1
|
Szablewski L. Changes in Cells Associated with Insulin Resistance. Int J Mol Sci 2024; 25:2397. [PMID: 38397072 PMCID: PMC10889819 DOI: 10.3390/ijms25042397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Insulin is a polypeptide hormone synthesized and secreted by pancreatic β-cells. It plays an important role as a metabolic hormone. Insulin influences the metabolism of glucose, regulating plasma glucose levels and stimulating glucose storage in organs such as the liver, muscles and adipose tissue. It is involved in fat metabolism, increasing the storage of triglycerides and decreasing lipolysis. Ketone body metabolism also depends on insulin action, as insulin reduces ketone body concentrations and influences protein metabolism. It increases nitrogen retention, facilitates the transport of amino acids into cells and increases the synthesis of proteins. Insulin also inhibits protein breakdown and is involved in cellular growth and proliferation. On the other hand, defects in the intracellular signaling pathways of insulin may cause several disturbances in human metabolism, resulting in several chronic diseases. Insulin resistance, also known as impaired insulin sensitivity, is due to the decreased reaction of insulin signaling for glucose levels, seen when glucose use in response to an adequate concentration of insulin is impaired. Insulin resistance may cause, for example, increased plasma insulin levels. That state, called hyperinsulinemia, impairs metabolic processes and is observed in patients with type 2 diabetes mellitus and obesity. Hyperinsulinemia may increase the risk of initiation, progression and metastasis of several cancers and may cause poor cancer outcomes. Insulin resistance is a health problem worldwide; therefore, mechanisms of insulin resistance, causes and types of insulin resistance and strategies against insulin resistance are described in this review. Attention is also paid to factors that are associated with the development of insulin resistance, the main and characteristic symptoms of particular syndromes, plus other aspects of severe insulin resistance. This review mainly focuses on the description and analysis of changes in cells due to insulin resistance.
Collapse
Affiliation(s)
- Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
2
|
Lee Y, Ni J, Wasinger VC, Graham P, Li Y. Comparison Study of Small Extracellular Vesicle Isolation Methods for Profiling Protein Biomarkers in Breast Cancer Liquid Biopsies. Int J Mol Sci 2023; 24:15462. [PMID: 37895140 PMCID: PMC10607056 DOI: 10.3390/ijms242015462] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are an important intercellular communicator, participating in all stages of cancer metastasis, immunity, and therapeutic resistance. Therefore, protein cargoes within sEVs are considered as a superior source for breast cancer (BC) biomarker discovery. Our study aimed to optimise the approach for sEV isolation and sEV proteomic analysis to identify potential sEV protein biomarkers for BC diagnosis. sEVs derived from BC cell lines, BC patients' plasma, and non-cancer controls were isolated using ultracentrifugation (UC), a Total Exosome Isolation kit (TEI), and a combined approach named UCT. In BC cell lines, the UC isolates showed a higher sEV purity and marker expression, as well as a higher number of sEV proteins. In BC plasma samples, the UCT isolates showed the highest proportion of sEV-related proteins and the lowest percentage of lipoprotein-related proteins. Our data suggest that the assessment of both the quantity and quality of sEV isolation methods is important in selecting the optimal approach for the specific sEV research purpose, depending on the sample types and downstream analysis.
Collapse
Affiliation(s)
- Yujin Lee
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Jie Ni
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Valerie C. Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Kensington, NSW 2052, Australia;
| | - Peter Graham
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| | - Yong Li
- School of Clinical Medicine, St George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW 2052, Australia; (Y.L.); (J.N.); (P.G.)
- Cancer Care Centre, St George Hospital, Kogarah, NSW 2217, Australia
| |
Collapse
|
3
|
Sankofi BM, Valencia-Rincón E, Sekhri M, Ponton-Almodovar AL, Bernard JJ, Wellberg EA. The impact of poor metabolic health on aggressive breast cancer: adipose tissue and tumor metabolism. Front Endocrinol (Lausanne) 2023; 14:1217875. [PMID: 37800138 PMCID: PMC10548218 DOI: 10.3389/fendo.2023.1217875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Obesity and type 2 diabetes are chronic metabolic diseases that impact tens to hundreds of millions of adults, especially in developed countries. Each condition is associated with an elevated risk of breast cancer and with a poor prognosis after treatment. The mechanisms connecting poor metabolic health to breast cancer are numerous and include hyperinsulinemia, inflammation, excess nutrient availability, and adipose tissue dysfunction. Here, we focus on adipose tissue, highlighting important roles for both adipocytes and fibroblasts in breast cancer progression. One potentially important mediator of adipose tissue effects on breast cancer is the fibroblast growth factor receptor (FGFR) signaling network. Among the many roles of FGFR signaling, we postulate that key mechanisms driving aggressive breast cancer include epithelial-to-mesenchymal transition and cellular metabolic reprogramming. We also pose existing questions that may help better understand breast cancer biology in people with obesity, type 2 diabetes, and poor metabolic health.
Collapse
Affiliation(s)
- Barbara Mensah Sankofi
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Estefania Valencia-Rincón
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Malika Sekhri
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Adriana L. Ponton-Almodovar
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Nicolas V. Perricone Division of Dermatology, Michigan State University, East Lansing, MI, United States
- Department of Medicine, Michigan State University, East Lansing, MI, United States
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Nicolas V. Perricone Division of Dermatology, Michigan State University, East Lansing, MI, United States
- Department of Medicine, Michigan State University, East Lansing, MI, United States
| | - Elizabeth A. Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
4
|
Saha A, Hamilton-Reeves J, DiGiovanni J. White adipose tissue-derived factors and prostate cancer progression: mechanisms and targets for interventions. Cancer Metastasis Rev 2022; 41:649-671. [PMID: 35927363 PMCID: PMC9474694 DOI: 10.1007/s10555-022-10056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022]
Abstract
Obesity represents an important risk factor for prostate cancer, driving more aggressive disease, chemoresistance, and increased mortality. White adipose tissue (WAT) overgrowth in obesity is central to the mechanisms that lead to these clinical observations. Adipose stromal cells (ASCs), the progenitors to mature adipocytes and other cell types in WAT, play a vital role in driving PCa aggressiveness. ASCs produce numerous factors, especially chemokines, including the chemokine CXCL12, which is involved in driving EMT and chemoresistance in PCa. A greater understanding of the impact of WAT in obesity-induced progression of PCa and the underlying mechanisms has begun to provide opportunities for developing interventional strategies for preventing or offsetting these critical events. These include weight loss regimens, therapeutic targeting of ASCs, use of calorie restriction mimetic compounds, and combinations of compounds as well as specific receptor targeting strategies.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Jill Hamilton-Reeves
- Departments of Urology and Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA.
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA.
| |
Collapse
|
5
|
Sunjaya AP, Sunjaya AF. Targeting ageing and preventing organ degeneration with metformin. DIABETES & METABOLISM 2021; 47:101203. [DOI: 10.1016/j.diabet.2020.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/29/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
|
6
|
Iwase T, Wang X, Shrimanker TV, Kolonin MG, Ueno NT. Body composition and breast cancer risk and treatment: mechanisms and impact. Breast Cancer Res Treat 2021; 186:273-283. [PMID: 33475878 DOI: 10.1007/s10549-020-06092-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this review is to clarify the association of body composition with breast cancer risk and treatment, including physiological mechanisms, and to elucidate strategies for overcoming unfavorable body composition changes that relate to breast cancer progression. METHODS We have summarized updated knowledge regarding the mechanism of the negative association of altered body composition with breast cancer risk and treatment. We also review strategies for reversing unfavorable body composition based on the latest clinical trial results. RESULTS Body composition changes in patients with breast cancer typically occur during menopause or as a result of chemotherapy or endocrine therapy. Dysfunction of visceral adipose tissue (VAT) in the setting of obesity underlies insulin resistance and chronic inflammation, which can lead to breast cancer development and progression. Insulin resistance and chronic inflammation are also observed in patients with breast cancer who have sarcopenia or sarcopenic obesity. Nutritional support and a personalized exercise program are the fundamental interventions for reversing unfavorable body composition. Other interventions that have been explored in specific situations include metformin, testosterone, emerging agents that directly target the adipocyte microenvironment, and bariatric surgery. CONCLUSIONS A better understanding of the biology of body composition phenotypes is key to determining the best intervention program for patients with breast cancer.
