1351
|
Kaplan HG, Malmgren JA, Atwood MK, Calip GS. Effect of treatment and mammography detection on breast cancer survival over time: 1990-2007. Cancer 2015; 121:2553-61. [PMID: 25872471 DOI: 10.1002/cncr.29371] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/14/2015] [Accepted: 03/04/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND The extent to which improvements over time in breast cancer survival are related to earlier detection by mammography or to more effective treatments is not known. METHODS At a comprehensive cancer care center, the authors conducted a retrospective cohort study of women ages 50 to 69 years who were diagnosed with invasive breast cancer (stages I through III) and were followed over 3 periods (1990-1994, 1995-1999, and 2000-2007). Data were abstracted from patient charts and included detection method, diagnosis, treatment, and follow-up for vital status in the institutional breast cancer registry (n = 2998). The method of detection was categorized as patient or physician detected or mammography detected. Cox proportional hazards models were used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for 5-year disease-specific survival in relation to detection method and treatment factors, and differences in survival were analyzed using the Kaplan-Meier method. RESULTS Fifty-eight percent of breast cancers were mammography detected, and 42% were patient or physician detected; 56% of tumors were stage I, 31% were stage II, and 13% were stage III. The average length of follow-up was 10.71 years. The combined 5-year disease-specific survival rate was 89% from 1990 to 1994, 94% from 1995 to 1999, and 96% from 2000 to 2007 (P < .001). In an adjusted model, mammography detection (HR, 0.43; 95% CI, 0.27-0.70), hormone therapy (HR, 0.47; 95% CI, 0.30-0.75), and taxane-containing chemotherapy (HR, 0.61; 95% CI, 0.37-0.99) were significantly associated with a decreased risk of disease-specific mortality. CONCLUSIONS Better breast cancer survival over time was related to mammography detection, hormone therapy, and taxane-containing chemotherapy. Treatment improvements alone are not sufficient to explain the observed survival improvements over time.
Collapse
Affiliation(s)
- Henry G Kaplan
- Department of Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Judith A Malmgren
- HealthStat Consulting, Seattle, Washington.,Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Mary K Atwood
- Department of Oncology, Swedish Cancer Institute, Seattle, Washington
| | - Gregory S Calip
- Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
1352
|
Kim Y, Yoo KY, Goodman MT. Differences in Incidence, Mortality and Survival of Breast Cancer by Regions and Countries in Asia and Contributing Factors. Asian Pac J Cancer Prev 2015; 16:2857-70. [DOI: 10.7314/apjcp.2015.16.7.2857] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
1353
|
Lucato MT, Freitas-Junior R, Moreira MA, Bernardes-Junior JR, Pinto SA, Paulinelli RR, Soares LR. Effect of tamoxifen and raloxifene on the proliferative activity of the breast epithelium in premenopausal women. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2015; 9:25-30. [PMID: 25861235 PMCID: PMC4360847 DOI: 10.4137/cmo.s22456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/26/2015] [Accepted: 01/31/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To compare the effects of tamoxifen and raloxifene on the proliferative activity of normal breast tissue in premenopausal women as measured by Ki-67/MIB-1 expression. STUDY DESIGN A total of 48 women with benign breast nodules and a recommendation for surgical removal of the lesion took part in this study. They were randomized to use tamoxifen or raloxifene for 22 days, after which they were submitted to surgery. During the surgical procedure, a 1-cm fragment of normal breast tissue was removed to study Ki-67 expression. RESULTS The mean percentage ratios between immunolabeled and non-labeled cells were 2.02 ± 1.09 and 3.13 ± 3.23 for the tamoxifen and raloxifene groups, respectively. There was no statistically significant difference between the tamoxifen (n = 16) and raloxifene (n = 14) groups in relation to the immunohistochemical analysis of Ki-67 (P = 0.205). CONCLUSION The results of this study showed no difference between tamoxifen and raloxifene with respect to the potential of these drugs to reduce the proliferative activity of the normal breast epithelium in premenopausal women.
Collapse
|
1354
|
Yuan JP, Wang LW, Qu AP, Chen JM, Xiang QM, Chen C, Sun SR, Pang DW, Liu J, Li Y. Quantum dots-based quantitative and in situ multiple imaging on ki67 and cytokeratin to improve ki67 assessment in breast cancer. PLoS One 2015; 10:e0122734. [PMID: 25856425 PMCID: PMC4391934 DOI: 10.1371/journal.pone.0122734] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/12/2015] [Indexed: 01/16/2023] Open
Abstract
Background As a marker for tumor cell proliferation, Ki67 has important impacts on breast cancer (BC) prognosis. Although immunohistochemical staining is the current standard method, variations in analytical practice make it difficult for pathologists to manually measure Ki67 index. This study was to develop a fluorescent spectrum-based quantitative analysis of Ki67 expression by quantum-dots (QDs) multiple imaging technique. Methods A QDs-based in situ multiple fluorescent imaging method was developed, which stained nuclear Ki67 as red signal and cytoplasmic cytokeratin (CK) as green signal. Both Ki67 and CK signals were automatically separated and quantified by professional spectrum analysis software. This technique was applied to tissue microarrays from 240 BC patients. Both Ki67 and CK values, and Ki67/CK ratio were obtained for each patient, and their prognostic value on 5-year disease free survival was assessed. Results This method simultaneously stains nuclear Ki67 and cytoplasmic CK with clear signal contrast, making it easy for signal separation and quantification. The total fluorescent signal intensities of both Ki67 sum and CK sum were obtained, and Ki67/CK ratio calculated. Ki67 sum and Ki67/CK ratio were each attributed into two grades by X-tile software based on the best P value principle. Multivariate analysis showed Ki67 grade (P = 0.047) and Ki67/CK grade (P = 0.004) were independent prognostic factors. Furthermore, area under curve (AUC) of ROC analysis for Ki67/CK grade (AUC: 0.683, 95%CI: 0.613–0.752) was higher than Ki67 grade (AUC: 0.665, 95%CI: 0.596–0.734) and HER-2 gene (AUC: 0.586, 95%CI: 0.510–0.661), but lower than N stage (AUC: 0.760, 95%CI: 0.696–0.823) and histological grade (AUC: 0.756, 95%CI: 0.692–0.820) on predicting the risk for recurrence. Conclusions A QDs-based quantitative and in situ multiple imaging on Ki67 and CK was developed to improve Ki67 assessment in BC, and Ki67/CK grade had better performance than Ki67 grade in predicting prognosis.
Collapse
Affiliation(s)
- Jing Ping Yuan
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Lin Wei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Ai Ping Qu
- School of Computer, Wuhan University, Wuhan, Hubei, China
| | - Jia Mei Chen
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Qing Ming Xiang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan, Hubei, China
| | - Juan Liu
- School of Computer, Wuhan University, Wuhan, Hubei, China
| | - Yan Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, Hubei, China
- * E-mail:
| |
Collapse
|
1355
|
Sehrawat A, Singh SV. Short-form RON overexpression augments benzyl isothiocyanate-induced apoptosis in human breast cancer cells. Mol Carcinog 2015; 55:473-85. [PMID: 25857724 DOI: 10.1002/mc.22295] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/26/2014] [Accepted: 01/14/2015] [Indexed: 11/08/2022]
Abstract
Chemoprevention of breast cancer is feasible with the use of non-toxic phytochemicals from edible and medicinal plants. Benzyl isothiocyanate (BITC) is one such plant compound that prevents mammary cancer development in a transgenic mouse model in association with tumor cell apoptosis. Prior studies from our laboratory have demonstrated a role for reactive oxygen species (ROS)-dependent Bax activation through the intermediary of c-Jun N-terminal kinases in BITC-induced apoptosis in human breast cancer cells. The present study demonstrates that truncated Recepteur d'Origine Nantais (sfRON) is a novel regulator of BITC-induced apoptosis in breast cancer cells. Overexpression of sfRON in MCF-7 and MDA-MB-361 cells resulted in augmentation of BITC-induced apoptosis when the apoptotic fraction was normalized against vehicle control for each cell type (untransfected and sfRON overexpressing cells). ROS generation and G2 /M phase cell cycle arrest resulting from BITC treatment were significantly attenuated in sfRON overexpressing cells after normalization with vehicle control for each cell type. Increased BITC-induced apoptosis by sfRON overexpression was independent of c-Jun N-terminal kinase or p38 mitogen-activated protein kinase hyperphosphorylation. On the other hand, activation of Bax and Bak following BITC exposure was markedly more pronounced in sfRON overexpressing cells than in controls. sfRON overexpression also augmented apoptosis induction by structurally diverse cancer chemopreventive phytochemicals including withaferin A, phenethyl isothiocyanate, and D,L-sulforaphane. In conclusion, the present study provides novel mechanistic insights into the role of sfRON in apoptosis regulation by BITC and other electrophilic phytochemicals.
Collapse
Affiliation(s)
- Anuradha Sehrawat
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
1356
|
Anitha J, Peter JD. Mammogram segmentation using maximal cell strength updation in cellular automata. Med Biol Eng Comput 2015; 53:737-49. [PMID: 25841356 DOI: 10.1007/s11517-015-1280-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 03/16/2015] [Indexed: 11/30/2022]
Abstract
Breast cancer is the most frequently diagnosed type of cancer among women. Mammogram is one of the most effective tools for early detection of the breast cancer. Various computer-aided systems have been introduced to detect the breast cancer from mammogram images. In a computer-aided diagnosis system, detection and segmentation of breast masses from the background tissues is an important issue. In this paper, an automatic segmentation method is proposed to identify and segment the suspicious mass regions of mammogram using a modified transition rule named maximal cell strength updation in cellular automata (CA). In coarse-level segmentation, the proposed method performs an adaptive global thresholding based on the histogram peak analysis to obtain the rough region of interest. An automatic seed point selection is proposed using gray-level co-occurrence matrix-based sum average feature in the coarse segmented image. Finally, the method utilizes CA with the identified initial seed point and the modified transition rule to segment the mass region. The proposed approach is evaluated over the dataset of 70 mammograms with mass from mini-MIAS database. Experimental results show that the proposed approach yields promising results to segment the mass region in the mammograms with the sensitivity of 92.25% and accuracy of 93.48%.
Collapse
Affiliation(s)
- J Anitha
- Department of Computer Science and Engineering, Karunya University, Coimbatore, India
| | | |
Collapse
|
1357
|
Quantum dots-based tissue and in vivo imaging in breast cancer researches: current status and future perspectives. Breast Cancer Res Treat 2015; 151:7-17. [PMID: 25833213 PMCID: PMC4408370 DOI: 10.1007/s10549-015-3363-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
As the most common malignant tumor for females, breast cancer (BC) is a highly heterogeneous disease regarding biological behaviors. Precisely targeted imaging on BC masses and biomarkers is critical to BC detection, treatment, monitoring, and prognostic evaluation. As an important imaging technique, quantum dots (QDs)-based imaging has emerged as a promising tool in BC researches owe to its outstanding optical properties. However, few reviews have been specifically devoted to discussing applications of QDs-based imaging in BC researches. This review summarized recent promising works in QDs-based tissue and in vivo imaging for BC studies. Physicochemical and optical properties of QDs and its potential applications were briefly described first. Then QDs-based imaging studies in BC were systematically reviewed, including tissue imaging for studying biomarkers interactions, and evaluating prognostic biomarkers, in vivo imaging for mapping axillary lymphatic system, showing BC xenograft tumor, and detecting BC metastases. At last, the future perspectives with special emphasis on the potential clinical applications have also been discussed. Potential applications of QDs-based imaging on clinical BC in the future are mainly focused on tissue study, especially in BC molecular pathology due to its optimal optical properties and quantitative information capabilities on multiple biomarkers.
