPA, vimentin, E-cadherin, N-cadherin, and MMP-2/9 proteins expression and regulation of the AMPK/m-TOR signaling pathways as expected from UA and Met. Moreover, UA-Met could reduce the progression of pulmonary metastasis by 4T1 cells (63.4 ± 3.52%) without influencing the glucose blood level in mice. Our study suggests that the codrug UA-Met is safe and effective in preventing cancer metastasis and possibly treatment of cancer.
Collapse
Affiliation(s)
- Guirong Zheng
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Zhichun Shen
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Aixiao Xu
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Kai Jiang
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Pengyu Wu
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Xiang Yang
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Xian Chen
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University , Fuzhou 350116, China
| |
Collapse
|
110
|
Bifunctional enzyme ATIC promotes propagation of hepatocellular carcinoma by regulating AMPK-mTOR-S6 K1 signaling. Cell Commun Signal 2017; 15:52. [PMID: 29246230 PMCID: PMC5732395 DOI: 10.1186/s12964-017-0208-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/07/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the cancer types with poor prognosis. To effectively treat HCC, new molecular targets and therapeutic approaches must be identified. 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate (IMP) cyclohydrolase (ATIC), a bifunctional protein enzyme, catalyzes the last two steps of the de novo purine biosynthetic pathway. Whether ATIC contributes to cancer development remains unclear. METHODS ATIC mRNA levels in different types of human HCC samples or normal tissues were determined from Gene Expression across Normal and Tumor tissue (GENT) database. The expression level of ATIC in human HCC samples or cell lines were examined by RT-PCR and western blot. Overall survival and disease-free survival of HCC patients in the ATIC low and ATIC high groups were determined by Kaplan-Meier analysis. Effects of ATIC knockdown by lentivirus infection were evaluated on cell-proliferation, cell-apoptosis, colony formation and migration. The mechanisms involved in HCC cells growth, apoptosis and migration were analyzed by western blot and Compound C (C-C) rescue assays. RESULTS Here, we first demonstrated that expression of ATIC is aberrantly up-regulated in HCC tissues and high level of ATIC is correlated with poor survival in HCC patients. Knockdown of ATIC expression resulted in a dramatic decrease in proliferation, colony formation and migration of HCC cells. We also identified ATIC as a novel regulator of adenosine monophosphate-activated protein kinase (AMPK) and its downstream signaling mammalian target of rapamycin (mTOR). ATIC suppresses AMPK activation, thus activates mTOR-S6 K1-S6 signaling and supports growth and motility activity of HCC cells. CONCLUSION Taken together, our results indicate that ATIC acts as an oncogenic gene that promotes survival, proliferation and migration by targeting AMPK-mTOR-S6 K1 signaling.
Collapse
|
111
|
Identification of human age-associated gene co-expressions in functional modules using liquid association. Oncotarget 2017; 9:1063-1074. [PMID: 29416677 PMCID: PMC5787419 DOI: 10.18632/oncotarget.23148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023] Open
Abstract
Aging is a major risk factor for age-related diseases such as certain cancers. In this study, we developed Age Associated Gene Co-expression Identifier (AAGCI), a liquid association based method to infer age-associated gene co-expressions at thousands of biological processes and pathways across 9 human tissues. Several hundred to thousands of gene pairs were inferred to be age co-expressed across different tissues, the genes involved in which are significantly enriched in functions like immunity, ATP binding, DNA damage, and many cancer pathways. The age co-expressed genes are significantly overlapped with aging genes curated in the GenAge database across all 9 tissues, suggesting a tissue-wide correlation between age-associated genes and co-expressions. Interestingly, age-associated gene co-expressions are significantly different from gene co-expressions identified through correlation analysis, indicating that aging might only contribute to a small portion of gene co-expressions. Moreover, the key driver analysis identified biologically meaningful genes in important function modules. For example, IGF1, ERBB2, TP53 and STAT5A were inferred to be key genes driving age co-expressed genes in the network module associated with function “T cell proliferation”. Finally, we prioritized a few anti-aging drugs such as metformin based on an enrichment analysis between age co-expressed genes and drug signatures from a recent study. The predicted drugs were partially validated by literature mining and can be readily used to generate hypothesis for further experimental validations.
Collapse
|
112
|
Valtorta S, Lo Dico A, Raccagni I, Gaglio D, Belloli S, Politi LS, Martelli C, Diceglie C, Bonanomi M, Ercoli G, Vaira V, Ottobrini L, Moresco RM. Metformin and temozolomide, a synergic option to overcome resistance in glioblastoma multiforme models. Oncotarget 2017; 8:113090-113104. [PMID: 29348889 PMCID: PMC5762574 DOI: 10.18632/oncotarget.23028] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with poor survival. Cytoreduction in association with radiotherapy and temozolomide (TMZ) is the standard therapy, but response is heterogeneous and life expectancy is limited. The combined use of chemotherapeutic agents with drugs targeting cell metabolism is becoming an interesting therapeutic option for cancer treatment. Here, we found that metformin (MET) enhances TMZ effect on TMZ-sensitive cell line (U251) and overcomes TMZ-resistance in T98G GBM cell line. In particular, combined-treatment modulated apoptosis by increasing Bax/Bcl-2 ratio, and reduced Reactive Oxygen Species (ROS) production. We also observed that MET associated with TMZ was able to reduce the expression of glioma stem cells (GSC) marker CD90 particularly in T98G cells but not that of CD133. In vivo experiments showed that combined treatment with TMZ and MET significantly slowed down growth of TMZ-resistant tumors but did not affect overall survival of TMZ-sensitive tumor bearing mice. In conclusion, our results showed that metformin is able to enhance TMZ effect in TMZ-resistant cell line suggesting its potential use in TMZ refractory GBM patients. However, the lack of effect on a GBM malignancy marker like CD133 requires further evaluation since it might influence response duration.
Collapse
Affiliation(s)
- Silvia Valtorta
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Isabella Raccagni
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Daniela Gaglio
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| | - Letterio S Politi
- Imaging Core, IRCCS San Raffaele Scientific Institute, Milan, Italy.,University of Massachusetts Medical School, Worcester, MA, USA.,Hematology/Oncology Division and Radiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.,Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy
| | | | - Giulia Ercoli
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.,Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ottobrini
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Rosa Maria Moresco
- Tecnomed Foundation and Medicine and Surgery Department, University of Milan-Bicocca, Monza, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy.,SYSBIO.IT, Centre of Systems Biology, Milan, Italy
| |
Collapse
|
113
|
Dogan Turacli I, Candar T, Yuksel BE, Demirtas S. Role of metformin on base excision repair pathway in p53 wild-type H2009 and HepG2 cancer cells. Hum Exp Toxicol 2017; 37:909-919. [DOI: 10.1177/0960327117737145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The antidiabetic agent metformin was shown to further possess chemopreventive and chemotherapeutic effects against cancer. Despite the advances, the underlying molecular mechanisms involved in decreasing tumor formation are still unclear. The understanding of the participation of oxidative stress in the action mechanism of metformin and its related effects on p53 and on DNA base excision repair (BER) system can help us to get closer to solve metformin puzzle in cancer. We investigated the effects of metformin in HepG2 and H2009 cells, verifying cytotoxicity, oxidative stress, antioxidant status, and DNA BER system. Our results showed metformin induced oxidative stress and reduced antioxidant capacity. Also, metformin treatment with hydrogen peroxide (H2O2) enhanced these effects. Although DNA BER enzyme activities were not changed accordantly together by metformin as a single agent or in combination with H2O2, activated p53 was decreased with increased oxidative stress in H2009 cells. Our study on the relationship between metformin/reactive oxygen species and DNA BER system in cancer cells would be helpful to understand the anticancer effects of metformin through cellular signal transduction pathways. These findings can be a model of the changes on oxidative stress that reflects p53’s regulatory role on DNA repair systems in cancer for the future studies.
Collapse
Affiliation(s)
- Irem Dogan Turacli
- Department of Medical Biology, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Tuba Candar
- Department of Medical Biochemistry, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Berrin Emine Yuksel
- Department of Medical Genetics, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Selda Demirtas
- Department of Medical Biochemistry, Faculty of Medicine, Ufuk University, Ankara, Turkey
| |
Collapse
|
114
|
Friend or foe? Mitochondria as a pharmacological target in cancer treatment. Future Med Chem 2017; 9:2197-2210. [PMID: 29182013 DOI: 10.4155/fmc-2017-0110] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mitochondria have acquired numerous functions over the course of evolution, such as those involved in controlling energy production, cellular metabolism, cell survival, apoptosis and autophagy within host cells. Tumor cells can develop defects in mitochondrial function, presenting a potential strategy for designing selective anticancer therapies. Therefore, cancer has been the main focus of recent research to uncover possible mitochondrial targets for therapeutic benefit. This comprehensive review covers not only the recent discoveries of the roles of mitochondria in cancer development, progression and therapeutic implications but also the findings regarding emerging mitochondrial therapeutic targets and mitochondria-targeted agents. Current challenges and future directions for developments and applications of mitochondrial-targeted therapeutics are also discussed.
Collapse
|
115
|
Metformin transiently inhibits colorectal cancer cell proliferation as a result of either AMPK activation or increased ROS production. Sci Rep 2017; 7:15992. [PMID: 29167573 PMCID: PMC5700100 DOI: 10.1038/s41598-017-16149-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 11/08/2017] [Indexed: 12/15/2022] Open
Abstract
Metformin is a widely used and well-tolerated anti-diabetic drug that can reduce cancer risk and improve the prognosis of certain malignancies. However, the mechanism underlying its anti-cancer effect is still unclear. We studied the anti-cancer activity of metformin on colorectal cancer (CRC) by using the drug to treat HT29, HCT116 and HCT116 p53−/− CRC cells. Metformin reduced cell proliferation and migration by inducing cell cycle arrest in the G0/G1 phase. This was accompanied by a sharp decrease in the expression of c-Myc and down-regulation of IGF1R. The anti-proliferative action of metformin was mediated by two different mechanisms: AMPK activation and increase in the production of reactive oxygen species, which suppressed the mTOR pathway and its downstream targets S6 and 4EBP1. A reduction in CD44 and LGR5 expression suggested that the drug had an effect on tumour cells with stem characteristics. However, a colony formation assay showed that metformin slowed the cells’ ability to form colonies without arresting cell growth, as confirmed by absence of apoptosis, autophagy or senescence. Our finding that metformin only transiently arrests CRC cell growth suggests that efforts should be made to identify compounds that combined with the biguanide can act synergistically to induce cell death.
