51
|
Ortiz-Mendoza CM, Pérez-Chávez E, Fuente-Vera TADL. Modified metabolic syndrome and second cancers in women: A case control study. South Asian J Cancer 2016; 5:189-191. [PMID: 28032086 PMCID: PMC5184756 DOI: 10.4103/2278-330x.195341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background: According to some studies, the metabolic syndrome causes diverse primary cancers; however, there is no evidence about metabolic syndrome impact on second cancers development in women. Aim: To find out the implication of the modified metabolic syndrome in women with second cancers. Materials and Methods: This was a case–control study, at a general hospital in Mexico City, in women with second cancers (cases) and age-matched women with only one neoplasm (controls). The analysis comprised: Tumor (s), anthropometric features, and body mass index (BMI); moreover, presence of diabetes mellitus, hypertension, and fasting serum levels of total cholesterol, triglycerides and glucose. Results: The sample was of nine cases and 27 controls. In cases, the metabolic syndrome (diabetes mellitus or glucose > 99 mg/dL + hypertension or blood pressure ≥ 135/85 mm Hg + triglycerides > 149 mg/dL or BMI ≥ 30 kg/m2) was more frequent (odds ratio 20.8, 95% confidence interval: 1.9–227.1). Conclusion: Our results suggest that in women, the modified metabolic syndrome may be a risk factor for second cancers.
Collapse
Affiliation(s)
| | - Ernesto Pérez-Chávez
- Department of General Surgery, Hospital General Tacuba, ISSSTE, Mexico City, Mexico
| | | |
Collapse
|
52
|
Zhou JC, Guo JF, Teng RY, Wang QC, Wang J, Wei Q, Li ZD, Shen JG, Wang LB. New utility of an old marker: serum low-density lipoprotein predicts histopathological response of neoadjuvant chemotherapy in locally advanced gastric cancer. Onco Targets Ther 2016; 9:5041-7. [PMID: 27574445 PMCID: PMC4990386 DOI: 10.2147/ott.s97061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Although the correlation between metabolic abnormality and gastric cancer has been extensively investigated, the question of whether metabolic parameters might influence the efficacy of chemotherapy in locally advanced gastric cancer is still unanswered. In our present study, we investigated the relationship between serum fasting glucose, lipid levels, and histopathological response of neoadjuvant chemotherapy (NAC) in locally advanced gastric cancers. Patients and methods A total of 128 patients were identified from a prospectively maintained database of patients with locally advanced gastric cancer who received NAC between July 2004 and December 2012. Histopathological response after NAC was analyzed according to Becker’s tumor-regression grade. Univariate analyses and multivariable regression analyses were performed to determine the correlation between tumor size, differentiation, fasting glucose, lipid levels, and tumor histopathological response after NAC. Results Univariate analysis revealed that low-density lipoprotein level and total cholesterol, as well as tumor size and differentiation, correlated significantly with histopathological response. Low-density lipoprotein levels and tumor size were found to be independent predictors for histopathological response, according to multivariable regression analyses. Conclusion In this observational, hypothesis-generating study, serum low-density lipoprotein measurement was found to be useful in predicting chemosensitivity to locally advanced gastric cancer patients undergoing NAC. Incorporation of serum low-density lipoprotein levels into individualized treatment protocols could be considered in clinical practice.
Collapse
Affiliation(s)
- Ji-Chun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine
| | - Ju-Feng Guo
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine; Department of Surgical Oncology, Hangzhou First People's Hospital, Hangzhou, People's Republic of China
| | - Rong-Yue Teng
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine
| | - Qin-Chuan Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine
| | - Ji Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine
| | - Qun Wei
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine; Department of International Medicine and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zi-Duo Li
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine; Dendritic Cell Biology and Therapeutic Group, ANZAC Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Jian-Guo Shen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine
| | - Lin-Bo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine; Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
53
|
Wen YS, Huang C, Zhang X, Qin R, Lin P, Rong T, Zhang LJ. Impact of metabolic syndrome on the survival of Chinese patients with resectable esophageal squamous cell carcinoma. Dis Esophagus 2016; 29:607-13. [PMID: 26123618 DOI: 10.1111/dote.12376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metabolic syndrome (MetS) is associated with the risk of esophageal squamous cell carcinoma (ESCC). However, the impact of MetS on survival has not been evaluated. A retrospective review was performed on 596 consecutive Chinese patients with esophageal squamous cell carcinoma who received surgery between January 2005 and October 2007. The clinical data and pretreatment information related to MetS were reviewed. The impact of MetS on overall survival (OS) was estimated by Kaplan-Meier and Cox proportional hazards analyses. MetS was a significant and independent predictor for better survival in patients with resectable ESCC. The 3-year OS and 5-year OS for patients with and without MetS were 75.0% versus 57.8% and 65.1% versus 44.6%, respectively (P = 0.005 in the univariate analysis, P = 0.010 in multivariate analysis). However, there was no apparent influence of any single component of MetS on OS. The other independent prognostic factors identified in the univariate analysis included the following: gender, smoking status, alcohol use, the extent of radical surgical resection, T and N stage, and tumor differentiation. The results of the multivariate analysis included the extent of radical surgery resection, T and N stage, and tumor differentiation. MetS was also associated with greater tumor cell differentiation (P = 0.036). There was no association found between MetS status and postoperative complications. MetS is an independent prognostic factor for OS in patients with ESCC and is associated with better tumor cell differentiation.
Collapse
Affiliation(s)
- Y-S Wen
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - C Huang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - X Zhang
- School of Medicine, University of Glasgow, Glasgow, UK
| | - R Qin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - P Lin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - T Rong
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - L-J Zhang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.,State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| |
Collapse
|
54
|
Associations between metabolic syndrome, breast cancer recurrence, and the 21-gene recurrence score assay. Breast Cancer Res Treat 2016; 157:597-603. [PMID: 27271766 DOI: 10.1007/s10549-016-3846-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 01/15/2023]
Abstract
The 21-gene recurrence score (RS) assay is prognostic in estrogen receptor-positive (HR+), HER2-negative, node-negative breast cancer (BC). The interaction between RS and host factors including metabolic syndrome (MS) is unclear. MS conditions such as obesity have been associated with worse BC prognosis. The aim of this study was to identify associations between presence of MS conditions and RS group or breast cancer recurrence. Demographic, pathologic, and treatment data, MS criteria, and menopausal status were abstracted from medical records of women with stage I-II, HR+, HER2-negative BC evaluated with the RS assay at a single institution since 2005. MS was defined as presence of ≥3 of the following within 2 years of diagnosis: body mass index ≥27.7 kg/m(2); hypertension; impaired fasting glucose; HDL <50 mg/dL; hypertriglyceridemia. Of 533 eligible women, 22 % had MS. MS was more common in post- vs premenopausal women (30 vs 9 %; P < 0.0001). There was no significant association between RS group and overall MS status or any individual criterion, controlling for stage, and no association after stratification by menopausal status. Postmenopausal status was associated with higher RS group (P = 0.039), independent of stage. With 4.2-year median follow-up, no association between disease recurrence and MS was identified. Although MS has been associated with worse BC outcomes, we were unable to identify associations between RS group and MS criteria. Identification of prognostic factors other than RS that underlie this higher risk will be important for optimizing breast cancer treatment decision-making in patients with MS.
Collapse
|
55
|
Dieli-Conwright CM, Wong L, Waliany S, Bernstein L, Salehian B, Mortimer JE. An observational study to examine changes in metabolic syndrome components in patients with breast cancer receiving neoadjuvant or adjuvant chemotherapy. Cancer 2016; 122:2646-53. [PMID: 27219902 PMCID: PMC4992442 DOI: 10.1002/cncr.30104] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/23/2016] [Accepted: 04/13/2016] [Indexed: 12/02/2022]
Abstract
BACKGROUND The authors sought to determine the effect of chemotherapy on the development of metabolic syndrome (MetS) in premenopausal and postmenopausal women undergoing (neo)adjuvant therapy for early‐stage breast cancer. METHODS A total of 86 women with early‐stage (AJCC stage I‐III) breast cancer who were free from clinically diagnosed MetS (defined as 3 of 5 components of MetS) were prospectively tested for the presence of the 5 components of MetS within 1 week before initiating and after completing (neo)adjuvant chemotherapy. The 5 components of MetS measured were waist circumference; blood pressure; and fasting levels of blood glucose, triglycerides, and high‐density lipoprotein cholesterol. Anthropometrics (body weight, percentage body fat, fat mass), lipid profile (total cholesterol, low‐density lipoprotein cholesterol), glucose metabolism (insulin, homeostatic model assessment of insulin resistance, glycated hemoglobin), and inflammation (C‐reactive protein) also were examined before initiating and after completing treatment. RESULTS The current study included 46 premenopausal and 40 postmenopausal women. All individual MetS components and the overall MetS score were found to be statistically significantly increased (P<.01) after chemotherapy. Body weight, percentage body fat, fat mass, lipids, glucose metabolism, and inflammation also were found to be statistically significantly increased (P<.01). CONCLUSIONS A 12‐week to 18‐week course of chemotherapy appears to statistically significantly increase MetS and related anthropometrics, biomarkers of glucose metabolism, and inflammation in patients with early‐stage breast cancer with no preexisting MetS. Lifestyle interventions such as diet and exercise may be preventive approaches for use during chemotherapy to reduce the onset of MetS in patients with breast cancer. Cancer 2016. © 2016 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. Cancer 2016;122:2646–2653. © 2016 American Cancer Society. Patients with breast cancer who receive (neo)adjuvant chemotherapy experience an impaired metabolic presentation as noted by worsened components of metabolic syndrome, anthropometrics, and biomarkers of glucose metabolism. Lifestyle interventions such as diet and exercise may be preventive approaches for use during chemotherapy to reduce the onset of metabolic syndrome in patients with breast cancer.
Collapse
Affiliation(s)
| | - Louise Wong
- Division of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Sarah Waliany
- Division of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Leslie Bernstein
- Beckman Research Institute, Division of Cancer Etiology, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Behrouz Salehian
- Department of Clinical Diabetes, Endocrinology and Metabolism, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Joanne E Mortimer
- Division of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
56
|
Nam S, Park S, Park HS, Kim S, Kim JY, Kim SI. Association Between Insulin Resistance and Luminal B Subtype Breast Cancer in Postmenopausal Women. Medicine (Baltimore) 2016; 95:e2825. [PMID: 26945364 PMCID: PMC4782848 DOI: 10.1097/md.0000000000002825] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Currently, there is limited information on the clinical characteristics of breast cancer patients with insulin resistance. Hence, the purpose of this study was to investigate the association between insulin resistance and clinicopathological factors in newly diagnosed breast cancer patients without diabetes. We assessed 760 patients with breast cancer treated between 2012 and 2014. We compared the clinicopathological characteristics between patients with and without insulin resistance using univariate and multivariate analyses, including after stratification by menopausal status. Insulin resistance was defined according to the homeostatic model assessment of insulin resistance. Of 760 patients, 26.4% had insulin resistance. Age, menopausal status, body mass index, tumor size, histologic grade, Ki-67 expression, and breast cancer subtype significantly differed according to the presence of insulin resistance. Multivariate analysis revealed that postmenopausal status and obesity were significantly associated with insulin resistance. In postmenopausal women, older age, obesity, larger tumor size, advanced stage, and high proliferative luminal B subtype were significantly associated with insulin resistance. In contrast, in premenopausal patients, only obesity was related to insulin resistance. Multivariate analysis indicated that insulin resistance was independently correlated with obesity, larger tumor size, and the luminal B/human epidermal growth factor receptor-2-negative subtype in postmenopausal but not premenopausal patients. Insulin resistance was significantly associated with larger tumors and proliferative luminal B subtype breast cancer in postmenopausal women only. These findings suggest that insulin resistance could mechanistically induce tumor progression and might be a good prognostic factor, and that it could represent a therapeutic target in postmenopausal patients with breast cancer.
