101
|
Mauro L, Naimo GD, Ricchio E, Panno ML, Andò S. Cross-Talk between Adiponectin and IGF-IR in Breast Cancer. Front Oncol 2015; 5:157. [PMID: 26236690 PMCID: PMC4502352 DOI: 10.3389/fonc.2015.00157] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/29/2015] [Indexed: 01/22/2023] Open
Abstract
Obesity is a chronic and multifactorial disorder that is reaching epidemic proportions. It is characterized by an enlarged mass of adipose tissue caused by a combination of size increase of preexisting adipocytes (hypertrophy) and de novo adipocyte differentiation (hyperplasia). Obesity is related to many metabolic disorders like hypertension, type 2 diabetes, metabolic syndrome, and cardiovascular disease, and it is associated with an increased risk of cancer development in different tissues including breast. Adipose tissue is now regarded as not just a storage reservoir for excess energy, but rather as an endocrine organ, secreting a large number of bioactive molecules called adipokines. Among these, adiponectin represents the most abundant adipose tissue-excreted protein, which exhibits insulin sensitizing, anti-inflammatory, and antiatherogenic properties. The serum concentrations of adiponectin are inversely correlated with body mass index. Recently, low levels of plasma adiponectin have been associated with an increased risk for obesity-related cancers and development of more aggressive phenotype, concomitantly with alterations in the bioavailability of insulin-like growth factor-I (IGF-I) and IGF-I receptor (IGF-IR) signaling pathways. In this review, we discuss the cross-talk between adiponectin/AdipoR1 and IGF-I/IGF-IR in breast cancer.
Collapse
Affiliation(s)
- Loredana Mauro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Cosenza , Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Cosenza , Italy
| | - Emilia Ricchio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Cosenza , Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Cosenza , Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Cosenza , Italy
| |
Collapse
|
102
|
Thompson HJ, Sedlacek SM, Wolfe P, Paul D, Lakoski SG, Playdon MC, McGinley JN, Matthews SB. Impact of Weight Loss on Plasma Leptin and Adiponectin in Overweight-to-Obese Post Menopausal Breast Cancer Survivors. Nutrients 2015; 7:5156-76. [PMID: 26132992 PMCID: PMC4516992 DOI: 10.3390/nu7075156] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/10/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022] Open
Abstract
Women who are obese at the time of breast cancer diagnosis have higher overall mortality than normal weight women and some evidence implicates adiponectin and leptin as contributing to prognostic disadvantage. While intentional weight loss is thought to improve prognosis, its impact on these adipokines is unclear. This study compared the pattern of change in plasma leptin and adiponectin in overweight-to-obese post-menopausal breast cancer survivors during weight loss. Given the controversies about what dietary pattern is most appropriate for breast cancer control and regulation of adipokine metabolism, the effect of a low fat versus a low carbohydrate pattern was evaluated using a non-randomized, controlled study design. Anthropometric data and fasted plasma were obtained monthly during the six-month weight loss intervention. While leptin was associated with fat mass, adiponectin was not, and the lack of correlation between leptin and adiponectin concentrations throughout weight loss implies independent mechanisms of regulation. The temporal pattern of change in leptin but not adiponectin was affected by magnitude of weight loss. Dietary pattern was without effect on either adipokine. Mechanisms not directly related to dietary pattern, weight loss, or fat mass appear to play dominant roles in the regulation of circulating levels of these adipokines.
Collapse
Affiliation(s)
- Henry J Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523-1173, USA.
| | - Scot M Sedlacek
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523-1173, USA.
- Rocky Mountain Cancer Centers, Denver, CO 80220, USA.
| | - Pamela Wolfe
- Colorado Biostatistics Consortium, University of Colorado, Denver, CO 80045, USA.
| | - Devchand Paul
- Rocky Mountain Cancer Centers, Denver, CO 80220, USA.
| | - Susan G Lakoski
- Department of Internal Medicine, University of Vermont, Burlington, VT 05405, USA.
| | - Mary C Playdon
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523-1173, USA.
