1
|
Pan C, Wei Y, Dai J, Yang L, Ding Z, Xinke Wang. Knowledge mapping of metformin use on cancers: a bibliometric analysis (2013-2023). Front Pharmacol 2024; 15:1388253. [PMID: 39193327 PMCID: PMC11347356 DOI: 10.3389/fphar.2024.1388253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
There is substantial evidence from clinical and preclinical studies suggesting an association between metformin use and a reduced risk of cancer. However, the effects of metformin use on cancers have not yet been subjected to bibliometric analysis. The goal of this study was to explore the potential effects of metformin use on cancers and to conduct a comprehensive assessment of research hotspots related to the use of metformin on cancers. The results of the literature analysis were visualized using various tools such as Adobe Illustrator CC 2018, VOSviewer, CiteSpace, and the R package "bibliometric." The average annual publications from 2013 to 2023 was 372. In terms of journals and co-cited journals, a total of 1,064 journals published 1958 papers, and Oncotarget published the highest number of papers (n = 153, 7.81%), while Cancer Research (Co-citation = 5,125) was the most frequently cited journal. A total of 25,665 authors participated in the research on metformin use on cancers. Metformin has demonstrated improved outcomes in various types of cancer, including breast cancer (BC), lung cancer (LC), colorectal cancer (CRC), prostate cancer (PC), and pancreatic cancer. This bibliometric analysis reviews the current literature on the clinical data on metformin use on cancers and describes the preclinical evidence illustrating the potential mechanisms of metformin use on various cancers directly or indirectly.
Collapse
Affiliation(s)
| | | | | | | | - Zhuoyu Ding
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Engin A. Obesity-Associated Breast Cancer: Analysis of Risk Factors and Current Clinical Evaluation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:767-819. [PMID: 39287872 DOI: 10.1007/978-3-031-63657-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Additionally, obese and postmenopausal women are at higher risk of all-cause and breast cancer-specific mortality compared with non-obese women with breast cancer. In this context, increased levels of estrogens, excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, adipocyte-derived adipokines, hypercholesterolemia, and excessive oxidative stress contribute to the development of breast cancer in obese women. Genetic evaluation is an integral part of diagnosis and treatment for patients with breast cancer. Despite trimodality therapy, the four-year cumulative incidence of regional recurrence is significantly higher. Axillary lymph nodes as well as primary lesions have diagnostic, prognostic, and therapeutic significance for the management of breast cancer. In clinical setting, because of the obese population primary lesions and enlarged lymph nodes could be less palpable, the diagnosis may be challenging due to misinterpretation of physical findings. Thereby, a nomogram has been created as the "Breast Imaging Reporting and Data System" (BI-RADS) to increase agreement and decision-making consistency between mammography and ultrasonography (USG) experts. Additionally, the "breast density classification system," "artificial intelligence risk scores," ligand-targeted receptor probes," "digital breast tomosynthesis," "diffusion-weighted imaging," "18F-fluoro-2-deoxy-D-glucose positron emission tomography," and "dynamic contrast-enhanced magnetic resonance imaging (MRI)" are important techniques for the earlier detection of breast cancers and to reduce false-positive results. A high concordance between estrogen receptor (ER) and progesterone receptor (PR) status evaluated in preoperative percutaneous core needle biopsy and surgical specimens is demonstrated. Breast cancer surgery has become increasingly conservative; however, mastectomy may be combined with any axillary procedures, such as sentinel lymph node biopsy (SLNB) and/or axillary lymph node dissection whenever is required. As a rule, SLNB-guided axillary dissection in breast cancer patients who have clinically axillary lymph node-positive to node-negative conversion following neoadjuvant chemotherapy is recommended, because lymphedema is the most debilitating complication after any axillary surgery. There is no clear consensus on the optimal treatment of occult breast cancer, which is much discussed today. Similarly, the current trend in metastatic breast cancer is that the main palliative treatment option is systemic therapy.
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
|
3
|
Wang F, Giskeødegård GF, Skarra S, Engstrøm MJ, Hagen L, Geisler J, Mikkola TS, Tikkanen MJ, Debik J, Reidunsdatter RJ, Bathen TF. Association of serum cortisol and cortisone levels and risk of recurrence after endocrine treatment in breast cancer. Clin Exp Med 2023; 23:3883-3893. [PMID: 37395895 PMCID: PMC10618334 DOI: 10.1007/s10238-023-01109-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/29/2023] [Indexed: 07/04/2023]
Abstract
Metabolic reprogramming in breast cancer involves changes in steroid hormone synthesis and metabolism. Alterations in estrogen levels in both breast tissue and blood may influence carcinogenesis, breast cancer growth, and response to therapy. Our aim was to examine whether serum steroid hormone concentrations could predict the risk of recurrence and treatment-related fatigue in patients with breast cancer. This study included 66 postmenopausal patients with estrogen receptor-positive breast cancer who underwent surgery, radiotherapy, and adjuvant endocrine treatment. Serum samples were collected at six different time points [before the start of radiotherapy (as baseline), immediately after radiotherapy, and then 3, 6, 12 months, and 7-12 years after radiotherapy]. Serum concentrations of eight steroid hormones (cortisol, cortisone, 17α-hydroxyprogesterone, 17β-estradiol, estrone, androstenedione, testosterone, and progesterone) were measured using a liquid chromatography-tandem mass spectrometry-based method. Breast cancer recurrence was defined as clinically proven relapse/metastatic breast cancer or breast cancer-related death. Fatigue was assessed with the QLQ-C30 questionnaire. Serum steroid hormone concentrations measured before and immediately after radiotherapy differed between relapse and relapse-free patients [(accuracy 68.1%, p = 0.02, and 63.2%, p = 0.03, respectively, partial least squares discriminant analysis (PLS-DA)]. Baseline cortisol levels were lower in patients who relapsed than in those who did not (p < 0.05). The Kaplan-Meier analysis showed that patients with high baseline concentrations of cortisol (≥ median) had a significantly lower risk of breast cancer recurrence than patients with low cortisol levels (
Collapse
Affiliation(s)
- Feng Wang
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.
- Department of Breast and Endocrine Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Guro F Giskeødegård
- Department of Breast and Endocrine Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Sissel Skarra
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Monica J Engstrøm
- Department of Breast and Endocrine Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Deprtment of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Lars Hagen
- Deprtment of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, Trondheim, Norway
| | - Jürgen Geisler
- Deparment of Oncology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tomi S Mikkola
- Department of Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Matti J Tikkanen
- Folkhälsan Research Center, University of Helsinki, Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Julia Debik
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Randi J Reidunsdatter
- 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.
- Department of Breast and Endocrine Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
4
|
Brown JC, Sturgeon K, Sarwer DB, Troxel AB, DeMichele AM, Denlinger CS, Schmitz KH. The effects of exercise and diet on sex steroids in breast cancer survivors. Endocr Relat Cancer 2022; 29:485-493. [PMID: 35639865 PMCID: PMC9339480 DOI: 10.1530/erc-22-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/08/2022]
Abstract
Insufficient physical activity and obesity are associated with an increased risk of cancer recurrence and death in breast cancer survivors. Sex steroid hormones may mediate these associations. This study tested the hypothesis that exercise and diet, as compared to control, favorably change sex steroid hormones. This analysis of data from a subset of participants in a 2 × 2 factorial trial compares 269 postmenopausal breast cancer survivors who were insufficiently physically active and had overweight or obesity and were randomized to one of four treatment groups for 52 weeks: control, exercise alone, diet alone, or exercise plus diet. Secondary sex steroid hormone endpoints included estradiol, sex hormone-binding globulin (SHBG), and testosterone. Treatment effects were quantified using a mixed model for repeated measures. Compared to control, exercise alone did not significantly change estradiol (-1.9%; 95% CI: -12.6, 8.8), SHBG (2.4%; 95% CI: -9.9, 14.6), or testosterone (1.2%; 95% CI: -12.2, 14.5). Compared to control, diet alone did not significantly change estradiol (-7.8%; 95% CI: -17.6, 1.9), SHBG (8.2%; 95% CI: -4.2, 20.6), or testosterone (-0.8%; 95% CI: -13.6, 12.0). Compared to control, exercise plus diet did not significantly change estradiol (-6.3%; 95% CI: -16.3, 3.6), SHBG (8.8%; 95% CI: -4.0, 21.7), or testosterone (-5.3%; 95% CI: -18.0, 7.4). In postmenopausal breast cancer survivors who were insufficiently physically active and had overweight or obesity, randomization to exercise alone, diet alone, or exercise plus diet did not statistically significantly change sex steroid hormone concentrations at week 52.
Collapse
Affiliation(s)
- Justin C. Brown
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, U.S.A
- LSU Health Sciences Center New Orleans School of Medicine, 1901 Perdido St, New Orleans, LA 70112, U.S.A
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 533 Bolivar St, New Orleans, LA, 70112, U.S.A
| | - Kathleen Sturgeon
- Penn State College of Medicine, 400 University Drive, Hershey, PA 17033, U.S.A
| | - David B. Sarwer
- Temple University College of Public Health, 1101 W. Montgomery Ave, Philadelphia, PA 1912, U.S.A
| | - Andrea B. Troxel
- New York University Grossman School of Medicine, 180 Madison Ave, New York, NY 10016, U.S.A
| | - Angela M. DeMichele
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, U.S.A
| | | | - Kathryn H. Schmitz
- Penn State College of Medicine, 400 University Drive, Hershey, PA 17033, U.S.A
| |
Collapse
|
5
|
Li J, Li C, Feng Z, Liu L, Zhang L, Kang W, Liu Y, Ma B, Li H, Huang Y, Zheng H, Song F, Song F, Chen K. Effect of estradiol as a continuous variable on breast cancer survival by menopausal status: a cohort study in China. Breast Cancer Res Treat 2022; 194:103-111. [PMID: 35467315 DOI: 10.1007/s10549-022-06593-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/31/2022] [Indexed: 11/26/2022]
Abstract
High levels of circulating estradiol (E2) are associated with increased risk of breast cancer, whereas its relationship with breast cancer prognosis is still unclear. We evaluated the effect of E2 concentration on survival endpoints among 8766 breast cancer cases diagnosed between 2005 and 2017 from the Tianjin Breast Cancer Cases Cohort. Levels of serum E2 were measured in pre-menopausal and post-menopausal women. Multivariable-adjusted Cox proportional hazards models were used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI) between quartile of E2 levels and overall survival (OS) and progression-free survival (PFS) of breast cancer. The penalized spline was then used to test for non-linear relationships between E2 (continuous variable) and survival endpoints. 612 deaths and 982 progressions occurred over follow-up through 2017. Compared to women in the quartile 3, the highest quartile of E2 was associated with reduced risk of both PFS in pre-menopausal women (HR 1.79, 95% CI 1.17-2.75, P = 0.008) and OS in post-menopausal women (HR 1.35, 95% CI 1.04-1.74, P = 0.023). OS and PFS in pre-menopausal women exhibited a nonlinear relation ("L-shaped" and "U-shaped", respectively) with E2 levels. However, there was a linear relationship in post-menopausal women. Moreover, patients with estrogen receptor-negative (ER-negative) breast cancer showed a "U-shaped" relationship with OS and PFS in pre-menopausal women. Pre-menopausal breast cancer patients have a plateau stage of prognosis at the intermediate concentrations of E2, whereas post-menopausal patients have no apparent threshold, and ER status may have an impact on this relationship.
Collapse
Affiliation(s)
- Junxian Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Chenyang Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Ziwei Feng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Luyang Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Liwen Zhang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Wenjuan Kang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Ya Liu
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Baoshan Ma
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Haixin Li
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Yubei Huang
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China.
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Key Laboratory of Molecular Cancer Epidemiology in Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huanhu Xi Road, Tiyuan Bei, Hexi District, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
6
|
Pre-treatment circulating reproductive hormones levels predict pathological and survival outcomes in breast cancer submitted to neoadjuvant chemotherapy. Int J Clin Oncol 2022; 27:899-910. [PMID: 35239089 DOI: 10.1007/s10147-022-02141-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/03/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE This study aimed to evaluate the correlation of pre-treatment circulating reproductive hormones levels with pathological and survival outcomes in breast cancer patients received neoadjuvant chemotherapy (NAC). METHODS Information from 196 premenopausal and 137 postmenopausal breast cancer patients who received NAC were retrospectively analyzed. Treatment response to NAC, with odds ratios (OR) and 95% confidence intervals (95% CI) was estimated using logistic regression adjusted for key confounders. Survival outcomes with hazard ratios (HR) and 95% CI were estimated using Cox regression adjusted for key confounders. The Kaplan-Meier method was applied in the survival analysis. RESULTS Premenopausal patients with lower testosterone levels (OR = 0.996, 95% CI 0.992-0.999, P = 0.026), and postmenopausal patients with higher follicle-stimulating hormone (FSH) levels (OR = 1.045, 95% CI 1.014-1.077, P = 0.005) were likely to achieve pathological complete response (pCR). In multivariate survival analysis, the lowest tertile (T) progesterone was associated with worse overall survival (OS) in premenopausal patients (T2 vs T1, HR = 0.113, 95% CI 0.013-0.953, P = 0.045; T3 vs T1, HR = 0.109, 95% CI 0.013-0.916, P = 0.041). Premenopausal patients with the lowest tertile progesterone exhibited worse 3-year OS compared with those with higher tertiles (72.9% vs 97.4%, log-rank, P = 0.007). CONCLUSION Pre-treatment testosterone and FSH are significant independent predictors for pCR to NAC in premenopausal and postmenopausal patients, respectively. Low progesterone levels are correlated with worse OS in premenopausal patients. These findings may provide a theoretical basis for pre-operative endocrine therapy combined with NAC in breast cancer.