Collapse
Affiliation(s)
- Sanggeun Nam
- From the Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
57
|
Zeng L, Zielinska HA, Arshad A, Shield JP, Bahl A, Holly JMP, Perks CM. Hyperglycaemia-induced chemoresistance in breast cancer cells: role of the estrogen receptor. Endocr Relat Cancer 2016; 23:125-34. [PMID: 26647383 DOI: 10.1530/erc-15-0507] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2015] [Indexed: 11/08/2022]
Abstract
Breast cancer patients with diabetes respond less well to chemotherapy; in keeping with this we determined previously that hyperglycaemia-induced chemoresistance in estrogen receptor (ERα) positive breast cancer cells and showed that this was mediated by fatty acid synthase (FASN). More recent evidence suggests that the effect of metabolic syndrome and diabetes is not the same for all subtypes of breast cancer with inferior disease-free survival and worse overall survival only found in women with ERα positive breast cancer and not for other subtypes. Here we examined the impact of hyperglycaemia on ERα negative breast cancer cells and further investigated the mechanism underlying chemoresistance in ERα with a view to identifying strategies to alleviate hyperglycaemia-induced chemoresistance. We found that hyperglycaemia-induced chemoresistance was only observed in ERα breast cancer cells and was dependent upon the expression of ERα as chemoresistance was negated when the ERα was silenced. Hyperglycaemia-induced an increase in activation and nuclear localisation of the ERα that was downstream of FASN and dependent on the activation of MAPK. We found that fulvestrant successfully negated the hyperglycaemia-induced chemoresistance, whereas tamoxifen had no effect. In summary our data suggests that the ERα may be a predictive marker of poor response to chemotherapy in breast cancer patients with diabetes. It further indicates that anti-estrogens could be an effective adjuvant to chemotherapy in such patients and indicates the importance for the personalised management of breast cancer patients with diabetes highlighting the need for clinical trials of tailored chemotherapy for diabetic patients diagnosed with ERα positive breast cancers.
Collapse
Affiliation(s)
- L Zeng
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - H A Zielinska
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - A Arshad
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - J P Shield
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - A Bahl
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - J M P Holly
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - C M Perks
- IGFs and Metabolic Endocrinology GroupSchool of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol BS10 1TD, UKDepartment of Clinical OncologyBristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| |
Collapse
|
58
|
Kim J, Lee YH, Park JHY, Sung MK. Estrogen deprivation and excess energy supply accelerate 7,12-dimethylbenz(a)anthracene-induced mammary tumor growth in C3H/HeN mice. Nutr Res Pract 2015; 9:628-36. [PMID: 26634052 PMCID: PMC4667204 DOI: 10.4162/nrp.2015.9.6.628] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/18/2015] [Accepted: 06/17/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/OBJECTIVES Obesity is a risk factor of breast cancer in postmenopausal women. Estrogen deprivation has been suggested to cause alteration of lipid metabolism thereby creating a cellular microenvironment favoring tumor growth. The aim of this study is to investigate the effects of estrogen depletion in combination with excess energy supply on breast tumor development. MATERIALS/METHODS Ovariectomized (OVX) or sham-operated C3H/HeN mice at 4 wks were provided with either a normal diet or a high-fat diet (HD) for 16 weeks. Breast tumors were induced by administration of 7,12-dimethylbenz(a)anthracene once a week for six consecutive weeks. RESULTS Study results showed higher serum concentrations of free fatty acids and insulin in the OVX+HD group compared to other groups. The average tumor volume was significantly larger in OVX+HD animals than in other groups. Expressions of mammary tumor insulin receptor and mammalian target of rapamycin proteins as well as the ratio of pAKT/AKT were significantly increased, while pAMPK/AMPK was decreased in OVX+HD animals compared to the sham-operated groups. Higher relative expression of liver fatty acid synthase mRNA was observed in OVX+HD mice compared with other groups. CONCLUSIONS These results suggest that excess energy supply affects the accelerated mammary tumor growth in estrogen deprived mice.
Collapse
Affiliation(s)
- Jin Kim
- Department of Food and Nutrition, Sookmyung Women's University, 100 Chungpa-ro 47-gil, Yongsan-gu, Seoul, 140-742, Korea
| | - Yoon Hee Lee
- Department of Food and Nutrition, Sookmyung Women's University, 100 Chungpa-ro 47-gil, Yongsan-gu, Seoul, 140-742, Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, College of Natural Sciences, Hallym University, 39 Hallymdaehak-gil, Chuncheon, 200-702, Korea
| | - Mi-Kyung Sung
- Department of Food and Nutrition, Sookmyung Women's University, 100 Chungpa-ro 47-gil, Yongsan-gu, Seoul, 140-742, Korea
| |
Collapse
|
59
|
Kim BK, Chang Y, Ahn J, Jung HS, Kim CW, Yun KE, Kwon MJ, Suh BS, Chung EC, Shin H, Ryu S. Metabolic syndrome, insulin resistance, and mammographic density in pre- and postmenopausal women. Breast Cancer Res Treat 2015; 153:425-34. [PMID: 26277917 DOI: 10.1007/s10549-015-3544-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 01/24/2023]
Abstract
Little is known about the association of metabolic syndrome (MetS) or insulin resistance (IR) with mammographic density, a strong risk factor for breast cancer. The goal of this study was to evaluate these associations in pre- and postmenopausal women. A cross-sectional study was performed in 73,974 adult women who underwent a comprehensive health screening examination that included a mammogram between 2011 and 2013 (mean age 42.6 years). MetS was defined according to the modified National Cholesterol Education Program Adult Treatment Panel III. IR was assessed with the homeostasis model assessment-insulin resistance (HOMA-IR). Adjusted odds ratios (ORs) with 95% confidence intervals (CIs) for dense breast were estimated using logistic regression models after adjustment for potential confounders. In premenopausal women, MetS and all its components except waist circumference were associated with dense breast. After adjustment for potential confounders, the OR (95% CI) for dense breast in women with MetS compared with those without MetS was 1.22 (1.06-1.39). In postmenopausal women, however, there was positive but non-significant association between MetS and dense breast. In both pre- and postmenopausal women, high blood glucose and IR were positively associated with dense breast. The OR (95% CI) for dense breast between the highest and lowest quartiles of HOMA-IR was 1.29 (1.20-1.39) for premenopausal women and 1.44 (1.05-1.97) for postmenopausal women. In a large sample of Korean women, MetS and IR were associated with mammographic dense breast, demonstrating that IR, a potentially modifiable risk factor, may increase breast cancer risk, possibly through high mammographic density.
Collapse
Affiliation(s)
- Bo-Kyoung Kim
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Samsung Main Building B2, 250, Taepyung-ro 2ga, Jung-gu, Seoul, 100-742, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Finocchiaro C, Ossola M, Monge T, Fadda M, Brossa L, Caudera V, De Francesco A. Effect of specific educational program on dietary change and weight loss in breast-cancer survivors. Clin Nutr 2015. [PMID: 26199085 DOI: 10.1016/j.clnu.2015.05.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
AIMS Consumption of Western foods with high dietary glycemic load is associated with breast cancer development, whereas adherence to Mediterranean diet has been linked to a reduced risk. Changing lifestyle can decrease risk of recurrences and mortality. Thus decreasing the weight, improving the diet and promoting lifestyle are among of the most important issues of public health. We evaluated the effects of a specific educational intervention conducted by dietitians, nutritionists physicians, oncologist and sport physician to promote lifestyle in breast cancer survivors. METHODS We recruited 100 pts in breast cancer follow up. The intervention program consisted of four meetings once a week including lectures, training sessions and workshops lasting overall a month. Dietary recommendations were provided on the basis of WCRF/AICR guidelines and were modelled on Mediterranean diet. Sport physician recommended adapted physical activity, based on clinical experiences and scientific evidences. RESULTS Significantly decrease of BMI and waist circumference was observed after 2 and 6 months. Adherence to Mediterranean diet was significantly improved, both in heightening typical Mediterranean foods, both in decreasing consumption of non typical foods. At baseline 63% of women was inactive, 37% was mild active and 0% active, while at the end inactive patients felt by half (30%) and mild active women almost doubled (67%). CONCLUSIONS We found this dietary intervention effective in reducing BMI and waist circumference, and enhancing healthy lifestyle in BC survivors. It has surely contributed to achieve these results besides the change in diet quality, mostly a marked reduction in sedentary habits.
Collapse
Affiliation(s)
- Concetta Finocchiaro
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy.
| | - Marta Ossola
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| | - Taira Monge
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| | - Maurizio Fadda
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| | - Laura Brossa
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| | - Vilma Caudera
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| | - Antonella De Francesco
- Department of Clinical Nutrition, Hospital Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
61
|
Zou L, Liu TR, Yang AK. Metabolic syndrome is associated with better prognosis in patients with tongue squamous cell carcinoma. CHINESE JOURNAL OF CANCER 2015; 34:184-8. [PMID: 25963193 PMCID: PMC4593369 DOI: 10.1186/s40880-015-0009-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/02/2014] [Indexed: 01/30/2023]
Abstract
Introduction Metabolic syndrome (MS) is associated with several cancers, but it is not clear whether MS affects the prognosis of tongue squamous cell carcinoma (TSCC). This study aimed to evaluate the prognostic value of MS in TSCC. Methods Clinical data from 252 patients with TSCC who were initially treated at the Sun Yat-sen University Cancer Center between April 1998 and June 2011 were collected, and the associations between MS and clinicopathologic factors were retrospectively analyzed. Prognostic outcomes were examined by Kaplan-Meier analysis and Cox regression analysis. Results Of the 252 patients, 48 were diagnosed with MS. MS was associated with early N category in TSCC (P < 0.001). The patients with MS showed longer survival than those without MS (P = 0.028). MS was an independent prognostic factor for patients with TSCC. Conclusions MS is associated with early N category in TSCC. It is an independent prognostic factor for better survival in patients with TSCC.