Collapse
Affiliation(s)
- Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Tushaar Vishal Shrimanker
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mikhail G Kolonin
- Center for Metabolic and Degenerative Diseases, The University of Texas Health Science Center at Houston, 7000 Fannin Street, Houston, TX, 77030, USA
| | - Naoto T Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1354, Houston, TX, 77030, USA. .,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Shaikh H, Bradhurst P, Ma LX, Tan SYC, Egger SJ, Vardy JL. Body weight management in overweight and obese breast cancer survivors. Cochrane Database Syst Rev 2020; 12:CD012110. [PMID: 33305350 PMCID: PMC8094215 DOI: 10.1002/14651858.cd012110.pub2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Studies suggest that overweight and obese breast cancer survivors are at increased risk of cancer recurrence and have higher all-cause mortality. Obesity has an impact on breast cancer survivor's quality of life (QOL) and increases the risk of longer-term morbidities such as type 2 diabetes mellitus and cardiovascular disease. Many cancer guidelines recommend survivors maintain a healthy weight but there is a lack of evidence regarding which weight loss method to recommend. OBJECTIVES To assess the effects of different body weight loss approaches in breast cancer survivors who are overweight or obese (body mass index (BMI) ≥ 25 kg/m2). SEARCH METHODS We carried out a search in the Cochrane Breast Cancer Group's (CBCG's) Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL, Issue 6), MEDLINE (2012 to June 2019), Embase (2015 to June 2019), the World Health Organisation International Clinical Trials Registry Platform (WHO ICTRP) and Clinicaltrials.gov on 17 June 2019. We also searched Mainland Chinese academic literature databases (CNKI), VIP, Wan Fang Data and SinoMed on 25 June 2019. We screened references in relevant manuscripts. SELECTION CRITERIA We included randomised controlled trials (RCTs), quasi-RCTs and randomised cross-over trials evaluating body weight management for overweight and obese breast cancer survivors (BMI ≥ 25 kg/m2). The aim of the intervention had to be weight loss. DATA COLLECTION AND ANALYSIS Two review authors independently performed data extraction and assessed risk of bias for the included studies, and applied the quality of the evidence using the GRADE approach. Dichotomous outcomes were analysed as proportions using the risk ratio (RR) as the measure of effect. Continuous data were analysed as means with the measure of effect being expressed as the mean differences (MDs) between treatment groups in change from baseline values with 95% confidence intervals (CIs), when all studies reported exactly the same outcomes on the same scale. If similar outcomes were reported on different scales the standardised mean difference (SMD) was used as the measure of effect. Quality of life data and relevant biomarkers were extracted where available. MAIN RESULTS We included a total of 20 studies (containing 23 intervention-comparisons) and analysed 2028 randomised women. Participants in the experimental groups received weight loss interventions using the core element of dietary changes, either in isolation or in combination with other core elements such as 'diet and exercise', 'diet and psychosocial support' or 'diet, exercise and psychosocial support'. Participants in the controls groups either received usual care, written materials or placebo, or wait-list controls. The duration of interventions ranged from 0.5 months to 24 months. The duration of follow-up ranged from three months to 36 months. There were no time-to-event data available for overall survival, breast cancer recurrence and disease-free survival. There was a relatively small amount of data available for breast cancer recurrence (281 participants from 4 intervention-comparisons with 14 recurrence events; RR 1.95, 95% CI 0.68 to 5.60; low-quality evidence) and the analysis was likely underpowered. Overall, we found low-quality evidence that weight loss interventions for overweight and obese breast cancer survivors resulted in a reduction in body weight (MD: -2.25 kg, 95% CI: -3.19 to -1.3 kg; 21 intervention-comparisons; 1751 women), body mass index (BMI) (MD: -1.08 kg/m2, 95% CI: -1.61 to -0.56 kg/m2; 17 intervention-comparisons; 1353 women), and waist circumference (MD:-1.73 cm, 95% CI: -3.17 to -0.29 cm; 13 intervention-comparisons; 1193 women), and improved overall quality of life (SMD: 0.74; 95% CI: 0.20 to 1.29; 10 intervention-comparisons; 867 women). No increase was seen in adverse events for women in the intervention groups compared to controls (RR 0.94, 95% CI: 0.76 to 1.17; 4 intervention-comparisons; 394 women; high-quality evidence). Subgroup analyses revealed that decreases in body weight, BMI and waist circumference were present in women regardless of their ethnicity and menopausal status. Multimodal weight loss interventions (which referred to 'diet, exercise and psychosocial support') appeared to result in greater reductions in body weight (MD: -2.88 kg, 95% CI: -3.98 to -1.77 kg; 13 intervention-comparisons; 1526 participants), BMI (MD: -1.44 kg/m2, 95% CI: -2.16 to -0.72 kg/m2; 11 studies; 1187 participants) and waist circumference (MD:-1.66 cm, 95% CI: -3.49 to -0.16 cm; 8 intervention-comparisons; 1021 participants) compared to dietary change alone, however the evidence was low quality. AUTHORS' CONCLUSIONS Weight loss interventions, particularly multimodal interventions (incorporating diet, exercise and psychosocial support), in overweight or obese breast cancer survivors appear to result in decreases in body weight, BMI and waist circumference and improvement in overall quality of life. There was no increase in adverse events. There is a lack of data to determine the impact of weight loss interventions on survival or breast cancer recurrence. This review is based on studies with marked heterogeneity regarding weight loss interventions. Due to the methods used in included studies, there was a high risk of bias regarding blinding of participants and assessors. Further research is required to determine the optimal weight loss intervention and assess the impact of weight loss on survival outcomes. Long-term follow-up in weight loss intervention studies is required to determine if weight changes are sustained beyond the intervention periods.
Collapse
Affiliation(s)
- Hassan Shaikh
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | - Li Xin Ma
- Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Australia
- Nutrition and Food Hygiene Department, Hebei University, Baoding, China
| | - Sim Yee Cindy Tan
- Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Australia
- Nutrition and Dietetics Department, Concord Repatriation General Hospital, Concord, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Sam J Egger
- Cancer Research Division, Cancer Council NSW, Sydney, Australia
| | - Janette L Vardy
- Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Australia
- Faculty of Medicine and Health, Concord Clinical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
8
|
Cheng TYD, Omilian AR, Yao S, Sanchez PV, Polk LZ, Zhang W, Datta S, Bshara W, Ondracek RP, Davis W, Liu S, Hong CC, Bandera EV, Khoury T, Ambrosone CB. Body fatness and mTOR pathway activation of breast cancer in the Women's Circle of Health Study. NPJ Breast Cancer 2020; 6:45. [PMID: 33024820 PMCID: PMC7505987 DOI: 10.1038/s41523-020-00187-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 08/19/2020] [Indexed: 12/30/2022] Open
Abstract
Energy imbalance has an important role in breast cancer prognosis. Hyperactive mechanistic Target of Rapamycin (mTOR) pathway is associated with breast tumor growth, but the extent to which body fatness is associated with mTOR pathway activities in breast cancer is unclear. We performed immunostaining for mTOR, phosphorylated (p)-mTOR, p-AKT, and p-p70S6K in tumor tissue from 590 women (464 African Americans/Blacks and 126 Whites) with newly diagnosed invasive breast cancer in the Women's Circle of Health Study. Anthropometric measures were taken by study staff, and body composition was measured by bioelectrical impedance analysis. Linear regressions were used to estimate percent differences in protein expression between categories of body mass index (BMI), waist circumference, waist/hip ratio, fat mass, fat mass index, and percent body fat. We observed that BMI ≥ 35.0 vs. <25 kg/m2 was associated with 108.3% (95% CI = 16.9%-270.9%) and 101.8% (95% CI = 17.0%-248.8%) higher expression in p-mTOR and normalized p-mTOR, i.e., p-mTOR/mTOR, respectively. Quartiles 4 vs. 1 of waist/hip ratio was associated with 41.8% (95% CI = 5.81%-89.9%) higher mTOR expression. Similar associations were observed for the body fat measurements, particularly in patients with estrogen receptor-negative (ER-) tumors, but not in those with ER+ tumors, although the differences in associations were not significant. This tumor-based study found positive associations between body fatness and mTOR pathway activation, evident by a p-mTOR expression, in breast cancer. Our findings suggest that mTOR inhibition can be a treatment strategy to prevent the recurrence of these tumors in obese individuals.
Collapse
Affiliation(s)
- Ting-Yuan David Cheng
- Department of Epidemiology, University of Florida, Gainesville, FL USA
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Angela R Omilian
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Pamela V Sanchez
- Department of Epidemiology, University of Florida, Gainesville, FL USA
| | - Latasia Z Polk
- Department of Epidemiology, University of Florida, Gainesville, FL USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL USA
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL USA
| | - Wiam Bshara
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Rochelle Payne Ondracek
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Warren Davis
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Elisa V Bandera
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, NJ USA
| | - Thaer Khoury
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| |
Collapse
|
9
|
Pan K, Chlebowski RT, Mortimer JE, Gunter MJ, Rohan T, Vitolins MZ, Adams-Campbell LL, Ho GYF, Cheng TYD, Nelson RA. Insulin resistance and breast cancer incidence and mortality in postmenopausal women in the Women's Health Initiative. Cancer 2020; 126:3638-3647. [PMID: 32530506 DOI: 10.1002/cncr.33002] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/22/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Insulin resistance is associated with higher all-cause and cancer-specific mortality in postmenopausal women. However, to the authors' knowledge, information regarding insulin resistance and breast cancer mortality risk is limited. Therefore, the authors examined associations between insulin resistance and breast cancer incidence and mortality in a subsample of Women's Health Initiative participants. METHODS A total of 22,837 postmenopausal women with fasting baseline glucose and insulin levels were followed for incident breast cancer and breast cancer mortality. Breast cancers were verified by medical record review and serial National Death Index linkage-enhanced mortality findings. Insulin resistance was estimated using the homeostatic model assessment of insulin resistance (HOMA-IR). Multivariable Cox proportional hazards models were used to compute hazard ratios (HRs) with 95% confidence intervals (95% CIs) for quartile comparisons. Outcomes included breast cancer incidence, deaths from breast cancer, and deaths after breast cancer (breast cancer followed by death from any cause). RESULTS During a median of 19.8 years of follow-up of 1328 breast cancer cases, there were 512 deaths reported, 151 of which were from breast cancer. Breast cancer incidence was higher in women in the highest HOMA-IR quartile (HR, 1.34; 95% CI, 1.12-1.61 [P for trend = .003]). Although HOMA-IR was not found to be associated with risk of death from breast cancer (HR, 1.04; 95% CI, 0.60-1.79), women in the highest versus those in the lowest HOMA-IR quartile were at a higher risk of death after breast cancer (HR, 1.78; 95% CI, 1.32-2.39 [P for trend <.001]). CONCLUSIONS Higher levels of insulin resistance in postmenopausal women are associated with higher breast cancer incidence and higher all-cause mortality after breast cancer.
Collapse
Affiliation(s)
- Kathy Pan
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, California
| | - Rowan T Chlebowski
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, California
| | | | - Marc J Gunter
- The International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Thomas Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Mara Z Vitolins
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Gloria Y F Ho
- Division of Epidemiology, Northwell Health, Great Neck, New York
| | - Ting-Yuan David Cheng
- Department of Epidemiology, University of Florida at Gainesville, Gainesville, Florida
| | | |
Collapse
|
10
|
Wahdan-Alaswad RS, Edgerton SM, Salem HS, Thor AD. Metformin Targets Glucose Metabolism in Triple Negative Breast Cancer. ACTA ACUST UNITED AC 2018; 4. [PMID: 29780974 PMCID: PMC5959056 DOI: 10.4172/2476-2261.1000129] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Metformin is the most widely administered anti-diabetic agent worldwide. In patients receiving metformin for metabolic syndrome or diabetes, it reduces the incidence and improves the survival of breast cancer (BC) patients. We have previously shown that metformin is particularly potent against triple negative breast cancer (TNBC), with a reduction of proliferation, oncogenicity and motility, inhibition of pro-oncogenic signaling pathways and induction of apoptosis. These BCs are well recognized to be highly dependent on glucose/glucosamine (metabolized through anaerobic glycolysis) and lipids, which are metabolized for the production of energy and cellular building blocks to sustain a high rate of proliferation. We have previously demonstrated that metformin inhibits lipid metabolism, specifically targeting fatty acid synthase (FASN), cholesterol biosynthesis and GM1 lipid rafts in TNBC. We also reported that glucose promotes phenotypic aggression and reduces metformin efficacy. We now show that metformin inhibits several key enzymes requisite to glucose metabolism in TNBC, providing additional insight into why metformin is especially toxic to this subtype of BC. Our data suggests that the use of metformin to target key metabolic defects in lipid and carbohydrate metabolism in cancer may be broadly applicable, especially against highly aggressive malignant cells.
