1
|
Imidazopyridazine Acetylcholinesterase Inhibitors Display Potent Anti-Proliferative Effects in the Human Neuroblastoma Cell-Line, IMR-32. Molecules 2021; 26:molecules26175319. [PMID: 34500749 PMCID: PMC8434581 DOI: 10.3390/molecules26175319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 11/29/2022] Open
Abstract
Imidazo[1,2-b]pyridazine compounds are a new class of promising lead molecules to which we have incorporated polar nitro and amino moieties to increase the scope of their biological activity. Two of these substituted 3-nitro-6-amino-imidazo[1,2-b]pyridazine compounds (5c and 5h) showed potent acetylcholinesterase (AChE) inhibitory activity (IC50 40–50 nM), which we have previously reported. In this study, we wanted to test the biological efficacy of these compounds. Cytotoxicity assays showed that compound 5h mediated greater cell death with over 43% of cells dead at 100 μM and activation of caspase 3-mediated apoptosis. On the other hand, compound 5c mediated a dose-dependent decrease in cell proliferation. Both compounds showed cell cycle arrest in the G0/G1 phase and reduced cellular ATP levels leading to activation of adenosine monophosphate-activated protein kinase (AMPK) and enhanced mitochondrial oxidative stress. It has to be noted that all these effects were observed at doses beyond 10 μM, 200-fold above the IC50 for AChE inhibition. Both compounds also inhibited bacterial lipopolysaccharide-mediated cyclooxygenase-2 and nitric oxide release in primary rat microglial cells. These results suggested that the substituted imidazo (1,2-b) pyridazine compounds, which have potent AChE inhibitory activity, were also capable of antiproliferative, anti-migratory, and anti-inflammatory effects at higher doses.
Collapse
|
2
|
Shi B, Hu X, He H, Fang W. Metformin suppresses breast cancer growth via inhibition of cyclooxygenase-2. Oncol Lett 2021; 22:615. [PMID: 34257723 PMCID: PMC8243079 DOI: 10.3892/ol.2021.12876] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Pre-clinical and on-going trials have indicated the advantage of using metformin as an anticancer drug alone or in combination with other chemotherapeutics for the treatment of patients with breast cancer. However, the mechanisms by which metformin attenuates tumorigenesis remain to be further elucidated. The present study investigated the anticancer effects of metformin in breast cancer and identified potential molecular targets of metformin using western blotting and immunohistochemical analysis. Metformin significantly decreased tumor cell proliferation in vitro and suppressed tumor growth in vivo. Moreover, it induced the activation of AMP-induced protein kinase and suppression of phosphorylated-eukaryotic translation initiation factor 4E-binding protein 1 (p-4E-BP1), a downstream effector of the mTOR signaling pathway, and decreased cyclin D1 levels in in vitro and in vivo experimental models. Additionally, metformin inhibited cyclooxygenase (COX)-2 expression. Clinically, high expression levels of COX-2 and p-4E-BP1 in tissues of patients with breast cancer were significantly associated with enhanced lymphatic metastasis and distant metastasis. Thus, the current data suggested that metformin may have potential value as a synergistic therapy targeting both the COX-2 and mTOR signaling pathways.