Collapse
Affiliation(s)
- Lan Zou
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.
| | - Tian-Run Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510655, P. R. China.
| | - An-Kui Yang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.
| |
Collapse
|
62
|
Abdominal adiposity and family income-to-poverty ratio in American women. Obes Res Clin Pract 2015; 8:e201-98. [PMID: 24847661 DOI: 10.1016/j.orcp.2012.12.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 10/02/2012] [Accepted: 12/08/2012] [Indexed: 11/23/2022]
Abstract
OBJECTIVE We examined (a) secular changes in abdominal fat accumulation (AFA) and family income-to-poverty ratio (PIR) across race/ethnicity, education and age in Mexican (MA), non-Hispanic Whites (NHW), non-Hispanic Black (NHB), and (b) association between PIR and AFA among American women. METHODS Data (n = 9787) from 2001-2002 to 2009-2010 NHANES were used. Rates of AFA and poverty by race/ethnic, age and education categories were determined across study time points. Subjects with low and medium PIR values were classified as poor. Linear trends in AFA and PIR were evaluated. Study time-specific odds ratios (OR) from logistic regression models were used to estimate risk of AFA due to low to medium PIR. Statistical adjustments were made for race/ethnicity, education, age, and marital status. RESULTS Increased trends in low to medium PIR and AFA in MA, NHW, and NHB American women were observed between 2001 and 2010. Poor women had much higher prevalence of AFA compared to richer women. For each of the studied periods, medium and low PIR were each associated with increased odds of AFA. The association between poverty and AFA was weakest in 2001-2002 (OR = 1.49, 95% CI: 1.05-2.11) compared to 2009-2010 (OR = 1.64, 95% CI: 1.21-2.22). Compared to NHW, being of MA and NHB race/ethnicity was also each associated with increased odds of AFA, controlling for other independent variables. CONCLUSIONS Increase in poverty and AFA, and positive association between decreased PIR and increased odds of AFA were observed in the period between 2001 and 2010 in MA, NHW, and NHB American women. A robust economic policy designed to alleviate poverty may be an important means of reducing the trajectory of AFA in American women.
Collapse
|
63
|
Ghose A, Kundu R, Toumeh A, Hornbeck C, Mohamed I. A review of obesity, insulin resistance, and the role of exercise in breast cancer patients. Nutr Cancer 2015; 67:197-202. [PMID: 25625592 DOI: 10.1080/01635581.2015.990569] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Breast cancer, the most common female malignancy in the world, has a strong association with obesity and insulin resistance. The importance of these risk factors goes up significantly in patients already affected by this cancer as they negatively affect the prognosis, recurrence rate, and survival by various mechanisms. The literature on the role of physical activity and aerobic exercise on modifying the above risks is debatable with data both for and against it. In this article, we have reviewed the risks of obesity and insulin resistance in breast cancer patients and the controversy associated with the impact of exercise. Ultimately, we have concluded that a randomized control trial is necessary with an individualized aerobic exercise program for a minimum duration of 20 wk on breast cancer patients, who are undergoing or recently completed chemotherapy, to study its effects on insulin resistance, weight, and clinical outcome.
Collapse
Affiliation(s)
- Abhimanyu Ghose
- a Department of Hematology/Oncology , University of Cincinnati , Cincinnati , Ohio , USA
| | | | | | | | | |
Collapse
|
64
|
Zielinska HA, Bahl A, Holly JM, Perks CM. Epithelial-to-mesenchymal transition in breast cancer: a role for insulin-like growth factor I and insulin-like growth factor-binding protein 3? BREAST CANCER-TARGETS AND THERAPY 2015; 7:9-19. [PMID: 25632238 PMCID: PMC4304531 DOI: 10.2147/bctt.s43932] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Evidence indicates that for most human cancers the problem is not that gene mutations occur but is more dependent upon how the body deals with damaged cells. It has been estimated that only about 1% of human cancers can be accounted for by unmistakable hereditary cancer syndromes, only up to 5% can be accounted for due to high-penetrance, single-gene mutations, and in total only 5%-15% of all cancers may have a major genetic component. The predominant contribution to the causation of most sporadic cancers is considered to be environmental factors contributing between 58% and 82% toward different cancers. A nutritionally poor lifestyle is associated with increased risk of many cancers, including those of the breast. As nutrition, energy balance, macronutrient composition of the diet, and physical activity levels are major determinants of insulin-like growth factor (IGF-I) bioactivity, it has been proposed that, at least in part, these increases in cancer risk and progression may be mediated by alterations in the IGF axis, related to nutritional lifestyle. Localized breast cancer is a manageable disease, and death from breast cancer predominantly occurs due to the development of metastatic disease as treatment becomes more complicated with poorer outcomes. In recent years, epithelial-to-mesenchymal transition has emerged as an important contributor to breast cancer progression and malignant transformation resulting in tumor cells with increased potential for migration and invasion. Furthermore, accumulating evidence suggests a strong link between components of the IGF pathway, epithelial-to-mesenchymal transition, and breast cancer mortality. Here, we highlight some recent studies highlighting the relationship between IGFs, IGF-binding protein 3, and epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Hanna A Zielinska
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Amit Bahl
- Department of Clinical Oncology, Bristol Haematology and Oncology Centre, University Hospitals Bristol, Bristol, UK
| | - Jeff Mp Holly
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol, UK
| | - Claire M Perks
- IGFs and Metabolic Endocrinology Group, School of Clinical Sciences, University of Bristol, Learning and Research Building, Southmead Hospital, Bristol, UK
| |
Collapse
|
65
|
Metabolic syndrome is associated with increased breast cancer risk: a systematic review with meta-analysis. Int J Breast Cancer 2014; 2014:189384. [PMID: 25653879 PMCID: PMC4295135 DOI: 10.1155/2014/189384] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 12/29/2022] Open
Abstract
Background. Although individual metabolic risk factors are reported to be associated with breast cancer risk, controversy surrounds risk of breast cancer from metabolic syndrome (MS). We report the first systematic review and meta-analysis of the association between MS and breast cancer risk in all adult females. Methods. Studies were retrieved by searching four electronic reference databases [PubMed, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, and ProQuest through June 30, 2012] and cross-referencing retrieved articles. Eligible for inclusion were longitudinal studies reporting associations between MS and breast cancer risk among females aged 18 years and older. Relative risks and 95% confidence intervals were calculated for each study and pooled using random-effects models. Publication bias was assessed quantitatively (Trim and Fill) and qualitatively (funnel plots). Heterogeneity was examined using Q and I2 statistics. Results. Representing nine independent cohorts and 97,277 adult females, eight studies met the inclusion criteria. A modest, positive association was observed between MS and breast cancer risk (RR: 1.47, 95% CI, 1.15–1.87; z = 3.13; p = 0.002; Q = 26.28, p = 0.001; I2 = 69.55%). No publication bias was observed. Conclusions. MS is associated with increased breast cancer risk in adult women.
Collapse
|
66
|
Effect of metabolic syndrome and its components on recurrence and survival in early-stage non-small cell lung cancer. Med Oncol 2014; 32:423. [DOI: 10.1007/s12032-014-0423-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 01/10/2023]
|
67
|
Calip GS, Malone KE, Gralow JR, Stergachis A, Hubbard RA, Boudreau DM. Metabolic syndrome and outcomes following early-stage breast cancer. Breast Cancer Res Treat 2014; 148:363-77. [PMID: 25301086 PMCID: PMC4236717 DOI: 10.1007/s10549-014-3157-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 09/30/2014] [Indexed: 12/18/2022]
Abstract
The prevalence of risk factors contributing to metabolic syndrome (MetS) is increasing, and numerous components of MetS are associated with increased primary breast cancer (BC) risk. However, less is known about the relationship of MetS to BC outcomes. The aim of this study was to evaluate whether MetS, characterized by increased weight, hypertension, low HDL-cholesterol, high triglycerides, and diabetes or impaired glucose tolerance, is associated with risk of second breast cancer events (SBCE) and BC-specific mortality. Retrospective cohort study of women diagnosed with incident early-stage (I-II) BC between 1990 and 2008, enrolled in an integrated health plan. Outcomes of interest were SBCE, defined as recurrence or second primary BC, and BC-specific mortality. We used multivariable Cox proportional hazards models to estimate adjusted hazard ratios (HR) and 95% confidence intervals (CI) for time-varying exposure to MetS components while accounting for potential confounders and competing risks. Among 4,216 women in the cohort, 26% had ≥3 MetS components and 13% developed SBCE during median follow-up of 6.3 years. Compared to women with no MetS components, presence of MetS (≥3 components) was associated with increased risk of SBCE (HR = 1.50, 95% CI 1.08-2.07) and BC-specific mortality (HR = 1.65, 95% CI 1.02-2.69). Of the individual components, only increased weight was associated with increased risk of SBCE (HR = 1.26, 95% CI 1.06-1.49). MetS is associated with modestly increased risk of SBCE and BC-specific mortality. Given the growing population of BC survivors, further research in larger and more diverse populations is warranted.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Body Mass Index
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/physiopathology
- Female
- Follow-Up Studies
- Humans
- Immunoenzyme Techniques
- Metabolic Syndrome/complications
- Metabolic Syndrome/metabolism
- Metabolic Syndrome/mortality
- Middle Aged
- Neoplasm Recurrence, Local/etiology
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm Staging
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/metabolism
- Neoplasms, Second Primary/mortality
- Neoplasms, Second Primary/pathology
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Survival Rate
- Young Adult
Collapse
Affiliation(s)
- Gregory S Calip
- Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago, 833 S. Wood St. M/C 871, Room 287, Chicago, IL, 60612, USA,
| | | | | | | | | | | |
Collapse
|
68
|
Gordon-Dseagu VLZ, Shelton N, Mindell J. Diabetes mellitus and mortality from all-causes, cancer, cardiovascular and respiratory disease: evidence from the Health Survey for England and Scottish Health Survey cohorts. J Diabetes Complications 2014; 28:791-7. [PMID: 25104237 DOI: 10.1016/j.jdiacomp.2014.06.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/03/2014] [Accepted: 06/25/2014] [Indexed: 01/23/2023]
Abstract
BACKGROUND Diabetes mellitus is associated with differing rates of all-cause and cause-specific mortality compared with the general population; although the strength of these associations requires further investigation. The effects of confounding factors, such as overweight and obesity and the presence of co-morbid cardiovascular disease (CVD), upon such associations also remain unclear. There is thus a need for studies which utilise data from nationally-representative samples to explore these associations further. METHODS A cohort study of 204,533 participants aged 16+ years (7,199 with diabetes) from the Health Survey for England (HSE) (1994-2008) and Scottish Health Survey (SHeS) (1995, 1998 and 2003) linked with UK mortality records. Odds ratios (ORs) of all-cause and cause-specific mortality and 95% confidence intervals were estimated using logistic and multinomial logistic regression. RESULTS There were 20,051 deaths (1,814 among those with diabetes). Adjusted (age, sex, and smoking status) ORs for all-cause mortality among those with diabetes was 1.68 (95%CI 1.57-1.79). Cause-specific mortality ORs were: cancer 1.26 (1.13-1.42), respiratory diseases 1.25 (1.08-1.46), CVD 1.96 (1.80-2.14) and 'other' causes 2.06 (1.84-2.30). These were not attenuated significantly after adjustment for generalised and/or central adiposity and other confounding factors. The odds of mortality differed between those with and without comorbid CVD at baseline; the ORs for the latter group were substantially increased. CONCLUSIONS In addition to the excess in CVD and all-cause mortality among those with diabetes, there is also increased mortality from cancer, respiratory diseases, and 'other' causes. This increase in mortality is independent of obesity and a range of other confounding factors. With falling CVD incidence and mortality, the raised risks of respiratory and cancer deaths in people with diabetes will become more important and require increased health care provision.