Collapse
Affiliation(s)
- R S Wahdan-Alaswad
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - S M Edgerton
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - H S Salem
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, USA
| | - A D Thor
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, USA
| |
Collapse
|
11
|
Hanawa S, Mitsuhashi A, Shozu M. Antitumor effects of metformin via indirect inhibition of protein phosphatase 2A in patients with endometrial cancer. PLoS One 2018; 13:e0192759. [PMID: 29444159 PMCID: PMC5812621 DOI: 10.1371/journal.pone.0192759] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/20/2017] [Indexed: 01/22/2023] Open
Abstract
Objective Metformin, an antidiabetic drug, inhibits the endometrial cancer cell growth in vivo by improving the insulin resistance; however, its mechanism of action is not completely understood. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase associated with insulin resistance and type 2 diabetes, and its inhibition restores the insulin resistance. This study investigated the antitumor effect of metformin on endometrial cancer with a focus on PP2A. Methods Metformin (1,500–2,250 mg/day) was preoperatively administered to patients with endometrial cancer for 4 to 6 weeks. Expression of the PP2A regulatory subunits, 4 (PPP2R4) and B (PP2A-B), was evaluated using real-time polymerase chain reaction (RT–PCR) and immunohistochemistry (IHC) using paired specimens obtained before and after metformin treatment. The effect of PPP2R4 inhibition with small interfering RNA was evaluated in the endometrial cancer cell lines HEC265 and HEC1B. P values of < .05 were considered statistically significant. Results Preoperative metformin treatment significantly reduced the expression of PP2A-B, as determined using IHC, and the mRNA expression of PPP2R4, as determined using RT–PCR, in the patients with endometrial cancer. However, metformin could not directly alter the PPP2R4 mRNA levels in the endometrial cancer cell lines in vitro. PPP2R4 knockdown reduced the proliferation and induced the apoptosis by activating caspases 3/7 in HEC265 and HEC1B cells. Conclusions Downregulation of the PP2A-B subunit, including PPP2R4, is an important indirect target of metformin. Inhibition of PP2A may be an option for the treatment of endometrial cancer patients with insulin resistance. Trial registration This trial is registered with UMIN-CTR (number UMIN000004852).
Collapse
Affiliation(s)
- Shinsuke Hanawa
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Mitsuhashi
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- * E-mail:
| | - Makio Shozu
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
12
|
Farr A, Stolz M, Baumann L, Bago-Horvath Z, Oppolzer E, Pfeiler G, Seifert M, Singer CF. The effect of obesity on pathological complete response and survival in breast cancer patients receiving uncapped doses of neoadjuvant anthracycline-taxane-based chemotherapy. Breast 2017; 33:153-158. [DOI: 10.1016/j.breast.2017.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 11/16/2022] Open
|
13
|
Sulfonylureas (not metformin) improve survival of patients with diabetes and resectable pancreatic adenocarcinoma. INTERNATIONAL JOURNAL OF SURGERY-ONCOLOGY 2017; 2:e15. [PMID: 29177213 PMCID: PMC5673127 DOI: 10.1097/ij9.0000000000000015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Patients with pancreatic adenocarcinoma have an increased propensity for diabetes. Recent studies suggest patients with diabetes and pancreatic adenocarcinoma treated with metformin have increased survival. This study was undertaken to determine whether metformin use is associated with increased survival in patients with pancreatic adenocarcinoma.
Collapse
|
14
|
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
|
15
|
Melatonin and Metformin Diminish Oxidative Stress in Heart Tissue in a Rat Model of High Fat Diet and Mammary Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1047:7-19. [PMID: 29151256 DOI: 10.1007/5584_2017_128] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of this study was to determine the effects of long-term administration of the oral antidiabetic metformin or the pineal hormone melatonin, and a combination thereof, in preventing oxidative stress in the heart tissue of female Sprague-Dawley rats with mammary tumors induced by N-methyl-N-nitrosourea (NMU) (50 mg/kg) given on the 42nd postnatal day. Metformin and melatonin were administered 12 days before and 16 weeks after the carcinogen. During the experiment, all animals were fed a high fat diet (10% total fat, 2.5% from lard, and 7.5% from palm oil). The findings are that mammary carcinogenesis generated oxidative stress. Reactive oxygen species (ROS) content, estimated from thiobarbituric acid reactive substances (TBARS), oxidatively modified protein content (aldehyde and ketone derivatives), and the activity of the antioxidant enzymes superoxide dismutase, glutathione reductase, and glutathione peroxidase were all augmented. Metformin caused a decrease in oxidative stress in the heart, accompanied by a decrease in diene conjugates, the elimination of ROS (stable total antioxidant status), and the activation of catalase and glutathione reductase. Melatonin caused an increase in total antioxidant status and a substantial reduction in ROS as estimated from aldehyde and ketone derivatives, lipid peroxidation at the initial (diene conjugates) and terminal stages (TBARS), and increased catalase and glutathione peroxidase activities. Metformin and melatonin combined reversed the effects of NMU on oxidative stress. In conclusion, melatonin reduces the level of oxidative stress in the heart tissue, caused by NMU carcinogenesis and a high fat diet, significantly stronger than metformin.
Collapse
|
16
|
Fukumura D, Incio J, Shankaraiah RC, Jain RK. Obesity and Cancer: An Angiogenic and Inflammatory Link. Microcirculation 2016; 23:191-206. [PMID: 26808917 DOI: 10.1111/micc.12270] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/20/2016] [Indexed: 12/15/2022]
Abstract
With the current epidemic of obesity, a large number of patients diagnosed with cancer are overweight or obese. Importantly, this excess body weight is associated with tumor progression and poor prognosis. The mechanisms for this worse outcome, however, remain poorly understood. We review here the epidemiological evidence for the association between obesity and cancer, and discuss potential mechanisms focusing on angiogenesis and inflammation. In particular, we will discuss how the dysfunctional angiogenesis and inflammation occurring in adipose tissue in obesity may promote tumor progression, resistance to chemotherapy, and targeted therapies such as anti-angiogenic and immune therapies. Better understanding of how obesity fuels tumor progression and therapy resistance is essential to improve the current standard of care and the clinical outcome of cancer patients. To this end, we will discuss how an anti-diabetic drug such as metformin can overcome these adverse effects of obesity on the progression and treatment resistance of tumors.
Collapse
Affiliation(s)
- Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology Group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal.,Department of Internal Medicine, Hospital S. João, Porto, Portugal
| | - Ram C Shankaraiah
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
|
18
|
Examining the prevalence of metabolic syndrome among overweight/obese African-American breast cancer survivors vs. matched non-cancer controls. J Cancer Surviv 2016; 11:102-110. [PMID: 27562474 DOI: 10.1007/s11764-016-0566-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/07/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE Metabolic Syndrome (MetS) is more predominant in overweight, obese and minority populations. This study examined the prevalence of MetS in an exclusively African-American (AA) cohort of breast cancer (BC) survivors; an underrepresented group in previous studies demonstrating negative BC outcomes disparities for females with MetS. METHODS Using a case-control design, overweight/obese AA women with treated Stage I-IIIa BC were matched 1:1 on age, race, sex, and body mass index (BMI) category with non-cancer population controls (n = 444). Three of the following conditions were used to define MetS: HDL cholesterol <50 mg/dL (1.3 mmol/L), serum triglycerides ≥150 mg/dL (1.7 mmol/L), blood glucose ≥100 mg/dL (or on treatment), waist circumference ≥88 cm, or ≥130 mmHg systolic or ≥85 mmHg diastolic blood pressure (or on treatment). Matched-pairs analyses were conducted. RESULTS For BC cases, most women had self-reported Stage I (n = 76) or Stage II (n = 91) disease and were 6.9 (±5.2) years post-diagnosis. MetS was significantly lower in BC survivors vs. their non-cancer population controls (43.2 vs. 51.4 %, respectively; p < 0.05). The diagnosis of MetS did not differ by BMI stratification. A lower prevalence of ≥2 risk factors (80.2 vs. 85.6 %, p < 0.05) was observed for all cases vs. CONTROLS CONCLUSIONS While MetS occurred less frequently in our BC cases vs. non-cancer controls, our estimates are nearly two times those reported in other BC survivors, suggesting important racial/ethnic differences. IMPLICATIONS FOR CANCER SURVIVORS The prognostic implications of MetS among AA BC survivors remain unknown and warrant further investigation.
Collapse
|
19
|
Karatas F, Erdem GU, Sahin S, Aytekin A, Yuce D, Sever AR, Babacan T, Ates O, Ozisik Y, Altundag K. Obesity is an independent prognostic factor of decreased pathological complete response to neoadjuvant chemotherapy in breast cancer patients. Breast 2016; 32:237-244. [PMID: 27318645 DOI: 10.1016/j.breast.2016.05.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/25/2016] [Accepted: 05/31/2016] [Indexed: 11/30/2022] Open
Abstract
PURPOSE The relation between higher body mass index (BMI) and pathological complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer (BC) is a controversial issue according to the data of Western and Asian patients. The aim of this study is to evaluate BMI and pCR to NAC and discuss the importance of pCR outcomes in Turkish BC patients as a bridging country between Europe and Asia. PATIENTS AND METHODS Of the 4423 BC patients diagnosed between the years 1994 and 2015 in Hacettepe University Cancer Institute, 295 female patients with stage II and III BC were enrolled in the study. Three different group divisions were done according to patients' BMI as normal or underweight (N/U) patients (BMI <25 kg/m2), overweight (OW) patients (BMI = 25-29.9 kg/m2) and obese (OB) patients (BMI ≥30 kg/m2). BC subtypes were defined as luminal-like (ER/PR-positive and HER2-negative), HER2/luminal (ER/PR-positive and HER2-positive), HER2-type (ER/PR-negative and HER2-positive), and triple-negative (TNBC; ER/PR- and HER2-negative). The analysis of overall survival (OS) and recurrence-free survival (RFS) was performed according to Kaplan-Meier method. The Log-rank test was used to compare the subgroup analysis and logistic regression analysis to determine the independent prognostic factors. RESULTS In this study, a total number of 93 (31.5%) patients were N/U, 107 (36.3%) patients were OW and 95 (32.2%) patients were OB. Among groups, except for the age, no baseline clinicopathological differences were found. In 70 (23.7%) patients, pCR was achieved. pCR rates in N/U, OW and OB were 31.2%, 22.4%, and 17.9% respectively, showing a considerable trend towards significance (P = 0.09 in chi-square test). In the multivariate logistic regression analysis, obesity was an independent adverse prognostic feature on pCR to NAC compared to N/U patients (OR, 0.34; 95% CI, 0.13 to 0.85, P = 0.02). The recurrence rates were slightly increased with the increase of BMI (N/U = 24.7%, OW = 29.0% and OB = 40%; P = 0.06 respectively). Median RFS was significantly higher in N/U group compared to OB patients (150 vs. 76 months respectively, P = 0.03) and was also higher in pCR group compared to non-pCR patients (151 vs. 77 months P = 0.004). Median OS was significantly higher in N/U patients compared to OB patients (N/U = not reached, OW = 211 and OB = 114 months; P = 0.01) and was also higher in pCR group compared to non-pCR patients (not reached vs. 211 months P = 0.04). In Cox regression analysis; pCR, histopathological grade and TNBC were found as independent prognostic factors on OS (HR, 0.29; 95% CI, 0.11 to 0.79, P = 0.015, HR, 2.09; 95% CI, 1.14 to 3.83, P = 0.017, HR, 1.95; 95% CI, 1.01 to 3.77, P = 0.046, respectively). CONCLUSION It was observed that obesity was an important independent prognostic factor which has an adverse effect on pCR. Moreover it causes decreasing RFS and OS in BC patients who had received NAC. The probability of inefficient treatment in obese patients should be considered.
