151
|
Journy NM, Bernier MO, Doody MM, Alexander BH, Linet MS, Kitahara CM. Hyperthyroidism, Hypothyroidism, and Cause-Specific Mortality in a Large Cohort of Women. Thyroid 2017; 27:1001-1010. [PMID: 28578598 PMCID: PMC5564026 DOI: 10.1089/thy.2017.0063] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The prevalence of hyperthyroidism and hypothyroidism is 0.5-4% in iodine-replete communities, but it is 5-10 times higher in women than in men. Those conditions are associated with a broad range of metabolic disorders and cardiovascular diseases. Biological evidence of a role of thyroid hormones in carcinogenesis also exists. However, the association between thyroid dysfunction and cardiovascular disease or cancer mortality risk remains controversial. In a large cohort of women, the associations of hyperthyroidism and hypothyroidism with cause-specific mortality were evaluated after nearly 30 years of follow-up. METHODS The prospective study included 75,076 women aged 20-89 years who were certified as radiologic technologists in the United States in 1926-1982, completed baseline questionnaires in 1983-1998 from which medical history was ascertained, and reported no malignant disease or benign thyroid disease except thyroid dysfunction. A passive follow-up of this cohort was performed through the Social Security Administration database and the National Death Index-Plus. Cause-specific mortality risks were compared according to self-reported thyroid status, with proportional hazards models adjusted for baseline year and age, race/ethnicity, body mass index, family history of breast cancer, and life-style and reproductive factors. RESULTS During a median follow-up of 28 years, 2609 cancer, 1789 cardiovascular or cerebrovascular, and 2442 other non-cancer deaths were recorded. Women with hyperthyroidism had an elevated risk of breast cancer mortality after 60 years of age (hazard ratio [HR] = 2.04 [confidence interval (CI) 1.16-3.60], 13 cases in hyperthyroid women) compared to women without thyroid disease. Hypothyroid women had increased mortality risks for diabetes mellitus (HR = 1.58 [CI 1.03-2.41], 27 cases in hypothyroid women), cardiovascular disease (HR = 1.20 [CI 1.01-1.42], 179 cases), and cerebrovascular disease (HR = 1.45 [CI 1.01-2.08], 35 cases, when restricting the follow-up to ≥10 years after baseline). Other causes of death were not associated with hyperthyroidism or hypothyroidism, though there was a suggestion of an elevated risk of ovarian cancer mortality in hyperthyroid women based on very few cases. CONCLUSION The excess mortality risks observed in a large, prospective 30-year follow-up of patients with thyroid dysfunction require confirmation, and, if replicated, further investigation will be needed because of the clinical implications.
Collapse
Affiliation(s)
- Neige M.Y. Journy
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Marie-Odile Bernier
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratoire d'épidémiologie des rayonnements ionisants, Service de Radiobiologie et d'Epidémiologie, Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
| | - Michele M. Doody
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bruce H. Alexander
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Martha S. Linet
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Cari M. Kitahara
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
152
|
Tsujimoto T, Kajio H, Sugiyama T. Association between hyperinsulinemia and increased risk of cancer death in nonobese and obese people: A population-based observational study. Int J Cancer 2017; 141:102-111. [PMID: 28390156 PMCID: PMC5435954 DOI: 10.1002/ijc.30729] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/18/2017] [Accepted: 04/03/2017] [Indexed: 12/12/2022]
Abstract
Obesity, metabolic syndrome and type 2 diabetes are associated with cancer-related mortality. We assessed whether hyperinsulinemia is a risk factor for cancer death in nonobese people without diabetes. We conducted a prospective cohort study using data from the National Health and Nutrition Examination Survey 1999-2010 and followed up the participants until December 31, 2011. For the primary analysis of cancer mortality, we used Cox proportional hazard models to estimate hazard ratios (HRs) in the participants with hyperinsulinemia and those without. Hyperinsulinemia was defined as a fasting insulin level of ≥10 μU/mL. To identify causes of deaths, the International Classification of Diseases, Tenth Revision codes were used. This study included 9,778 participants aged 20 years or older without diabetes or a history of cancer: 6,718 nonobese participants (2,057 with hyperinsulinemia [30.6%]) and 3,060 obese participants (2,303 with hyperinsulinemia [75.3%]). A total of 99.9% completed follow-up. Among all study participants, cancer mortality was significantly higher in those with hyperinsulinemia than in those without hyperinsulinemia (adjusted HR 2.04, 95% CI 1.24-3.34, p = 0.005). Similarly, among nonobese participants, multivariable analysis showed that cancer mortality was significantly higher in those with hyperinsulinemia than in those without (adjusted HR 1.89, 95% CI 1.07-3.35, p = 0.02). Considering that nonobese people with hyperinsulinemia were at higher risk of cancer mortality than those without hyperinsulinemia, improvement of hyperinsulinemia may be an important approach for preventing cancer regardless of the presence or absence of obesity.
Collapse
Affiliation(s)
- Tetsuro Tsujimoto
- Department of Diabetes, Endocrinology, and MetabolismCenter Hospital, National Center for Global Health and MedicineTokyoJapan
| | - Hiroshi Kajio
- Department of Diabetes, Endocrinology, and MetabolismCenter Hospital, National Center for Global Health and MedicineTokyoJapan
| | - Takehiro Sugiyama
- Diabetes and Metabolism Information Center, Research Institute, National Center for Global Health and MedicineTokyoJapan
- Department of Public Health/Health Policythe University of TokyoTokyoJapan
| |
Collapse
|
153
|
Christian AH, O'Malley D, Barac A, Miller SM, Hudson SV. Cardiovascular risk and communication among early stage breast cancer survivors. PATIENT EDUCATION AND COUNSELING 2017; 100:1360-1366. [PMID: 28215826 PMCID: PMC5568653 DOI: 10.1016/j.pec.2017.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/01/2017] [Accepted: 02/09/2017] [Indexed: 06/06/2023]
Abstract
OBJECTIVE African American (AA) women have disproportionately high rates of cardiovascular (CV) risk factors that, coupled with potentially cardiotoxic breast cancer (BC) therapies, place them at risk of adverse CV outcomes. The purpose of this study was to assess CV risk factors among white and AA BC survivors and perceptions of patient-provider CV-related communication and post-treatment information needs. METHODS Early-stage, English-speaking BC survivors were recruited through community-based survivorship organizations/clinics in New Jersey. Qualitative, semi-structured interviews and a brief survey of socio-demographics and comorbidities were conducted. RESULTS BC survivors reported high rates of CV risk factors. Most reported their health as good/excellent despite their cancer history and CV risk profile. Approximately half of overweight/obese BC survivors reported no weight-related conversation with their health care team and, among those who reported discussion, 36% indicated it was self-initiated and dismissed. More than half of BC survivors' reported a desire for lifestyle-related information and guidance. CONCLUSION BC survivors may not understand how their separate, yet cumulative, health issues can impact their CV risk. PRACTICE IMPLICATIONS Patient-provider communication on behavioral risk factor modification and CV risk is needed, particularly among at-risk subgroups such as AA and obese BC survivors, to improve BC and CV outcomes.
Collapse
Affiliation(s)
- Allison H Christian
- Department of Health Education and Behavioral Science, Rutgers, The State University, School of Public Health, Piscataway, NJ, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.
| | | | - Ana Barac
- MedStar Heart & Vascular Institute, MedStar Washington Hospital Center, Washington, DC, United States
| | - Suzanne M Miller
- Fox Chase Cancer Center/Temple University Health System, Philadelphia, PA, United States
| | - Shawna V Hudson
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States; Department of Family Medicine and Community Health, Rutgers Robert Wood Johnson Medical School, Somerset, NJ, United States
| |
Collapse
|
154
|
Abstract
PURPOSE OF REVIEW Adipocytes have adapted to store energy in the form of lipid and also secrete circulating factors called adipokines that signal to other tissues to coordinate energy homeostasis. These functions are disrupted in the setting of obesity, promoting the development of diseases such as diabetes, cardiovascular disease, and cancer. RECENT FINDINGS Obesity is linked to an increased risk of many types of cancer and increased cancer-related mortality. The basis for the striking association between obesity and cancer is not well understood. Here, we review the cellular and molecular pathways that appear to be involved in obesity-driven cancer. We also describe possible therapeutic considerations and highlight important unanswered questions in the field.
Collapse
Affiliation(s)
- Sarah E Ackerman
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Olivia A Blackburn
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
- The Rockefeller University, 1230 York Avenue, Box 223, New York, NY, 10065, USA.
| |
Collapse
|
155
|
Interaction of insulin-like growth factor-I and insulin resistance-related genetic variants with lifestyle factors on postmenopausal breast cancer risk. Breast Cancer Res Treat 2017; 164:475-495. [PMID: 28478612 DOI: 10.1007/s10549-017-4272-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/29/2017] [Indexed: 12/31/2022]
Abstract
PURPOSE Genetic variants and traits in metabolic signaling pathways may interact with obesity, physical activity, and exogenous estrogen (E), influencing postmenopausal breast cancer risk, but these inter-related pathways are incompletely understood. METHODS We used 75 single-nucleotide polymorphisms (SNPs) in genes related to insulin-like growth factor-I (IGF-I)/insulin resistance (IR) traits and signaling pathways, and data from 1003 postmenopausal women in Women's Health Initiative Observation ancillary studies. Stratifying via obesity and lifestyle modifiers, we assessed the role of IGF-I/IR traits (fasting IGF-I, IGF-binding protein 3, insulin, glucose, and homeostatic model assessment-insulin resistance) in breast cancer risk as a mediator or influencing factor. RESULTS Seven SNPs in IGF-I and INS genes were associated with breast cancer risk. These associations differed between non-obese/active and obese/inactive women and between exogenous E non-users and users. The mediation effects of IGF-I/IR traits on the relationship between these SNPs and cancer differed between strata, but only roughly 35% of the cancer risk due to the SNPs was mediated by traits. Similarly, carriers of 20 SNPs in PIK3R1, AKT1/2, and MAPK1 genes (signaling pathways-genetic variants) had different associations with breast cancer between strata, and the proportion of the SNP-cancer relationship explained by traits varied 45-50% between the strata. CONCLUSIONS Our findings suggest that IGF-I/IR genetic variants interact with obesity and lifestyle factors, altering cancer risk partially through pathways other than IGF-I/IR traits. Unraveling gene-phenotype-lifestyle interactions will provide data on potential genetic targets in clinical trials for cancer prevention and intervention strategies to reduce breast cancer risk.
Collapse
|
156
|
Ferreira ECS, Bortolin RH, Freire-Neto FP, Souza KSC, Bezerra JF, Ururahy MAG, Ramos AMO, Himelfarb ST, Abreu BJ, Didone TVN, Pedrosa LFC, Medeiros AC, Doi SQ, Brandão-Neto J, Hirata RDC, Rezende LA, Almeida MG, Hirata MH, Rezende AA. Zinc supplementation reduces RANKL/OPG ratio and prevents bone architecture alterations in ovariectomized and type 1 diabetic rats. Nutr Res 2017; 40:48-56. [PMID: 28473060 DOI: 10.1016/j.nutres.2017.03.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/16/2017] [Accepted: 03/09/2017] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes mellitus (T1DM) and estrogen deficiency are associated with several alterations in bone turnover. Zinc (Zn) is required for growth, development, and overall health. Zinc has been used in complementary therapy against bone loss in several diseases. We hypothesized that Zn supplementation represents a potential therapy against severe bone loss induced by the combined effect of estrogen deficiency and T1DM. We evaluated the protective effect of Zn against bone alterations in a chronic model of these disorders. Female Wistar rats were ramdomized into 3 groups (5 rats each): control, OVX/T1DM (ovariectomized rats with streptozotocin-induced T1DM), and OVX/T1DM+Zn (OVX/T1DM plus daily Zn supplementation). Serum biochemical, bone histomorphometric, and molecular analyses were performed. Histomorphometric parameters were similar between the control and OVX/T1DM+Zn groups, suggesting that Zn prevents bone architecture alterations. In contrast, the OVX/T1DM group showed significantly lower trabecular width and bone area as well as greater trabecular separation than the control. The OVX/T1DM and OVX/T1DM+Zn groups had significantly higher serum alkaline phosphatase activity than the control. The supplemented group had higher levels of serum-ionized calcium and phosphorus than the nonsupplemented group. The RANKL/OPG ratio was similar between the control and OVX/T1DM+Zn groups, whereas it was higher in the OVX/T1DM group. In conclusion, Zn supplementation prevents bone alteration in chronic OVX/T1DM rats, as demonstrated by the reduced RANKL/OPG ratio and preservation of bone architecture. The findings may represent a novel therapeutic approach to preventing OVX/T1DM-induced bone alterations.