- Department of Chronic Disease Epidemiology, Yale University, New Haven, CT 06520, USA.
| | - John N McGinley
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523-1173, USA.
| | - Shawna B Matthews
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523-1173, USA.
| |
Collapse
|
103
|
Schmidt S, Monk JM, Robinson LE, Mourtzakis M. The integrative role of leptin, oestrogen and the insulin family in obesity-associated breast cancer: potential effects of exercise. Obes Rev 2015; 16:473-87. [PMID: 25875578 PMCID: PMC4691342 DOI: 10.1111/obr.12281] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/06/2015] [Accepted: 02/24/2015] [Indexed: 12/13/2022]
Abstract
Obesity is an established risk factor for postmenopausal breast cancer. The mechanisms through which obesity influences the development and progression of breast cancer are not fully elucidated; however, several factors such as increased oestrogen, concentrations of various members of the insulin family and inflammation that are associated with adiposity are purported to be important factors in this relationship. Emerging research has also begun to focus on the role of adipokines, (i.e. adipocyte secreted factors), in breast cancer. Leptin secretion is directly related to adiposity and is believed to promote breast cancer directly and independently, as well as through involvement with the oestrogen and insulin signalling pathways. As leptin is secreted from white adipose tissue, any intervention that reduces adiposity may be favourable. However, it is also important to consider that energy expenditure through exercise, independent of fat loss, may improve leptin regulation. The purpose of this narrative review was to explore the role of leptin in breast cancer development and progression, identify key interactions with oestrogen and the insulin family, and distinguish the potential effects of exercise on these interactions.
Collapse
Affiliation(s)
- S Schmidt
- Department of Kinesiology, University of Waterloo, Waterloo, Canada
| | - J M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - L E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - M Mourtzakis
- Department of Kinesiology, University of Waterloo, Waterloo, Canada
| |
Collapse
|
104
|
Herlevic VC, Mowad R, Miller JK, Darensburg NA, Li BDL, Kim RH. Breast cancer outcomes in a population with high prevalence of obesity. J Surg Res 2015; 198:371-6. [PMID: 25959832 DOI: 10.1016/j.jss.2015.03.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Obesity has been associated with poor prognosis in breast cancer. However, most previous studies examined populations with relatively low proportions of obese patients. Given that forecasts predict obesity rates to exceed 50% by 2030, it is important to examine breast cancer outcomes in populations with higher rates of obesity. We hypothesized that obesity, as measured by body mass index (BMI), is associated with decreased overall survival and disease-free survival in patients with invasive breast cancer in a population with a high prevalence of obesity. METHODS A retrospective review of a prospectively maintained database was conducted on patients treated for invasive breast cancer at an academic medical center between 1997 and 2013. BMI was calculated from each patient's height and weight at the time of diagnosis. Patients were categorized as normal (BMI <25 kg/m(2)), overweight (BMI 25-30 kg/m(2)), or obese (BMI >30 kg/m(2)), as per the definitions established by the World Health Organization. The end points of overall survival and disease-free survival were analyzed. RESULTS A total of 523 patients with invasive breast cancer were included for analysis. Based on BMI, 87 (16%) were categorized as normal, 150 (29%) were overweight, and 286 (55%) were obese. The median follow-up was 49 mo. There were 16 deaths (18.4%) in normal patients, 25 (16.7 %) in overweight patients, and 45 (15.7%) in obese patients (P = 0.84). By Kaplan-Meier survival analysis, there were no differences in overall survival (P = 0.49) or in disease-free survival (P = 0.33) among the three groups. CONCLUSIONS Obesity is not associated with decreased overall or disease-free survival in a patient population with a high prevalence of obesity. These findings suggest that there may be other factors that contribute to the poor prognosis of obese breast cancer patients observed in populations with lower rates of obesity.
Collapse
Affiliation(s)
- Vincent C Herlevic
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana
| | - Ronald Mowad
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana
| | - J Karen Miller
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana
| | - Nicholas A Darensburg
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana
| | - Benjamin D L Li
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana
| | - Roger H Kim
- Department of Surgery, Louisiana State University Health Sciences Center - Shreveport and the Feist-Weiller Cancer Center, Shreveport, Louisiana.