Collapse
|
7
|
Febvey-Combes O, Jobard E, Rossary A, Pialoux V, Foucaut AM, Morelle M, Delrieu L, Martin A, Caldefie-Chézet F, Touillaud M, Berthouze SE, Boumaza H, Elena-Herrmann B, Bachmann P, Trédan O, Vasson MP, Fervers B. Effects of an Exercise and Nutritional Intervention on Circulating Biomarkers and Metabolomic Profiling During Adjuvant Treatment for Localized Breast Cancer: Results From the PASAPAS Feasibility Randomized Controlled Trial. Integr Cancer Ther 2021; 20:1534735420977666. [PMID: 33655799 PMCID: PMC7934026 DOI: 10.1177/1534735420977666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Purpose: Exercise has been shown to improve physical and psychological conditions during cancer therapy, but mechanisms remain poorly understood. The purpose of the present study was to report the results of cancer-related biomarkers and metabolomics outcomes from the PASAPAS feasibility study. Methods: In the PASAPAS randomized controlled trial, 61 women beginning adjuvant chemotherapy for localized breast cancer were randomized in a 6-month program of weekly aerobic exercises associated with nutritional counseling versus usual care with nutritional counseling. In the present analysis of 58 women for whom blood samples were available, first, circulating levels of biomarkers (ie, insulin, insulin-like growth factor 1, estradiol, adiponectin, leptin, interleukin-6, and tumor necrosis factor α) were measured at baseline and 6-month follow-up. Changes in biomarkers were compared between exercisers (n = 40) and controls (n = 18) using mixed-effect models. Second, serum metabolites were studied using an untargeted 1H nuclear magnetic resonance spectroscopy, and orthogonal partial least squares analyses were performed to discriminate exercisers and controls at baseline and at 6 months. Results: Over the 6-month intervention, no statistically significant differences were observed between exercisers and controls regarding changes in biomarkers and metabolomic profiles. Conclusion: The present analysis of the PASAPAS feasibility trial did not reveal any improvement in circulating biomarkers nor identified metabolic signatures in exercisers versus controls during adjuvant breast cancer treatment. Larger studies preferably in women with poor physical activity level to avoid ceiling effect, testing different doses and types of exercise on additional biological pathways, could allow to clarify the mechanisms mediating beneficial effects of physical exercise during cancer treatment. Trial registration: ClinicalTrials.gov Identifier: NCT01331772. Registered 8 April 2011, https://clinicaltrials.gov/ct2/show/NCT01331772?term=pasapas&rank=1
Collapse
Affiliation(s)
| | - Elodie Jobard
- Léon Bérard Cancer Center, Lyon, France.,University of Lyon, Villeurbanne, France
| | - Adrien Rossary
- University of Clermont Auvergne, Clermont-Ferrand, France
| | - Vincent Pialoux
- University of Lyon, Villeurbanne, France.,Institut Universitaire de France, Paris, France
| | | | | | - Lidia Delrieu
- Léon Bérard Cancer Center, Lyon, France.,University of Lyon, Villeurbanne, France
| | | | | | - Marina Touillaud
- Léon Bérard Cancer Center, Lyon, France.,Inserm UA8, Lyon, France
| | | | | | | | | | | | - Marie-Paule Vasson
- University of Clermont Auvergne, Clermont-Ferrand, France.,Clermont-Ferrand University Hospital, Jean Perrin Cancer Center, Clermont-Ferrand, France
| | - Béatrice Fervers
- Léon Bérard Cancer Center, Lyon, France.,Inserm UA8, Lyon, France
| |
Collapse
|
8
|
Pimentel I, Chen BE, Lohmann AE, Ennis M, Ligibel J, Shepherd L, Hershman DL, Whelan T, Stambolic V, Mayer I, Hobday T, Lemieux J, Thompson A, Rastogi P, Gelmon K, Rea D, Rabaglio M, Ellard S, Mates M, Bedard P, Pitre L, Vandenberg T, Dowling RJO, Parulekar W, Goodwin PJ. The Effect of Metformin vs Placebo on Sex Hormones in Canadian Cancer Trials Group MA.32. J Natl Cancer Inst 2021; 113:192-198. [PMID: 33527137 PMCID: PMC7850529 DOI: 10.1093/jnci/djaa082] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/08/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metformin has been associated with lower breast cancer (BC) risk and improved outcomes in observational studies. Multiple biologic mechanisms have been proposed, including a recent report of altered sex hormones. We evaluated the effect of metformin on sex hormones in MA.32, a phase III trial of nondiabetic BC subjects who were randomly assigned to metformin or placebo. METHODS We studied the subgroup of postmenopausal hormone receptor-negative BC subjects not receiving endocrine treatment who provided fasting blood at baseline and at 6 months after being randomly assigned. Sex hormone-binding globulin, bioavailable testosterone, and estradiol levels were assayed using electrochemiluminescence immunoassay. Change from baseline to 6 months between study arms was compared using Wilcoxon sum rank tests and regression models. RESULTS 312 women were eligible (141 metformin vs 171 placebo); the majority of subjects in each arm had T1/2, N0, HER2-negative BC and had received (neo)adjuvant chemotherapy. Mean age was 58.1 (SD=6.9) vs 57.5 (SD=7.9) years, mean body mass index (BMI) was 27.3 (SD=5.5) vs 28.9 (SD=6.4) kg/m2 for metformin vs placebo, respectively. Median estradiol decreased between baseline and 6 months on metformin vs placebo (-5.7 vs 0 pmol/L; P < .001) in univariable analysis and after controlling for baseline BMI and BMI change (P < .001). There was no change in sex hormone-binding globulin or bioavailable testosterone. CONCLUSION Metformin lowered estradiol levels, independent of BMI. This observation suggests a new metformin effect that has potential relevance to estrogen sensitive cancers.
Collapse
Affiliation(s)
- Isabel Pimentel
- Vall d`Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bingshu E Chen
- Canadian Cancer Trials Group, Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | | | | | | | - Lois Shepherd
- Canadian Cancer Trials Group, Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | - Dawn L Hershman
- Herbert Irving Cancer Center, Columbia University, New York, NY, USA
| | - Timothy Whelan
- Juravinski Cancer Centre at Hamilton Health Sciences, Hamilton, ON, Canada
| | - Vuk Stambolic
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - Ingrid Mayer
- Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | - Julie Lemieux
- CHA-Hopital Du St-Sacrement, Hopital Enfant Jesus Site, Quebec City, Canada
| | | | - Priya Rastogi
- National Surgical Adjuvant Breast and Bowel Project, Pittsburgh, PA, USA
| | - Karen Gelmon
- BCCA–Vancouver Cancer Centre, Vancouver, BC, Canada
| | - Daniel Rea
- Institute of Cancer Research, Clinical Trials and Statistics Unit, Sutton, UK
| | | | - Susan Ellard
- BCCA-Cancer Centre for the Southern Interior, Kelowna, BC, Canada
| | - Mihaela Mates
- Cancer Centre of Southeastern Ontario, Kingston, ON, Canada
| | - Philippe Bedard
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | | | | | - Ryan J O Dowling
- University Health Network, Princess Margaret Hospital, Toronto, ON, Canada
| | - Wendy Parulekar
- Canadian Cancer Trials Group , Queen’s University–Cancer Research Institute, Kingston, ON, Canada
| | - Pamela J Goodwin
- Lunenfeld Tanenbaum Research Institute at Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Hooper L, Martin N, Jimoh OF, Kirk C, Foster E, Abdelhamid AS. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev 2020; 8:CD011737. [PMID: 32827219 PMCID: PMC8092457 DOI: 10.1002/14651858.cd011737.pub3] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Reducing saturated fat reduces serum cholesterol, but effects on other intermediate outcomes may be less clear. Additionally, it is unclear whether the energy from saturated fats eliminated from the diet are more helpfully replaced by polyunsaturated fats, monounsaturated fats, carbohydrate or protein. OBJECTIVES To assess the effect of reducing saturated fat intake and replacing it with carbohydrate (CHO), polyunsaturated (PUFA), monounsaturated fat (MUFA) and/or protein on mortality and cardiovascular morbidity, using all available randomised clinical trials. SEARCH METHODS We updated our searches of the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (Ovid) and Embase (Ovid) on 15 October 2019, and searched Clinicaltrials.gov and WHO International Clinical Trials Registry Platform (ICTRP) on 17 October 2019. SELECTION CRITERIA Included trials fulfilled the following criteria: 1) randomised; 2) intention to reduce saturated fat intake OR intention to alter dietary fats and achieving a reduction in saturated fat; 3) compared with higher saturated fat intake or usual diet; 4) not multifactorial; 5) in adult humans with or without cardiovascular disease (but not acutely ill, pregnant or breastfeeding); 6) intervention duration at least 24 months; 7) mortality or cardiovascular morbidity data available. DATA COLLECTION AND ANALYSIS Two review authors independently assessed inclusion, extracted study data and assessed risk of bias. We performed random-effects meta-analyses, meta-regression, subgrouping, sensitivity analyses, funnel plots and GRADE assessment. MAIN RESULTS We included 15 randomised controlled trials (RCTs) (16 comparisons, 56,675 participants), that used a variety of interventions from providing all food to advice on reducing saturated fat. The included long-term trials suggested that reducing dietary saturated fat reduced the risk of combined cardiovascular events by 17% (risk ratio (RR) 0.83; 95% confidence interval (CI) 0.70 to 0.98, 12 trials, 53,758 participants of whom 8% had a cardiovascular event, I² = 67%, GRADE moderate-quality evidence). Meta-regression suggested that greater reductions in saturated fat (reflected in greater reductions in serum cholesterol) resulted in greater reductions in risk of CVD events, explaining most heterogeneity between trials. The number needed to treat for an additional beneficial outcome (NNTB) was 56 in primary prevention trials, so 56 people need to reduce their saturated fat intake for ~four years for one person to avoid experiencing a CVD event. In secondary prevention trials, the NNTB was 53. Subgrouping did not suggest significant differences between replacement of saturated fat calories with polyunsaturated fat or carbohydrate, and data on replacement with monounsaturated fat and protein was very limited. We found little or no effect of reducing saturated fat on all-cause mortality (RR 0.96; 95% CI 0.90 to 1.03; 11 trials, 55,858 participants) or cardiovascular mortality (RR 0.95; 95% CI 0.80 to 1.12, 10 trials, 53,421 participants), both with GRADE moderate-quality evidence. There was little or no effect of reducing saturated fats on non-fatal myocardial infarction (RR 0.97, 95% CI 0.87 to 1.07) or CHD mortality (RR 0.97, 95% CI 0.82 to 1.16, both low-quality evidence), but effects on total (fatal or non-fatal) myocardial infarction, stroke and CHD events (fatal or non-fatal) were all unclear as the evidence was of very low quality. There was little or no effect on cancer mortality, cancer diagnoses, diabetes diagnosis, HDL cholesterol, serum triglycerides or blood pressure, and small reductions in weight, serum total cholesterol, LDL cholesterol and BMI. There was no evidence of harmful effects of reducing saturated fat intakes. AUTHORS' CONCLUSIONS The findings of this updated review suggest that reducing saturated fat intake for at least two years causes a potentially important reduction in combined cardiovascular events. Replacing the energy from saturated fat with polyunsaturated fat or carbohydrate appear to be useful strategies, while effects of replacement with monounsaturated fat are unclear. The reduction in combined cardiovascular events resulting from reducing saturated fat did not alter by study duration, sex or baseline level of cardiovascular risk, but greater reduction in saturated fat caused greater reductions in cardiovascular events.
Collapse
Affiliation(s)
- Lee Hooper
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Nicole Martin
- Institute of Health Informatics Research, University College London, London, UK
| | - Oluseyi F Jimoh
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Christian Kirk
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Eve Foster
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
10
|
The effect of synbiotic on glycemic profile and sex hormones in overweight and obese breast cancer survivors following a weight-loss diet: A randomized, triple-blind, controlled trial. Clin Nutr 2020; 40:394-403. [PMID: 32698957 DOI: 10.1016/j.clnu.2020.05.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The investigation was designed to assess the effects of synbiotic supplementation on glycemic profile, insulin-like growth factor-1 (IGF-1) and sex hormones in overweight and obese postmenopausal breast cancer survivors (BCSs) who had hormone-receptor-positive breast cancer. METHODS This randomized, triple-blind, placebo-controlled trial was conducted on 76 overweight and obese BCSs aged 57.43 (5.82) years. All participants were given a specified low calorie diet and were randomly assigned into two groups to intake 109 CFU/day of synbiotic supplement (n = 38) or placebo (n = 38) for 8 weeks. Body composition, physical activity, glycemic profile, IGF-1, estradiol, testosterone and dehydroepiandrosterone sulfate (DHEA-S) were measured at baseline and after 8 weeks. RESULTS A significant reduction in serum insulin (median change (Q1, Q3) from baseline of -1.05 (-2.36, 0.32) μIU/mL; P = 0.006) and insulin resistance (HOMA-IR) (mean change (SD) from baseline of -4.0 (0.9); P = 0.007) were seen over the 8 weeks in the synbiotic group. However, no significant changes were observed in serum insulin, fasting plasma glucose, HbA1c, HOMA-IR, IGF-1, estradiol, testosterone, DHEA-S and sex hormone binding globulin between-groups at the end of the intervention. CONCLUSIONS Overall, as the 8-week synbiotic consumption compared with placebo had insignificant-reducing effects on glycemic profile, IGF-1 and sex hormones among overweight and obese postmenopausal BCSs, synbiotics may exert considerable beneficial consequences, which need to be further assessed in future clinical trials. TRIAL REGISTRATION IRCT, IRCT2015090223861N1. Registered 02 February 2017, http://www.irct.ir: IRCT2015090223861N1.
Collapse
|
11
|
Hooper L, Abdelhamid AS, Jimoh OF, Bunn D, Skeaff CM. Effects of total fat intake on body fatness in adults. Cochrane Database Syst Rev 2020; 6:CD013636. [PMID: 32476140 PMCID: PMC7262429 DOI: 10.1002/14651858.cd013636] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The ideal proportion of energy from fat in our food and its relation to body weight is not clear. In order to prevent overweight and obesity in the general population, we need to understand the relationship between the proportion of energy from fat and resulting weight and body fatness in the general population. OBJECTIVES To assess the effects of proportion of energy intake from fat on measures of body fatness (including body weight, waist circumference, percentage body fat and body mass index) in people not aiming to lose weight, using all appropriate randomised controlled trials (RCTs) of at least six months duration. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, Clinicaltrials.gov and the WHO International Clinical Trials Registry Platform (ICTRP) to October 2019. We did not limit the search by language. SELECTION CRITERIA Trials fulfilled the following criteria: 1) randomised intervention trial, 2) included adults aged at least 18 years, 3) randomised to a lower fat versus higher fat diet, without the intention to reduce weight in any participants, 4) not multifactorial and 5) assessed a measure of weight or body fatness after at least six months. We duplicated inclusion decisions and resolved disagreement by discussion or referral to a third party. DATA COLLECTION AND ANALYSIS We extracted data on the population, intervention, control and outcome measures in duplicate. We extracted measures of body fatness (body weight, BMI, percentage body fat and waist circumference) independently in duplicate at all available time points. We performed random-effects meta-analyses, meta-regression, subgrouping, sensitivity, funnel plot analyses and GRADE assessment. MAIN RESULTS We included 37 RCTs (57,079 participants). There is consistent high-quality evidence from RCTs that reducing total fat intake results in small reductions in body fatness; this was seen in almost all included studies and was highly resistant to sensitivity analyses (GRADE high-consistency evidence, not downgraded). The effect of eating less fat (compared with higher fat intake) is a mean body weight reduction of 1.4 kg (95% confidence interval (CI) -1.7 to -1.1 kg, in 53,875 participants from 26 RCTs, I2 = 75%). The heterogeneity was explained in subgrouping and meta-regression. These suggested that greater weight loss results from greater fat reductions in people with lower fat intake at baseline, and people with higher body mass index (BMI) at baseline. The size of the effect on weight does not alter over time and is mirrored by reductions in BMI (MD -0.5 kg/m2, 95% CI -0.6 to -0.3, 46,539 participants in 14 trials, I2 = 21%), waist circumference (MD -0.5 cm, 95% CI -0.7 to -0.2, 16,620 participants in 3 trials; I2 = 21%), and percentage body fat (MD -0.3% body fat, 95% CI -0.6 to 0.00, P = 0.05, in 2350 participants in 2 trials; I2 = 0%). There was no suggestion of harms associated with low fat diets that might mitigate any benefits on body fatness. The reduction in body weight was reflected in small reductions in LDL (-0.13 mmol/L, 95% CI -0.21 to -0.05), and total cholesterol (-0.23 mmol/L, 95% CI -0.32 to -0.14), with little or no effect on HDL cholesterol (-0.02 mmol/L, 95% CI -0.03 to 0.00), triglycerides (0.01 mmol/L, 95% CI -0.05 to 0.07), systolic (-0.75 mmHg, 95% CI -1.42 to -0.07) or diastolic blood pressure(-0.52 mmHg, 95% CI -0.95 to -0.09), all GRADE high-consistency evidence or quality of life (0.04, 95% CI 0.01 to 0.07, on a scale of 0 to 10, GRADE low-consistency evidence). AUTHORS' CONCLUSIONS Trials where participants were randomised to a lower fat intake versus a higher fat intake, but with no intention to reduce weight, showed a consistent, stable but small effect of low fat intake on body fatness: slightly lower weight, BMI, waist circumference and percentage body fat compared with higher fat arms. Greater fat reduction, lower baseline fat intake and higher baseline BMI were all associated with greater reductions in weight. There was no evidence of harm to serum lipids, blood pressure or quality of life, but rather of small benefits or no effect.