Collapse
Affiliation(s)
- Elaine C S Ferreira
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Raul H Bortolin
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Francisco P Freire-Neto
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Karla S C Souza
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - João F Bezerra
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Marcela A G Ururahy
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Ana M O Ramos
- Department of Clinical Pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Silvia T Himelfarb
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Bento J Abreu
- Department of Morphology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Thiago V N Didone
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Lucia F C Pedrosa
- Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Aldo C Medeiros
- Department of Clinical Medicine, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Sonia Q Doi
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - José Brandão-Neto
- Department of Clinical Medicine, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Rosário D C Hirata
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Luciana A Rezende
- Department of Chemistry, University of Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| | - Maria G Almeida
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Mario H Hirata
- School of Pharmaceutical Science, University of São Paulo, São Paulo, Brazil
| | - Adriana A Rezende
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
157
|
Oh H, Coburn SB, Matthews CE, Falk RT, LeBlanc ES, Wactawski-Wende J, Sampson J, Pfeiffer RM, Brinton LA, Wentzensen N, Anderson GL, Manson JE, Chen C, Zaslavsky O, Xu X, Trabert B. Anthropometric measures and serum estrogen metabolism in postmenopausal women: the Women's Health Initiative Observational Study. Breast Cancer Res 2017; 19:28. [PMID: 28284224 PMCID: PMC5346241 DOI: 10.1186/s13058-017-0810-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 01/28/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Several anthropometric measures have been associated with hormone-related cancers. However, it is unknown whether estrogen metabolism plays an important role in these relationships. We examined whether measured current body mass index (BMI), waist-to-hip ratio (WHR), height, and self-reported BMI at age 18 years were associated with serum estrogens/estrogen metabolites using baseline, cross-sectional data from 1835 postmenopausal women enrolled in the Women's Health Initiative Observational Study. METHODS Fifteen estrogens/estrogen metabolites were quantified using liquid chromatography-tandem mass spectrometry. Geometric means (GMs) of estrogens/estrogen metabolites (in picomoles per liter) were estimated using inverse probability weighted linear regression, adjusting for potential confounders and stratified on menopausal hormone therapy (MHT) use. RESULTS Among never or former MHT users, current BMI (≥30 vs. <25 kg/m2) was positively associated with parent estrogens (multivariable adjusted GM 432 vs. 239 pmol/L for estrone, 74 vs. 46 pmol/L for estradiol; p-trend < 0.001 for both) and all of the 2-, 4-, and 16-pathway estrogen metabolites evaluated (all p-trend ≤ 0.02). After additional adjustment for estradiol, unconjugated methylated 2-catechols were inversely associated (e.g., 2-methoxyestrone multivariable GM 9.3 vs. 12.0 pmol/L; p-trend < 0.001). Among current MHT users, current BMI was not associated with parent estrogens but was inversely associated with methylated catechols (e.g., 2-methoxyestrone multivariable GM 216 vs. 280 pmol/L; p-trend = 0.008). Similar patterns of association were found with WHR; however, the associations were not independent of BMI. Height and BMI at age 18 years were not associated with postmenopausal estrogens/estrogen metabolite levels. CONCLUSIONS Our data suggest that postmenopausal BMI is associated with increased circulating levels of parent estrogens and reduced methylation of catechol estrogen metabolites, the estrogen metabolism patterns that have previously been associated with higher breast cancer risk.
Collapse
Affiliation(s)
- Hannah Oh
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA.
| | - Sally B Coburn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Charles E Matthews
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Erin S LeBlanc
- Center for Health Research, Kaiser Permanente NW, Portland, OR, USA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY, USA
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| | - Garnet L Anderson
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Oleg Zaslavsky
- School of Nursing, University of Washington, Seattle, WA, USA
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E332, Bethesda, MD, 20892, USA
| |
Collapse
|
158
|
Ghavami S, Lahouti F. Abnormality Detection in Correlated Gaussian Molecular Nano-Networks: Design and Analysis. IEEE Trans Nanobioscience 2017; 16:189-202. [PMID: 28278478 DOI: 10.1109/tnb.2017.2659678] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A nano-abnormality detection scheme (NADS) in molecular nano-networks is studied. This is motivated by the fact that early detection of diseases such as cancer plays a crucial role in their successful treatment. The proposed NADS is in fact a two-tier network of sensor nano-machines (SNMs) in the first tier and a data-gathering node (DGN) at the sink. The SNMs detect the presence of competitor cells (abnormality) by variations in input and/or parameters of a nano-communications channel. The noise of SNMs as their nature suggest is considered correlated in time and space and herein assumed additive Gaussian. In the second step, the SNMs transmit micro-scale messages over a noisy micro-communications channel (MCC) to the DGN, where a decision is made upon fusing the received signals. We find an optimum design of detectors for each of the NADS tiers based on the end-to-end NADS performance. The detection performance of each SNM is analyzed by setting up a generalized likelihood ratio test. Next, taking into account the effect of the MCC, the overall performance of the NADS is analyzed in terms of probabilities of misdetection and false alarm. In addition, computationally efficient expressions to quantify the NADS performance are derived by providing, respectively, an approximation and an upper bound for the probabilities of misdetection and false alarm. This in turn enables formulating a design problem, where the optimized concentration of SNMs in a sample is obtained for a high probability of detection and a limited probability of false alarm. The results indicate that otherwise ignoring the spatial and temporal correlation of SNM noise in the analysis, leads to an NADS that noticeably underperforms in operations.The results indicate how effective fusion of the noisy observations collected from a number of SNMs with limited capabilities could provide an acceptable detection performance.
Collapse
|
159
|
Jankowska MM, Natarajan L, Godbole S, Meseck K, Sears DD, Patterson RE, Kerr J. Kernel Density Estimation as a Measure of Environmental Exposure Related to Insulin Resistance in Breast Cancer Survivors. Cancer Epidemiol Biomarkers Prev 2017; 26:1078-1084. [PMID: 28258052 DOI: 10.1158/1055-9965.epi-16-0927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/11/2017] [Accepted: 02/20/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Environmental factors may influence breast cancer; however, most studies have measured environmental exposure in neighborhoods around home residences (static exposure). We hypothesize that tracking environmental exposures over time and space (dynamic exposure) is key to assessing total exposure. This study compares breast cancer survivors' exposure to walkable and recreation-promoting environments using dynamic Global Positioning System (GPS) and static home-based measures of exposure in relation to insulin resistance.Methods: GPS data from 249 breast cancer survivors living in San Diego County were collected for one week along with fasting blood draw. Exposure to recreation spaces and walkability was measured for each woman's home address within an 800 m buffer (static), and using a kernel density weight of GPS tracks (dynamic). Participants' exposure estimates were related to insulin resistance (using the homeostatic model assessment of insulin resistance, HOMA-IR) controlled by age and body mass index (BMI) in linear regression models.Results: The dynamic measurement method resulted in greater variability in built environment exposure values than did the static method. Regression results showed no association between HOMA-IR and home-based, static measures of walkability and recreation area exposure. GPS-based dynamic measures of both walkability and recreation area were significantly associated with lower HOMA-IR (P < 0.05).Conclusions: Dynamic exposure measurements may provide important evidence for community- and individual-level interventions that can address cancer risk inequities arising from environments wherein breast cancer survivors live and engage.Impact: This is the first study to compare associations of dynamic versus static built environment exposure measures with insulin outcomes in breast cancer survivors. Cancer Epidemiol Biomarkers Prev; 26(7); 1078-84. ©2017 AACR.
Collapse
Affiliation(s)
- Marta M Jankowska
- Qualcomm Institute, California Institute for Telecommunications and Information Technology, University of California, San Diego, California.
| | - Loki Natarajan
- Department of Family Medicine and Public Health, University of California, San Diego, California.,Moores UC San Diego Cancer Center, University of California San Diego, California
| | - Suneeta Godbole
- Department of Family Medicine and Public Health, University of California, San Diego, California
| | - Kristin Meseck
- Department of Family Medicine and Public Health, University of California, San Diego, California
| | - Dorothy D Sears
- Department of Family Medicine and Public Health, University of California, San Diego, California.,Department of Medicine, University of California, San Diego, California
| | - Ruth E Patterson
- Department of Family Medicine and Public Health, University of California, San Diego, California.,Moores UC San Diego Cancer Center, University of California San Diego, California
| | - Jacqueline Kerr
- Department of Family Medicine and Public Health, University of California, San Diego, California.,Moores UC San Diego Cancer Center, University of California San Diego, California
| |
Collapse
|
160
|
Kang DW, Lee J, Suh SH, Ligibel J, Courneya KS, Jeon JY. Effects of Exercise on Insulin, IGF Axis, Adipocytokines, and Inflammatory Markers in Breast Cancer Survivors: A Systematic Review and Meta-analysis. Cancer Epidemiol Biomarkers Prev 2017; 26:355-365. [PMID: 27742668 DOI: 10.1158/1055-9965.epi-16-0602] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/04/2016] [Accepted: 10/04/2016] [Indexed: 12/11/2022] Open
Abstract
Background: Insulin, IGF axis, adiponectin, and inflammatory markers are associated with breast cancer. Given that physical activity improves prognosis of breast cancer survivors, we investigated the effects of exercise on these markers as potential mediators between physical activity and breast cancer.Methods: PubMed, EMBASE, CENTRAL, CINAHL, and SportDiscus were searched up to December 3, 2015, to identify randomized controlled trials (RCT) that investigated the effect of exercise on insulin, IGF axis, and cytokines in breast cancer survivors. Weighted mean difference (WMD) was calculated using either fixed- or random-effects models on the basis of the heterogeneity of the studies.Results: A total of 18 studies involving 681 breast cancer survivors were included, and these numbers were reduced for individual biomarker analyses. We found that exercise significantly reduced fasting insulin levels [WMD, -3.46 μU/mL; 95% confidence interval (CI), -5.97 to -0.95; P = 0.007]. Furthermore, potentially meaningful but statistically nonsignificant changes were observed in insulin resistance (WMD, -0.73; 95% CI, -0.54 to 0.13; P = 0.23), adiponectin (WMD, 1.17 μg/mL; 95% CI, -0.87 to 3.20; P = 0.26), and C-reactive protein (WMD, -1.10 mg/L; 95% CI, -2.39 to 0.20; P = 0.10). Subgroup analyses showed that fasting insulin levels were significantly more impacted in studies in which intervention participants experienced a weight reduction (WMD, -7.10 μU/mL; 95% CI, -10.31 to -3.90; P < 0.001).Conclusions: Exercise reduces fasting insulin levels in breast cancer survivors. This may be due to exercise-induced reductions in body weight.Impact: Practitioners and clinicians may better help breast cancer prognosis be improved through exercise, anticipating physiological effects on cancer. Cancer Epidemiol Biomarkers Prev; 26(3); 355-65. ©2016 AACR.
Collapse
Affiliation(s)
- Dong-Woo Kang
- Faculty of Physical Education and Recreation, University of Alberta, Edmonton, Alberta, Canada
| | - Junga Lee
- Department of Sport and Leisure Studies, Yonsei University, Seoul, Korea.,Exercise Medicine Center for Diabetes and Cancer Patients, Institute of Convergence of Science (ICONS), Yonsei University, Seoul, Korea
| | - Sang-Hoon Suh
- Department of Physical Education, Yonsei University, Seoul, Korea
| | - Jennifer Ligibel
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kerry S Courneya
- Faculty of Physical Education and Recreation, University of Alberta, Edmonton, Alberta, Canada
| | - Justin Y Jeon
- Department of Sport and Leisure Studies, Yonsei University, Seoul, Korea. .,Exercise Medicine Center for Diabetes and Cancer Patients, Institute of Convergence of Science (ICONS), Yonsei University, Seoul, Korea.,Cancer Prevention Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
161
|
Dethlefsen C, Pedersen KS, Hojman P. Every exercise bout matters: linking systemic exercise responses to breast cancer control. Breast Cancer Res Treat 2017; 162:399-408. [PMID: 28138894 DOI: 10.1007/s10549-017-4129-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 12/27/2022]
Abstract
Cumulative epidemiological evidence shows that regular exercise lowers the risk of developing breast cancer and decreases the risk of disease recurrence. The causality underlying this relation has not been fully established, and the exercise recommendations for breast cancer patients follow the general physical activity guidelines, prescribing 150 min of exercise per week. Thus, elucidations of the causal mechanisms are important to prescribe and implement the most optimal training regimen in breast cancer prevention and treatment. The prevailing hypothesis on the positive association within exercise oncology has focused on lowering of the basal systemic levels of cancer risk factors with exercise training. However, another rather overlooked systemic exercise response is the marked acute increases in several potential anti-cancer components during each acute exercise bout. Here, we review the evidence of the exercise-mediated changes in systemic components with the ability to influence breast cancer progression. In the first part, we focus on systemic risk factors for breast cancer, i.e., sex hormones, insulin, and inflammatory markers, and their adaptation to long-term training. In the second part, we describe the systemic factors induced acutely during exercise, including catecholamines and myokines. In conclusion, we propose that the transient increases in exercise factors during acute exercise appear to be mediating the positive effect of regular exercise on breast cancer progression.
Collapse
Affiliation(s)
- Christine Dethlefsen
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Rigshospitalet, Faculty of Health Science, Copenhagen University Hospital, 7641, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Katrine Seide Pedersen
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Rigshospitalet, Faculty of Health Science, Copenhagen University Hospital, 7641, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Pernille Hojman
- Centre of Inflammation and Metabolism (CIM) and Centre for Physical Activity Research (CFAS), Rigshospitalet, Faculty of Health Science, Copenhagen University Hospital, 7641, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
162
|
The Mechanisms and Effects of Physical Activity on Breast Cancer. Clin Breast Cancer 2017; 17:272-278. [PMID: 28233686 DOI: 10.1016/j.clbc.2017.01.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/13/2017] [Indexed: 12/15/2022]
Abstract
Breast cancer is the most common cancer in women. An extensive part of this health problem can be prevented by an active lifestyle. Physical activity can reduce the risk of breast cancer, reduce the rate of recurrence, and increase the survival rate of patients with breast cancer. The aim of this review was to summarize our current knowledge regarding the effects of physical activity on breast cancer risk, recurrence, and survival. Furthermore, we investigated 5 possible underlying mechanisms through which physical activity has an influence on breast cancer (ie, a reduction of sex hormones, metabolic hormones, adipokines and oxidative stress, and an improvement of the immune function). In this review, we give a complete overview of this subject.