Collapse
|
116
|
Sanchez-Salazar L, Gonzales GF. Aqueous extract of yellow maca (Lepidium meyenii
) improves sperm count in experimental animals but response depends on hypocotyl size, pH and routes of administration. Andrologia 2017; 50. [DOI: 10.1111/and.12929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 12/01/2022] Open
Affiliation(s)
- L. Sanchez-Salazar
- Faculty of Sciences and Philosophy; Department of Biological and Physiological Sciences; Universidad Peruana Cayetano Heredia; Lima Peru
- Research Circle on Plants with effects on Health; Universidad Peruana Cayetano Heredia; Lima Peru
| | - G. F. Gonzales
- Faculty of Sciences and Philosophy; Department of Biological and Physiological Sciences; Universidad Peruana Cayetano Heredia; Lima Peru
- Research Circle on Plants with effects on Health; Universidad Peruana Cayetano Heredia; Lima Peru
- Instituto de Investigaciones de la Altura; Universidad Peruana Cayetano Heredia; Lima Peru
| |
Collapse
|
117
|
Targeting AMPK, mTOR and β-Catenin by Combined Metformin and Aspirin Therapy in HCC: An Appraisal in Egyptian HCC Patients. Mol Diagn Ther 2017; 22:115-127. [PMID: 29094287 DOI: 10.1007/s40291-017-0307-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
118
|
Casadei Gardini A, Faloppi L, De Matteis S, Foschi FG, Silvestris N, Tovoli F, Palmieri V, Marisi G, Brunetti O, Vespasiani-Gentilucci U, Perrone G, Valgiusti M, Granato AM, Ercolani G, Negrini G, Tamburini E, Aprile G, Passardi A, Santini D, Cascinu S, Frassineti GL, Scartozzi M. Metformin and insulin impact on clinical outcome in patients with advanced hepatocellular carcinoma receiving sorafenib: Validation study and biological rationale. Eur J Cancer 2017; 86:106-114. [PMID: 28985579 DOI: 10.1016/j.ejca.2017.09.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/28/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE In 2015, we published a study on a small series of patients with hepatocellular carcinoma (HCC) treated chronically with metformin for type II diabetes mellitus (DM2) who showed a poorer response to sorafenib. The aim of the present study was to validate the prognostic significance of metformin in HCC patients treated with sorafenib, providing a biological rationale for the mechanism of resistance to sorafenib in patients on chronic metformin therapy, and to clarify the role of sirtuin-3 (SIRT-3), a protein involved in metabolic diseases and acknowledged as a tumour suppressor in HCC, in this resistance. PATIENTS AND METHODS We analysed 279 patients consecutively treated with sorafenib for the clinical analysis. Of the 86 (30%) patients with DM2, 52 (19%) were on chronic treatment with metformin and 34 (12%) with insulin. We included 43 patients with HCC for the biological study: 19 (44.1%) were diabetic and 14 (73.7%) of these received metformin for DM2. SIRT-3 expression was investigated by immunohistochemistry (IHC) in formalin-fixed and paraffin-embedded (FFPE) samples. RESULTS In HCC patients undergoing chronic treatment with metformin, the use of sorafenib was associated with poor progression-free survival (PFS) and overall survival (OS) (1.9 and 6.6 months, respectively) compared to 3.7 months and 10.8 months, respectively, for patients without DM2 and 8.4 months and 16.6 months, respectively, for patients on insulin (P < .0001). We also observed that SIRT-3 protein expression was significantly higher in patients treated with metformin than in those not taking this medication (65% versus 25%, respectively) (P = .013). CONCLUSIONS Our findings could be attributed to increased tumour aggressiveness and resistance to sorafenib caused by chronic treatment with metformin.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Databases, Factual
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/mortality
- Disease-Free Survival
- Drug Interactions
- Drug Resistance, Neoplasm
- Female
- Humans
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/therapeutic use
- Immunohistochemistry
- Insulin/adverse effects
- Insulin/therapeutic use
- Italy
- Kaplan-Meier Estimate
- Liver Neoplasms/drug therapy
- Liver Neoplasms/enzymology
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Male
- Metformin/adverse effects
- Metformin/therapeutic use
- Middle Aged
- Niacinamide/adverse effects
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Phenylurea Compounds/adverse effects
- Phenylurea Compounds/therapeutic use
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Retrospective Studies
- Sirtuin 3/analysis
- Sorafenib
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - Luca Faloppi
- Department of Medical Oncology, University of Cagliari, Italy
| | - Serena De Matteis
- Biosciences Laboratory, Istituto Scientifico Romagnolo per Lo Studio e La Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Nicola Silvestris
- Department of Medical Oncology, National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Francesco Tovoli
- Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, University of Bologna, Italy
| | - Vincenzo Palmieri
- Department of Biomedical Sciences and Human Oncology, Clinica Medica A. Murri, University of Bari Medical School, Italy
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per Lo Studio e La Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Oronzo Brunetti
- Department of Medical Oncology, National Cancer Institute Giovanni Paolo II, Bari, Italy
| | | | - Giuseppe Perrone
- Department of Pathology, Università Campus Bio-Medico, Rome, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Anna Maria Granato
- Immunotherapy Unit, Istituto Scientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgio Ercolani
- Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, University of Bologna, Italy; Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Giulia Negrini
- Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, University of Bologna, Italy
| | | | - Giuseppe Aprile
- Department of Oncology, University and General Hospital, Udine, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Daniele Santini
- Medical Oncology Unit, Università Campus Bio-Medico, Rome, Italy
| | - Stefano Cascinu
- Modena Cancer Center, Policlinico di Modena, Università di Modena e Reggio Emilia, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Italy
| |
Collapse
|
119
|
Kunisada Y, Eikawa S, Tomonobu N, Domae S, Uehara T, Hori S, Furusawa Y, Hase K, Sasaki A, Udono H. Attenuation of CD4 +CD25 + Regulatory T Cells in the Tumor Microenvironment by Metformin, a Type 2 Diabetes Drug. EBioMedicine 2017. [PMID: 29066174 DOI: 10.1016/j.ebiom.2017.10.009.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
CD4+CD25+ regulatory T cells (Treg), an essential subset for preventing autoimmune diseases, is implicated as a negative regulator in anti-tumor immunity. We found that metformin (Met) reduced tumor-infiltrating Treg (Ti-Treg), particularly the terminally-differentiated CD103+KLRG1+ population, and also decreased effector molecules such as CTLA4 and IL-10. Met inhibits the differentiation of naïve CD4+ T cells into inducible Treg (iTreg) by reducing forkhead box P3 (Foxp3) protein, caused by mTORC1 activation that was determined by the elevation of phosphorylated S6 (pS6), a downstream molecule of mTORC1. Rapamycin and compound C, an inhibitor of AMP-activated protein kinase (AMPK) restored the iTreg generation, further indicating the involvement of mTORC1 and AMPK. The metabolic profile of iTreg, increased Glut1-expression, and reduced mitochondrial membrane-potential and ROS production of Ti-Treg aided in identifying enhanced glycolysis upon Met-treatment. The negative impact of Met on Ti-Treg may help generation of the sustained antitumor immunity.
Collapse
Affiliation(s)
- Yuki Kunisada
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shingo Eikawa
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nahoko Tomonobu
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shohei Domae
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takenori Uehara
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shohei Hori
- Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yukihiro Furusawa
- Division of Biochemistry, Keio University Graduate School of Pharmaceutical Science, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Keio University Graduate School of Pharmaceutical Science, Tokyo, Japan
| | - Akira Sasaki
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan. @cc.okayama-u.ac.jp
| |
Collapse
|
120
|
Chikara S, Mamidi S, Sreedasyam A, Chittem K, Pietrofesa R, Zuppa A, Moorthy G, Dyer N, Christofidou-Solomidou M, Reindl KM. Flaxseed Consumption Inhibits Chemically Induced Lung Tumorigenesis and Modulates Expression of Phase II Enzymes and Inflammatory Cytokines in A/J Mice. Cancer Prev Res (Phila) 2017; 11:27-37. [PMID: 29074535 DOI: 10.1158/1940-6207.capr-17-0119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/26/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022]
Abstract
Flaxseed consumption is associated with reduced oxidative stress and inflammation in lung injury models and has shown anticancer effects for breast and prostate tissues. However, the chemopreventive potential of flaxseed remains unexplored for lung cancer. In this study, we investigated the effect of flaxseed on tobacco smoke carcinogen (NNK)-induced lung tumorigenesis in an A/J mouse model. Mice exposed to NNK were fed a control diet or a 10% flaxseed-supplemented diet for 26 weeks. Flaxseed-fed mice showed reduced lung tumor incidence (78%) and multiplicity, with an average of 2.7 ± 2.3 surface lung tumor nodules and 1.0 ± 0.9 H&E cross-section nodules per lung compared with the control group, which had 100% tumor incidence and an average of 10.2 ± 5.7 surface lung tumor nodules and 3.9 ± 2.6 H&E cross-section nodules per lung. Furthermore, flaxseed-fed mice had a lower incidence of adenocarcinomas compared with control-fed mice. Western blotting performed on normal lung tissues showed flaxseed suppressed phosphorylation (activation) of p-AKT, p-ERK, and p-JNK kinases. RNA-Seq data obtained from normal lung and lung tumors of control and flaxseed-fed mice suggested that flaxseed intake resulted in differential expression of genes involved in inflammation-mediated cytokine signaling (IL1, 6, 8, 9, and 12α), xenobiotic metabolism (several CYPs, GSTs, and UGTs), and signaling pathways (AKT and MAPK) involved in tumor cell proliferation. Together, our results indicate that dietary flaxseed supplementation may be an effective chemoprevention strategy for chemically induced lung carcinogenesis by altering signaling pathways, inflammation, and oxidative stress. Cancer Prev Res; 11(1); 27-37. ©2017 AACR.