Collapse
Affiliation(s)
- Lee Hooper
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | | - Oluseyi F Jimoh
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Diane Bunn
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
12
|
Wang T, Nichols HB, Nyante SJ, Bradshaw PT, Moorman PG, Kabat GC, Parada H, Khankari NK, Teitelbaum SL, Terry MB, Santella RM, Neugut AI, Gammon MD. Urinary Estrogen Metabolites and Long-Term Mortality Following Breast Cancer. JNCI Cancer Spectr 2020; 4:pkaa014. [PMID: 32455334 PMCID: PMC7236781 DOI: 10.1093/jncics/pkaa014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/11/2019] [Accepted: 02/26/2020] [Indexed: 12/09/2022] Open
Abstract
Background Estrogen metabolite concentrations of 2-hydroxyestrone (2-OHE1) and 16-hydroxyestrone (16-OHE1) may be associated with breast carcinogenesis. However, no study has investigated their possible impact on mortality after breast cancer. Methods This population-based study was initiated in 1996–1997 with spot urine samples obtained shortly after diagnosis (mean = 96 days) from 683 women newly diagnosed with first primary breast cancer and 434 age-matched women without breast cancer. We measured urinary concentrations of 2-OHE1 and 16-OHE1 using an enzyme-linked immunoassay. Vital status was determined via the National Death Index (n = 244 deaths after a median of 17.7 years of follow-up). We used multivariable-adjusted Cox proportional hazards to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the estrogen metabolites-mortality association. We evaluated effect modification using likelihood ratio tests. All statistical tests were two-sided. Results Urinary concentrations of the 2-OHE1 to 16-OHE1 ratio (>median of 1.8 vs ≤median) were inversely associated with all-cause mortality (HR = 0.74, 95% CI = 0.56 to 0.98) among women with breast cancer. Reduced hazard was also observed for breast cancer mortality (HR = 0.73, 95% CI = 0.45 to 1.17) and cardiovascular diseases mortality (HR = 0.76, 95% CI = 0.47 to 1.23), although the 95% confidence intervals included the null. Similar findings were also observed for women without breast cancer. The association with all-cause mortality was more pronounced among breast cancer participants who began chemotherapy before urine collection (n = 118, HR = 0.42, 95% CI = 0.22 to 0.81) than among those who had not (n = 559, HR = 0.98, 95% CI = 0.72 to 1.34; Pinteraction = .008). Conclusions The urinary 2-OHE1 to 16-OHE1 ratio may be inversely associated with long-term all-cause mortality, which may depend on cancer treatment status at the time of urine collection.
Collapse
Affiliation(s)
- Tengteng Wang
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hazel B Nichols
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Sarah J Nyante
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Patricia G Moorman
- Department of Community and Family Medicine, Duke University, Durham, NC, USA
| | | | - Humberto Parada
- Division of Epidemiology and Biostatistics, San Diego State University, San Diego, CA, USA
| | - Nikhil K Khankari
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susan L Teitelbaum
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Regina M Santella
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Alfred I Neugut
- Department of Epidemiology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
13
|
Hooper L, Martin N, Jimoh OF, Kirk C, Foster E, Abdelhamid AS. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev 2020; 5:CD011737. [PMID: 32428300 PMCID: PMC7388853 DOI: 10.1002/14651858.cd011737.pub2] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Reducing saturated fat reduces serum cholesterol, but effects on other intermediate outcomes may be less clear. Additionally, it is unclear whether the energy from saturated fats eliminated from the diet are more helpfully replaced by polyunsaturated fats, monounsaturated fats, carbohydrate or protein. OBJECTIVES To assess the effect of reducing saturated fat intake and replacing it with carbohydrate (CHO), polyunsaturated (PUFA), monounsaturated fat (MUFA) and/or protein on mortality and cardiovascular morbidity, using all available randomised clinical trials. SEARCH METHODS We updated our searches of the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (Ovid) and Embase (Ovid) on 15 October 2019, and searched Clinicaltrials.gov and WHO International Clinical Trials Registry Platform (ICTRP) on 17 October 2019. SELECTION CRITERIA Included trials fulfilled the following criteria: 1) randomised; 2) intention to reduce saturated fat intake OR intention to alter dietary fats and achieving a reduction in saturated fat; 3) compared with higher saturated fat intake or usual diet; 4) not multifactorial; 5) in adult humans with or without cardiovascular disease (but not acutely ill, pregnant or breastfeeding); 6) intervention duration at least 24 months; 7) mortality or cardiovascular morbidity data available. DATA COLLECTION AND ANALYSIS Two review authors independently assessed inclusion, extracted study data and assessed risk of bias. We performed random-effects meta-analyses, meta-regression, subgrouping, sensitivity analyses, funnel plots and GRADE assessment. MAIN RESULTS We included 15 randomised controlled trials (RCTs) (16 comparisons, ~59,000 participants), that used a variety of interventions from providing all food to advice on reducing saturated fat. The included long-term trials suggested that reducing dietary saturated fat reduced the risk of combined cardiovascular events by 21% (risk ratio (RR) 0.79; 95% confidence interval (CI) 0.66 to 0.93, 11 trials, 53,300 participants of whom 8% had a cardiovascular event, I² = 65%, GRADE moderate-quality evidence). Meta-regression suggested that greater reductions in saturated fat (reflected in greater reductions in serum cholesterol) resulted in greater reductions in risk of CVD events, explaining most heterogeneity between trials. The number needed to treat for an additional beneficial outcome (NNTB) was 56 in primary prevention trials, so 56 people need to reduce their saturated fat intake for ~four years for one person to avoid experiencing a CVD event. In secondary prevention trials, the NNTB was 32. Subgrouping did not suggest significant differences between replacement of saturated fat calories with polyunsaturated fat or carbohydrate, and data on replacement with monounsaturated fat and protein was very limited. We found little or no effect of reducing saturated fat on all-cause mortality (RR 0.96; 95% CI 0.90 to 1.03; 11 trials, 55,858 participants) or cardiovascular mortality (RR 0.95; 95% CI 0.80 to 1.12, 10 trials, 53,421 participants), both with GRADE moderate-quality evidence. There was little or no effect of reducing saturated fats on non-fatal myocardial infarction (RR 0.97, 95% CI 0.87 to 1.07) or CHD mortality (RR 0.97, 95% CI 0.82 to 1.16, both low-quality evidence), but effects on total (fatal or non-fatal) myocardial infarction, stroke and CHD events (fatal or non-fatal) were all unclear as the evidence was of very low quality. There was little or no effect on cancer mortality, cancer diagnoses, diabetes diagnosis, HDL cholesterol, serum triglycerides or blood pressure, and small reductions in weight, serum total cholesterol, LDL cholesterol and BMI. There was no evidence of harmful effects of reducing saturated fat intakes. AUTHORS' CONCLUSIONS The findings of this updated review suggest that reducing saturated fat intake for at least two years causes a potentially important reduction in combined cardiovascular events. Replacing the energy from saturated fat with polyunsaturated fat or carbohydrate appear to be useful strategies, while effects of replacement with monounsaturated fat are unclear. The reduction in combined cardiovascular events resulting from reducing saturated fat did not alter by study duration, sex or baseline level of cardiovascular risk, but greater reduction in saturated fat caused greater reductions in cardiovascular events.
Collapse
Affiliation(s)
- Lee Hooper
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Nicole Martin
- Institute of Health Informatics Research, University College London, London, UK
| | - Oluseyi F Jimoh
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Christian Kirk
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Eve Foster
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
14
|
Pavlović R, Janković S, Milovanović J, Stefanović S, Folić M, Milovanović O, Mamillapalli C, Milosavljević M. The Safety of Local Hormonal Treatment for Vulvovaginal Atrophy in Women With Estrogen Receptor-positive Breast Cancer Who Are on Adjuvant Aromatase Inhibitor Therapy: Meta-analysis. Clin Breast Cancer 2019; 19:e731-e740. [DOI: 10.1016/j.clbc.2019.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/13/2019] [Accepted: 07/14/2019] [Indexed: 01/01/2023]
|
15
|
Prague JK, Abbara A, Comninos AN, Jayasena CN, Higham CE, Adaway J, Keevil BG, Veldhuis JD, Dhillo WS. Neurokinin 3 Receptor Antagonists Do Not Increase FSH or Estradiol Secretion in Menopausal Women. J Endocr Soc 2019; 4:bvz009. [PMID: 32318647 PMCID: PMC7159071 DOI: 10.1210/jendso/bvz009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/13/2019] [Indexed: 01/19/2023] Open
Abstract
Background Neurokinin 3 receptor (NK3R) antagonism is a promising novel treatment for menopausal flashes. However, to avoid adverse hormonal effects it is clinically important to first confirm whether gonadotropin and estradiol concentrations change as a result of their administration. Methods Single-center, randomized, double-blind, placebo-controlled, crossover trial of an oral NK3R antagonist (MLE4901) in 28 women aged 40 to 62 years, experiencing >7 hot flashes/24 h; some bothersome or severe (Clinicaltrials.gov, NCT02668185). Weekly serum gonadotropins and estradiol levels were measured using commercially available automated immunoassays a priori. Serum estradiol was also measured post hoc using a highly sensitive direct assay by liquid chromatography tandem mass spectrometry. Hormone levels were compared by the paired sample t tests or by the Wilcoxon matched-pairs signed rank test, as appropriate for the distribution of the data. Results Mean (standard deviation) serum follicle-stimulating hormone (FSH) concentration was not significantly increased when taking MLE4901 (72.07 ± 19.81 IU/L) compared to placebo (70.03 ± 19.56 IU/L), P = .26. Serum estradiol was also not significantly altered, irrespective of which assay method was used (median interquartile range of serum estradiol by immunoassay: placebo 36 ± 3 pmol/L, MLE4901 36 ± 1 pmol/L, P = .21; median serum highly sensitive estradiol: placebo 12 ± 16 pmol/L, MLE4901 13 ± 15 pmol/L, P = .70). However, mean (standard deviation) serum luteinizing hormone concentration significantly decreased with MLE4901 (27.63 ± 9.76 IU/L) compared to placebo (30.26 ± 9.75 IU/L), P = .0024. Implication NK3R antagonists do not increase serum estradiol or FSH despite their reduction in hot flashes. This is clinically significant and highly reassuring for women who have a contraindication to conventional hormone therapy such as prior/existing breast cancer and/or thromboembolism.
Collapse
Affiliation(s)
- Julia K Prague
- Section of Endocrinology & Investigative Medicine, Imperial College, London, UK
| | - Ali Abbara
- Section of Endocrinology & Investigative Medicine, Imperial College, London, UK
| | | | - Channa N Jayasena
- Section of Endocrinology & Investigative Medicine, Imperial College, London, UK
| | - Claire E Higham
- Department of Endocrinology, The Christie NHS Foundation Trust, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jo Adaway
- Biochemistry Department, Wythenshawe Hospital, Wythenshawe, UK.,School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Brian G Keevil
- Biochemistry Department, Wythenshawe Hospital, Wythenshawe, UK.,School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Waljit S Dhillo
- Section of Endocrinology & Investigative Medicine, Imperial College, London, UK
| |
Collapse
|
16
|
The estrogen effect; clinical and histopathological evidence of dichotomous influences in dogs with spontaneous mammary carcinomas. PLoS One 2019; 14:e0224504. [PMID: 31652293 PMCID: PMC6814212 DOI: 10.1371/journal.pone.0224504] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/10/2019] [Indexed: 11/21/2022] Open
Abstract
The purpose of this study was to investigate the associations and explore the relationships between hormonal factors (serum estrogen, estrogen receptors and ovariohysterectomy) and other clinical/histological prognostic factors and their impact on outcome in dogs with mammary carcinomas. Data from two separate prospective studies on dogs with spontaneous mammary carcinomas were used for this research. All dogs underwent standardized diagnostic testing, staging, surgery and follow-up examinations. Serum estrogen was analyzed by competitive enzyme immunoassay or radioimmunoassay, and tumor estrogen receptor (ER) expression was analyzed by immunohistochemistry. A total of 159 dogs were included; 130 were spayed and 29 remained. High serum estrogen was associated with an overall longer time to metastasis (p = 0.021). When stratifying based on spay group, the effect was only significant in spayed dogs, (p = 0.019). Positive tumor ER expression was also associated with a longer time to metastasis (p = 0.025), but similar to above, only in dogs that were spayed (p = 0.049). Further subgroup analysis revealed that high serum estrogen was significantly associated with improved survival in dogs with ER positive tumors, but only in spayed dogs (p = 0.0052). Interestingly, the effect of spaying was the opposite in dogs with ER negative tumors; here, intact dogs with high serum estrogen but ER negative tumors had a significantly longer time to metastasis (p = 0.036). Low serum estrogen was associated with increased risk for the development of non-mammary tumors in the post-operative period (p = 0.012). These results highlight the dual effect of estrogen in cancer: Estrogen acts as a pro-carcinogen in ER positive mammary tumors, but a may have a protective effect in ER negative tumors, potentially via non-receptor mechanisms. The latter is supported by the decreased risk for non-mammary tumors in dogs with high serum estrogen, and explains the increased incidence of certain non-mammary tumors in in dogs spayed at an early age.