Collapse
Affiliation(s)
- Vanessa L Z Gordon-Dseagu
- UCL (University College London), Research Department of Epidemiology and Public Health, 1-19 Torrington Place, London, WC1E 6BT.
| | - Nicola Shelton
- UCL (University College London), Research Department of Epidemiology and Public Health, 1-19 Torrington Place, London, WC1E 6BT.
| | - Jennifer Mindell
- UCL (University College London), Research Department of Epidemiology and Public Health, 1-19 Torrington Place, London, WC1E 6BT.
| |
Collapse
|
69
|
Tsai MS, Chen HP, Hung CM, Lee PH, Lin CL, Kao CH. Hospitalization for Inflammatory Bowel Disease is Associated with Increased Risk of Breast Cancer: A Nationwide Cohort Study of an Asian Population. Ann Surg Oncol 2014; 22:1996-2002. [PMID: 25354573 DOI: 10.1245/s10434-014-4198-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Indexed: 12/28/2022]
Abstract
PURPOSE To learn whether women with inflammatory bowel disease (IBD) exhibit a higher risk of breast cancer. METHODS We identified 4,856 women with IBD symptoms from 1998 to 2008 and 19,424 control patients without the disorder, frequency matched by age, sex, and admission year. Both cohorts were followed-up until the end of 2010 to estimate the risk of breast cancer. RESULTS Overall, the incidence of breast cancer was similar in the IBD and control cohorts (1.31 vs. 1.25 per 1,000 person-years). The adjusted hazard ratio of breast cancer was 0.95 (95 % confidence interval 0.66-1.36) for the IBD patients. Further analysis revealed that neither Crohn disease nor ulcerative colitis was associated with the risk of developing breast cancer in women. The age-specific analysis indicated that the incidence of breast cancer was highest in the 45- to 65-year-old age group in both cohorts. The incidence of breast cancer was significantly increased in patients who required hospitalization twice or more per year, compared with the control cohort (adjusted hazard ratio 8.45; 95 % confidence interval 4.64-15.4). Moreover, age-specific analysis showed that patients aged less than 65 years old (≤44 or 45-65 years of age) exhibited a strong association between IBD hospitalization and breast cancer risk. CONCLUSIONS The risk of breast cancer was positively proportional to the frequency of admission for IBD. Therefore, careful surveillance of breast cancer should be sought for female IBD patients with 2 or more annual hospitalizations.
Collapse
Affiliation(s)
- Ming-Shian Tsai
- Department of General Surgery, E-Da Hospital and I-Shou University, Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
70
|
Omega-3 fatty acids reduce obesity-induced tumor progression independent of GPR120 in a mouse model of postmenopausal breast cancer. Oncogene 2014; 34:3504-13. [PMID: 25220417 PMCID: PMC4362785 DOI: 10.1038/onc.2014.283] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/27/2014] [Accepted: 07/27/2014] [Indexed: 01/07/2023]
Abstract
Obesity and inflammation are both risk factors for a variety of cancers, including breast cancer in postmenopausal women. Intake of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) decreases the risk of breast cancer, and also reduces obesity-associated inflammation and insulin resistance, but whether the two effects are related is currently unknown. We tested this hypothesis in a postmenopausal breast cancer model using ovariectomized, immune-competent female mice orthotopically injected with Py230 mammary tumor cells. Obesity, whether triggered genetically or by high-fat diet (HFD) feeding, increased inflammation in the mammary fat pad and promoted mammary tumorigenesis. The presence of tumor cells in the mammary fat pad further enhanced the local inflammatory milieu. Tumor necrosis factor-alpha (TNF-α) was the most highly upregulated cytokine in the obese mammary fat pad, and we observed that TNF-α dose-dependently stimulated Py230 cell growth in vitro. An ω-3 PUFA-enriched HFD (referred to as fish oil diet, FOD) reduced inflammation in the obese mammary fat pad in the absence of tumor cells and inhibited Py230 tumor growth in vivo. Although some anti-inflammatory effects of ω-3 PUFAs were previously shown to be mediated by the G-protein-coupled receptor 120 (GPR120), the FOD reduced Py230 tumor burden in GPR120-deficient mice to a similar degree as observed in wild-type mice, indicating that the effect of FOD to reduce tumor growth does not require GPR120 in the host mouse. Instead, in vitro studies demonstrated that ω-3 PUFAs act directly on tumor cells to activate c-Jun N-terminal kinase, inhibit proliferation and induce apoptosis. Our results show that obesity promotes mammary tumor progression in this model of postmenopausal breast cancer and that ω-3 PUFAs, independent of GPR120, inhibit mammary tumor progression in obese mice.
Collapse
|
71
|
Berrino F, Villarini A, Traina A, Bonanni B, Panico S, Mano MP, Mercandino A, Galasso R, Barbero M, Simeoni M, Bassi MC, Consolaro E, Johansson H, Zarcone M, Bruno E, Gargano G, Venturelli E, Pasanisi P. Metabolic syndrome and breast cancer prognosis. Breast Cancer Res Treat 2014; 147:159-65. [PMID: 25104441 DOI: 10.1007/s10549-014-3076-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/21/2014] [Indexed: 01/06/2023]
Abstract
Metabolic syndrome (MS), conventionally defined by the presence of at least three out of five dysmetabolic traits (abdominal obesity, hypertension, low plasma HDL-cholesterol, high plasma glucose and high triglycerides), has been associated with an increased risk of several age-related chronic diseases, including breast cancer (BC). This may have prognostic implications for BC survivors. 2,092 early stage BC survivors aged 35-70, recruited in eleven Italian centres 0-5 years after surgical treatment (1.74 years on average), were followed-up over 2.8 years on average for additional BC-related events, including BC-specific mortality, distant metastasis, local recurrences and contralateral BC. At recruitment, 20 % of the patients had MS. Logistic regression models were carried out to generate OR and 95 % confidence intervals (CI) for new BC events associated with MS, adjusting for baseline pathological prognostic factors. New BC events occurred in 164 patients, including 89 distant metastases. The adjusted ORs for women with MS versus women without any MS traits were 2.17 (CI 1.31-3.60) overall, and 2.45 (CI 1.24-4.82) for distant metastasis. The OR of new BC events for women with only one or two MS traits was 1.40 (CI 0.91-2.16). All MS traits were positively associated with new BC events, and significantly so for low HDL and high triglycerides. MS is an important prognostic factor in BC. As MS is reversible through lifestyle changes, interventions to decrease MS traits in BC patients should be implemented in BC clinics.
Collapse
Affiliation(s)
- Franco Berrino
- Epidemiology & Prevention Unit, Department of Preventive & Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Hiatt RA, Porco TC, Liu F, Balke K, Balmain A, Barlow J, Braithwaite D, Diez-Roux AV, Kushi LH, Moasser MM, Werb Z, Windham GC, Rehkopf DH. A multilevel model of postmenopausal breast cancer incidence. Cancer Epidemiol Biomarkers Prev 2014; 23:2078-92. [PMID: 25017248 DOI: 10.1158/1055-9965.epi-14-0403] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Breast cancer has a complex etiology that includes genetic, biologic, behavioral, environmental, and social factors. Etiologic factors are frequently studied in isolation with adjustment for confounding, mediating, and moderating effects of other factors. A complex systems model approach may present a more comprehensive picture of the multifactorial etiology of breast cancer. METHODS We took a transdisciplinary approach with experts from relevant fields to develop a conceptual model of the etiology of postmenopausal breast cancer. The model incorporated evidence of both the strength of association and the quality of the evidence. We operationalized this conceptual model through a mathematical simulation model with a subset of variables, namely, age, race/ethnicity, age at menarche, age at first birth, age at menopause, obesity, alcohol consumption, income, tobacco use, use of hormone therapy (HT), and BRCA1/2 genotype. RESULTS In simulating incidence for California in 2000, the separate impact of individual variables was modest, but reduction in HT, increase in the age at menarche, and to a lesser extent reduction in excess BMI >30 kg/m(2) were more substantial. CONCLUSIONS Complex systems models can yield new insights on the etiologic factors involved in postmenopausal breast cancer. Modification of factors at a population level may only modestly affect risk estimates, while still having an important impact on the absolute number of women affected. IMPACT This novel effort highlighted the complexity of breast cancer etiology, revealed areas of challenge in the methodology of developing complex systems models, and suggested additional areas for further study.
Collapse
Affiliation(s)
- Robert A Hiatt
- Department of Epidemiology and Biostatistics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
| | - Travis C Porco
- Department of Epidemiology and Biostatistics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California. Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Fengchen Liu
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California
| | - Kaya Balke
- Department of Epidemiology and Biostatistics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Allan Balmain
- Department of Biochemistry and Biophysics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | - Dejana Braithwaite
- Department of Epidemiology and Biostatistics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Ana V Diez-Roux
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | | | - Mark M Moasser
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Zena Werb
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Gayle C Windham
- Division of Environmental and Occupational Disease Control, California Department of Public Health, Richmond, California
| | - David H Rehkopf
- Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
73
|
The muscle mass, omega-3, diet, exercise and lifestyle (MODEL) study - a randomised controlled trial for women who have completed breast cancer treatment. BMC Cancer 2014; 14:264. [PMID: 24739260 PMCID: PMC4006632 DOI: 10.1186/1471-2407-14-264] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/26/2014] [Indexed: 02/01/2023] Open
Abstract
Background Loss of lean body mass (LBM) is a common occurrence after treatment for breast cancer and is related to deleterious metabolic health outcomes [Clin Oncol, 22(4):281–288, 2010; Appl Physiol Nutr Metab, 34(5):950–956, 2009]. The aim of this research is to determine the effectiveness of long chain omega-3 fatty acids (LCn-3s) and exercise training alone, or in combination, in addressing LBM loss in breast cancer survivors. Methods/design A total of 153 women who have completed treatment for breast cancer in the last 12 months, with a Body Mass Index (BMI) of 20 to 35 kg/m2, will be randomly assigned to one of 3 groups: 3g/d LCn-3s (N-3), a 12-week nutrition and exercise education program plus olive oil (P-LC) or the education program plus LCn-3s (EX+N-3). Participants randomised to the education groups will be blinded to treatment, and will receive either olive oil placebo (OO+N-3) or LCn-3 provision, while the N-3 group will be open label. The education program includes nine 60-75min sessions over 12 weeks that will involve breast cancer specific healthy eating advice, plus a supervised exercise session run as a resistance exercise circuit. They will also be advised to conduct the resistance training and aerobic training 5 to 7 days per week collectively. Outcome measures will be taken at baseline, 12-weeks and 24-weeks. The primary outcome is % change in LBM as measured by the air displacement plethysmograhy. Secondary outcomes include quality of life (FACT-B + 4) and inflammation (C-Reactive protein: CRP). Additional measures taken will be erythrocyte fatty acid analysis, fatigue, physical activity, menopausal symptoms, dietary intake, joint pain and function indices. Discussion This research will provide the first insight into the efficacy of LCn-3s alone or in combination with exercise in breast cancer survivors with regards to LBM and quality of life. In addition, this study is designed to improve evidence-based dietetic practice, and how specific dietary prescription may link with appropriate exercise interventions. Trials registration ACTRN12610001005044; and World Health Organisation Universal trial number:
U1111-1116-8520.