| |
Collapse
|
105
|
Dietary lipids and adipocytes: potential therapeutic targets in cancers. J Nutr Biochem 2014; 26:303-11. [PMID: 25524629 DOI: 10.1016/j.jnutbio.2014.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/12/2014] [Accepted: 11/18/2014] [Indexed: 12/22/2022]
Abstract
Lipids play an important role to support the rapid growth of cancer cells, which can be derived from both the endogenous synthesis and exogenous supplies. Enhanced de novo fatty acid synthesis and mobilization of stored lipids in cancer cells promote tumorigenesis. Besides, lipids and fatty acids derived from diet or transferred from neighboring adipocytes also influence the proliferation and metastasis of cancer cells. Indeed, the pathogenic roles of adipocytes in the tumor microenvironment have been recognized recently. The adipocyte-derived mediators or the cross talk between adipocytes and cancer cells in the microenvironment is gaining attention. This review will focus on the impacts of lipids on cancers and the pathogenic roles of adipocytes in tumorigenesis and discuss the possible anticancer therapeutic strategies targeting lipids in the cancer cells.
Collapse
|
106
|
Madeddu C, Gramignano G, Floris C, Murenu G, Sollai G, Macciò A. Role of inflammation and oxidative stress in post-menopausal oestrogen-dependent breast cancer. J Cell Mol Med 2014; 18:2519-2529. [PMID: 25338520 PMCID: PMC4302656 DOI: 10.1111/jcmm.12413] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 07/30/2014] [Indexed: 12/19/2022] Open
Abstract
Weight gain and obesity are among the most important risk factors for post-menopausal oestrogen-dependent breast cancer (EDBC). Weight gain is associated with oxidative stress, which in turn promotes breast cancer progression. We carried out a prospective study in 216 consecutive post-menopausal breast cancer patients aiming to examine the correlations between traditional prognostic factors (tumour size, T, nodal, N, grading, G, and metastasis status, M), and body mass index (BMI), leptin, pro-inflammatory cytokines (Interleukin, IL,-6 and tumour necrosis factor-alpha, TNF-α), and oxidative stress (reactive oxygen species, ROS, glutathione peroxidase, GPx, superoxide dismutase, SOD) among patients with oestrogen receptor (ER)+ and ER- breast cancers. Distribution of T, N and M categories did not differ between ER+ and ER- breast cancer patients. ER- patients showed a higher incidence of G3 tumours. Weight, BMI, leptin, IL-6 and ROS were higher in ER+ compared with ER- patients. Among ER+ patients, BMI, leptin, IL-6 and ROS correlated with T and M. Leptin, IL-6 and ROS were positively correlated also with N. Among ER- patients, BMI and leptin did not correlate with any of prognostic parameters, whereas a positive correlation between IL-6, ROS and M was found. Multivariate regression analysis showed that BMI, leptin, IL-6 and ROS were predictive for T, N and M in ER+ patients. Weight gain, inflammation and oxidative stress are involved in EDBC prognosis. Their modulation through antidiabetic, anti-inflammatory and antioxidants drugs combined with endocrine therapy may constitute a targeted approach in post-menopausal EDBC.
Collapse
Affiliation(s)
- Clelia Madeddu
- Department of Medical Sciences “Mario Aresu”, University of CagliariCagliari, Italy
| | | | - Carlo Floris
- Department of Medical Oncology, Nuova Casa di CuraCagliari, Italy
| | - Giuseppe Murenu
- Department of Experimental Surgery, “A. Businco” Oncological HospitalCagliari, Italy
| | - Giuseppe Sollai
- Department of Oncological Surgery, “A. Businco” Oncological HospitalCagliari, Italy
| | - Antonio Macciò
- Department of Oncological Surgery, “A. Businco” Oncological HospitalCagliari, Italy
- Department of Gynecologic Oncology, “A. Businco” Oncological HospitalCagliari, Italy
| |
Collapse
|
107
|
Jiang N, Sun R, Sun Q. Leptin signaling molecular actions and drug target in hepatocellular carcinoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:2295-302. [PMID: 25484575 PMCID: PMC4238752 DOI: 10.2147/dddt.s69004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Previous reports indicate that over 13 different tumors, including hepatocellular carcinoma (HCC), are related to obesity. Obesity-associated inflammatory, metabolic, and endocrine mediators, as well as the functioning of the gut microbiota, are suspected to contribute to tumorigenesis. In obese people, proinflammatory cytokines/chemokines including tumor necrosis factor-alpha, interleukin (IL)-1 and IL-6, insulin and insulin-like growth factors, adipokines, plasminogen activator inhibitor-1, adiponectin, and leptin are found to play crucial roles in the initiation and development of cancer. The cytokines induced by leptin in adipose tissue or tumor cells have been intensely studied. Leptin-induced signaling pathways are critical for biological functions such as adiposity, energy balance, endocrine function, immune reaction, and angiogenesis as well as oncogenesis. Leptin is an activator of cell proliferation and anti-apoptosis in several cell types, and an inducer of cancer stem cells; its critical roles in tumorigenesis are based on its oncogenic, mitogenic, proinflammatory, and pro-angiogenic actions. This review provides an update of the pathological effects of leptin signaling with special emphasis on potential molecular mechanisms and therapeutic targeting, which could potentially be used in future clinical settings. In addition, leptin-induced angiogenic ability and molecular mechanisms in HCC are discussed. The stringent binding affinity of leptin and its receptor Ob-R, as well as the highly upregulated expression of both leptin and Ob-R in cancer cells compared to normal cells, makes leptin an ideal drug target for the prevention and treatment of HCC, especially in obese patients.