Collapse
Affiliation(s)
- Fatih Karatas
- Diskapi Yildirim Beyazit Education and Training Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Gokmen Umut Erdem
- Ankara Numune Education and Training Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Suleyman Sahin
- Diskapi Yildirim Beyazit Education and Training Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Aydin Aytekin
- Gazi University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Deniz Yuce
- Hacettepe University Faculty of Medicine, Department of Preventive Oncology, Ankara, Turkey
| | - Ali R Sever
- Hacettepe University Faculty of Medicine, Department of Radiology, Ankara, Turkey
| | - Taner Babacan
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Ozturk Ates
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Yavuz Ozisik
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey
| | - Kadri Altundag
- Hacettepe University Faculty of Medicine, Department of Medical Oncology, Ankara, Turkey.
| |
Collapse
|
20
|
Wang Y, Wei J, Li L, Fan C, Sun Y. Combined Use of Metformin and Everolimus Is Synergistic in the Treatment of Breast Cancer Cells. Oncol Res 2016; 22:193-201. [PMID: 26351208 PMCID: PMC7838426 DOI: 10.3727/096504015x14348950540999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Everolimus inhibits mammalian target of rapamycin (mTOR) and leads to decreased protein synthesis and decreased cancer cell proliferation in many experimental systems. Adenosine 5′-monophosphate-activated protein kinase (AMPK) activators such as metformin have similar actions in keeping with the TSC2/1 pathway linking activation of AMPK to inhibition of mTOR. Histopathological and biochemical studies of breast cancer show frequent dysregulation of the AMPK and the mTOR pathway. Therefore, we investigated the efficacy of the mTOR inhibitor everolimus and metformin in the treatment of breast cancer cells. This study evaluated the in vitro and in vivo effects of everolimus alone or in combination with metformin on breast cancer cells. MTT assay was used to quantify the inhibitory effect of the drugs on breast cancer cells in vitro. SCID mice injected with HCC1428 cells followed by different treatments were used to assess the in vivo efficacy of different agents. Data showed that the combination of everolimus and metformin exerted synergistic inhibitory effects on the growth of breast cancer cells both in culture and in a mouse xenograft model. Further, this combination abrogated S6 and 4EBP1phosphorylation. Collectively, we suggest that the combination of everolimus and metformin may be an effective regimen for treatment of breast cancer, hence warranting further evaluation of the combination in the clinic.
Collapse
Affiliation(s)
- Yunshan Wang
- Department of Human Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, Shandong, China
| | | | | | | | | |
Collapse
|
21
|
Imaoka T, Nishimura M, Daino K, Morioka T, Nishimura Y, Uemura H, Akimoto K, Furukawa Y, Fukushi M, Wakabayashi K, Mutoh M, Shimada Y. A Rat Model to Study the Effects of Diet-Induced Obesity on Radiation-Induced Mammary Carcinogenesis. Radiat Res 2016; 185:505-15. [PMID: 27135968 DOI: 10.1667/rr14309.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
A detailed understanding of the relationship between radiation-induced breast cancer and obesity is needed for appropriate risk management and to prevent the development of a secondary cancer in patients who have been treated with radiation. Our goal was to develop an animal model to study the relationship by combining two existing Sprague-Dawley rat models of radiation-induced mammary carcinogenesis and diet-induced obesity. Female rats were fed a high-fat diet for 4 weeks and categorized as obesity prone or obesity resistant based on their body weight at 7 weeks of age, at which time the rats were irradiated with 4 Gy. Control rats were fed a standard diet and irradiated at the same time and in the same manner. All rats were maintained on their initial diets and assessed for palpable mammary cancers once a week for the next 30 weeks. The obesity-prone rats were heavier than those in the other groups. The obesity-prone rats were also younger than the other animals at the first detection of mammary carcinomas and their carcinoma weights were greater. A tendency toward higher insulin and leptin blood levels were observed in the obesity-prone rats compared to the other two groups. Blood angiotensin II levels were elevated in the obesity-prone and obesity-resistant rats. Genes related to translation and oxidative phosphorylation were upregulated in the carcinomas of obesity-prone rats. Expression profiles from human breast cancers were used to validate this animal model. As angiotensin is potentially an important factor in obesity-related morbidities and breast cancer, a second set of rats was fed in a similar manner, irradiated and then treated with an angiotensin-receptor blocker, losartan and candesartan. Neither blocker altered mammary carcinogenesis; analyses of losartan-treated animals indicated that expression of renin in the renal cortex and of Agtr1a (angiotensin II receptor, type 1) in cancer tissue was significantly upregulated, suggesting the presence of compensating mechanisms for blocking angiotensin-receptor signaling. Thus, obesity-related elevation of insulin and leptin blood levels and an increase in available energy may facilitate sustained protein synthesis in cancer cells, which is required for rapid cancer development.
Collapse
Affiliation(s)
- Tatsuhiko Imaoka
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,b Radiation Effect Accumulation and Prevention Project, Fukushima Project Headquarters, NIRS, Chiba, Japan
| | - Mayumi Nishimura
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,b Radiation Effect Accumulation and Prevention Project, Fukushima Project Headquarters, NIRS, Chiba, Japan
| | - Kazuhiro Daino
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,b Radiation Effect Accumulation and Prevention Project, Fukushima Project Headquarters, NIRS, Chiba, Japan
| | - Takamitsu Morioka
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,b Radiation Effect Accumulation and Prevention Project, Fukushima Project Headquarters, NIRS, Chiba, Japan
| | - Yukiko Nishimura
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan
| | - Hiroji Uemura
- c Department of Urology and Renal Transplantation, Yokohama City University Medical Center, Yokohama, Japan
| | - Kenta Akimoto
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,d Division of Radiological Sciences, Faculty of Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yuki Furukawa
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,d Division of Radiological Sciences, Faculty of Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Masahiro Fukushi
- d Division of Radiological Sciences, Faculty of Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Keiji Wakabayashi
- e Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan; and
| | - Michihiro Mutoh
- f Epidemiology and Prevention Division, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | - Yoshiya Shimada
- a Radiobiology for Children's Health Program, Research Center for Radiation Protection, National Institute of Radiological Sciences (NIRS) Chiba, Japan;,b Radiation Effect Accumulation and Prevention Project, Fukushima Project Headquarters, NIRS, Chiba, Japan
| |
Collapse
|
22
|
Imaoka T, Ishii N, Kawaguchi I, Homma-Takeda S, Doi K, Daino K, Nakanishi I, Tagami K, Kokubo T, Morioka T, Hosoki A, Takabatake M, Yoshinaga S. Biological measures to minimize the risk of radiotherapy-associated second cancer: A research perspective. Int J Radiat Biol 2016; 92:289-301. [DOI: 10.3109/09553002.2016.1152413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Tatsuhiko Imaoka
- National Institute of Radiological Sciences, Radiobiology for Children's Health Program, Research Center for Radiation Protection, Chiba, Japan
- Radiation Effect Accumulation and Prevention Project, Fukushima Projects Headquarters, Chiba, Japan
| | - Nobuyoshi Ishii
- Waste Management Research Team, Research Center for Radiation Protection, Chiba, Japan
| | - Isao Kawaguchi
- Regulatory Sciences Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Shino Homma-Takeda
- National Institute of Radiological Sciences, Radiobiology for Children's Health Program, Research Center for Radiation Protection, Chiba, Japan
- Radiation Effect Accumulation and Prevention Project, Fukushima Projects Headquarters, Chiba, Japan
| | - Kazutaka Doi
- Regulatory Sciences Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
- Project for Human Health, Fukushima Projects Headquarters, National Institute of Radiological Sciences, Chiba, Japan
| | - Kazuhiro Daino
- National Institute of Radiological Sciences, Radiobiology for Children's Health Program, Research Center for Radiation Protection, Chiba, Japan
- Radiation Effect Accumulation and Prevention Project, Fukushima Projects Headquarters, Chiba, Japan
| | - Ikuo Nakanishi
- Advanced Radiation Biology Research Program, Research Center for Charged Particle Therapy, National Institute of Radiological Sciences, Chiba, Japan
| | - Keiko Tagami
- Waste Management Research Team, Research Center for Radiation Protection, Chiba, Japan
| | - Toshiaki Kokubo
- Department of Technical Support and Development, Research Development and Support Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Takamitsu Morioka
- National Institute of Radiological Sciences, Radiobiology for Children's Health Program, Research Center for Radiation Protection, Chiba, Japan
- Radiation Effect Accumulation and Prevention Project, Fukushima Projects Headquarters, Chiba, Japan
| | - Ayaka Hosoki
- Radiation Effect Accumulation and Prevention Project, Fukushima Projects Headquarters, Chiba, Japan
| | - Masaru Takabatake
- National Institute of Radiological Sciences, Radiobiology for Children's Health Program, Research Center for Radiation Protection, Chiba, Japan
| | - Shinji Yoshinaga
- Regulatory Sciences Research Program, Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
- Project for Human Health, Fukushima Projects Headquarters, National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
23
|
Ma LX, Bulsara MK, Tan SY, Vardy J. Body weight management in overweight and obese breast cancer survivors. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2016. [DOI: 10.1002/14651858.cd012110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Li Xin Ma
- Concord Repatriation General Hospital; Concord Cancer Centre; Hospital Rd, Concord, Sydney New South Wales Australia 2139
- Hebei University; Nutrition and Food Hygiene Department; No. 342 East Yuhua Rd Baoding Hebei Province China 071000
| | - Max K Bulsara
- The University of Notre Dame Australia; Institute for Health Research; 19 Mouat Street PO Box 1125 Fremantle WA Australia 6959
| | - Sim Yee Tan
- Concord Repatriation General Hospital; Concord Cancer Centre; Hospital Rd, Concord, Sydney New South Wales Australia 2139
- Concord Repatriation General Hospital; Nutrition and Dietetics Department; Building 38 Hospital Road Concord NSW Australia 2139
- University of Sydney; Sydney Medical School; Sydney Australia
| | - Janette Vardy
- Concord Repatriation General Hospital; Concord Cancer Centre; Hospital Rd, Concord, Sydney New South Wales Australia 2139
- University of Sydney; Sydney Medical School; Sydney Australia
| |
Collapse
|
24
|
Incio J, Tam J, Rahbari NN, Suboj P, McManus DT, Chin SM, Vardam TD, Batista A, Babykutty S, Jung K, Khachatryan A, Hato T, Ligibel JA, Krop IE, Puchner SB, Schlett CL, Hoffmman U, Ancukiewicz M, Shibuya M, Carmeliet P, Soares R, Duda DG, Jain RK, Fukumura D. PlGF/VEGFR-1 Signaling Promotes Macrophage Polarization and Accelerated Tumor Progression in Obesity. Clin Cancer Res 2016; 22:2993-3004. [PMID: 26861455 DOI: 10.1158/1078-0432.ccr-15-1839] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/19/2016] [Indexed: 12/25/2022]
Abstract
PURPOSE Obesity promotes pancreatic and breast cancer progression via mechanisms that are poorly understood. Although obesity is associated with increased systemic levels of placental growth factor (PlGF), the role of PlGF in obesity-induced tumor progression is not known. PlGF and its receptor VEGFR-1 have been shown to modulate tumor angiogenesis and promote tumor-associated macrophage (TAM) recruitment and activity. Here, we hypothesized that increased activity of PlGF/VEGFR-1 signaling mediates obesity-induced tumor progression by augmenting tumor angiogenesis and TAM recruitment/activity. EXPERIMENTAL DESIGN We established diet-induced obese mouse models of wild-type C57BL/6, VEGFR-1 tyrosine kinase (TK)-null, or PlGF-null mice, and evaluated the role of PlGF/VEGFR-1 signaling in pancreatic and breast cancer mouse models and in human samples. RESULTS We found that obesity increased TAM infiltration, tumor growth, and metastasis in pancreatic cancers, without affecting vessel density. Ablation of VEGFR-1 signaling prevented obesity-induced tumor progression and shifted the tumor immune environment toward an antitumor phenotype. Similar findings were observed in a breast cancer model. Obesity was associated with increased systemic PlGF, but not VEGF-A or VEGF-B, in pancreatic and breast cancer patients and in various mouse models of these cancers. Ablation of PlGF phenocopied the effects of VEGFR-1-TK deletion on tumors in obese mice. PlGF/VEGFR-1-TK deletion prevented weight gain in mice fed a high-fat diet, but exacerbated hyperinsulinemia. Addition of metformin not only normalized insulin levels but also enhanced antitumor immunity. CONCLUSIONS Targeting PlGF/VEGFR-1 signaling reprograms the tumor immune microenvironment and inhibits obesity-induced acceleration of tumor progression. Clin Cancer Res; 22(12); 2993-3004. ©2016 AACR.
