201
|
Denis GV, Palmer JR. "Obesity-Associated" Breast Cancer in Lean Women: Metabolism and Inflammation as Critical Modifiers of Risk. Cancer Prev Res (Phila) 2017; 10:267-269. [PMID: 28408379 DOI: 10.1158/1940-6207.capr-17-0083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 03/29/2017] [Accepted: 03/29/2017] [Indexed: 01/27/2023]
Abstract
Why is obesity only weakly associated with certain "obesity-driven" cancers? Recent population studies identify cohorts of high body mass index (BMI) subjects with unexpectedly reduced risk for breast and colon cancer, and normal BMI subjects with unexpectedly elevated risk for breast cancer, provoking hard thinking about cellular and molecular mechanisms that most strongly couple obesity to cancer occurrence or progression. Emerging work suggests that abnormal metabolism and its associated chronic inflammation make the difference. Type II diabetes, for example, is a chronic inflammatory disease with specific imbalances in T-cell and myeloid-origin cytokines. Inflammation is elevated systemically, measured through blood biomarkers, and locally in adipose tissue. Here, cytokines and chemokines likely modify tumor microenvironments in dangerous ways. High BMI subjects with low inflammation and less disturbed metabolism appear to have reduced risk for certain obesity-associated cancers, whereas lean or slightly overweight subjects with high inflammation and metabolic abnormalities have elevated risk. This latter phenotype is prevalent among South Asian adults and suggests we are not monitoring certain normal weight adults sufficiently for risks of "obesity-associated" cancers. Profiling of patient metabolism and inflammation should accompany measures of body composition when considering cancer risk; the evidence base for these refinements must be extended through new, prospective observational studies. Cancer Prev Res; 10(5); 267-9. ©2017 AACRSee related article by Iyengar et al., Cancer Prev Res 2017;10(4):235-43.
Collapse
Affiliation(s)
- Gerald V Denis
- Cancer Center, Boston University School of Medicine, Boston, Massachusetts.
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, Boston, Massachusetts
| |
Collapse
|
202
|
Iyengar NM, Brown KA, Zhou XK, Gucalp A, Subbaramaiah K, Giri DD, Zahid H, Bhardwaj P, Wendel NK, Falcone DJ, Wang H, Williams S, Pollak M, Morrow M, Hudis CA, Dannenberg AJ. Metabolic Obesity, Adipose Inflammation and Elevated Breast Aromatase in Women with Normal Body Mass Index. Cancer Prev Res (Phila) 2017; 10:235-243. [PMID: 28270386 PMCID: PMC5380584 DOI: 10.1158/1940-6207.capr-16-0314] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/25/2017] [Accepted: 02/04/2017] [Indexed: 01/25/2023]
Abstract
Obesity is associated with breast white adipose tissue (WAT) inflammation, elevated levels of the estrogen biosynthetic enzyme, aromatase, and systemic changes that have been linked to the pathogenesis of breast cancer. Here, we determined whether metabolic obesity, including changes in breast biology and systemic effects, occurs in a subset of women with normal body mass index (BMI). Breast WAT and fasting blood were collected from 72 women with normal BMI (<25 kg/m2) undergoing mastectomy for breast cancer risk reduction or treatment. WAT inflammation was defined by the presence of crown-like structures of the breast (CLS-B) which are composed of dead or dying adipocytes surrounded by macrophages. Severity of inflammation was measured as CLS-B/cm2 The primary objective was to determine whether breast WAT inflammation is associated with aromatase expression and activity. Secondary objectives included assessment of circulating factors and breast adipocyte size. Breast WAT inflammation was present in 39% of women. Median BMI was 23.0 kg/m2 (range, 18.4-24.9 kg/m2) in women with breast WAT inflammation versus 21.8 kg/m2 (range, 17.3-24.6 kg/m2) in those without inflammation (P = 0.04). Breast WAT inflammation was associated with elevated aromatase expression and activity, which increased with severity of inflammation (P < 0.05). Breast WAT inflammation correlated with larger adipocytes (P = 0.01) and higher circulating levels of C-reactive protein, leptin, insulin, and triglycerides (P ≤ 0.05). A subclinical inflammatory state associated with elevated aromatase in the breast, adipocyte hypertrophy, and systemic metabolic dysfunction occurs in some normal BMI women and may contribute to the pathogenesis of breast cancer. Cancer Prev Res; 10(4); 235-43. ©2017 AACRSee related article by Berger, p. 223-25.
Collapse
Affiliation(s)
- Neil M Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kristy A Brown
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Ayca Gucalp
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Dilip D Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heba Zahid
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Nils K Wendel
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Domenick J Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Hanhan Wang
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Samantha Williams
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Pollak
- Department of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | | |
Collapse
|
203
|
Ronco AL, Espinosa E, Calderon JM, Lasalvia- Galante E, De Rosa A, Sanchez G. ‘Mate’ Intake, Hormone-Based Risk Factors and Breast Cancer: a Case-Control Study. Asian Pac J Cancer Prev 2017; 18:941-948. [PMID: 28545191 PMCID: PMC5494243 DOI: 10.22034/apjcp.2017.18.4.941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Previous reports on the inverse association between ‘mate’ intake (infusion of Ilex Paraguariensis herb) and breast cancer (BC) risk led us to consider two main roles for the infusion: as a substantial antioxidant contributor and as a hormone regulator, particularly through anti-aromatase capacities. Since menstrual-reproductive risk factors for BC reflect women’s estrogenic exposure during the reproductive lifespan, and considering that ‘mate’ intake exerts putative stronger protection among high antioxidant contributors, we attempted to analyze interactions among the infusion, hormon-linked reproductive factors and BC risk, which have hitherto remained unexplored. We analyzed a database of 572 BC incident cases and 889 controls. Women were interviewed with a specific questionnaire featuring socio-demographic, lifestyle and reproductive variables (age at menarche, 1st live birth and menopause; number of live births; breastfeeding months), and a food frequency questionnaire, focusing on ‘mate’ intake (consumer status, daily intake, age at start, age at quit, duration of habit). Odds ratios (OR) and their 95% confidence Intervals were calculated through unconditional logistic regression, adjusting for relevant potential confounders. ‘Mate’ intake showed strong inverse associations with some high-risk hormone-related factors: early menarche, nulliparity, low breastfeeding, long reproductive years and high number of ovulatory cycles. Moreover, all subsets of high dietary energy demonstrated even stronger associations. In conclusion, regarding exposure to known hormone risk factors, we found strong inverse associations between high ‘mate’ intake and BC, which were greater among those consuming higher calorific diets. Our analyses support possible combined antioxidant and antiestrogenic effects for ‘mate’ infusions.
Collapse
Affiliation(s)
- Alvaro L Ronco
- Unit of Oncology and Radiotherapy, Pereira Rossell Women’s Hospital, Bvar. Artigas 1550, Montevideo 11600, Uruguay
- IUCLAEH School of Medicine, Prado and Salt Lake, Maldonado 20100, Uruguay
- Biomedical Sciences Center, University of Montevideo, Puntas de Santiago 1604, Montevideo 11500, Uruguay.
| | | | | | | | | | | |
Collapse
|
204
|
Berger NA. Crown-like Structures in Breast Adipose Tissue from Normal Weight Women: Important Impact. Cancer Prev Res (Phila) 2017; 10:223-225. [PMID: 28274935 DOI: 10.1158/1940-6207.capr-17-0062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 11/16/2022]
Abstract
Crown-like structures (CLS), composed of macrophages surrounding dead or dying adipocytes, are a histologic hallmark of the proinflammatory process by which adipose tissue contributes to the increased risk and worse prognosis of breast cancer in obese, postmenopausal patients. In this issue of Cancer Prevention Research, Iyengar and colleagues report the intriguing finding that CLS can be identified in a significant proportion of normal-BMI women undergoing mastectomy for breast cancer risk reduction or therapy. This surprising observation suggests that some normal weight women may have similar mechanisms driving initiation and/or progression of breast cancer as those contributing to the increased incidence and worse prognosis of breast cancer in obese postmenopausal women. The possibility of a common mechanism in both lean and obese women provides added impetus to more fully define this process and evaluate its important implications for prevention and screening strategies as well as therapeutic interventions. Cancer Prev Res; 10(4); 223-5. ©2017 AACRSee related article by Iyengar et al., p. 235-43.