Collapse
Affiliation(s)
- Shireen Chikara
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota
| | - Sujan Mamidi
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | | | - Kishore Chittem
- Department of Plant Pathology, North Dakota State University, Fargo, North Dakota
| | - Ralph Pietrofesa
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Athena Zuppa
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ganesh Moorthy
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Neil Dyer
- Department of Animal Sciences, North Dakota State University, Fargo, North Dakota
| | | | - Katie M Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota.
| |
Collapse
|
121
|
Kunisada Y, Eikawa S, Tomonobu N, Domae S, Uehara T, Hori S, Furusawa Y, Hase K, Sasaki A, Udono H. Attenuation of CD4 +CD25 + Regulatory T Cells in the Tumor Microenvironment by Metformin, a Type 2 Diabetes Drug. EBioMedicine 2017; 25:154-164. [PMID: 29066174 PMCID: PMC5704053 DOI: 10.1016/j.ebiom.2017.10.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022] Open
Abstract
CD4+ CD25+ regulatory T cells (Treg), an essential subset for preventing autoimmune diseases, is implicated as a negative regulator in anti-tumor immunity. We found that metformin (Met) reduced tumor-infiltrating Treg (Ti-Treg), particularly the terminally-differentiated CD103+ KLRG1+ population, and also decreased effector molecules such as CTLA4 and IL-10. Met inhibits the differentiation of naïve CD4+ T cells into inducible Treg (iTreg) by reducing forkhead box P3 (Foxp3) protein, caused by mTORC1 activation that was determined by the elevation of phosphorylated S6 (pS6), a downstream molecule of mTORC1. Rapamycin and compound C, an inhibitor of AMP-activated protein kinase (AMPK) restored the iTreg generation, further indicating the involvement of mTORC1 and AMPK. The metabolic profile of iTreg, increased Glut1-expression, and reduced mitochondrial membrane-potential and ROS production of Ti-Treg aided in identifying enhanced glycolysis upon Met-treatment. The negative impact of Met on Ti-Treg may help generation of the sustained antitumor immunity. Metformin downregulates CD4+ CD25+ regulatory T cells (Treg) in tumors but not in peripheral lymphoid tissues. Metformin administration results in activation of mTORC1 in Treg in tumors. Metformin administration results in elevation of glycolysis, while suppressing oxidative phosphorylation in Treg in tumors.
CD4+ CD25+ regulatory T cells (Treg) is a negative regulator that inhibits T cell mediated anti-tumor immunity. Therefore, targeting Treg is one of the important therapeutic intervention in cancers. We found that metformin reduces Treg in the number and the function in tumors. Metabolism of Treg is usually dependent on oxidative phosphorylation through fatty acid oxidation (FAO). However, metformin treatment causes metabolic reprogramming of Treg toward the glycolysis, resulting in the failure in survival in tumors. Metformin as a metabolic modifier for Treg may contribute to generation of sustained anti-tumor immunity, combined with currently emerging cancer immunotherapy.
Collapse
Affiliation(s)
- Yuki Kunisada
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shingo Eikawa
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nahoko Tomonobu
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shohei Domae
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Takenori Uehara
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shohei Hori
- Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yukihiro Furusawa
- Division of Biochemistry, Keio University Graduate School of Pharmaceutical Science, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Keio University Graduate School of Pharmaceutical Science, Tokyo, Japan
| | - Akira Sasaki
- Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan. @cc.okayama-u.ac.jp
| |
Collapse
|
122
|
Parikh AB, Kozuch P, Rohs N, Becker DJ, Levy BP. Metformin as a repurposed therapy in advanced non-small cell lung cancer (NSCLC): results of a phase II trial. Invest New Drugs 2017; 35:813-819. [DOI: 10.1007/s10637-017-0511-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 09/14/2017] [Indexed: 11/24/2022]
|
123
|
Asiedu MK, Barron M, Aubry MC, Wigle DA. Patient- and Cell Type-Specific Heterogeneity of Metformin Response. Basic Clin Pharmacol Toxicol 2017; 122:214-222. [PMID: 28862803 DOI: 10.1111/bcpt.12898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022]
Abstract
Most FDA-approved drugs are not equally effective in all patients, suggesting that identification of biomarkers to predict responders to a chemoprevention agent will be needed to stratify patients and achieve maximum benefit. The goal of this study was to investigate both patient-specific and cell context-specific heterogeneity of metformin response, using fibroblast cell lines and induced pluripotent stem cells differentiated into lung epithelial lineages. We performed cell survival analysis, transcriptome and whole exome sequencing analysis on both patient-derived cell lines and cancer cell lines to assess differential metformin response and identify response genes. We found differences in response to metformin treatment across a variety of cell lines and cellular contexts, suggesting that heterogeneity may be patient- and cell type-specific. Gene expression profiling and analysis of metformin-sensitive and metformin-resistant cells identified differentially expressed genes that may be able to stratify patients into metformin responders and non-responders. Sequencing analysis found genomic alterations that correlated with metformin response. These results suggest that the identification of genomic biomarkers for patients who may respond to metformin treatment can provide an opportunity for individualizing metformin chemoprevention in the clinical setting.
Collapse
Affiliation(s)
- Michael K Asiedu
- Division of General Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Matthew Barron
- Division of General Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Marie Christine Aubry
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dennis A Wigle
- Division of General Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
124
|
Rajeshkumar NV, Yabuuchi S, Pai SG, De Oliveira E, Kamphorst JJ, Rabinowitz JD, Tejero H, Al-Shahrour F, Hidalgo M, Maitra A, Dang CV. Treatment of Pancreatic Cancer Patient-Derived Xenograft Panel with Metabolic Inhibitors Reveals Efficacy of Phenformin. Clin Cancer Res 2017; 23:5639-5647. [PMID: 28611197 PMCID: PMC6540110 DOI: 10.1158/1078-0432.ccr-17-1115] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 12/12/2022]
Abstract
Purpose: To identify effective metabolic inhibitors to suppress the aggressive growth of pancreatic ductal adenocarcinoma (PDAC), we explored the in vivo antitumor efficacy of metabolic inhibitors, as single agents, in a panel of patient-derived PDAC xenograft models (PDX) and investigated whether genomic alterations of tumors correlate with the sensitivity to metabolic inhibitors.Experimental Design: Mice with established PDAC tumors from 6 to 13 individual PDXs were randomized and treated, once daily for 4 weeks, with either sterile PBS (vehicle) or the glutaminase inhibitor bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES), transaminase inhibitor aminooxyacetate (AOA), pyruvate dehydrogenase kinase inhibitor dichloroacetate (DCA), autophagy inhibitor chloroquine (CQ), and mitochondrial complex I inhibitor phenformin/metformin.Results: Among the agents tested, phenformin showed significant tumor growth inhibition (>30% compared with vehicle) in 5 of 12 individual PDXs. Metformin, at a fivefold higher dose, displayed significant tumor growth inhibition in 3 of 12 PDXs similar to BPTES (2/8 PDXs) and DCA (2/6 PDXs). AOA and CQ had the lowest response rates. Gene set enrichment analysis conducted using the baseline gene expression profile of pancreatic tumors identified a gene expression signature that inversely correlated with phenformin sensitivity, which is in agreement with the phenformin gene expression signature of NIH Library of Integrated Network-based Cellular Signatures (LINCS). The PDXs that were more sensitive to phenformin showed a baseline reduction in amino acids and elevation in oxidized glutathione. There was no correlation between phenformin response and genetic alterations in KRAS, TP53, SMAD4, or PTENConclusions: Phenformin treatment showed relatively higher antitumor efficacy against established PDAC tumors, compared with the efficacy of other metabolic inhibitors and metformin. Phenformin treatment significantly diminished PDAC tumor progression and prolonged tumor doubling time. Overall, our results serve as a foundation for further evaluation of phenformin as a therapeutic agent in pancreatic cancer. Clin Cancer Res; 23(18); 5639-47. ©2017 AACR.
Collapse
Affiliation(s)
- N V Rajeshkumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shinichi Yabuuchi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shweta G Pai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth De Oliveira
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jurre J Kamphorst
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, United Kingdom
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, New Jersey
| | - Héctor Tejero
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Manuel Hidalgo
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Division of Hematology-Oncology, Rosenberg Clinical Cancer Center, Beth Israel Deaconess Medical Center (BIDMC), Boston, Massachusetts
| | - Anirban Maitra
- Department of Pathology and Translational Molecular Pathology, Sheikh Ahmad Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chi V Dang
- Abramson Cancer Center, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania.
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
125
|
Results from 11C-metformin-PET scans, tissue analysis and cellular drug-sensitivity assays questions the view that biguanides affects tumor respiration directly. Sci Rep 2017; 7:9436. [PMID: 28842630 PMCID: PMC5573362 DOI: 10.1038/s41598-017-10010-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 08/02/2017] [Indexed: 11/16/2022] Open
Abstract
The anti-diabetic biguanide drugs metformin (METF) and phenformin (PHEN) may have anti-cancer effects. Biguanides suppress plasma growth factors, but nonetheless, the view that these mitochondrial inhibitors accumulate in tumor tissue to an extent that leads to severe energetic stress or alleviation of hypoxia-induced radioresistance is gaining ground. Our cell studies confirm that biguanides inhibits cell proliferation by targeting respiration, but only at highly suprapharmacological concentrations due to low drug retention. Biodistribution/PET studies of 11C-labeled metformin (11C-METF) revealed that plasma bioavailability remained well below concentrations with metabolic/anti-proliferative in vitro effects, following a high oral dose. Intraperitoneal administration resulted in higher drug concentrations, which affected metabolism in normal organs with high METF uptake (e.g., kidneys), but tumor drug retention peaked at low levels comparable to plasma levels and hypoxia was unaffected. Prolonged intraperitoneal treatment reduced tumor growth in two tumor models, however, the response did not reflect in vitro drug sensitivity, and tumor metabolism and hypoxia was unaffected. Our results do not support that direct inhibition of tumor cell respiration is responsible for reduced tumor growth, but future studies using 11C-METF-PET are warranted, preferably in neoplasia’s originating from tissue with high drug transport capacity, to investigate the controversial idea of direct targeting.