Collapse
Affiliation(s)
- Bin Shi
- Department of Medical Oncology, Fuzhou General Hospital of Fujian Medical University, East Hospital Affiliated to Xiamen University (The 900th Hospital of The Joint Logistics Support Force of The Chinese PLA), Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China.,Department of Medical Oncology, Longyan People's Hospital, Longyan, Fujian 364000, P.R. China
| | - Xinyu Hu
- Department of Medical Oncology, Fuzhou General Hospital of Fujian Medical University, East Hospital Affiliated to Xiamen University (The 900th Hospital of The Joint Logistics Support Force of The Chinese PLA), Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Huimin He
- Department of Medical Oncology, Fuzhou General Hospital of Fujian Medical University, East Hospital Affiliated to Xiamen University (The 900th Hospital of The Joint Logistics Support Force of The Chinese PLA), Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Wenzheng Fang
- Department of Medical Oncology, Fuzhou General Hospital of Fujian Medical University, East Hospital Affiliated to Xiamen University (The 900th Hospital of The Joint Logistics Support Force of The Chinese PLA), Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
3
|
Li W, Zhao L, Li Y, Zhai Z. Artesunate attenuates proliferation of epithelial cells by downregulating the NF-κB and AKT signaling pathways in benign mammary gland hyperplasia rats. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:848. [PMID: 34164482 PMCID: PMC8184455 DOI: 10.21037/atm-21-1447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background The aim of this study was to investigate the effects of artesunate (ART) on breast epithelial cell proliferation in vitro and in vivo. Methods Immortalized human non-cancer mammary epithelial (MCF-10A) cells were used to determine the effect of ART on estrogen-induced mammary hyperplasia cells. We investigated the effect of ART on the synthesis of cyclooxygenase-2 (COX-2) and proliferating cell nuclear antigen (PCNA) in MCF-10A by treating MCF-10A 36 h with different concentrations of ART (0, 100, 200, 400 µm, n=12/group). We then investigated the effect of ART on estrogen induced COX-2, PCNA, nuclear factor-kappa B (NF-κB), and pNF-κB synthesis by treating MCF-10A with both estrogen and ART (0, 50, 100, 200 µm, n=12/group). A mammary hyperplasia model (MGH) was established in rats. All rats (n=12) were divided into 4 groups [group A: negative control (NC) + Art −; group B: NC + Art +; group C: MGH + Art −; group D: MGH + Art +] by the random number table method and the effects of ART on estradiol-induced mammary hyperplasia, fibrosis, and phosphorylation of AKT and NF-κB were studied by histopathological staining, Masson trichrome staining, immunohistochemistry (IHC), and western blotting. Results The proliferation and inflammation of mammary epithelial cells were blocked by ART (P<0.05). The phosphorylation of NF-κB induced by estradiol in MCF-10A was attenuated by ART (P<0.05). In the rat MGH, ART reduced cell proliferation and fibrosis (P<0.05) and inhibited the phosphorylation of AKT and NF-κB (P<0.05). Conclusions The drug ART inhibits estrogen-induced breast hyperplasia by blocking AKT and NFkB phosphorylation.
Collapse
Affiliation(s)
- Wei Li
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Lina Zhao
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yiliang Li
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Zhai
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Jindal S, Pennock ND, Klug A, Narasimhan J, Calhoun A, Roberts MR, Tamimi RM, Eliassen AH, Weinmann S, Borges VF, Schedin P. S-nitrosylated and non-nitrosylated COX2 have differential expression and distinct subcellular localization in normal and breast cancer tissue. NPJ Breast Cancer 2020; 6:62. [PMID: 33298921 PMCID: PMC7686348 DOI: 10.1038/s41523-020-00204-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Immunohistochemical (IHC) staining in breast cancer shows both gain and loss of COX2 expression with disease risk and progression. We investigated four common COX2 antibody clones and found high specificity for purified human COX2 for three clones; however, recognition of COX2 in cell lysates was clone dependent. Biochemical characterization revealed two distinct forms of COX2, with SP21 recognizing an S-nitrosylated form, and CX229 and CX294 recognizing non-nitrosylated COX2 antigen. We found S-nitrosylated and non-nitrosylated COX2 occupy different subcellular locations in normal and breast cancer tissue, implicating distinct synthetic/trafficking pathways and function. Dual stains of ~2000 breast cancer cases show early-onset breast cancer had increased expression of both forms of COX2 compared to postmenopausal cases. Our results highlight the strengths of using multiple, highly characterized antibody clones for COX2 IHC studies and raise the prospect that S-nitrosylation of COX2 may play a role in breast cancer biology.