Collapse
Affiliation(s)
- Nathan A Berger
- Departments of Medicine, Biochemistry, Genetics, and Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
205
|
Miyahira AK, Roychowdhury S, Goswami S, Ippolito JE, Priceman SJ, Pritchard CC, Sfanos KS, Subudhi SK, Simons JW, Pienta KJ, Soule HR. Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer; Report From the 2016 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2017; 77:123-144. [PMID: 27679977 DOI: 10.1002/pros.23260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The 2016 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, "Beyond Seed and Soil: Understanding and Targeting Metastatic Prostate Cancer," was held from June 23 to June 26, 2016, in Coronado, California. METHODS For the 4th year in a row, the Prostate Cancer Foundation (PCF) hosted the CHPCA Meeting, a think tank-structured scientific conference, which focuses on a specific topic of critical unmet need on the biology and treatment of advanced prostate cancer. The 2016 CHPCA Meeting was attended by 71 investigators from prostate cancer and other fields, who discussed the biology, study methodologies, treatment strategies, and critical unmet needs concerning metastatic prostate cancer, with the ultimate goal of advancing strategies to treat and eliminate this disease. RESULTS The major topics of discussion included: the molecular landscape and molecular heterogeneity of metastatic prostate cancer, the role of the metastatic microenvironment, optimizing immunotherapy in metastatic prostate cancer, learning from exceptional responders and non-responders, targeting DNA repair deficiency in advanced prostate cancer, developing and applying novel biomarkers and imaging techniques, and potential roles for the microbiome in prostate cancer. DISCUSSION This article reviews the topics presented and discussions held at the CHPCA Meeting, with a focus on the unknowns and next steps needed to advance our understanding of the biology and most effective treatment strategies for metastatic prostate cancer. Prostate 77:123-144, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Sameek Roychowdhury
- Comprehensive Cancer Center, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Division of Medical Oncology, Ohio State University, Cincinnati, Ohio
| | - Sangeeta Goswami
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Joseph E Ippolito
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Saul J Priceman
- Departments of Hematology and Hematopoietic Cell Transplantation, and Immuno-Oncology, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Kenneth J Pienta
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, The James Buchanan Brady Urological Institute, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
206
|
Chuffa LGDA, Lupi-Júnior LA, Costa AB, Amorim JPDA, Seiva FRF. The role of sex hormones and steroid receptors on female reproductive cancers. Steroids 2017; 118:93-108. [PMID: 28041951 DOI: 10.1016/j.steroids.2016.12.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/10/2016] [Accepted: 12/24/2016] [Indexed: 02/08/2023]
Abstract
Sex steroids have been widely described to be associated with a number of human diseases, including hormone-dependent tumors. Several studies have been concerned about the factors regulating the availability of sex steroids and its importance in the pathophysiological aspects of the reproductive cancers in women. In premenopausal women, large fluctuations in the concentration of circulating estradiol (E2) and progesterone (P4) orchestrate many events across the menstrual cycle. After menopause, the levels of circulating E2 and P4 decline but remain at high concentration in the peripheral tissues. Notably, there is a strong relationship between circulating sex hormones and female reproductive cancers (e.g. ovarian, breast, and endometrial cancers). These hormones activate a number of specific signaling pathways after binding either to estrogen receptors (ERs), especially ERα, ERα36, and ERβ or progesterone receptors (PRs). Importantly, the course of the disease will depend on particular transactivation pathway. Identifying ER- or PR-positive tumors will benefit patients in terms of proper endocrine therapy. Based on hormonal responsiveness, effective prevention methods for ovarian, breast, and endometrial cancers represent a special opportunity for women at risk of malignancies. Hormone replacement therapy (HRT) might significantly increase the risk of these cancer types, and endocrine treatments targeting ER signaling may be helpful against E2-dependent tumors. This review will present the role of sex steroids and their receptors associated with the risk of developing female reproductive cancers, with emphasis on E2 levels in pre and postmenopausal women. In addition, new therapeutic strategies for improving the survival rate outcomes in women will be addressed.
Collapse
Affiliation(s)
| | - Luiz Antonio Lupi-Júnior
- Department of Anatomy, IBB/UNESP, Institute of Biosciences of Botucatu, Univ. Estadual Paulista, SP, Brazil
| | - Aline Balandis Costa
- Department of Nursing, UENP/CLM - Universidade Estadual do Norte do Paraná, PR, Brazil
| | | | | |
Collapse
|
207
|
Mullooly M, Yang HP, Falk RT, Nyante SJ, Cora R, Pfeiffer RM, Radisky DC, Visscher DW, Hartmann LC, Carter JM, Degnim AC, Stanczyk FZ, Figueroa JD, Garcia-Closas M, Lissowska J, Troester MA, Hewitt SM, Brinton LA, Sherman ME, Gierach GL. Relationship between crown-like structures and sex-steroid hormones in breast adipose tissue and serum among postmenopausal breast cancer patients. Breast Cancer Res 2017; 19:8. [PMID: 28103902 PMCID: PMC5244534 DOI: 10.1186/s13058-016-0791-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/05/2016] [Indexed: 01/16/2023] Open
Abstract
Background Postmenopausal obesity is associated with increased circulating levels of androgens and estrogens and elevated breast cancer risk. Crown-like structures (CLS; microscopic foci of dying adipocytes surrounded by macrophages) are proposed to represent sites of increased aromatization of androgens to estrogens. Accordingly, we examined relationships between CLS and sex-steroid hormones in breast adipose tissue and serum from postmenopausal breast cancer patients. Methods Formalin-fixed paraffin embedded benign breast tissues collected for research from postmenopausal women (n = 83) diagnosed with invasive breast cancer in the Polish Breast Cancer Study (PBCS) were evaluated. Tissues were immunohistochemically stained for CD68 to determine the presence of CLS per unit area of adipose tissue. Relationships were assessed between CD68 density and CLS and previously reported sex-steroid hormones quantified using radioimmunoassays in serum taken at the time of diagnosis and in fresh frozen adipose tissue taken at the time of surgery. Logistic regression analysis was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the relationships between hormones (in tertiles) and CLS. Results CLS were observed in 36% of benign breast tissues, with a higher frequency among obese versus lean women (54% versus 17%, p = 0.03). Detection of CLS was not related to individual hormone levels or breast tumor pathology characteristics. However, detection of CLS was associated with hormone ratios. Compared with women in the highest tertile of estrone:androstenedione ratio in fat, those in the lowest tertile were less likely to have CLS (OR 0.12, 95% CI 0.03–0.59). A similar pattern was observed with estradiol:testosterone ratio in serum and CLS (lowest versus highest tertile, OR 0.18, 95% CI 0.04–0.72). Conclusions CLS were more frequently identified in the breast fat of obese women and were associated with increased ratios of select estrogens:androgens in the blood and tissues, but not with individual hormones. Additional studies on CLS, tissue and blood hormone levels, and breast cancer risk are needed to understand and confirm these findings. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0791-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maeve Mullooly
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA. .,Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA.
| | - Hannah P Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Sarah J Nyante
- Department of Radiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Renata Cora
- Independent contractor, CT(ASCP), MB(ASCP), Stamford, CT, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | | | | | | | | | | | - Frank Z Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jonine D Figueroa
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Medical School, Teviot Place, Edinburgh, UK
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, Cancer Center and M. Sklodowska-Curie Institute of Oncology, Warsaw, Poland
| | - Melissa A Troester
- Department of Epidemiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Mark E Sherman
- Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
208
|
Au CC, Furness JB, Brown KA. Ghrelin and Breast Cancer: Emerging Roles in Obesity, Estrogen Regulation, and Cancer. Front Oncol 2017; 6:265. [PMID: 28119851 PMCID: PMC5220482 DOI: 10.3389/fonc.2016.00265] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/26/2023] Open
Abstract
Local and systemic factors have been shown to drive the growth of breast cancer cells in postmenopausal obese women, who have increased risk of estrogen receptor-positive breast cancer. Estrogens, produced locally in the breast fat by the enzyme aromatase, have an important role in promoting cancer cell proliferation. Ghrelin, a 28-amino acid peptide hormone, may also influence cancer growth. This peptide is produced in the stomach and acts centrally to regulate appetite and growth hormone release. Circulating levels of ghrelin, and its unacylated form, des-acyl ghrelin, are almost always inversely correlated with obesity, and these peptide hormones have recently been shown to inhibit adipose tissue aromatase expression. Ghrelin and des-acyl ghrelin have also been shown to be produced by some tumor cells and influence tumor growth. The ghrelin/des-acyl ghrelin–cancer axis is complex, one reason being that tumor cells have been shown to express splice variants of ghrelin, and ghrelin and des-acyl ghrelin might act at receptors other than the cognate ghrelin receptor, growth hormone secretagogue receptor 1a, in tumors. Effects of ghrelin and des-acyl ghrelin on energy homeostasis may also affect tumor development and growth. This review will summarize our current understanding of the role of ghrelin and des-acyl ghrelin in hormone-dependent cancers, breast cancer in particular.
Collapse
Affiliation(s)
- CheukMan Cherie Au
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne and Florey Institute of Neuroscience and Mental Health , Parkville, VIC , Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
209
|
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: 88] [Impact Index Per Article: 11.0] [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
|
210
|
Abstract
Obesity rates are increasing in the developed and developing world; this has implications for breast cancer risk and outcome. Areas covered: Recent advances relating to the association of obesity with breast cancer are reviewed. Expert commentary: Obesity has been associated with increased risk of postmenopausal hormone receptor positive and premenopausal triple negative breast cancer and with poor prognosis of most types of breast cancer. Obese individuals may present with breast cancer at a more advanced stage and their breast cancer may differ biologically from cancers diagnosed in nonobese women. A picture of a complex, multifactorial biology underlying the obesity-cancer link is emerging, with the identification of obesity-associated tissue and systemic changes that are cancer promoting, enhancing proliferation, invasion and metastasis. Intervention research to ascertain effects of weight loss and of pharmacologic interventions that reverse the metabolic changes of obesity is needed.