Collapse
|
74
|
Dieli-Conwright CM, Mortimer JE, Schroeder ET, Courneya K, Demark-Wahnefried W, Buchanan TA, Tripathy D, Bernstein L. Randomized controlled trial to evaluate the effects of combined progressive exercise on metabolic syndrome in breast cancer survivors: rationale, design, and methods. BMC Cancer 2014; 14:238. [PMID: 24708832 PMCID: PMC3985576 DOI: 10.1186/1471-2407-14-238] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 03/21/2014] [Indexed: 12/18/2022] Open
Abstract
Background Metabolic syndrome (MetS) is increasingly present in breast cancer survivors, possibly worsened by cancer-related treatments, such as chemotherapy. MetS greatly increases risk of cardiovascular disease and diabetes, co-morbidities that could impair the survivorship experience, and possibly lead to cancer recurrence. Exercise has been shown to positively influence quality of life (QOL), physical function, muscular strength and endurance, reduce fatigue, and improve emotional well-being; however, the impact on MetS components (visceral adiposity, hyperglycemia, low serum high-density lipoprotein cholesterol, hypertriglyceridemia, and hypertension) remains largely unknown. In this trial, we aim to assess the effects of combined (aerobic and resistance) exercise on components of MetS, as well as on physical fitness and QOL, in breast cancer survivors soon after completing cancer-related treatments. Methods/Design This study is a prospective randomized controlled trial (RCT) investigating the effects of a 16-week supervised progressive aerobic and resistance exercise training intervention on MetS in 100 breast cancer survivors. Main inclusion criteria are histologically-confirmed breast cancer stage I-III, completion of chemotherapy and/or radiation within 6 months prior to initiation of the study, sedentary, and free from musculoskeletal disorders. The primary endpoint is MetS; secondary endpoints include: muscle strength, shoulder function, cardiorespiratory fitness, body composition, bone mineral density, and QOL. Participants randomized to the Exercise group participate in 3 supervised weekly exercise sessions for 16 weeks. Participants randomized to the Control group are offered the same intervention after the 16-week period of observation. Discussion This is the one of few RCTs examining the effects of exercise on MetS in breast cancer survivors. Results will contribute a better understanding of metabolic disease-related effects of resistance and aerobic exercise training and inform intervention programs that will optimally improve physiological and psychosocial health during cancer survivorship, and that are ultimately aimed at improving prognosis. Trial registration NCT01140282; Registration: June 10, 2010
Collapse
Affiliation(s)
- Christina M Dieli-Conwright
- Division of Biokinesiology and Physical Therapy, University of Southern California, 1540 E, Alcazar St, CHP 155, 90089 Los Angeles, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
OBJECTIVE The aim of this study was to assess the risk of metabolic syndrome (MetS) in postmenopausal breast cancer survivors as compared with postmenopausal women without breast cancer. METHODS In this cross-sectional study, 104 postmenopausal breast cancer survivors were compared with 208 postmenopausal women (controls) attending a university hospital. Eligibility criteria included the following: amenorrhea longer than 12 months and aged 45 years or older, treated for breast cancer, and metastasis-free for at least 5 years. The control group consisted of women with amenorrhea longer than 12 months and aged 45 years or older and without breast cancer, matched by age and menopause status (in a proportion of 1:2 as sample calculation). Clinical and anthropometric data were collected. Biochemical parameters, including total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, glucose, and C-reactive protein, were measured. Women showing three or more diagnostic criteria were diagnosed as having MetS: waist circumference of 88 cm or larger, blood pressure of 130/85 mm Hg or higher, triglycerides level of 150 mg/dL or higher, high-density lipoprotein cholesterol level lower than 50 mg/dL, and glucose level of 100 mg/dL or higher. For statistical analysis, Student's t test, χ2 test, and logistic regression (odds ratio [OR]) were used. RESULTS The mean (SD) age of breast cancer survivors was 60.6 (8.6) years, with a mean (SD) follow-up of 9.4 (4.4) years. A higher percentage of breast cancer survivors (46.2%) were obese as compared with controls (32.7%; P < 0.05), and a smaller percentage showed optimal values for low-density lipoprotein cholesterol, glucose, and C-reactive protein versus controls (P < 0.05). MetS was diagnosed in 50% of breast cancer survivors and in 37.5% of control group women (P < 0.05). Among the MetS diagnostic criteria, the most prevalent was abdominal obesity (waist circumference >88 cm), affecting 62.5% and 67.8% of the participants, respectively. In the control group, breast cancer survivors had a higher risk for MetS (OR, 1.66; 95% CI, 1.04-2.68), dysglycemia (OR, 1.05; 95% CI, 1.09-3.03), and hypertension (OR, 1.71; 95% CI, 1.02-2.89). CONCLUSIONS Postmenopausal breast cancer survivors present a higher risk of developing MetS as compared with women without breast cancer.
Collapse
|
76
|
Wei XL, Qiu MZ, Lin HX, Zhang Y, Liu JX, Yu HM, Liang WP, Jin Y, Ren C, He MM, Chen WW, Luo HY, Wang ZQ, Zhang DS, Wang FH, Li YH, Xu RH. Patients with old age or proximal tumors benefit from metabolic syndrome in early stage gastric cancer. PLoS One 2014; 9:e89965. [PMID: 24599168 PMCID: PMC3943843 DOI: 10.1371/journal.pone.0089965] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/25/2014] [Indexed: 12/12/2022] Open
Abstract
Background Metabolic syndrome and/or its components have been demonstrated to be risk factors for several cancers. They are also found to influence survival in breast, colon and prostate cancer, but the prognostic value of metabolic syndrome in gastric cancer has not been investigated. Methods Clinical data and pre-treatment information of metabolic syndrome of 587 patients diagnosed with early stage gastric cancer were retrospectively collected. The associations of metabolic syndrome and/or its components with clinical characteristics and overall survival in early stage gastric cancer were analyzed. Results Metabolic syndrome was identified to be associated with a higher tumor cell differentiation (P = 0.036). Metabolic syndrome was also demonstrated to be a significant and independent predictor for better survival in patients aged >50 years old (P = 0.009 in multivariate analysis) or patients with proximal gastric cancer (P = 0.047 in multivariate analysis). No association was found between single metabolic syndrome component and overall survival in early stage gastric cancer. In addition, patients with hypertension might have a trend of better survival through a good control of blood pressure (P = 0.052 in univariate analysis). Conclusions Metabolic syndrome was associated with a better tumor cell differentiation in patients with early stage gastric cancer. Moreover, metabolic syndrome was a significant and independent predictor for better survival in patients with old age or proximal tumors.
Collapse
Affiliation(s)
- Xiao-li Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Miao-zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huan-xin Lin
- Department of Preventive Care, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Zhang
- Department of Preventive Care, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jian-xin Liu
- Department of Preventive Care, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hong-mei Yu
- Department of Preventive Care, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wei-ping Liang
- Department of Preventive Care, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Jin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chao Ren
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ming-ming He
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wei-wei Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui-yan Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhi-qiang Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Dong-sheng Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Feng-hua Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Rui-hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- * E-mail:
| |
Collapse
|
77
|
Tehranifar P, Reynolds D, Fan X, Boden-Albala B, Engmann NJ, Flom JD, Terry MB. Multiple metabolic risk factors and mammographic breast density. Ann Epidemiol 2014; 24:479-83. [PMID: 24698111 DOI: 10.1016/j.annepidem.2014.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 01/20/2014] [Accepted: 02/12/2014] [Indexed: 12/29/2022]
Abstract
PURPOSE We examined whether obesity and a history of diabetes, hypertension, and elevated cholesterol, individually and in combination, are associated with breast density, a strong risk factor for breast cancer. METHODS We measured percent density and dense area using a computer-assisted method (n = 191; age range = 40-61 years). We used linear regression models to examine the associations of each metabolic condition and the number of metabolic conditions (zero, one, two, and three or four conditions) with breast density. RESULTS Among individual metabolic conditions, only high blood cholesterol was inversely associated with percent density (β = -5.4, 95% confidence interval [CI]: -8.5, -2.2) and dense area (β = -6.7, 95% CI = -11.1, -2.4). Having multiple metabolic conditions was also associated with lower breast density, with two conditions and three or four conditions versus zero conditions associated with 6.4% (95% CI: -11.2, -1.6) and 7.4% (95% CI: -12.9, -1.9) reduction in percent density and with 6.5 cm(2) (95% CI: -13.1, -0.1) and 9.5 cm(2) (95% CI: -17.1, -1.9) decrease in dense area. CONCLUSIONS A history of high blood cholesterol and multiple metabolic conditions were associated with lower relative and absolute measures of breast density. The positive association between metabolic abnormalities and breast cancer risk may be driven by pathways unrelated to mammographic breast density.
Collapse
Affiliation(s)
- Parisa Tehranifar
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY.
| | - Diane Reynolds
- School of Nursing, Long Island University, Brooklyn Campus, Brooklyn, NY
| | - Xiaozhou Fan
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| | - Bernadette Boden-Albala
- Division of Social Epidemiology, Department of Health Evidence and Policy, Department of Neurology, ICAHN School of Medicine at Mount Sinai, New York, NY
| | - Natalie J Engmann
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| | - Julie D Flom
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| |
Collapse
|
78
|
Llanos AA, Peng J, Pennell ML, Krok JL, Vitolins MZ, Degraffinreid CR, Paskett ED. Effects of tomato and soy on serum adipokine concentrations in postmenopausal women at increased breast cancer risk: a cross-over dietary intervention trial. J Clin Endocrinol Metab 2014; 99:625-32. [PMID: 24423335 PMCID: PMC3913803 DOI: 10.1210/jc.2013-3222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
CONTEXT Breast cancer risk among postmenopausal women increases as body mass index increases. Practical preventive methods to reduce risk of breast cancer are lacking. Few studies have investigated the effects of carotenoids and isoflavones on circulating adipokines in postmenopausal women. OBJECTIVE The aim was to examine the effects of lycopene- and isoflavone-rich diets on serum adipokines. DESIGN This was a 26-week, two-arm, longitudinal crossover trial. SETTING Participants were recruited from clinics at The Ohio State University Comprehensive Cancer Center. PARTICIPANTS Seventy postmenopausal women at increased breast cancer risk participated in the study. The mean age and body mass index of participants was 57.2 years and 30.0 kg/m(2), respectively; the study was comprised of 81.4% whites. INTERVENTIONS The interventions included 10 weeks of consumption of a tomato-based diet (≥25 mg lycopene daily) and 10 weeks of consumption of a soy-based diet (≥40 g of soy protein daily), with a 2-week washout in between. MAIN OUTCOME MEASURES Changes in serum adiponectin, leptin, and the adiponectin to leptin ratio were examined for each intervention through linear mixed models, with ratio estimates corresponding to postintervention adipokine concentrations relative to preintervention concentrations. RESULTS After the tomato intervention, among all women, adiponectin concentration increased (ratio 1.09, 95% confidence interval (CI) 1.00-1.18), with a stronger effect observed among nonobese women (ratio 1.13, 95% CI 1.02-1.25). After the soy intervention, adiponectin decreased overall (ratio 0.91, 95% CI 0.84-0.97), with a larger reduction observed among nonobese women (ratio 0.89, 95% CI 0.81-0.98). Overall, no significant changes in leptin or the adiponectin to leptin ratio were observed after either intervention. CONCLUSIONS Increasing dietary consumption of tomato-based foods may beneficially increase serum adiponectin concentrations among postmenopausal women at increased breast cancer risk, especially those who are not obese. Additional studies are essential to confirm these effects and to elucidate the specific mechanisms that may make phytonutrients found in tomatoes practical as breast cancer chemopreventive agents.