Collapse
|
163
|
Li G, Xu Z, Zhuang A, Chang S, Hou L, Chen Y, Polat M, Wu D. Magnetic Resonance Spectroscopy-Detected Change in Marrow Adiposity Is Strongly Correlated to Postmenopausal Breast Cancer Risk. Clin Breast Cancer 2017; 17:239-244. [PMID: 28188108 DOI: 10.1016/j.clbc.2017.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/30/2016] [Accepted: 01/09/2017] [Indexed: 01/01/2023]
Abstract
PURPOSE To determine whether marrow fat fraction (FF) is correlated with postmenopausal breast cancer risk and clinicopathological characteristics of breast cancer. METHODS Fifty-six patients with newly diagnosed and histologically confirmed postmenopausal breast cancer and 56 healthy controls underwent serologic test and magnetic resonance spectroscopy-based FF measurements. Data were analyzed by logistic multivariate regression models to determine the independent predictors of breast cancer risk and clinicopathological characters of breast cancer. RESULTS Patients with breast cancer had higher FF than that of the controls. Marrow FF showed positive association with serum leptin levels (r = 0.607, P < .001) in the cases, but no relationship was found in the controls. In the univariate analysis, both levels of leptin and marrow FF were significantly associated with breast cancer risk and clinicopathological characteristics of breast cancer. In the multivariable model with adjustment for established breast cancer risk factors, serum leptin was a significant predictor of breast cancer risk (OR 1.746; 95% CI, 1.226-2.556) and clinicopathological characteristics of breast cancer including TNM, tumor size, lymph node status, and histological grade (OR 1.461-1.695); but when marrow FF was additionally added to the regression model, marrow FF but not leptin levels was observed to be an independent risk factor for breast cancer risk (OR 1.940; 95% CI, 1.306-2.910) and clinicopathological characteristics of breast cancer (OR 1.770-1.903). CONCLUSION Marrow adiposity is a predictor of postmenopausal breast cancer risk and clinicopathological characteristics of breast cancer.
Collapse
Affiliation(s)
- Guanwu Li
- Department of Radiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zheng Xu
- Xinzhuang Community Health Center, Shanghai, China
| | - Alex Zhuang
- Department of Radiology, Wayne State University, Detroit, MI
| | - Shixin Chang
- Department of Radiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingmi Hou
- Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Sichuan, China
| | - Yongsheng Chen
- Department of Radiology, Wayne State University, Detroit, MI
| | - Maki Polat
- School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - Dongmei Wu
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, China
| |
Collapse
|
164
|
Engin A. Obesity-associated Breast Cancer: Analysis of risk factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:571-606. [PMID: 28585217 DOI: 10.1007/978-3-319-48382-5_25] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Furthermore, obese women are at higher risk of all-cause and breast cancer specific mortality when compared to non-obese women with breast cancer. In this context, increased levels of estrogens due to excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, hyperactivation of insulin-like growth factors (IGFs) pathways, adipocyte-derived adipokines, hypercholesterolemia and excessive oxidative stress contribute to the development of breast cancer in obese women. While higher breast cancer risk with hormone replacement therapy is particularly evident among lean women, in postmenopausal women who are not taking exogenous hormones, general obesity is a significant predictor for breast cancer. Moreover, increased plasma cholesterol leads to accelerated tumor formation and exacerbates their aggressiveness. In contrast to postmenopausal women, premenopausal women with high BMI are inversely associated with breast cancer risk. Nevertheless, life-style of women for breast cancer risk is regulated by avoiding the overweight and a high-fat diet. Estrogen-plus-progestin hormone therapy users for more than 5 years have elevated risks of both invasive ductal and lobular breast cancer. Additionally, these cases are more commonly node-positive and have a higher cancer-related mortality. Collectively, in this chapter, the impacts of obesity-related estrogen, cholesterol, saturated fatty acid, leptin and adiponectin concentrations, aromatase activity, leptin and insulin resistance on breast cancer patients are evaluated. Obesity-related prognostic factors of breast cancer also are discussed at molecular basis.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey. .,, Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
165
|
Costa-Silva DR, Barros-Oliveira MDAC, Borges RS, Tavares CB, Borges US, Alves-Ribeiro FA, Silva VC, Silva BBDA. Insulin-like Growth Factor 1 gene polymorphism and breast cancer risk. AN ACAD BRAS CIENC 2016; 88:2349-2356. [PMID: 27925035 DOI: 10.1590/0001-3765201620160169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/23/2016] [Indexed: 11/22/2022] Open
Abstract
Insulin-like Growth Factor-1 (IGF-1) gene polymorphism has been associated with an increased risk for breast cancer. IGF-1 is a key regulator of proliferation, cell differentiation and apoptosis. It has important mitogenic and anti-apoptotic activities in normal cells and in breast cancer cells, acting synergistically with estrogen to increase neoplastic cell proliferation. This review aims to present the recent finds of IGF-1 gene polymorphism and its relationship with the risk of breast cancer through following the polymorphic dinucleotide repeat cytosine-adenine (CA) and single nucleotide polymorphisms (SNPs) by searching in the PubMed database publications focused studies published from 2010 to 2015 related to IGF-1 gene polymorphism and breast cancer risk. A growing number of studies support an association between IGF-1 gene polymorphism and breast cancer risk with conflicting results, nevertheless elucidation of the patterns of IGF-1 gene expression may permit characterization of women at high-risk for breast cancer, as well as the development of strategies for early diagnosis and efficient treatment against the disease.
Collapse
Affiliation(s)
- Danylo R Costa-Silva
- Programa de Pós-Graduação em Ciências e Saúde, Universidade Federal do Piauí, Av. Frei Serafim, 2280, Centro, 64001-020 Teresina, PI, Brazil
| | - Maria DA Conceição Barros-Oliveira
- Programa de Pós-Graduação em Ciências e Saúde, Universidade Federal do Piauí, Av. Frei Serafim, 2280, Centro, 64001-020 Teresina, PI, Brazil
| | - Rafael S Borges
- Setor de Mastologia do Hospital Getulio Vargas, Universidade Federal do Piauí, Av. Frei Serafim, 2352, Centro, 64001-020 Teresina, PI, Brazil
| | - Cléciton B Tavares
- Programa de Pós-Graduação em Ciências e Saúde, Universidade Federal do Piauí, Av. Frei Serafim, 2280, Centro, 64001-020 Teresina, PI, Brazil
| | - Umbelina S Borges
- Programa de Pós-Graduação em Ciências e Saúde, Universidade Federal do Piauí, Av. Frei Serafim, 2280, Centro, 64001-020 Teresina, PI, Brazil
| | - Francisco A Alves-Ribeiro
- Setor de Mastologia do Hospital Getulio Vargas, Universidade Federal do Piauí, Av. Frei Serafim, 2352, Centro, 64001-020 Teresina, PI, Brazil
| | - Vladimir C Silva
- Serviço de Biologia Molecular, Hospital Natan Portella, Universidade Federal do Piauí, Rua Governador Raimundo Artur de Vasconcelos, 151, Centro/Sul, 64001-450 Teresina, PI, Brazil
| | - Benedito B DA Silva
- Programa de Pós-Graduação em Ciências e Saúde, Universidade Federal do Piauí, Av. Frei Serafim, 2280, Centro, 64001-020 Teresina, PI, Brazil.,Setor de Mastologia do Hospital Getulio Vargas, Universidade Federal do Piauí, Av. Frei Serafim, 2352, Centro, 64001-020 Teresina, PI, Brazil
| |
Collapse
|
166
|
Hunt KK, Euhus DM, Boughey JC, Chagpar AB, Feldman SM, Hansen NM, Kulkarni SA, McCready DR, Mamounas EP, Wilke LG, Van Zee KJ, Morrow M. Society of Surgical Oncology Breast Disease Working Group Statement on Prophylactic (Risk-Reducing) Mastectomy. Ann Surg Oncol 2016; 24:375-397. [PMID: 27933411 DOI: 10.1245/s10434-016-5688-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Indexed: 12/15/2022]
Abstract
Over the past several years, there has been an increasing rate of bilateral prophylactic mastectomy (BPM) and contralateral prophylactic mastectomy (CPM) surgeries. Since publication of the 2007 SSO position statement on the use of risk-reducing mastectomy, there have been significant advances in the understanding of breast cancer biology and treatment. The purpose of this manuscript is to review the current literature as a resource to facilitate a shared and informed decision-making process regarding the use of risk-reducing mastectomy.
Collapse
Affiliation(s)
- Kelly K Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | - Monica Morrow
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
167
|
Murphy N, Falk RT, Messinger DB, Pollak M, Xue X, Lin J, Sgueglia R, Strickler HD, Gaudet MM, Gunter MJ. Influence of Fasting Status and Sample Preparation on Metabolic Biomarker Measurements in Postmenopausal Women. PLoS One 2016; 11:e0167832. [PMID: 27930694 PMCID: PMC5145182 DOI: 10.1371/journal.pone.0167832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/21/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Epidemiologic data linking metabolic markers-such as insulin, insulin-like growth factors (IGFs)-and adipose tissue-derived factors with cancer are inconsistent. Between-study differences in blood collection protocols, in particular participant's fasting status, may influence measurements. METHODS We investigated the impact of fasting status and blood sample processing time on components of the insulin/IGF axis and in adipokines in a controlled feeding study of 45 healthy postmenopausal-women aged 50-75 years. Fasting blood samples were drawn (T0), after which subjects ate a standardized breakfast; subsequent blood draws were made at 1 hour (T1), 3 hours (T3), and 6 hours (T6) after breakfast. Serum samples were assayed for insulin, C-peptide, total- and free-IGF-I, IGF-binding protein [BP]-1 and -3, total and high molecular weight (HMW)-adiponectin, retinol binding protein-4, plasminogen activator inhibitor (PAI)-1, and resistin. RESULTS Insulin and C-peptide levels followed similar postprandial trajectories; intra-class correlation coefficients [ICC] for insulin = 0.75, (95%CI:0.64-0.97) and C-peptide (ICC = 0.66, 95%CI:0.54-0.77) were similarly correlated in fasting (Spearman correlation, r = 0.78, 95%CI:0.64-0.88) and postprandial states (T1, r = 0.77 (95%CI: 0.62-0.87); T3,r = 0.78 (95%CI: 0.63-0.87); T6,r = 0.77 (95%CI: 0.61-0.87)). Free-IGF-I and IGFBP-1 levels were also affected by fasting status, whereas total-IGF-I and IGFBP-3 levels remained unchanged. Levels of adipokines were largely insensitive to fasting status and blood sample processing delays. CONCLUSION Several components of the insulin/IGF axis were significantly impacted by fasting state and in particular, C-peptide levels were substantially altered postprandially and in a similar manner to insulin.
Collapse
Affiliation(s)
- Neil Murphy
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- International Agency for Research on Cancer, Lyon, France
| | - Roni T. Falk
- Division of Cancer Epidemiology and Genetics, Hormonal and Reproductive Epidemiology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Diana B. Messinger
- The Rockefeller University Hospital, New York, New York, United States of America
| | - Michael Pollak
- Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Juan Lin
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Robin Sgueglia
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Howard D. Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Mia M. Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, United States of America
| | - Marc J. Gunter
- International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
168
|
Buschard K, Thomassen K, Lynge E, Vejborg I, Tjønneland A, von Euler-Chelpin M, Andersen ZJ. Diabetes, diabetes treatment, and mammographic density in Danish Diet, Cancer, and Health cohort. Cancer Causes Control 2016; 28:13-21. [PMID: 27832382 PMCID: PMC5219016 DOI: 10.1007/s10552-016-0829-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/01/2016] [Indexed: 11/04/2022]
Abstract
Purpose We examined whether diabetes and diabetes treatment are associated with MD in a cohort study of Danish women above age of 50 years. Methods Study cohort consisted of 5,644 women (4,500 postmenopausal) who participated in the Danish Diet, Cancer, and Health cohort (1993–1997) and subsequently attended mammographic screening in Copenhagen (1993–2001). We used MD assessed at the first screening after the cohort entry, defined as mixed/dense or fatty. Diabetes diagnoses and diabetes treatments (diet, insulin, or oral antidiabetic agents) were self-reported at the time of recruitment (1993–1997). The association between MD and diabetes was analyzed by logistic regression adjusted for potential confounders. Effect modification by menopausal status and body mass index (BMI) was performed by introducing an interaction term into the model and tested by Wald test. Results Of 5,644 women with mean age of 56 years, 137 (2.4%) had diabetes and 3,180 (56.3%) had mixed/dense breasts. Having diabetes was significantly inversely associated with having mixed/dense breasts, in both, the crude model (odds ratio; 95% confidence interval: 0.33; 0.23–0.48), and after adjustment for adiposity and other risk factors (0.61; 0.40–0.92). Similar inverse associations were observed for 44 women who controlled diabetes by diet only and did not receive any medication (0.56; 0.27–1.14), and 62 who took oral antidiabetic agents only for diabetes (0.59; 0.32–1.09), while women taking insulin had increased odds of mixed/dense breasts (2.08; 0.68–6.35). There was no effect modification of these associations by menopausal status or BMI. Conclusions Having diabetes controlled by diet or oral antidiabetic agents is associated with a decrease in MD, whereas taking insulin is associated with an increase in MD.