Collapse
Affiliation(s)
- Pamela J Goodwin
- a Department of Medicine, Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, University of Toronto , Toronto , Canada
| |
Collapse
|
211
|
Casagrande D, Moehlecke M, Mottin C, Rosa D, Schaan B. Cancer and Bariatric Surgery. METABOLISM AND PATHOPHYSIOLOGY OF BARIATRIC SURGERY 2017:333-342. [DOI: 10.1016/b978-0-12-804011-9.00049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
212
|
Nicholas DA, Andrieu G, Strissel KJ, Nikolajczyk BS, Denis GV. BET bromodomain proteins and epigenetic regulation of inflammation: implications for type 2 diabetes and breast cancer. Cell Mol Life Sci 2017; 74:231-243. [PMID: 27491296 PMCID: PMC5222701 DOI: 10.1007/s00018-016-2320-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/16/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022]
Abstract
Chronic inflammation drives pathologies associated with type 2 diabetes (T2D) and breast cancer. Obesity-driven inflammation may explain increased risk and mortality of breast cancer with T2D reported in the epidemiology literature. Therapeutic approaches to target inflammation in both T2D and cancer have so far fallen short of the expected improvements in disease pathogenesis or outcomes. The targeting of epigenetic regulators of cytokine transcription and cytokine signaling offers one promising, untapped approach to treating diseases driven by inflammation. Recent work has deeply implicated the Bromodomain and Extra-Terminal domain (BET) proteins, which are acetylated histone "readers", in epigenetic regulation of inflammation. This review focuses on inflammation associated with T2D and breast cancer, and the possibility of targeting BET proteins as an approach to regulating inflammation in the clinic. Understanding inflammation in the context of BET protein regulation may provide a basis for designing promising therapeutics for T2D and breast cancer.
Collapse
Affiliation(s)
- Dequina A Nicholas
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Guillaume Andrieu
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Katherine J Strissel
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA
| | - Barbara S Nikolajczyk
- Department of Microbiology, Training Program in Inflammatory Disorders, 72 East Concord Street, K520, Boston, MA, 02118, USA
| | - Gerald V Denis
- Cancer Center, Boston University School of Medicine, 72 East Concord Street, Room K520, Boston, MA, 02118, USA.
- Section of Hematology/Oncology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, K520, Boston, MA, 02118, USA.
| |
Collapse
|
213
|
Zahid H, Simpson ER, Brown KA. Inflammation, dysregulated metabolism and aromatase in obesity and breast cancer. Curr Opin Pharmacol 2016; 31:90-96. [PMID: 27875786 DOI: 10.1016/j.coph.2016.11.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/17/2022]
Abstract
Obesity is associated with an increased risk of estrogen-dependent breast cancer after menopause. Adipose tissue undergoes important changes in obesity due to excess storage of lipids, leading to adipocyte cell death and the recruitment of macrophages. The resultant state of chronic low-grade inflammation is associated with the activation of NFkB signaling and elevated levels of aromatase, the rate-limiting enzyme in estrogen biosynthesis. This occurs not only in the visceral and subcutaneous fat, but also in the breast fat. The regulation of aromatase in the breast adipose stromal cell in response to inflammatory mediators is under the control of complex signaling pathways, including metabolic pathways involving LKB1/AMPK, p53, HIF1α and PKM2. Interventions aimed at modifying weight, including diet and exercise, are associated with changes in adipose tissue inflammation and estrogen production that are likely to impact breast cancer risk. This review will present an overview of these topics.
Collapse
Affiliation(s)
- Heba Zahid
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia; Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Centre for Endocrinology and Metabolism, Hudson Institute for Medical Research, Clayton, Victoria, Australia; Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
214
|
Tahergorabi Z, Khazaei M, Moodi M, Chamani E. From obesity to cancer: a review on proposed mechanisms. Cell Biochem Funct 2016; 34:533-545. [PMID: 27859423 DOI: 10.1002/cbf.3229] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022]
Abstract
Nowadays, obesity is considered as a serious and growing global health problem. It is documented that the overweight and obesity are major risk factors for a series of noncommunicable diseases, and in recent years, the obesity-cancer link has received much attention. Numerous epidemiological studies have shown that obesity is associated with increased risk of several cancer types, including colon, breast, endometrium, liver, kidney, esophagus, gastric, pancreatic, gallbladder, and leukemia, and can also lead to poorer treatment. We review here the epidemiological and experimental evidences for the association between obesity and cancer. Specifically, we discuss potential mechanisms focusing how dysfunctional angiogenesis, chronic inflammation, interaction of proinflammatory cytokines, endocrine hormones, and adipokines including leptin, adiponectin insulin, growth factors, estrogen, and progesterone and strikingly, cell metabolism alteration in obesity participate in tumor development and progression, resistance to chemotherapy, and targeted therapies such as antiangiogenic and immune therapies.
Collapse
Affiliation(s)
- Zoya Tahergorabi
- Department of Physiology, Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Khazaei
- Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mitra Moodi
- Social Determinants of Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Elham Chamani
- Department of Biochemistry, Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
215
|
Iyengar NM, Gucalp A, Dannenberg AJ, Hudis CA. Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J Clin Oncol 2016; 34:4270-4276. [PMID: 27903155 DOI: 10.1200/jco.2016.67.4283] [Citation(s) in RCA: 591] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose There is growing evidence that inflammation is a central and reversible mechanism through which obesity promotes cancer risk and progression. Methods We review recent findings regarding obesity-associated alterations in the microenvironment and the local and systemic mechanisms through which these changes support tumor growth. Results Locally, hyperadiposity is associated with altered adipose tissue function, adipocyte death, and chronic low-grade inflammation. Most individuals who are obese harbor inflamed adipose tissue, which resembles chronically injured tissue, with immune cell infiltration and remodeling. Within this distinctly altered local environment, several pathophysiologic changes are found that may promote breast and other cancers. Consistently, adipose tissue inflammation is associated with a worse prognosis in patients with breast and tongue cancers. Systemically, the metabolic syndrome, including dyslipidemia and insulin resistance, occurs in the setting of adipose inflammation and operates in concert with local mechanisms to sustain the inflamed microenvironment and promote tumor growth. Importantly, adipose inflammation and its protumor consequences can be found in some individuals who are not considered to be obese or overweight by body mass index. Conclusion The tumor-promoting effects of obesity occur at the local level via adipose inflammation and associated alterations in the microenvironment, as well as systemically via circulating metabolic and inflammatory mediators associated with adipose inflammation. Accurately characterizing the obese state and identifying patients at increased risk for cancer development and progression will likely require more precise assessments than body mass index alone. Biomarkers of adipose tissue inflammation would help to identify high-risk populations. Moreover, adipose inflammation is a reversible process and represents a novel therapeutic target that warrants further study to break the obesity-cancer link.
Collapse
Affiliation(s)
- Neil M Iyengar
- Neil M. Iyengar, Ayca Gucalp, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center; Neil M. Iyengar, Ayca Gucalp, Andrew J. Dannenberg, and Clifford A. Hudis, Weill Cornell Medical College, New York, NY
| | - Ayca Gucalp
- Neil M. Iyengar, Ayca Gucalp, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center; Neil M. Iyengar, Ayca Gucalp, Andrew J. Dannenberg, and Clifford A. Hudis, Weill Cornell Medical College, New York, NY
| | - Andrew J Dannenberg
- Neil M. Iyengar, Ayca Gucalp, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center; Neil M. Iyengar, Ayca Gucalp, Andrew J. Dannenberg, and Clifford A. Hudis, Weill Cornell Medical College, New York, NY
| | - Clifford A Hudis
- Neil M. Iyengar, Ayca Gucalp, and Clifford A. Hudis, Memorial Sloan Kettering Cancer Center; Neil M. Iyengar, Ayca Gucalp, Andrew J. Dannenberg, and Clifford A. Hudis, Weill Cornell Medical College, New York, NY
| |
Collapse
|
216
|
Simapivapan P, Boltong A, Hodge A. To what extent is alcohol consumption associated with breast cancer recurrence and second primary breast cancer?: A systematic review. Cancer Treat Rev 2016; 50:155-167. [DOI: 10.1016/j.ctrv.2016.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/10/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022]
|
217
|
Iyengar NM, Ghossein RA, Morris LG, Zhou XK, Kochhar A, Morris PG, Pfister DG, Patel SG, Boyle JO, Hudis CA, Dannenberg AJ. White adipose tissue inflammation and cancer-specific survival in patients with squamous cell carcinoma of the oral tongue. Cancer 2016; 122:3794-3802. [PMID: 27508351 DOI: 10.1002/cncr.30251] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/08/2016] [Accepted: 07/18/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Obesity is associated with increased adipose tissue in the tongue. Chronic white adipose tissue (WAT) inflammation commonly occurs in the obese. We investigated whether WAT inflammation in the tongue impacts survival in patients with squamous cell carcinoma (SCC) of the oral tongue. METHODS In a retrospective cohort study, patients with T1 and T2 SCC of the oral tongue who underwent curative-intent resection were included. Tongue WAT inflammation was defined by the presence of dead or dying adipocytes surrounded by macrophages forming crown-like structures. The primary and secondary endpoints were disease-specific survival (DSS) and overall survival (OS), respectively. Subgroup analyses were carried out in patients without lymph node involvement for whom adjuvant therapies were not indicated. RESULTS Archived tissue was available from 125 patients. The median follow-up was 55 months (range, 3-156 months). Overall, 49 of 125 patients (39%) had tongue WAT inflammation, which was associated with higher body mass index, increased tumor thickness, and vascular invasion (P < .05). The 3-year DSS rate for patients with tongue WAT inflammation was 59% (95% confidence interval [CI], 46%-76%) versus 82% (95% CI, 73%-92%) for those without inflammation. For patients without lymph node involvement for whom adjuvant therapy was not indicated (N = 70), tongue WAT inflammation was associated with shortened DSS and OS (P < .05). When adjusted for body mass index and potential prognostic covariates, the hazard ratio for DSS and OS was 5.40 (95% CI, 1.20-24.26) and 2.97 (95% CI, 1.02-8.65), respectively. CONCLUSIONS Tongue WAT inflammation is associated with worse DSS and OS in patients who have early stage SCC of the oral tongue. Cancer 2016;122:3794-3802. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Neil M Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc G Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xi K Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Amit Kochhar
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Patrick G Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David G Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Snehal G Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jay O Boyle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | | |
Collapse
|
218
|
Stewart DA, Winnike JH, McRitchie SL, Clark RF, Pathmasiri WW, Sumner SJ. Metabolomics Analysis of Hormone-Responsive and Triple-Negative Breast Cancer Cell Responses to Paclitaxel Identify Key Metabolic Differences. J Proteome Res 2016; 15:3225-40. [PMID: 27447733 DOI: 10.1021/acs.jproteome.6b00430] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To date, no targeted therapies are available to treat triple negative breast cancer (TNBC), while other breast cancer subtypes are responsive to current therapeutic treatment. Metabolomics was conducted to reveal differences in two hormone receptor-negative TNBC cell lines and two hormone receptor-positive Luminal A cell lines. Studies were conducted in the presence and absence of paclitaxel (Taxol). TNBC cell lines had higher levels of amino acids, branched-chain amino acids, nucleotides, and nucleotide sugars and lower levels of proliferation-related metabolites like choline compared with Luminal A cell lines. In the presence of paclitaxel, each cell line showed unique metabolic responses, with some similarities by type. For example, in the Luminal A cell lines, levels of lactate and creatine decreased while certain choline metabolites and myo-inositol increased with paclitaxel. In the TNBC cell lines levels of glutamine, glutamate, and glutathione increased, whereas lysine, proline, and valine decreased in the presence of drug. Profiling secreted inflammatory cytokines in the conditioned media demonstrated a greater response to paclitaxel in the hormone-positive Luminal cells compared with a secretion profile that suggested greater drug resistance in the TNBC cells. The most significant differences distinguishing the cell types based on pathway enrichment analyses were related to amino acid, lipid and carbohydrate metabolism pathways, whereas several biological pathways were differentiated between the cell lines following treatment.