Collapse
|
126
|
Zhao Y, Gong C, Wang Z, Zhang J, Wang L, Zhang S, Cao J, Tao Z, Li T, Wang B, Hu X. A randomized phase II study of aromatase inhibitors plus metformin in pre-treated postmenopausal patients with hormone receptor positive metastatic breast cancer. Oncotarget 2017; 8:84224-84236. [PMID: 29137418 PMCID: PMC5663590 DOI: 10.18632/oncotarget.20478] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/19/2017] [Indexed: 12/15/2022] Open
Abstract
Background Everolimus significantly improves progression-free survival (PFS) and has been approved to use in aromatase inhibitor pretreated patients with hormone receptor positive advanced breast cancer. Metformin has been shown to inhibit mTOR pathway, with more favorable safety profile, leading to this hypothesis-generating trial to assess whether metformin enhances the efficacy of aromatase inhibitors. Methods 60 postmenopausal women with hormone receptor positive locally advanced or metastatic breast cancer were randomly assigned 1:1 to aromatase inhibitor (exemestane 25mg/d or letrozole 2.5mg/d depending on the most recent treatment) plus metformin (0.5g bid, orally) or placebo. The primary endpoint was PFS, and secondary endpoints were objective response rate, clinical benefit rate, overall survival and safety. Results Median PFS was 4.7 months in the combination group and 6.0 months in the control group (hazard ratio, 1.2; 95% confidence interval [CI], 0.7 to 2.1; P =0.48). ORR was 6.7% in the combination group and 0% in the control group (odds ratio for ORR not available; P =0.99), and CBR was 33.3% and 50.0%, respectively (OR for CBR 0.5; 95% CI, 0.2 to 1.4; P=0.15). No significant difference in overall survival was observed between the combination and control groups (median OS, 30.9 vs. 32.4 months; P = 0.81). Subgroup analyses didn't find any specific population favoring the combination treatment. No substantial difference in incidence or severity of adverse events was seen between the two treatment groups. Conclusion This randomized phase II clinical trial failed to show an improved efficacy with the addition of metformin to endocrine therapy, although with excellent tolerability.
Collapse
Affiliation(s)
- Yannan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chengcheng Gong
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhonghua Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sheng Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ting Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
127
|
Liang X, Kong P, Wang J, Xu Y, Gao C, Guo G. Effects of metformin on proliferation and apoptosis of human megakaryoblastic Dami and MEG-01 cells. J Pharmacol Sci 2017; 135:14-21. [PMID: 28927780 DOI: 10.1016/j.jphs.2017.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/11/2017] [Accepted: 08/02/2017] [Indexed: 01/07/2023] Open
Abstract
Metformin has received increasing attention for its potential anticancer activity against certain human leukemia cells, but its effects on human megakaryoblastic cells are unclear. This study aimed to investigate the effects of metformin on proliferation and apoptosis of human megakaryoblastic cells (Dami and MEG-01) and the underlying molecular mechanisms. CCK8 assay was employed to measure cell proliferation. Flow cytometry was adopted to detect cell apoptosis. Western blot was further employed to measure apoptosis-related proteins. In Dami and MEG-01 cells, metformin significantly inhibited proliferation and promoted apoptosis in a dose- and time-dependent manner, and metformin (4 mM) was selected for subsequent experiments. Metformin inhibited ERK1/2, JNK, and PI3K/Akt, but activated p38 pathway in these two cells. Moreover, inhibition of ERK1/2, JNK or PI3K/Akt pathway alone induced cell apoptosis compared to the control group. The combination of specific inhibitors of ERK1/2, JNK or PI3K/Akt pathway and metformin further promoted cell apoptosis and the up-regulation of p21, Bax, Bad, cleaved caspase-3 and -9 as well as the down-regulation of Bcl-2 mediated by metformin alone, but inhibition of p38 pathway exhibited the opposite results. These findings support the possibility of metformin treatment as a new therapeutic strategy against acute megakaryoblastic leukemia (AMKL).
Collapse
Affiliation(s)
- Xue Liang
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Peiyan Kong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Jin Wang
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Yulin Xu
- Department of Hematology, No. 150 Central Hospital of Chinese People's Liberation Army, Luoyang, China
| | - Chunfang Gao
- Institute of Anal-Colorectal Surgery, No. 150 Central Hospital of Chinese People's Liberation Army, No. 2 Huaxia Road, Luoyang, 471000, China.
| | - Guozhen Guo
- Department of Radiation Medicine, School of Public Health, The Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
128
|
Zhang Y, Feng X, Li T, Yi E, Li Y. Metformin synergistic pemetrexed suppresses non-small-cell lung cancer cell proliferation and invasion in vitro. Cancer Med 2017; 6:1965-1975. [PMID: 28719077 PMCID: PMC5548881 DOI: 10.1002/cam4.1133] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 05/03/2017] [Accepted: 05/26/2017] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to investigate whether metformin in combination with pemetrexed has an effect on the treatment of non-small-cell lung cancer (NSCLC) models and to explore the related molecular mechanism. The half maximal inhibitory concentration (IC50) and combination index (CI) of metformin and pemetrexed were detected by the CCK8 assay to assess the antiproliferative and therapeutic effects of the two-drug combination. Flow cytometry (FCM) and invasion assays were used to estimate the variation in apoptosis rate and invasion ability of the differently treated NSCLC cell lines. Apoptotic markers were detected by western blotting to validate the data related to the antiproliferation and proapoptosis effects. Metformin monotherapy inhibited the growth of NSCLC cell lines and reduced the invasion ability to different degrees compared with the control groups (P < 0.05). Metformin in combination with pemetrexed produced a synergistic effect (CI < 0.90) compared with the two drugs in monotherapy in the three tested NSCLC cell lines. Metformin in combination with pemetrexed significantly increased the cell numbers of HCC827 cells at S phase (P < 0.001), and the combination therapy had no influence on the A549 and H1975 cell lines. We found that combining metformin with pemetrexed induced more cell apoptosis than metformin or pemetrexed used alone (P < 0.05), which was validated by the apoptotic markers. These results demonstrate that the combination of metformin and pemetrexed has a synergistic effect on the treatment of NSCLC cell lines by inducing apoptosis or blocking the cell cycle. Our data indicate that the combination of metformin and pemetrexed could have beneficial antitumor effects on NSCLC cells in vitro.
Collapse
Affiliation(s)
- Yan Zhang
- Departments of Respiratory MedicineQilu hospital of Shandong UniversityJinanShandongChina
| | - Xiuli Feng
- Departments of Respiratory MedicineQilu hospital of Shandong UniversityJinanShandongChina
- Departments of Respiratory MedicinePeople's Hospital of QingzhouWeifangShandongChina
| | - Tao Li
- Departments of Respiratory MedicineQilu hospital of Shandong UniversityJinanShandongChina
| | - Erpan Yi
- Departments of Respiratory MedicineQilu hospital of Shandong UniversityJinanShandongChina
| | - Yu Li
- Departments of Respiratory MedicineQilu hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
129
|
Chen K, Qian W, Jiang Z, Cheng L, Li J, Sun L, Zhou C, Gao L, Lei M, Yan B, Cao J, Duan W, Ma Q. Metformin suppresses cancer initiation and progression in genetic mouse models of pancreatic cancer. Mol Cancer 2017; 16:131. [PMID: 28738823 PMCID: PMC5525317 DOI: 10.1186/s12943-017-0701-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-associated mortality worldwide with an overall five-year survival rate less than 7%. Accumulating evidence has revealed the cancer preventive and therapeutic effects of metformin, one of the most widely prescribed medications for type 2 diabetes mellitus. However, its role in pancreatic cancer is not fully elucidated. Herein, we aimed to further study the preventive and therapeutic effects of metformin in genetically engineered mouse models of pancreatic cancer. Methods LSL-KrasG12D/+; Pdx1-Cre (KC) mouse model was established to investigate the effect of metformin in pancreatic tumorigenesis suppression; LSL-KrasG12D/+; Trp53fl/+; Pdx1-Cre (KPC) mouse model was used to evaluate the therapeutic efficiency of metformin in PDAC. Chronic pancreatitis was induced in KC mice by peritoneal injection of cerulein. Results Following metformin treatment, pancreatic acinar-to-ductal metaplasia (ADM) and mouse pancreatic intraepithelial neoplasia (mPanIN) were decreased in KC mice. Chronic pancreatitis induced a stroma-rich and duct-like structure and increased the formation of ADM and mPanIN lesions, in line with an increased cytokeratin 19 (CK19)-stained area. Metformin treatment diminished chronic pancreatitis-mediated ADM and mPanIN formation. In addition, it alleviated the percent area of Masson’s trichrome staining, and decreased the number of Ki67-positive cells. In KPC mice, metformin inhibited tumor growth and the incidence of abdominal invasion. More importantly, it prolonged the overall survival. Conclusions Metformin inhibited pancreatic cancer initiation, suppressed chronic pancreatitis-induced tumorigenesis, and showed promising therapeutic effect in PDAC. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0701-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ke Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liang Cheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Jie Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Liankang Sun
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Luping Gao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Meng Lei
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Bin Yan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Junyu Cao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
130
|