Collapse
Affiliation(s)
- Karsten Buschard
- Bartholin Institute, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Katrin Thomassen
- Department of Public Health, Center for Epidemiology and Screening, University of Copenhagen, Øster Farimagsgade 5, 1014, Copenhagen, Denmark
| | - Elsebeth Lynge
- Department of Public Health, Center for Epidemiology and Screening, University of Copenhagen, Øster Farimagsgade 5, 1014, Copenhagen, Denmark
| | - Ilse Vejborg
- Department of Radiology, Diagnostic Imaging Centre, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Anne Tjønneland
- Danish Cancer Research Center, Danish Cancer Society, Strandboulevarden 49, Copenhagen, Denmark
| | - My von Euler-Chelpin
- Department of Public Health, Center for Epidemiology and Screening, University of Copenhagen, Øster Farimagsgade 5, 1014, Copenhagen, Denmark
| | - Zorana Jovanovic Andersen
- Department of Public Health, Center for Epidemiology and Screening, University of Copenhagen, Øster Farimagsgade 5, 1014, Copenhagen, Denmark.
| |
Collapse
|
169
|
Hopkins BD, Goncalves MD, Cantley LC. Obesity and Cancer Mechanisms: Cancer Metabolism. J Clin Oncol 2016; 34:4277-4283. [PMID: 27903152 DOI: 10.1200/jco.2016.67.9712] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for cancer development and is associated with poor prognosis in multiple tumor types. The positive energy balance linked with obesity induces a variety of systemic changes including altered levels of insulin, insulin-like growth factor-1, leptin, adiponectin, steroid hormones, and cytokines. Each of these factors alters the nutritional milieu and has the potential to create an environment that favors tumor initiation and progression. Although the complete ramifications of obesity as it relates to cancer are still unclear, there is convincing evidence that reducing the magnitude of the systemic hormonal and inflammatory changes has significant clinical benefits. This review will examine the changes that occur in the obese state and review the biologic mechanisms that connect these changes to increased cancer risk. Understanding the metabolic changes that occur in obese individuals may also help to elucidate more effective treatment options for these patients when they develop cancer. Moving forward, targeted clinical trials examining the effects of behavioral modifications such as reduced carbohydrate intake, caloric restriction, structured exercise, and/or pharmacologic interventions such as the use of metformin, in obese populations may help to reduce their cancer risk.
Collapse
Affiliation(s)
- Benjamin D Hopkins
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Marcus D Goncalves
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Lewis C Cantley
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| |
Collapse
|
170
|
Hartman TJ, Gapstur SM, Gaudet MM, Shah R, Flanders WD, Wang Y, McCullough ML. Dietary Energy Density and Postmenopausal Breast Cancer Incidence in the Cancer Prevention Study II Nutrition Cohort. J Nutr 2016; 146:2045-2050. [PMID: 27629577 DOI: 10.3945/jn.116.234344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/04/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Dietary energy density (ED) is a measure of diet quality that estimates the amount of energy per unit of food (kilocalories per gram) consumed. Low-ED diets are generally high in fiber and fruits and vegetables and low in fat. Dietary ED has been positively associated with body mass index (BMI) and other risk factors for postmenopausal breast cancer. OBJECTIVE We evaluated the associations of total dietary ED and energy-dense (high-ED) foods with postmenopausal breast cancer incidence. METHODS Analyses included 56,795 postmenopausal women from the Cancer Prevention Study II Nutrition Cohort with no previous history of breast or other cancers and who provided information on diet, lifestyle, and medical history in 1999. Multivariable-adjusted breast cancer incidence rate ratios (RRs and 95% CIs) were estimated for quintiles of total dietary ED and for the consumption of high-ED foods in Cox proportional hazards regression models. RESULTS During a median follow-up of 11.7 y, 2509 invasive breast cancer cases were identified, including 1857 estrogen receptor-positive and 277 estrogen receptor-negative tumors. Median dietary ED was 1.5 kcal/g (IQR: 1.3-1.7 kcal/g). After adjusting for age, race, education, reproductive characteristics, and family history, high compared with low dietary ED was associated with a statistically significantly higher risk of breast cancer (RR for fifth quintile compared with first quintile: 1.20; 95% CI: 1.05, 1.36; P-trend = 0.03). The association between the amount of high-ED foods consumed and breast cancer risk was not statistically significant. We observed no differences by estrogen receptor status or effect modification by BMI, age, or physical activity. CONCLUSION These results suggest a modest positive association between total dietary ED and risk of postmenopausal breast cancer.
Collapse
Affiliation(s)
- Terryl J Hartman
- Department of Epidemiology, Rollins School of Public Health, Winship Cancer Institute, Emory University, Atlanta, GA; and
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Mia M Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Roma Shah
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - W Dana Flanders
- Department of Epidemiology, Rollins School of Public Health, Winship Cancer Institute, Emory University, Atlanta, GA; and
| | - Ying Wang
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | | |
Collapse
|
171
|
Sun W, Lu J, Wu S, Bi Y, Mu Y, Zhao J, Liu C, Chen L, Shi L, Li Q, Yang T, Yan L, Wan Q, Liu Y, Wang G, Luo Z, Tang X, Chen G, Huo Y, Gao Z, Su Q, Ye Z, Wang Y, Qin G, Deng H, Yu X, Shen F, Chen L, Zhao L, Wang T, Sun J, Xu M, Xu Y, Chen Y, Dai M, Zhang J, Zhang D, Lai S, Li D, Ning G, Wang W. Association of insulin resistance with breast, ovarian, endometrial and cervical cancers in non-diabetic women. Am J Cancer Res 2016; 6:2334-2344. [PMID: 27822422 PMCID: PMC5088296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 06/15/2016] [Indexed: 06/06/2023] Open
Abstract
Hyperinsulinemia and insulin resistance were reported to play a crucial role in diabetes-cancer relationship. This study aimed to explore the associations between insulin resistance and several female cancers in a non-diabetic population. This cross-sectional study was conducted in 121,230 middle-aged and elderly non-diabetic women. Cancer diagnosis was self-reported and further validated by medical records. Insulin resistance was defined as homeostasis model assessment of insulin resistance (HOMA-IR) ≥ 2.50. The prevalence of both premenopausal and postmenopausal breast cancer, postmenopausal ovarian cancer and premenopausal endometrial cancer were higher in insulin-resistant participants than in insulin-sensitive participants (premenopausal breast cancer, 0.45 vs 0.28%; postmenopausal breast cancer, 0.86 vs 0.63%; postmenopausal ovarian cancer, 0.17 vs 0.09%; premenopausal endometrial cancer, 0.43 vs 0.25%, respectively, all P < 0.05). Individuals with insulin resistance had higher odds ratio (OR) of breast cancer, both premenopausal and postmenopausal (OR 1.98, 95% confidence interval (CI) 1.19-3.32; OR 1.29, 95% CI 1.01-1.63), postmenopausal ovarian cancer (OR 2.17, 95% CI 1.10-3.40) as well as total endometrial cancer (OR 1.47, 95% CI 1.02-2.12). Subgroup analysis revealed that the possitive association between insulin resistance and risk of prevalent breast cancer was observed in popualtion with younger age, overweight or obesity, higher education and impaired glucose tolerance (IGR). No relationships were observed for the risk of prevalent cervical cancers with insulin resistance. Non-diabetic women with insulin resistance had higher risk of prevalent breast, ovarian and endomatrial cancer, which suggests special attentions to these female cancer screening and prevention.
Collapse
Affiliation(s)
- Wanwan Sun
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Jieli Lu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Shengli Wu
- Karamay Municipal People’s HospitalXinjiang 834000, China
| | - Yufang Bi
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Yiming Mu
- Chinese People’s Liberation Army General HospitalBeijing 100000, China
| | - Jiajun Zhao
- Shandong Provincial Hospital Affiliated to Shandong UniversityJinan 250000, China
| | - Chao Liu
- Jiangsu Province Hospital on Integration of Chinese and Western MedicineNanjing 210000, China
| | - Lulu Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430000, China
| | - Lixin Shi
- Affiliated Hospital of Guiyang Medical CollegeGuiyang 550000, China
| | - Qiang Li
- The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150000, China
| | - Tao Yang
- The First Affiliated Hospital with Nanjing Medical University, Jiangsu Province HospitalNanjing 210000, China
| | - Li Yan
- Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510000, China
| | - Qin Wan
- The Affiliated Hospital of Luzhou Medical CollegeLuzhou 646000, China
| | - Yan Liu
- The First Hospital of Jilin UniversityChangchun 130000, China
| | - Guixia Wang
- The First Hospital of Jilin UniversityChangchun 130000, China
| | - Zuojie Luo
- The First Affiliated Hospital of Guangxi Medical UniversityNanning 530000, China
| | - Xulei Tang
- The First Hospital of Lanzhou UniversityLanzhou 730000, China
| | - Gang Chen
- Fujian Provincial Hospital, Fujian Medical UniversityFuzhou 350000, China
| | - Yanan Huo
- Jiangxi People’s HospitalNanchang 330000, China
| | - Zhengnan Gao
- Dalian Municipal Central HospitalDalian 116000, China
| | - Qing Su
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of MedicineShanghai 200082, China
| | - Zhen Ye
- Zhejiang Provincial Center for Disease Control and PreventionZhejiang 314000, China
| | - Youmin Wang
- The First Affiliated Hospital of Anhui Medical UniversityHefei 230000, China
| | - Guijun Qin
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450000, China
| | - Huacong Deng
- The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400000, China
| | - Xuefeng Yu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430000, China
| | - Feixia Shen
- The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, China
| | - Li Chen
- Qilu Hospital of Shandong UniversityJinan 250000, China
| | - Liebin Zhao
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Tiange Wang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Jichao Sun
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
- Laboratory of Endocrine and Metabolic Diseases, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of MedicineShanghai 200030,China
| | - Min Xu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Yu Xu
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Yuhong Chen
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Meng Dai
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Jie Zhang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Di Zhang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| | - Shenghan Lai
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
- Johns Hopkins University School of MedicineBaltimore, Maryland 21201, USA
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77036, USA
| | - Guang Ning
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
- Laboratory of Endocrine and Metabolic Diseases, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of MedicineShanghai 200030,China
| | - Weiqing Wang
- National Clinical Research Center for Metabolic Diseases, Rui Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200025, China
| |
Collapse
|
172
|
Wei R, De Vivo I, Huang S, Zhu X, Risch H, Moore JH, Yu H, Garmire LX. Meta-dimensional data integration identifies critical pathways for susceptibility, tumorigenesis and progression of endometrial cancer. Oncotarget 2016; 7:55249-55263. [PMID: 27409342 PMCID: PMC5342415 DOI: 10.18632/oncotarget.10509] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 05/02/2016] [Indexed: 11/25/2022] Open
Abstract
Endometrial Cancer (EC) is one of the most common female cancers. Genome-wide association studies (GWAS) have been investigated to identify genetic polymorphisms that are predictive of EC risks. Here we utilized a meta-dimensional integrative approach to seek genetically susceptible pathways that may be associated with tumorigenesis and progression of EC. We analyzed GWAS data obtained from Connecticut Endometrial Cancer Study (CECS) and identified the top 20 EC susceptible pathways. To further verify the significance of top 20 EC susceptible pathways, we conducted pathway-level multi-omics analyses using EC exome-Seq, RNA-Seq and survival data, all based on The Cancer Genome Atlas (TCGA) samples. We measured the overall consistent rankings of these pathways in all four data types. Some well-studied pathways, such as p53 signaling and cell cycle pathways, show consistently high rankings across different analyses. Additionally, other cell signaling pathways (e.g. IGF-1/mTOR, rac-1 and IL-5 pathway), genetic information processing pathway (e.g. homologous recombination) and metabolism pathway (e.g. sphingolipid metabolism) are also highly associated with EC risks, diagnosis and prognosis. In conclusion, the meta-dimensional integration of EC cohorts has suggested some common pathways that may be associated from predisposition, tumorigenesis to progression.
Collapse
Affiliation(s)
- Runmin Wei
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, HI, USA
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Sijia Huang
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, HI, USA
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Xun Zhu
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, HI, USA
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Harvey Risch
- Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Jason H. Moore
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Herbert Yu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Lana X. Garmire
- Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at Manoa, Honolulu, HI, USA
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| |
Collapse
|
173
|
Gillessen S, Gilson C, James N, Adler A, Sydes MR, Clarke N. Repurposing Metformin as Therapy for Prostate Cancer within the STAMPEDE Trial Platform. Eur Urol 2016; 70:906-908. [PMID: 27450106 DOI: 10.1016/j.eururo.2016.07.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 07/11/2016] [Indexed: 01/18/2023]
Abstract
Metformin is a safe, well-tolerated, inexpensive treatment that can be given in addition to current standard-of-care therapies for prostate cancer. Its use might mitigate the deleterious side effects of castration and exert an additional anticancer effect. It will be incorporated in the STAMPEDE trial platform in summer 2016. This will test its true utility as a repurposed treatment for men with high-risk locally advanced or metastatic prostate cancer at first presentation.