Collapse
Affiliation(s)
- Delisha A Stewart
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, RTI International , Research Triangle Park, North Carolina 27709, United States
| | - Jason H Winnike
- David H. Murdock Research Institute , Kannapolis, North Carolina 28081, United States
| | - Susan L McRitchie
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, RTI International , Research Triangle Park, North Carolina 27709, United States
| | - Robert F Clark
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, RTI International , Research Triangle Park, North Carolina 27709, United States
| | - Wimal W Pathmasiri
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, RTI International , Research Triangle Park, North Carolina 27709, United States
| | - Susan J Sumner
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, RTI International , Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
219
|
Strissel KJ, Nicholas DA, Castagne-Charlotin M, Ko N, Denis GV. Barriers to Obtaining Sera and Tissue Specimens of African-American Women for the Advancement of Cancer Research. CLINICAL MEDICINE INSIGHTS. WOMEN'S HEALTH 2016; 9:57-61. [PMID: 27441007 PMCID: PMC4946585 DOI: 10.4137/cmwh.s34698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 01/13/2023]
Abstract
African-American women, a historically understudied and underserved group, have increased risk for triple-negative breast cancer and obesity-associated disease. Obesity-associated metabolic diseases share a common link of low grade chronic inflammation, but not all obese women have metabolic disturbances or are inflamed. One goal of our ongoing research is to identify blood biomarkers that can predict increased risk of breast cancer in women who have obesity or metabolic dysfunction. However, vulnerable populations that stand to benefit most from advances in biomedical research are also underrepresented in research studies. The development of effective, novel approaches for cancer prevention and treatment will require significant basic medical research effort to establish the necessary evidence base in multiple populations. Work with vulnerable human subjects at a safety net hospital enabled us to comment on potential obstacles to obtaining serological and tissue specimens from African-American women. Here, we report some unexpected barriers to participation in our ongoing research study that might inform future efforts.
Collapse
Affiliation(s)
| | - Dequina A Nicholas
- Cancer Center, Boston University School of Medicine, Boston, MA, USA.; Department of Microbiology, Training Program in Inflammatory Disorders, Boston, MA, USA
| | | | - Naomi Ko
- Department of Medicine, Section of Hematology Oncology, Boston, MA, USA
| | - Gerald V Denis
- Cancer Center, Boston University School of Medicine, Boston, MA, USA.; Department of Pharmacology and Experimental Therapeutics, Section of Hematology/Oncology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
220
|
Dieli-Conwright CM, Lee K, Kiwata JL. Reducing the Risk of Breast Cancer Recurrence: an Evaluation of the Effects and Mechanisms of Diet and Exercise. CURRENT BREAST CANCER REPORTS 2016; 8:139-150. [PMID: 27909546 PMCID: PMC5112289 DOI: 10.1007/s12609-016-0218-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
With recent medical advances in diagnosis and treatment, the increasing numbers of long-term survivors of breast cancer is considerable and has resulted in the expansion of scientific research to include examination of lifestyle modifications as means of prevention of recurrence, new breast cancer events, and mortality. The objective of this report is to review randomized controlled trials (RCTs) including diet and/or exercise interventions on breast cancer recurrence in women with a history of breast cancer as well as pertinent recent epidemiologic evidence. Implicated biologic mechanisms are discussed to elucidate the impact of diet and exercise on disease recurrence.
Collapse
Affiliation(s)
- Christina M Dieli-Conwright
- Division of Biokinesiology and Physical Therapy, University of Southern California, 1540 E. Alcazar St., CHP-155, Los Angeles, CA 90033 USA
| | - Kyuwan Lee
- Division of Biokinesiology and Physical Therapy, University of Southern California, 1540 E. Alcazar St., CHP-155, Los Angeles, CA 90033 USA
| | - Jacqueline L Kiwata
- Division of Biokinesiology and Physical Therapy, University of Southern California, 1540 E. Alcazar St., CHP-155, Los Angeles, CA 90033 USA
| |
Collapse
|
221
|
Koru-Sengul T, Santander AM, Miao F, Sanchez LG, Jorda M, Glück S, Ince TA, Nadji M, Chen Z, Penichet ML, Cleary MP, Torroella-Kouri M. Breast cancers from black women exhibit higher numbers of immunosuppressive macrophages with proliferative activity and of crown-like structures associated with lower survival compared to non-black Latinas and Caucasians. Breast Cancer Res Treat 2016; 158:113-126. [PMID: 27283835 DOI: 10.1007/s10549-016-3847-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 12/16/2022]
Abstract
Racial disparities in breast cancer incidence and outcome are a major health care challenge. Patients in the black race group more likely present with an early onset and more aggressive disease. The occurrence of high numbers of macrophages is associated with tumor progression and poor prognosis in solid malignancies. Macrophages are observed in adipose tissues surrounding dead adipocytes in "crown-like structures" (CLS). Here we investigated whether the numbers of CD163+ tumor-associated macrophages (TAMs) and/or CD163+ CLS are associated with patient survival and whether there are significant differences across blacks, non-black Latinas, and Caucasians. Our findings confirm that race is statistically significantly associated with the numbers of TAMs and CLS in breast cancer, and demonstrate that the highest numbers of CD163+ TAM/CLS are found in black breast cancer patients. Our results reveal that the density of CD206 (M2) macrophages is a significant predictor of progression-free survival univariately and is also significant after adjusting for race and for HER2, respectively. We examined whether the high numbers of TAMs detected in tumors from black women were associated with macrophage proliferation, using the Ki-67 nuclear proliferation marker. Our results reveal that TAMs actively divide when in contact with tumor cells. There is a higher ratio of proliferating macrophages in tumors from black patients. These findings suggest that interventions based on targeting TAMs may not only benefit breast cancer patients in general but also serve as an approach to remedy racial disparity resulting in better prognosis patients from minority racial groups.
Collapse
Affiliation(s)
- Tulay Koru-Sengul
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ana M Santander
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue Rosenstiel Medical School Building Suite 3123A, P.O. Box 016960 (R-138), Miami, FL, 33101, USA
| | - Feng Miao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lidia G Sanchez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue Rosenstiel Medical School Building Suite 3123A, P.O. Box 016960 (R-138), Miami, FL, 33101, USA
| | - Merce Jorda
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stefan Glück
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Celgene Corporation, Summit, NJ, USA
| | - Tan A Ince
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mehrad Nadji
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue Rosenstiel Medical School Building Suite 3123A, P.O. Box 016960 (R-138), Miami, FL, 33101, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, CA, USA.,Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, CA, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.,UCLA AIDS Institute, UCLA, Los Angeles, CA, USA.,The Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Margot P Cleary
- Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Marta Torroella-Kouri
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA. .,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, 1600 NW 10th Avenue Rosenstiel Medical School Building Suite 3123A, P.O. Box 016960 (R-138), Miami, FL, 33101, USA. .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
222
|
Chen L, Cook LS, Tang MTC, Porter PL, Hill DA, Wiggins CL, Li CI. Body mass index and risk of luminal, HER2-overexpressing, and triple negative breast cancer. Breast Cancer Res Treat 2016; 157:545-54. [PMID: 27220749 DOI: 10.1007/s10549-016-3825-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/05/2016] [Indexed: 12/27/2022]
Abstract
Triple negative (TN, tumors that do not express estrogen receptor (ER), progesterone receptor (PR), or human epidermal growth factor receptor 2 (HER2)) and HER2-overexpressing (H2E, ER-/HER2+) tumors are two particularly aggressive subtypes of breast cancer. There is a lack of knowledge regarding the etiologies of these cancers and in particular how anthropometric factors are related to risk. We conducted a population-based case-case study consisting of 2659 women aged 20-69 years diagnosed with invasive breast cancer from 2004 to 2012. Four case groups defined based on joint ER/PR/HER2 status were included: TN, H2E, luminal A (ER+/HER2-), and luminal B (ER+/HER2+). Polytomous logistic regression was used to estimate odds ratios (ORs) and associated 95 % confidence intervals (CIs) where luminal A patients served as the reference group. Obese premenopausal women [body mass index (BMI) ≥30 kg/m(2)] had an 82 % (95 % CI 1.32-2.51) increased risk of TN breast cancer compared to women whose BMI <25 kg/m(2), and those in the highest weight quartile (quartiles were categorized based on the distribution among luminal A patients) had a 79 % (95 % CI 1.23-2.64) increased risk of TN disease compared to those in the lowest quartile. Among postmenopausal women obesity was associated with reduced risks of both TN (OR = 0.74, 95 % CI 0.54-1.00) and H2E (OR = 0.47, 95 % CI 0.32-0.69) cancers. Our results suggest obesity has divergent impacts on risk of aggressive subtypes of breast cancer in premenopausal versus postmenopausal women, which may contribute to the higher incidence rates of TN cancers observed among younger African American and Hispanic women.