Siddappa G, Kulsum S, Ravindra DR, Kumar VV, Raju N, Raghavan N, Sudheendra HV, Sharma A, Sunny SP, Jacob T, Kuruvilla BT, Benny M, Antony B, Seshadri M, Lakshminarayan P, Hicks W, Suresh A, Kuriakose MA. Curcumin and metformin-mediated chemoprevention of oral cancer is associated with inhibition of cancer stem cells. Mol Carcinog 2017; 56:2446-2460. [DOI: 10.1002/mc.22692] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/23/2017] [Accepted: 06/13/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Gangotri Siddappa
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- Head and Neck Oncology; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Safeena Kulsum
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- School of Biosciences and Technology; VIT University; Vellore Tamil Nadu India
| | - Doddathimmasandra Ramanjanappa Ravindra
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Vinay V. Kumar
- Department of Oral Surgery; Dr. BR Ambedkar Medical College; Bangalore Karnataka India
| | - Nalini Raju
- Department of Histopathology; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Nisheena Raghavan
- Department of Histopathology; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Holalugunda Vittalamurthy Sudheendra
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Anupam Sharma
- Stem Cell Research Laboratory; GROW Laboratory; Narayana Nethralaya; Narayana Health; Bangalore Karnataka India
| | - Sumsum P. Sunny
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- Head and Neck Oncology; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
| | - Tina Jacob
- Department of Oral Pathology and Microbiology; Bangalore Institute of Dental Sciences; Bangalore Karnataka India
| | | | - Merina Benny
- Arjuna Natural Extracts Ltd.; Alwaye, Kochi Kerala India
| | - Benny Antony
- Arjuna Natural Extracts Ltd.; Alwaye, Kochi Kerala India
| | - Mukund Seshadri
- Department of Pharmacology and Therapeutics; Roswell Park Cancer Institute; Buffalo New York
- Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program; Roswell Park Cancer Institute; Buffalo New York
| | - Padma Lakshminarayan
- Department of Pharmacology; Dr. BR Ambedkar Medical College; Bangalore Karnataka India
| | - Wesley Hicks
- Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program; Roswell Park Cancer Institute; Buffalo New York
- Department of Head and Neck/Plastic & Reconstructive Surgery; Roswell Park Cancer Institute; Buffalo New York
| | - Amritha Suresh
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program; Roswell Park Cancer Institute; Buffalo New York
| | - Moni A. Kuriakose
- Integrated Head and Neck Oncology Research Program, DSRG-5; Mazumdar Shaw Centre for Translational Research; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- Head and Neck Oncology; Mazumdar Shaw Medical Centre; Narayana Health; Bangalore Karnataka India
- Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program; Roswell Park Cancer Institute; Buffalo New York
| |
Collapse
|
131
|
Tang JC, An R, Jiang YQ, Yang J. Effects and Mechanisms of Metformin on the Proliferation of Esophageal Cancer Cells In Vitro and In Vivo. Cancer Res Treat 2017; 49:778-789. [PMID: 27857021 PMCID: PMC5512357 DOI: 10.4143/crt.2015.485] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 10/21/2016] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The purpose of this study was to observe the effects of metformin on human esophageal cancer cell and to investigate its possible mechanisms. MATERIALS AND METHODS Cell viability was detected by using a Cell Counting Kit-8, while cell cycle and apoptosis were assessed by flow cytometry and western blot was used to measure the expression of the related proteins. RNAi was used to knockout pyruvate kinase muscle isozyme 2 (PKM2). An Eca109 tumor model was established to evaluate the antitumor effect in vivo. Immunohistochemistry was determined based on the expression of PKM2 and Bim in tumor tissues. Tunnel was used to assess tumor cell apoptosis. RESULTS Esophageal cancer cells viability was reduced after metformin treatment. The cell cycle was arrested in the G0/G1 phase, apoptosis was induced, caspase 3 was activated, caspase 9 was downregulated, and the pro-apoptotic protein Bim increased. Further study revealed that metformin could suppress the expression of insulin-like growth factor 1 receptor and its downstream proteins, phosphoinositide 3-kinase (PI3K), protein kinase B (AKT/PKB), phosphorylation of AKT (pAKT), mammalian target of rapamycin (mTOR), p70S6K, and PKM2. Insulin-like growth factor 1 partly reversed metfromin-induced apoptosis and attenuated the repression effect of metfomin to PI3K, pAKT, and PKM2. Knockout PKM2 resulted in the activation of caspase 3, down-regulation of caspase 9, and increased expression of Bim. In the Eca109 xenograft model, metformin significantly reduced tumor growth. Furthermore, we found that metformin treatment increased the rate of apoptosis, down-regulation of PKM2, and up-regulation of Bim in tumor tissues. CONCLUSION Metformin restrained esophageal cancer cell proliferation partly by suppressing the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Jian-Cai Tang
- Department of Biochemistry, North of Sichuan Medical University, Nanchong, China
| | - Rui An
- School of Basic Medical Sciences, North of Sichuan Medical University, Nanchong, China
| | - Yi-Qing Jiang
- School of Basic Medical Sciences, North of Sichuan Medical University, Nanchong, China
| | - Jian Yang
- Pathogenic Biology and Immunology Experiment Teaching Center, North of Sichuan Medical University, Nanchong, China
| |
Collapse
|
132
|
Mohamed Suhaimi NA, Phyo WM, Yap HY, Choy SHY, Wei X, Choudhury Y, Tan WJ, Tan LAPY, Foo RSY, Tan SHS, Tiang Z, Wong CF, Koh PK, Tan MH. Metformin Inhibits Cellular Proliferation and Bioenergetics in Colorectal Cancer Patient-Derived Xenografts. Mol Cancer Ther 2017; 16:2035-2044. [PMID: 28533437 DOI: 10.1158/1535-7163.mct-16-0793] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 02/06/2023]
Abstract
There is increasing preclinical evidence suggesting that metformin, an antidiabetic drug, has anticancer properties against various malignancies, including colorectal cancer. However, the majority of evidence, which was derived from cancer cell lines and xenografts, was likely to overestimate the benefit of metformin because these models are inadequate and require supraphysiologic levels of metformin. Here, we generated patient-derived xenograft (PDX) lines from 2 colorectal cancer patients to assess the properties of metformin and 5-fluorouracil (5-FU), the first-line drug treatment for colorectal cancer. Metformin (150 mg/kg) as a single agent inhibits the growth of both PDX tumors by at least 50% (P < 0.05) when administered orally for 24 days. In one of the PDX models, metformin given concurrently with 5-FU (25 mg/kg) leads to an 85% (P = 0.054) growth inhibition. Ex vivo culture of organoids generated from PDX demonstrates that metformin inhibits growth by executing metabolic changes to decrease oxygen consumption and activating AMPK-mediated pathways. In addition, we also performed genetic characterizations of serial PDX samples with corresponding parental tissues from patients using next-generation sequencing (NGS). Our pilot NGS study demonstrates that PDX represents a useful platform for analysis in cancer research because it demonstrates high fidelity with parental tumor. Furthermore, NGS analysis of PDX may be useful to determine genetic identifiers of drug response. This is the first preclinical study using PDX and PDX-derived organoids to investigate the efficacy of metformin in colorectal cancer. Mol Cancer Ther; 16(9); 2035-44. ©2017 AACR.
Collapse
Affiliation(s)
| | - Wai Min Phyo
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Hao Yun Yap
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | | | - Xiaona Wei
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Yukti Choudhury
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | - Wai Jin Tan
- Institute of Bioengineering and Nanotechnology Singapore, Singapore
| | | | - Roger Sik Yin Foo
- Genome Institute of Singapore, Singapore.,Cardiovascular Research Institute, National University Health Systems, Singapore
| | | | | | | | | | - Min-Han Tan
- Institute of Bioengineering and Nanotechnology Singapore, Singapore. .,Concord Cancer Hospital Singapore, Singapore.,National Cancer Centre Singapore, Singapore
| |
Collapse
|
133
|
Lin J, Gill A, Zahm SH, Carter CA, Shriver CD, Nations JA, Anderson WF, McGlynn KA, Zhu K. Metformin use and survival after non-small cell lung cancer: A cohort study in the US Military health system. Int J Cancer 2017; 141:254-263. [PMID: 28380674 DOI: 10.1002/ijc.30724] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/17/2017] [Accepted: 03/09/2017] [Indexed: 12/28/2022]
Abstract
Research suggests that metformin may be associated with improved survival in cancer patients with type II diabetes. This study assessed whether metformin use after non-small cell lung cancer (NSCLC) diagnosis is associated with overall survival among type II diabetic patients with NSCLC in the U.S. military health system (MHS). The study included 636 diabetic patients with histologically confirmed NSCLC diagnosed between 2002 and 2007, identified from the linked database from the Department of Defense's Central Cancer Registry (CCR) and the Military Health System Data Repository (MDR). Time-dependent multivariate Cox proportional hazards models were used to assess the association between metformin use and overall survival during follow-up. Among the 636 patients, 411 died during the follow-up. The median follow-up time was 14.6 months. Increased post-diagnosis cumulative use (per 1 year of use) conferred a significant reduction in mortality (adjusted hazard ratio (HR) = 0.76; 95% CI = 0.65-0.88). Further analysis by duration of use revealed that compared to non-users, the lowest risk reduction occurred among patients with the longest duration of use (i.e. use for more than 2 years) (HR = 0.19; 95% CI = 0.09-0.40). Finally, the reduced mortality was particularly observed only among patients who also used metformin before lung cancer diagnosis and among patients at early stage of diagnosis. Prolonged duration of metformin use in the study population was associated with improved survival, especially among early stage patients. Future research with a larger number of patients is warranted.