Collapse
|
1358
|
Dawood S, Haaland B, Albaracin C, Gupta S, Cortes J, Sim YY, Dent RA. Is the Proportion of Patients Diagnosed with Synchronous Stage IV Breast Cancer Who Survive More than Two Years Increasing over Time? Oncology 2015; 89:79-87. [PMID: 25832113 DOI: 10.1159/000371746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/19/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Studies have shown a moderate increase in survival over time among patients with stage IV breast cancer. Median survival is approximately 2 years. The aim of this study was to evaluate trends over time in survival of >2 years of patients with synchronous stage IV disease. METHODS Using the SEER (Surveillance, Epidemiology and End Results) registry, we identified patients with synchronous stage IV breast cancer diagnosed between 1990 and 2007. Patients were divided into 3 groups according to the year of diagnosis (1990-1995, 1996-2000, and 2001-2007). The probability of survival of >2 years was computed within each group of diagnosis years. A multivariate logistic regression model was then fit to determine the association between year of diagnosis and probability of surviving >2 years after adjusting for important prognostic factors. RESULTS 22,601 patients were identified, of whom 9,435 (41.7%) had an overall survival of >2 years. The probability of breast cancer-specific survival (BCSS) of >2 years was 40.1, 44, and 48.1% among patients diagnosed in the periods 1990-1995, 1996-2000, and 2001-2007, respectively (p < 0.001). In the multivariate model, the probability of BCSS of >2 years increased with the more recent year of diagnosis (OR 1.058, 95% CI 1.046-1.069, p < 0.001). Other factors significantly associated with an increased probability of surviving >2 years included surgery of the primary tumor, lower grade, younger age, hormone receptor-positive disease, and noninflammatory disease. CONCLUSION Our results indicate that among the patients with synchronous stage IV breast cancer, the probability of BCSS of >2 years in the US has increased over time. Attributable factors may be the increasing number of efficacious agents and improved palliative care services over time.
Collapse
Affiliation(s)
- Shaheenah Dawood
- Department of Medical Oncology, Dubai Hospital, Dubai, United Arab Emirates
| | | | | | | | | | | | | |
Collapse
|
1359
|
Reaves DK, Ginsburg E, Bang JJ, Fleming JM. Persistent organic pollutants and obesity: are they potential mechanisms for breast cancer promotion? Endocr Relat Cancer 2015; 22:R69-86. [PMID: 25624167 PMCID: PMC4352112 DOI: 10.1530/erc-14-0411] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dietary ingestion of persistent organic pollutants (POPs) is correlated with the development of obesity. Obesity alters metabolism, induces an inflammatory tissue microenvironment, and is also linked to diabetes and breast cancer risk/promotion of the disease. However, no direct evidence exists with regard to the correlation among all three of these factors (POPs, obesity, and breast cancer). Herein, we present results from current correlative studies indicating a causal link between POP exposure through diet and their bioaccumulation in adipose tissue that promotes the development of obesity and ultimately influences breast cancer development and/or progression. Furthermore, as endocrine disruptors, POPs could interfere with hormonally responsive tissue functions causing dysregulation of hormone signaling and cell function. This review highlights the critical need for advanced in vitro and in vivo model systems to elucidate the complex relationship among obesity, POPs, and breast cancer, and, more importantly, to delineate their multifaceted molecular, cellular, and biochemical mechanisms. Comprehensive in vitro and in vivo studies directly testing the observed correlations as well as detailing their molecular mechanisms are vital to cancer research and, ultimately, public health.
Collapse
Affiliation(s)
- Denise K Reaves
- Department of BiologyNorth Carolina Central University, MTSC Room 2247, 1801 Fayetteville Street, Durham, North Carolina 27707, USANational Cancer InstituteNational Institutes of Health, Center for Cancer Training, Bethesda, Maryland 20892, USADepartment of BiologyNorth Carolina Central University, Durham, North Carolina 27707, USA
| | - Erika Ginsburg
- Department of BiologyNorth Carolina Central University, MTSC Room 2247, 1801 Fayetteville Street, Durham, North Carolina 27707, USANational Cancer InstituteNational Institutes of Health, Center for Cancer Training, Bethesda, Maryland 20892, USADepartment of BiologyNorth Carolina Central University, Durham, North Carolina 27707, USA
| | - John J Bang
- Department of BiologyNorth Carolina Central University, MTSC Room 2247, 1801 Fayetteville Street, Durham, North Carolina 27707, USANational Cancer InstituteNational Institutes of Health, Center for Cancer Training, Bethesda, Maryland 20892, USADepartment of BiologyNorth Carolina Central University, Durham, North Carolina 27707, USA
| | - Jodie M Fleming
- Department of BiologyNorth Carolina Central University, MTSC Room 2247, 1801 Fayetteville Street, Durham, North Carolina 27707, USANational Cancer InstituteNational Institutes of Health, Center for Cancer Training, Bethesda, Maryland 20892, USADepartment of BiologyNorth Carolina Central University, Durham, North Carolina 27707, USA
| |
Collapse
|
1360
|
Soni A, Ren Z, Hameed O, Chanda D, Morgan CJ, Siegal GP, Wei S. Breast cancer subtypes predispose the site of distant metastases. Am J Clin Pathol 2015; 143:471-8. [PMID: 25779997 DOI: 10.1309/ajcpyo5fsv3upexs] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES The distant organs to which breast cancer preferentially metastasizes are of significant clinical importance. METHODS We explored the relationship between the clinicopathologic factors and the common sites of distant metastasis in 531 consecutive patients with advanced breast cancer. RESULTS Breast cancer subtype as a variable was significantly associated with all five common sites of relapse by multivariate analysis. The luminal tumors were remarkable for their significant bone-seeking phenotype and were less frequently observed in lung, brain, and pleural metastases and less likely to be associated with multiorgan relapse. The HER2 subtype demonstrated a significant liver-homing characteristic. African Americans were significantly less likely to have brain-only metastasis in patients with brain relapse. CONCLUSIONS These findings further articulate that breast cancer subtypes differ not only in tumor characteristics but also in their metastatic behavior, thus raising the possibility that this knowledge could potentially be used in determining the appropriate strategy for follow-up of patients with newly diagnosed breast cancer.
Collapse
Affiliation(s)
- Abha Soni
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Nashville, TN
| | - Zhiyong Ren
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Nashville, TN
| | - Omar Hameed
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Diptiman Chanda
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Nashville, TN
| | - Charity J. Morgan
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Nashville, TN
| | - Gene P. Siegal
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Nashville, TN
| | - Shi Wei
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Nashville, TN
| |
Collapse
|
1361
|
McLean LS, Watkins CN, Campbell P, Zylstra D, Rowland L, Amis LH, Scott L, Babb CE, Livingston WJ, Darwanto A, Davis WL, Senthil M, Sowers LC, Brantley E. Aryl Hydrocarbon Receptor Ligand 5F 203 Induces Oxidative Stress That Triggers DNA Damage in Human Breast Cancer Cells. Chem Res Toxicol 2015; 28:855-71. [PMID: 25781201 DOI: 10.1021/tx500485v] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Breast tumors often show profound sensitivity to exogenous oxidative stress. Investigational agent 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203) induces aryl hydrocarbon receptor (AhR)-mediated DNA damage in certain breast cancer cells. Since AhR agonists often elevate intracellular oxidative stress, we hypothesize that 5F 203 increases reactive oxygen species (ROS) to induce DNA damage, which thwarts breast cancer cell growth. We found that 5F 203 induced single-strand break formation. 5F 203 enhanced oxidative DNA damage that was specific to breast cancer cells sensitive to its cytotoxic actions, as it did not increase oxidative DNA damage or ROS formation in nontumorigenic MCF-10A breast epithelial cells. In contrast, AhR agonist and procarcinogen benzo[a]pyrene and its metabolite, 1,6-benzo[a]pyrene quinone, induced oxidative DNA damage and ROS formation, respectively, in MCF-10A cells. In sensitive breast cancer cells, 5F 203 activated ROS-responsive kinases: c-Jun-N-terminal kinase (JNK) and p38 mitogen activated protein kinase (p38). AhR antagonists (alpha-naphthoflavone, CH223191) or antioxidants (N-acetyl-l-cysteine, EUK-134) attenuated 5F 203-mediated JNK and p38 activation, depending on the cell type. Pharmacological inhibition of AhR, JNK, or p38 attenuated 5F 203-mediated increases in intracellular ROS, apoptosis, and single-strand break formation. 5F 203 induced the expression of cytoglobin, an oxidative stress-responsive gene and a putative tumor suppressor, which was diminished with AhR, JNK, or p38 inhibition. Additionally, 5F 203-mediated increases in ROS production and cytoglobin were suppressed in AHR100 cells (AhR ligand-unresponsive MCF-7 breast cancer cells). Our data demonstrate 5F 203 induces ROS-mediated DNA damage at least in part via AhR, JNK, or p38 activation and modulates the expression of oxidative stress-responsive genes such as cytoglobin to confer its anticancer action.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lawrence C Sowers
- ⊥Department of Pharmacology and Toxicology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555, United States
| | | |
Collapse
|
1362
|
Chalasani P, Marron M, Roe D, Clarke K, Iannone M, Livingston RB, Shan JS, Stopeck AT. A phase I clinical trial of bavituximab and paclitaxel in patients with HER2 negative metastatic breast cancer. Cancer Med 2015; 4:1051-9. [PMID: 25826750 PMCID: PMC4529343 DOI: 10.1002/cam4.447] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 12/20/2022] Open
Abstract
Bavituximab is a chimeric monoclonal antibody that targets phosphatidylserine (PS). PS is externalized on cells in the tumor microenvironment when exposed to hypoxia and/or other physiological stressors. On attaching to PS, bavituximab is thought to promote antitumor immunity through its effects on PS receptors in monocytes, and myeloid-derived suppressor cells, as well as trigger antitumor effects by inducing an antibody-dependent cellular cytotoxicity on tumor-associated endothelial cells. We conducted a phase I clinical trial of bavituximab in combination with paclitaxel in patients with HER2-negative metastatic breast cancer. Patients were treated with weekly paclitaxel (80 mg/m2 for 3/4 weeks) and weekly bavituximab (3 mg/kg for 4/4 weeks). Correlative studies included the measurement of circulating microparticles, endothelial cells, and apoptotic tumor cells by flow cytometry. Fourteen patients with metastatic breast cancer were enrolled; all were evaluable for toxicity and 13 were evaluable for response. Treatment resulted in an overall response rate (RR) of 85% with a median progression-free survival (PFS) of 7.3 months. Bone pain, fatigue, headache, and neutropenia were the most common adverse effects. Infusion-related reactions were the most common adverse event related to bavituximab therapy. Correlative studies showed an increase in the PS-expressing apoptotic circulating tumor cells in response to bavituximab, but not with paclitaxel. No changes in the number of circulating endothelial cells or apoptotic endothelial cells were observed with therapy. Platelet and monocyte-derived microparticles decreased after initiation of bavituximab. Bavituximab in combination with paclitaxel is well tolerated for treatment of patients with metastatic breast cancer with promising results observed in terms of clinical RRs and PFS. The toxicity profile of bavituximab is notable for manageable infusion-related reactions with no evidence for increased thrombogenicity. Recent preclinical data suggest that bavituximab can also promote antitumor immune activity that should be explored in future clinical trials.