Collapse
Affiliation(s)
- Lu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Mail Stop M4-C308, P.O. Box 19024, Seattle, WA, 98109, USA.
| | - Linda S Cook
- Department of Internal Medicine, University of New Mexico and the University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Mei-Tzu C Tang
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Mail Stop M4-C308, P.O. Box 19024, Seattle, WA, 98109, USA
| | - Peggy L Porter
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Mail Stop M4-C308, P.O. Box 19024, Seattle, WA, 98109, USA.,Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Deirdre A Hill
- Department of Internal Medicine, University of New Mexico and the University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Charles L Wiggins
- Department of Internal Medicine, University of New Mexico and the University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Christopher I Li
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Mail Stop M4-C308, P.O. Box 19024, Seattle, WA, 98109, USA
| |
Collapse
|
223
|
Seo BR, Bhardwaj P, Choi S, Gonzalez J, Andresen Eguiluz RC, Wang K, Mohanan S, Morris PG, Du B, Zhou XK, Vahdat LT, Verma A, Elemento O, Hudis CA, Williams RM, Gourdon D, Dannenberg AJ, Fischbach C. Obesity-dependent changes in interstitial ECM mechanics promote breast tumorigenesis. Sci Transl Med 2016; 7:301ra130. [PMID: 26290412 DOI: 10.1126/scitranslmed.3010467] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity and extracellular matrix (ECM) density are considered independent risk and prognostic factors for breast cancer. Whether they are functionally linked is uncertain. We investigated the hypothesis that obesity enhances local myofibroblast content in mammary adipose tissue and that these stromal changes increase malignant potential by enhancing interstitial ECM stiffness. Indeed, mammary fat of both diet- and genetically induced mouse models of obesity were enriched for myofibroblasts and stiffness-promoting ECM components. These differences were related to varied adipose stromal cell (ASC) characteristics because ASCs isolated from obese mice contained more myofibroblasts and deposited denser and stiffer ECMs relative to ASCs from lean control mice. Accordingly, decellularized matrices from obese ASCs stimulated mechanosignaling and thereby the malignant potential of breast cancer cells. Finally, the clinical relevance and translational potential of our findings were supported by analysis of patient specimens and the observation that caloric restriction in a mouse model reduces myofibroblast content in mammary fat. Collectively, these findings suggest that obesity-induced interstitial fibrosis promotes breast tumorigenesis by altering mammary ECM mechanics with important potential implications for anticancer therapies.
Collapse
Affiliation(s)
- Bo Ri Seo
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Siyoung Choi
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jacqueline Gonzalez
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Karin Wang
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sunish Mohanan
- Department of Biological and Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Patrick G Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Baoheng Du
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xi K Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, NY 10065, USA
| | - Linda T Vahdat
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Akanksha Verma
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rebecca M Williams
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Delphine Gourdon
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Claudia Fischbach
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA. Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
224
|
Troester MA, Hoadley KA, D'Arcy M, Cherniack AD, Stewart C, Koboldt DC, Robertson AG, Mahurkar S, Shen H, Wilkerson MD, Sandhu R, Johnson NB, Allison KH, Beck AH, Yau C, Bowen J, Sheth M, Hwang ES, Perou CM, Laird PW, Ding L, Benz CC. DNA defects, epigenetics, and gene expression in cancer-adjacent breast: a study from The Cancer Genome Atlas. NPJ Breast Cancer 2016; 2:16007. [PMID: 28721375 PMCID: PMC5515343 DOI: 10.1038/npjbcancer.2016.7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 01/04/2016] [Accepted: 02/18/2016] [Indexed: 12/11/2022] Open
Abstract
Recurrence rates after breast-conserving therapy may depend on genomic characteristics of cancer-adjacent, benign-appearing tissue. Studies have not evaluated recurrence in association with multiple genomic characteristics of cancer-adjacent breast tissue. To estimate the prevalence of DNA defects and RNA expression subtypes in cancer-adjacent, benign-appearing breast tissue at least 2 cm from the tumor margin, cancer-adjacent, pathologically well-characterized, benign-appearing breast tissue specimens from The Cancer Genome Atlas project were analyzed for DNA sequence, copy-number variation, DNA methylation, messenger RNA (mRNA) sequence, and mRNA/microRNA expression. Additional samples were also analyzed by at least one of these genomic data types and associations between genomic characteristics of normal tissue and overall survival were assessed. Approximately 40% of cancer-adjacent, benign-appearing tissues harbored genomic defects in DNA copy number, sequence, methylation, or in RNA sequence, although these defects did not significantly predict 10-year overall survival. Two mRNA/microRNA expression phenotypes were observed, including an active mRNA subtype that was identified in 40% of samples. Controlling for tumor characteristics and the presence of genomic defects, this active subtype was associated with significantly worse 10-year survival among estrogen receptor (ER)-positive cases. This multi-platform analysis of breast cancer-adjacent samples produced genomic findings consistent with current surgical margin guidelines, and provides evidence that extratumoral RNA expression patterns in cancer-adjacent tissue predict overall survival among patients with ER-positive disease.
Collapse
Affiliation(s)
- Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Monica D'Arcy
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew D Cherniack
- The Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Chip Stewart
- The Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel C Koboldt
- The McDonnell Genome Institute, Washington University, St Louis, MO, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada
| | - Swapna Mahurkar
- USC Epigenome Center, University of Southern California, Los Angeles, CA, USA
| | - Hui Shen
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Matthew D Wilkerson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rupninder Sandhu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicole B Johnson
- Department of Pathology, Division of Anatomical Pathology, Beth Isreal Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew H Beck
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Christina Yau
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Jay Bowen
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Margi Sheth
- National Cancer Institute, Rockville, MD, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Comprehensive Cancer Center, Durham, NC, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Li Ding
- The McDonnell Genome Institute, Washington University, St Louis, MO, USA.,Department of Genetics, Washington University, St Louis, MO, USA
| | | |
Collapse
|
225
|
Iyengar NM, Zhou XK, Gucalp A, Morris PG, Howe LR, Giri DD, Morrow M, Wang H, Pollak M, Jones LW, Hudis CA, Dannenberg AJ. Systemic Correlates of White Adipose Tissue Inflammation in Early-Stage Breast Cancer. Clin Cancer Res 2016; 22:2283-9. [PMID: 26712688 PMCID: PMC4854755 DOI: 10.1158/1078-0432.ccr-15-2239] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/16/2015] [Indexed: 02/06/2023]
Abstract
PURPOSE Obesity, insulin resistance, and elevated levels of circulating proinflammatory mediators are associated with poorer prognosis in early-stage breast cancer. To investigate whether white adipose tissue (WAT) inflammation represents a potential unifying mechanism, we examined the relationship between breast WAT inflammation and the metabolic syndrome and its prognostic importance. EXPERIMENTAL DESIGN WAT inflammation was defined by the presence of dead/dying adipocytes surrounded by macrophages forming crown-like structures (CLS) of the breast. Two independent groups were examined in cross-sectional (cohort 1) and retrospective (cohort 2) studies. Cohort 1 included 100 women undergoing mastectomy for breast cancer risk reduction (n = 10) or treatment (n = 90). Metabolic syndrome-associated circulating factors were compared by CLS-B status. The association between CLS of the breast and the metabolic syndrome was validated in cohort 2, which included 127 women who developed metastatic breast cancer. Distant recurrence-free survival (dRFS) was compared by CLS-B status. RESULTS In cohorts 1 and 2, breast WAT inflammation was detected in 52 of 100 (52%) and 52 of 127 (41%) patients, respectively. Patients with breast WAT inflammation had elevated insulin, glucose, leptin, triglycerides, C-reactive protein, and IL6 and lower high-density lipoprotein cholesterol and adiponectin (P < 0.05) in cohort 1. In cohort 2, breast WAT inflammation was associated with hyperlipidemia, hypertension, and diabetes (P < 0.05). Compared with patients without breast WAT inflammation, the adjusted HR for dRFS was 1.83 (95% CI, 1.07-3.13) for patients with inflammation. CONCLUSIONS WAT inflammation, a clinically occult process, helps to explain the relationship between metabolic syndrome and worse breast cancer prognosis. Clin Cancer Res; 22(9); 2283-9. ©2015 AACR.