Collapse
Affiliation(s)
- Adana A Llanos
- Division of Population Sciences (A.A.L., J.L.K., C.R.D., E.D.P.), and Divisions of Biostatistics (J.P., M.L.P.) and Epidemiology (E.D.P.), College of Public Health, and Division of Cancer Prevention and Control (E.D.P.), College of Medicine, Ohio State University Comprehensive Cancer Center, The Ohio State University Columbus, Ohio 43210; Department of Epidemiology (A.A.L.), School of Public Health, Rutgers University, Piscataway, New Jersey 08854; Rutgers Cancer Institute of New Jersey (A.A.L.), New Brunswick, New Jersey 08903; and Department of Public Health Sciences (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | | | | | | | | | | | | |
Collapse
|
79
|
Suba Z. Triple-negative breast cancer risk in women is defined by the defect of estrogen signaling: preventive and therapeutic implications. Onco Targets Ther 2014; 7:147-64. [PMID: 24482576 PMCID: PMC3905095 DOI: 10.2147/ott.s52600] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epidemiologic studies strongly support that triple-negative breast cancers (TNBCs) may be distinct entities as compared with estrogen receptor (ER)+ tumors, suggesting that the etiologic factors, clinical characteristics, and therapeutic possibilities may vary by molecular subtypes. Many investigations propose that reproductive factors and exogenous hormone use differently or even quite inversely affect the risk of TNBCs and ER+ cancers. Controversies concerning the exact role of even the same risk factor in TNBC development justify that the biological mechanisms behind the initiation of both TNBCs and non-TNBCs are completely obscure. To arrive at a comprehensive understanding of the etiology of different breast cancer subtypes, we should also reconsider our traditional concepts and beliefs regarding cancer risk factors. Malignancies are multicausal, but the disturbance of proper estrogen signaling seems to be a crucial risk factor for the development of mammary cancers. The grade of defect in metabolic and hormonal equilibrium is directly associated with TNBC risk for women during their whole life. Inverse impact of menopausal status or parity on the development of ER+ and ER− breast cancers may not be possible; these controversial results derive from the misinterpretation of percentage-based statistical evaluations. Exogenous or parity-associated excessive estrogen supply is suppressive against breast cancer, though the lower the ER expression of tumors, the weaker the anticancer capacity. In women, the most important preventive strategy against breast cancers – included TNBCs – is the strict control and maintenance of hormonal equilibrium from early adolescence through the whole lifetime, particularly during the periods of great hormonal changes.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- National Institute of Oncology, Surgical and Molecular Tumor Pathology Centre, Budapest, Hungary
| |
Collapse
|
80
|
McDonald C, Bauer J, Capra S, Waterhouse M. Muscle function and omega-3 fatty acids in the prediction of lean body mass after breast cancer treatment. SPRINGERPLUS 2014; 2:681. [PMID: 24404435 PMCID: PMC3882342 DOI: 10.1186/2193-1801-2-681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 11/08/2013] [Indexed: 12/04/2022]
Abstract
Background Decreased lean body mass (LBM) is common in breast cancer survivors yet currently there is a lack of information regarding the determinants of LBM after treatment, in particular, the effect of physical activity and dietary factors, such as long-chain omega-3 fatty acids (LCn-3) on LBM and LBM function. This cross-sectional study explored associations of LBM and function with LCn-3 intake, dietary intake, inflammation, quality of life (QOL) and physical fitness in breast cancer survivors to improve clinical considerations when addressing body composition change. Methods Forty-nine women who had completed treatment (surgery, radiation and/or chemotherapy) were assessed for body composition (BODPOD), LCn-3 content of erythrocytes, C-reactive protein (CRP), QOL, dietary intake, objective physical activity, 1-min push-ups, 1-min sit-stand, sub-maximal treadmill (TM) test, and handgrip strength. Results After adjustment for age, LBM was associated with push-ups (r = 0.343, p = 0.000), stage reached on treadmill (StageTM) (r = 0.302, 0.001), % time spent ≥ moderate activity (Mod + Vig) (r = 0.228, p = 0.024). No associations were seen between anthropometric values and any treatment, diagnostic and demographical variables. Body mass, push-ups and StageTM accounted for 76.4% of the variability in LBM (adjusted r-square: 0.764, p = 0.000). After adjustment docosahexanoic acid (DHA) was positively associated with push-ups (β=0.399, p = 0.001), eicosapentanoic acid (EPA) was negatively associated with squats (r = −0.268, p = 0.041), with no other significant interactions found between LCn-3 and physical activity for LBM or LBM function. Conclusion This is the first investigation to report that a higher weight adjusted LBM is associated with higher estimated aerobic fitness and ability to perform push-ups in breast cancer survivors. Potential LCn-3 and physical activity interactions on LBM require further exploration.
Collapse
Affiliation(s)
| | - Judy Bauer
- University of Queensland, Brisbane, 4059 Australia
| | - Sandra Capra
- University of Queensland, Brisbane, 4059 Australia
| | | |
Collapse
|
81
|
Qu YY, Dai B, Kong YY, Chang K, Ye DW, Yao XD, Zhang SL, Zhang HL, Yang WY. Influence of obesity on localized prostate cancer patients treated with radical prostatectomy. Asian J Androl 2013; 15:747-52. [PMID: 24036920 DOI: 10.1038/aja.2013.104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 06/26/2013] [Accepted: 07/12/2013] [Indexed: 11/09/2022] Open
Abstract
This study aimed to investigate the association between different anthropometric measures of obesity and clinicopathological characteristics in Chinese patients with clinically localized prostate cancer (PCa). A total of 734 patients with clinically localized PCa who underwent radical prostatectomy (RP) were included in this study. Clinical and pathological data from each patient were collected. Anthropometric measures of abdominal adiposity were measured from T2-weighted sagittal localisation images from magnetic resonance imaging (MRI) for 413 (56.3%) patients. Patient clinical and pathological characteristics were compared across body mass index (BMI) groups. Univariable and multivariable logistic regression models were used to address the influence of the preoperative total testosterone level and anthropometric measures of obesity on pathological outcomes. In the multivariate analysis, BMI was not significantly associated with any pathological outcomes. However, the percentage of visceral adipose tissue (VAT%) was an independent predictor of a pathological Gleason score ≥8 (P<0.001), extracapsular extension (ECE; P=0.002) and seminal vesicle invasion (SVI; P=0.007). More importantly, we found that the preoperative total testosterone level was significantly correlated with the VAT% (Pearson's correlation coefficient: -0.485, P<0.001) and subcutaneous adipose tissue (SAT; Pearson's correlation coefficient: 0.413, P<0.001). In conclusion, the results of this study suggest that abdominal fat distribution, and particularly VAT%, is associated with a risk of advanced PCa. Moreover, our present study confirms a significant inverse correlation between visceral adiposity and testosterone. Further studies are warranted to elucidate the biological mechanisms underlying the relationship between abdominal adiposity and the aggressiveness of PCa.
Collapse
Affiliation(s)
- Yuan-Yuan Qu
- 1] Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, China [2] Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Suba Z. Circulatory estrogen level protects against breast cancer in obese women. Recent Pat Anticancer Drug Discov 2013; 8:154-67. [PMID: 23061769 PMCID: PMC3636519 DOI: 10.2174/1574892811308020004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 10/08/2012] [Accepted: 10/09/2012] [Indexed: 12/15/2022]
Abstract
Literary data suggest apparently ambiguous interaction between menopausal status and obesity-associated breast cancer risk based on the principle of the carcinogenic capacity of estrogen. Before menopause, breast cancer incidence is relatively low and adiposity is erroneously regarded as a protective factor against this tumor conferred by the obesity associated defective estrogen-synthesis. By contrast, in postmenopausal cases, obesity presents a strong risk factor for breast cancer being mistakenly attributed to the presumed excessive estrogen-production of their adipose-tissue mass. Obesity is associated with dysmetabolism and endangers the healthy equilibrium of sexual hormone-production and regular menstrual cycles in women, which are the prerequisites not only for reproductive capacity but also for somatic health. At the same time, literary data support that anovulatory infertility is a very strong risk for breast cancer in young women either with or without obesity. In the majority of premenopausal women, obesity associated insulin resistance is moderate and may be counteracted by their preserved circulatory estrogen level. Consequently, it is not obesity but rather the still sufficient estrogen-level, which may be protective against breast cancer in young adult females. In obese older women, never using hormone replacement therapy (HRT) the breast cancer risk is high, which is associated with their continuous estrogen loss and increasing insulin-resistance. By contrast, obese postmenopausal women using HRT, have a decreased risk for breast cancer as the protective effect of estrogen-substitution may counteract to their obesity associated systemic alterations. The revealed inverse correlation between circulatory estrogen-level and breast cancer risk in obese women should advance our understanding of breast cancer etiology and promotes primary prevention measures. New patents recommend various methods for the prevention and treatment of obesity-related systemic disorders and the associated breast cancer.