Collapse
Affiliation(s)
- Sonali Jindal
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-ADM, Portland, OR, 97201, USA
| | - Nathan D Pennock
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Alex Klug
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Jayasri Narasimhan
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Andrea Calhoun
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA
| | - Michelle R Roberts
- Department of Dermatology, Massachusetts General Hospital, 50 Staniford Street, Suite 200, Boston, MA, 02114, USA
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02114, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02114, USA
| | - Sheila Weinmann
- Center for Health Research, Kaiser Permanente Northwest, 3800 N. Interstate Ave., Portland, OR, 97227, USA
| | - Virginia F Borges
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA
- Young Women's Breast Cancer Translational Program, School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA
| | - Pepper Schedin
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-CDCB, Portland, OR, 97201, USA.
- Knight Cancer Institute, Oregon Health & Science University, 2720 SW Moody Ave., Mailing Code: KR-ADM, Portland, OR, 97201, USA.
- School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA.
- Young Women's Breast Cancer Translational Program, School of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, MS8117, RC-1S, 8401K, 12801 E 17th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
5
|
Deciphering of Key Pharmacological Pathways of Poria Cocos Intervention in Breast Cancer Based on Integrated Pharmacological Method. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4931531. [PMID: 33149754 PMCID: PMC7603580 DOI: 10.1155/2020/4931531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 08/25/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Poria cocos (Fuling), a natural plant, has recently emerged as a promising strategy for cancer treatment. However, the molecular mechanisms of Poria cocos action in breast cancer remain poorly understood. METHODS TCMSP database was used to screen the potential active ingredients in Poria cocos. GEO database was used to identify differentially expressed genes. Network pharmacology was used to identify the specific pathways and key target proteins related to breast cancer. Finally, molecular docking was used to validate the results. RESULTS In our study, 237 targets were predicted for 15 potential active ingredients found in Poria cocos. An interaction network of predicted targets and genes differentially regulated in breast cancers was constructed. Based on the constructed network and further analysis including network topology, KEGG, survival analysis, and gene set enrichment analysis, 3 primary nodes were identified as key potential targets that were significantly enriched in the PPAR signaling pathway. CONCLUSION The results showed that potential active ingredients of Poria cocos might interfere with breast cancer through synergistic regulation of PTGS2, ESR1, and FOS.
Collapse
|
6
|
Sherman ME, Vierkant RA, Kaggal S, Hoskin TL, Frost MH, Denison L, Visscher DW, Carter JM, Winham SJ, Jensen MR, Radisky DC, Vachon CM, Degnim AC. Breast Cancer Risk and Use of Nonsteroidal Anti-inflammatory Agents After a Benign Breast Biopsy. Cancer Prev Res (Phila) 2020; 13:967-976. [PMID: 32718942 PMCID: PMC9509660 DOI: 10.1158/1940-6207.capr-20-0178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/04/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
Over one million women in the United States receive biopsy diagnoses of benign breast disease (BBD) each year, which confer a 1.5-4.0-fold increase in breast cancer risk. Studies in the general population suggest that nonsteroidal anti-inflammatory agents (NSAID) lower breast cancer risk; however, associations among women with BBD are unknown. We assessed whether NSAID use among women diagnosed with BBD is associated with lower breast cancer risk. Participants included 3,080 women (mean age = 50.3 ± 13.5 years) in the Mayo BBD surgical biopsy cohort diagnosed between January 1, 1992 and December 31, 2001 who completed breast cancer risk factor questionnaires that assessed NSAID use, and whose biopsies underwent detailed pathology review, masked to outcome. Women were followed from date of BBD biopsy to breast cancer diagnosis (main outcome) or censoring (death, prophylactic mastectomy, reduction mammoplasty, lobular carcinoma in situ or last contact). Median follow-up time was 16.4 ± 6.0 years. Incident breast cancer was diagnosed among 312 women over a median follow-up of 9.9 years. Regular non-aspirin NSAID use was associated with lower breast cancer risk [HR = 0.63; 95% confidence interval (CI) = 0.46-0.85; P = 0.002] with trends of lower risk (highest tertiles of use vs. nonuse) for greater number of years used [HR = 0.55; 95% CI = 0.31-0.97; P trend = 0.003), days used per month (HR = 0.51; 95% CI = 0.33-0.80; P trend = 0.001) and lifetime number of doses taken (HR = 0.53; 95% CI = 0.31-0.89; P trend = 0.003). We conclude that nonaspirin NSAID use is associated with statistically significant lower breast cancer risk after a BBD biopsy, including a dose-response effect, suggesting a potential role for NSAIDs in breast cancer prevention among patients with BBD.