Collapse
|
17
|
Uomori T, Horimoto Y, Arakawa A, Iijima K, Saito M. Breast Cancer in Lean Postmenopausal Women Might Have Specific Pathological Features. In Vivo 2019; 33:483-487. [PMID: 30804130 DOI: 10.21873/invivo.11499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM The rate of obesity, an independent risk factor for breast cancer in post-menopausal women, is quite low in Japan, indicating breast cancer in lean women to be more common in the Japanese than in Westerners. However, biological characteristics of such breast cancers have not been adequately investigated. PATIENTS AND METHODS We retrospectively investigated clinicopathological breast cancer features of 566 postmenopausal Japanese women, who underwent curative surgery, in relation to patient physique based on the body mass index. RESULTS There were no differences in several factors examined such as tumour size according to patient physique. On the other hand, mean values of the Ki67 labelling index were significantly higher in lean compared to obese patients (p=0.027). Likewise, HER2-positive tumours were more often observed in lean patients (p=0.051). CONCLUSION Lean postmenopausal women had more aggressive tumours, apparently contradicting the widely held view for breast cancer in obese women.
Collapse
Affiliation(s)
- Toshitaka Uomori
- Deartment of Breast Oncology, Juntendo University School of Medcine, Tokyo, Japan
| | - Yoshiya Horimoto
- Deartment of Breast Oncology, Juntendo University School of Medcine, Tokyo, Japan .,Deartment of Pathology and Oncology, Juntendo University School of Medcine, Tokyo, Japan
| | - Atsushi Arakawa
- Deartment of Human Pathology, Juntendo University School of Medcine, Tokyo, Japan
| | - Kotaro Iijima
- Deartment of Breast Oncology, Juntendo University School of Medcine, Tokyo, Japan
| | - Mitsue Saito
- Deartment of Breast Oncology, Juntendo University School of Medcine, Tokyo, Japan
| |
Collapse
|
18
|
Ecker BL, Lee JY, Sterner CJ, Solomon AC, Pant DK, Shen F, Peraza J, Vaught L, Mahendra S, Belka GK, Pan TC, Schmitz KH, Chodosh LA. Impact of obesity on breast cancer recurrence and minimal residual disease. Breast Cancer Res 2019; 21:41. [PMID: 30867005 PMCID: PMC6416940 DOI: 10.1186/s13058-018-1087-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/13/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Obesity is associated with an increased risk of breast cancer recurrence and cancer death. Recurrent cancers arise from the pool of residual tumor cells, or minimal residual disease (MRD), that survives primary treatment and persists in the host. Whether the association of obesity with recurrence risk is causal is unknown, and the impact of obesity on MRD and breast cancer recurrence has not been reported in humans or in animal models. METHODS Doxycycline-inducible primary mammary tumors were generated in intact MMTV-rtTA;TetO-HER2/neu (MTB/TAN) mice or orthotopic recipients fed a high-fat diet (HFD; 60% kcal from fat) or a control low-fat diet (LFD; 10% kcal from fat). Following oncogene downregulation and tumor regression, mice were followed for clinical recurrence. Body weight was measured twice weekly and used to segregate HFD mice into obese (i.e., responders) and lean (i.e., nonresponders) study arms, and obesity was correlated with body fat percentage, glucose tolerance (measured using intraperitoneal glucose tolerance tests), serum biomarkers (measured by enzyme-linked immunosorbent assay), and tissue transcriptomics (assessed by RNA sequencing). MRD was quantified by droplet digital PCR. RESULTS HFD-Obese mice weighed significantly more than HFD-Lean and LFD control mice (p < 0.001) and had increased body fat percentage (p < 0.001). Obese mice exhibited fasting hyperglycemia, hyperinsulinemia, and impaired glucose tolerance, as well as decreased serum levels of adiponectin and increased levels of leptin, resistin, and insulin-like growth factor 1. Tumor recurrence was accelerated in HFD-Obese mice compared with HFD-Lean and LFD control mice (median relapse-free survival 53.0 days vs. 87.0 days vs. 80.0 days, log-rank p < 0.001; HFD-Obese compared with HFD-Lean HR 2.52, 95% CI 1.52-4.16; HFD-Obese compared with LFD HR 2.27, 95% CI 1.42-3.63). HFD-Obese mice harbored a significantly greater number of residual tumor cells than HFD-Lean and LFD mice (12,550 ± 991 vs. 7339 ± 2182 vs. 4793 ± 1618 cells, p < 0.001). CONCLUSION These studies provide a genetically engineered mouse model for study of the association of diet-induced obesity with breast cancer recurrence. They demonstrate that this model recapitulates physiological changes characteristic of obese patients, establish that the association between obesity and recurrence risk is causal in nature, and suggest that obesity is associated with the increased survival and persistence of residual tumor cells.
Collapse
MESH Headings
- Animals
- Body Mass Index
- Body Weight
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Cell Line, Tumor/transplantation
- Datasets as Topic
- Diet, High-Fat/adverse effects
- Disease-Free Survival
- Female
- Humans
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Mice, Obese
- Mice, Transgenic
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm, Residual
- Obesity/etiology
- Obesity/pathology
- Receptor, ErbB-2/genetics
- Survival Analysis
Collapse
Affiliation(s)
- Brett L. Ecker
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
| | - Jun Y. Lee
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Christopher J. Sterner
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Aaron C. Solomon
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Dhruv K. Pant
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Fei Shen
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Javier Peraza
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Lauren Vaught
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Samyukta Mahendra
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - George K. Belka
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Tien-chi Pan
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| | - Kathryn H. Schmitz
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033 USA
| | - Lewis A. Chodosh
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA USA
- 2-PREVENT Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
- The Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-6160 USA
| |
Collapse
|
19
|
Pre-diagnostic sex hormone levels and survival among breast cancer patients. Breast Cancer Res Treat 2019; 174:749-758. [PMID: 30604001 DOI: 10.1007/s10549-018-05121-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Higher levels of circulating sex steroid hormones are associated with increased breast cancer risk, though their association with prognosis remains unclear. We evaluated the association between circulating sex hormone levels and breast cancer survival in two large cohorts. METHODS We evaluated this association among 2073 breast cancer cases from the Nurses' Health Study (NHS) and Nurses' Health Study II (NHSII) cohorts. Women in this analysis provided a blood sample in 1989-1990 (NHS) or in 1996-1999 (NHSII) and were subsequently diagnosed with breast cancer. Levels of estradiol (postmenopausal women only), testosterone, dehydroepiandrosterone-sulfate (DHEAS), and sex hormone-binding globulin (SHBG) were measured in plasma. Cox proportional hazards models were used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI) for survival, adjusting for patient and tumor characteristics. RESULTS A total of 639 deaths and 160 breast cancer deaths occurred over follow-up through 2015. Compared to women in the lowest quartile, postmenopausal women in the highest quartile of estradiol experienced a 1.43-fold overall mortality rate (HR 1.43, 95% CI 1.03-1.97, P-trend = 0.04) and a nonsignificantly higher breast cancer mortality rate (HR 1.50, 95% CI 0.75-2.98, P-trend = 0.12). Higher DHEAS levels were nonsignificantly associated with better overall survival (HRQ4vsQ1=0.79, 95% CI 0.57-1.10, P-trend = 0.05), though not with breast cancer survival. No associations were observed between testosterone or SHBG and survival. CONCLUSIONS Pre-diagnostic postmenopausal circulating estradiol levels were modestly associated with worse survival among breast cancer patients. Further studies should evaluate whether circulating hormone levels at diagnosis predict cancer prognosis or treatment response.
Collapse
|
20
|
Patterson RE, Marinac CR, Sears DD, Kerr J, Hartman SJ, Cadmus-Bertram L, Villaseñor A, Flatt SW, Godbole S, Li H, Laughlin GA, Oratowski-Coleman J, Parker BA, Natarajan L. The Effects of Metformin and Weight Loss on Biomarkers Associated With Breast Cancer Outcomes. J Natl Cancer Inst 2018; 110:1239-1247. [PMID: 29788487 PMCID: PMC6235688 DOI: 10.1093/jnci/djy040] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/05/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background This study investigated the effects of metformin and weight loss on biomarkers associated with breast cancer prognosis. Methods Overweight/obese postmenopausal breast cancer survivors (n = 333) were randomly assigned to metformin vs placebo and to a weight loss intervention vs control (ie, usual care). The 2 × 2 factorial design allows a single randomized trial to investigate the effect of two factors and interactions between them. Outcomes were changes in fasting insulin, glucose, C-reactive protein (CRP), estradiol, testosterone, and sex-hormone binding globulin (SHBG). The trial was powered for a main effects analysis of metformin vs placebo and weight loss vs control. All tests of statistical significance were two-sided. Results A total of 313 women (94.0%) completed the six-month trial. High prescription adherence (ie, ≥80% of pills taken) ranged from 65.9% of participants in the metformin group to 81.3% of those in the placebo group (P < .002). Mean percent weight loss was statistically significantly higher in the weight loss group (-5.5%, 95% confidence interval [CI] = -6.3% to -4.8%) compared with the control group (-2.7%, 95% CI = -3.5% to -1.9%). Statistically significant group differences (ie, percent change in metformin group minus placebo group) were -7.9% (95% CI = -15.0% to -0.8%) for insulin, -10.0% (95% CI = -18.5% to -1.5%) for estradiol, -9.5% (95% CI = -15.2% to -3.8%) for testosterone, and 7.5% (95% CI = 2.4% to 12.6%) for SHBG. Statistically significant group differences (ie, percent change in weight loss group minus placebo group) were -12.5% (95% CI = -19.6% to -5.3%) for insulin and 5.3% (95% CI = 0.2% to 10.4%) for SHBG. Conclusions As adjuvant therapy, weight loss and metformin were found to be a safe combination strategy that modestly lowered estrogen levels and advantageously affected other biomarkers thought to be on the pathway for reducing breast cancer recurrence and mortality.
Collapse
Affiliation(s)
- Ruth E Patterson
- Department of Family Medicine and Public Health, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | - Catherine R Marinac
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Dorothy D Sears
- Department of Family Medicine and Public Health, La Jolla, CA
- Department of Medicine, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | - Jacqueline Kerr
- Department of Family Medicine and Public Health, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | - Sheri J Hartman
- Department of Family Medicine and Public Health, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | | | - Adriana Villaseñor
- Department of Family Medicine and Public Health, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | | | | | | | - Gail A Laughlin
- Department of Family Medicine and Public Health, La Jolla, CA
| | | | - Barbara A Parker
- Department of Medicine, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| | - Loki Natarajan
- Department of Family Medicine and Public Health, La Jolla, CA
- Moores UC San Diego Cancer Center, La Jolla, CA
| |
Collapse
|
21
|
Abstract
Weight, weight change and physical activity may affect prognosis among women who are diagnosed with breast cancer. Observational studies show associations between overweight/obesity and weight gain with several measures of reduced prognosis in women with breast cancer, and some suggestions of lower survival in women who are underweight or who experience unexplained weight loss after diagnosis. Observational studies have also shown an association between higher levels of physical activity and reduced breast cancer-specific and all-cause mortality, although a dose–response relationship has not been established. The effects of purposive dietary weight loss and increase in physical activity on survival or recurrence in breast cancer are not yet established, and randomised controlled trials are needed for definitive data. This paper presents the epidemiologic evidence on weight status, weight change, and physical activity and breast cancer survival; suggests potential mediating mechanisms; summarises evidence on weight loss interventions in breast cancer survivors; describes ongoing randomised clinical trials designed to test the effects of weight loss or physical activity on breast cancer survival; and provides information on available guidelines on weight and physical activity for cancer survivors.
Collapse
|
22
|
Yerushalmi R, Dong B, Chapman JW, Goss PE, Pollak MN, Burnell MJ, Levine MN, Bramwell VHC, Pritchard KI, Whelan TJ, Ingle JN, Shepherd LE, Parulekar WR, Han L, Ding K, Gelmon KA. Impact of baseline BMI and weight change in CCTG adjuvant breast cancer trials. Ann Oncol 2018; 28:1560-1568. [PMID: 28379421 DOI: 10.1093/annonc/mdx152] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Indexed: 12/12/2022] Open
Abstract
Background We hypothesized that increased baseline BMI and BMI change would negatively impact clinical outcomes with adjuvant breast cancer systemic therapy. Methods Data from chemotherapy trials MA.5 and MA.21; endocrine therapy MA.12, MA.14 and MA.27; and trastuzumab HERA/MA.24 were analyzed. The primary objective was to examine the effect of BMI change on breast cancer-free interval (BCFI) landmarked at 5 years; secondary objectives included BMI changes at 1 and 3 years; BMI changes on disease-specific survival (DSS) and overall survival (OS); and effects of baseline BMI. Stratified analyses included trial therapy and composite trial stratification factors. Results In pre-/peri-/early post-menopausal chemotherapy trials (N = 2793), baseline BMI did not impact any endpoint and increased BMI from baseline did not significantly affect BCFI (P = 0.85) after 5 years although it was associated with worse BCFI (P = 0.03) and DSS (P = 0.07) after 1 year. BMI increase by 3 and 5 years was associated with better DSS (P = 0.01; 0.01) and OS (P = 0.003; 0.05). In pre-menopausal endocrine therapy trial MA.12 (N = 672), patients with higher baseline BMI had worse BCFI (P = 0.02) after 1 year, worse DSS (P = 0.05; 0.004) after 1 and 5 years and worse OS (P = 0.01) after 5 years. Increased BMI did not impact BCFI (P = 0.90) after 5 years, although it was associated with worse BCFI (P = 0.01) after 1 year. In post-menopausal endocrine therapy trials MA.14 and MA.27 (N = 8236), baseline BMI did not significantly impact outcome for any endpoint. BMI change did not impact BCFI or DSS after 1 or 3 years, although a mean increased BMI of 0.3 was associated with better OS (P = 0.02) after 1 year. With the administration of trastuzumab (N = 1395) baseline BMI and BMI change did not significantly impact outcomes. Conclusions Higher baseline BMI and BMI increases negatively affected outcomes only in pre-/peri-/early post-menopausal trial patients. Otherwise, BMI increases similar to those expected in healthy women either did not impact outcome or were associated with better outcomes. Clinical Trials numbers CAN-NCIC-MA5; National Cancer Institute (NCI)-V90-0027; MA.12-NCT00002542; MA.14-NCT00002864; MA.21-NCT00014222; HERA, NCT00045032;CAN-NCIC-MA24; MA-27-NCT00066573.