Collapse
Affiliation(s)
- Pavani Chalasani
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Marilyn Marron
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Denise Roe
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Kathryn Clarke
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Maria Iannone
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Robert B Livingston
- University of Arizona Cancer Center, 1515 N Campbell Ave, Tucson, Arizona, 85724
| | - Joseph S Shan
- Peregrine Pharmaceuticals, Inc., 14282 Franklin Avenue, Tustin, California, 92780
| | - Alison T Stopeck
- Stonybrook Medicine University, PO Box 1554, Stonybrook, New York, 11790
| |
Collapse
|
1363
|
Kim EK, Tang Y, Kim YS, Hwang JW, Choi EJ, Lee JH, Lee SH, Jeon YJ, Park PJ. First evidence that Ecklonia cava-derived dieckol attenuates MCF-7 human breast carcinoma cell migration. Mar Drugs 2015; 13:1785-97. [PMID: 25830682 PMCID: PMC4413187 DOI: 10.3390/md13041785] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/09/2015] [Accepted: 03/18/2015] [Indexed: 12/23/2022] Open
Abstract
We investigated the effect of Ecklonia cava (E. cava)-derived dieckol on movement behavior and the expression of migration-related genes in MCF-7 human breast cancer cell. Phlorotannins (e.g., dieckol, 6,6'-biecko, and 2,7″-phloroglucinol-6,6'-bieckol) were purified from E. cava by using centrifugal partition chromatography. Among the phlorotannins, we found that dieckol inhibited breast cancer cell the most and was selected for further study. Radius™-well was used to assess cell migration, and dieckol (1-100 µM) was found to suppress breast cancer cell movement. Metastasis-related gene expressions were evaluated by RT-PCR and Western blot analysis. In addition, dieckol inhibited the expression of migration-related genes such as matrix metalloproteinase (MMP)-9 and vascular endothelial growth factor (VEGF). On the other hand, it stimulated the expression of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2. These results suggest that dieckol exerts anti-breast cancer activity via the regulation of the expressions of metastasis-related genes, and this is the first report on the anti-breast cancer effect of dieckol.
Collapse
Affiliation(s)
- Eun-Kyung Kim
- Division of Food Bio Science, College of Biomedical and Health Sciences, Konkuk University, Chungju 380-701, Korea.
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
| | - Yujiao Tang
- Division of Food Bio Science, College of Biomedical and Health Sciences, Konkuk University, Chungju 380-701, Korea.
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
| | - Yon-Suk Kim
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
- Department of Biotechnology, Konkuk University, Chungju 380-701, Korea.
| | - Jin-Woo Hwang
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
- Department of Biotechnology, Konkuk University, Chungju 380-701, Korea.
| | - Eun-Ju Choi
- Division of Sport Science, Konkuk University, Chungju, 380-701, Korea.
| | - Ji-Hyeok Lee
- Department of Marine Life Science, Jeju National University, Jeju 690-756, Korea.
| | - Seung-Hong Lee
- Division of Food Bio Science, College of Biomedical and Health Sciences, Konkuk University, Chungju 380-701, Korea.
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 690-756, Korea.
| | - Pyo-Jam Park
- Korea Nokyong Research Center, Konkuk University, Chungju 380-701, Korea.
- Department of Biotechnology, Konkuk University, Chungju 380-701, Korea.
| |
Collapse
|
1364
|
Necela BM, Crozier JA, Andorfer CA, Lewis-Tuffin L, Kachergus JM, Geiger XJ, Kalari KR, Serie DJ, Sun Z, Aspita AM, O’Shannessy DJ, Maltzman JD, McCullough AE, Pockaj BA, Cunliffe HE, Ballman KV, Thompson EA, Perez EA. Folate receptor-α (FOLR1) expression and function in triple negative tumors. PLoS One 2015; 10:e0122209. [PMID: 25816016 PMCID: PMC4376802 DOI: 10.1371/journal.pone.0122209] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022] Open
Abstract
Folate receptor alpha (FOLR1) has been identified as a potential prognostic and therapeutic target in a number of cancers. A correlation has been shown between intense overexpression of FOLR1 in breast tumors and poor prognosis, yet there is limited examination of the distribution of FOLR1 across clinically relevant breast cancer subtypes. To explore this further, we used RNA-seq data from multiple patient cohorts to analyze the distribution of FOLR1 mRNA across breast cancer subtypes comprised of estrogen receptor positive (ER+), human epidermal growth factor receptor positive (HER2+), and triple negative (TNBC) tumors. FOLR1 expression varied within breast tumor subtypes; triple negative/basal tumors were significantly associated with increased expression of FOLR1 mRNA, compared to ER+ and HER2+ tumors. However, subsets of high level FOLR1 expressing tumors were observed in all clinical subtypes. These observations were supported by immunohistochemical analysis of tissue microarrays, with the largest number of 3+ positive tumors and highest H-scores of any subtype represented by triple negatives, and lowest by ER+ tumors. FOLR1 expression did not correlate to common clinicopathological parameters such as tumor stage and nodal status. To delineate the importance of FOLR1 overexpression in triple negative cancers, RNA-interference was used to deplete FOLR1 in overexpressing triple negative cell breast lines. Loss of FOLR1 resulted in growth inhibition, whereas FOLR1 overexpression promoted folate uptake and growth advantage in low folate conditions. Taken together, our data suggests patients with triple negative cancers expressing high FOLR1 expression represent an important population of patients that may benefit from targeted anti-FOLR1 therapy. This may prove particularly helpful for a large number of patients who would typically be classified as triple negative and who to this point have been left without any targeted treatment options.
Collapse
Affiliation(s)
- Brian M. Necela
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Jennifer A. Crozier
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Cathy A. Andorfer
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Laura Lewis-Tuffin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Jennifer M. Kachergus
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| | - Xochiquetzal J. Geiger
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Krishna R. Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Daniel J. Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida United States of America
| | - Zhifu Sun
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alvaro Moreno Aspita
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Daniel J. O’Shannessy
- Department of Translational Medicine and Diagnostics, Morphotek, Exton, Pennsylvania, United States of America
| | - Julia D. Maltzman
- Department of Clinical Development, Morphotek, Exton, Pennsylvania, United States of America
| | - Ann E. McCullough
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Barbara A. Pockaj
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Heather E. Cunliffe
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Karla V. Ballman
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - E. Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
- * E-mail:
| | - Edith A. Perez
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, United Sates of America
| |
Collapse
|
1365
|
Wang T, Ma Y, Wang L, Liu H, Chen M, Niu R. Strong adverse effect of epidermal growth factor receptor 2 overexpression on prognosis of patients with invasive lobular breast cancer: a comparative study with invasive ductal breast cancer in Chinese population. Tumour Biol 2015; 36:6113-24. [DOI: 10.1007/s13277-015-3293-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/26/2015] [Indexed: 12/31/2022] Open
|
1366
|
Khankari NK, Bradshaw PT, Steck SE, He K, Olshan AF, Shen J, Ahn J, Chen Y, Ahsan H, Terry MB, Teitelbaum SL, Neugut AI, Santella RM, Gammon MD. Dietary intake of fish, polyunsaturated fatty acids, and survival after breast cancer: A population-based follow-up study on Long Island, New York. Cancer 2015; 121:2244-52. [PMID: 25809414 DOI: 10.1002/cncr.29329] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 01/24/2015] [Accepted: 02/02/2015] [Indexed: 11/11/2022]
Abstract
BACKGROUND In laboratory experiments, ω-3 polyunsaturated fatty acids (PUFAs) have been found to reduce inflammatory eicosanoids resulting from ω-6 PUFA metabolism via competitive inhibition, and the ω-3-induced cytotoxic environment increases apoptosis and reduces cell growth in breast cancer cells. To the authors' knowledge, epidemiologic investigations regarding whether dietary ω-3 PUFA intake benefits survival after breast cancer are limited and inconsistent. METHODS The authors used resources from a population-based follow-up study conducted on Long Island, New York, among 1463 women newly diagnosed with first primary breast cancer who were interviewed an average of approximately 3 months after diagnosis to assess risk and prognostic factors, including dietary intake (using a food frequency questionnaire). Vital status was determined through 2011, yielding a median follow-up of 14.7 years and 485 deaths. Adjusted hazard ratios (HRs) and 95% confidence intervals (95% CIs) were estimated using Cox proportional hazards regression. RESULTS All-cause mortality was reduced among women with breast cancer reporting the highest quartile of intake (compared with never) for tuna (HR, 0.71; 95% CI, 0.55-0.92), other baked/broiled fish (HR, 0.75; 95% CI, 0.58-0.97), and the dietary long-chain ω-3 PUFAs docosahexaenoic acid (HR, 0.71; 95% CI, 0.55-0.92) and eicosapentaenoic acid (HR, 0.75; 95% CI, 0.58-0.97). CONCLUSIONS All-cause mortality was reduced by 16% to 34% among women with breast cancer who reported a high intake of fish and long-chain ω-3 PUFAs. Long-chain ω-3 PUFA intake from fish and other dietary sources may provide a potential strategy to improve survival after breast cancer.
Collapse
Affiliation(s)
- Nikhil K Khankari
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Patrick T Bradshaw
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Susan E Steck
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, South Carolina
| | - Ka He
- Department of Epidemiology and Biostatistics, Indiana University, Bloomington, Indiana
| | - Andrew F Olshan
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Jing Shen
- Department of Environmental Health, Columbia University, New York, New York
| | - Jiyoung Ahn
- Department of Population Health, New York University, New York, New York.,Department of Environmental Medicine, New York University, New York, New York
| | - Yu Chen
- Department of Population Health, New York University, New York, New York.,Department of Environmental Medicine, New York University, New York, New York
| | - Habibul Ahsan
- Department of Health Studies and Comprehensive Cancer Center, University of Chicago, Chicago, Illinois
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, New York
| | - Susan L Teitelbaum
- Department of Preventive Medicine, Mount Sinai School of Medicine, New York, New York
| | - Alfred I Neugut
- Department of Epidemiology, Columbia University, New York, New York.,Department of Medicine, Columbia University, New York, New York
| | - Regina M Santella
- Department of Environmental Health, Columbia University, New York, New York
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
1367
|
Gambogic Acid lysinate induces apoptosis in breast cancer mcf-7 cells by increasing reactive oxygen species. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:842091. [PMID: 25866542 PMCID: PMC4381976 DOI: 10.1155/2015/842091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/02/2014] [Accepted: 12/15/2014] [Indexed: 11/17/2022]
Abstract
Gambogic acid (GA) inhibits the proliferation of various human cancer cells. However, because of its water insolubility, the antitumor efficacy of GA is limited. Objectives. To investigate the antitumor activity of gambogic acid lysinate (GAL) and its mechanism. Methods. Inhibition of cell proliferation was determined by MTT assay; intracellular ROS level was detected by staining cells with DCFH-DA; cell apoptosis was determined by flow cytometer and the mechanism of GAL was investigated by Western blot. Results. GAL inhibited the proliferation of MCF-7 cells with IC50 values 1.46 μmol/L comparable with GA (IC50, 1.16 μmol/L). GAL promoted the production of ROS; however NAC could remove ROS and block the effect of GAL. GAL inhibited the expression of SIRT1 but increased the phosphorylation of FOXO3a and the expression of p27Kip1. At knockdown of FOXO3a, cell apoptosis induced by GAL can be partly blocked. In addition it also enhanced the cleavage of caspase-3. Conclusions. GAL inhibited MCF-7 cell proliferation and induced MCF-7 cell apoptosis by increasing ROS level which could induce cell apoptosis by both SIRT1/FOXO3a/p27Kip1 and caspase-3 signal pathway. These results suggested that GAL might be useful as a modulation agent in cancer chemotherapy.