Collapse
Affiliation(s)
- Mark E Sherman
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida.
| | | | | | | | - Marlene H Frost
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Lori Denison
- Department of Information Technology, Mayo Clinic, Rochester, Minnesota
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
7
|
Ren Q, Hou Y, Li X, Fan X. Silence of TPPP3 suppresses cell proliferation, invasion and migration via inactivating NF-κB/COX2 signal pathway in breast cancer cell. Cell Biochem Funct 2020; 38:773-781. [PMID: 32515139 DOI: 10.1002/cbf.3546] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/01/2020] [Accepted: 04/24/2020] [Indexed: 12/16/2022]
Abstract
Malignant phenotypes are leading causes of death in patients with breast cancer (BC). Previously, it has been proved that tubulin polymerization promoting protein 3 (TPPP3) participates in cell progressions in several human cancers. Little is known about the functions of TPPP3 in BC. Herein, we detected the expression of TPPP3 in 54 clinical BC tissues and two BC cell lines by immunohistochemistry and Western blot. CCK-8, wound healing, colony formation and Transwell assays were used to assess cell proliferation, clone formation, invasion and migration of MCF-7 and T47D cells after transfection with TPPP3 siRNA. Meanwhile, related-proteins expression was detected using Western blot. TPPP3 was found to be highly expressed in the tissues from the patients with BC. Poor outcomes were associated with the high expression of TPPP3 in all patients with BC. When MCF-7 and T47D cells receiving TPPP3 siRNA transfection, the capacities of proliferation, clone formation, invasion and migration were suppressed and the expression of MMP-2/-9 and NF-κB p65/COX2 was notably reduced. The dual-luciferase reporter assay indicated that the promoter regions of NF-κB p65 could combine to TPPP3. Overall, the present study demonstrated that TPPP3 played a significant role in BC, and its inhibition lead to the suppression of NF-κB/COX-2 signalling pathway along with the reduction of malignant phenotypes. SIGNIFICANCE OF THIS STUDY: Previously, it has been proved that tubulin polymerization promoting protein 3 (TPPP3) participates in cell progression in several human cancers. Little is known about the function of TPPP3 in BC. Our study was the first direct evidence to support the role of TPPP3 in tumorigenesis and metastasis of BC. Although the underlying mechanism has not been fully delineated, these findings suggested that TPPP3 was an important factor in the tumour progression and metastasis of BC cells and provided a molecular basis for potential therapeutic implications in the treatment of patients with BC.