Collapse
Affiliation(s)
- Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal. Department of Internal Medicine, Hospital S. João, Porto, Portugal
| | - Josh Tam
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nuh N Rahbari
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Surgery, Dresden University of Technology, Dresden, Germany
| | - Priya Suboj
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Botany and Biotechnology, St. Xaviers College, Thumba, Trivandrum, Kerala, India
| | - Dan T McManus
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. University of Massachusetts, Boston, Massachusetts
| | - Shan M Chin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Trupti D Vardam
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Mayo Clinic College of Medicine, Scottsdale, Arizona
| | - Ana Batista
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suboj Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Zoology, Mar Ivanios College, Nalanchira, Trivandrum, Kerala, India
| | - Keehoon Jung
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anna Khachatryan
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Surgery, KeioUniversity School of Medicine, Tokyo, Japan
| | - Jennifer A Ligibel
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Ian E Krop
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Stefan B Puchner
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Biomedical Imaging and Image-Guided Therapy, Medical University Vienna, Vienna, Austria
| | - Christopher L Schlett
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Udo Hoffmman
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marek Ancukiewicz
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. PAREXEL International, Billerica, Massachusetts
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Takasaki, Gunma, Japan
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, Department of Oncology, K.U. Leuven and VIB, Leuven, Belgium
| | - Raquel Soares
- I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Dan G Duda
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
So EY, Ouchi M, Cuesta-Sancho S, Olson SL, Reif D, Shimomura K, Ouchi T. Tumor suppression by resistant maltodextrin, Fibersol-2. Cancer Biol Ther 2016; 16:460-5. [PMID: 25692338 DOI: 10.1080/15384047.2015.1009269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Resistant maltodextrin Fibersol-2 is a soluble and fermentable dietary fiber that is Generally Recognized As Safe (GRAS) in the United States. We tested whether Fibersol-2 contains anti-tumor activity. Human colorectal cancer cell line, HCT116, and its isogenic cells were treated with FIbersol-2. Tumor growth and tumorigenesis were studied in vitro and in vivo. Apoptotic pathway and generation of reactive oxygen species (ROS) were investigated. We discovered that Fibersol-2 significantly inhibits tumor growth of HCT116 cells by inducing apoptosis. Fibersol-2 strongly induces mitochondrial ROS and Bax-dependent cleavage of caspase 3 and 9, which is shown by isogenic HCT116 variants. Fibersol-2 induces phosphorylation of Akt, mTOR in parental HCT116 cells, but not in HCT116 deficient for Bax or p53. It prevents growth of tumor xenograft without any apparent signs of toxicity in vivo. These results identify Fibersol-2 as a mechanism-based dietary supplement agent that could prevent colorectal cancer development.
Collapse
Affiliation(s)
- Eui Young So
- a Department of Cancer Genetics ; Roswell Park Cancer Institute ; Buffalo , NY USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Patterson RE, Marinac CR, Natarajan L, Hartman SJ, Cadmus-Bertram L, Flatt SW, Li H, Parker B, Oratowski-Coleman J, Villaseñor A, Godbole S, Kerr J. Recruitment strategies, design, and participant characteristics in a trial of weight-loss and metformin in breast cancer survivors. Contemp Clin Trials 2015; 47:64-71. [PMID: 26706665 DOI: 10.1016/j.cct.2015.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 01/03/2023]
Abstract
Weight loss and metformin are hypothesized to improve breast cancer outcomes; however the joint impacts of these treatments have not been investigated. Reach for Health is a randomized trial using a 2 × 2 factorial design to investigate the effects of weight loss and metformin on biomarkers associated with breast cancer prognosis among overweight/obese postmenopausal breast cancer survivors. This paper describes the trial recruitment strategies, design, and baseline sample characteristics. Participants were randomized in equal numbers to (1) placebo, (2) metformin, (3) weight loss intervention and placebo, or (4) weight-loss intervention and metformin. The lifestyle intervention was a personalized, telephone-based program targeting a 7% weight-loss in the intervention arm. The metformin dose was 1500 mg/day. The duration of the intervention was 6 months. Main outcomes were biomarkers representing 3 metabolic systems putatively related to breast cancer mortality: glucoregulation, inflammation, and sex hormones. Between August 2011 and May 2015, we randomized 333 breast cancer survivors. Mass mailings from the California Cancer Registry were the most successful recruitment strategy with over 25,000 letters sent at a cost of $191 per randomized participant. At baseline, higher levels of obesity were significantly associated with worse sleep disturbance and impairment scores, lower levels of physical activity and higher levels of sedentary behavior, hypertension, hypercholesterolemia, and lower quality of life (p<0.05 for all). These results illustrate the health burden of obesity. Results of this trial will provide mechanistic data on biological pathways and circulating biomarkers associated with lifestyle and pharmacologic interventions to improve breast cancer prognosis.
Collapse
Affiliation(s)
- Ruth E Patterson
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA; Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA.
| | - Catherine R Marinac
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA; Graduate School of Public Health, San Diego State University, San Diego, CA, USA
| | - Loki Natarajan
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA; Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Sheri J Hartman
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA; Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | | | - Shirley W Flatt
- Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Hongying Li
- Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Barbara Parker
- Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | | | - Adriana Villaseñor
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA; Moores UC San Diego Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Suneeta Godbole
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA
| | - Jacqueline Kerr
- Department of Family Medicine and Public Health, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Incio J, Suboj P, Chin SM, Vardam-Kaur T, Liu H, Hato T, Babykutty S, Chen I, Deshpande V, Jain RK, Fukumura D. Metformin Reduces Desmoplasia in Pancreatic Cancer by Reprogramming Stellate Cells and Tumor-Associated Macrophages. PLoS One 2015; 10:e0141392. [PMID: 26641266 PMCID: PMC4671732 DOI: 10.1371/journal.pone.0141392] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 10/06/2015] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is a highly desmoplastic tumor with a dismal prognosis for most patients. Fibrosis and inflammation are hallmarks of tumor desmoplasia. We have previously demonstrated that preventing the activation of pancreatic stellate cells (PSCs) and alleviating desmoplasia are beneficial strategies in treating PDAC. Metformin is a widely used glucose-lowering drug. It is also frequently prescribed to diabetic pancreatic cancer patients and has been shown to associate with a better outcome. However, the underlying mechanisms of this benefit remain unclear. Metformin has been found to modulate the activity of stellate cells in other disease settings. In this study, we examine the effect of metformin on PSC activity, fibrosis and inflammation in PDACs. Methods/Results In overweight, diabetic PDAC patients and pre-clinical mouse models, treatment with metformin reduced levels of tumor extracellular matrix (ECM) components, in particular hyaluronan (HA). In vitro, we found that metformin reduced TGF-ß signaling and the production of HA and collagen-I in cultured PSCs. Furthermore, we found that metformin alleviates tumor inflammation by reducing the expression of inflammatory cytokines including IL-1β as well as infiltration and M2 polarization of tumor-associated macrophages (TAMs) in vitro and in vivo. These effects on macrophages in vitro appear to be associated with a modulation of the AMPK/STAT3 pathway by metformin. Finally, we found in our preclinical models that the alleviation of desmoplasia by metformin was associated with a reduction in ECM remodeling, epithelial-to-mesenchymal transition (EMT) and ultimately systemic metastasis. Conclusion Metformin alleviates the fibro-inflammatory microenvironment in obese/diabetic individuals with pancreatic cancer by reprogramming PSCs and TAMs, which correlates with reduced disease progression. Metformin should be tested/explored as part of the treatment strategy in overweight diabetic PDAC patients.