Collapse
Affiliation(s)
- Nan Jiang
- Shandong University School of Medicine, Jinan, Shandong Province, People's Republic of China
| | - Rongtong Sun
- Weihai Municipal Hospital, Weihai, Shandong Province, People's Republic of China
| | - Qing Sun
- Department of Pathology, QianFoShan Hospital Affiliated to Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
108
|
Growth hormone receptor inhibition decreases the growth and metastasis of pancreatic ductal adenocarcinoma. Exp Mol Med 2014; 46:e117. [PMID: 25301264 PMCID: PMC4221692 DOI: 10.1038/emm.2014.61] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 07/12/2014] [Accepted: 07/31/2014] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is the only major cancer with very low survival rates (1%). It is the fourth leading cause of cancer-related death. Hyperactivated growth hormone receptor (GHR) levels have been shown to increase the risk of cancer in general and this pathway is a master regulator of key cellular functions like proliferation, apoptosis, differentiation, metastasis, etc. However, to date there is no available data on how GHR promotes pancreatic cancer pathogenesis. Here, we used an RNA interference approach targeted to GHR to determine whether targeting GHR is an effective method for controlling pancreatic cancer growth and metastasis. For this, we used an in vitro model system consisting of HPAC and PANC-1 pancreatic cancer cells lines. GHR is upregulated in both of these cell lines and silencing GHR significantly reduced cell proliferation and viability. Inhibition of GHR also reduced the metastatic potential of pancreatic cancer cells, which was aided through decreased colony-forming ability and reduced invasiveness. Flow cytometric and western blot analyses revealed the induction of apoptosis in GHR silenced cells. GHR silencing affected phosphatidylinositol 3 kinase/AKT, mitogen extracellular signal-regulated kinase/extracellular signal-regulated kinase, Janus kinase/signal transducers and activators of transcription and mammalian target of rapamycin signaling, as well as, epithelial to mesenchymal transition. Interestingly, silencing GHR also suppressed the expression of insulin receptor-β and cyclo-oxygenease-2. Altogether, GHR silencing controls the growth and metastasis of pancreatic cancer and reveals its importance in pancreatic cancer pathogenesis.