Collapse
Affiliation(s)
- Qianfeng Ren
- Department of Pathology, Jincheng People's Hospital, Jincheng, China
| | - Yugui Hou
- Department of Pathology, Jincheng People's Hospital, Jincheng, China
| | - Xiaoying Li
- Department of Pathology, Jincheng People's Hospital, Jincheng, China
| | - Xiaoe Fan
- Department of Ophthalmology, Jincheng People's Hospital, Shanxi Province, China
| |
Collapse
|
8
|
Li ZY, Yin YF, Guo Y, Li H, Xu MQ, Liu M, Wang JR, Feng ZH, Duan XC, Zhang S, Zhang SQ, Wang GX, Liao A, Wang SM, Zhang X. Enhancing Anti-Tumor Activity of Sorafenib Mesoporous Silica Nanomatrix in Metastatic Breast Tumor and Hepatocellular Carcinoma via the Co-Administration with Flufenamic Acid. Int J Nanomedicine 2020; 15:1809-1821. [PMID: 32214813 PMCID: PMC7083629 DOI: 10.2147/ijn.s240436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/21/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Because tumor-associated inflammation is a hallmark of cancer treatment, in the present study, sorafenib mesoporous silica nanomatrix (MSNM@SFN) co-administrated with flufenamic acid (FFA, a non-steroidal anti-inflammatory drug (NSAID)) was investigated to enhance the anti-tumor activity of MSNM@SFN. METHODS Metastatic breast tumor 4T1/luc cells and hepatocellular carcinoma HepG2 cells were selected as cell models. The effects of FFA in vitro on cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in 4T1/luc and HepG2 cells were investigated. The in vivo anti-tumor activity of MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) was evaluated in a 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model, respectively. RESULTS The results indicated that FFA could markedly decrease cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in both 4T1/luc and HepG2 cells. The enhanced anti-tumor activity of MSNM@SFN+FFA compared with that of MSNM@SFN was confirmed in the 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model in vivo, respectively. DISCUSSION MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) developed in this study is an alternative strategy for improving the therapeutic efficacy of MSNM@SFN via co-administration with NSAIDs.
Collapse
Affiliation(s)
- Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yi-Fan Yin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Yang Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Man Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Xiao-Chuan Duan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shuai-Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Guang-Xue Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Ai Liao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| | - Shu-Min Wang
- Department of Ultrasound, Peking University Third Hospital, Peking University, Beijing100191, People’s Republic of China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing100191, People’s Republic of China
| |
Collapse
|
9
|
Wyatt GL, Crump LS, Young CM, Wessells VM, McQueen CM, Wall SW, Gustafson TL, Fan YY, Chapkin RS, Porter WW, Lyons TR. Cross-talk between SIM2s and NFκB regulates cyclooxygenase 2 expression in breast cancer. Breast Cancer Res 2019; 21:131. [PMID: 31783895 PMCID: PMC6884910 DOI: 10.1186/s13058-019-1224-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 11/07/2019] [Indexed: 02/10/2023] Open
Abstract
Background Breast cancer is a leading cause of cancer-related death for women in the USA. Thus, there is an increasing need to investigate novel prognostic markers and therapeutic methods. Inflammation raises challenges in treating and preventing the spread of breast cancer. Specifically, the nuclear factor kappa b (NFκB) pathway contributes to cancer progression by stimulating proliferation and preventing apoptosis. One target gene of this pathway is PTGS2, which encodes for cyclooxygenase 2 (COX-2) and is upregulated in 40% of human breast carcinomas. COX-2 is an enzyme involved in the production of prostaglandins, which mediate inflammation. Here, we investigate the effect of Singleminded-2s (SIM2s), a transcriptional tumor suppressor that is implicated in inhibition of tumor growth and metastasis, in regulating NFκB signaling and COX-2. Methods For in vitro experiments, reporter luciferase assays were utilized in MCF7 cells to investigate promoter activity of NFκB and SIM2. Real-time PCR, immunoblotting, immunohistochemistry, and chromatin immunoprecipitation assays were performed in SUM159 and MCF7 cells. For in vivo experiments, MCF10DCIS.COM cells stably expressing SIM2s-FLAG or shPTGS2 were injected into SCID mice and subsequent tumors harvested for immunostaining and analysis. Results Our results reveal that SIM2 attenuates the activation of NFκB as measured using NFκB-luciferase reporter assay. Furthermore, immunostaining of lysates from breast cancer cells overexpressing SIM2s showed reduction in various NFκB signaling proteins, as well as pAkt, whereas knockdown of SIM2 revealed increases in NFκB signaling proteins and pAkt. Additionally, we show that NFκB signaling can act in a reciprocal manner to decrease expression of SIM2s. Likewise, suppressing NFκB translocation in DCIS.COM cells increased SIM2s expression. We also found that NFκB/p65 represses SIM2 in a dose-dependent manner, and when NFκB is suppressed, the effect on the SIM2 is negated. Additionally, our ChIP analysis confirms that NFκB/p65 binds directly to SIM2 promoter site and that the NFκB sites in the SIM2 promoter are required for NFκB-mediated suppression of SIM2s. Finally, overexpression of SIM2s decreases PTGS2 in vitro, and COX-2 staining in vivo while decreasing PTGS2 and/or COX-2 activity results in re-expression of SIM2. Conclusion Our findings identify a novel role for SIM2s in NFκB signaling and COX-2 expression.