Collapse
Affiliation(s)
- Joao Incio
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Internal Medicine, Hospital S. Joao, Porto, Portugal
- I3S, Institute for Innovation and Research in Heath, Metabolism, Nutrition and Endocrinology group, Biochemistry Department, Faculty of Medicine, Porto University, Porto, Portugal
| | - Priya Suboj
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Botany and Biotechnology, St. Xaviers College, Thumba, Trivandrum, Kerala, India
| | - Shan M. Chin
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Trupti Vardam-Kaur
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hao Liu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Program of Biology and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tai Hato
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Suboj Babykutty
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Zoology, Mar Ivanios College, Nalanchira, Trivandrum, Kerala, India
| | - Ivy Chen
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RKJ); (DF)
| |
Collapse
|
28
|
Forouzandeh F, Salazar G, Patrushev N, Xiong S, Hilenski L, Fei B, Alexander RW. Metformin beyond diabetes: pleiotropic benefits of metformin in attenuation of atherosclerosis. J Am Heart Assoc 2015; 3:e001202. [PMID: 25527624 PMCID: PMC4338706 DOI: 10.1161/jaha.114.001202] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Clinical studies show that metformin attenuates all‐cause mortality and myocardial infarction compared with other medications for type 2 diabetes, even at similar glycemic levels. However, there is paucity of data in the euglycemic state on the vasculoprotective effects of metformin. The objectives of this study are to evaluate the effects of metformin on ameliorating atherosclerosis. Methods and Results Using ApoE−/− C57BL/6J mice, we found that metformin attenuates atherosclerosis and vascular senescence in mice fed a high‐fat diet and prevents the upregulation of angiotensin II type 1 receptor by a high‐fat diet in the aortas of mice. Thus, considering the known deleterious effects of angiotensin II mediated by angiotensin II type 1 receptor, the vascular benefits of metformin may be mediated, at least in part, by angiotensin II type 1 receptor downregulation. Moreover, we found that metformin can cause weight loss without hypoglycemia. We also found that metformin increases the antioxidant superoxide dismutase‐1. Conclusion Pleiotropic effects of metformin ameliorate atherosclerosis and vascular senescence.
Collapse
|
29
|
Wang X, Simpson ER, Brown KA. Aromatase overexpression in dysfunctional adipose tissue links obesity to postmenopausal breast cancer. J Steroid Biochem Mol Biol 2015. [PMID: 26209254 DOI: 10.1016/j.jsbmb.2015.07.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The number of breast cancer cases has increased in the last a few decades and this is believed to be associated with the increased prevalence of obesity worldwide. The risk of breast cancer increases with age beyond menopause and the relationship between obesity and the risk of breast cancer in postmenopausal women is well established. The majority of postmenopausal breast cancers are estrogen receptor (ER) positive and estrogens produced in the adipose tissue promotes tumor formation. Obesity results in the secretion of inflammatory factors that stimulate the expression of the aromatase enzyme, which converts androgens into estrogens in the adipose tissue. Evidence demonstrating a link between obesity and breast cancer has led to the investigation of metabolic pathways as novel regulators of estrogen production, including pathways that can be targeted to inhibit aromatase specifically within the breast. This review aims to present some of the key findings in this regard.
Collapse
Affiliation(s)
- Xuyi Wang
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of biochemistry & Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
30
|
Ko KP, Ma SH, Yang JJ, Hwang Y, Ahn C, Cho YM, Noh DY, Park BJ, Han W, Park SK. Metformin intervention in obese non-diabetic patients with breast cancer: phase II randomized, double-blind, placebo-controlled trial. Breast Cancer Res Treat 2015; 153:361-70. [DOI: 10.1007/s10549-015-3519-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/21/2015] [Indexed: 11/25/2022]
|
31
|
Vici P, Crispo A, Giordano A, Di Lauro L, Sperati F, Terrenato I, Pizzuti L, Sergi D, Mottolese M, Botti C, Grimaldi M, Capasso I, D'Aiuto G, Di Bonito M, Di Paola F, Maugeri-Saccà M, Montella M, Barba M. Anthropometric, metabolic and molecular determinants of human epidermal growth factor receptor 2 expression in luminal B breast cancer. J Cell Physiol 2015; 230:1708-12. [PMID: 25510909 DOI: 10.1002/jcp.24891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022]
Abstract
Genomic and trascriptomic profiling has recently contributed details to the characterization of luminal B breast cancer. We explored the contribution of anthropometric, metabolic, and molecular determinants to the multifaceted heterogeneity of this breast cancer subtype, with a specific focus on the association between body mass index (BMI), pre-treatment fasting glucose, hormone receptors, and expression of human epidermal growth factor receptor 2 (HER2). Extensively annotated specimens were obtained from 154 women with luminal B breast cancer diagnosed at two Italian comprehensive cancer centres. Participants' characteristics were descriptively analyzed overall and by HER2 status (positive vs. negative). BMI (<25 vs ≥25), pre-treatment fasting glucose (<median value of 94 mg/dl vs. ≥94) and percentage of hormone receptors were tested for association with HER2 expression in regression models. In univariate models, BMI, fasting glucose and, at a lesser extent, percentage of estrogen receptors (ER) were significantly and inversely associated with HER2 expression (OR: 0.32, 95% CI: 0.16-0.66; 0.43, 0.23-0.82; 0.96, 0.94-0.97, respectively). The multivariate models confirmed the protective role of BMI and ER on HER2 expression, with luminal B HER2 positive patients being significantly less frequent among women within the highest category of BMI and percentage expression of ER compared with their counterparts (OR: 0.22, 95% CI: 0.09-0.53; 0.95, 0.93-0.97). In conclusions, BMI and percentage of ER representation are inversely associated with HER2 expression in luminal B breast cancers. Upon confirmatory findings, this might help identify patient subgroups who may best benefit from the use of interventions targeting insulin resistance in well depicted breast cancer scenarios.
Collapse
Affiliation(s)
- Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kim HJ, Kwon H, Lee JW, Kim HJ, Lee SB, Park HS, Sohn G, Lee Y, Koh BS, Yu JH, Son BH, Ahn SH. Metformin increases survival in hormone receptor-positive, HER2-positive breast cancer patients with diabetes. Breast Cancer Res 2015; 17:64. [PMID: 25935404 PMCID: PMC4504447 DOI: 10.1186/s13058-015-0574-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
Introduction Metformin use has recently been observed to decrease both the rate and mortality of breast cancer. Our study was aim to determine whether metformin use is associated with survival in diabetic breast cancer patients by breast cancer subtype and systemic treatment. Methods Data from the Asan Medical Center Breast Cancer Database from 1997 to 2007 were analyzed. The study cohort comprised 6,967 nondiabetic patients, 202 diabetic patients treated with metformin, and 184 diabetic patients that did not receive metformin. Patients who were divided into three groups by diabetes status and metformin use were also divided into four subgroups by hormone receptor and HER2-neu status. Results In Kaplan-Meier analysis, the metformin group had a significantly better overall and cancer specific survival outcome compared with non metformin diabetic group (P <0.005 for both). There was no difference in survival between the nondiabetic and metformin groups. In multivariate analysis, Compared with metformin group, patients who did not receive metformin tended to have a higher risk of metastasis with HR 5.37 (95 % CI, 1.88 to 15.28) and breast cancer death with HR 6.51 (95 % CI, 1.88 to 15.28) on the hormone receptor-positive and HER2-negative breast cancer. The significant survival benefit of metformin observed in diabetic patients who received chemotherapy and endocrine therapy (HR for disease free survival 2.14; 95 % CI 1.14 to 4.04) was not seen in diabetic patients who did not receive these treatments. Conclusion Patients receiving metformin treatment when breast cancer diagnosis show a better prognosis only if they have hormone receptor-positive, HER2-positive tumors. Metformin treatment might provide a survival benefit when added to systemic therapy in diabetic patients.
Collapse
Affiliation(s)
- Hee Jeong Kim
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hyunwook Kwon
- Division of Vascular Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jong Won Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Sae Byul Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Hee Sung Park
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Guiyun Sohn
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Yura Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Beom Seok Koh
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Jong Han Yu
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Byung Ho Son
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| | - Sei Hyun Ahn
- Division of Breast and Endocrine Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
33
|
Abstract
Epidemiological data suggest a close link between obesity and breast cancer, the most frequently occurring cancer in women. The metabolic syndrome is typically associated with abdominal obesity and comprises disturbances in glucose and/or lipid metabolism and/or hypertension. Recent studies have established a specific association between the metabolic syndrome - as well as its components - and breast cancer, indicating both an increased risk of developing breast cancer and a poorer prognosis. In premenopausal women, obesity might have a protective effect only on receptor-positive tumors, whereas a positive association was observed between obesity/abdominal obesity and an increased risk of triple-negative breast cancer (TNBC). Overall survival and disease-free survival were reported to be significantly shorter in premenopausal obese women with TNBC compared to non-obese women, but these results are still inconsistent and need further research. The metabolic syndrome is characterized by a state of insulin resistance/hyperinsulinemia and subacute chronic inflammation, with both conditions offering a plausible mechanistic link towards breast cancer. Thus, in addition to their increased risk of cardiovascular morbidity and mortality, women with this syndrome represent a group at elevated risk of developing breast cancer and with poorer prognosis.
Collapse
Affiliation(s)
- Dagmar Hauner
- Else Kröner-Fresenius-Center for Nutritional Medicine, Klinikum rechts der Isar, Technical University Munich, Germany
| | - Hans Hauner
- Else Kröner-Fresenius-Center for Nutritional Medicine, Klinikum rechts der Isar, Technical University Munich, Germany
| |
Collapse
|
34
|
Modeling of hypo/hyperglycemia and their impact on breast cancer progression related molecules. PLoS One 2014; 9:e113103. [PMID: 25401697 PMCID: PMC4234670 DOI: 10.1371/journal.pone.0113103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 10/13/2014] [Indexed: 01/08/2023] Open
Abstract
Breast cancer (BC) arises commonly in women with metabolic dysfunction. The underlying mechanism by which glycemic load can exert its action on tumor metastasis is under investigated. In this study we showed that glycemic microenvironment alters the expression of three classes of proteins, VEGF and its receptors, cell to cell, and cell to extracellular matrix (ECM) adhesion proteins in MDA-MB-231 parental cells and its two metastatic variants to the bone and brain (MDA-MB-231BO and MDA-MB-231BR, respectively). Using western blotting, we showed that VEGFR2 levels were higher in these variant cells and persisted in the cells under extreme hypoglycemia. Hypoglycemia did not alter VEGFR2 expression per se but rather suppressed its posttranslational glycosylation. This was reversed rapidly upon the restoration of glucose, and cyclohexamide (CHX) treatment demonstrated that this deglycosylated VEGFR2 was not a product of de-novo protein synthesis. VEGFR2 co-receptor Neuropilin-1 was up-regulated four-fold in all MDA-MB-231 cells (parental and two variants) compared to VEGFR2 expression, and was also susceptible to glycemic changes but resistant to CHX treatment for up to 72 hrs. Hypoglycemia also resulted in a significant decrease in specific catenin, cadherin, and integrin proteins, as well as cellular proliferation and colony forming ability. However, MDA-MB-231BR cells showed a unique sensitivity to hypo/hyperglycemia in terms of morphological changes, colony formation ability, integrin β3 expression and secreted VEGF levels. In conclusion, this study can be translated clinically to provide insight into breast cancer cell responses to glycemic levels relevant for our understanding of the interaction between diabetes and cancer.