Collapse
Affiliation(s)
- Jie Lin
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD
| | - Abegail Gill
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD
| | - Shelia H Zahm
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Corey A Carter
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD
| | - Craig D Shriver
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD.,Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD
| | - Joel A Nations
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD
| | - William F Anderson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Kangmin Zhu
- John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD.,Department of Preventive Medicine and Biostatistics, Uniformed Services University of Health Sciences, Bethesda, MD
| |
Collapse
|
134
|
Morgillo F, Fasano M, Della Corte CM, Sasso FC, Papaccio F, Viscardi G, Esposito G, Di Liello R, Normanno N, Capuano A, Berrino L, Vicidomini G, Fiorelli A, Santini M, Ciardiello F. Results of the safety run-in part of the METAL (METformin in Advanced Lung cancer) study: a multicentre, open-label phase I-II study of metformin with erlotinib in second-line therapy of patients with stage IV non-small-cell lung cancer. ESMO Open 2017; 2:e000132. [PMID: 28761738 PMCID: PMC5519802 DOI: 10.1136/esmoopen-2016-000132] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 01/14/2023] Open
Abstract
Purpose Our previous works demonstrated the ability of metformin to revert resistance to gefitinib, a selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, in non-small-cell lung cancer (NSCLC) EGFR/LKB1 wild-type (WT) cell lines. However, the optimal dose of metformin to be used in non-diabetic patients still remains to be defined. The phase I–II trial METformin in Advanced Lung cancer (METAL) was designed to identify the maximum tolerated dose and to evaluate safety and activity of metformin combined with erlotinib in second-line treatment of patients with stage IV NSCLC, whose tumours harbour the WT EGFR gene. Patients and methods We report results from the safety run-in part designed to detect acute toxicities, to study pharmacokinetics and to identify the recommended phase II dose (RPD) to be used for the following phase of the study. In the run-in phase, metformin treatment was administered according to a dose escalation scheme and, subsequently, combined with erlotinib. Results Twelve patients were enrolled. Common adverse events were diarrhoea, decreased appetite, abdominal pain, vomiting and skin toxicity, mostly reversible with symptomatic medical treatment. Dose-limiting toxicities were vomiting and diarrhoea registered in the initial cohort receiving metformin 2000 mg plus erlotinib at 150 mg die, which was declared the maximum administered dose. Only one of nine patients treated at the next lower dose of 1500 mg of metformin plus erlotinib at 150 mg experienced G3 gastrointestinal toxicity. Metformin plasma-concentration profile confirmed the trend already observed in non-diabetic population. Glycemic profiles showed stability of the blood glucose level within the physiological range for non-diabetic subjects. At a follow-up of 30 weeks, six (50%) patients experienced a disease control (5 SD and 1 partial response). Conclusions The RP2D of metformin dose was defined at 1500 mg/day to be combined with erlotinib 150 mg. Trial registration number EudraCT number: 2014-000349-59.
Collapse
Affiliation(s)
- Floriana Morgillo
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Morena Fasano
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Carminia Maria Della Corte
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Medicina interna, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Federica Papaccio
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Giuseppe Viscardi
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Giovanna Esposito
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Raimondo Di Liello
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Nicola Normanno
- Biologia cellulare e bioterapie, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Annalisa Capuano
- Dipartimento di Medicina Sperimentale, Sezione di Farmacologia, Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Liberato Berrino
- Dipartimento di Medicina Sperimentale, Sezione di Farmacologia, Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Giovanni Vicidomini
- Chirurgia toracica, Dipartimento di Scienze Cardio-Toraciche e Respiratorie, Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Alfonso Fiorelli
- Chirurgia toracica, Dipartimento di Scienze Cardio-Toraciche e Respiratorie, Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Mario Santini
- Chirurgia toracica, Dipartimento di Scienze Cardio-Toraciche e Respiratorie, Seconda Universita degli Studi di Napoli, Naples, Italy
| | - Fortunato Ciardiello
- Oncologia medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Seconda Universita degli Studi di Napoli, Naples, Italy
| |
Collapse
|
135
|
Liu SF, Kuo HC, Lin MC, Ho SC, Tu ML, Chen YM, Chen YC, Fang WF, Wang CC, Liu GH. Inhaled corticosteroids have a protective effect against lung cancer in female patients with chronic obstructive pulmonary disease: a nationwide population-based cohort study. Oncotarget 2017; 8:29711-29721. [PMID: 28412726 PMCID: PMC5444697 DOI: 10.18632/oncotarget.15386] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 01/31/2017] [Indexed: 01/01/2023] Open
Abstract
Whether the use of inhaled corticosteroids (ICS) protects patients with chronic obstructive pulmonary disease (COPD) from lung cancer remains undetermined. In this retrospective nationwide population-based cohort study, we extracted data of 13,686 female COPD patients (ICS users, n = 1,290, ICS non-users, n = 12,396) diagnosed between 1997 and 2009 from the Taiwan's National Health Insurance database. These patients were followed-up until 2011, and lung cancer incidence was determined. Cox regression analysis was used to estimate hazard ratios (HRs) for lung cancer incidence. The time to lung cancer diagnosis was significantly different between ICS users and non-users (10.75 vs. 9.68 years, P < 0.001). Per 100,000 person-years, the lung cancer incidence rate was 235.92 for non-users and 158.67 for users [HR = 0.70 (95% confidence interval {CI}: 0.46-1.09)]. After adjusting for patients' age, income, and comorbidities, a cumulative ICS dose > 39.48 mg was significantly associated with a lower risk of lung cancer [ICS users > 39.48 mg, HR = 0.45 (95% CI: 0.21-0.96)]. Age ≥ 60 years, pneumonia, diabetes mellitus, and hypertension decreased lung cancer risk, whereas pulmonary tuberculosis increased the risk. Our results suggest that ICS have a potential role in lung cancer prevention among female COPD patients.
Collapse
Affiliation(s)
- Shih-Feng Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ho-Chang Kuo
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Chen Ho
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Mei-Lien Tu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Mu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yung-Che Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Feng Fang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chin-Chou Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Guan-Heng Liu
- Department of Senior High School, Li-Chih Valuable School, Kaohsiung, Taiwan
| |
Collapse
|
136
|
Targeting metabolism and AMP-activated kinase with metformin to sensitize non-small cell lung cancer (NSCLC) to cytotoxic therapy: translational biology and rationale for current clinical trials. Oncotarget 2017; 8:57733-57754. [PMID: 28915708 PMCID: PMC5593680 DOI: 10.18632/oncotarget.17496] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/19/2017] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the most fatal malignancy worldwide, in part, due to high resistance to cytotoxic therapy. There is need for effective chemo-radio-sensitizers in lung cancer. In recent years, we began to understand the modulation of metabolism in cancer and its importance in tumor progression and survival after cytotoxic therapy. The activity of biosynthetic pathways, driven by the Growth Factor Receptor/Ras/PI3k/Akt/mTOR pathway, is balanced by the energy stress sensor pathway of LKB1/AMPK/p53. AMPK responds both to metabolic and genotoxic stress. Metformin, a well-tolerated anti-diabetic agent, which blocks mitochondria oxidative phosphorylation complex I, became the poster child agent to elicit AMPK activity and tumor suppression. Metformin sensitizes NSCLC models to chemotherapy and radiation. Here, we discuss the rationale for targeting metabolism, the evidence supporting metformin as an anti-tumor agent and adjunct to cytotoxic therapy in NSCLC and we review retrospective evidence and on-going clinical trials addressing this concept.
Collapse
|
137
|
Wokoun U, Hellriegel M, Emons G, Gründker C. Co-treatment of breast cancer cells with pharmacologic doses of 2-deoxy-D-glucose and metformin: Starving tumors. Oncol Rep 2017; 37:2418-2424. [PMID: 28350075 DOI: 10.3892/or.2017.5491] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/26/2017] [Indexed: 11/05/2022] Open
Abstract
A characteristic of tumor cells is the increased aerobic glycolysis for energy production. Thus, inhibition of glycolysis represents a selective therapeutic option. It has been shown that glycolysis inhibitor 2-deoxy-D-glucose (2DG) induces apoptotic cell death in different tumor entities. In addition, the antitumor activity of the anti-diabetic drug metformin has been demonstrated. In the present study, we aimed to ascertain whether the combination of pharmacologic doses of 2DG with metformin increases the antitumor efficacy. Cell viability of MDA-MB-231 and HCC1806 triple-negative breast cancer (TNBC) cells treated without or with 2DG or with metformin alone or with the combination of both agents was measured using Alamar Blue assay. Induction of apoptosis was quantified by measurement of the loss of mitochondrial membrane potential and cleavage of PARP. Treatment of breast cancer cells with glycolysis inhibitor 2DG or with the anti-diabetic drug metformin resulted in a significant decrease in cell viability and an increase in apoptosis. Treatment with 2DG in combination with metformin resulted in significantly reduced viability compared with the single agent treatments. The observed reduction in viability was due to induction of apoptosis. In addition, in regards to apoptosis induction a stronger effect in the case of co-treatment compared with single agent treatments was observed. The glycolytic phenotype of human breast cancer cells can be targeted for therapeutic intervention. Co-treatment with doses of the glycolysis inhibitor 2DG and anti-diabetic drug metformin is tolerable in humans and may be a suitable therapy for human breast cancers.
Collapse
Affiliation(s)
- Ulrike Wokoun
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Martin Hellriegel
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| | - Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, D-37075 Göttingen, Germany
| |
Collapse
|
138
|
Howell JJ, Hellberg K, Turner M, Talbott G, Kolar MJ, Ross DS, Hoxhaj G, Saghatelian A, Shaw RJ, Manning BD. Metformin Inhibits Hepatic mTORC1 Signaling via Dose-Dependent Mechanisms Involving AMPK and the TSC Complex. Cell Metab 2017; 25:463-471. [PMID: 28089566 PMCID: PMC5299044 DOI: 10.1016/j.cmet.2016.12.009] [Citation(s) in RCA: 305] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/02/2016] [Accepted: 12/10/2016] [Indexed: 02/08/2023]
Abstract
Metformin is the most widely prescribed drug for the treatment of type 2 diabetes. However, knowledge of the full effects of metformin on biochemical pathways and processes in its primary target tissue, the liver, is limited. One established effect of metformin is to decrease cellular energy levels. The AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) are key regulators of metabolism that are respectively activated and inhibited in acute response to cellular energy depletion. Here we show that metformin robustly inhibits mTORC1 in mouse liver tissue and primary hepatocytes. Using mouse genetics, we find that at the lowest concentrations of metformin that inhibit hepatic mTORC1 signaling, this inhibition is dependent on AMPK and the tuberous sclerosis complex (TSC) protein complex (TSC complex). Finally, we show that metformin profoundly inhibits hepatocyte protein synthesis in a manner that is largely dependent on its ability to suppress mTORC1 signaling.