Collapse
Affiliation(s)
- R Yerushalmi
- Department of Medical Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva and Tel-Aviv University, Tel Aviv, Israel
| | - B Dong
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - J W Chapman
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - P E Goss
- Massachusetts General Hospital Cancer Center, Boston, USA
| | - M N Pollak
- Department of Medical Oncology, Jewish General Hospital, McGill University, Montreal
| | - M J Burnell
- Department of Medical Oncology, Saint John Regional Hospital, Saint John
| | - M N Levine
- Department of Oncology, McMaster University, Juravinski Cancer Center, Hamilton, Ontario
| | - V H C Bramwell
- Department of Medical Oncology, Tom Baker Cancer Centre, Alberta Health Services and University of Calgary, Calgary
| | - K I Pritchard
- Department of Medical Oncology, Sunnybrook Odette Cancer Centre and the University of Toronto, Toronto, Canada
| | - T J Whelan
- Department of Oncology, Juravinski Cancer Center, McMaster University, Hamilton, Ontario
| | - J N Ingle
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - L E Shepherd
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - W R Parulekar
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - L Han
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - K Ding
- Canadian Cancer Trials Group (CCTG; Formerly, NCIC Clinical Trials Group), Queen's University, Kingston, Canada
| | - K A Gelmon
- Department of Medical Oncology, British Columbia Cancer Agency, Vancouver, Canada
| |
Collapse
|
23
|
Allemani C, Berrino F, Krogh V, Sieri S, Pupa SM, Tagliabue E, Tagliabue G, Sant M. Do Pre-Diagnostic Drinking Habits Influence Breast Cancer Survival? TUMORI JOURNAL 2018; 97:142-8. [DOI: 10.1177/030089161109700202] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Alcohol consumption increases the risk of developing breast cancer and may also be associated with late diagnosis, recurrence, distant metastases and death. Many studies have examined the role of alcohol as a risk factor for the development of breast cancer, but very few studies have addressed the role of alcohol as a prognostic factor for survival among women diagnosed with breast cancer. The aim of this study was to investigate the survival of women with breast cancer in relation to pre-diagnostic alcohol intake and other factors known to influence prognosis. Methods We analyzed data for 264 women in the EUROCARE and ORDET studies who were diagnosed with breast cancer from 1987 up to 31 December 2001 and for whom information was available on follow-up, stage at diagnosis, HER-2 and hormone receptor status, and pre-diagnostic dietary alcohol intake, categorized as zero (0 g/day, non-drinkers), moderate (up to 13 g/day, about 1 serving) and high (>13 g/day). Ten-year relative survival was estimated using the maximum-likelihood approach. The excess risk of death within 10 years of diagnosis was modeled by level of alcohol intake, adjusting separately for age, stage, body mass index and tumor subtype. Results Ten-year relative survival was lowerin women who drank more than 13g/day (65%; 95% CI, 47–78) than in non-drinkers (88%; 95% CI, 75–95). The excess risk of death within 10 years was significantly higher in women who drank more than 13 g/day than non-drinkers (relative excess risk, 4.13; 95% CI, 1.69–10.10) and was not altered by adjustment for other prognostic factors. The excess risk within 10 years was higher for women with a body mass index of 25 kg/m2 or higher (relative excess risk, 2.20; 95% CI, 1.01–4.70) and higher for those with more advanced disease. Conclusions Women who drank more than 13 g alcohol per day had lower survival than non-drinkers. The excess risk of death within 10 years of diagnosis was unaffected by other known risk factors. High alcohol consumption may be an adverse prognostic factor for breast cancer.
Collapse
Affiliation(s)
- Claudia Allemani
- Department of Preventive and Predictive Medicine, Analytical Epidemiology Unit, Milan
| | - Franco Berrino
- Department of Preventive and Predictive Medicine, Unit of Etiological Epidemiology and Prevention, Milan
| | - Vittorio Krogh
- Department of Preventive and Predictive Medicine, Nutritional Epidemiology Unit, Milan
| | - Sabina Sieri
- Department of Preventive and Predictive Medicine, Nutritional Epidemiology Unit, Milan
| | - Serenella M Pupa
- Laboratories and Department of Experimental Oncology, Molecular Biology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Elda Tagliabue
- Laboratories and Department of Experimental Oncology, Molecular Biology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Giovanna Tagliabue
- Department of Preventive and Predictive Medicine, Lombardy Cancer Registry, Varese Province, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Milena Sant
- Department of Preventive and Predictive Medicine, Analytical Epidemiology Unit, Milan
| |
Collapse
|
24
|
Liu LN, Lin YC, Miaskowski C, Chen SC, Chen ML. Association between changes in body fat and disease progression after breast cancer surgery is moderated by menopausal status. BMC Cancer 2017; 17:863. [PMID: 29254480 PMCID: PMC5735658 DOI: 10.1186/s12885-017-3869-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/29/2017] [Indexed: 01/17/2023] Open
Abstract
Background Obesity is linked to poor disease outcomes in breast cancer patients. However, this link was mostly based on body weight or BMI rather than body-fat. The purpose of this study was to evaluate the relationship between body-fat gain and disease progression in Taiwanese women after breast cancer surgery and how this relationship is influenced by menopausal status. Methods Body fat percentage was measured 1 day before and 6 months after surgery in 131 women with stages 0–III breast cancer. Disease outcomes (metastasis and death) were assessed by chart review and telephone contact 7 to 8 years after diagnosis. These data were analyzed by multivariate Cox proportional hazard model analysis. Results The percentage of women with over 5% gain in body-fat was 56% for premenopausal and 42% for postmenopausal. Rates of distant metastasis and all-cause mortality were 17.6 and 9.9%, respectively over the follow-up period. Distant metastases were predicted in postmenopausal but not premenopausal women with breast cancer by increased body fat percentage (HR = 1.3, p = 0.035), after controlling other potential covariates, including disease severity, estrogen receptor expression, progesterone receptors expression, age, and exercise habit before diagnosis. Survival was not significantly associated with body-fat percentage gains. Conclusions Our results suggest that increased body fat percentage 6 months after breast surgery is an important predictor of distant metastasis in postmenopausal Taiwanese women with breast cancer. Clinicians may need to measure patients’ body fat periodically. Our findings should be validated in studies with a longer follow-up time.
Collapse
Affiliation(s)
- Li-Ni Liu
- Department of Nursing, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yung-Chang Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Christine Miaskowski
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, USA
| | - Shin-Cheh Chen
- Department of Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Mei-Ling Chen
- Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan. .,School of Nursing, College of Medicine, Chang Gung University, No.259, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33302, Taiwan, Republic of China. .,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
25
|
Secreto G, Muti P, Sant M, Meneghini E, Krogh V. Medical ovariectomy in menopausal breast cancer patients with high testosterone levels: a further step toward tailored therapy. Endocr Relat Cancer 2017; 24:C21-C29. [PMID: 28814452 DOI: 10.1530/erc-17-0251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/16/2017] [Indexed: 11/08/2022]
Abstract
Five years of adjuvant therapy with anti-estrogens reduce the incidence of disease progression by about 50% in estrogen receptor-positive breast cancer patients, but late relapse can still occur after anti-estrogens have been discontinued. In these patients, excessive androgen production may account for renewed excessive estrogen formation and increased risks of late relapse. In the 50% of patients who do not benefit with anti-estrogens, the effect of therapy is limited by de novo or acquired resistance to treatment. Androgen receptor and epidermal growth factor receptor overexpression are recognized mechanisms of endocrine resistance suggesting the involvement of androgens as activators of the androgen receptor pathway and as stimulators of epidermal growth factor synthesis and function. Data from a series of prospective studies on operable breast cancer patients, showing high serum testosterone levels are associated to increased risk of recurrence, provide further support to a role for androgens in breast cancer progression. According to the above reported evidence, we proposed to counteract excessive androgen production in the adjuvant setting of estrogen receptor-positive patients and suggested selecting postmenopausal patients with elevated levels of serum testosterone, marker of ovarian hyperandrogenemia, for adjuvant treatment with a gonadotropins-releasing hormone analogue (medical oophorectomy) in addition to standard therapy with anti-estrogens. The proposed approach provides an attempt of personalized medicine that needs to be further investigated in clinical trials.
Collapse
Affiliation(s)
- Giorgio Secreto
- Epidemiology and Prevention UnitFondazione IRCCS - Istituto Nazionale dei Tumori, Milano, Italy
| | - Paola Muti
- Department of OncologyMcMaster University, Hamilton, Ontario, Canada
| | - Milena Sant
- Analytical Epidemiology and Health Impact UnitFondazione IRCCS, Istituto Nazionale dei Tumori, Milano, Italy
| | - Elisabetta Meneghini
- Analytical Epidemiology and Health Impact UnitFondazione IRCCS, Istituto Nazionale dei Tumori, Milano, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention UnitFondazione IRCCS - Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
26
|
Winkels RM, Sturgeon KM, Kallan MJ, Dean LT, Zhang Z, Evangelisti M, Brown JC, Sarwer DB, Troxel AB, Denlinger C, Laudermilk M, Fornash A, DeMichele A, Chodosh LA, Schmitz KH. The women in steady exercise research (WISER) survivor trial: The innovative transdisciplinary design of a randomized controlled trial of exercise and weight-loss interventions among breast cancer survivors with lymphedema. Contemp Clin Trials 2017; 61:63-72. [PMID: 28739540 PMCID: PMC5817634 DOI: 10.1016/j.cct.2017.07.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/05/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Breast cancer survivors face dual challenges: long term sequelae of treatment, and risk of recurrent disease. Obesity and a sedentary lifestyle complicate both challenges. The WISER Survivor trial assessed the effects of exercise and/or weight-loss on lymphedema, biomarkers of breast cancer recurrence, and quality of life. We report on the innovative transdisciplinary design of this trial and report attrition rates. METHODS This one year trial randomized breast cancer survivors who had a BMI of ≥25kg/m2, were sedentary and had breast-cancer-related-lymphedema to 1) exercise (weight training and aerobic exercise) 2) weight-loss 3) exercise and weight-loss 4) or control group. Innovative aspects included: adaptation of a community-based weight training program to a largely home-based program; use of a commercial meal replacement system as part of the lifestyle modification weight-loss program; inclusion of measures of cost-effectiveness to enable economic evaluations; and alignment with a parallel mouse model for breast cancer recurrence to enable transdisciplinary research. In this model, mice bearing dormant residual tumor cells, which spontaneously relapse, were placed on a high-fat diet. Overweight animals were randomly assigned to exercise, calorie restriction, both, or control group and followed for cancer recurrence. The animal model will guide mechanistic biomarkers to be tested in the human trial. RESULTS & DISCUSSION 351 participants were randomized; 13 experienced breast cancer recurrence during the trial. Of the 338 participants without recurrence, 83% completed the trial. The WISER Survivor trial will show the effects of exercise and weight-loss on lymphedema outcomes, biomarkers of recurrence and quality of life. NCT ClinicalTrials.gov registration #: NCT01515124.
Collapse
Affiliation(s)
- Renate M Winkels
- Department of Public Health Sciences, Pennsylvania State University, Hershey, PA, United States
| | - Kathleen M Sturgeon
- Department of Public Health Sciences, Pennsylvania State University, Hershey, PA, United States
| | - Michael J Kallan
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, United States
| | - Lorraine T Dean
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Zi Zhang
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, United States
| | - Margaret Evangelisti
- Center for Human Phenomic Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Justin C Brown
- Division of Population Sciences, Dana Farber Cancer Institute, Boston, MA, United States
| | - David B Sarwer
- Center for Obesity Research and Education, College of Public Health, Temple University, Philadelphia, PA, United States
| | - Andrea B Troxel
- Department of Population Health, New York University School of Medicine, New York City, NY, United States
| | - Crystal Denlinger
- Department of Hematology/Oncology, Fox Chase Cancer Centre, Philadelphia, PA, United States
| | | | - Anna Fornash
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, United States
| | - Angela DeMichele
- Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lewis A Chodosh
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kathryn H Schmitz
- Department of Public Health Sciences, Pennsylvania State University, Hershey, PA, United States.
| |
Collapse
|
27
|
Shao X, Luo L, Guo Y, Xu X, Deng D, Feng J, Ding Y, Mou H, Huang P, Shi L, Huang Y, Ye W, Lou C, Chen Z, Zheng Y, Wang X. Rs1008805 polymorphism of CYP19A1 gene is associated with the efficacy of hormone therapy in stage I-II and operable stage III breast cancer. Oncol Lett 2017; 14:6156-6162. [PMID: 29113261 DOI: 10.3892/ol.2017.6984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 05/23/2017] [Indexed: 11/06/2022] Open
Abstract
It has been hypothesized that single nucleotide polymorphisms in CYP19A1 gene may alter aromatase activity and circulating steroid hormone levels in females. Therefore, it is biologically reasonable that CYP19A1 rs1008805 (A/G) polymorphism may be associated with the clinical outcome of hormone therapy. Genotyping for the CYP19A1 rs1008805 polymorphism was performed for 287 females with hormone receptor (HR)-positive early breast cancer, and potential associations were evaluated between CYP19A1 rs1008805 genotypes and disease-free survival (DFS). Based on the analysis of the whole cohort, no significant differences were observed between rs1008805 genotypes and DFS. However, in postmenopausal females, rs1008805 variants were significantly associated with DFS (AA vs. AG vs. GG, 89.2 vs. 58.2 vs. 32.7 months; P=0.019). In addition, when the population was divided into two cohorts, females with the GG variant exhibited a significantly poorer DFS [GG vs. AA or AG, 32.7 vs. 70.6 months; hazard ratio (HR), 3.613; 95% confidence interval (CI), 1.380-9.457; P=0.005]. Furthermore, when adjusted for other patient features in multivariate analyses, GG genotype remained an independent prognostic marker for DFS (HR, 3.439; 95% CI, 1.251-9.456; P=0.017). However, there were no significant differences in DFS between patients harboring the minor allele and those with the homozygous common allele (AG or GG vs. AA, 52.4 vs. 89.2 months; HR, 1.288; 95% CI, 0.705-2.353; P=0.408). There were also no associations between rs1008805 polymorphism and DFS for premenopausal females. In conclusion, the homozygous minor allele (GG) of CYP19A1 rs1008805 was identified to be significantly associated with an inferior clinical outcome of hormone therapy in postmenopausal hormone receptor-positive patients with early breast cancer. If confirmed by further study, genotyping for CYP19A1 rs1008805 polymorphism may provide predictive information to improve the selection of endocrine treatment.