Collapse
|
35
|
Bao B, Azmi AS, Ali S, Zaiem F, Sarkar FH. Metformin may function as anti-cancer agent via targeting cancer stem cells: the potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:59. [PMID: 25333034 DOI: 10.3978/j.issn.2305-5839.2014.06.05] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/27/2014] [Indexed: 12/13/2022]
Abstract
Metformin is one of the most used diabetic drugs for the management of type II diabetes mellitus (DM) in the world. Increased numbers of epidemiological and clinical studies have provided convincing evidence supporting the role of metformin in the development and progression of a variety of human tumors including breast and pancreatic cancer. Substantial pre-clinical evidence from in vitro and in vivo experimental studies strongly suggests that metformin has an anti-cancer activity mediated through the regulation of several cell signaling pathways including activation of AMP kinase (AMPK), and other direct and indirect mechanisms; however, the detailed mechanism(s) has not yet been fully understood. The concept of cancer stem cells (CSCs) has gained significant attention in recent years due its identification and defining its clinical implications in many different tumors including breast cancer and pancreatic cancer. In this review, we will discuss the protective role of metformin in the development of breast and pancreatic cancers. We will further discuss the role of metformin as an anti-cancer agent, which is in part mediated through targeting CSCs. Finally, we will discuss the potential role of metformin in the modulation of tumor-associated or CSC-associated microRNAs (miRNAs) as part of the novel mechanism of action of metformin in the development and progression of breast and pancreatic cancers.
Collapse
Affiliation(s)
- Bin Bao
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Asfar S Azmi
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Shadan Ali
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Feras Zaiem
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Fazlul H Sarkar
- 1 Department of Pathology, 2 Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
36
|
Cufí S, Corominas-Faja B, Lopez-Bonet E, Bonavia R, Pernas S, López IÁ, Dorca J, Martínez S, López NB, Fernández SD, Cuyàs E, Visa J, Rodríguez-Gallego E, Quirantes-Piné R, Segura-Carretero A, Joven J, Martin-Castillo B, Menendez JA. Dietary restriction-resistant human tumors harboring the PIK3CA-activating mutation H1047R are sensitive to metformin. Oncotarget 2014; 4:1484-95. [PMID: 23986086 PMCID: PMC3824528 DOI: 10.18632/oncotarget.1234] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer cells expressing constitutively active phosphatidylinositol-3 kinase (PI3K) are proliferative regardless of the absence of insulin, and they form dietary restriction (DR)-resistant tumors in vivo. Because the binding of insulin to its receptors activates the PI3K/AKT/mammalian target of rapamycin (mTOR) signaling cascade, activating mutations in the PIK3CA oncogene may determine tumor response to DR-like pharmacological strategies targeting the insulin and mTOR pathways. The anti-diabetic drug metformin is a stereotypical DR mimetic that exerts its anti-cancer activity through a dual mechanism involving insulin-related (systemic) and mTOR-related (cell-autonomous) effects. However, it remains unclear whether PIK3CA-activating mutations might preclude the anti-cancer activity of metformin in vivo. To model the oncogenic PIK3CA-driven early stages of cancer, we used the clonal breast cancer cell line MCF10DCIS.com, which harbors the gain-of-function H1047R hot-spot mutation in the catalytic domain of the PI3KCA gene and has been shown to form DR-refractory xenotumors. To model PIK3CA-activating mutations in late stages of cancer, we took advantage of the isogenic conversion of a PIK3CA-wild-type tumor into a PIK3CA H1047R-mutated tumor using the highly metastatic colorectal cancer cell line SW48. MCF10DCIS.com xenotumors, although only modestly affected by treatment with oral metformin (approximately 40% tumor growth inhibition), were highly sensitive to the intraperitoneal (i.p.) administration of metformin, the anti-cancer activity of which increased in a time-dependent manner and reached >80% tumor growth inhibition by the end of the treatment. Metformin treatment via the i.p. route significantly reduced the proliferation factor mitotic activity index (MAI) and decreased tumor cellularity in MCF10DCIS.com cancer tissues. Whereas SW48-wild-type (PIK3CA+/+) cells rapidly formed metformin-refractory xenotumors in mice, ad libitum access to water containing metformin significantly reduced the growth of SW48-mutated (PIK3CAH1047R/+) xenotumors by approximately 50%. Thus, metformin can no longer be considered as a bona fide DR mimetic, at least in terms of anti-cancer activity, because tumors harboring the insulin-unresponsive, DR-resistant, PIK3CA-activating mutation H1047R remain sensitive to the anti-tumoral effects of the drug. Given the high prevalence of PIK3CA mutations in human carcinomas and the emerging role of PIK3CA mutation status in the treatment selection process, these findings might have a significant impact on the design of future trials evaluating the potential of combining metformin with targeted therapy.
Collapse
Affiliation(s)
- Sílvia Cufí
- Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Neilson HK, Conroy SM, Friedenreich CM. The Influence of Energetic Factors on Biomarkers of Postmenopausal Breast Cancer Risk. Curr Nutr Rep 2013; 3:22-34. [PMID: 24563822 PMCID: PMC3921460 DOI: 10.1007/s13668-013-0069-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Strong and consistent evidence exists that physical activity reduces breast cancer risk by 10-25 %, and several proposed biologic mechanisms have now been investigated in randomized, controlled, exercise intervention trials. Leading hypothesized mechanisms relating to postmenopausal breast cancer include adiposity, endogenous sex hormones, insulin resistance, and chronic low-grade inflammation. In addition, other pathways are emerging as potentially important, including those involving oxidative stress and telomere length, global DNA hypomethylation, immune function, and vitamin D exposure. Recent exercise trials in overweight/obese postmenopausal women implicate weight loss as a mechanism whereby exercise induces favorable changes in circulating estradiol levels and other biomarkers as well. Still it is plausible that some exercise-induced biomarker changes do not require loss of body fat, whereas others depend on abdominal fat loss. We highlight the latest findings from randomized, controlled trials of healthy postmenopausal women, relating exercise to proposed biomarkers for postmenopausal breast cancer risk.
Collapse
Affiliation(s)
- Heather K Neilson
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada
| | - Shannon M Conroy
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada
| | - Christine M Friedenreich
- Department of Population Health Research, CancerControl Alberta, Alberta Health Services, Quarry Park, c/o 10101 Southport Rd SW, Calgary, Alberta T2W 3N2 Canada ; Department of Community Health Sciences, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N2 Canada ; Department of Oncology, Faculty of Medicine, University of Calgary, 1331 29 St. N.W., Calgary, Alberta T2N 4N2 Canada
| |
Collapse
|
38
|
Jain R, Strickler HD, Fine E, Sparano JA. Clinical studies examining the impact of obesity on breast cancer risk and prognosis. J Mammary Gland Biol Neoplasia 2013; 18:257-66. [PMID: 24221746 DOI: 10.1007/s10911-013-9307-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/24/2013] [Indexed: 10/26/2022] Open
Abstract
Obesity is associated with an increased risk of breast cancer, and increased risk of recurrence in women who develop breast cancer. Evidence suggests that the risk of estrogen-receptor (ER)-positive breast cancer is increased in obese postmenopausal women, whereas in premenopausal women the risk of triple negative breast cancer is increased. Nonetheless, the presence of obesity at diagnosis, and possibly weight gain after diagnosis, may independently contribute to an individual's risk of recurrence of both pre- and postmenopausal breast cancer. Factors associated with adiposity that are likely contributing factors include hyperinsulinemia, inflammation, and relative hyperestrogenemia. Some studies suggest that some aromatase inhibitors may be less effective in obese women than lean women. Clinical trials have evaluated pharmacologic (eg, metformin) and dietary/lifestyle interventions to reduce breast cancer recurrence, although these interventions have not been tested in obese women who may be most likely to benefit from them. Further research is required in order to identify adiposity-associated factors driving recurrence, and design clinical trials to specifically test interventions in obese women at highest risk of recurrence.
Collapse
Affiliation(s)
- Rishi Jain
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | | | | |
Collapse
|
39
|
Abstract
This paper proposes entitymetrics to measure the impact of knowledge units. Entitymetrics highlight the importance of entities embedded in scientific literature for further knowledge discovery. In this paper, we use Metformin, a drug for diabetes, as an example to form an entity-entity citation network based on literature related to Metformin. We then calculate the network features and compare the centrality ranks of biological entities with results from Comparative Toxicogenomics Database (CTD). The comparison demonstrates the usefulness of entitymetrics to detect most of the outstanding interactions manually curated in CTD.
Collapse
|
40
|
Blagosklonny MV. Common drugs and treatments for cancer and age-related diseases: revitalizing answers to NCI's provocative questions. Oncotarget 2013; 3:1711-24. [PMID: 23565531 PMCID: PMC3681506 DOI: 10.18632/oncotarget.890] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In 2011, The National Cancer Institute (NCI) has announced 24 provocative questions on cancer. Some of these questions have been already answered in “NCI's provocative questions on cancer: some answers to ignite discussion” (published in Oncotarget, 2011, 2: 1352.) The questions included “Why do many cancer cells die when suddenly deprived of a protein encoded by an oncogene?” “Can we extend patient survival by using approaches that keep tumors static?” “Why are some disseminated cancers cured by chemotherapy alone?” “Can we develop methods to rapidly test interventions for cancer treatment or prevention?” “Can we use our knowledge of aging to enhance prevention or treatment of cancer?” “What is the mechanism by which some drugs commonly and chronically used for other indications protect against cancer?” “How does obesity contribute to cancer risk?” I devoted a single subchapter to each the answer. As expected, the provocative questions were very diverse and numerous. Now I choose and combine, as a single problem, only three last questions, all related to common mechanisms and treatment of age-related diseases including obesity and cancer. Can we use common existing drugs for cancer prevention and treatment? Can we use some targeted “cancer-selective” agents for other diseases and … aging itself.