Collapse
Affiliation(s)
- Silke Gillessen
- Department of Oncology/Haematology, Kantonsspital, St. Gallen, Switzerland
| | - Clare Gilson
- MRC Clinical Trials Unit, University College London, London, UK
| | - Nick James
- Warwick Medical School, University of Warwick, Coventry, UK; University Hospitals Birmingham NHS Foundation Trust, The Medical School, University of Birmingham, Birmingham, UK
| | - Amanda Adler
- Diabetes Service, Addenbrooke's Hospital, Cambridge, UK
| | - Matthew R Sydes
- MRC Clinical Trials Unit, University College London, London, UK
| | - Noel Clarke
- Department of Urology, The Christie and Salford Royal NHS Foundation Trusts, Manchester, UK.
| |
Collapse
|
174
|
Ferroni P, Riondino S, Laudisi A, Portarena I, Formica V, Alessandroni J, D'Alessandro R, Orlandi A, Costarelli L, Cavaliere F, Guadagni F, Roselli M. Pretreatment Insulin Levels as a Prognostic Factor for Breast Cancer Progression. Oncologist 2016; 21:1041-9. [PMID: 27388232 DOI: 10.1634/theoncologist.2015-0462] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/09/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Based on the hypothesis that impaired glucose metabolism might be associated with survival outcomes independently of overt diabetes, we sought to investigate the prognostic value of routinely used glycemic parameters in a prospective study of breast cancer (BC) patients. PATIENTS AND METHODS Fasting blood glucose, insulin and HbA1c levels, and insulin resistance (assessed by the Homeostasis Model Assessment [HOMA] index) at diagnosis were evaluated in 286 nondiabetic BC patients (249 with primary cancer, 37 with metastatic) with respect to those parameters' possible associations with clinicopathological features and survival outcomes. As a control group, 143 healthy women matched in a 2:1 ratio for age, blood lipid levels, and body mass index were also investigated. RESULTS Fasting blood glucose level (mean ± SD: 99 ± 26 vs. 85 ± 15 mg/dL), insulin level (median: 10.0 vs. 6.8 μIU/mL), and HOMA index (median: 2.2 vs. 1.4), but not HbA1c level, were significantly elevated in BC patients compared with control subjects. Receiver operating characteristics analysis showed comparable areas for blood glucose and insulin levels, and HOMA index (ranging from 0.668 to 0.671). Using a cutoff level of 13 μIU/mL, insulin had the best specificity (92%) and sensitivity (41%), was significantly associated with disease stage, and acted as a negative prognostic marker of progression-free survival (hazard ratio: 2.17; 95% confidence interval: 1.13-4.20) independently of menopausal status, disease stage, hormone receptor status, and human epidermal growth factor receptor 2 and Ki67 expression. CONCLUSION These results suggest that insulin determination might provide prognostic information in BC and support the hypothesis that lifestyle and/or pharmacological interventions targeting glucose metabolism could be considered to improve survival outcome of selected BC patients. IMPLICATIONS FOR PRACTICE Pretreatment insulin levels may represent a biomarker of adverse prognosis in nondiabetic women with breast cancer, independently of other well-established prognostic factors (i.e., stage, hormone receptors, HER2/neu, and Ki67). This finding has important implications, because it provides the rationale for lifestyle or insulin-targeting pharmacologic interventions as a means of improving breast cancer outcomes not only in early stages, but also in advanced-stage breast cancer patients with aggressive tumor phenotypes (HER2-negative hormone-resistant, or triple-negative breast cancer), in which treatments are still challenging. The possibility of using insulin as a biomarker to guide insulin-targeted interventions also should be taken into account.
Collapse
Affiliation(s)
| | - Silvia Riondino
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Anastasia Laudisi
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Ilaria Portarena
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Vincenzo Formica
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| | - Jhessica Alessandroni
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Roberta D'Alessandro
- Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | | | | | - Fiorella Guadagni
- San Raffaele Roma Open University, Rome, Italy Interinstitutional Multidisciplinary Biobank, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Mario Roselli
- Department of Systems Medicine, Medical Oncology, Tor Vergata Clinical Center, Tor Vergata University of Rome, Rome, Italy
| |
Collapse
|
175
|
Zhao H, Zhou L, Shangguan AJ, Bulun SE. Aromatase expression and regulation in breast and endometrial cancer. J Mol Endocrinol 2016; 57:R19-33. [PMID: 27067638 PMCID: PMC5519084 DOI: 10.1530/jme-15-0310] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 12/12/2022]
Abstract
Long-term exposure to excess estrogen increases the risk of breast cancer and type 1 endometrial cancer. Most of the estrogen in premenopausal women is synthesized by the ovaries, while extraovarian subcutaneous adipose tissue is the predominant tissue source of estrogen after menopause. Estrogen and its metabolites can cause hyperproliferation and neoplastic transformation of breast and endometrial cells via increased proliferation and DNA damage. Several genetically modified mouse models have been generated to help understand the physiological and pathophysiological roles of aromatase and estrogen in the normal breast and in the development of breast cancers. Aromatase, the key enzyme for estrogen production, is comprised of at least ten partially tissue-selective and alternatively used promoters. These promoters are regulated by distinct signaling pathways to control aromatase expression and estrogen formation via recruitment of various transcription factors to their cis-regulatory elements. A shift in aromatase promoter use from I.4 to I.3/II is responsible for the excess estrogen production seen in fibroblasts surrounding malignant epithelial cells in breast cancers. Targeting these distinct pathways and/or transcription factors to modify aromatase activity may lead to the development of novel therapeutic remedies that inhibit estrogen production in a tissue-specific manner.
Collapse
Affiliation(s)
- Hong Zhao
- Division of Reproductive Science in MedicineDepartment of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ling Zhou
- Division of Reproductive Science in MedicineDepartment of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Anna Junjie Shangguan
- Division of Reproductive Science in MedicineDepartment of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Serdar E Bulun
- Division of Reproductive Science in MedicineDepartment of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
176
|
The Obesity-Breast Cancer Conundrum: An Analysis of the Issues. Int J Mol Sci 2016; 17:ijms17060989. [PMID: 27338371 PMCID: PMC4926517 DOI: 10.3390/ijms17060989] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 02/07/2023] Open
Abstract
Breast cancer develops over a timeframe of 2-3 decades prior to clinical detection. Given this prolonged latency, it is somewhat unexpected from a biological perspective that obesity has no effect or reduces the risk for breast cancer in premenopausal women yet increases the risk for breast cancer in postmenopausal women. This conundrum is particularly striking in light of the generally negative effects of obesity on breast cancer outcomes, including larger tumor size at diagnosis and poorer prognosis in both pre- and postmenopausal women. This review and analysis identifies factors that may contribute to this apparent conundrum, issues that merit further investigation, and characteristics of preclinical models for breast cancer and obesity that should be considered if animal models are used to deconstruct the conundrum.
Collapse
|
177
|
Słowik A, Frączek PA, Krzemieniecki K. Body mass index and aromatase inhibitors: a step forward in individualizing therapy for breast cancer patients? Expert Rev Anticancer Ther 2016; 16:759-66. [PMID: 27196669 DOI: 10.1080/14737140.2016.1191949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Progress made in breast cancer management along with treatment-related symptoms has drawn a lot of attention from both scientists and clinicians. Establishing predictive factors for treatment response facilitate tailoring of therapy to each individual patient and leads to a reduction in unnecessary treatments. Body mass index is confirmed to be a risk factor for breast cancer development as well as for disease recurrence, which additionally negatively influence the overall survival. Due to the increased level of fatty tissue in obese and overweight patients, their total level of body aromatase is elevated. This lead to the hypothesis about a worse response to aromatase inhibitors in these groups as compared to normal weight patients, due to incomplete aromatase blockage and thus higher peripheral androgen aromatization. AREAS COVERED This review aims to summarize the data from clinical trials assessing the effect of BMI on response to AI-based therapy in the setting of breast cancer. Expert commentary: Our conclusion made on the data available to date does not exclude BMI from the list of potential predictive factors however further research in this area is warranted.
Collapse
Affiliation(s)
- Agnieszka Słowik
- a Department of Oncology , Jagiellonian University Medical College , Krakow , Poland
| | - Paulina A Frączek
- a Department of Oncology , Jagiellonian University Medical College , Krakow , Poland
| | | |
Collapse
|
178
|
Villarini M, Lanari C, Nucci D, Gianfredi V, Marzulli T, Berrino F, Borgo A, Bruno E, Gargano G, Moretti M, Villarini A. Community-based participatory research to improve life quality and clinical outcomes of patients with breast cancer (DianaWeb in Umbria pilot study). BMJ Open 2016; 6:e009707. [PMID: 27251681 PMCID: PMC4893863 DOI: 10.1136/bmjopen-2015-009707] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Breast cancer (BC) is the most frequent cancer in Europe and the International Agency for Research on Cancer (IARC) has estimated over 460 000 incident cases per year. Survival among patients with BC has increased in the past decades and EUROCARE-5 has estimated a 5-year relative survival rate of 82% for patients diagnosed in 2000-2007. There is growing evidence that lifestyle (such as a diet based on Mediterranean principles associated with moderate physical activity) may influence prognosis of BC; however, this information is not currently available to patients and is not considered in oncology protocols. Only a few epidemiological studies have investigated the role of diet in BC recurrence and metastasis. METHODS AND ANALYSIS DianaWeb is a community-based participatory research dedicated to patients with BC and represents a collaborative effort between participants and research institutions to determine if specified changes in lifestyle would result in improved outcomes in terms of quality of life or survival. The aim of the study is to recruit a large number of participants, to monitor their lifestyle and health status over time, to provide them tips to encourage sustainable lifestyle changes, to analyse clinical outcomes as a function of baseline risk factors and subsequent changes, and to share with patients methodologies and results. DianaWeb uses a specific interactive website (http://www.dianaweb.org/) and, with very few exceptions, all communications will be made through the web. In this paper we describe the pilot study, namely DianaWeb in Umbria. ETHICS AND DISSEMINATION DianaWeb does not interfere with prescribed oncological treatments; rather, it recommends that participants should follow the received prescriptions. The results will be used to plan guidelines for nutrition and physical activity for patients with BC. The pilot study was approved by the ethics committee of the University of Perugia (reference number 2015-002), and is supported by Fondazione Cassa di Risparmio di Perugia (2013.0185 021).
Collapse
Affiliation(s)
- Milena Villarini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Chiara Lanari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Daniele Nucci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vincenza Gianfredi
- School of Specialization in Hygiene and Preventive Medicine, University of Perugia, Perugia, Italy
| | - Tiziana Marzulli
- School of Specialization in Hygiene and Preventive Medicine, University of Perugia, Perugia, Italy
| | | | - Alessandra Borgo
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eleonora Bruno
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giuliana Gargano
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Moretti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Anna Villarini
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
179
|
COSKUN TEOMAN, KOSOVA FUNDA, ARI ZEKI, SAKARYA ASLAN, KAYA YAVUZ. Effect of oncological treatment on serum adipocytokine levels in patients with stage II-III breast cancer. Mol Clin Oncol 2016; 4:893-897. [PMID: 27123303 PMCID: PMC4840827 DOI: 10.3892/mco.2016.815] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 03/08/2016] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue-derived hormones (adipocytokines), such as adiponectin, leptin, resistin and visfatin, and the pancreatic hormone insulin, have been suggested to play a role in carcinogenesis. we therefore hypothesized that the oncological treatment of breast cancer may alter the serum levels of these adipocytokines and insulin. In this study, we aimed to compare the serum levels of adipocytokines and insulin between the pre- and post-treatment period in patients with breast cancer. In this prospective study, 20 consecutive patients with stage II and III breast cancer underwent breast-conserving surgery or total mastectomy and/or axillary dissection. The patients received adjuvant chemotherapy and radiotherapy, if necessary. Blood samples were obtained during the preoperative period and postoperatively after completion of the adjuvant therapy. There was no statistically significant difference between the pre- and post-treatment levels of visfatin, adiponectin and leptin. However, the serum insulin and resistin levels and insulin resistance were found to be statistically significantly increased following treatment (P<0.05). Post-treatment resistin levels were positively correlated with insulin resistance (r=0.45, P<0.05). Therefore, oncological treatment of stage II and III breast cancer did not affect visfatin, adiponectin and leptin levels, but statistically significantly increased resistin levels and insulin resistance. In addition, the post-treatment resistin levels were positively correlated with insulin resistance, suggesting that resistin may be involved in the development of insulin resistance in breast cancer patients following treatment.
Collapse
Affiliation(s)
- TEOMAN COSKUN
- Department of General Surgery, Faculty of Medicine, Celal Bayar University, 45030 Manisa, Turkey
| | - FUNDA KOSOVA
- School of Health, Celal Bayar University, 45030 Manisa, Turkey
| | - ZEKI ARI
- Department of Biochemistry, Faculty of Medicine, Celal Bayar University, 45030 Manisa, Turkey
| | - ASLAN SAKARYA
- Department of General Surgery, Faculty of Medicine, Celal Bayar University, 45030 Manisa, Turkey
| | - YAVUZ KAYA
- Department of General Surgery, Faculty of Medicine, Celal Bayar University, 45030 Manisa, Turkey
| |
Collapse
|
180
|
Dyson JK, Anstee QM, McPherson S. Republished: Non-alcoholic fatty liver disease: a practical approach to treatment. Postgrad Med J 2016; 91:92-101. [PMID: 25655252 PMCID: PMC4345831 DOI: 10.1136/postgradmedj-2013-100404rep] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects up to a third of the population in many developed countries. Between 10% and 30% of patients with NAFLD have non-alcoholic steatohepatitis (NASH) that can progress to cirrhosis. There are metabolic risk factors common to both NAFLD and cardiovascular disease, so patients with NASH have an increased risk of liver-related and cardiovascular death. Management of patients with NAFLD depends largely on the stage of disease, emphasising the importance of careful risk stratification. There are four main areas to focus on when thinking about management strategies in NAFLD: lifestyle modification, targeting the components of the metabolic syndrome, liver-directed pharmacotherapy for high risk patients and managing the complications of cirrhosis.