Collapse
Affiliation(s)
- Neil M Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Ayca Gucalp
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Patrick G Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Louise R Howe
- Department of Cell & Developmental Biology, Weill Cornell Medical College, New York, New York
| | - Dilip D Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hanhan Wang
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Michael Pollak
- Departments of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | - Lee W Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | | |
Collapse
|
226
|
Abstract
As prevalence of obesity continues to rise in the United States, we are beginning to elucidate the complex role of obesity-associated chronic inflammation, endocrine dysfunction, and hormone production as a driver for increased breast cancer risk. Epidemiological data suggest that obesity (BMI > 30) is associated with increased breast cancer incidence, worse prognosis, and higher mortality rates. Mechanistically, obesity and excess fat mass represent a state of chronic inflammation, insulin resistance, adipokine imbalance, and increased estrogen signaling. This pro-tumorigenic environment stimulates cancer development through abnormal growth, proliferation, and survival of mammary tissue. Importantly, obesity is a modifiable risk factor; alterations in cell proliferation, apoptosis, circulating estrogen, and insulin sensitivity are observed in response to weight loss attainable through behavior modification including dietary and exercise changes.
Collapse
Affiliation(s)
- Victoria M Gershuni
- Department of Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA 19104, USA
| | - Rexford S Ahima
- Department of Endocrinology, Diabetes and Metabolism, Division, Hospital of the University of Pennsylvania, 3400, Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Julia Tchou
- Department of Surgery Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, Perelman Center for Advanced Medicine, 34th & Civic Center Blvd, 3 South, Philadelphia, PA 19104, USA
| |
Collapse
|
227
|
Flote VG, Vettukattil R, Bathen TF, Egeland T, McTiernan A, Frydenberg H, Husøy A, Finstad SE, Lømo J, Garred Ø, Schlichting E, Wist EA, Thune I. Lipoprotein subfractions by nuclear magnetic resonance are associated with tumor characteristics in breast cancer. Lipids Health Dis 2016; 15:56. [PMID: 26970778 PMCID: PMC4789271 DOI: 10.1186/s12944-016-0225-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/08/2016] [Indexed: 12/25/2022] Open
Abstract
Background High-Density Lipoprotein (HDL)-cholesterol, has been associated with breast cancer development, but the association is under debate, and whether lipoprotein subfractions is associated with breast tumor characteristics remains unclear. Methods Among 56 women with newly diagnosed invasive breast cancer stage I/II, aged 35–75 years, pre-surgery overnight fasting serum concentrations of lipids were assessed, and body mass index (BMI) was measured. All breast tumors were immunohistochemically examined in the surgical specimen. Serum metabolomics of lipoprotein subfractions and their contents of cholesterol, free cholesterol, phospholipids, apolipoprotein-A1 and apolipoprotein-A2, were assessed using nuclear magnetic resonance. Principal component analysis, partial least square analysis, and uni- and multivariable linear regression models were used to study whether lipoprotein subfractions were associated with breast cancer tumor characteristics. Results The breast cancer patients had following means: age at diagnosis: 55.1 years; BMI: 25.1 kg/m2; total-Cholesterol: 5.74 mmol/L; HDL-Cholesterol: 1.78 mmol/L; Low-Density Lipoprotein (LDL)-Cholesterol: 3.45 mmol/L; triglycerides: 1.18 mmol/L. The mean tumor size was 16.4 mm, and the mean Ki67 hotspot index was 26.5 %. Most (93 %) of the patients had estrogen receptor (ER) positive tumors (≥1 % ER+), and 82 % had progesterone receptor (PgR) positive tumors (≥10 % PgR+). Several HDL subfraction contents were strongly associated with PgR expression: Apolipoprotein-A1 (β 0.46, CI 0.22–0.69, p < 0.001), HDL cholesterol (β 0.95, CI 0.51–1.39, p < 0.001), HDL free cholesterol (β 2.88, CI 1.28–4.48, p = 0.001), HDL phospholipids (β 0.70, CI 0.36–1.04, p < 0.001). Similar results were observed for the subfractions of HDL1-3. We observed inverse associations between HDL phospholipids and Ki67 (β -0.25, p = 0.008), and in particular between HDL1’s contents of cholesterol, phospholipids, apolipoprotein-A1, apolipoprotein-A2 and Ki67. No association was observed between lipoproteins and ER expression. Conclusion Our findings hypothesize associations between different lipoprotein subfractions, and PgR expression, and Ki 67 % in breast tumors. These findings may have clinical implications, but require confirmation in larger studies. Electronic supplementary material The online version of this article (doi:10.1186/s12944-016-0225-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vidar G Flote
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway.
| | - Riyas Vettukattil
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thore Egeland
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432, Aas, Norway
| | - Anne McTiernan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hanne Frydenberg
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Anders Husøy
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Sissi E Finstad
- Norwegian Directorate of Health, PO Box 7000, St. Olavs plass, N-0130, Oslo, Norway
| | - Jon Lømo
- Department of Pathology, Oslo University Hospital, N-0424, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, N-0424, Oslo, Norway
| | - Ellen Schlichting
- Department of Breast and Endocrine Surgery, Oslo University Hospital, N-0424, Oslo, Norway
| | - Erik A Wist
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway
| | - Inger Thune
- The Cancer Centre, Oslo University Hospital HF, N-0424, Oslo, Norway.,Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, N-9037, Tromsø, Norway
| |
Collapse
|
228
|
Woodward WA. Inflammatory breast cancer: unique biological and therapeutic considerations. Lancet Oncol 2016; 16:e568-e576. [PMID: 26545845 DOI: 10.1016/s1470-2045(15)00146-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/29/2015] [Accepted: 07/06/2015] [Indexed: 12/29/2022]
Abstract
Through the concerted efforts of many patients, health-care providers, legislators, and other supporters, the past decade has seen the development of the first clinics dedicated to the care of patients with inflammatory breast cancer in the USA and other countries. Together with social networking, advocacy, and education, a few specialised centres have had substantial increases in patient numbers (in some cases ten times higher), which has further expanded the community of science and advocacy and increased the understanding of the disease process. Although inflammatory breast cancer is considered rare, constituting only 2-4% of breast cancer cases, poor prognosis means that patients with the disease account for roughly 10% of breast cancer mortality annually in the USA. I propose that the unique presentation of inflammatory breast cancer might require specific, identifiable changes in the breast parenchyma that occur before the tumour-initiating event. This would make the breast tissue itself a tumour-promoting medium that should be treated as a component of the pathology in multidisciplinary treatment and should be further studied for complementary targets to inhibit the pathobiology that is specific to inflammatory breast cancer.
Collapse
Affiliation(s)
- Wendy A Woodward
- Department of Radiation Oncology and MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
229
|
Fentiman IS. Perioperative progesterone for obese women with breast cancer may improve survival. ACTA ACUST UNITED AC 2016; 12:179-84. [PMID: 26935012 DOI: 10.2217/whe.15.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Ian S Fentiman
- Department of Surgical Oncology, Guy's Hospital, London, SE1 9RT, UK
| |
Collapse
|
230
|
Fentiman IS. Industry is not the dark side, but an essential partner to make progress in reproductive health. WOMEN'S HEALTH (LONDON, ENGLAND) 2016; 12:175-8. [PMID: 26939018 PMCID: PMC5375047 DOI: 10.2217/whe.15.106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/20/2015] [Indexed: 01/08/2023]
Abstract
For the last 20 years, Thomas D'Hooghe has been coordinator of the Leuven University Fertility Center at Leuven University Hospitals, Belgium, one of the largest teaching hospitals in Europe. Since 1995, he has also been Professor of Reproductive Medicine and Biology at KU Leuven (University of Leuven) and Adjunct Professor at Yale University, USA. Since 1 October 2015, he has been the Vice President and Head of Global Medical Affairs Fertility at Merck's headquarters in Darmstadt, Germany. He has published nearly 300 papers in internationally peer-reviewed journals and has contributed to reproductive health serving major international organizations such as the WHO, the European Society of Human Reproduction and Embryology, the Society of Reproductive Investigation and the World Endometriosis Research Foundation.
Collapse
Affiliation(s)
- Ian S Fentiman
- Department of Surgical Oncology, Guy's Hospital, London, SE1 9RT, UK
| |
Collapse
|
231
|
Robust Early Inflammation of the Peripancreatic Visceral Adipose Tissue During Diet-Induced Obesity in the KrasG12D Model of Pancreatic Cancer. Pancreas 2016; 45:458-65. [PMID: 26495779 PMCID: PMC4854638 DOI: 10.1097/mpa.0000000000000497] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Obesity increases the incidence of multiple types of cancer. Our previous work has shown that a high-fat, high-calorie diet (HFCD) leads to visceral obesity, pancreatic inflammation, and accelerated pancreatic neoplasia in KrasG12D (KC) mice. In this study, we aimed to investigate the effects of an HFCD on visceral adipose inflammation with emphasis on potential differences between distinct visceral adipose depots. METHODS We examined the weight and visceral obesity in both wild-type and KC mice on either control diet (CD) or HFCD. After 3 months, mice were killed for histological examination. Multiplex assays were also performed to obtain cytokine profiles between different adipose depots. RESULTS Both wild-type and KC mice on an HFCD exhibited significantly increased inflammation in the visceral adipose tissue, particularly in the peripancreatic fat (PPF), compared with animals on a CD. This was associated with significantly increased inflammation in the pancreas. Cytokine profiles were different between visceral adipose depots and between mice on the HFCD and CD. CONCLUSIONS Our results clearly demonstrate that an HFCD leads to obesity and inflammation in the visceral adipose tissue, particularly the PPF. These data suggest that obesity-associated inflammation in PPF may accelerate pancreatic neoplasia in KC mice.