Collapse
|
1368
|
Choron RL, Chang S, Khan S, Villalobos MA, Zhang P, Carpenter JP, Tulenko TN, Liu Y. Paclitaxel impairs adipose stem cell proliferation and differentiation. J Surg Res 2015; 196:404-15. [PMID: 25891676 DOI: 10.1016/j.jss.2015.03.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/20/2015] [Accepted: 03/12/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer patients with chemotherapy-induced immunosuppression have poor surgical site wound healing. Prior literature supports the use of human adipose-derived stem cell (hASC) lipoinjection to improve wound healing. It has been established that multipotent hASCs facilitate neovascularization, accelerate epithelialization, and quicken wound closure in animal models. Although hASC wound therapy may benefit surgical cancer patients, the chemotherapeutic effects on hASCs are unknown. We hypothesized that paclitaxel, a chemotherapeutic agent, impairs hASC growth, multipotency, and induces apoptosis. METHODS hASCs were isolated and harvested from consented, chemotherapy and radiation naive patients. Growth curves, MTT (3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide), and EdU (5-ethynyl-2-deoxyguridine) assays measured cytotoxicity and proliferation. Oil Red O stain, Alizarin Red stain, matrigel tube formation assay, and quantitative polymerase chain reaction analyzed hASC differentiation. Annexin V assay measured apoptosis. Immunostaining and Western blot determined tumor necrosis factor α (TNF-α) expression. RESULTS hASCs were selectively more sensitive to paclitaxel (0.01-30 μM) than fibroblasts (P < 0.05). After 12 d, paclitaxel caused hASC growth arrest, whereas control hASCs proliferated (P = 0.006). Paclitaxel caused an 80.6% reduction in new DNA synthesis (P < 0.001). Paclitaxel severely inhibited endothelial differentiation and capillary-like tube formation. Differentiation markers, lipoprotein lipase (adipogenic), alkaline phosphatase (osteogenic), CD31, and van Willebrand factor (endothelial), were significantly decreased (all P < 0.05) confirming paclitaxel impaired differentiation. Paclitaxel was also found to induce apoptosis and TNF-α was upregulated in paclitaxel-treated hASCs (P < 0.001). CONCLUSIONS Paclitaxel is more cytotoxic to hASCs than fibroblasts. Paclitaxel inhibits hASC proliferation, differentiation, and induces apoptosis, possibly through the TNF-α pathway. Paclitaxel's severe inhibition of endothelial differentiation indicates neovascularization disruption, possibly causing poor wound healing in cancer patients receiving chemotherapy.
Collapse
Affiliation(s)
- Rachel L Choron
- Department of Surgery, Cooper University Hospital, Camden, New Jersey.
| | - Shaohua Chang
- Department of Surgical Research, Cooper University Hospital, Camden, New Jersey
| | - Sophia Khan
- Department of Surgery, Cooper University Hospital, Camden, New Jersey
| | | | - Ping Zhang
- Department of Surgical Research, Cooper University Hospital, Camden, New Jersey
| | | | - Thomas N Tulenko
- Department of Surgical Research, Cooper University Hospital, Camden, New Jersey
| | - Yuan Liu
- Department of Plastic Surgery, Cooper University Hospital, Camden, New Jersey
| |
Collapse
|
1369
|
Zhao G, Shi A, Fan Z, Du Y. Salidroside inhibits the growth of human breast cancer in vitro and in vivo. Oncol Rep 2015; 33:2553-60. [PMID: 25814002 DOI: 10.3892/or.2015.3857] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/26/2015] [Indexed: 11/05/2022] Open
Abstract
Salidroside has been identified as one of the most potent compounds isolated from the plant Rhodiola rosea, and was found to have several important biological properties, including antioxidant and anti-inflammatory activity; however, its anticancer effects are poorly understood. Thus, the present study focused on evaluating the effects of purified salidroside on the growth of human breast cancer in vitro and in vivo, and on further investigating its possible molecular mechanisms. The human breast cancer cell line, MCF-7, was incubated with various concentrations of salidroside, and cell proliferation, colony formation, cell cycle distribution, apoptosis, migration and invasion were assayed by several in vitro approaches. As a result, it was found that salidroside treatment significantly inhibited cell proliferation, colony formation, migration and invasion, as well as induced cell apoptosis and cell cycle arrest at the G0/G1 phase in vitro. In addition, we also evaluated the effect of salidroside on tumor growth in a nude mouse model, and found that salidroside treatment significantly suppressed tumor growth in vivo. We also further disclosed that salidroside treatment significantly inhibited the intracellular reactive oxygen species (ROS) formation and MAPK pathway activation, which may contribute to the inhibition of tumor growth of breast cancer and reduction of oxidative stress. In conclusion, these findings suggest that salidroside may be a promising candidate target for the prevention and treatment of human breast cancer.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Aiping Shi
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ye Du
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
1370
|
KONG XIANGNAN, DING XIA, YANG QIFENG. Identification of multi-target effects of Huaier aqueous extract via microarray profiling in triple-negative breast cancer cells. Int J Oncol 2015; 46:2047-56. [DOI: 10.3892/ijo.2015.2932] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/16/2015] [Indexed: 11/06/2022] Open
|
1371
|
Li JP, Yang YX, Liu QL, Pan ST, He ZX, Zhang X, Yang T, Chen XW, Wang D, Qiu JX, Zhou SF. The investigational Aurora kinase A inhibitor alisertib (MLN8237) induces cell cycle G2/M arrest, apoptosis, and autophagy via p38 MAPK and Akt/mTOR signaling pathways in human breast cancer cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1627-52. [PMID: 25834401 PMCID: PMC4365748 DOI: 10.2147/dddt.s75378] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alisertib (ALS) is an investigational potent Aurora A kinase inhibitor currently undergoing clinical trials for the treatment of hematological and non-hematological malignancies. However, its antitumor activity has not been tested in human breast cancer. This study aimed to investigate the effect of ALS on the growth, apoptosis, and autophagy, and the underlying mechanisms in human breast cancer MCF7 and MDA-MB-231 cells. In the current study, we identified that ALS had potent growth-inhibitory, pro-apoptotic, and pro-autophagic effects in MCF7 and MDA-MB-231 cells. ALS arrested the cells in G2/M phase in MCF7 and MDA-MB-231 cells which was accompanied by the downregulation of cyclin-dependent kinase (CDK)1/cell division cycle (CDC) 2, CDK2, and cyclin B1 and upregulation of p21 Waf1/Cip1, p27 Kip1, and p53, suggesting that ALS induces G2/M arrest through modulation of p53/p21/CDC2/cyclin B1 pathways. ALS induced mitochondria-mediated apoptosis in MCF7 and MDA-MB-231 cells; ALS significantly decreased the expression of B-cell lymphoma 2 (Bcl-2), but increased the expression of B-cell lymphoma 2-associated X protein (Bax) and p53-upregulated modulator of apoptosis (PUMA), and increased the expression of cleaved caspases 3 and 9. ALS significantly increased the expression level of membrane-bound microtubule-associated protein 1 light chain 3 (LC3)-II and beclin 1 and induced inhibition of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and p38 mitogen-activated protein kinase (MAPK) pathways in MCF7 and MDA-MB-231 cells as indicated by their altered phosphorylation, contributing to the pro-autophagic activities of ALS. Furthermore, treatment with wortmannin markedly downregulated ALS-induced p38 MAPK activation and LC3 conversion. In addition, knockdown of the p38 MAPK gene by ribonucleic acid interference upregulated Akt activation and resulted in LC3-II accumulation. These findings indicate that ALS promotes cellular apoptosis and autophagy in breast cancer cells via modulation of p38 MAPK/Akt/mTOR pathways. Further studies are warranted to further explore the molecular targets of ALS in the treatment of breast cancer.
Collapse
Affiliation(s)
- Jin-Ping Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China ; Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Qi-Lun Liu
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Shu-Ting Pan
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China ; Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Xueji Zhang
- Research Center for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, People's Republic of China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Salt Lake Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Xiao-Wu Chen
- Department of General Surgery, The First People's Hospital of Shunde, Southern Medical University, Shunde, Foshan, Guangdong, People's Republic of China
| | - Dong Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA ; Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
1372
|
Zhou L, Huang A, Zhang D, Yao J, Zhang Y, Li X. Genetic variability of glutathione S-transferases influences treatment outcome of breast cancer. Tumour Biol 2015; 36:5925-9. [DOI: 10.1007/s13277-015-3266-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/17/2015] [Indexed: 11/28/2022] Open
|
1373
|
Liu L, Song Y, Gao S, Ji T, Zhang H, Ji B, Chen B, Jia B, Wang F, Xu Z, Ma Q. (99)mTc-3PRGD2 scintimammography in palpable and nonpalpable breast lesions. Mol Imaging 2015; 13. [PMID: 24825112 DOI: 10.2310/7290.2014.00010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The aim of this study was to explore the diagnostic performance of 99mTc-3(poly-(ethylene glycol),PEG)4-RGD2 (99mTc-3PRGD2) scintimammography (SMM) in patients with either palpable or nonpalpable breast lesions and compare SMM to mammography to assess the possible incremental value of SMM in breast cancer detection. We also investigated the αvβ3 expression in malignant and benign breast lesions. Ninety-four patients with 110 lesions were included in this study. Mammograms were evaluated according to the Breast Imaging Reporting and Data System (BI-RADS) by a specialized imaging radiologist. Prone SMM was performed 1 hour after injection of 99mTc-3PRGD2. Scintigraphic images were interpreted independently by two experienced nuclear medicine physicians using a three-point system, and the kappa value was calculated to determine the interreader agreement. The McNemar test was used to compare SMM and mammography with respect to sensitivity, specificity, and accuracy. Diagnostic values for breast cancer detection were evaluated for each lesion. Immunohistochemistry was performed to evaluate integrin αvβ3 expression. Histopathology revealed 46 malignant lesions and 64 benign lesions. The overall sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of SMM were 83%, 73%, 77%, 69%, and 85%, respectively. The kappa value between the two reviewers was 0.63. The diagnostic values of SMM were higher than those of mammography in evaluating overall breast lesions. A sensitivity of 91% was achieved when SMM and mammography results were combined with 60% of all false-negative mammography findings classified as true-positive results by SMM. Integrin αvβ3 expression was positively identified using SMM imaging. SMM is a promising tool to avoid unnecessary biopsies when used in addition to mammography and can be used to image αvβ3 expression in breast cancer with good image quality.
Collapse
|
1374
|
Wu M, Austin H, Eheman CR, Myles Z, Miller J, Royalty J, Ryerson AB. A comparative analysis of breast cancer stage between women enrolled in the National Breast and Cervical Cancer Early Detection Program and women not participating in the program. Cancer Causes Control 2015; 26:751-8. [PMID: 25761406 DOI: 10.1007/s10552-015-0548-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 02/28/2015] [Indexed: 11/30/2022]
Abstract
PURPOSE To determine the proportional distribution of early- and late-stage breast cancers diagnosed in years 2004-2009 among women enrolled in the National Breast and Cervical Cancer Early Detection Program (NBCCEDP) and to compare this distribution to that of geographically comparable non-enrolled women diagnosed with breast cancer. METHODS Using data from the National Program of Cancer Registries, we compared the demographic characteristics and cancer stage distribution of women enrollees and non-enrollees by use of conditional logistic regression using the odds ratio as a measure of association. RESULTS NBCCEDP enrollees were slightly younger and more likely to identify as African-American, API and AIAN than were non-enrollees. The proportion of late-stage breast cancer (regional and distant) decreased slightly over the study period. NBCCEDP enrollees generally were diagnosed at a later stage of breast cancer than were those not enrolled in the NBCCEDP. CONCLUSIONS The NBCCEDP has been effective in achieving its goal of enrolling racial and ethnic populations; however, enrollees had a poorer stage distribution of breast cancer than did non-enrollees underscoring the need to expand breast cancer control efforts among low-income, underserved populations.