Collapse
Affiliation(s)
- J K Dyson
- Liver Unit, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Q M Anstee
- Liver Unit, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - S McPherson
- Liver Unit, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
181
|
Hann A, Frawley A, Spedding G. Not very NICE: deviance, stigma and nutritional guidelines related to healthy weight and obesity. Int J Health Plann Manage 2016; 32:416-432. [PMID: 27062379 DOI: 10.1002/hpm.2350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 11/09/2022] Open
Abstract
This paper critically examines the current National Institute for Health and Clinical Excellence and National Health Service guidelines on weight management and the avoidance of obesity (NG7). We demonstrate that the guidance is unlikely to produce the desired effect of enabling people to reduce or control their weight through the twin strategies of dieting (primarily using the calories-in, calories-out approach) and increasing their levels of exercise. The paper provides a critical examination of these guidelines and concludes that they are unlikely to encourage maintenance of 'healthy' weights or prevent obesity, are not based upon particularly strong evidence and are misguided in maintaining a persistent focus upon weight rather than other indicators of health. Moreover, we suggest their promotion may produce a number of unintended consequences, including perpetuating body-related stigmatisation and anxieties. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Alison Hann
- College of Health and Human Sciences, Swansea University, Swansea, UK
| | - Ashley Frawley
- College of Health and Human Sciences, Swansea University, Swansea, UK
| | - Gillian Spedding
- College of Health and Human Sciences, Swansea University, Swansea, UK
| |
Collapse
|
182
|
Ho GYF, Zheng SL, Cushman M, Perez-Soler R, Kim M, Xue X, Wang T, Schlecht NF, Tinker L, Rohan TE, Wassertheil-Smoller S, Wallace R, Chen C, Xu J, Yu H. Associations of Insulin and IGFBP-3 with Lung Cancer Susceptibility in Current Smokers. J Natl Cancer Inst 2016; 108:djw012. [PMID: 27071409 DOI: 10.1093/jnci/djw012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 01/19/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) signaling network is involved in lung carcinogenesis. This study examined whether ligands that activate or suppress the EGFR signaling network were associated with lung cancer risk in ever smokers. METHODS A nested case-control study within the Women's Health Initiative assessed baseline plasma levels of insulin, insulin-like growth factor (IGF)-1, insulin-like growth factor binding protein (IGFBP)-3, interleukin (IL)-6, hepatocyte growth factor (HGF), and nerve growth factor (NGF) in 1143 ever-smoking lung cancer cases and 1143 controls. Leptin was measured as an adiposity biomarker. Conditional logistic regression was used in data analyses. RESULTS Leptin was inversely associated with lung cancer risk (odds ratio [ORcontinuous] per Ln [pg/mL] = 0.85, 95% confidence interval [CI] = 0.74 to 0.98). After adjusting for adiposity and other risk factors, null associations were found for IL-6, HGF, and NGF. In current smokers, but not former smokers, high insulin levels were associated with increased lung cancer risk (OR for 4th quartile vs others [ORq4] = 2.06, 95% CI = 1.30 to 3.26) whereas IGFBP-3 had a linear inverse association (ORcontinuous per μg/mL = 0.64, 95% CI = 0.41 to 0.98). The insulin association was consistent across subgroups defined by body mass index and histological type, but the IGFBP-3 association was specific to small cell lung cancer. There was a modest positive association between IGF-1 and lung cancer risk in current smokers (ORq4 = 1.44, 95% CI = 0.90 to 2.29). CONCLUSIONS Independent of obesity, high insulin levels but reduced levels of IGFBP-3 were associated with increased lung cancer risk in current smokers.
Collapse
Affiliation(s)
- Gloria Y F Ho
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Siqun L Zheng
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Mary Cushman
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Roman Perez-Soler
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Mimi Kim
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Xiaonan Xue
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Tao Wang
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Nicolas F Schlecht
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Lesley Tinker
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Thomas E Rohan
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Sylvia Wassertheil-Smoller
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Robert Wallace
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Chu Chen
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Jianfeng Xu
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| | - Herbert Yu
- Affiliations of authors:Department of Occupational Medicine, Epidemiology & Prevention, Feinstein Institute for Medical Research, Northwell Health; Hofstra-Northwell School of Medicine , Great Neck, NY (GYFH); Department of Epidemiology and Population Health (GYFH, MK, XX, TW, NFS, TER, SWS) and Department of Medicine (RPS, NFS), Albert Einstein College of Medicine, Bronx, NY; Center for Genomics and Personalized Medicine Research, Wake Forest University , Winston-Salem, NC (SLZ); Departments of Medicine & Pathology, University of Vermont , Burlington, VT (MC); Public Health Sciences Division, Fred Hutchinson Cancer Research Center , Seattle, WA (LT, CC); Department of Epidemiology, College of Public Health, University of Iowa , Iowa City, IA (RW); NorthShore University HealthSystem , Evanston, IL (JX); University of Hawaii Cancer Center , Honolulu, HI (HY)
| |
Collapse
|
183
|
Monzavi-Karbassi B, Gentry R, Kaur V, Siegel ER, Jousheghany F, Medarametla S, Fuhrman BJ, Safar AM, Hutchins LF, Kieber-Emmons T. Pre-diagnosis blood glucose and prognosis in women with breast cancer. Cancer Metab 2016; 4:7. [PMID: 27054036 PMCID: PMC4822279 DOI: 10.1186/s40170-016-0147-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 03/16/2016] [Indexed: 12/02/2022] Open
Abstract
Background The effect of moderately elevated blood glucose levels among non-diabetic subjects on cancer prognosis is not well described. The goal of this study was to examine the association of elevated random blood glucose (RBG) levels in non-diabetic breast cancer patients with overall survival (OS) and time to tumor recurrence (TTR). Results Forty-nine deaths and 32 recurrences occurred among 148 eligible study subjects during 855.44 person-years of follow-up, with median follow-up of 5.97 years. We observed that patients with elevated RBG levels experienced significantly shorter OS (hazard ratio [HR], 3.01; 95 % confidence interval [CI] (1.70–5.33); P < 0.001) and shorter TTR (HR, 2.08; CI (1.04–4.16); P = 0.04) as compared to patients with non-elevated RBG levels. After controlling for tumor grade, tumor stage, race, and BMI, elevated RBG continued to display high and statistically significant association with shorter OS (HR, 3.50; CI (1.87–6.54); P < 0.001). Adjustment for age, race, and BMI strengthened HR of RBG for TTR. The association of RGB with TTR lost its borderline statistical significance upon controlling for both tumor grade and stage. Conclusions The data suggest that elevated blood glucose is associated with poor prognosis of breast cancer patients. Given the potential clinical implication, these findings warrant further investigation.
Collapse
Affiliation(s)
- Behjatolah Monzavi-Karbassi
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Department of Pathology, University of Arkansas for Medical Sciences, 4301 West Markham St., Slot #824, Little Rock, AR 72205 USA
| | - Rhonda Gentry
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Varinder Kaur
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Eric R Siegel
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Fariba Jousheghany
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Srikanth Medarametla
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Barbara J Fuhrman
- Department of Epidemiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - A Mazin Safar
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Laura F Hutchins
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Thomas Kieber-Emmons
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA ; Department of Pathology, University of Arkansas for Medical Sciences, 4301 West Markham St., Slot #824, Little Rock, AR 72205 USA
| |
Collapse
|
184
|
Payne R, Yang M, Zheng Y, Jensen MK, Cai T. Robust risk prediction with biomarkers under two-phase stratified cohort design. Biometrics 2016; 72:1037-1045. [PMID: 27037494 DOI: 10.1111/biom.12515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/01/2015] [Accepted: 02/01/2016] [Indexed: 11/27/2022]
Abstract
Identification of novel biomarkers for risk prediction is important for disease prevention and optimal treatment selection. However, studies aiming to discover which biomarkers are useful for risk prediction often require the use of stored biological samples from large assembled cohorts, and thus the depletion of a finite and precious resource. To make efficient use of such stored samples, two-phase sampling designs are often adopted as resource-efficient sampling strategies, especially when the outcome of interest is rare. Existing methods for analyzing data from two-phase studies focus primarily on single marker analysis or fitting the Cox regression model to combine information from multiple markers. However, the Cox model may not fit the data well. Under model misspecification, the composite score derived from the Cox model may not perform well in predicting the outcome. Under a general two-phase stratified cohort sampling design, we present a novel approach to combining multiple markers to optimize prediction by fitting a flexible nonparametric transformation model. Using inverse probability weighting to account for the outcome-dependent sampling, we propose to estimate the model parameters by maximizing an objective function which can be interpreted as a weighted C-statistic for survival outcomes. Regardless of model adequacy, the proposed procedure yields a sensible composite risk score for prediction. A major obstacle for making inference under two phase studies is due to the correlation induced by the finite population sampling, which prevents standard inference procedures such as the bootstrap from being used for variance estimation. We propose a resampling procedure to derive valid confidence intervals for the model parameters and the C-statistic accuracy measure. We illustrate the new methods with simulation studies and an analysis of a two-phase study of high-density lipoprotein cholesterol (HDL-C) subtypes for predicting the risk of coronary heart disease.
Collapse
Affiliation(s)
- Rebecca Payne
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, U.S.A
| | - Ming Yang
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, U.S.A
| | - Yingye Zheng
- Fred Hutchinson Cancer Research Center, Seattle, Washington, U.S.A
| | - Majken K Jensen
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, U.S.A
| | - Tianxi Cai
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, U.S.A
| |
Collapse
|
185
|
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: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/21/2016] [Accepted: 01/21/2016] [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
|
186
|
Simone V, D'Avenia M, Argentiero A, Felici C, Rizzo FM, De Pergola G, Silvestris F. Obesity and Breast Cancer: Molecular Interconnections and Potential Clinical Applications. Oncologist 2016; 21:404-17. [PMID: 26865587 DOI: 10.1634/theoncologist.2015-0351] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/23/2015] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Obesity is an important risk factor for breast cancer (BC) in postmenopausal women; interlinked molecular mechanisms might be involved in the pathogenesis. Increased levels of estrogens due to aromatization of the adipose tissue, inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and prostaglandin E2, insulin resistance and hyperactivation of insulin-like growth factors pathways, adipokines, and oxidative stress are all abnormally regulated in obese women and contribute to cancerogenesis. These molecular factors interfere with intracellular signaling in the mitogen-activated protein kinase and phosphatydilinositol-3-phosphate/mammalian target of rapamycin (mTOR) pathways, which regulate the progression of the cell cycle, apoptosis, and protein synthesis. In this context, structural defects of typical genes related to both BC and obesity, such as leptin, leptin receptor, serum paraoxonase/arylesterase 1, the fat mass and obesity-associated gene and melanocortin receptor 4, have been associated with a high or low risk of BC development. The early detection of these gene alterations might be useful as risk predictors in obese women, and targeting these pathways involved in the BC pathogenesis in obese women is a potential therapeutic tool. In particular, mTOR pathway deregulation concurs in both obesity and BC, and inhibition of this might disrupt the molecular interlinks in a similar manner to that of metformin, which exerts definite anticancer activity and is currently used as an antidiabetic drug with a weight-reducing property. The identification of both genetic and pharmacological implications on the prevention and management of BC is the ultimate aim of these studies. IMPLICATIONS FOR PRACTICE Obese women are at risk of breast cancer, but clinicians lack concrete tools for the prevention or early diagnosis of this risk. The present study, starting from the biology and the molecular defects characterizing both obesity and breast cancer, analyzed the potential molecules and genetic defects whose early identification could delineate a risk profile. Three steps are proposed that are potentially achievable in the clinical assessment of obese women, namely the evaluation of altered levels of serum molecules, the identification of genetic polymorphisms, and the study of the transcriptomic profile of premalignant lesions. Finally, the therapeutic implications of this molecular assessment were evaluated.
Collapse
Affiliation(s)
- Valeria Simone
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Morena D'Avenia
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Antonella Argentiero
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Claudia Felici
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Francesca Maria Rizzo
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Giovanni De Pergola
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| |
Collapse
|
187
|
Othman EM, Oli RG, Arias-Loza PA, Kreissl MC, Stopper H. Metformin Protects Kidney Cells From Insulin-Mediated Genotoxicity In Vitro and in Male Zucker Diabetic Fatty Rats. Endocrinology 2016; 157:548-59. [PMID: 26636185 DOI: 10.1210/en.2015-1572] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hyperinsulinemia is thought to enhance cancer risk. A possible mechanism is induction of oxidative stress and DNA damage by insulin, Here, the effect of a combination of metformin with insulin was investigated in vitro and in vivo. The rationales for this were the reported antioxidative properties of metformin and the aim to gain further insights into the mechanisms responsible for protecting the genome from insulin-mediated oxidative stress and damage. The comet assay, a micronucleus frequency test, and a mammalian gene mutation assay were used to evaluate the DNA damage produced by insulin alone or in combination with metformin. For analysis of antioxidant activity, oxidative stress, and mitochondrial disturbances, the cell-free ferric reducing antioxidant power assay, the superoxide-sensitive dye dihydroethidium, and the mitochondrial membrane potential-sensitive dye 5,5',6,6'tetrachloro-1,1',3,3'-tetraethylbenzimidazol-carbocyanine iodide were applied. Accumulation of p53 and pAKT were analyzed. As an in vivo model, hyperinsulinemic Zucker diabetic fatty rats, additionally exposed to insulin during a hyperinsulinemic-euglycemic clamp, were treated with metformin. In the rat kidney samples, dihydroethidium staining, p53 and pAKT analysis, and quantification of the oxidized DNA base 8-oxo-7,8-dihydro-2'-deoxyguanosine were performed. Metformin did not show intrinsic antioxidant activity in the cell-free assay, but protected cultured cells from insulin-mediated oxidative stress, DNA damage, and mutation. Treatment of the rats with metformin protected their kidneys from oxidative stress and genomic damage induced by hyperinsulinemia. Metformin may protect patients from genomic damage induced by elevated insulin levels. This may support efforts to reduce the elevated cancer risk that is associated with hyperinsulinemia.