Collapse
Affiliation(s)
- Jessica J Howell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kristina Hellberg
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Marc Turner
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - George Talbott
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Matthew J Kolar
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Debbie S Ross
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Gerta Hoxhaj
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
139
|
Lei Y, Yi Y, Liu Y, Liu X, Keller ET, Qian CN, Zhang J, Lu Y. Metformin targets multiple signaling pathways in cancer. CHINESE JOURNAL OF CANCER 2017; 36:17. [PMID: 28126011 PMCID: PMC5270304 DOI: 10.1186/s40880-017-0184-9] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022]
Abstract
Metformin, an inexpensive and well-tolerated oral agent commonly used in the first-line treatment of type 2 diabetes, has become the focus of intense research as a candidate anticancer agent. Here, we discuss the potential of metformin in cancer therapeutics, particularly its functions in multiple signaling pathways, including AMP-activated protein kinase, mammalian target of rapamycin, insulin-like growth factor, c-Jun N-terminal kinase/mitogen-activated protein kinase (p38 MAPK), human epidermal growth factor receptor-2, and nuclear factor kappaB pathways. In addition, cutting-edge targeting of cancer stem cells by metformin is summarized.
Collapse
Affiliation(s)
- Yong Lei
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Yanhua Yi
- School for International Education, Guangxi Medical University, Nanning, 530021, Guangxi, P. R. China
| | - Yang Liu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Xia Liu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China.,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China
| | - Evan T Keller
- Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chao-Nan Qian
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jian Zhang
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China. .,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China. .,Department of Urology and Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Yi Lu
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, 530021, Guangxi, P. R. China. .,Center for Translational Medicine, Guangxi Medical University, 14th Floor, Pharmacology and Biomedical Sciences Building, No. 22 Shuangyong Road, Nanning, 530021, Guangxi, P. R. China.
| |
Collapse
|
140
|
Seabloom DE, Galbraith AR, Haynes AM, Antonides JD, Wuertz BR, Miller WA, Miller KA, Steele VE, Miller MS, Clapper ML, O'Sullivan MG, Ondrey FG. Fixed-Dose Combinations of Pioglitazone and Metformin for Lung Cancer Prevention. Cancer Prev Res (Phila) 2017; 10:116-123. [PMID: 28052934 DOI: 10.1158/1940-6207.capr-16-0232] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 12/01/2016] [Accepted: 12/11/2016] [Indexed: 01/06/2023]
Abstract
Combination treatment with pioglitazone and metformin is utilized clinically in the treatment of type II diabetes. Treatment with this drug combination reduced the development of aerodigestive cancers in this patient population. Our goal is to expand this treatment into clinical lung cancer chemoprevention. We hypothesized that dietary delivery of metformin/pioglitazone would prevent lung adenoma formation in A/J mice in a benzo[a]pyrene (B[a]P)-induced carcinogenesis model while modulating chemoprevention and anti-inflammatory biomarkers in residual adenomas. We found that metformin (500 and 850 mg/kg/d) and pioglitazone (15 mg/kg/d) produced statistically significant decreases in lung adenoma formation both as single-agent treatments and in combination, compared with untreated controls, after 15 weeks. Treatment with metformin alone and in combination with pioglitazone resulted in statistically significant decreases in lung adenoma formation at both early- and late-stage interventions. Pioglitazone alone resulted in significant decreases in adenoma formation only at early treatment intervention. We conclude that oral metformin is a viable chemopreventive treatment at doses ranging from 500 to 1,000 mg/kg/d. Pioglitazone at 15 mg/kg/d is a viable chemopreventive agent at early-stage interventions. Combination metformin and pioglitazone performed equal to metformin alone and better than pioglitazone at 15 mg/kg/d. Because the drugs are already FDA-approved, rapid movement to human clinical studies is possible. Cancer Prev Res; 10(2); 116-23. ©2017 AACR.
Collapse
Affiliation(s)
- Donna E Seabloom
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,AeroCore Inhalation Testing, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | | | - Anna M Haynes
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,AeroCore Inhalation Testing, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota
| | | | - Beverly R Wuertz
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,AeroCore Inhalation Testing, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota.,Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Wendy A Miller
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Kimberly A Miller
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Vernon E Steele
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Mark Steven Miller
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | | | - M Gerard O'Sullivan
- Comparative Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Frank G Ondrey
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota. .,AeroCore Inhalation Testing, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota.,Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
141
|
Triggle CR, Ding H. Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiol (Oxf) 2017; 219:138-151. [PMID: 26680745 DOI: 10.1111/apha.12644] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/18/2015] [Accepted: 12/11/2015] [Indexed: 12/28/2022]
Abstract
Metformin, a synthetic dimethyl biguanide, has been in clinical use for over 55 years, and today is considered the first-choice drug for the treatment of type 2 diabetes used by an estimated 125 million people worldwide. Metformin is orally effective, not metabolized, excreted unchanged by the kidney, relatively free of side effects and well tolerated by the majority of patients. Of importance is that the United Kingdom Prospective Diabetes Study 20-year study of type 2 diabetics, completed in 1998, compared patients treated with insulin, sulfonylureas and metformin and concluded that metformin provided vascular protective actions. Cardiovascular disease is the primary basis for the high morbidity and mortality that is associated with diabetes and that metformin proved to be protective resulted in a dramatic increase in its use. The vascular protective actions of metformin are thought to be secondary to the antihyperglycaemic effects of metformin that are mediated via activation of AMP kinase and subsequent inhibition of hepatic gluconeogenesis, fatty acid oxidation as well as an insulin sensitizing action in striated muscle and adipose tissue. As reflected by a number of clinical studies, patients treated with metformin also have improvement in endothelial function as measured by the use of plethysmography and measurement of flow-mediated vasodilatation. These data as well as data from animal studies are supportive that metformin has a direct protective action on the vascular endothelium. In this review article, we discuss the pharmacology of metformin and critique the literature as to its cellular sites and mechanism(s) of action.
Collapse
Affiliation(s)
- C. R. Triggle
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| | - H. Ding
- Departments of Pharmacology and Medical Education; Weill Cornell Medicine in Qatar; Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
142
|
Zou J, Hong L, Luo C, Li Z, Zhu Y, Huang T, Zhang Y, Yuan H, Hu Y, Wen T, Zhuang W, Cai B, Zhang X, Huang J, Cheng J. Metformin inhibits estrogen-dependent endometrial cancer cell growth by activating the AMPK-FOXO1 signal pathway. Cancer Sci 2016; 107:1806-1817. [PMID: 27636742 PMCID: PMC5198961 DOI: 10.1111/cas.13083] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 02/05/2023] Open
Abstract
Metformin is an oral biguanide commonly used for treating type II diabetes and has recently been reported to possess antiproliferative properties that can be exploited for the prevention and treatment of a variety of cancers. The mechanisms underlying this effect have not been fully elucidated. Our study shows a marked loss of AMP-activated protein kinase (AMPK) phosphorylation and nuclear human Forkhead box O1 (FOXO1) protein in estrogen-dependent endometrial cancer (EC) tumors compared to normal control endometrium. Metformin treatment suppressed EC cell growth in a time-dependent manner in vitro; this effect was cancelled by cotreatment with an AMPK inhibitor, compound C. Metformin decreased FOXO1 phosphorylation and increased FOXO1 nuclear localization in Ishikawa and HEC-1B cells, with non-significant increase in FOXO1 mRNA expression. Moreover, compound C blocked the metformin-induced changes of FOXO1 and its phosphorylation protein, suggesting that metformin upregulated FOXO1 activity by AMPK activation. Similar results were obtained after treatment with insulin. In addition, transfection with siRNA for FOXO1 cancelled metformin-inhibited cell growth, indicating that FOXO1 mediated metformin to inhibit EC cell proliferation. A xenograft mouse model further revealed that metformin suppressed HEC-1B tumor growth, accompanied by downregulated ki-67 and upregulated AMPK phosphorylation and nuclear FOXO1 protein. Taken together, these data provide a novel mechanism of antineoplastic effect for metformin through the regulation of FOXO1, and suggest that the AMPK-FOXO1 pathway may be a therapeutic target to the development of new antineoplastic drugs.
Collapse
Affiliation(s)
- Jingfang Zou
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Liangli Hong
- Departments of PathologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Chaohuan Luo
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Zhi Li
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yuzhang Zhu
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Tianliang Huang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yongneng Zhang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Huier Yuan
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yaqiu Hu
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Tengfei Wen
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Wanling Zhuang
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Bozhi Cai
- The Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xin Zhang
- The Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Jiexiong Huang
- Departments of PathologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Jidong Cheng
- Departments of Internal MedicineThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
143
|
Sacco F, Calderone A, Castagnoli L, Cesareni G. The cell-autonomous mechanisms underlying the activity of metformin as an anticancer drug. Br J Cancer 2016; 115:1451-1456. [PMID: 27875520 PMCID: PMC5155371 DOI: 10.1038/bjc.2016.385] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/12/2016] [Accepted: 10/23/2016] [Indexed: 12/23/2022] Open
Abstract
The biguanide drug metformin profoundly affects cell metabolism, causing an impairment of the cell energy balance and triggering a plethora of pleiotropic effects that vary depending on the cellular or environmental context. Interestingly, a decade ago, it was observed that metformin-treated diabetic patients have a significantly lower cancer risk. Although a variety of in vivo and in vitro observations emphasising the role of metformin as anticancer drug have been reported, the underlying mechanisms are still poorly understood. Here, we discuss our current understanding of the molecular mechanisms that are perturbed by metformin treatment and that might be relevant to understand its antitumour activities. We focus on the cell-autonomous mechanisms modulating growth and death of cancer cells.