Collapse
Affiliation(s)
- Xiying Shao
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Lei Luo
- Zhejiang Institute for Food and Drug Control, Hangzhou, Zhejiang 310052, P.R. China
| | - Yong Guo
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Traditional Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaohong Xu
- Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Dehou Deng
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Jianguo Feng
- Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yuheng Ding
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Traditional Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Hanzhou Mou
- Cancer Research Institute, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Ping Huang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Lei Shi
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yuan Huang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Weiwu Ye
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Caijin Lou
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Zhanhong Chen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yabing Zheng
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiaojia Wang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
28
|
Role of Plasma D-Dimer Levels in Breast Cancer Patients and Its Correlation with Clinical and Histopathological Stage. Indian J Surg Oncol 2017; 9:307-311. [PMID: 30287988 DOI: 10.1007/s13193-017-0682-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer, the most common female malignancy, represents a heterogeneous group of tumors, which presen both varied behaviors and response to therapy. This study was done with an attempt to evaluate the role of D-dimer in patients of carcinoma breast, in predicting lymph node metastasis in carcinoma patients and to look for relationship of these markers with histopathologic parameters. Clinical data was obtained from clinical examination of patients admitted in the Department of Surgery with history of breast lump and confirmed with fine needle aspiration cytology (FNAC). Clinical staging was done using TNM staging. D-dimer level was measured prior to commencement of treatment, i.e., modified radical mastectomy (MRM) or neoadjuvant chemotherapy and finally 6 months after surgery or completion of 6 cycles of chemotherapy. The characteristics of the study population Out of 60 study cases minimum age of the patient being 30 years and maximum being 74 years. Of all histopathologic variables examined, D-dimer levels directly correlated with extent of lymph node involvement and lymphovascular invasion, D-dimer levels correlated strongest with the number of positive lymph nodes, but not with tumor size, estrogen receptor status, and progesterone receptor status. This study clearly shows that plasma D-Dimer levels are elevated in carcinoma breast patients. Increased D-Dimer levels are an important marker of clinical stage, lymphovascular invasion, lymph node involvement, and tumor metastasis.
Collapse
|
29
|
Xu H, Han Y, Lou J, Zhang H, Zhao Y, Győrffy B, Li R. PDGFRA, HSD17B4 and HMGB2 are potential therapeutic targets in polycystic ovarian syndrome and breast cancer. Oncotarget 2017; 8:69520-69526. [PMID: 29050221 PMCID: PMC5642496 DOI: 10.18632/oncotarget.17846] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 04/26/2017] [Indexed: 11/25/2022] Open
Abstract
To explore the key genes associated with both PCOS and breast cancer, we overlapped the synchronously differently expressed genes in two obese insulin-resistant GEO datasets in muscle tissue and genes exert essential roles in breast cancer prognosis together base on the following reasons: (1) Androgens excess is believed to contribute to the onset of both PCOS and breast cancer. (2) PCOS is usually complicated with metabolic symptoms, such as obesity and insulin-resistance. (3) Muscle is the main place where energy metabolism and material metabolism take place. Consequently, 53 genes were found, functionally enriched in pathways such as pyruvate metabolism, muscle system process and development of primary male sexual characteristics etc. We further lay our eyes on genes correlated with male sexual characteristics, which may be involved in the onset of both PCOS and breast cancer. Three genes were indicated to be associated with this process, including hydroxysteroid (17-beta) dehydrogenase 4/HSD17B4, platelet-derived growth factor receptor, alpha polypeptide/PDGFRA and high-mobility group box 2/HMGB2. Gene-drug interaction network about the three genes were then constructed. Drugs or chemicals that contribute to correcting the disorder of lipid metabolism were detected to restore the abnormal expression of the three genes in PCOS, such as simvastatin, bezafibrate, fenofibrate et al, which provide further choices for managing patients with PCOS.
Collapse
Affiliation(s)
- Huiyu Xu
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Peking University Third Hospital, Beijing, P.R. China
| | - Yong Han
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang Province, P.R. China
| | - Jiaying Lou
- Department of Clinical Laboratory, Renmin Hospital of Xiaoshan District, Hangzhou, Zhejiang Province, P.R. China
| | - Hongxian Zhang
- Department of Urology, Peking University Third Hospital, Beijing, P.R. China
| | - Yue Zhao
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Peking University Third Hospital, Beijing, P.R. China
| | - Balázs Győrffy
- Momentum Cancer Biomarker Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Second Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Rong Li
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Peking University Third Hospital, Beijing, P.R. China
| |
Collapse
|
30
|
Melisko ME, Goldman ME, Hwang J, De Luca A, Fang S, Esserman LJ, Chien AJ, Park JW, Rugo HS. Vaginal Testosterone Cream vs Estradiol Vaginal Ring for Vaginal Dryness or Decreased Libido in Women Receiving Aromatase Inhibitors for Early-Stage Breast Cancer. JAMA Oncol 2017; 3:313-319. [DOI: 10.1001/jamaoncol.2016.3904] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Michelle E. Melisko
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Mindy E. Goldman
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Jimmy Hwang
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Amy De Luca
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Sally Fang
- Samuel Merritt University, Oakland, California
| | - Laura J. Esserman
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Amy J. Chien
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - John W. Park
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| | - Hope S. Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco
| |
Collapse
|
31
|
Lee SA, Sung H, Han W, Noh DY, Ahn SH, Kang D. Serum adiponectin but not leptin at diagnosis as a predictor of breast cancer survival. Asian Pac J Cancer Prev 2017; 15:6137-43. [PMID: 25124587 DOI: 10.7314/apjcp.2014.15.15.6137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Limited numbers of epidemiological studies have examined the relationship between adipokines and breast cancer survival. Preoperative serum levels of obesity-related adipokines (leptin and adiponectin) were here measured in 370 breast cancer patients, recruited from two hospitals in Korea. We examined the association between those adipokines and disease-free survival (DFS). The TNM stage, ER status and histological grade were aslo assessed in relation to breast cancer survival. Elevated adiponectin levels were associated with reduced DFS of breast cancer (Ptrend=0.03) among patients with normal body weight, predominantly in postmenopausal women. There was no association of leptin with breast cancer survival. In conclusion, our study suggests that high levels of adiponectin at diagnosis are associated with breast cancer survival among women with normal body weight.
Collapse
Affiliation(s)
- Sang-Ah Lee
- Department of Preventive Medicine, Kangwon National University School of Medicine, Korea E-mail :
| | | | | | | | | | | |
Collapse
|
32
|
Associations of obesity and physical activity with serum and intratumoral sex steroid hormone levels among postmenopausal women with breast cancer: analysis of paired serum and tumor tissue samples. Breast Cancer Res Treat 2017; 162:115-125. [PMID: 28044214 DOI: 10.1007/s10549-016-4094-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/23/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE It has been hypothesized that intratumoral estrogens may play important roles in the growth of breast cancer. However, few studies have investigated such intratumoral hormones, or their association with risk factors of breast cancer. METHODS In this cross-sectional study, hormone levels in paired serum and tumor tissue samples from 146 postmenopausal women with breast cancer were measured by liquid chromatography-tandem mass spectrometry and compared between estrogen/progesterone (ER/PgR) subtypes. The associations of risk factors including body mass index (BMI) and other lifestyle factors with these hormone levels were investigated using analysis of covariance. RESULTS The level of estradiol (E2) in tumor tissue was extremely high in women with ER+ (geometric mean 95.6 pg/g) relative to women with ER-/PgR- (8.9 pg/g), whereas serum E2 level did not differ much between the two groups (3.1 and 2.8 pg/ml, respectively). Serum levels of precursors for E2, including testosterone (T) and androstenedione (Adione), and tissue Adione level, were high among women with ER+. After adjustment for confounding variables, BMI was found to be positively associated with tissue levels of E2, estrone (E1), T, and Adione among women with ER+ (P trend < 0.0001 for E2; 0.0016 for E1; 0.0002 for T; and 0.03 for Adione). CONCLUSION The data suggest that tissue E2 is related to the growth of receptor-positive breast cancer and that risk factors such as BMI affect tissue levels of E2 and its precursors. Understanding of hormonal environments within tumor tissue may be important for elucidating hormonal etiology of breast cancer and improving the prognosis of patients.
Collapse
|
33
|
Engin A. Obesity-associated Breast Cancer: Analysis of risk factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:571-606. [PMID: 28585217 DOI: 10.1007/978-3-319-48382-5_25] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several studies show that a significantly stronger association is obvious between increased body mass index (BMI) and higher breast cancer incidence. Furthermore, obese women are at higher risk of all-cause and breast cancer specific mortality when compared to non-obese women with breast cancer. In this context, increased levels of estrogens due to excessive aromatization activity of the adipose tissue, overexpression of pro-inflammatory cytokines, insulin resistance, hyperactivation of insulin-like growth factors (IGFs) pathways, adipocyte-derived adipokines, hypercholesterolemia and excessive oxidative stress contribute to the development of breast cancer in obese women. While higher breast cancer risk with hormone replacement therapy is particularly evident among lean women, in postmenopausal women who are not taking exogenous hormones, general obesity is a significant predictor for breast cancer. Moreover, increased plasma cholesterol leads to accelerated tumor formation and exacerbates their aggressiveness. In contrast to postmenopausal women, premenopausal women with high BMI are inversely associated with breast cancer risk. Nevertheless, life-style of women for breast cancer risk is regulated by avoiding the overweight and a high-fat diet. Estrogen-plus-progestin hormone therapy users for more than 5 years have elevated risks of both invasive ductal and lobular breast cancer. Additionally, these cases are more commonly node-positive and have a higher cancer-related mortality. Collectively, in this chapter, the impacts of obesity-related estrogen, cholesterol, saturated fatty acid, leptin and adiponectin concentrations, aromatase activity, leptin and insulin resistance on breast cancer patients are evaluated. Obesity-related prognostic factors of breast cancer also are discussed at molecular basis.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey. .,, Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
34
|
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
|
35
|
Rock CL, Flatt SW, Pakiz B, Quintana EL, Heath DD, Rana BK, Natarajan L. Effects of diet composition on weight loss, metabolic factors and biomarkers in a 1-year weight loss intervention in obese women examined by baseline insulin resistance status. Metabolism 2016; 65:1605-1613. [PMID: 27733248 PMCID: PMC5802865 DOI: 10.1016/j.metabol.2016.07.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Obesity is a risk factor for postmenopausal breast cancer incidence and premenopausal and postmenopausal breast cancer mortality, which may be explained by several metabolic and hormonal factors (sex hormones, insulin resistance, and inflammation) that are biologically related. Differential effects of dietary composition on weight loss and these metabolic factors may occur in insulin-sensitive vs. insulin-resistant obese women. OBJECTIVE To examine the effect of diet composition on weight loss and metabolic, hormonal and inflammatory factors in overweight/obese women stratified by insulin resistance status in a 1-year weight loss intervention. METHODS AND RESULTS Nondiabetic women who were overweight/obese (n=245) were randomly assigned to a lower fat (20% energy), higher carbohydrate (65% energy) diet; a lower carbohydrate (45% energy), higher fat (35% energy) diet; or a walnut-rich (18% energy), higher fat (35% energy), lower carbohydrate (45% energy) diet. All groups lost weight at follow-up (P<0.0001), with mean (SEM) percent loss of 9.2(1.1)% in lower fat, 6.5(0.9)% in lower carbohydrate, and 8.2(1.0)% in walnut-rich groups at 12months. The diet×time×insulin resistance status interaction was not statistically significant in the model for overall weight loss, although insulin sensitive women at 12months lost more weight in the lower fat vs. lower carbohydrate group (7.5kg vs. 4.3kg, P=0.06), and in the walnut-rich vs. lower carbohydrate group (8.1kg vs. 4.3kg, P=0.04). Sex hormone binding globulin increased within each group except in the lower carbohydrate group at 12months (P<0.01). C-reactive protein and interleukin-6 decreased at follow-up in all groups (P<0.01). CONCLUSIONS Findings provide some support for differential effects of diet composition on weight loss depending on insulin resistance status. Prescribing walnuts is associated with weight loss comparable to a standard lower fat diet in a behavioral weight loss intervention. Weight loss itself may be the most critical factor for reducing the chronic inflammation associated with increased breast cancer risk and progression.
Collapse
Affiliation(s)
- Cheryl L Rock
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Shirley W Flatt
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bilge Pakiz
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Elizabeth L Quintana
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Dennis D Heath
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brinda K Rana
- Department of Psychiatry, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Loki Natarajan
- Department of Family Medicine and Public Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
36
|
Gnagnarella P, Dragà D, Baggi F, Simoncini MC, Sabbatini A, Mazzocco K, Bassi FD, Pravettoni G, Maisonneuve P. Promoting weight loss through diet and exercise in overweight or obese breast cancer survivors (InForma): study protocol for a randomized controlled trial. Trials 2016; 17:363. [PMID: 27464488 PMCID: PMC4963992 DOI: 10.1186/s13063-016-1487-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/21/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Most women with breast cancer experience a progressive weight gain during and after treatment. Obesity is associated with an increased risk of recurrence, contralateral breast cancer, and death. Physical activity after cancer diagnosis has been reported to have positive effects on body composition and quality of life. We present the protocol of the InForma study, a trial testing the efficacy of an intervention on weight loss (≥5 % of the baseline body weight) in a group of overweight or obese breast cancer survivors. METHODS/DESIGN This is a four-arm randomized controlled trial. Patients will receive a 6-month intervention and be followed for a further 18 months. Intervention is designed to improve adherence to a healthy diet and/or to increase physical activity, taking advantage of a wrist-based activity monitor. Participants will be recruited among overweight or obese breast cancer patients treated at the European Institute of Oncology, after completion of eventual adjuvant chemotherapy and/or radiotherapy. It is envisaged that 260 patients will be randomized into four arms: Dietary Intervention; Physical Activity Intervention; Physical Activity and Dietary Intervention; and Less Intensive Intervention. Women will be offered individualized counseling consisting of face-to face discussion and phone calls in addition to group meetings. A motivational interviewing approach will be used to encourage health behavior change. All participants will be given a pedometer device to monitor their physical activity. Participants' dietary intake will be repeatedly assessed using a validated food frequency questionnaire. Participants' quality of life and anxiety will be assessed with the Functional Assessment of Cancer Therapy-Breast and the State-Trait Anxiety Inventory questionnaires. Blood samples will be collected at baseline and follow-up visits to assess lipid and hormone profiles. Body composition will be repeatedly assessed using bioelectrical impedance vector analysis for identifying changes of fat and fat-free mass. Women allocated to the less intensive intervention arm will be considered as the control group. DISCUSSION While there is a rising concern about the role of obesity in cancer recurrence and survival, this trial with its multi-arm design, motivational approach and use of a pedometer device will provide important insights regarding the most effective approach in promoting weight control in breast cancer survivors. TRIAL REGISTRATION ISRCTN53325751 (registration date: 16 October 2015); ClinicalTrials.gov NCT02622711 (registration date: 2 December 2015).