Collapse
Affiliation(s)
- Manxia Wu
- Centers for Disease Control and Prevention, Atlanta, GA, USA,
| | | | | | | | | | | | | |
Collapse
|
1375
|
Seo HS, Woo JK, Shin YC, Ko SG. Identification of biomarkers regulated by rexinoids (LGD1069, LG100268 and Ro25-7386) in human breast cells using Affymetrix microarray. Mol Med Rep 2015; 12:800-18. [PMID: 25778982 PMCID: PMC4438952 DOI: 10.3892/mmr.2015.3480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 02/26/2015] [Indexed: 11/05/2022] Open
Abstract
Retinoids possess anti-proliferative properties, which suggests that they possess chemopreventive and therapeutic potential against cancer. In the current study, genes modulated by rexinoids (retinoid X receptor (RXR)-pan agonists, LGD1069 and LG100268; and the RXRα agonist, Ro25-7386) were identified using an Affymetrix microarray in normal and malignant breast cells. It was observed that LGD1069, LG100268 and Ro25-7386 suppressed the growth of breast cells. Secondly, several rexinoid-regulated genes were identified, which are involved in cell death, cell growth/maintenance, signal transduction and response to stimulus. These genes may be associated with the growth-suppressive activity of rexinoids. Therefore, the identified genes may serve as biomarkers and novel molecular targets for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Hye-Sook Seo
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, College of Korean Medicine, Kyung Hee University, Dongdaemun‑gu, Seoul 130‑701, Republic of Korea
| | - Jong-Kyu Woo
- Laboratory of Preventive Pharmacy, College of Pharmacy, Gachon University of Medicine and Science, Yeonsu‑gu, Incheon 406‑840, Republic of Korea
| | - Yong Cheol Shin
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, College of Korean Medicine, Kyung Hee University, Dongdaemun‑gu, Seoul 130‑701, Republic of Korea
| | - Seong-Gyu Ko
- Laboratory of Clinical Biology and Pharmacogenomics and Center for Clinical Research and Genomics, College of Korean Medicine, Kyung Hee University, Dongdaemun‑gu, Seoul 130‑701, Republic of Korea
| |
Collapse
|
1376
|
Xuan Q, Ji H, Tao X, Xu Y, Zhang Q. Quantitative assessment of HER2 amplification in HER2-positive breast cancer: its association with clinical outcomes. Breast Cancer Res Treat 2015; 150:581-8. [PMID: 25762478 DOI: 10.1007/s10549-015-3334-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is an effective therapeutic target in breast cancer. However, not all patients benefit from trastuzumab-based therapy. We aimed to investigate whether patients with different levels of HER2 amplification would experience different clinical outcomes with trastuzumab-based chemotherapy. We quantified the HER2 gene copy number (GCN) and HER2/centromere chromosome probe 17 (CEP17) ratio in 291 breast cancer patients with HER2 amplification confirmed by immunohistochemistry and fluorescence in situ hybridization. The optimal cutoff points for HER2 GCN and the HER2/CEP17 ratios for distinguishing positive results were determined by receiver operating characteristic (ROC) curve analyses. ROC analysis identified optimal cutoff points for HER2 GCN and HER2/CEP17 ratios as 11.5 and 6.5 (P = 0.039 and P = 0.012), respectively. The DFS in patients with HER2 GCN <11.5 was significantly longer than in HER2 GCN ≥11.5 patients (P = 0.015) according to Kaplan-Meier survival curves analysis. Similarly, patients with HER2/CEP17 ratios <6.5 had a significantly longer DFS than those with HER2/CEP17 ratios ≥6.5 (P = 0.013). Moreover, patients with HER2 cluster amplification showed a worse survival than those with HER2 non-cluster amplification (P = 0.041). This study demonstrated a significant association between the level of HER2 amplification and survival time in a relatively large cohort of HER2-positive breast cancer patients undergoing trastuzumab-based chemotherapy. Further investigations of more precise quantitative measurements and larger cohorts are required to define this threshold.
Collapse
Affiliation(s)
- Qijia Xuan
- Department of Internal Medicine, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, 150081, Heilongjiang, China
| | | | | | | | | |
Collapse
|
1377
|
Dormancy and growth of metastatic breast cancer cells in a bone-like microenvironment. Clin Exp Metastasis 2015; 32:335-44. [PMID: 25749879 DOI: 10.1007/s10585-015-9710-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/28/2015] [Indexed: 12/19/2022]
Abstract
Breast cancer can reoccur, often as bone metastasis, many years if not decades after the primary tumor has been treated. The factors that stimulate dormant metastases to grow are not known, but bone metastases are often associated with skeletal trauma. We used a dormancy model of MDA-MB-231BRMS1, a metastasis-suppressed human breast cancer cell line, co-cultured with MC3T3-E1 osteoblasts in a long term, three dimensional culture system to test the hypothesis that bone remodeling cytokines could stimulate dormant cells to grow. The cancer cells attached to the matrix produced by MC3T3-E1 osteoblasts but grew slowly or not at all until the addition of bone remodeling cytokines, TNFα and IL-β. Stimulation of cell proliferation by these cytokines was suppressed with indomethacin, an inhibitor of cyclooxygenase and of prostaglandin production, or a prostaglandin E2 (PGE2) receptor antagonist. Addition of PGE2 directly to the cultures also stimulated cell proliferation. MCF-7, non-metastatic breast cancer cells, remained dormant when co-cultured with normal human osteoblast and fibroblast growth factor. Similar to the MDA-MB-231BRMS1 cells, MCF-7 proliferation increased in response to TNFα and IL-β. These findings suggest that changes in the bone microenvironment due to inflammatory cytokines associated with bone repair or excess turnover may trigger the occurrence of latent bone metastasis.
Collapse
|
1378
|
Yang Y, Deng Q, Feng X, Sun J. Use of the disulfiram/copper complex for breast cancer chemoprevention in MMTV-erbB2 transgenic mice. Mol Med Rep 2015; 12:746-52. [PMID: 25738885 DOI: 10.3892/mmr.2015.3426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
The disulfiram/copper complex (DS/Cu) has been demonstrated to exert potent anti-tumor effects in various types of cancer. At present, whether DS/Cu has chemopreventive effects on breast cancer development remains to be fully elucidated. In the present study, using MMTV-erbB2 transgenic mice, it was identified for the first time that DS/Cu treatment was able to inhibit cell growth in breast cancer cells while sparing normal cells in vitro, in addition to delaying the development of mammary tumor development in MMTV-erbB2 transgenic mice in vivo. Morphological examination demonstrated that DS/Cu treatment resulted in cell proliferation inhibition and apoptosis activation in vitro and in vivo. Furthermore, the present study observed that DS/Cu may inhibit proliferation via inhibition of AKT and cyclin D1 signaling and promote apoptosis via c-Jun N-terminal kinase activation and suppression of nuclear factor κB signaling. These results suggested that DS/Cu treatment may be a promising therapy for the prevention of erbB2-positive breast cancer.
Collapse
Affiliation(s)
- Yanhui Yang
- Department of General Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Qian Deng
- Department of Pharmacy, The Central Hospital of Luoyang, Luoyang, Henan 471000, P.R. China
| | - Xiaoshan Feng
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Junjun Sun
- Department of General Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
1379
|
Feng T, Xu D, Tu C, Li W, Ning Y, Ding J, Wang S, Yuan L, Xu N, Qian K, Wang Y, Qi C. MiR-124 inhibits cell proliferation in breast cancer through downregulation of CDK4. Tumour Biol 2015; 36:5987-97. [PMID: 25731732 DOI: 10.1007/s13277-015-3275-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/17/2015] [Indexed: 12/19/2022] Open
Abstract
Studies have shown that microRNAs (miRNAs) are involved in the malignant progression of human cancer. However, little is known about the potential role of miRNAs in breast carcinogenesis. miR-124 expression in breast cancer tissue was measured by quantitative real-time PCR (qRT-PCR). Target prediction algorithms and luciferase reporter gene assays were used to investigate the target of miR-124. Breast cancer cells growth was regulated by overexpression or knockdown miR-124. At the end of the study, tumor-bearing mice were tested to confirm the function of miR-124 in breast cancer. In this study, we demonstrated that the expression of miR-124 was significantly downregulated in breast cancer tissues compared with matched adjacent non-neoplastic tissues. We identified and confirmed that cyclin-dependent kinase 4 (CDK4) was a direct target of miR-124. Overexpression of miR-124 suppressed CDK4 protein expression and attenuated cell viability, proliferation, and cell cycle progression in MCF-7 and MDA-MB-435S breast cancer cells in vitro. Overexpression of CDK4 partially rescued the inhibitory effect of miR-124 in the breast cancer cells. Moreover, we found that miR-124 overexpression effectively repressed tumor growth in xenograft animal experiments. Our results demonstrate that miR-124 functions as a growth-suppressive miRNA and plays an important role in inhibiting tumorigenesis by targeting CDK4.
Collapse
Affiliation(s)
- Tongbao Feng
- Oncology Institute, The Affiliated Hospital of Nanjing Medical University, Changzhou No.2 People's Hospital, Changzhou, 213003, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1380
|
Improvement in Overall Survival from Extremity Soft Tissue Sarcoma over Twenty Years. Sarcoma 2015; 2015:279601. [PMID: 25821397 PMCID: PMC4363656 DOI: 10.1155/2015/279601] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/06/2015] [Accepted: 02/17/2015] [Indexed: 12/31/2022] Open
Abstract
Several patient demographic factors, including marital status, have been demonstrated to have prognostic significance for survival in extremity soft tissue sarcoma (ESTS). A study population of 12,546 adult patients diagnosed with ESTS from 1991 to 2010 was identified from the SEER database, a large population-based registry, in order to determine whether overall survival had changed over this recent 20-year period. The study population was divided into three groups by year of diagnosis: 1991–1996, 1997–2003, and 2004–2010. We used the Kaplan-Meier method and Cox proportional hazards regression to assess survival differences between different demographic groups and prognostic clinical characteristics. Over the course of time, the 5-year overall survival rates have increased from 28% in the earliest time period to 62% in the latest (P < 0.0001). On multivariate analysis, the mortality rate progressively declined from the 1991–1996 group (HR: 3.02, CI: 2.78–3.29) to the 1997–2003 group (HR: 2.21, CI: 2.06–2.37), with the 2004–2010 group having the best overall survival, despite increases in the proportion of patients with tumors greater than 5 cm in size (P < 0.0001), and those presenting with metastasis (P < 0.0001).