Collapse
Affiliation(s)
- Zsuzsanna Suba
- National Institute of Oncology, Surgical and Molecular Tumor Pathology Centre, Address: H-1122 Rath Gyorgy str. 7-9, Budapest, Hungary.
| |
Collapse
|
83
|
Strong AL, Semon JA, Strong TA, Santoke TT, Zhang S, McFerrin HE, Gimble JM, Bunnell BA. Obesity-associated dysregulation of calpastatin and MMP-15 in adipose-derived stromal cells results in their enhanced invasion. Stem Cells 2013; 30:2774-83. [PMID: 22969001 DOI: 10.1002/stem.1229] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 08/21/2012] [Indexed: 01/29/2023]
Abstract
Adipose tissue maintains a subpopulation of cells, referred to as adipose-derived stromal/stem cells (ASCs), which have been associated with increased breast cancer tumorigenesis and metastasis. For ASCs to affect breast cancer cells, it is necessary to delineate how they mobilize and home to cancer cells, which requires mobilization and invasion through extracellular matrix barriers. In this study, ASCs were separated into four different categories based on the donor's obesity status and depot site of origin. ASCs isolated from the subcutaneous abdominal adipose tissue of obese patients (Ob(+)Ab(+)) demonstrated increased invasion through Matrigel as well as a chick chorioallantoic membrane, a type I collagen-rich extracellular matrix barrier. Detailed mRNA and protein analyses revealed that calpain-4, calpastatin, and MMP-15 were associated with increased invasion, and the silencing of each protease or protease inhibitor confirmed their role in ASC invasion. Thus, the data indicate that both the donor's obesity status and depot site of origin distinguishes the properties of subcutaneous-derived ASCs with respect to enhanced invasion and this is associated with the dysregulation of calpain-4, calpastatin, and MMP-15.
Collapse
Affiliation(s)
- Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Nagaraju GP, Zafar SF, El-Rayes BF. Pleiotropic effects of genistein in metabolic, inflammatory, and malignant diseases. Nutr Rev 2013; 71:562-72. [PMID: 23865800 DOI: 10.1111/nure.12044] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genistein is a soy-derived biologically active isoflavone that exhibits diverse health-promoting effects. An increasing body of evidence shows that genistein influences lipid homeostasis and insulin resistance, counteracts inflammatory cytokines, and possesses antidiabetic properties. Genistein also impedes cancer progression by promoting apoptosis, inducing cell cycle arrest, modulating intracellular signaling pathways, and inhibiting angiogenesis and metastasis of neoplastic cells. This review summarizes the pleiotropic functions of genistein in common health disorders such as metabolic syndrome, chronic inflammatory diseases, and cancer. In the current era of uncontrolled health expenditure, a focus on the clinical development of nutritional agents with the capacity to prevent a variety of common health disorders is needed. As a micronutrient that exerts multifaceted effects ranging from antidiabetic to anticarcinogenic functions, genistein should be clinically developed further for use in the prevention and treatment of a variety of health disorders.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
85
|
Seim I, Lubik AA, Lehman ML, Tomlinson N, Whiteside EJ, Herington AC, Nelson CC, Chopin LK. Cloning of a novel insulin-regulated ghrelin transcript in prostate cancer. J Mol Endocrinol 2013; 50:179-91. [PMID: 23267039 DOI: 10.1530/jme-12-0150] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ghrelin is a multifunctional hormone, with roles in stimulating appetite and regulating energy balance, insulin secretion and glucose homoeostasis. The ghrelin gene locus (GHRL) is highly complex and gives rise to a range of novel transcripts derived from alternative first exons and internally spliced exons. The wild-type transcript encodes a 117 amino acid preprohormone that is processed to yield the 28 amino acid peptide ghrelin. Here, we identified insulin-responsive transcription corresponding to cryptic exons in intron 2 of the human ghrelin gene. A transcript, termed in2c-ghrelin (intron 2-cryptic), was cloned from the testis and the LNCaP prostate cancer cell line. This transcript may encode an 83 amino acid preproghrelin isoform that codes for ghrelin, but not obestatin. It is expressed in a limited number of normal tissues and in tumours of the prostate, testis, breast and ovary. Finally, we confirmed that in2c-ghrelin transcript expression, as well as the recently described in1-ghrelin transcript, is significantly upregulated by insulin in cultured prostate cancer cells. Metabolic syndrome and hyperinsulinaemia have been associated with prostate cancer risk and progression. This may be particularly significant after androgen deprivation therapy for prostate cancer, which induces hyperinsulinaemia, and this could contribute to castrate-resistant prostate cancer growth. We have previously demonstrated that ghrelin stimulates prostate cancer cell line proliferation in vitro. This study is the first description of insulin regulation of a ghrelin transcript in cancer and should provide further impetus for studies into the expression, regulation and function of ghrelin gene products.
Collapse
Affiliation(s)
- Inge Seim
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | | | | | | | | | | | | | | |
Collapse
|
86
|
|
87
|
Fini MA, Elias A, Johnson RJ, Wright RM. Contribution of uric acid to cancer risk, recurrence, and mortality. Clin Transl Med 2012; 1:16. [PMID: 23369448 PMCID: PMC3560981 DOI: 10.1186/2001-1326-1-16] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/17/2012] [Indexed: 02/07/2023] Open
Abstract
Two risk factors for the development and progression of cancers that are amenable to life style modification are chronic inflammation and the metabolic syndrome. This review proposes two new targets that may mechanistically integrate inflammation and metabolic syndrome, have been largely ignored, and are known to be druggable. Recent evidence has demonstrated that elevated serum uric acid (hyperuricemia) is associated with excess cancer risk, recurrence, and mortality. Although uric acid (UA) can function as a systemic antioxidant, its pro-inflammatory properties have been postulated to play an important role in the pathogenesis of cancer. Furthermore, obesity, Type 2 Diabetes Mellitus (T2DM), and the metabolic syndrome (MetS) are also associated with excess cancer, chronic inflammation, and with hyperuricemia, suggesting that UA may represent an important link between these disorders and the development of cancer. While pharmacological modulation of hyperuricemia could in principal augment anti-cancer therapeutic strategies, some cancer cells express low intracellular levels of the enzyme Xanthine Oxidoreductase (XOR) that are associated with increased cancer aggressiveness and poor clinical outcome. Thus, systemic pharmacological inhibition of XOR may worsen clinical outcome, and specific strategies that target serum uric acid (SUA) without inhibiting tumor cell XOR may create new therapeutic opportunities for cancer associated with hyperuricemia. This review will summarize the evidence that elevated SUA may be a true risk factor for cancer incidence and mortality, and mechanisms by which UA may contribute to cancer pathogenesis will be discussed in the hope that these will identify new opportunities for cancer management.
Collapse
Affiliation(s)
- Mehdi A Fini
- Department of Medicine, Pulmonary Division and Webb-Waring Center, University of Colorado Denver, Anschutz Medical Campus, V20, Room 3105, Mail stop C-322 12850 East Montview Boulevard, Aurora, CO, 80045-0511, USA.
| | | | | | | |
Collapse
|
88
|
Caan BJ, Kwan ML, Shu XO, Pierce JP, Patterson RE, Nechuta SJ, Poole EM, Kroenke CH, Weltzien EK, Flatt SW, Quesenberry CP, Holmes MD, Chen WY. Weight change and survival after breast cancer in the after breast cancer pooling project. Cancer Epidemiol Biomarkers Prev 2012; 21:1260-71. [PMID: 22695738 PMCID: PMC3433249 DOI: 10.1158/1055-9965.epi-12-0306] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Weight change after a breast cancer diagnosis has been linked to lower survival. To further understand effects of postdiagnostic weight variation on survival, we examined the relationship by comorbid status and initial body mass index (BMI). METHODS The current analysis included 12,915 patients with breast cancer diagnosed between 1990 and 2006 with stage I-III tumors from four prospective cohorts in the United States and China. HRs and 95% confidence intervals (CI) representing the associations of five weight change categories [within <5% (reference); 5%-<10% and ≥10% loss and gain] with mortality were estimated using Cox proportional hazards models. RESULTS Mean weight change was 1.6 kg. About 14.7% women lost and 34.7% gained weight. Weight stability in the early years postdiagnosis was associated with the lowest overall mortality risk. Weight loss ≥10% was related to a 40% increased risk of death (HR, 1.41; 95% CI, 1.14-1.75) in the United States and over three times the risk of death (HR, 3.25; 95% CI: 2.24, 4.73) in Shanghai. This association varied by prediagnosis BMI, and in the United States, lower survival was seen for women who lost weight and had comorbid conditions. Weight gain ≥10% was associated with a nonsignificant increased risk of death. CONCLUSIONS Prevention of excessive weight gain is a valid public health goal for breast cancer survivors. Although intentionality of weight loss could not be determined, women with comorbid conditions may be particularly at risk of weight loss and mortality. IMPACT Weight control strategies for breast cancer survivors should be personalized to the individual's medical history.
Collapse
Affiliation(s)
- Bette J Caan
- Division of Research, Kaiser Permanente, Oakland, California 94612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL, Bandera EV, Hamilton KK, Grant B, McCullough M, Byers T, Gansler T. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin 2012; 62:243-74. [PMID: 22539238 DOI: 10.3322/caac.21142] [Citation(s) in RCA: 1327] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer survivors are often highly motivated to seek information about food choices, physical activity, and dietary supplements to improve their treatment outcomes, quality of life, and overall survival. To address these concerns, the American Cancer Society (ACS) convened a group of experts in nutrition, physical activity, and cancer survivorship to evaluate the scientific evidence and best clinical practices related to optimal nutrition and physical activity after the diagnosis of cancer. This report summarizes their findings and is intended to present health care providers with the best possible information with which to help cancer survivors and their families make informed choices related to nutrition and physical activity. The report discusses nutrition and physical activity guidelines during the continuum of cancer care, briefly highlighting important issues during cancer treatment and for patients with advanced cancer, but focusing largely on the needs of the population of individuals who are disease free or who have stable disease following their recovery from treatment. It also discusses select nutrition and physical activity issues such as body weight, food choices, food safety, and dietary supplements; issues related to selected cancer sites; and common questions about diet, physical activity, and cancer survivorship.
Collapse
Affiliation(s)
- Cheryl L Rock
- Department of Family and Preventive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Pituskin E, Paterson I, Haykowsky M. The Role of Exercise Interventions in Reducing the Risk for Cardiometabolic Disease in Cancer Survivors. CURRENT CARDIOVASCULAR RISK REPORTS 2012. [DOI: 10.1007/s12170-012-0244-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
91
|
Given breast cancer, does breast size matter? Data from a prospective breast cancer cohort. Cancer Causes Control 2012; 23:1307-16. [DOI: 10.1007/s10552-012-0008-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 05/31/2012] [Indexed: 10/28/2022]
|
92
|
Reeves KW, McLaughlin V, Fredman L, Ensrud K, Cauley JA. Components of metabolic syndrome and risk of breast cancer by prognostic features in the study of osteoporotic fractures cohort. Cancer Causes Control 2012; 23:1241-51. [PMID: 22661101 DOI: 10.1007/s10552-012-0002-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/18/2012] [Indexed: 11/26/2022]
Abstract
PURPOSE Metabolic syndrome (MetS) and most of its components have been previously associated with increased breast cancer risk. We hypothesized that increasing number of MetS components would be positively associated with breast cancer risk. METHODS Data were obtained from the Study of Osteoporotic Fractures, a prospective cohort of women aged ≥65 enrolled between 1986 and 1988 and still being followed prospectively (n = 8,956). MetS components evaluated at baseline were elevated waist circumference, hypertension, and diabetes. Data were not available on hyperlipidemia. Incident breast cancers were confirmed by pathology report. We compared women with 0, 1, and 2 or 3 MetS components. We used Cox proportional hazards regression to calculate associations for breast cancer overall and classified by prognostic features. RESULTS At baseline, 28.8 % of participants had 2 or 3 MetS components. Over an average follow-up of 14.4 years, 551 breast cancer cases were identified. Compared to those with no components, women with 2 or 3 components had increased breast cancer risk (hazard ratio (HR), 1.30; 95 % confidence interval (CI), 1.01-1.68) and increased risk of ER+ (HR, 1.48; 95 % CI, 1.09-2.03) and PR+ (HR, 1.56; 95 % CI, 1.10-2.20) cancer, adjusting for age, hormone use, and family history of breast cancer. These results became attenuated and not statistically significant when additionally adjusted for body mass index. CONCLUSIONS MetS is associated with increased postmenopausal breast cancer risk, especially for ER+ and PR+ cancers, though this effect may not be independent of the effect of body mass index. Managing the components of MetS could be efficacious for breast cancer risk reduction.