Collapse
|
232
|
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
|
233
|
Haka AS, Sue E, Zhang C, Bhardwaj P, Sterling J, Carpenter C, Leonard M, Manzoor M, Walker J, Aleman JO, Gareau D, Holt PR, Breslow JL, Zhou XK, Giri D, Morrow M, Iyengar N, Barman I, Hudis CA, Dannenberg AJ. Noninvasive Detection of Inflammatory Changes in White Adipose Tissue by Label-Free Raman Spectroscopy. Anal Chem 2016; 88:2140-8. [PMID: 26752499 DOI: 10.1021/acs.analchem.5b03696] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
White adipose tissue inflammation (WATi) has been linked to the pathogenesis of obesity-related diseases, including type 2 diabetes, cardiovascular disease, and cancer. In addition to the obese, a substantial number of normal and overweight individuals harbor WATi, putting them at increased risk for disease. We report the first technique that has the potential to detect WATi noninvasively. Here, we used Raman spectroscopy to detect WATi with excellent accuracy in both murine and human tissues. This is a potentially significant advance over current histopathological techniques for the detection of WATi, which rely on tissue excision and, therefore, are not practical for assessing disease risk in the absence of other identifying factors. Importantly, we show that noninvasive Raman spectroscopy can diagnose WATi in mice. Taken together, these results demonstrate the potential of Raman spectroscopy to provide objective risk assessment for future cardiometabolic complications in both normal weight and overweight/obese individuals.
Collapse
Affiliation(s)
- Abigail S Haka
- Department of Biochemistry, Weill Cornell Medical College , New York, New York 10065, United States
| | - Erika Sue
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| | - Chi Zhang
- Department of Mechanical Engineering, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| | - Joshua Sterling
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| | - Cassidy Carpenter
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| | - Madeline Leonard
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| | - Maryem Manzoor
- Department of Biochemistry, Weill Cornell Medical College , New York, New York 10065, United States
| | - Jeanne Walker
- Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University , New York, New York 10065, United States
| | - Jose O Aleman
- Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University , New York, New York 10065, United States
| | - Daniel Gareau
- Department of Investigative Dermatology, The Rockefeller University , New York, New York 10065, United States
| | - Peter R Holt
- Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University , New York, New York 10065, United States
| | - Jan L Breslow
- Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University , New York, New York 10065, United States
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College , New York, New York 10065, United States
| | - Dilip Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Monica Morrow
- Department of Surgery, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Neil Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University , Baltimore, Maryland 21218, United States.,Department of Oncology, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Clifford A Hudis
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States.,Department of Medicine, Memorial Sloan Kettering Cancer Center , New York, New York 10065, United States
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medical College , New York, New York 10065, United States
| |
Collapse
|
234
|
|
235
|
Epidemiological risk factors associated with inflammatory breast cancer subtypes. Cancer Causes Control 2016; 27:359-66. [PMID: 26797453 DOI: 10.1007/s10552-015-0712-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 12/30/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND In this single-institution case-control study, we identified risk factors associated with inflammatory breast cancer (IBC) subtypes based on staining of estrogen receptor (ER), progesterone receptor (PR) and expression of human epidermal growth factor 2 (HER2neu) to determine distinct etiologic pathways. METHODS We identified 224 women with IBC and 396 cancer-free women seen at the MD Anderson Cancer Center. Multinomial logistic regression was used to estimate odds ratios (ORs) and 95 % confidence intervals (CIs) for associations between breast cancer risk factors and the IBC tumor subtypes: luminal (ER+ and/or PR+/HER2neu-), HER2neu+ (any ER and PR, HER2neu+), and triple-negative (ER-/PR-/HER2neu-). RESULTS In multivariable analysis, compared with women age ≥26 at first pregnancy, women age <26 had a higher risk of triple-negative IBC (OR 3.32, 95% CI 1.37-8.05). Women with a history of breast-feeding had a lower risk of triple-negative (OR 0.30; 95% CI 0.15-0.62) and luminal IBC (OR 0.35, 95% CI 0.18-0.68). A history of smoking was associated with an increased risk of luminal IBC (OR 2.37; 95% CI 1.24-4.52). Compared with normal-weight women, those who were overweight or obese (body mass index ≥25 kg/m(2)) had a higher risk of all three tumor subtypes (p < 0.01 for all subtypes). CONCLUSION Overweight or obese status is important modifiable risk factor for IBC of any subtype. Modifiable risk factors, age at first pregnancy (≥26), breast-feeding, and smoking may be associated with specific IBC subtypes. These results highlight the importance of evaluating epidemiologic risk factors for IBC for the identification of subtype-specific prevention strategies.
Collapse
|
236
|
Williams CB, Yeh ES, Soloff AC. Tumor-associated macrophages: unwitting accomplices in breast cancer malignancy. NPJ Breast Cancer 2016; 2:15025. [PMID: 26998515 PMCID: PMC4794275 DOI: 10.1038/npjbcancer.2015.25] [Citation(s) in RCA: 331] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 12/12/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023] Open
Abstract
Deleterious inflammation is a primary feature of breast cancer. Accumulating evidence demonstrates that macrophages, the most abundant leukocyte population in mammary tumors, have a critical role at each stage of cancer progression. Such tumor-associated macrophages facilitate neoplastic transformation, tumor immune evasion and the subsequent metastatic cascade. Herein, we discuss the dynamic process whereby molecular and cellular features of the tumor microenvironment act to license tissue-repair mechanisms of macrophages, fostering angiogenesis, metastasis and the support of cancer stem cells. We illustrate how tumors induce, then exploit trophic macrophages to subvert innate and adaptive immune responses capable of destroying malignant cells. Finally, we discuss compelling evidence from murine models of cancer and early clinical trials in support of macrophage-targeted intervention strategies with the potential to dramatically reduce breast cancer morbidity and mortality.
Collapse
Affiliation(s)
- Carly Bess Williams
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Adam C Soloff
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
237
|
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
|
238
|
|
239
|
Ben-Shmuel S, Scheinman EJ, Rashed R, Orr ZS, Gallagher EJ, LeRoith D, Rostoker R. Ovariectomy is associated with metabolic impairments and enhanced mammary tumor growth in MKR mice. J Endocrinol 2015; 227:143-151. [PMID: 26383532 PMCID: PMC4618719 DOI: 10.1530/joe-15-0310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2015] [Indexed: 12/29/2022]
Abstract
Obesity and type 2 diabetes (T2D) are associated with an increased risk of breast cancer incidence and mortality. Common features of obesity and T2D are insulin resistance and hyperinsulinemia. A mammary tumor promoting effect of insulin resistance and hyperinsulinemia was demonstrated in the transgenic female MKR mouse model of pre-diabetes inoculated with mammary cancer cells. Interestingly, in MKR mice, as well as in other diabetic mouse models, males exhibit severe hyperglycemia, while females display insulin resistance and hyperinsulinemia with only a mild increase in blood glucose levels. This gender-specific protection from hyperglycemia may be attributed to estradiol, a key player in the regulation of the metabolic state, including obesity, glucose homeostasis, insulin resistance, and lipid profile. The aim of this study was to investigate the effects of ovariectomy (including the removal of endogenous estradiol) on the metabolic state of MKR female mice and subsequently on the growth of Mvt-1 mammary cancer cells, inoculated into the mammary fat pad of ovariectomized mice, compared with sham-operated mice. The results showed an increase in body weight, accompanied by increased fat mass, elevated blood glucose levels, and hypercholesterolemia, in ovariectomized MKR mice. In addition, mammary tumor growth was significantly higher in these mice. The results suggest that ovarian hormone deficiency may promote impaired metabolic homeostasis in the hyperinsulinemic MKR female mice, which in turn is associated with an increased growth of mammary tumors.
Collapse
Affiliation(s)
- Sarit Ben-Shmuel
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
| | - Eyal J. Scheinman
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
| | - Rola Rashed
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
| | - Zila Shen Orr
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1055, Atran 4-36, New York, NY 10029, USA
| | - Derek LeRoith
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1055, Atran 4-36, New York, NY 10029, USA
- Corresponding author: Derek LeRoith, Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa, 31096, Israel.
| | - Ran Rostoker
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Reseasch Institute at Rambam (CRIR), Rambam Medical Center, P.O.B 9602, Haifa 31096, Israel
| |
Collapse
|
240
|
Argolo DF, Iyengar NM, Hudis CA. Obesity and Cancer: Concepts and Challenges. Indian J Surg Oncol 2015; 6:390-8. [PMID: 27081257 DOI: 10.1007/s13193-015-0483-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/17/2015] [Indexed: 12/17/2022] Open
Abstract
The rates of overweight and obesity are increasing worldwide in both developed and developing countries. Obesity is a major public health problem as it is associated with many diseases, including diabetes, hypertension, dyslipidemia, atherosclerosis, and some types of cancer. Breast cancer is a malignancy in which both the risk of development and the prognosis are negatively impacted by the obese state. The precise mechanisms pathophysiologically linking obesity and cancer are still under investigation. The biological basis for these associations includes both systemic and local tissue effects and white adipose tissue inflammation appears to be a critical component. A comprehensive understanding of the mechanisms linking obesity, inflammation and cancer may provide an opportunity for the development of strategies to attenuate the negative impact of obesity.