Collapse
|
1381
|
Sridharan A, Eisenbrey JR, Machado P, Ojeda-Fournier H, Wilkes A, Sevrukov A, Mattrey RF, Wallace K, Chalek CL, Thomenius KE, Forsberg F. Quantitative analysis of vascular heterogeneity in breast lesions using contrast-enhanced 3-D harmonic and subharmonic ultrasound imaging. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2015; 62:502-10. [PMID: 25935933 PMCID: PMC4607037 DOI: 10.1109/tuffc.2014.006886] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Ability to visualize breast lesion vascularity and quantify the vascular heterogeneity using contrast-enhanced 3-D harmonic (HI) and subharmonic (SHI) ultrasound imaging was investigated in a clinical population. Patients (n = 134) identified with breast lesions on mammography were scanned using power Doppler imaging, contrast-enhanced 3-D HI, and 3-D SHI on a modified Logiq 9 scanner (GE Healthcare). A region of interest corresponding to ultrasound contrast agent flow was identified in 4D View (GE Medical Systems) and mapped to raw slice data to generate a map of time-intensity curves for the lesion volume. Time points corresponding to baseline, peak intensity, and washout of ultrasound contrast agent were identified and used to generate and compare vascular heterogeneity plots for malignant and benign lesions. Vascularity was observed with power Doppler imaging in 84 lesions (63 benign and 21 malignant). The 3-D HI showed flow in 8 lesions (5 benign and 3 malignant), whereas 3-D SHI visualized flow in 68 lesions (49 benign and 19 malignant). Analysis of vascular heterogeneity in the 3-D SHI volumes found benign lesions having a significant difference in vascularity between central and peripheral sections (1.71 ± 0.96 vs. 1.13 ± 0.79 dB, p < 0.001, respectively), whereas malignant lesions showed no difference (1.66 ± 1.39 vs. 1.24 ± 1.14 dB, p = 0.24), indicative of more vascular coverage. These preliminary results suggest quantitative evaluation of vascular heterogeneity in breast lesions using contrast-enhanced 3-D SHI is feasible and able to detect variations in vascularity between central and peripheral sections for benign and malignant lesions.
Collapse
Affiliation(s)
- Anush Sridharan
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA. Department of Electrical and Computer Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - John R. Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Priscilla Machado
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Annina Wilkes
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexander Sevrukov
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert F. Mattrey
- Department of Radiology, University of California, San Diego, CA 92103, USA
| | | | | | | | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
1382
|
Yuri T, Tsubura A. Relation between parity and pregnancy-related hormones and breast cancer control. BREAST CANCER MANAGEMENT 2015. [DOI: 10.2217/bmt.14.52] [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 Epidemiological research has indicated the beneficial effects of full-term pregnancy at an early age for a reduction in breast cancer risk. Experimental data have shown that pregnancy and pregnancy-related hormones, such as estrogen plus progesterone, estrogen alone and human chorionic gonadotropin, are involved in parity-induced protection. Pregnancy and short-duration treatment of a young host with pregnancy-related hormones to mimic the pregnancy environment provide mammary cancer protection by making cells refractory to carcinogenic stimuli and causing growth arrest and programmed cell death. Experimental data concerning pregnancy and pregnancy-related hormones are reviewed in relation to intrinsic subtypes of mammary cancer.
Collapse
Affiliation(s)
- Takashi Yuri
- Department of Pathology II, Kansai Medical University, Hirakata, Osaka, Japan
| | - Airo Tsubura
- Department of Pathology II, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
1383
|
Rapoport BL. Sorafenib: a brief review with emphasis on its possible role in breast cancer. BREAST CANCER MANAGEMENT 2015. [DOI: 10.2217/bmt.15.1] [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 The treatment of metastatic breast cancer is palliative and usually requires hormone treatment, and/or chemotherapy and trastuzumab for HER2-positive disease. Randomized studies only demonstrate marginal survival benefit with combination chemotherapy, as opposed to sequential single agents or nontaxane versus taxane-containing regimens. Metastatic breast cancer remains an unmet medical need. Sorafenib is an oral multikinase inhibitor that exhibits antiangiogenic activity by targeting numerous proangiogenic pathways. Sorafenib has marketing authorization for the treatment of patients with advanced renal cell, hepatocellular and thyroid carcinomas. Recent studies have explored the usage of sorafenib as a single agent and in combination with chemotherapy. An overview of sorafenib is presented in this article with the current data and potential future usage in patients with metastatic breast cancer.
Collapse
|
1384
|
Smoking and survival in female breast cancer patients. Breast Cancer Res Treat 2015; 150:395-403. [PMID: 25724306 DOI: 10.1007/s10549-015-3317-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 02/19/2015] [Indexed: 10/23/2022]
Abstract
The purpose of this study was to determine if smoking affects survival in female breast cancer patients, both overall and stratified by race, ethnicity, and socioeconomic status. We linked data from the 1996-2007 Florida cancer data system, the Florida Agency for Health Care Administration, and the U.S. census. Inclusion criteria were females ≥18 years, diagnosed with breast cancer, and residing in Florida (n = 127,754). To analyze the association between smoking and survival, we performed sequential multivariate Cox proportional hazard regression models with progressive adjustment for main confounders. Compared to never smokers, worse survival was found in current (hazard ratio 1.33; 95 % CI 1.28-1.38) and former smokers (1.09; 1.06-1.13). Those who smoked <1, 1-2, and >2 packs/day had worse survival (HR 1.28; 1.20-1.36; HR 1.40; 1.33-1.47 and 1.70; 1.45-1.99, respectively) (p for linear trend <0.001), than never smokers. Among Whites, current and former smokers had worse survival (HR 1.38; 1.33-1.44 and HR 1.11; 1.07-1.15, respectively) than never smokers. Worse survival was also found for current and former smokers (HR 1.34; 1.29-1.40 and HR 1.10; 1.06-1.15, respectively) compared with never smokers among non-Hispanics; similarly, worse survival was found among current Hispanic smokers (HR 1.13; 1.01-1.26). The association was not significant for Blacks. Current smoking is associated with worse survival in White breast cancer patients and through all socioeconomic status categories and ethnicities compared to never smoking. Former smoking is associated with worse survival in White and non-Hispanic females. Blacks had similar survival regardless of smoking status. Nonetheless, all female breast cancer patients should be advised to quit smoking.
Collapse
|
1385
|
Tordonato C, Di Fiore PP, Nicassio F. The role of non-coding RNAs in the regulation of stem cells and progenitors in the normal mammary gland and in breast tumors. Front Genet 2015; 6:72. [PMID: 25774169 PMCID: PMC4343025 DOI: 10.3389/fgene.2015.00072] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/11/2015] [Indexed: 12/17/2022] Open
Abstract
The outlook on stem cell (SC) biology is shifting from a rigid hierarchical to a more flexible model in which the identity and the behavior of adult SCs, far from being fixed, are determined by the dynamic integration of cell autonomous and non-autonomous mechanisms. Within this framework, the recent discovery of thousands of non-coding RNAs (ncRNAs) with regulatory function is redefining the landscape of transcriptome regulation, highlighting the interplay of epigenetic, transcriptional, and post-transcriptional mechanisms in the specification of cell fate and in the regulation of developmental processes. Furthermore, the expression of ncRNAs is often tissue- or even cell type-specific, emphasizing their involvement in defining space, time and developmental stages in gene regulation. Such a role of ncRNAs has been investigated in embryonic and induced pluripotent SCs, and in numerous types of adult SCs and progenitors, including those of the breast, which will be the topic of this review. We will focus on ncRNAs with an important role in breast cancer, in particular in mammary cancer SCs and progenitors, and highlight the ncRNA-based circuitries whose subversion alters a number of the epigenetic, transcriptional, and post-transcriptional events that control “stemness” in the physiological setting.
Collapse
Affiliation(s)
- Chiara Tordonato
- Department of Experimental Oncology, European Institute of Oncology, Milan Italy
| | - Pier Paolo Di Fiore
- Department of Experimental Oncology, European Institute of Oncology, Milan Italy ; Fondazione Istituto FIRC di Oncologia Molecolare, Milan Italy ; Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan Italy
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan Italy
| |
Collapse
|
1386
|
Fernandes AS, Flórido A, Saraiva N, Cerqueira S, Ramalhete S, Cipriano M, Cabral MF, Miranda JP, Castro M, Costa J, Oliveira NG. Role of the Copper(II) Complex Cu[15]pyN5 in Intracellular ROS and Breast Cancer Cell Motility and Invasion. Chem Biol Drug Des 2015; 86:578-88. [PMID: 25600158 DOI: 10.1111/cbdd.12521] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/02/2014] [Accepted: 01/08/2015] [Indexed: 12/29/2022]
Abstract
Multiple mechanisms related to metastases undergo redox regulation. Cu[15]pyN5 is a redox-active copper(II) complex previously studied as a chemotherapy sensitizer in mammary cells. The effects of a cotreatment with Cu[15]pyN5 and doxorubicin (dox) were evaluated in two human breast cancer cell lines: MCF7 (low aggressiveness) and MDA-MB-231 (highly aggressive). Cu[15]pyN5 decreased MCF7-directed cell migration. In addition, a cotreatment with dox and Cu[15]pyN5 reduced the proteolytic invasion of MDA-MB-231 cells. Cell detachment was not affected by exposure to these agents. Cu[15]pyN5 and dox significantly increased intracellular ROS in both cell lines. This increase could be at least partially due to H2 O2 accumulation. The combination of Cu[15]pyN5 with dox may be beneficial in breast cancer treatment as it could help reduce cancer cell migration and invasion. Moreover, the ligand [15]pyN5 has a high affinity for copper(II) and displays potential anti-angiogenic properties. Overall, we present a potential drug that might arrest the progression of breast cancer by different and complementary mechanisms.
Collapse
Affiliation(s)
- Ana S Fernandes
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
| | - Ana Flórido
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal.,Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
| | - Sara Cerqueira
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa, 1749-024, Portugal
| | - Sérgio Ramalhete
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Madalena Cipriano
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Maria Fátima Cabral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Joana P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Judite Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
1387
|
Quantification of the binding potential of cell-surface receptors in fresh excised specimens via dual-probe modeling of SERS nanoparticles. Sci Rep 2015; 5:8582. [PMID: 25716578 PMCID: PMC4341215 DOI: 10.1038/srep08582] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/26/2015] [Indexed: 12/12/2022] Open
Abstract
The complete removal of cancerous tissue is a central aim of surgical oncology, but is difficult to achieve in certain cases, especially when the removal of surrounding normal tissues must be minimized. Therefore, when post-operative pathology identifies residual tumor at the surgical margins, re-excision surgeries are often necessary. An intraoperative approach for tumor-margin assessment, insensitive to nonspecific sources of molecular probe accumulation and contrast, is presented employing kinetic-modeling analysis of dual-probe staining using surface-enhanced Raman scattering nanoparticles (SERS NPs). Human glioma (U251) and epidermoid (A431) tumors were implanted subcutaneously in six athymic mice. Fresh resected tissues were stained with an equimolar mixture of epidermal growth factor receptor (EGFR)-targeted and untargeted SERS NPs. The binding potential (BP; proportional to receptor concentration) of EGFR – a cell-surface receptor associated with cancer – was estimated from kinetic modeling of targeted and untargeted NP concentrations in response to serial rinsing. EGFR BPs in healthy, U251, and A431 tissues were 0.06 ± 0.14, 1.13 ± 0.40, and 2.23 ± 0.86, respectively, which agree with flow-cytometry measurements and published reports. The ability of this approach to quantify the BP of cell-surface biomarkers in fresh tissues opens up an accurate new approach to analyze tumor margins intraoperatively.