Collapse
Affiliation(s)
- Francesca Sacco
- Department of Biochemistry, Max Plank Institute, Martinsried (Munich) 82152, Germany
| | - Alberto Calderone
- IBBE-CNR at the Bioinformatics and Computational Biology Unit, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| |
Collapse
|
144
|
Wang PY, Li J, Walcott FL, Kang JG, Starost MF, Talagala SL, Zhuang J, Park JH, Huffstutler RD, Bryla CM, Mai PL, Pollak M, Annunziata CM, Savage SA, Fojo AT, Hwang PM. Inhibiting mitochondrial respiration prevents cancer in a mouse model of Li-Fraumeni syndrome. J Clin Invest 2016; 127:132-136. [PMID: 27869650 DOI: 10.1172/jci88668] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/06/2016] [Indexed: 12/14/2022] Open
Abstract
Li-Fraumeni syndrome (LFS) is a cancer predisposition disorder caused by germline mutations in TP53 that can lead to increased mitochondrial metabolism in patients. However, the implications of altered mitochondrial function for tumorigenesis in LFS are unclear. Here, we have reported that genetic or pharmacologic disruption of mitochondrial respiration improves cancer-free survival in a mouse model of LFS that expresses mutant p53. Mechanistically, inhibition of mitochondrial function increased autophagy and decreased the aberrant proliferation signaling caused by mutant p53. In a pilot study, LFS patients treated with metformin exhibited decreases in mitochondrial activity concomitant with activation of antiproliferation signaling, thus reproducing the effects of disrupting mitochondrial function observed in LFS mice. These observations indicate that a commonly prescribed diabetic medicine can restrain mitochondrial metabolism and tumorigenesis in an LFS model, supporting its further consideration for cancer prevention in LFS patients.
Collapse
|
145
|
Gui DY, Sullivan LB, Luengo A, Hosios AM, Bush LN, Gitego N, Davidson SM, Freinkman E, Thomas CJ, Vander Heiden MG. Environment Dictates Dependence on Mitochondrial Complex I for NAD+ and Aspartate Production and Determines Cancer Cell Sensitivity to Metformin. Cell Metab 2016; 24:716-727. [PMID: 27746050 PMCID: PMC5102768 DOI: 10.1016/j.cmet.2016.09.006] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 07/26/2016] [Accepted: 09/14/2016] [Indexed: 12/25/2022]
Abstract
Metformin use is associated with reduced cancer mortality, but how metformin impacts cancer outcomes is controversial. Although metformin can act on cells autonomously to inhibit tumor growth, the doses of metformin that inhibit proliferation in tissue culture are much higher than what has been described in vivo. Here, we show that the environment drastically alters sensitivity to metformin and other complex I inhibitors. We find that complex I supports proliferation by regenerating nicotinamide adenine dinucleotide (NAD)+, and metformin's anti-proliferative effect is due to loss of NAD+/NADH homeostasis and inhibition of aspartate biosynthesis. However, complex I is only one of many inputs that determines the cellular NAD+/NADH ratio, and dependency on complex I is dictated by the activity of other pathways that affect NAD+ regeneration and aspartate levels. This suggests that cancer drug sensitivity and resistance are not intrinsic properties of cancer cells, and demonstrates that the environment can dictate sensitivity to therapies that impact cell metabolism.
Collapse
Affiliation(s)
- Dan Y Gui
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lucas B Sullivan
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Alba Luengo
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Aaron M Hosios
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lauren N Bush
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Nadege Gitego
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Shawn M Davidson
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Craig J Thomas
- NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
146
|
Metformin suppresses hypoxia-induced stabilization of HIF-1α through reprogramming of oxygen metabolism in hepatocellular carcinoma. Oncotarget 2016; 7:873-84. [PMID: 26621849 PMCID: PMC4808039 DOI: 10.18632/oncotarget.6418] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 11/16/2015] [Indexed: 12/16/2022] Open
Abstract
Overexpression of hypoxia-induced factor 1α (HIF-1α) has been shown to be involved in the development and progression of hepatocellular carcinoma (HCC). HIF-1α should therefore be a promising molecular target for the development of anti-HCC agents. Metformin, an established antidiabetic drug, has proved to also be effective in treating cancer although the precise underlying mechanisms of this activity are not fully elucidated. The aim of this study was to investigate the effects of metformin on the expression of HIF-1α and oxygen metabolism in HCC. The results showed that metformin inhibited hypoxia-induced HIF-1α accumulation and activation independent of AMP-activated protein kinase (AMPK). Moreover, this decrease in HIF-1α accumulation was accompanied by promotion of HIF-1α protein degradation. In addition, metformin significantly decreased oxygen consumption, ultimately leading to increased intracellular oxygen tension and decreased staining with the hypoxia marker pimonidazole. In vivo studies demonstrated that metformin delayed tumor growth and attenuated the expression of HIF-1α in HCC tumor xenografts. Together, these findings suggest that metformin decreases hypoxia-induced HIF-1α accumulation by actively suppressing mitochondrial oxygen consumption and enhancing cellular oxygenation ability, providing a fundamental mechanism of metformin activity against HCC.
Collapse
|
147
|
Metformin increases antitumor activity of MEK inhibitors through GLI1 downregulation in LKB1 positive human NSCLC cancer cells. Oncotarget 2016; 7:4265-78. [PMID: 26673006 PMCID: PMC4826204 DOI: 10.18632/oncotarget.6559] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/25/2015] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Metformin, widely used as antidiabetic drug, showed antitumoral effects expecially in combination with chemotherapy. Our group recently has demonstrated that metformin and gefitinib are synergistic in LKB1-wild-type NSCLC cells. In these models, metformin as single agent induced an activation and phosphorylation of mitogen-activated-protein-kinase (MAPK) through an increased C-RAF/B-RAF heterodimerization. EXPERIMENTAL DESIGN Since single agent metformin enhances proliferating signals through the RAS/RAF/MAPK pathway, and several MEK inhibitors (MEK-I) demonstrated clinical efficacy in combination with other agents in NSCLC, we tested the effects of metformin plus MEK-I (selumetinib or pimasertib) on proliferation, invasiveness, migration abilities in vitro and in vivo in LKB1 positive NSCLC models harboring KRAS wild type and mutated gene. RESULTS The combination of metformin with MEK-I showed a strong anti-proliferative and proapoptotic effect in Calu-3, H1299, H358 and H1975 human NSCLC cell lines, independently from the KRAS mutational status. The combination reduced the metastatic behaviour of NSCLC cells, via a downregulation of GLI1 trascritional activity, thus affecting the transition from an epithelial to a mesenchymal phenotype. Metformin and MEK-Is combinations also decreased the production and activity of MMP-2 and MMP-9 by reducing the NF-jB (p65) binding to MMP-2 and MMP-9 promoters. CONCLUSIONS Metformin potentiates the antitumor activity of MEK-Is in human LKB1-wild-type NSCLC cell lines, independently from the KRAS mutational status, through GLI1 downregulation and by reducing the NF-jB (p65)-mediated transcription of MMP-2 and MMP-9.
Collapse
|
148
|
Daugan M, Dufaÿ Wojcicki A, d’Hayer B, Boudy V. Metformin: An anti-diabetic drug to fight cancer. Pharmacol Res 2016; 113:675-685. [DOI: 10.1016/j.phrs.2016.10.006] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/22/2016] [Accepted: 10/04/2016] [Indexed: 12/22/2022]
|
149
|
Figueiredo RADO, Weiderpass E, Tajara EH, Ström P, Carvalho AL, de Carvalho MB, Kanda JL, Moyses RA, Wünsch-Filho V. Diabetes mellitus, metformin and head and neck cancer. Oral Oncol 2016; 61:47-54. [DOI: 10.1016/j.oraloncology.2016.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/17/2016] [Accepted: 08/20/2016] [Indexed: 12/25/2022]
|
150
|
Bruno S, Ledda B, Tenca C, Ravera S, Orengo AM, Mazzarello AN, Pesenti E, Casciaro S, Racchi O, Ghiotto F, Marini C, Sambuceti G, DeCensi A, Fais F. Metformin inhibits cell cycle progression of B-cell chronic lymphocytic leukemia cells. Oncotarget 2016; 6:22624-40. [PMID: 26265439 PMCID: PMC4673187 DOI: 10.18632/oncotarget.4168] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/23/2015] [Indexed: 12/20/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL) was believed to result from clonal accumulation of resting apoptosis-resistant malignant B lymphocytes. However, it became increasingly clear that CLL cells undergo, during their life, iterative cycles of re-activation and subsequent clonal expansion. Drugs interfering with CLL cell cycle entry would be greatly beneficial in the treatment of this disease. 1, 1-Dimethylbiguanide hydrochloride (metformin), the most widely prescribed oral hypoglycemic agent, inexpensive and well tolerated, has recently received increased attention for its potential antitumor activity. We wondered whether metformin has apoptotic and anti-proliferative activity on leukemic cells derived from CLL patients. Metformin was administered in vitro either to quiescent cells or during CLL cell activation stimuli, provided by classical co-culturing with CD40L-expressing fibroblasts. At doses that were totally ineffective on normal lymphocytes, metformin induced apoptosis of quiescent CLL cells and inhibition of cell cycle entry when CLL were stimulated by CD40-CD40L ligation. This cytostatic effect was accompanied by decreased expression of survival- and proliferation-associated proteins, inhibition of signaling pathways involved in CLL disease progression and decreased intracellular glucose available for glycolysis. In drug combination experiments, metformin lowered the apoptotic threshold and potentiated the cytotoxic effects of classical and novel antitumor molecules. Our results indicate that, while CLL cells after stimulation are in the process of building their full survival and cycling armamentarium, the presence of metformin affects this process.
Collapse
Affiliation(s)
- Silvia Bruno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Bernardetta Ledda
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Claudya Tenca
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Silvia Ravera
- Department of Pharmacology, University of Genova, Genova, Italy
| | - Anna Maria Orengo
- IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Andrea Nicola Mazzarello
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,The Feinstein Institute for Medical Research, North Shore-Long Island, Experimental Immunology, Manhasset, NY, USA
| | - Elisa Pesenti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Salvatore Casciaro
- Department of Internal Medicine and Medical Specialty, University of Genova, Genova, Italy
| | - Omar Racchi
- Hematology-Oncology Unit - Ospedale Villa Scassi, Genova, Italy
| | - Fabio Ghiotto
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Cecilia Marini
- CNR Institute of Bioimages and Molecular Physiology, Milan, Section of Genoa, Genoa, Italy
| | - Gianmario Sambuceti
- IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.,Department of Health Science, University of Genova, Genova, Italy
| | - Andrea DeCensi
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy.,Division of Medical Oncology, Ospedali Galliera, Genova, Italy
| | - Franco Fais
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|