Collapse
|
109
|
Bouguerra H, Guissouma H, Labidi S, Stambouli N, Marrakchi R, Chouaib S, Elgaaied ABA, Boussen H, Gati A. Breast Cancer in Tunisia: Association of Body Mass Index with Histopathological Aspects of Tumors. Asian Pac J Cancer Prev 2014; 15:6805-10. [DOI: 10.7314/apjcp.2014.15.16.6805] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
110
|
Pande M, Bondy ML, Do KA, Sahin AA, Ying J, Mills GB, Thompson PA, Brewster AM. Association between germline single nucleotide polymorphisms in the PI3K-AKT-mTOR pathway, obesity, and breast cancer disease-free survival. Breast Cancer Res Treat 2014; 147:381-7. [PMID: 25108739 DOI: 10.1007/s10549-014-3081-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/30/2022]
Abstract
Obesity-related hormones and cytokines alter PI3 K-AKT-mTOR pathway activation in breast tumors contributing to poorer disease-free survival (DFS) and decreased responsiveness to tamoxifen and trastuzumab. We hypothesized that single nucleotide polymorphisms (SNPs) in candidate genes in the PI3 K-AKT-mTOR signaling pathway may act as genetic modifiers of breast cancer DFS. We analyzed the association of 106 tagging SNPs in 13 genes (ADIPOQ, IGF1, INS, IRS1, LEP, LEPR, LEPROT, PIK3CA, PIK3R5, PTEN, TSC1, TSC2, and AKT1) in the P13K-AKT-mTOR pathway with DFS in a sample of 1,019 women with stage I-II breast cancer. SNPs significantly associated with DFS in any genetic model (additive, dominant, or recessive) after correcting for false discovery rate (FDR = 0.10) were included in Cox proportional hazards multivariable analyses. After adjusting for race/ethnicity, age at diagnosis, tumor stage, and treatment, rs1063539 in ADIPOQ, rs11585329 in LEPR, and rs2519757 in TSC1 were associated with improved DFS, and rs1520220 in IGF1 and rs2677760 in PIK3CA were associated with worse DFS. The associations were not significantly modified by the type of systemic treatment received or body mass index. The SNPs were not associated with tumor characteristics such as tumor size, lymph node status, nuclear grade, or hormone receptor status. In this study, germline SNPs in the PI3 K-AKT-mTOR pathway were associated with breast cancer DFS and may be potential prognostic markers. Future studies are needed to replicate our results and to evaluate the relationship between these polymorphisms and activation of the PI3 K-AKT-mTOR pathway in breast tumors.
Collapse
Affiliation(s)
- Mala Pande
- Department of Gastroenterology - Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
111
|
Zhou W, Tian Y, Gong H, Guo S, Luo C. Oncogenic role and therapeutic target of leptin signaling in colorectal cancer. Expert Opin Ther Targets 2014; 18:961-71. [PMID: 24946986 DOI: 10.1517/14728222.2014.926889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Obesity is characterized by high secretion of several cytokines from adipose tissue and is a recognized risk factor for many cancers. Among these cytokines, leptin mainly produced by adipose tissue and cancer cells is the most studied adipokine. Leptin is an activator of cell proliferation, an antiapoptotic molecule and inducer of cancer stem cells in many cell types, and its critical roles in obesity-related tumorigenesis are based on its oncogenic, mitogenic, pro-inflammatory and pro-angiogenic actions. AREAS COVERED These leptin-induced signals and action are critical for their biological effects on energy balance, adiposity, endocrine systems, immunity, angiogenesis as well as oncogenesis. This review focuses on the up-to-date knowledge on the oncogenic role of leptin signaling, clinical significance and specific drug target development in colorectal cancer (CRC). Additionally, leptin-induced angiogenic ability and molecular mechanisms in CRC cells are discussed. EXPERT OPINION Stringent binding affinity of leptin/Ob-R and overexpression of leptin/Ob-R and their targets in cancer cells make it a unique drug target for prevention and treatment of CRC, particularly in obesity colorectal patients.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Shenyang Medical College, Key Laboratory of Environmental Pollution and Microecology of Liaoning Province , No.146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034 , PR China
| | | | | | | | | |
Collapse
|
112
|
Aggressive estrogen-receptor-positive breast cancer arising in patients with elevated body mass index. Int J Clin Oncol 2014; 20:317-23. [PMID: 24913910 DOI: 10.1007/s10147-014-0712-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/14/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Obese women with estrogen receptor (ER)-positive breast cancer may experience worse disease-free and overall survival. We hypothesize that this observation is due to intrinsically aggressive disease and that obesity will be associated with higher histologic grade and Ki67. METHODS A sequential cohort of women with breast cancer diagnosed over 2 years was assembled from institutional tumor registries. Patient and tumor characteristics were abstracted from medical records; those with non-invasive tumors, or lacking body mass index (BMI), Ki67 or histologic grade data, were excluded. Univariate and multivariate analysis was performed to investigate the relationship between markers of aggressive disease (grade and Ki67) and multiple variables associated with obesity. A subgroup analysis was performed to investigate further whether ER and menopausal status influenced associations between BMI and aggressive phenotypes. RESULTS Of the 1007 patients initially identified, 668 (68 %) met the eligibility criteria. In univariate analysis, histologic grade and Ki67 were strongly associated with increased BMI, younger age, and African-American race, but less so with diabetes, hypertension, and hyperlipidemia. Multivariate analysis confirmed that higher histologic grade was associated with increased BMI (p = 0.02), and that increased Ki67 was associated with younger age (p = 0.0003) and African-American race (p = 0.002). Additional analysis found that the association between increased BMI and higher-grade tumors was particularly significant in premenopausal women with ER-positive disease. CONCLUSION This study concludes that increased BMI is associated with aggressive-phenotype breast cancer and may be particularly relevant to ER-positive breast cancer developing in premenopausal African-American women.