Collapse
Affiliation(s)
- Katherine W Reeves
- Division of Biostatistics and Epidemiology, University of Massachusetts Amherst, 715 North Pleasant Street, Amherst, MA 01003, USA.
| | | | | | | | | |
Collapse
|
93
|
Stebbing J, Sharma A, North B, Athersuch TJ, Zebrowski A, Pchejetski D, Coombes RC, Nicholson JK, Keun HC. A metabolic phenotyping approach to understanding relationships between metabolic syndrome and breast tumour responses to chemotherapy. Ann Oncol 2012; 23:860-6. [PMID: 21821546 DOI: 10.1093/annonc/mdr347] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Breast cancer is associated with adverse outcomes in patients with the metabolic syndrome phenotype. To study this further, we examined the relationship between serum metabolite levels and the components of metabolic syndrome with treatment outcomes in breast cancer. METHODS A total of 88 women with measurable breast cancer were studied; their serum metabolites as assessed by (1)H nuclear magnetic resonance spectroscopy, blood pressure, lipids, glucose, body mass index and waist circumference were recorded and correlated with treatment response. RESULTS We identified metabolic syndrome in approximately half of our cohort (42 patients) and observed a significant trend (P = 0.03) of increased incidence of metabolic syndrome in partial response (33.3%), stable disease (42.9%) and progressive disease groups (66.1%). High blood sugar predicted a poor response (P < 0.001). Logistic regression of metabonomic data demonstrated that high lactate (P = 0.03) and low alanine (P = 0.01) combined with high glucose (P = 0.01) were associated with disease progression. CONCLUSIONS Metabolic syndrome is commonly observed in metastatic breast cancer and these patients have poorer outcomes. These data, which support our previous findings, suggest that high blood glucose as part of metabolic syndrome is associated with a poor response in breast cancer. They also validate new therapeutic approaches that focus on metabolism.
Collapse
Affiliation(s)
- J Stebbing
- Section of Oncology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Aronis KN, Mantzoros CS. A brief history of insulin resistance: from the first insulin radioimmunoassay to selectively targeting protein kinase C pathways. Metabolism 2012; 61:445-9. [PMID: 22304840 DOI: 10.1016/j.metabol.2012.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 01/03/2012] [Indexed: 12/29/2022]
|
95
|
Anderson GL, Neuhouser ML. Obesity and the Risk for Premenopausal and Postmenopausal Breast Cancer: Table 1. Cancer Prev Res (Phila) 2012; 5:515-21. [DOI: 10.1158/1940-6207.capr-12-0091] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
96
|
Neuhouser ML, Perrigue MM. The Role of Energy Balance in Cancer Prevention. Curr Nutr Rep 2012. [DOI: 10.1007/s13668-011-0001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
97
|
Healy LA, Howard JM, Ryan AM, Beddy P, Mehigan B, Stephens R, Reynolds JV. Metabolic syndrome and leptin are associated with adverse pathological features in male colorectal cancer patients. Colorectal Dis 2012; 14:157-65. [PMID: 21689278 DOI: 10.1111/j.1463-1318.2011.02562.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM Metabolic syndrome (MetS) describes a clustering of factors including central obesity, hypertension and raised plasma glucose, triglycerides and high-density lipoprotein (HDL) cholesterol. Central obesity is associated with a risk for colorectal cancer, but the impact of MetS on colorectal cancer biology and outcomes is unclear. METHOD A prospective observational study of colorectal cancer patients was carried out in an Irish population. Patients underwent metabolic and anthropometric assessment before treatment, including measurement of serum hormones and adipokines and CT measurement of visceral fat. MetS was defined according to the International Diabetes Federation definition(3) . RESULTS One-hundred and thirty consecutive colorectal cancer patients (66 men and 64 women) were recruited. MetS was diagnosed in 38% patients compared with the population norms reported at 21%(21) . Male patients had a significantly greater visceral fat area compared with female patients. MetS was associated with node-positive disease (P = 0.026), percentage nodal involvement (P = 0.033) and extramural vascular invasion (P = 0.049) in male patients but no significant association was observed in female patients. HDL cholesterol was also significantly associated with a more advanced pathological stage (P = 0.014) and node-positive disease (P = 0.028). Leptin was associated with nodal status (P = 0.036), microvascular invasion (P = 0.054), advanced pathological stage (P = 0.046) and more advanced Dukes stage (P = 0.042). CONCLUSION We report a high prevalence of MetS and visceral obesity in a colorectal cancer population. MetS and plasma leptin are associated with a more aggressive tumour phenotype in male patients only.
Collapse
Affiliation(s)
- L A Healy
- Department of Clinical Nutrition, St James's Hospital and Trinity College Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
98
|
Fischer L, Clemens G, Gehrig T, Kenngott H, Becker K, Bruckner T, Gutt CN, Büchler MW, Müller-Stich B. Challenges and pitfalls of experimental bariatric procedures in rats. Obes Facts 2012; 5:359-71. [PMID: 22722345 DOI: 10.1159/000339531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 11/15/2011] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The impact of Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) on obesity and obesity-related diseases is unquestionable. Up to now, the technical descriptions of these techniques in animals/rats have not been very comprehensive. METHODS For SG and RYGB, operating time, learning curve, and intraoperative mortality in relation to weight of the rat and type of anesthesia were recorded. Furthermore, a review of the literature on experimental approaches towards SG and RYGB in rats was carried out, merging in a detailed technical description for both procedures. RESULTS The data presented here revealed that the mean operating time for SG (69.4 ± 22.2 min (SD)) was shorter than for RYGB (123.0 ± 20.7 min). There is a learning curve for both procedures, resulting in a reduced operating time of up to 60% in SG and 35% in RYGB (p < 0.05; t-test). However, with increased weight, operating time increases to about 80 min for SG and about 120 min for RYGB. Obese rats have an increased intraoperative mortality rate of up to 50%. After gaseous anesthesia the mortality can be even higher. The literature search revealed 40 papers dealing with SG and RYGB in rats. 18 articles (45%) contained neither photographs nor illustrations; 14 articles (35%) did not mention the applied type of anesthesia. The mortality rate was described in 15 papers (37.5%). CONCLUSION Experimental obesity surgery in rats is challenging. Because of the high mortality in obese rats operated under gaseous anesthesia, exercises to establish the techniques should be performed in small rats using intraperitoneal anesthesia.
Collapse
Affiliation(s)
- Lars Fischer
- Department of Surgery, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
This paper presents emerging evidence linking visceral adiposity and the metabolic syndrome (MetSyn) with carcinogenesis. The link between obesity and cancer has been clearly identified in a multitude of robust epidemiological studies. Research is now focusing on the role of visceral adipose tissue in carcinogenesis; as it is recognised as an important metabolic tissue that secretes factors that systemically alter the immunological, metabolic and endocrine milieu. Excess visceral adipose tissue gives rise to a state of chronic systemic inflammation with associated insulin resistance and dysmetabolism, collectively known as the MetSyn. Prospective cohort studies have shown associations between visceral adiposity, the MetSyn and increased risk of breast cancer, colorectal cancer and oesophageal adenocarcinoma. Furthermore, visceral adiposity and the MetSyn have been associated with increased tumour progression and reduced survival. The mechanisms by which visceral adiposity and the MetSyn are thought to promote tumorigenesis are manifold. These include alterations in adipokine secretion and cell signalling pathways. In addition, hyperinsulinaemia, subsequent insulin resistance and stimulation of the insulin-like growth factor-1 axis have all been linked with visceral adiposity and promote tumour progression. Furthermore, the abundance of inflammatory cells in visceral adipose tissue, including macrophages and T-cells, create systemic inflammation and a pro-tumorigenic environment. It is clear from current research that excess visceral adiposity and associated dysmetabolism play a central role in the pathogenesis of certain cancer types. Further research is required to elucidate the exact mechanisms at play and identify potential targets for intervention.
Collapse
|
100
|
A retrospective review of the metabolic syndrome in women diagnosed with breast cancer and correlation with estrogen receptor. Breast Cancer Res Treat 2011; 131:325-31. [PMID: 21964613 DOI: 10.1007/s10549-011-1790-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 09/15/2011] [Indexed: 02/07/2023]
Abstract
Women diagnosed with obesity and breast cancer have an increased risk of recurrence and death (Protani et al., Breast Cancer Res Treat 123:627-635, 1). Obesity is associated with the metabolic syndrome--a pathophysiologically distinct inflammatory process comprised of central obesity, insulin resistance, hypertension, and atherogenic dyslipidemia. The relationship of obesity as a risk factor for breast cancer is complex with a protective effect for younger women in contrast to a risk for older women (Kabat et al., Cancer Epidemiol Biomarkers Prev 18:2046-2053, 2; Ursin et al., Epidemiology 6:137-141, 3). The metabolic syndrome has been associated with the risk of cancer, and pro-inflammatory circulating factors may be associated with risk of more aggressive breast cancer (Capasso et al., Cancer Biol Ther 10:1240-1243, 4; Healy et al., Clin Oncol (R Coll Radiol) 22:281-288, 5; Laukkanen et al., Cancer Epidemiol Biomarkers Prev 13:1646-1650, 6). We conducted a retrospective review of 860 breast cancer patients to determine the relationship between estrogen receptor status and the metabolic syndrome. We collected the relevant metabolic diagnoses, medications, physical findings, and laboratory values and adapted the National Cholesterol Education Program criteria to define the metabolic syndrome retrospectively. No relationship was found between estrogen receptor status and the individual components of the metabolic syndrome. Based on findings in the medical records, 15% of the women with breast cancer had the metabolic syndrome, and 26% of the women were considered obese, 16% hyperglycemic, 54% hypertensive, and 30% dyslipidemic. The metabolic syndrome was associated with advanced age and African-American race (P < 0.001). When adjusted for age, race, and stage, the metabolic syndrome was marginally associated with estrogen receptor-positive tumors (P = 0.054). Our findings do not support the concern that the metabolic syndrome may contribute to more biologically aggressive breast cancer.
Collapse
|