Collapse
Affiliation(s)
- Patrizia Gnagnarella
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Via Adamello, 16, 20139 Milan, Italy
| | - Daniele Dragà
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Via Adamello, 16, 20139 Milan, Italy
| | - Federica Baggi
- Physiotherapy Unit, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy
| | | | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Ketti Mazzocco
- Department of Oncology and Hemato-oncology, University of Milan, Via Festa del Perdono, 7, 20122 Milan, Italy
- Applied Research Division for Cognitive and Psychological Sciences, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Fabio Domenico Bassi
- Division of Breast Surgery, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Gabriella Pravettoni
- Department of Oncology and Hemato-oncology, University of Milan, Via Festa del Perdono, 7, 20122 Milan, Italy
- Applied Research Division for Cognitive and Psychological Sciences, European Institute of Oncology, Via Ripamonti, 435, 20141 Milan, Italy
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Via Adamello, 16, 20139 Milan, Italy
| |
Collapse
|
37
|
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
|
38
|
Aristarco V, Serrano D, Gandini S, Johansson H, Macis D, Guerrieri-Gonzaga A, Lazzeroni M, Feroce I, Pruneri G, Pagani G, Toesca A, Caldarella P, DeCensi A, Bonanni B. A Randomized, Placebo-Controlled, Phase II, Presurgical Biomarker Trial of Celecoxib Versus Exemestane in Postmenopausal Breast Cancer Patients. Cancer Prev Res (Phila) 2016; 9:349-56. [PMID: 26928670 DOI: 10.1158/1940-6207.capr-15-0311] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 02/15/2016] [Indexed: 11/16/2022]
Abstract
In breast cancer presurgical trials, the Ki-67 labeling index predicts disease outcome and offers clues to the preventive potential of drugs. We conducted a placebo-controlled trial to evaluate the activity of exemestane and celecoxib before surgery. The main endpoint was the change in Ki-67. Secondary endpoints were the modulation of circulating biomarkers. Postmenopausal women with histologically confirmed estrogen receptor-positive breast cancer were randomly assigned to exemestane 25 mg/day (n = 50), or celecoxib 800 mg/day (n = 50), or placebo (n = 25) for 6 weeks before surgery. Changes in biomarkers were analyzed through an ANCOVA model adjusting for baseline values. Exemestane showed a median absolute 10% reduction in Ki-67 [from 22 (interquartile range, IQR, 16-27), to 8 (IQR 5-18)], and a 15% absolute reduction in PgR expression [from 50 (IQR 3-90) to 15 (IQR -0-30)] after 6 weeks of treatment. Exemestane significantly increased testosterone [median change 0.21 ng/mL, (IQR 0.12-0.35)], decreased SHBG [median change -14.6 nmol/L, (IQR -23.1 to -8.6)], decreased total and HDL cholesterol by -10 mg/dL (IQR -21-2) and -7 mg/dL, (IQR -14 to -2), respectively. Triglycerides were reduced by both agents [median change -0.5 mg/dL (IQR -17.5-13.5) and -8 mg/dL (IQR -28-9) for celecoxib and exemestane, respectively]. Exemestane showed a remarkable antiproliferative effect on breast cancer, whereas celecoxib did not affect breast cancer proliferation. Given the proven preventive efficacy of exemestane, these findings support the use of Ki-67 to explore the optimal exemestane dose and schedule in the prevention setting. Cancer Prev Res; 9(5); 349-56. ©2016 AACR.
Collapse
Affiliation(s)
- Valentina Aristarco
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy.
| | - Davide Serrano
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Debora Macis
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | | | - Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Irene Feroce
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | | | - Gianmatteo Pagani
- Division of Breast Cancer Surgery, European Institute of Oncology, Milan, Italy
| | - Antonio Toesca
- Division of Breast Cancer Surgery, European Institute of Oncology, Milan, Italy
| | - Pietro Caldarella
- Division of Breast Cancer Surgery, European Institute of Oncology, Milan, Italy
| | - Andrea DeCensi
- Division of Medical Oncology, Galliera Hospital, Genoa, Italy. Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| |
Collapse
|
39
|
Associations of sex steroid hormones with mortality in women with breast cancer. Breast Cancer Res Treat 2016; 155:559-67. [PMID: 26865065 DOI: 10.1007/s10549-016-3704-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
Abstract
Epidemiological studies have demonstrated associations between circulating levels of sex steroid hormones and risk of breast cancer in postmenopausal women. However, data on associations with breast cancer survival are limited. We measured levels of estradiol, estrone, testosterone, and sex hormone-binding globulin (SHBG), in serum collected on average 30 months after diagnosis from 358 postmenopausal women diagnosed with stage I-IIIA breast cancer between 1995 and 1998 who participated in a multiethnic, prospective cohort study. Women were followed through December, 2012. We evaluated associations between log-transformed analytes and breast cancer-specific and all-cause mortality fitting multivariable Cox proportional hazards models. Over a median of 14.5 years of follow-up, 102 deaths occurred; 43 of these were due to breast cancer. In models adjusted for ethnicity/study site, age, body mass index, and tumor stage, increased levels of log-transformed SHBG were associated with reduced risk of both breast cancer-specific mortality (hazard ratio, HR 0.48; 95 % confidence interval, CI 0.26-0.89) and all-cause mortality (HR 0.64, 95 % CI 0.43-0.97). There were no associations between levels of estradiol, estrone, or testosterone for either endpoint. In subgroup analyses, after correction for multiple testing, increased estrone was significantly associated with reduced risk for breast cancer-specific mortality among participants with ER-negative tumors (HR 0.16, 95 % CI 0.05-0.63) but not among participants with ER-positive tumors. Increased serum levels of SHBG were associated with decreased risk of breast cancer-specific and all-cause mortality in women with breast cancer. These results should be confirmed in larger breast cancer survivor cohorts.
Collapse
|
40
|
Serra MC, Goldberg AP, Ryan AS. Increased depression and metabolic risk in postmenopausal breast cancer survivors. Diabetol Metab Syndr 2016; 8:44. [PMID: 27453736 PMCID: PMC4957862 DOI: 10.1186/s13098-016-0170-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/14/2016] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Breast cancer survivors (BCS) are at high risk for the development of obesity, type 2 diabetes mellitus, and metabolic syndrome. There is increasing interest in the association between depression and metabolic dysfunction, which is relevant in this population as depression is often present in the chronic phase of cancer recovery. Thus, the aim of this study was to evaluate metabolic risk in BCS with and without depression compared to non-cancer controls. METHODS African American (46 %) and Caucasian (54 %) postmenopausal BCS (N = 28; age: 60 ± 2 years; mean ± SEM) were matched for race, age (±2 years), and BMI (±2 kg/m(2)) to non-cancer controls (N = 28). Center for Epidemiologic Studies Depression Scale (CES-D) >16 or antidepressant medication usage was used to classify depression. Metabolic status was defined by 2-hr glucose during an OGTT and classification of metabolic syndrome. RESULTS Compared to non-cancer controls, BCS had similar 2-hr glucose, but higher fasting glucose and total cholesterol, and were 2.5 times more likely to have metabolic syndrome (21 vs. 52 %)(P's < 0.05). Conversely, HDL-C was 16 % higher in BCS (P < 0.05). Forty three % of BCS were on antidepressants compared to 14 % in non-cancer controls, despite similar mean CES-D scores (6 ± 1). Depressed BCS (46 %) had a higher BMI, waist circumference, fasting glucose, and more metabolic syndrome components than non-depressed BCS (P's < 0.05). CONCLUSIONS BCS have a heightened prevalence of depression that may be associated with an increased prevalence of metabolic syndrome. These results support the need to monitor weight gain, depression, and the progression of metabolic abnormalities after cancer diagnosis and treatment. Further studies into the mechanistic link between depression and metabolic disease are necessary to identify strategies that can offset their impact on obesity and associated cardiovascular risk following a breast cancer diagnosis.
Collapse
Affiliation(s)
- Monica C. Serra
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and Geriatric Research and Education Clinical Center, Baltimore VA Medical Center, 10 N Greene St. (BT/18/GR), Baltimore, MD 21201 USA
| | - Andrew P. Goldberg
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and Geriatric Research and Education Clinical Center, Baltimore VA Medical Center, 10 N Greene St. (BT/18/GR), Baltimore, MD 21201 USA
| | - Alice S. Ryan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine and Geriatric Research and Education Clinical Center, Baltimore VA Medical Center, 10 N Greene St. (BT/18/GR), Baltimore, MD 21201 USA
| |
Collapse
|
41
|
Dan TD, Wright CM, Simone NL. What benefits could caloric restriction bring to cancer patients? Future Oncol 2015; 10:2543-6. [PMID: 25531042 DOI: 10.2217/fon.14.241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Tu D Dan
- Department of Radiation Oncology, Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, 1020 Walnut Street, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
42
|
Hooper L, Abdelhamid A, Bunn D, Brown T, Summerbell CD, Skeaff CM. Effects of total fat intake on body weight. Cochrane Database Syst Rev 2015; 2016:CD011834. [PMID: 26250104 PMCID: PMC10403157 DOI: 10.1002/14651858.cd011834] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In order to prevent overweight and obesity in the general population we need to understand the relationship between the proportion of energy from fat and resulting weight and body fatness in the general population. OBJECTIVES To assess the effects of proportion of energy intake from fat on measures of weight and body fatness (including obesity, waist circumference and body mass index) in people not aiming to lose weight, using all appropriate randomised controlled trials (RCTs) and cohort studies in adults, children and young people SEARCH METHODS We searched CENTRAL to March 2014 and MEDLINE, EMBASE and CINAHL to November 2014. We did not limit the search by language. We also checked the references of relevant reviews. SELECTION CRITERIA Trials fulfilled the following criteria: 1) randomised intervention trial, 2) included children (aged ≥ 24 months), young people or adults, 3) randomised to a lower fat versus usual or moderate fat diet, without the intention to reduce weight in any participants, 4) not multifactorial and 5) assessed a measure of weight or body fatness after at least six months. We also included cohort studies in children, young people and adults that assessed the proportion of energy from fat at baseline and assessed the relationship with body weight or fatness after at least one year. We duplicated inclusion decisions and resolved disagreement by discussion or referral to a third party. DATA COLLECTION AND ANALYSIS We extracted data on the population, intervention, control and outcome measures in duplicate. We extracted measures of weight and body fatness independently in duplicate at all available time points. We performed random-effects meta-analyses, meta-regression, subgrouping, sensitivity and funnel plot analyses. MAIN RESULTS We included 32 RCTs (approximately 54,000 participants) and 30 sets of analyses of 25 cohorts. There is consistent evidence from RCTs in adults of a small weight-reducing effect of eating a smaller proportion of energy from fat; this was seen in almost all included studies and was highly resistant to sensitivity analyses. The effect of eating less fat (compared with usual diet) is a mean weight reduction of 1.5 kg (95% confidence interval (CI) -2.0 to -1.1 kg), but greater weight loss results from greater fat reductions. The size of the effect on weight does not alter over time and is mirrored by reductions in body mass index (BMI) (-0.5 kg/m(2), 95% CI -0.7 to -0.3) and waist circumference (-0.3 cm, 95% CI -0.6 to -0.02). Included cohort studies in children and adults most often do not suggest any relationship between total fat intake and later measures of weight, body fatness or change in body fatness. However, there was a suggestion that lower fat intake was associated with smaller increases in weight in middle-aged but not elderly adults, and in change in BMI in the highest validity child cohort. AUTHORS' CONCLUSIONS Trials where participants were randomised to a lower fat intake versus usual or moderate fat intake, but with no intention to reduce weight, showed a consistent, stable but small effect of low fat intake on body fatness: slightly lower weight, BMI and waist circumference compared with controls. Greater fat reduction and lower baseline fat intake were both associated with greater reductions in weight. This effect of reducing total fat was not consistently reflected in cohort studies assessing the relationship between total fat intake and later measures of body fatness or change in body fatness in studies of children, young people or adults.
Collapse
Affiliation(s)
- Lee Hooper
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK, NR4 7TJ
| | | | | | | | | | | |
Collapse
|
43
|
Hooper L, Martin N, Abdelhamid A, Davey Smith G. Reduction in saturated fat intake for cardiovascular disease. Cochrane Database Syst Rev 2015:CD011737. [PMID: 26068959 DOI: 10.1002/14651858.cd011737] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Reducing saturated fat reduces serum cholesterol, but effects on other intermediate outcomes may be less clear. Additionally it is unclear whether the energy from saturated fats that are lost in the diet are more helpfully replaced by polyunsaturated fats, monounsaturated fats, carbohydrate or protein. This review is part of a series split from and updating an overarching review. OBJECTIVES To assess the effect of reducing saturated fat intake and replacing it with carbohydrate (CHO), polyunsaturated (PUFA) or monounsaturated fat (MUFA) and/or protein on mortality and cardiovascular morbidity, using all available randomised clinical trials. SEARCH METHODS We updated our searches of the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (Ovid) and EMBASE (Ovid) on 5 March 2014. We also checked references of included studies and reviews. SELECTION CRITERIA Trials fulfilled the following criteria: 1) randomised with appropriate control group; 2) intention to reduce saturated fat intake OR intention to alter dietary fats and achieving a reduction in saturated fat; 3) not multifactorial; 4) adult humans with or without cardiovascular disease (but not acutely ill, pregnant or breastfeeding); 5) intervention at least 24 months; 6) mortality or cardiovascular morbidity data available. DATA COLLECTION AND ANALYSIS Two review authors working independently extracted participant numbers experiencing health outcomes in each arm, and we performed random-effects meta-analyses, meta-regression, subgrouping, sensitivity analyses and funnel plots. MAIN RESULTS We include 15 randomised controlled trials (RCTs) (17 comparisons, ˜59,000 participants), which used a variety of interventions from providing all food to advice on how to reduce saturated fat. The included long-term trials suggested that reducing dietary saturated fat reduced the risk of cardiovascular events by 17% (risk ratio (RR) 0.83; 95% confidence interval (CI) 0.72 to 0.96, 13 comparisons, 53,300 participants of whom 8% had a cardiovascular event, I² 65%, GRADE moderate quality of evidence), but effects on all-cause mortality (RR 0.97; 95% CI 0.90 to 1.05; 12 trials, 55,858 participants) and cardiovascular mortality (RR 0.95; 95% CI 0.80 to 1.12, 12 trials, 53,421 participants) were less clear (both GRADE moderate quality of evidence). There was some evidence that reducing saturated fats reduced the risk of myocardial infarction (fatal and non-fatal, RR 0.90; 95% CI 0.80 to 1.01; 11 trials, 53,167 participants), but evidence for non-fatal myocardial infarction (RR 0.95; 95% CI 0.80 to 1.13; 9 trials, 52,834 participants) was unclear and there were no clear effects on stroke (any stroke, RR 1.00; 95% CI 0.89 to 1.12; 8 trials, 50,952 participants). These relationships did not alter with sensitivity analysis. Subgrouping suggested that the reduction in cardiovascular events was seen in studies that primarily replaced saturated fat calories with polyunsaturated fat, and no effects were seen in studies replacing saturated fat with carbohydrate or protein, but effects in studies replacing with monounsaturated fats were unclear (as we located only one small trial). Subgrouping and meta-regression suggested that the degree of reduction in cardiovascular events was related to the degree of reduction of serum total cholesterol, and there were suggestions of greater protection with greater saturated fat reduction or greater increase in polyunsaturated and monounsaturated fats. There was no evidence of harmful effects of reducing saturated fat intakes on cancer mortality, cancer diagnoses or blood pressure, while there was some evidence of improvements in weight and BMI. AUTHORS' CONCLUSIONS The findings of this updated review are suggestive of a small but potentially important reduction in cardiovascular risk on reduction of saturated fat intake. Replacing the energy from saturated fat with polyunsaturated fat appears to be a useful strategy, and replacement with carbohydrate appears less useful, but effects of replacement with monounsaturated fat were unclear due to inclusion of only one small trial. This effect did not appear to alter by study duration, sex or baseline level of cardiovascular risk. Lifestyle advice to all those at risk of cardiovascular disease and to lower risk population groups should continue to include permanent reduction of dietary saturated fat and partial replacement by unsaturated fats. The ideal type of unsaturated fat is unclear.