Collapse
Affiliation(s)
- Eman Maher Othman
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - R G Oli
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Paula-Anahi Arias-Loza
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Michael C Kreissl
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology (E.M.O., R.G.O., H.S.), University of Würzburg, D-97078 Würzburg, Germany; Department of Analytical Chemistry (E.M.O.), Faculty of Pharmacy, University of El-Minia, 61519 Minia, Egypt; and Department of Nuclear Medicine (P.-A.A.-L., M.C.K.), University Hospital Würzburg, Würzburg 97080, Germany
| |
Collapse
|
188
|
Polycystic ovarian syndrome (PCOS), related symptoms/sequelae, and breast cancer risk in a population-based case-control study. Cancer Causes Control 2016; 27:403-14. [PMID: 26797454 DOI: 10.1007/s10552-016-0716-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/09/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE Despite the overlap between the clinical symptoms/sequelae of polycystic ovarian syndrome (PCOS) and many known reproductive risk factors for breast cancer, the relationship between PCOS and breast cancer remains unclear, possibly because of the complex heterogeneity and challenges in diagnosing PCOS over time. We hypothesized that PCOS, specific PCOS-related symptoms/sequelae, or clusters of PCOS-related symptoms/sequelae may be differentially associated with pre- versus postmenopausal breast cancer risk. MATERIALS AND METHODS Cases were 1,508 women newly diagnosed with a first primary in situ or invasive breast, and the 1,556 population-based controls were frequency-matched by age. RESULTS History of physician-diagnosed PCOS was reported by 2.2 % (n = 67), among whom oral contraceptive (OC) use, irregular menstruation, and infertility due to ovulatory dysfunction were common. Using unconditional logistic regression, adjusted odds ratios (95 % CI) for PCOS were increased for premenopausal [2.74 (1.13, 6.63)], but not postmenopausal breast cancer [0.87 (0.44, 1.71)]. We used cluster analysis to investigate whether risk among all women varied by PCOS-related symptoms/sequelae, such as reproductive irregularities, OC use, and components of insulin resistance. In the cluster analysis, odds ratios were elevated among premenopausal women who had a history of OC use and no ovulatory dysfunction [1.39 (1.03, 1.88)], compared to those with fewer number of PCOS-related symptoms/sequelae. CONCLUSION PCOS and associated PCOS-related symptoms/sequelae including OC use may play a role in the development of premenopausal breast cancer. Our findings require confirmation in studies with a larger number of premenopausal women with systematically applied diagnostic criteria for PCOS.
Collapse
|
189
|
Schairer C, Fuhrman BJ, Boyd-Morin J, Genkinger JM, Gail MH, Hoover RN, Ziegler RG. Quantifying the Role of Circulating Unconjugated Estradiol in Mediating the Body Mass Index-Breast Cancer Association. Cancer Epidemiol Biomarkers Prev 2016; 25:105-13. [PMID: 26637268 PMCID: PMC5555590 DOI: 10.1158/1055-9965.epi-15-0687] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/23/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Higher body mass index (BMI) and circulating estrogen levels each increase postmenopausal breast cancer risk, particularly estrogen receptor-positive (ER(+)) tumors. Higher BMI also increases estrogen production. METHODS We estimated the proportion of the BMI-ER(+) breast cancer association mediated through estrogen in a case-control study nested within the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Participants included 143 women with invasive ER(+) breast cancer and 268 matched controls, all postmenopausal and never having used hormone therapy at baseline. We used liquid chromatography-tandem mass spectrometry to measure 15 estrogens and estrogen metabolites in baseline serum. We calculated BMI from self-reported height and weight at baseline. We estimated the mediating effect of unconjugated estradiol on the BMI-ER(+) breast cancer association using Aalen additive hazards and Cox regression models. RESULTS All estrogens and estrogen metabolites were statistically significantly correlated with BMI, with unconjugated estradiol most strongly correlated [Pearson correlation (r) = 0.45]. Approximately 7% to 10% of the effect of overweight, 12% to 15% of the effect of obesity, and 19% to 20% of the effect of a 5 kg/m(2) BMI increase on ER(+) breast cancer risk was mediated through unconjugated estradiol. The BMI-breast cancer association, once adjusted for unconjugated estradiol, was not modified by further adjustment for two metabolic ratios statistically significantly associated with both breast cancer and BMI. CONCLUSION Circulating unconjugated estradiol levels partially mediate the BMI-breast cancer association, but other potentially important estrogen mediators (e.g., bioavailable estradiol) were not evaluated. IMPACT Further research is required to identify mechanisms underlying the BMI-breast cancer association.
Collapse
Affiliation(s)
- Catherine Schairer
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland.
| | - Barbara J Fuhrman
- Department of Epidemiology, College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Jeanine M Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York. Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Mitchell H Gail
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Robert N Hoover
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Regina G Ziegler
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| |
Collapse
|
190
|
Wairagu PM, Phan ANH, Kim MK, Han J, Kim HW, Choi JW, Kim KW, Cha SK, Park KH, Jeong Y. Insulin priming effect on estradiol-induced breast cancer metabolism and growth. Cancer Biol Ther 2016; 16:484-92. [PMID: 25701261 DOI: 10.1080/15384047.2015.1016660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Diabetes is a risk factor for breast cancer development and is associated with poor prognosis for breast cancer patients. However, the molecular and biochemical mechanisms underlying the association between diabetes and breast cancer have not been fully elucidated. Here, we investigated estradiol response in MCF-7 breast cancer cells with or without chronic exposure to insulin. We found that insulin priming is necessary and specific for estradiol-induced cancer cell growth, and induces anaplerotic shunting of glucose into macromolecule biosynthesis in the estradiol treated cells. Treatment with ERK or Akt specific inhibitors, U0126 or LY294002, respectively, suppressed estradiol-induced growth. Interestingly, molecular analysis revealed that estradiol treatment markedly increases expression of cyclin A and B, and decreases p21 and p27 in the insulin-primed cells. In addition, estradiol treatment activated metabolic genes in pentose phosphate (PPP) and serine biosynthesis pathways in the insulin-primed cells while insulin priming decreased metabolic gene expression associated with glucose catabolism in the breast cancer cells. Finally, we found that anti-diabetic drug metformin and AMPK ligand AICAR, but not thiazolidinediones (TZDs), specifically suppress the estradiol-induced cellular growth in the insulin-primed cells. These findings suggest that estrogen receptor (ER) activation under chronic hyperinsulinemic condition increases breast cancer growth through the modulation of cell cycle and apoptotic factors and nutrient metabolism, and further provide a mechanistic evidence for the clinical benefit of metformin use for ER-positive breast cancer patients with diabetes.
Collapse
Key Words
- AR, androgen receptor
- CKI, cyclin dependent-kinase inhibitor
- DHT, dihydrotestosterone
- ER, estrogen receptor
- G6PD, glucose-6-phosphodehydrogenase
- GLUT1, glucose transporter 1
- HER2, human epidermal growth factor receptor 2
- IGF-1, insulin-like growth factor 1
- LDHA, lactate dehydrogenase A
- MCF-7
- PDK1, pyruvate dehydrogenase kinase 1
- PFK, phosphofructokinase
- PFKFB3, 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphate 3
- PGD, 6-phosphogluconate dehydrogenase
- PHGDH, phosphoglycerate dehydrogenase
- PKM, pyruvate kinase M
- PPAR γ, peroxisome proliferator-activated receptor gamma
- PPP, pentose phosphate pathway
- PR, progesterone receptor
- PSPH, phosphoserine phosphatase
- RPE, ribulose-5-phosphate-3-epimerase
- RPIA, ribulose-5-phosphate isomerase A
- SHMT, serine hydroxymethyltransferase
- TALDO1, transaldolase 1
- TKT, transketolase
- TZDs, thiazolidinediones
- breast cancer
- diabetes
- estradiol
- estrogen receptor
- iInsulin priming
- metformin
Collapse
Affiliation(s)
- Peninah M Wairagu
- a Department of Biochemistry ; Wonju College of Medicine ; Yonsei University ; Wonju , Gangwon-do , Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Ben-Shmuel S, Rostoker R, Scheinman EJ, LeRoith D. Metabolic Syndrome, Type 2 Diabetes, and Cancer: Epidemiology and Potential Mechanisms. Handb Exp Pharmacol 2016; 233:355-372. [PMID: 25903410 DOI: 10.1007/164_2015_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Obesity is associated with multiple metabolic disorders that drive cardiovascular disease, T2D and cancer. The doubling in the number of obese adults over the past 3 decades led to the recognition of obesity as a "disease". With over 42 million children obese or overweight, this epidemic is rapidly growing worldwide. Obesity and T2D are both associated together and independently with an increased risk for cancer and a worse prognosis. Accumulating evidence from epidemiological studies revealed potential factors that may explain the association between obesity-linked metabolic disorders and cancer risk. Studies based on the insulin resistance MKR mice, highlighted the roe of the insulin receptor and its downstream signaling proteins in mediating hyperinsulinemia's mitogenic effects. Hypercholesterolemia was also shown to promote the formation of larger tumors and enhancement in metastasis. Furthermore, the conversion of cholesterol into 27-Hydroxycholesterol was found to link high fat diet-induced hypercholesterolemia with cancer pathophysiology. Alteration in circulating adipokines and cytokines are commonly found in obesity and T2D. Adipokines are involved in tumor growth through multiple mechanisms including mTOR, VEGF and cyclins. In addition, adipose tissues are known to recruit and alter macrophage phenotype; these macrophages can promote cancer progression by secreting inflammatory cytokines such as TNF-α and IL-6. Better characterization on the above factors and their downstream effects is required in order to translate the current knowledge into the clinic, but more importantly is to understand which are the key factors that drive cancer in each patient. Until we reach this point, policies and activities toward healthy diets and physical activities remain the best medicine.
Collapse
Affiliation(s)
- Sarit Ben-Shmuel
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Ran Rostoker
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Eyal J Scheinman
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Derek LeRoith
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
192
|
Abstract
The relationship between adiposity and breast cancer risk and prognosis is complex, with associations that differ depending on when body size is assessed (e.g., pre- vs. postmenopausal obesity) and when breast cancer is diagnosed (i.e., pre- vs. postmenopausal disease). Further, the impact of obesity on risk differs by tumor hormone receptor status (e.g., estrogen (ER) and progesterone (PR) receptor) and, among postmenopausal women, use of exogenous hormones (i.e., hormone replacement therapy (HRT)). In the context of these complexities, this review focuses on associations between childhood and adolescent adiposity, general adiposity, weight changes (i.e., loss and gain), abdominal adiposity, and breast cancer risk and survival. Finally, we discuss potential mechanisms linking adiposity to breast cancer.
Collapse
Affiliation(s)
- Renée T Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
193
|
Nimptsch K, Pischon T. Obesity Biomarkers, Metabolism and Risk of Cancer: An Epidemiological Perspective. Recent Results Cancer Res 2016; 208:199-217. [PMID: 27909909 DOI: 10.1007/978-3-319-42542-9_11] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Obesity is associated with metabolic alterations that may pose a biological link between body fatness and risk of cancer. Elucidating the role of obesity-related biomarkers in cancer development is essential for developing targeted strategies aiming at obesity-associated cancer prevention. Molecular epidemiological studies of the past decades have provided evidence that major hormonal pathways linking obesity and cancer risk include the insulin and insulin-like growth factor-1 (IGF-1) axis, sex-steroid hormones, adipokines and chronic low-grade inflammation. These pathways are interrelated with each other, and their importance varies by obesity-related cancer type. The insulin/IGF-1 axis has been implicated to play an important mediating role in the association between obesity and risk of pancreatic, colorectal and prostate cancer. Endogenous sex-steroid hormone concentrations, in particular obesity-associated pre-diagnostic elevations of estrogens and androgens, play an important role in postmenopausal breast cancer and endometrial cancer development. The adipokines adiponectin and leptin and adipocyte-mediated chronic low-grade inflammation represented by the acute-phase C-reactive protein may explain a substantial part of the association between obesity and risk of colorectal cancer. There is less evidence on whether these hormonal pathways play a mediating role in other obesity-associated types of cancer. In this chapter, the molecular epidemiologic evidence from prospective studies relating circulating obesity-related biomarkers to cancer risk is summarized, taking into account available evidence from Mendelian Randomization investigations aiming at improving causal inference.