Collapse
Affiliation(s)
- Garhett L Wyatt
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Chloe M Young
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Veronica M Wessells
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Cole M McQueen
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Steven W Wall
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Tanya L Gustafson
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Yang-Yi Fan
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Weston W Porter
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA. .,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA.
| |
Collapse
|
10
|
|
11
|
Marzbany M, Bishayee A, Rasekhian M. Increased expression of ZNF 703 in breast cancer tissue: An opportunity for RNAi-NSAID combinatorial therapy. Biotechnol Appl Biochem 2019; 66:808-814. [PMID: 31195426 DOI: 10.1002/bab.1790] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/08/2019] [Indexed: 12/15/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are known to exhibit antitumor activities. Among the very well-known oncogenes in breast cancer is zinc finger protein 703 (ZNF703) and cyclooxygenase-2 (COX-2). Numerous reports indicate a direct link among apoptosis resistance, chemotherapy resistance, and increased expression of ZNF703. In the present study, the expression level of ZNF703 was compared in human breast cancer tissue, healthy breast tissue, and MCF-7 breast cancer cell line by a real-time PCR. We also investigated the inhibitory effect of anti-ZNF703 RNAi interference (RNAi) and ibuprofen, either individually or in combination, on MCF-7 cell survival and apoptosis. Results showed a 93.3% and fourfold increase in the expression of ZNF703 in breast cancer tissue and MCF-7 cell line, respectively. Ibuprofen inhibited the viability of MCF-7 cells in a concentration-dependent manner. Ibuprofen alone or in combination with anti-ZNF703 RNA reduced the expression of ZNF703, induced apoptosis, reduced mitochondrial membrane potential, and elevated BAX and LC3A in MCF-7 cells. Our results show that the combination of ibuprofen and anti-ZNF703 siRNA is more effective in promoting apoptosis than each treatment alone. We report that the combination of anti-ZNF703 RNAi with ibuprofen as the inhibitor of COX-2 is highly effective in inhibiting MCF-7 as a breast cancer cell line and shows therapeutic potential for breast cancer.
Collapse
Affiliation(s)
- Marzieh Marzbany
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermansha, Iran
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida
| | - Mahsa Rasekhian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermansha, Iran
| |
Collapse
|
12
|
Mojeiko G, de Brito M, Salata GC, Lopes LB. Combination of microneedles and microemulsions to increase celecoxib topical delivery for potential application in chemoprevention of breast cancer. Int J Pharm 2019; 560:365-376. [DOI: 10.1016/j.ijpharm.2019.02.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/23/2019] [Accepted: 02/06/2019] [Indexed: 12/16/2022]
|
13
|
Xu H, Lin F, Wang Z, Yang L, Meng J, Ou Z, Shao Z, Di G, Yang G. CXCR2 promotes breast cancer metastasis and chemoresistance via suppression of AKT1 and activation of COX2. Cancer Lett 2017; 412:69-80. [PMID: 28964785 DOI: 10.1016/j.canlet.2017.09.030] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 09/17/2017] [Accepted: 09/21/2017] [Indexed: 01/07/2023]
Abstract
Metastasis and chemoresistance are two major causes of breast cancer death. We show here that the chemokine receptor CXCR2 was overexpressed in breast cancer cell lines and tissues. CXCR2 promoted anti-apoptosis, anti-senescence, and epithelial-to-mesenchymal transition (EMT) of breast cancer cells, leading to the enhanced metastasis and chemoresistance. Further study suggested that AKT1 and cyclooxygenase-2 (COX2; PTGS2) might mediate the CXCR2 signaling to inversely control the breast cancer metastasis and chemoresistance through the regulation of EMT, apoptosis, and senescence. Analyses of clinical data indicate that the high expression of CXCR2 was correlated with the high expression of COX2 and the low expression of AKT1, P85α, E-cadherin, and β-catenin in cancer tissues. Poor outcomes were associated with the high expression of CXCR2 or COX2 while favorable survivals were associated with the high expression of P85α, AKT1, or E-cadherin in all cancer patients. Cox multivariate analysis demonstrated that CXCR2, COX2, and AKT1 could be independent predictors for disease free survivals. All these data suggest that CXCR2 promotes breast cancer metastasis and chemoresistance via suppressing AKT1 and activating COX2. Thus, antagonists of the CXCR2 signaling molecules may be used to treat breast cancer patients particularly with high metastasis and chemoresistance.