Collapse
|
1388
|
Azevedo C, Correia-Branco A, Araújo JR, Guimarães JT, Keating E, Martel F. The chemopreventive effect of the dietary compound kaempferol on the MCF-7 human breast cancer cell line is dependent on inhibition of glucose cellular uptake. Nutr Cancer 2015; 67:504-13. [PMID: 25719685 DOI: 10.1080/01635581.2015.1002625] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Our aim was to investigate the effect of several dietary polyphenols on glucose uptake by breast cancer cells. Uptake of (3)H-deoxy-D-glucose ((3)H-DG) by MCF-7 cells was time-dependent, saturable, and inhibited by cytochalasin B plus phloridzin. In the short-term (26 min), myricetin, chrysin, genistein, resveratrol, kaempferol, and xanthohumol (10-100 µM) inhibited (3)H-DG uptake. Kaempferol was found to be the most potent inhibitor of (3)H-DG uptake [IC50 of 4 µM (1.6-9.8)], behaving as a mixed-type inhibitor. In the long-term (24 h), kaempferol (30 µM) was also able to inhibit (3)H-DG uptake, associated with a 40% decrease in GLUT1 mRNA levels. Interestingly enough, kaempferol (100 µM) revealed antiproliferative (sulforhodamine B and (3)H-thymidine incorporation assays) and cytotoxic (extracellular lactate dehydrogenase activity determination) properties, which were mimicked by low extracellular (1 mM) glucose conditions and reversed by high extracellular (20 mM) glucose conditions. Finally, exposure of cells to kaempferol (30 µM) induced an increase in extracellular lactate levels over time (to 731 ± 32% of control after a 24 h exposure), due to inhibition of MCT1-mediated lactate cellular uptake. In conclusion, kaempferol potently inhibits glucose uptake by MCF-7 cells, apparently by decreasing GLUT1-mediated glucose uptake. The antiproliferative and cytotoxic effect of kaempferol in these cells appears to be dependent on this effect.
Collapse
Affiliation(s)
- Cláudia Azevedo
- a Department of Biochemistry , Faculty of Medicine , University of Porto , Porto , Portugal
| | | | | | | | | | | |
Collapse
|
1389
|
Xu C, Wei Q, Guo J, Zhou JC, Mei J, Jiang ZN, Shen JG, Wang LB. FOXA1 Expression Significantly Predict Response to Chemotherapy in Estrogen Receptor-Positive Breast Cancer Patients. Ann Surg Oncol 2015; 22:2034-9. [PMID: 25707489 DOI: 10.1245/s10434-014-4313-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Indexed: 01/09/2023]
Abstract
PURPOSE Most estrogen receptor (ER)-positive breast cancer responds poorly to chemotherapy and no single cost-effective biomarker capable of selecting chemosensitive ones has been found yet. We investigated FOXA1 for its role in predicting chemosensitivity of this subgroup in neoadjuvant chemotherapy settings. METHODS We reviewed pathologic slides of 123 patients who were diagnosed with ER-positive breast cancer on core needle biopsy and underwent neoadjuvant chemotherapy at our institution between 2002 and 2012. FOXA1 expression and pathologic response were evaluated. We then statistically analyzed FOXA1 expression and its relationship with chemosensitivity. RESULTS FOXA1 expression before NAC was correlated with poor chemoresponse in ER-positive as well as luminal A and luminal B breast cancer patients (p = 0.002, 0.001, and 0.049 respectively). Significant association between change of FOXA1 staining position after NAC and chemosensitivity also was observed (p = 0.024). Multivariate analysis identified FOXA1 expression before NAC as an independent predictor of chemosensitivity in ER-positive and luminal A breast cancer patients [p = 0.002; relative risk (RR) 0.163; 95 % confidence interval (CI) 0.053-0.500, and p = 0.002; RR 0.055; 95 % CI 0.008-0.353, respectively]. Additionally, change of FOXA1 staining position after NAC was shown to be an independent predictor of chemoresponse in luminal B subtype breast cancer patients (p = 0.012; RR 0.153; 95 % CI 0.035-0.665). CONCLUSIONS FOXA1 expression can independently predict chemosensitivity of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Chenpu Xu
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
1390
|
Bonilla C, Bertoni B, Hidalgo PC, Artagaveytia N, Ackermann E, Barreto I, Cancela P, Cappetta M, Egaña A, Figueiro G, Heinzen S, Hooker S, Román E, Sans M, Kittles RA. Breast cancer risk and genetic ancestry: a case-control study in Uruguay. BMC WOMENS HEALTH 2015; 15:11. [PMID: 25783644 PMCID: PMC4341228 DOI: 10.1186/s12905-015-0171-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 01/27/2015] [Indexed: 01/26/2023]
Abstract
Background Uruguay exhibits one of the highest rates of breast cancer in Latin America, similar to those of developed nations, the reasons for which are not completely understood. In this study we investigated the effect that ancestral background has on breast cancer susceptibility among Uruguayan women. Methods We carried out a case–control study of 328 (164 cases, 164 controls) women enrolled in public hospitals and private clinics across the country. We estimated ancestral proportions using a panel of nuclear and mitochondrial ancestry informative markers (AIMs) and tested their association with breast cancer risk. Results Nuclear individual ancestry in cases was (mean ± SD) 9.8 ± 7.6% African, 13.2 ± 10.2% Native American and 77.1 ± 13.1% European, and in controls 9.1 ± 7.5% African, 14.7 ± 11.2% Native American and 76.2 ± 14.2% European. There was no evidence of a difference in nuclear or mitochondrial ancestry between cases and controls. However, European mitochondrial haplogroup H was associated with breast cancer (OR = 2.0; 95% CI 1.1, 3.5). Conclusions We have not found evidence that overall genetic ancestry differs between breast cancer patients and controls in Uruguay but we detected an association of the disease with a European mitochondrial lineage, which warrants further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s12905-015-0171-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carolina Bonilla
- School of Social and Community Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1391
|
Reeder-Hayes KE, Wheeler SB, Mayer DK. Health disparities across the breast cancer continuum. Semin Oncol Nurs 2015; 31:170-7. [PMID: 25951746 DOI: 10.1016/j.soncn.2015.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES To provide a brief overview of disparities across the spectrum of breast cancer incidence, treatment, and long-term care during the survivorship period. DATA SOURCES Review of the literature including research reports, review articles, and clinically based articles available through PubMed and CINAHL. CONCLUSION Minority women generally experience worse breast cancer outcomes despite a lower incidence of breast cancer than whites. A variety of factors contribute to this disparity, including advanced stage at diagnosis, higher rates of aggressive breast cancer subtypes, and lower receipt of appropriate therapies including surgery, chemotherapy, and radiation. Disparities in breast cancer care also extend into the survivorship trajectory, including lower rates of endocrine therapy use among some minority groups, as well as differences in follow-up and survivorship care. IMPLICATIONS FOR NURSING PRACTICE Breast cancer research should include improved minority representation and analyses by race, ethnicity, and socioeconomic status. While we cannot yet change the biology of this disease, we can encourage adherence to screening and treatment and help address the many physical, psychological, spiritual, and social issues minority women face in a culturally sensitive manner.
Collapse
|
1392
|
Lou C, Yokoyama S, Saiki I, Hayakawa Y. Selective anticancer activity of hirsutine against HER2‑positive breast cancer cells by inducing DNA damage. Oncol Rep 2015; 33:2072-6. [PMID: 25672479 DOI: 10.3892/or.2015.3796] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/05/2015] [Indexed: 11/05/2022] Open
Abstract
Hirsutine is one of the major alkaloids isolated from plants of the Uncaria genus and is known for its cardioprotective, anti‑hypertensive and anti-arrhythmic activities. We recently reported that hirsutine is an anti-metastatic phytochemical by targeting NF-κB activation in a murine breast cancer model. In the present study, we further examined the clinical utility of hirsutine against human breast cancer. Among six distinct human breast cancer cell lines, hirsutine showed strong cytotoxicity against HER2-positive/p53-mutated MDA-MB‑453 and BT474 cell lines. Conversely, HER2-negative/p53 wild‑type MCF-7 and ZR-75-1 cell lines showed resistance against hirsutine-induced cytotoxicity. Hirsutine induced apoptotic cell death in the MDA-MB-453 cells, but not in the MCF-7 cells, through activation of caspases. Furthermore, hirsutine induced the DNA damage response in the MDA-MB-453 cells, but not in the MCF-7 cells, as highlighted by the upregulation of γH2AX expression. Along with the induction of the DNA damage response, the suppression of HER2, NF-κB and Akt pathways and the activation of the p38 MAPK pathway in the MDA-MB-453 cells were observed. Considering that there was no difference between MDA-MB-453 and MCF-7 cells in regards to irinotecan‑induced DNA damage response, our present results indicate the selective anticancer activity of hirsutine in HER2-positive breast cancer by inducing a DNA damage response.
Collapse
Affiliation(s)
- Chenghua Lou
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Satoru Yokoyama
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Ikuo Saiki
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Toyama 930-0194, Japan
| |
Collapse
|
1393
|
Chang CF, Diers AR, Hogg N. Cancer cell metabolism and the modulating effects of nitric oxide. Free Radic Biol Med 2015; 79:324-36. [PMID: 25464273 PMCID: PMC5275750 DOI: 10.1016/j.freeradbiomed.2014.11.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/31/2014] [Accepted: 11/08/2014] [Indexed: 12/18/2022]
Abstract
Altered metabolic phenotype has been recognized as a hallmark of tumor cells for many years, but this aspect of the cancer phenotype has come into greater focus in recent years. NOS2 (inducible nitric oxide synthase of iNOS) has been implicated as a component in many aggressive tumor phenotypes, including melanoma, glioblastoma, and breast cancer. Nitric oxide has been well established as a modulator of cellular bioenergetics pathways, in many ways similar to the alteration of cellular metabolism observed in aggressive tumors. In this review we attempt to bring these concepts together with the general hypothesis that one function of NOS2 and NO in cancer is to modulate metabolic processes to facilitate increased tumor aggression. There are many mechanisms by which NO can modulate tumor metabolism, including direct inhibition of respiration, alterations in mitochondrial mass, oxidative inhibition of bioenergetic enzymes, and the stimulation of secondary signaling pathways. Here we review metabolic alterations in the context of cancer cells and discuss the role of NO as a potential mediator of these changes.
Collapse
Affiliation(s)
- Ching-Fang Chang
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Anne R Diers
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Neil Hogg
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
1394
|
Autophagy inhibition re-sensitizes pulse stimulation-selected paclitaxel-resistant triple negative breast cancer cells to chemotherapy-induced apoptosis. Breast Cancer Res Treat 2015; 149:619-29. [PMID: 25638397 DOI: 10.1007/s10549-015-3283-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/20/2015] [Indexed: 01/07/2023]
Abstract
Chemotherapy is the mainstay of systemic treatment for triple negative breast cancer (TNBC); however, the development of drug resistance limits its effectiveness. Therefore, we investigated the underlying mechanism for drug resistance and potential approaches to overcome it for a more effective treatment for TNBCs. Using a pulse-stimulated selection strategy to mimic chemotherapy administration in the clinic, we developed a new paclitaxel-resistant MDA-MB-231 cell line and analyzed these cells for changes in autophagy activity, and the role and mechanisms of the increased autophagy in promoting drug resistance were determined. We found that the pulse-stimulated selection strategy with paclitaxel resulted in MDA-MB-231 variant cells with enhanced resistance to paclitaxel. These resistant cells were found to have enhanced basal autophagy activity, which confers a cytoprotective function under paclitaxel treatment stress. Inhibition of autophagy enhanced paclitaxel-induced cell death in these paclitaxel-resistant cells. We further revealed that up-regulated autophagy in resistant cells enhanced the clearance of damaged mitochondria. Last, we showed that the paclitaxel-resistant cancer cells acquired cross resistance to epirubicin and cisplatin. Together, these results suggest that combining autophagy inhibition with chemotherapy may be an effective strategy to improve treatment outcome in paclitaxel-resistant TNBC patients.