Collapse
Affiliation(s)
- Katharina Nimptsch
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany
| |
Collapse
|
194
|
Şendur MAN, Aksoy S, Akıncı MB, Ozdemir N, Dede DŞ, Altundag K, Zengin N, Yalçın B. Role of obesity on the efficacy of exemestane plus ovarian suppression in hormone receptor-positive premenopausal breast cancer. Future Oncol 2015; 11:905-7. [PMID: 25760972 DOI: 10.2217/fon.15.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mehmet Ali Nahit Şendur
- Department of Medical Oncology, Faculty of Medicine, Yıldırım Beyazıt University, 06100 Sihhiye, Ankara, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
195
|
Jansen N, Walach H. The development of tumours under a ketogenic diet in association with the novel tumour marker TKTL1: A case series in general practice. Oncol Lett 2015; 11:584-592. [PMID: 26870251 PMCID: PMC4726921 DOI: 10.3892/ol.2015.3923] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 07/21/2015] [Indexed: 01/01/2023] Open
Abstract
Since the initial observations by Warburg in 1924, it has become clear in recent years that tumour cells require a high level of glucose to proliferate. Therefore, a ketogenic diet that provides the body with energy mainly through fat and proteins, but contains a reduced amount of carbohydrates, has become a dietary option for supporting tumour treatment and has exhibited promising results. In the present study, the first case series of such a treatment in general practice is presented, in which 78 patients with tumours were treated within a time window of 10 months. The patients were monitored regarding their levels of transketolase-like-1 (TKTL1), a novel tumour marker associated with aerobic glycolysis of tumour cells, and the patients' degree of adherence to a ketogenic diet. Tumour progression was documented according to oncologists' reports. Tumour status was correlated with TKTL1 expression (Kruskal-Wallis test, P<0.0001), indicating that more progressed and aggressive tumours may require a higher level of aerobic glycolysis. In palliative patients, a clear trend was observed in patients who adhered strictly to a ketogenic diet, with one patient experiencing a stagnation in tumour progression and others an improvement in their condition. The adoption of a ketogenic diet was also observed to affect the levels of TKTL1 in those patients. In conclusion, the results from the present case series in general practice suggest that it may be beneficial to advise tumour patients to adopt a ketogenic diet, and that those who adhere to it may have positive results from this type of diet. Thus, the use of a ketogenic diet as a complementary treatment to tumour therapy must be further studied in rigorously controlled trials.
Collapse
Affiliation(s)
- Natalie Jansen
- Dr Natalie Jansen Private Practice, Heilbronn, Baden-Württemberg, D-74074 Heilbronn, Germany; Institute of Transcultural Health Studies, European University Viadrina, D-15207 Frankfurt (Oder), Germany
| | - Harald Walach
- Institute of Transcultural Health Studies, European University Viadrina, D-15207 Frankfurt (Oder), Germany
| |
Collapse
|
196
|
Ligibel JA, Strickler HD. Obesity and its impact on breast cancer: tumor incidence, recurrence, survival, and possible interventions. Am Soc Clin Oncol Educ Book 2015:52-9. [PMID: 23714455 DOI: 10.14694/edbook_am.2013.33.52] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A positive association between obesity and the risk of incident postmenopausal breast cancer has been consistently observed in epidemiologic studies. Although most studies of premenopausal women have not found a similar relationship between breast cancer and obesity, the prognosis for both pre- and postmenopausal breast cancer is substantially worse among obese than normal-weight individuals. Increasing evidence suggests that these associations may be mechanistically related to sex hormones, insulin, and certain adipokines. Insulin, for example, has important mitogenic/antiapoptotic activity in addition to its metabolic effects, and many breast tumors express high levels of the insulin receptor (IR)-A isoform. Further, the use of metformin, a diabetes medication that reduces insulin levels, has been epidemiologically associated with reduced breast cancer risk among patients with diabetes, and a recent observational study found a higher rate of pathologic complete responses among patients with diabetes and breast cancer who were using metformin. Formal clinical trials of metformin as adjuvant breast cancer therapy have been initiated and are ongoing. Similarly, the effect of lifestyle changes on breast cancer outcomes is actively being investigated. Several lifestyle intervention studies have demonstrated that weight loss, increased physical activity, and dietary changes are feasible in breast cancer populations, and that individuals who make lifestyle changes after breast cancer diagnosis experience several physical and psychologic benefits. In this article, the authors review the evidence linking obesity with breast cancer risk and outcomes and provide an overview of lifestyle intervention studies in patients with breast cancer.
Collapse
Affiliation(s)
- Jennifer A Ligibel
- From the Department of Adult Oncology, Dana-Farber Cancer Institute, Boston, MA; the Department of Epidemiology & Population Health, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY
| | | |
Collapse
|
197
|
Rose DP, Gracheck PJ, Vona-Davis L. The Interactions of Obesity, Inflammation and Insulin Resistance in Breast Cancer. Cancers (Basel) 2015; 7:2147-68. [PMID: 26516917 PMCID: PMC4695883 DOI: 10.3390/cancers7040883] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/21/2015] [Accepted: 10/19/2015] [Indexed: 12/29/2022] Open
Abstract
Obese postmenopausal women have an increased breast cancer risk, the principal mechanism for which is elevated estrogen production by adipose tissue; also, regardless of menstrual status and tumor estrogen dependence, obesity is associated with biologically aggressive breast cancers. Type 2 diabetes has a complex relationship with breast cancer risk and outcome; coexisting obesity may be a major factor, but insulin itself induces adipose aromatase activity and estrogen production and also directly stimulates breast cancer cell growth and invasion. Adipose tissue inflammation occurs frequently in obesity and type 2 diabetes, and proinflammatory cytokines and prostaglandin E2 produced by cyclooxygenase-2 in the associated infiltrating macrophages also induce elevated aromatase expression. In animal models, the same proinflammatory mediators, and the chemokine monocyte chemoattractant protein-1, also stimulate tumor cell proliferation and invasion directly and promote tumor-related angiogenesis. We postulate that chronic adipose tissue inflammation, rather than body mass index-defined obesity per se, is associated with an increased risk of type 2 diabetes and postmenopausal estrogen-dependent breast cancer. Also, notably before the menopause, obesity and type 2 diabetes, or perhaps the associated inflammation, promote estrogen-independent, notably triple-negative, breast cancer development, invasion and metastasis by mechanisms that may involve macrophage-secreted cytokines, adipokines and insulin.
Collapse
Affiliation(s)
- David P Rose
- Mary Babb Randolph Cancer Center,West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Peter J Gracheck
- Mary Babb Randolph Cancer Center,West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Linda Vona-Davis
- Mary Babb Randolph Cancer Center,West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Department of Surgery, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| |
Collapse
|
198
|
Kang HW, Jung HD, Ha YS, Kim TH, Kwon TG, Byun SS, Yun SJ, Kim WJ, Choi YD. Preoperative Underweight Patients with Upper Tract Urothelial Carcinoma Survive Less after Radical Nephroureterectomy. J Korean Med Sci 2015; 30:1483-9. [PMID: 26425047 PMCID: PMC4575939 DOI: 10.3346/jkms.2015.30.10.1483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/18/2015] [Indexed: 11/20/2022] Open
Abstract
The prognostic impact of body mass index (BMI) in patients with upper tract urothelial carcinoma (UTUC) is an ongoing debate. Our study aimed to investigate the prognostic role of BMI in patients treated with radical nephroureterectomy (RNU) for UTUC from a multi-institutional Korean collaboration. We retrospectively reviewed data from 440 patients who underwent RNU for UTUC at four institutions in Korea. To avoid biasing the survival estimates, patients who had previous or concomitant muscle-invasive bladder tumors were excluded. BMI was categorized into approximate quartiles with the lowest quartile assigned to the reference group. Kaplan-Meier and multivariate Cox regression analyses were performed to assess the influence of BMI on survival. The lower quartile BMI group showed significantly increased overall mortality (OM) and cancer specific mortality (CSM) compared to the 25%-50% quartiles and upper quartile BMI groups. Kaplan-Meier estimates showed similar results. Based on multivariate Cox regression analysis, preoperative BMI as a continuous variable was an independent predictor for OM and CSM. In conclusion, preoperative underweight patients with UTUC in Korea survive less after RNU. Preoperative BMI may provide additional prognostic information to establish risk factors.
Collapse
Affiliation(s)
- Ho Won Kang
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Hae Do Jung
- Department of Urology, Severance Hospital, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Tae-Hwan Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seok-Joong Yun
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Korea
| | - Young Deuk Choi
- Department of Urology, Severance Hospital, Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Robot and Minimal Invasive Surgery Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
199
|
DeCensi A, Puntoni M, Guerrieri-Gonzaga A, Cazzaniga M, Serrano D, Lazzeroni M, Vingiani A, Gentilini O, Petrera M, Viale G, Cuzick J, Bonanni B, Pruneri G. Effect of Metformin on Breast Ductal Carcinoma In Situ Proliferation in a Randomized Presurgical Trial. Cancer Prev Res (Phila) 2015; 8:888-94. [PMID: 26276754 DOI: 10.1158/1940-6207.capr-15-0048] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/06/2015] [Indexed: 11/16/2022]
Abstract
Metformin is associated with lower breast cancer risk in epidemiologic studies and showed decreased proliferation in HER2-positive breast cancer in a presurgical trial. To provide insight into its preventive potential, we measured proliferation by Ki-67 labeling index (LI) of intraepithelial lesions surrounding breast cancer. We randomly assigned 200 nondiabetic patients diagnosed with invasive breast cancer in core biopsies to metformin, 1,700 mg or placebo once daily for 28 days before surgery. Upon surgery, five to seven specimens of cancer adjacent (≤1 cm) and distant (>1 cm) tissue were screened for LCIS, ductal carcinoma in situ (DCIS), and ductal hyperplasia (DH). The prevalence of LCIS, DCIS, and DH was 4.5% (9/200), 67% (133/200), and 35% (69/200), respectively. Overall, metformin did not affect Ki-67 LI in premalignant disorders. The median posttreatment Ki-67 LI (IQR) in the metformin and placebo arm was, respectively, 15% (5-15) versus 5% (4-6) in LCIS (P = 0.1), 12% (8-20) versus 10% (7-24) in DCIS (P = 0.9), and 3% (1-4) versus 3% (1-4) in DH (P = 0.5). However, posttreatment Ki-67 in HER2-positive DCIS lesions was significantly lower in women randomized to metformin especially when ER was coexpressed: 22% (11-32) versus 35% (30-40) in HER2-positive DCIS (n = 22, P = .06); 12% (7-18) versus 32% (27-42) in ER-positive/HER2-positive DCIS (n = 15, P = .004). Eight of 22 (36%) HER2-positive DCIS were adjacent to HER2-negative invasive breast cancer. In tissue samples obtained following 4 weeks of study drug, proliferation was lower in HER2-positive DCIS for women randomized to metformin versus placebo. An adjuvant trial incorporating metformin in HER2-positive DCIS is warranted.
Collapse
Affiliation(s)
- Andrea DeCensi
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy. Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom.
| | - Matteo Puntoni
- Office of the Scientific Director, E.O. Ospedali Galliera, Genoa, Italy
| | | | - Massimiliano Cazzaniga
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Davide Serrano
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Matteo Lazzeroni
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Andrea Vingiani
- Division of Pathology, European Institute of Oncology, Milan, Italy
| | - Oreste Gentilini
- Division of Breast Surgery, European Institute of Oncology, Milan, Italy
| | - Marilena Petrera
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology, Milan, Italy. University of Milan, School of Medicine, Milan, Italy
| | - Jack Cuzick
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
| | - Bernardo Bonanni
- Divisions of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Giancarlo Pruneri
- Division of Pathology, European Institute of Oncology, Milan, Italy. University of Milan, School of Medicine, Milan, Italy
| |
Collapse
|
200
|
Setoyama T, Miyamoto S, Nikaido M, Muto M, Chiba T. Instability of IGF-IGFBP complex as a cause of the different performance of serum and EDTA-plasma after storage: EDTA-plasma is preferable for evaluating bioactive IGF especially in the mouse. Growth Horm IGF Res 2015; 25:227-231. [PMID: 26144570 DOI: 10.1016/j.ghir.2015.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 06/16/2015] [Accepted: 06/26/2015] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The insulin-like growth factor (IGF) signaling pathway is recognized as a potential target for treating several cancers, and strategies targeting the IGF type 1 receptor (IGF-1R) have been evaluated in many clinical trials. These suggested that the pretreatment level of circulating free IGF gives an estimate of IGF bioactivity and might be a predictive biomarker of the response to anti-IGF-1R antibodies. However, there is no defined protocol for measuring free and bioactive IGF concentrations, partly because the measurement procedures, including sample collection and handling, have not been standardized. We investigated the effects of sample collection methods and storage conditions on bioactive IGF measurement using a modified kinase receptor activation (KIRA) assay in human and mouse samples. DESIGN Blood samples were obtained from healthy men and women, and from healthy male and female wild-type BALB/c mice. Serum and ethylenediaminetetraacetic acid (EDTA)-plasma samples were collected and used immediately or stored in small quantities at 4 °C or -80 °C for 3, 7, or 14 days. A bioassay directed against the phosphorylated IGF-1R using western blot analysis was developed as a modification of the KIRA assay, in which the level of phosphorylation of IGF-1R represented the IGF bioactivity in blood samples. RESULTS The levels of bioactive IGFs in mouse serum stored at 4 °C increased markedly in a time-dependent manner; the increase was slightly reduced in samples stored at -80 °C. Analysis of mouse EDTA-plasma stored at 4 °C showed a similar pattern, but the time-dependent increase was less than in the serum samples. By contrast, the levels of bioactive IGFs in EDTA-plasma stored at -80 °C were stable over 14 days. The levels of human bioactive IGFs in both serum and EDTA-plasma stored at 4 °C increased slightly with time, but the increases were much smaller than in mouse samples. The levels of human bioactive IGF in both serum and EDTA-plasma stored at -80 °C were stable over 14 days. CONCLUSIONS The use of EDTA-plasma avoids the problems with long-term storage. Therefore, EDTA-plasma should be used when measuring circulating IGF bioactivity, especially in mouse samples. All samples should be stored at -80 °C when long-term storage is unavoidable. Because of the large difference in the stability of the IGF-IGF-binding protein complex between the human and mouse in vitro, all samples should be handled carefully to ensure the accurate evaluation of IGF bioactivity, especially in mouse samples.
Collapse
Affiliation(s)
- Takeshi Setoyama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin'ichi Miyamoto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Mitsuhiro Nikaido
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|