Collapse
Affiliation(s)
- Mikhail V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA.
| |
Collapse
|
41
|
Xie L, Wang W. Weight control and cancer preventive mechanisms: role of insulin growth factor-1-mediated signaling pathways. Exp Biol Med (Maywood) 2013; 238:127-32. [PMID: 23576795 DOI: 10.1177/1535370213477602] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Overweight and obese not only increase the risk of cardiovascular disease and type-2 diabetes mellitus, but are also now known risk factors for a variety of cancers. Weight control, via dietary calorie restriction and/or exercise, has been demonstrated to be beneficial for cancer prevention in various experimental models, but the underlying mechanisms are still not well defined. Recent studies conducted in a mouse skin carcinogenesis model show that weight loss induced a significant reduction of the circulating levels of insulin growth factor (IGF)-1 and other hormones, including insulin and leptin, resulting in reduced IGF-1-dependent signaling pathways, i.e. Ras-MAPK proliferation and protein kinase B-phosphoinositide 3-kinase (Akt-PI3K) antiapoptosis. Selective targeting IGF-1 to Akt/mammalian target of rapamycin and AMP-activated protein kinase pathways, via negative energy balance, might inactivate cell cycle progression and ultimately suppress tumor development. This review highlights the current studies focused on the major role of reducing IGF-1-activated signaling via weight control as a potential cancer preventive mechanism.
Collapse
Affiliation(s)
- Linglin Xie
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, ND 58202, USA.
| | | |
Collapse
|
42
|
Sanchez-Alvarez R, Martinez-Outschoorn UE, Lamb R, Hulit J, Howell A, Gandara R, Sartini M, Rubin E, Lisanti MP, Sotgia F. Mitochondrial dysfunction in breast cancer cells prevents tumor growth: understanding chemoprevention with metformin. Cell Cycle 2012; 12:172-82. [PMID: 23257779 DOI: 10.4161/cc.23058] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Metformin is a well-established diabetes drug that prevents the onset of most types of human cancers in diabetic patients, especially by targeting cancer stem cells. Metformin exerts its protective effects by functioning as a weak "mitochondrial poison," as it acts as a complex I inhibitor and prevents oxidative mitochondrial metabolism (OXPHOS). Thus, mitochondrial metabolism must play an essential role in promoting tumor growth. To determine the functional role of "mitochondrial health" in breast cancer pathogenesis, here we used mitochondrial uncoupling proteins (UCPs) to genetically induce mitochondrial dysfunction in either human breast cancer cells (MDA-MB-231) or cancer-associated fibroblasts (hTERT-BJ1 cells). Our results directly show that all three UCP family members (UCP-1/2/3) induce autophagy and mitochondrial dysfunction in human breast cancer cells, which results in significant reductions in tumor growth. Conversely, induction of mitochondrial dysfunction in cancer-associated fibroblasts has just the opposite effect. More specifically, overexpression of UCP-1 in stromal fibroblasts increases β-oxidation, ketone body production and the release of ATP-rich vesicles, which "fuels" tumor growth by providing high-energy nutrients in a paracrine fashion to epithelial cancer cells. Hence, the effects of mitochondrial dysfunction are truly compartment-specific. Thus, we conclude that the beneficial anticancer effects of mitochondrial inhibitors (such as metformin) may be attributed to the induction of mitochondrial dysfunction in the epithelial cancer cell compartment. Our studies identify cancer cell mitochondria as a clear target for drug discovery and for novel therapeutic interventions.
Collapse
|
43
|
Choe YJ, Ha TJ, Ko KW, Lee SY, Shin SJ, Kim HS. Anthocyanins in the black soybean (Glycine max L.) protect U2OS cells from apoptosis by inducing autophagy via the activation of adenosyl monophosphate-dependent protein kinase. Oncol Rep 2012; 28:2049-56. [PMID: 22992992 DOI: 10.3892/or.2012.2034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/23/2012] [Indexed: 11/06/2022] Open
Abstract
Anthocyanins (ATCs) have been reported to induce apoptosis in various types of cancer cells, stimulating the development of ATCs as a cancer chemotherapeutic or chemopreventive agent. It was recently reported that ATCs can induce autophagy, however, the mechanism for this remains unclear. In the present report, we carried out mechanistic studies of the mechanism involved in ATC-induced autophagy using ATCs extracted from black soybeans (cv. Cheongja 3, Glycine max L.). ATCs clearly induced hallmarks of autophagy, including LC3 puncta formation and the conversion of LC3-I to LC3-II in U2OS human osteosarcoma cells. The induction of autophagy was accompanied by the phosphorylation of multiple protein kinases including extracellular signal-regulated kinase (ERK)1/2, p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), protein kinase B (AKT) and adenosyl mono-phosphate-dependent protein kinase (AMPK). While chemical inhibitors against ERK1/2, p38 MAPK, JNK and AKT failed to inhibit ATC-induced autophagy, the suppression of AMPK by compound C (CC) as well as siRNA against AMPK reduced ATC-induced autophagy. The treatment of ATCs resulted in a decrease in intracellular ATP contents and the activation of AMPK by AICAR treatment also induced autophagy. It is noteworthy that the reduction of autophagy via the inhibition of AMPK resulted in enhanced apoptosis in ATC-treated cells. In addition, siRNA against forkhead box O3A (FOXO3a), a downstream target of AMPK, suppressed ATC-induced autophagy and p27KIP1 siRNA increased apoptosis in ATC-treated cells. Collectively, it can be concluded that ATCs induce autophagy in U2OS cells via activation of the AMPK-FOXO3a pathway and protect cells from ATC-induced apoptosis via the AMPK-p27KIP1 pathway. These results also suggest that autophagy-modulating agents could contribute to the efficient development of ATCs as anticancer therapy.
Collapse
Affiliation(s)
- Yun-Jeong Choe
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
The hydrolysis of ATP drives virtually all of the energy-requiring processes in living cells. A prerequisite of living cells is that the concentration of ATP needs to be maintained at sufficiently high levels to sustain essential cellular functions. In eukaryotic cells, the AMPK (AMP-activated protein kinase) cascade is one of the systems that have evolved to ensure that energy homoeostasis is maintained. AMPK is activated in response to a fall in ATP, and recent studies have suggested that ADP plays an important role in regulating AMPK. Once activated, AMPK phosphorylates a broad range of downstream targets, resulting in the overall effect of increasing ATP-producing pathways whilst decreasing ATP-utilizing pathways. Disturbances in energy homoeostasis underlie a number of disease states in humans, e.g. Type 2 diabetes, obesity and cancer. Reflecting its key role in energy metabolism, AMPK has emerged as a potential therapeutic target. In the present review we examine the recent progress aimed at understanding the regulation of AMPK and discuss some of the latest developments that have emerged in key areas of human physiology where AMPK is thought to play an important role.
Collapse
|
45
|
Afzal M, Kazmi I, Gupta G, Rahman M, Kimothi V, Anwar F. Preventive effect of Metformin against N-nitrosodiethylamine-initiated hepatocellular carcinoma in rats. Saudi Pharm J 2012; 20:365-70. [PMID: 23960811 DOI: 10.1016/j.jsps.2012.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/31/2012] [Indexed: 12/11/2022] Open
Abstract
Effect of Metformin in chemically induced hepatocarcinogenesis was assessed in Wistar rats. Intraperitoneal administration of chemical carcinogen diethyl nitrosamine (DENA) (200 mg/kg) in single dose elevated the levels of serum glutamate oxaloacetate transaminase (SGOT), serum glutamate pyruvate transaminase (SGPT), alkaline phosphatase (ALP), total cholesterol (TC), triglycerides (TG) and reduced high density lipoproteins (HDL), total proteins (TPR) and blood glucose level in tested animals. Histopathological examinations of the liver tissue showed marked carcinogenicity of the chemical carcinogen. Food and water intake, animal weights and serum albumin (ALB) were also assessed. The animals exposed to DENA showed a significant decrease in the body weights and, there were no significant alterations found in the total bilirubin (TBR) levels and gamma-glutamyltranspeptidase (GGTP), whereas the decreased levels of serum ALB were maintained by Metformin treatment. The elevated levels of serum SGOT, SGPT, ALP, AFP, TC and TG were restored by administration of Metformin in reduced dose (125 mg/kg) daily for 16 weeks p.o. Physiological and biochemical analysis showed the beneficial effects of Metformin in the animals exposed to DENA.
Collapse
Affiliation(s)
- Muhammad Afzal
- Siddhartha Institute of Pharmacy, Dehra Dun, Uttarakhand, India
| | | | | | | | | | | |
Collapse
|
46
|
Berstein LM. Metformin in obesity, cancer and aging: addressing controversies. Aging (Albany NY) 2012; 4:320-9. [PMID: 22589237 PMCID: PMC3384433 DOI: 10.18632/aging.100455] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/29/2012] [Indexed: 12/25/2022]
Abstract
Metformin, an oral anti-diabetic drug, is being considered increasingly for treatment and prevention of cancer, obesity as well as for the extension of healthy lifespan. Gradually accumulating discrepancies about its effect on cancer and obesity can be explained by the shortage of randomized clinical trials, differences between control groups (reference points), gender- and age-associated effects and pharmacogenetic factors. Studies of the potential antiaging effects of antidiabetic biguanides, such as metformin, are still experimental for obvious reasons and their results are currently ambiguous. Here we discuss whether the discrepancies in different studies are merely methodological or inherently related to individual differences in responsiveness to the drug.
Collapse
Affiliation(s)
- Lev M Berstein
- Laboratory of Oncoendocrinology, N.N.Petrov Research Institute of Oncology, St. Petersburg, Russia.
| |
Collapse
|
47
|
What’s new with physical exercise in 2012? SPORT SCIENCES FOR HEALTH 2012. [DOI: 10.1007/s11332-012-0104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Cufí S, Vazquez-Martin A, Oliveras-Ferraros C, Quirantes R, Segura-Carretero A, Micol V, Joven J, Bosch-Barrera J, Del Barco S, Martin-Castillo B, Vellon L, Menendez JA. Metformin lowers the threshold for stress-induced senescence: A role for the microRNA-200 family and miR-205. Cell Cycle 2012; 11:1235-46. [DOI: 10.4161/cc.11.6.19665] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
49
|
Potter DA, Yee D, Guo Z, Rodriguez M. Should diabetic women with breast cancer have their own intervention studies? Endocr Relat Cancer 2012; 19:C13-7. [PMID: 22180498 PMCID: PMC3680369 DOI: 10.1530/erc-11-0309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
This commentary on 'Calorie restriction and rapamycin inhibit MMTV-Wnt-1 mammary tumor growth in a mouse model of postmenopausal obesity' by Nogueira et al., published in this issue of Endocrine-Related Cancer, addresses the challenges of translating diet, exercise, and pharmacologic trials in diabetic mouse mammary tumor models to human studies. We propose that trials specifically designed to test such interventions in diabetic women with breast cancer would be valuable and informative.
Collapse
Affiliation(s)
- David A Potter
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, 420 Delaware Street SouthEast, MMC 480, Minneapolis, Minnesota 55455-0392, USA.
| | | | | | | |
Collapse
|