Collapse
|
1395
|
Seksenyan A, Kadavallore A, Walts AE, de la Torre B, Berel D, Strom SP, Aliahmad P, Funari VA, Kaye J. TOX3 is expressed in mammary ER(+) epithelial cells and regulates ER target genes in luminal breast cancer. BMC Cancer 2015; 15:22. [PMID: 25632947 PMCID: PMC4324787 DOI: 10.1186/s12885-015-1018-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023] Open
Abstract
Background A breast cancer susceptibility locus has been mapped to the gene encoding TOX3. Little is known regarding the expression pattern or biological role of TOX3 in breast cancer or in the mammary gland. Here we analyzed TOX3 expression in murine and human mammary glands and in molecular subtypes of breast cancer, and assessed its ability to alter the biology of breast cancer cells. Methods We used a cell sorting strategy, followed by quantitative real-time PCR, to study TOX3 gene expression in the mouse mammary gland. To study the expression of this nuclear protein in human mammary glands and breast tumors, we generated a rabbit monoclonal antibody specific for human TOX3. In vitro studies were performed on MCF7, BT474 and MDA-MB-231 cell lines to study the effects of TOX3 modulation on gene expression in the context of breast cancer cells. Results We found TOX3 expression in estrogen receptor-positive mammary epithelial cells, including progenitor cells. A subset of breast tumors also highly expresses TOX3, with poor outcome associated with high expression of TOX3 in luminal B breast cancers. We also demonstrate the ability of TOX3 to alter gene expression in MCF7 luminal breast cancer cells, including cancer relevant genes TFF1 and CXCR4. Knockdown of TOX3 in a luminal B breast cancer cell line that highly expresses TOX3 is associated with slower growth. Surprisingly, TOX3 is also shown to regulate TFF1 in an estrogen-independent and tamoxifen-insensitive manner. Conclusions These results demonstrate that high expression of this protein likely plays a crucial role in breast cancer progression. This is in sharp contrast to previous studies that indicated breast cancer susceptibility is associated with lower expression of TOX3. Together, these results suggest two different roles for TOX3, one in the initiation of breast cancer, potentially related to expression of TOX3 in mammary epithelial cell progenitors, and another role for this nuclear protein in the progression of cancer. In addition, these results can begin to shed light on the reported association of TOX3 expression and breast cancer metastasis to the bone, and point to TOX3 as a novel regulator of estrogen receptor-mediated gene expression. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1018-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akop Seksenyan
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Asha Kadavallore
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Brian de la Torre
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Dror Berel
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Center for Applied Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA.
| | - Samuel P Strom
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Pathology and Laboratory Medicine, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Parinaz Aliahmad
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA.
| | - Vincent A Funari
- Genomics Core Facility, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Jonathan Kaye
- Research Division of Immunology, Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Davis 5089, Los Angeles, 90048, CA, USA. .,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
1396
|
Gomes LR, Vessoni AT, Menck CFM. Three-dimensional microenvironment confers enhanced sensitivity to doxorubicin by reducing p53-dependent induction of autophagy. Oncogene 2015; 34:5329-40. [PMID: 25619836 DOI: 10.1038/onc.2014.461] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 12/02/2014] [Accepted: 12/19/2014] [Indexed: 12/19/2022]
Abstract
Preclinical studies of anticancer drugs are typically performed using cancer cell lines maintained in two-dimensional (2D) cultures, ignoring the influences of the extracellular matrix (ECM) and three-dimensional (3D) microenvironment. In this study, we evaluated the microenvironmental control of human breast cancer cells responses to doxorubicin (DOXO) using the 3D laminin-rich ECM (3D lrECM) cell culture model. Under 3D culture conditions, MCF-7 cells displayed drastic morphological alterations, a decrease in proliferation and elevated sensitivity to DOXO. Interestingly, the chemotherapy-mediated activation of autophagy was compromised in the 3D matrix, suggesting an association between the increased cytotoxicity of DOXO and hindered autophagy induction. Indeed, while chloroquine or ATG5 knockdown potentiated DOXO-induced cell death under the 2D culture conditions, the autophagy inducer rapamycin improved the resistance of 3D-cultured cells to this drug. Moreover, in the monolayer-cultured cells, DOXO treatment led to increases in p53 and DRAM-1 expression, which is a p53-dependent activator of autophagy that functions in response to DNA damage. Conversely, p53 and DRAM-1 expression was impaired in 3D-cultured cells. The knockdown of p53 by shRNA blocked DRAM-1 activation, impaired autophagy induction and sensitized only those cells maintained under 2D conditions to DOXO. In addition, 2D-cultured MDA-MB-231 cells (a p53-mutated breast cancer cell line) not only showed increased sensitivity to DOXO compared with MCF-7 cells but also failed to induce DRAM-1 expression or autophagy. Similar to p53 silencing, DRAM-1 knockdown potentiated DOXO cytotoxicity only in 2D-cultured cells. These results suggest that the 3D tissue microenvironment controls tumor cell sensitivity to DOXO treatment by preventing p53-DRAM-autophagy axis activation.
Collapse
Affiliation(s)
- L R Gomes
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - A T Vessoni
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - C F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
1397
|
Caro-Morán E, Fernández-Lao C, Galiano-Castillo N, Cantarero-Villanueva I, Arroyo-Morales M, Díaz-Rodríguez L. Heart Rate Variability in Breast Cancer Survivors After the First Year of Treatments: A Case-Controlled Study. Biol Res Nurs 2015; 18:43-9. [PMID: 25616419 DOI: 10.1177/1099800414568100] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The same aggressive treatments that have led to a reduction in the breast cancer may also have adverse effects on cardiac autonomic balance. The objective of this study was to compare heart rate variability (HRV) between breast cancer survivors in the first year posttreatment and healthy women, controlling for known confounders. This descriptive case-controlled study included 22 breast cancer survivors and 22 healthy age- and sex-matched controls. Short-term HRV was measured using an accepted methodology to assess the cardiac autonomic balance. One-way analysis of covariance results revealed that heart rate was significantly higher (F = 15.86, p < .001) and the standard deviation of normal-to-normal (NN) interval (F = 19.93, p = .001), square root of mean squared differences of successive NN intervals (F = 18.72, p = .001), HRV index (F = 5.44, p = .025), and high-frequency (F = 5.77, p = .03) values were significantly lower in the breast cancer survivors than in the matched controls. The principal finding of the presence of a cardiovascular imbalance in breast cancer survivors in comparison to healthy age-matched controls suggests that HRV study could be a clinically useful tool to detect cardiovascular disease in early-stage breast cancer survivors.
Collapse
Affiliation(s)
- Elena Caro-Morán
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Carolina Fernández-Lao
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain Sport and Health University Research Institute, University of Granada, Granada, Spain
| | - Noelia Galiano-Castillo
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain Sport and Health University Research Institute, University of Granada, Granada, Spain
| | - Irene Cantarero-Villanueva
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain Sport and Health University Research Institute, University of Granada, Granada, Spain
| | - Manuel Arroyo-Morales
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain Sport and Health University Research Institute, University of Granada, Granada, Spain
| | - Lourdes Díaz-Rodríguez
- Instituto Biosanitario Granada, Faculty of Health Sciences, University of Granada, Granada, Spain Sport and Health University Research Institute, University of Granada, Granada, Spain Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| |
Collapse
|
1398
|
Spivey TL, Carlson KA, Janssen I, Witt TR, Jokich P, Madrigrano A. Breast Imaging Second Opinions Impact Surgical Management. Ann Surg Oncol 2015; 22:2359-64. [DOI: 10.1245/s10434-014-4205-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Indexed: 11/18/2022]
|
1399
|
Chang HY, Li MH, Huang TC, Hsu CL, Tsai SR, Lee SC, Huang HC, Juan HF. Quantitative proteomics reveals middle infrared radiation-interfered networks in breast cancer cells. J Proteome Res 2015; 14:1250-62. [PMID: 25556991 DOI: 10.1021/pr5011873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Breast cancer is one of the leading cancer-related causes of death worldwide. Treatment of triple-negative breast cancer (TNBC) is complex and challenging, especially when metastasis has developed. In this study, we applied infrared radiation as an alternative approach for the treatment of TNBC. We used middle infrared (MIR) with a wavelength range of 3-5 μm to irradiate breast cancer cells. MIR significantly inhibited cell proliferation in several breast cancer cells but did not affect the growth of normal breast epithelial cells. We performed iTRAQ-coupled LC-MS/MS analysis to investigate the MIR-triggered molecular mechanisms in breast cancer cells. A total of 1749 proteins were identified, quantified, and subjected to functional enrichment analysis. From the constructed functionally enriched network, we confirmed that MIR caused G2/M cell cycle arrest, remodeled the microtubule network to an astral pole arrangement, altered the actin filament formation and focal adhesion molecule localization, and reduced cell migration activity and invasion ability. Our results reveal the coordinative effects of MIR-regulated physiological responses in concentrated networks, demonstrating the potential implementation of infrared radiation in breast cancer therapy.
Collapse
Affiliation(s)
- Hsin-Yi Chang
- Department of Life Science, National Taiwan University , Taipei 10617, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
1400
|
Hong JY, Chung HJ, Bae SY, Trung TN, Bae K, Lee SK. Induction of Cell Cycle Arrest and Apoptosis by Physcion, an Anthraquinone Isolated From Rhubarb (Rhizomes of Rheum tanguticum), in MDA-MB-231 Human Breast Cancer Cells. J Cancer Prev 2015; 19:273-8. [PMID: 25574462 PMCID: PMC4285958 DOI: 10.15430/jcp.2014.19.4.273] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022] Open
Abstract
Background: Physcion is an anthraquinone from rhubarb (rhizomes of Rheum tanguticum) and has been reported to have anti-inflammatory, hepatoprotective, antifungal, and anti-cancer activities. However, the growth inhibitory activity against human cancer cells and the underlying molecular mechanisms have been poorly determined. This study was designed to investigate the anti-proliferative activity of physcion by induction of cell cycle arrest and apoptosis in human MDA-MB-231 triple negative breast cancer cell line. Methods: MDA-MB-231 cells were treated with physcion, and the anti-proliferative activity was evaluated by the sulforhodamine B assay. The mechanisms of action for the growth inhibitory activity of physcion were evaluated by flow cytometry for cell cycle distribution, and by Western blot for the assessment of potential target proteins. Results: Physcion showed a significant anti-proliferative activity against MDA-MB-231 human breast cancer cells. Flow cytometric analysis indicated that physcion markedly induced the accumulation of cells in the G0/G1 phase and the increase of cell population in the sub-G1 phase. The G0/G1 cell cycle arrest by physcion was associated with the down-regulation of Cyclin D1, Cyclin A, CDK4, CDK2, c-Myc and phosphorylated Rb protein expressions. The increase of sub-G1 peak by physcion was closely correlated with the induction of apoptosis, which was confirmed by the induction of cleaved poly-(adenosine diphosphate ribose) polymerase, activation of Caspases, and suppression of Bid and Bcl-2 expression. Conclusions: The induction of G0/G1 cell cycle arrest and apoptosis might be one of the plausible mechanisms of actions for the anti-proliferative activity of physcion in human breast cancer cells.
Collapse
Affiliation(s)
- Ji-Young Hong
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hwa-Jin Chung
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Song Yi Bae
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Trinh Nam Trung
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - KiHwan Bae
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|