Collapse
Affiliation(s)
- Daniel F Argolo
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; CLION - CAM Group, Salvador, BA Brazil
| | - Neil M Iyengar
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; Weill Cornell Medical College, New York, NY USA
| | - Clifford A Hudis
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street - 8th floor, New York, NY 10065 USA ; Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
241
|
Iyengar NM, Hudis CA, Dannenberg AJ. Obesity and inflammation: new insights into breast cancer development and progression. Am Soc Clin Oncol Educ Book 2015:46-51. [PMID: 23714453 DOI: 10.14694/edbook_am.2013.33.46] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The importance of inflammation in promoting carcinogenesis and tumor progression is well recognized. Chronic inflammation caused by a variety of infectious agents can lead to the development of several common malignancies. Similarly, inflammatory bowel disease is a well-known risk factor for colorectal cancer. Much less is known about the link between inflammation and the development of breast cancer. Recent data suggest that obesity causes both in-breast and systemic inflammation that contribute to the development and progression of breast cancer. This observation has potentially important implications in terms of prevention and treatment of breast cancer, especially given the rising worldwide overweight and obesity rates. Inflamed white adipose tissue (WAT) within the breast is associated with elevated levels of proinflammatory mediators, enhanced expression of aromatase (the rate-limiting enzyme for estrogen biosynthesis), and increased estrogen receptor-α (ER-α)-dependent gene expression. Systemic consequences of obesity including altered adipokine levels, elevated circulating estrogen levels, and insulin resistance are also believed to play a role in the pathogenesis of breast cancer. Collectively, these findings suggest a significant role for inflammation in the pathogenesis of breast cancer in obese and overweight patients.
Collapse
Affiliation(s)
- Neil M Iyengar
- From the Memorial Sloan-Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| | | | | |
Collapse
|
242
|
Iyengar NM, Hudis CA, Dannenberg AJ. Obesity and inflammation: new insights into breast cancer development and progression. AMERICAN SOCIETY OF CLINICAL ONCOLOGY EDUCATIONAL BOOK. AMERICAN SOCIETY OF CLINICAL ONCOLOGY. ANNUAL MEETING 2015. [PMID: 23714453 DOI: 10.1200/edbook_am.2013.33.46] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The importance of inflammation in promoting carcinogenesis and tumor progression is well recognized. Chronic inflammation caused by a variety of infectious agents can lead to the development of several common malignancies. Similarly, inflammatory bowel disease is a well-known risk factor for colorectal cancer. Much less is known about the link between inflammation and the development of breast cancer. Recent data suggest that obesity causes both in-breast and systemic inflammation that contribute to the development and progression of breast cancer. This observation has potentially important implications in terms of prevention and treatment of breast cancer, especially given the rising worldwide overweight and obesity rates. Inflamed white adipose tissue (WAT) within the breast is associated with elevated levels of proinflammatory mediators, enhanced expression of aromatase (the rate-limiting enzyme for estrogen biosynthesis), and increased estrogen receptor-α (ER-α)-dependent gene expression. Systemic consequences of obesity including altered adipokine levels, elevated circulating estrogen levels, and insulin resistance are also believed to play a role in the pathogenesis of breast cancer. Collectively, these findings suggest a significant role for inflammation in the pathogenesis of breast cancer in obese and overweight patients.
Collapse
Affiliation(s)
- Neil M Iyengar
- From the Memorial Sloan-Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| | | | | |
Collapse
|
243
|
Glaser R, Dimitrakakis C. Testosterone and breast cancer prevention. Maturitas 2015; 82:291-5. [DOI: 10.1016/j.maturitas.2015.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 01/03/2023]
|
244
|
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: 93] [Impact Index Per Article: 9.3] [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
|
245
|
Leitner L, Jürets A, Itariu BK, Keck M, Prager G, Langer F, Grablowitz V, Zeyda M, Stulnig TM. Osteopontin promotes aromatase expression and estradiol production in human adipocytes. Breast Cancer Res Treat 2015; 154:63-9. [DOI: 10.1007/s10549-015-3603-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
|
246
|
Abstract
Obesity is a leading modifiable risk factor for the development of several epithelial malignancies. In addition to increasing risk, obesity also confers worse prognosis for many cancers. Obesity represents an overall state of energy imbalance frequently associated with systemic effects including insulin resistance, altered hormone signaling, and high circulating levels of proinflammatory mediators. In addition to its systemic effects, obesity causes subclinical white adipose inflammation including increased tissue levels of proinflammatory mediators. Both local and systemic effects are likely to contribute to the development and progression of cancer. An understanding of the interplay between local and systemic alterations involved in the obesity-cancer link provides the basis for developing interventions aimed at mitigating the protumorigenic effects.
Collapse
Affiliation(s)
- Neil M Iyengar
- Memorial Sloan Kettering Cancer Center, New York, NY 10065; ,
| | | | | |
Collapse
|
247
|
Gucalp A, Iyengar NM, Hudis CA, Dannenberg AJ. Targeting obesity-related adipose tissue dysfunction to prevent cancer development and progression. Semin Oncol 2015; 43:154-160. [PMID: 26970134 DOI: 10.1053/j.seminoncol.2015.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The incidence of obesity, a leading modifiable risk factor for common solid tumors, is increasing. Effective interventions are needed to minimize the public health implications of obesity. Although the mechanisms linking increased adiposity to malignancy are incompletely understood, growing evidence points to complex interactions among multiple systemic and tissue-specific pathways including inflamed white adipose tissue. The metabolic and inflammatory consequences of white adipose tissue dysfunction collectively provide a plausible explanation for the link between overweight/obesity and carcinogenesis. Gaining a better understanding of these underlying molecular pathways and developing risk assessment tools that identify at-risk populations will be critical in implementing effective and novel cancer prevention and management strategies.
Collapse
Affiliation(s)
- Ayca Gucalp
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Neil M Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
248
|
Polari L, Yatkin E, Martínez Chacón MG, Ahotupa M, Smeds A, Strauss L, Zhang F, Poutanen M, Saarinen N, Mäkelä SI. Weight gain and inflammation regulate aromatase expression in male adipose tissue, as evidenced by reporter gene activity. Mol Cell Endocrinol 2015; 412:123-30. [PMID: 26054748 DOI: 10.1016/j.mce.2015.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/11/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022]
Abstract
Obesity and white adipose tissue (WAT) inflammation are associated with enhanced aromatization in women, but little is known about the regulation of aromatase (CYP19A1) gene expression in male WAT. We investigated the impact of weight gain and WAT inflammation on the regulation of CYP19A1 in males, by utilizing the hARO-Luc aromatase reporter mouse model containing a >100-kb 5'-region of the human CYP19A1 gene. We show that hARO-Luc reporter activity is enhanced in WAT of mice with increased adiposity and inflammation. Dexamethasone and TNFα, as well as forskolin and phorbol 12-myristate 13-acetate, upregulate hARO-Luc activity, suggesting the involvement of promoters I.4 and I.3/II. Furthermore, we show that diet enriched with antioxidative plant polyphenols attenuates WAT inflammation and hARO-Luc activity in obese males. In conclusion, our data suggest that obesity-associated WAT inflammation leads to increased peripheral CYP19A1 expression in males, and that polyphenol-enriched diet may have the potential to attenuate excessive aromatization in WAT of obese men.
Collapse
Affiliation(s)
- L Polari
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - E Yatkin
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - M G Martínez Chacón
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - M Ahotupa
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - A Smeds
- Åbo Akademi University, Process Chemistry Centre, Laboratory of Wood and Paper Chemistry, Turku, Finland
| | - L Strauss
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - F Zhang
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - M Poutanen
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - N Saarinen
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland; Department of Physiology, Institute of Biomedicine, University of Turku, Finland
| | - S I Mäkelä
- Functional Foods Forum, University of Turku, Turku, Finland; Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
249
|
Wang X, Simpson ER, Brown KA. Aromatase overexpression in dysfunctional adipose tissue links obesity to postmenopausal breast cancer. J Steroid Biochem Mol Biol 2015. [PMID: 26209254 DOI: 10.1016/j.jsbmb.2015.07.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The number of breast cancer cases has increased in the last a few decades and this is believed to be associated with the increased prevalence of obesity worldwide. The risk of breast cancer increases with age beyond menopause and the relationship between obesity and the risk of breast cancer in postmenopausal women is well established. The majority of postmenopausal breast cancers are estrogen receptor (ER) positive and estrogens produced in the adipose tissue promotes tumor formation. Obesity results in the secretion of inflammatory factors that stimulate the expression of the aromatase enzyme, which converts androgens into estrogens in the adipose tissue. Evidence demonstrating a link between obesity and breast cancer has led to the investigation of metabolic pathways as novel regulators of estrogen production, including pathways that can be targeted to inhibit aromatase specifically within the breast. This review aims to present some of the key findings in this regard.
Collapse
Affiliation(s)
- Xuyi Wang
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of biochemistry & Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Metabolism & Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia; Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
250
|
Laforest S, Labrecque J, Michaud A, Cianflone K, Tchernof A. Adipocyte size as a determinant of metabolic disease and adipose tissue dysfunction. Crit Rev Clin Lab Sci 2015; 52:301-13. [DOI: 10.3109/10408363.2015.1041582] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|