Collapse
|
113
|
Linking adiponectin and autophagy in the regulation of breast cancer metastasis. J Mol Med (Berl) 2014; 92:1015-23. [PMID: 24903246 DOI: 10.1007/s00109-014-1179-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 12/17/2022]
Abstract
Adipokines within the tumor microenvironment may play important roles in regulating the early steps of breast cancer metastasis. Adiponectin (AdipoQ) is the most abundant adipokine and exists in multiple forms: full-length multimers (fAd) and a cleaved, globular isoform (gAd). While these isoforms are observed as having distinct biological properties, nearly all investigation into AdipoQ in breast cancer has focused on the antitumor roles of fAd, while mostly ignoring gAd. However, evidence from other disease settings suggests that gAd is linked to processes known to promote metastasis. Here, we discuss key areas in which knowledge about AdipoQ in breast cancer is lacking, expressly focusing on data suggesting that gAd is elevated in the microenvironment and may act directly on invasive breast cancer cells to support their initial metastatic progression. We discuss autophagy as a potential mechanism of action for this effect. Overall, given that AdipoQ and AdipoQ receptor agonists have been proposed as therapeutic strategies, it is necessary to better understand the various functions of these regulatory molecules in metastatic breast cancer. Doing so will help ensure the most effective approaches to treating this disease, for which there remain no curative options.
Collapse
|
114
|
Subramani R, Lopez-Valdez R, Arumugam A, Nandy S, Boopalan T, Lakshmanaswamy R. Targeting insulin-like growth factor 1 receptor inhibits pancreatic cancer growth and metastasis. PLoS One 2014; 9:e97016. [PMID: 24809702 PMCID: PMC4014591 DOI: 10.1371/journal.pone.0097016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 04/15/2014] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers. Increasing incidence and mortality indicates that there is still much lacking in detection and management of the disease. This is partly due to a lack of specific symptoms during early stages of the disease. Several growth factor receptors have been associated with pancreatic cancer. Here, we have investigated if an RNA interference approach targeted to IGF-IR could be effective and efficient against pancreatic cancer growth and metastasis. For that, we evaluated the effects of IGF-1R inhibition using small interfering RNA (siRNAs) on tumor growth and metastasis in HPAC and PANC-1 pancreatic cancer cell lines. We found that silencing IGF-1R inhibits pancreatic cancer growth and metastasis by blocking key signaling pathways such AKT/PI3K, MAPK, JAK/STAT and EMT. Silencing IGF-1R resulted in an anti-proliferative effect in PANC-1 and HPAC pancreatic cancer cell lines. Matrigel invasion, transwell migration and wound healing assays also revealed a role for IGF-1R in metastatic properties of pancreatic cancer. These results were further confirmed using Western blotting analysis of key intermediates involved in proliferation, epithelial mesenchymal transition, migration, and invasion. In addition, soft agar assays showed that silencing IGF-1R also blocks the colony forming capabilities of pancreatic cancer cells in vitro. Western blots, as well as, flow cytometric analysis revealed the induction of apoptosis in IGF-1R silenced cells. Interestingly, silencing IGF-1R also suppressed the expression of insulin receptor β. All these effects together significantly control pancreatic cancer cell growth and metastasis. To conclude, our results demonstrate the significance of IGF-1R in pancreatic cancer.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Rebecca Lopez-Valdez
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Arunkumar Arumugam
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Sushmita Nandy
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Thiyagarajan Boopalan
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
| | - Rajkumar Lakshmanaswamy
- Center of Excellence in Cancer Research, Department of Biomedical Sciences MSB1, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States of America
- * E-mail:
| |
Collapse
|