Collapse
Affiliation(s)
- Lee Hooper
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK, NR4 7TJ
| | | | | | | |
Collapse
|
44
|
Shao X, Guo Y, Xu X, Zheng Y, Wang J, Chen Z, Huang J, Huang P, Cai J, Wang X. The CYP19 RS4646 polymorphism IS related to the prognosis of stage I-II and operable stage III breast cancer. PLoS One 2015; 10:e0121535. [PMID: 25793413 PMCID: PMC4368615 DOI: 10.1371/journal.pone.0121535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 02/03/2015] [Indexed: 12/03/2022] Open
Abstract
Purpose Aromatase, encoded by the CYP19 gene, catalyzes the final step of the conversion of androgens to estrogens. Given the critical role of CYP19 in estrogen synthesis, the potential influence of CYP19 rs4646 polymorphism on breast cancer survival, deserves further study. Methods Genotyping for CYP19 rs4646 variants was performed on 406 Chinese women with stage I–II and operable stage III breast cancer. Associations were evaluated between CYP19 rs4646 genotypes and disease-free survival (DFS). Results In premenopausal patients, women who are homozygous for the minor allele (AA) have a longer DFS compared with those carrying the major allele (CC or AC) (87 months versus 48.7 months; Hazard ratio (HR) = 0.56, 95 % CI = 0.318-0.985, P = 0.041). These differences were further demonstrated by a multivariate analysis (HR = 0.456, 95 % CI = 0.249-0.836, P = 0.011). Conversely, the same variant (AA) was estimated to be associated with a poorer DFS in postmenopausal women (AA versus AC or CC: 13.7 months versus 56.3 months; HR = 2.758, 95 % CI = 1.432-5.313, P = 0.002). Furthermore, the differences were confirmed by the COX proportional hazards model (HR = 2.983, 95% CI =1.494-5.955, P = 0.002). Conclusions The present study indicates that CYP19 rs4646 polymorphism is related to DFS in early breast cancer and that the prognosis index of the homozygous for the minor allele (AA) may depend on menopause status. The findings are novel, if confirmed, rs4646 genotypes may provide useful information for routine management in breast cancer.
Collapse
Affiliation(s)
- Xiying Shao
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yong Guo
- Department of Oncology, The First Affiliated Hospital of Zhejiang Traditional Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaohong Xu
- Clinical laboratory, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yabing Zheng
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jiwen Wang
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Zhanhong Chen
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jian Huang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Ping Huang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jufen Cai
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaojia Wang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- * E-mail:
| |
Collapse
|
45
|
Abstract
There is growing appreciation that the current obesity epidemic is associated with increases in cancer incidence at a population level and may lead to poor cancer outcomes; concurrent decreases in cancer mortality at a population level may represent a paradox, i.e., they may also reflect improvements in the diagnosis and treatment of cancer that mask obesity effects. An association of obesity with cancer is biologically plausible because adipose tissue is biologically active, secreting estrogens, adipokines, and cytokines. In obesity, adipose tissue reprogramming may lead to insulin resistance, with or without diabetes, and it may contribute to cancer growth and progression locally or through systemic effects. Obesity-associated changes impact cancer in a complex fashion, potentially acting directly on cells through pathways, such as the phosphoinositide 3-kinase (PI3K) and Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathways, or indirectly via changes in the tumor microenvironment. Approaches to obesity management are discussed, and the potential for pharmacologic interventions that target the obesity-cancer link is addressed.
Collapse
Affiliation(s)
- Pamela J Goodwin
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X4, Canada;
| | | |
Collapse
|
46
|
Buckland G, Travier N, Agudo A. The role of diet, weight control and physical activity in breast cancer survivors. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.14.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Recent advances in breast cancer (BC) treatment and improved screening have resulted in an increasing number of BC survivors. However, since recurrences are still a relatively common event there is a critical need to investigate modifiable factors that could impact disease recurrence and long-term prognosis. There is substantial evidence from observational studies and increasingly from randomized controlled trials, showing that weight management, increased physical activity and dietary modification may be effective methods to improve BC survival and reduce recurrences, due to their interrelated beneficial effects on systemic inflammation, circulating reproductive hormones and metabolic imbalances. Although ongoing randomized controlled trials should be able to confirm the role of these lifestyle factors on BC prognosis, further research is needed to establish specific lifestyle recommendations.
Collapse
Affiliation(s)
- Genevieve Buckland
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Noemie Travier
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Agudo
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| |
Collapse
|
47
|
Campagnoli C, Berrino F, Venturelli E, Abbà C, Biglia N, Brucato T, Cogliati P, Danese S, Donadio M, Zito G, Pasanisi P. Metformin decreases circulating androgen and estrogen levels in nondiabetic women with breast cancer. Clin Breast Cancer 2014; 13:433-8. [PMID: 24267731 DOI: 10.1016/j.clbc.2013.08.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/08/2013] [Accepted: 08/26/2013] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Diabetic patients treated with metformin have a lower risk of developing BC or a better BC prognosis. Metformin might reduce cancer growth through direct antiproliferative effects or through indirect mechanisms, particularly the reduction of insulin. In a randomized study on nondiabetic BC patients in natural menopause with high testosterone levels, we observed a significant decrease in insulin and in testosterone levels with metformin 1500 mg/d compared with 1000 mg/d. We present the results of a new analysis of our study on the effect of metformin on the bioavailability of sex hormones. PATIENTS AND METHODS One hundred twenty-four eligible women were initially invited to take metformin 500 mg/d for 3 months. The 108 women who completed the first 3 months continued the study using 1000 mg/d for 1 month. The women were then randomized into 2 groups, and, for the subsequent 5 months, 1 group increased the dose to 1500 mg/d, and the other group continued with 1000 mg/d. RESULTS Ninety-six women completed the study, 43 receiving metformin 1500 mg/day, and 53 receiving 1000 mg/day. The women receiving 1500 mg/d showed a greater and significant reduction of free testosterone (-29%) and estradiol (-38%), a borderline significant reduction of estrone and insulin-like growth factor-1, and a nonsignificant reduction of androstenedione. They also showed a nonsignificant increase of dehydroepiandrosterone sulfate. CONCLUSION Metformin does not interfere with the production of dehydroepiandrosterone sulfate. Besides, it decreases estradiol levels, basically through the reduction of testosterone. These hormonal changes might have clinical relevance.
Collapse
Affiliation(s)
- Carlo Campagnoli
- Unit of Endocrinological Gynecology, Ospedale Sant'Anna di Torino, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Goodwin PJ, Segal RJ, Vallis M, Ligibel JA, Pond GR, Robidoux A, Blackburn GL, Findlay B, Gralow JR, Mukherjee S, Levine M, Pritchard KI. Randomized trial of a telephone-based weight loss intervention in postmenopausal women with breast cancer receiving letrozole: the LISA trial. J Clin Oncol 2014; 32:2231-9. [PMID: 24934783 DOI: 10.1200/jco.2013.53.1517] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Obesity is associated with poor outcomes in women with operable breast cancer. Lifestyle interventions (LIs) that help women reduce their weight may improve outcomes. PATIENTS AND METHODS We conducted a multicenter randomized trial comparing mail-based delivery of general health information alone or combined with a 24-month standardized, telephone-based LI that included diet (500 to 1,000 kcal per day deficit) and physical activity (150 to 200 minutes of moderate-intensity physical activity per week) goals to achieve weight loss (up to 10%). Women receiving adjuvant letrozole for T1-3N0-3M0 breast cancer with a body mass index (BMI) ≥ 24 kg/m(2) were eligible. Weight was measured in the clinic, and self-report physical activity, quality-of-life (QOL), and diet questionnaires were completed. The primary outcome was disease-free survival. Accrual was terminated at 338 of 2,150 planned patients because of loss of funding. RESULTS Mean weight loss was significantly (P < .001) greater in the LI arm versus the comparison arm (4.3 v 0.6 kg or 5.3% v 0.7% at 6 months and 3.1 v 0.3 kg or 3.6% v 0.4% at 24 months) and occurred consistently across strata (BMI 24 to < 30 v ≥ 30 kg/m(2); prior v no prior adjuvant chemotherapy). Weight loss was greatest in those with higher baseline levels of moderate-intensity physical activity or improvement in QOL. Hospitalization rates and medical events were similar. CONCLUSION A telephone-based LI led to significant weight loss that was still evident at 24 months, without adverse effects on QOL, hospitalizations, or medical events. Adequately powered randomized trials with cancer end points are needed.
Collapse
Affiliation(s)
- Pamela J Goodwin
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA.
| | - Roanne J Segal
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Michael Vallis
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Jennifer A Ligibel
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Gregory R Pond
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - André Robidoux
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - George L Blackburn
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Brian Findlay
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Julie R Gralow
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Som Mukherjee
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Mark Levine
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| | - Kathleen I Pritchard
- Pamela J. Goodwin, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital and Princess Margaret Hospital; Kathleen I. Pritchard, Sunnybrook Odette Cancer Center; Pamela J. Goodwin and Kathleen I. Pritchard, University of Toronto, Toronto; Roanne J. Segal, Ottawa Hospital Regional Cancer Center, University of Ottawa, Ottawa; Gregory R. Pond, Som Mukherjee, and Mark Levine, Juravinski Hospital and Cancer Center; Brian Findlay, Niagara Health System, Walker Family Cancer Center; Gregory R. Pond, Mark Levine, Brian Findlay, and Som Mukherjee, McMaster University, Hamilton, Ontario; Michael Vallis, Dalhousie University, Halifax, Nova Scotia; André Robidoux, Centre Hospitalier de l'Université de Montréal, Montréal, Quebec, Canada; Jennifer A. Ligibel, Dana-Farber Cancer Institute; George L. Blackburn, Beth Israel Deaconess Medical Center; Jennifer A. Ligibel and George L. Blackburn, Harvard Medical School, Boston, MA; and Julie R. Gralow, University of Washington, Seattle, WA
| |
Collapse
|
49
|
Goodwin PJ. Obesity and endocrine therapy: host factors and breast cancer outcome. Breast 2014; 22 Suppl 2:S44-7. [PMID: 24074791 DOI: 10.1016/j.breast.2013.07.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Obesity is becoming increasingly prevalent and it has been linked to poor breast cancer outcomes. Because obesity is associated with increased adipose tissue mass and aromatase activity [the target of aromatase inhibitors (AIs)], there is concern that these agents may be less effective in women who are overweight or obese. Four of the randomized trials of AIs vs. tamoxifen conducted in the adjuvant breast cancer setting (ATAC, BIG 1-98 and TEAM in the postmenopausal setting and ABCSG-12 in the premenopausal setting) have reported effects of body mass index (BMI) on the relative effectiveness of an AI vs. tamoxifen. Obesity was confirmed as an adverse prognostic factor in ATAC and BIG 1-98 but not the TEAM study; in ABSCG-12, obesity was associated with poor outcomes in the anastrozole arm only. In the three postmenopausal trials, the use of an AI vs. tamoxifen was associated with better outcomes at all levels of BMI [all hazard ratios for recurrence <1, although 95% confidence intervals often included 1 due to lower power and smaller reductions in risk]. Of note, there was no significant interaction of BMI with letrozole (vs. tamoxifen) in the BIG 1-98 trial; while ATAC investigators concluded that the relative benefit of anastrozole (vs. tamoxifen) might be better in thinner (vs. heavier) women. In ABCSG-12, the use of anastrozole (vs. tamoxifen) was associated with significantly worse outcomes in women with BMI ≥25 kg/m(2) (similar to the detrimental effect of anastrozole on overall survival seen in the parent trial). These findings do not support the use of BMI as a predictor of AI (vs. tamoxifen) benefit in the adjuvant setting in postmenopausal breast cancer.
Collapse
Affiliation(s)
- Pamela J Goodwin
- Department of Medicine, Division of Medical Oncology and Hematology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; Division of Clinical Epidemiology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
50
|
Elliott KM, Dent J, Stanczyk FZ, Woodley L, Coombes RC, Purohit A, Palmieri C. Effects of aromatase inhibitors and body mass index on steroid hormone levels in women with early and advanced breast cancer. Br J Surg 2014; 101:939-48. [PMID: 24687409 DOI: 10.1002/bjs.9477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Aromatase inhibitors (AIs) are central to the management of oestrogen receptor-positive breast cancer in the adjuvant and metastatic setting. Levels of circulating steroid hormones (SHs) were measured in patients established on AIs to investigate: the influence of body mass index (BMI) in both the adjuvant and metastatic setting; the class of AI utilized in the adjuvant setting (steroidal versus non-steroidal); and differences in SH levels between women treated adjuvantly and those receiving a second-line AI for locally advanced/metastatic disease. METHODS Plasma levels of androstenedione, 5-androstene-3β,17β-diol, dehydroepiandrosterone, oestradiol and testosterone were measured by radioimmunoassay in women with breast cancer who were receiving AIs in either an adjuvant or a metastatic setting. Differences between mean SH levels by class of AI, BMI, and second-line versus adjuvant therapy were assessed. RESULTS Sixty-four women were receiving AI therapy, 45 (70 per cent) in an adjuvant setting and 19 (30 per cent) were taking a second-line AI. There was no significant correlation between BMI and SH levels. However, BMI was significantly higher in the second-line AI cohort compared with the adjuvant cohort (29.8 versus 26.2 kg/m2 respectively; P = 0.026). In the adjuvant setting, patients receiving a steroidal AI had significantly higher levels of all five hormones (P < 0.050). In the second-line AI cohort, oestradiol levels were significantly higher than in the adjuvant cohort (4.5 versus 3.3 pg/ml respectively; P = 0.022). Multivariable analysis adjusted for BMI confirmed the higher residual oestradiol level in the second-line AI group (P = 0.063) and a significantly higher androstenedione level (P = 0.022). CONCLUSION Residual levels of SH were not significantly influenced by BMI. However, the significant differences in residual SH levels between the second-line and adjuvant AI cohort is of relevance in the context of resistance to AI therapy, and warrants further investigation.
Collapse
Affiliation(s)
- K M Elliott
- Department of Biosurgery and Surgical Technology, Imperial College London, London, UK; Cancer Research UK Laboratories, Imperial Centre for Translational and Experimental Medicine, Division of Cancer, Imperial College London, London, UK
| | | | | | | | | | | | | |
Collapse
|