Collapse
Affiliation(s)
- Han Xu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Fengjuan Lin
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Ziliang Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Lina Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Jiao Meng
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Zhouluo Ou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China
| | - Zhimin Shao
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China.
| | - Genhong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China.
| | - Gong Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 200032, China; Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, 200240, China.
| |
Collapse
|
14
|
Molecular profile of atypical hyperplasia of the breast. Breast Cancer Res Treat 2017; 167:9-29. [DOI: 10.1007/s10549-017-4488-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022]
|
15
|
Winham SJ, Mehner C, Heinzen EP, Broderick BT, Stallings-Mann M, Nassar A, Vierkant RA, Hoskin TL, Frank RD, Wang C, Denison LA, Vachon CM, Frost MH, Hartmann LC, Aubrey Thompson E, Sherman ME, Visscher DW, Degnim AC, Radisky DC. NanoString-based breast cancer risk prediction for women with sclerosing adenosis. Breast Cancer Res Treat 2017; 166:641-650. [PMID: 28798985 PMCID: PMC5668350 DOI: 10.1007/s10549-017-4441-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/04/2017] [Indexed: 01/13/2023]
Abstract
Purpose Sclerosing adenosis (SA), found in ¼ of benign breast disease (BBD) biopsies, is a histological feature characterized by lobulocentric proliferation of acini and stromal fibrosis and confers a two-fold increase in breast cancer risk compared to women in the general population. We evaluated a NanoString-based gene expression assay to model breast cancer risk using RNA derived from formalin-fixed, paraffin-embedded (FFPE) biopsies with SA. Methods The study group consisted of 151 women diagnosed with SA between 1967 and 2001 within the Mayo BBD cohort, of which 37 subsequently developed cancer within 10 years (cases) and 114 did not (controls). RNA was isolated from benign breast biopsies, and NanoString-based methods were used to assess expression levels of 61 genes, including 35 identified by previous array-based profiling experiments and 26 from biological insight. Diagonal linear discriminant analysis of these data was used to predict cancer within 10 years. Predictive performance was assessed with receiver operating characteristic area under the curve (ROC-AUC) values estimated from 5-fold cross-validation. Results Gene expression prediction models achieved cross-validated ROC-AUC estimates ranging from 0.66 to 0.70. Performing univariate associations within each of the five folds consistently identified genes DLK2, EXOC6, KIT, RGS12, and SORBS2 as significant; a model with only these five genes showed cross-validated ROC-AUC of 0.75, which compared favorably to risk prediction using established clinical models (Gail/BCRAT: 0.57; BBD-BC: 0.67). Conclusions Our results demonstrate that biomarkers of breast cancer risk can be detected in benign breast tissue years prior to cancer development in women with SA. These markers can be assessed using assay methods optimized for RNA derived from FFPE biopsy tissues which are commonly available. Electronic supplementary material The online version of this article (doi:10.1007/s10549-017-4441-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stacey J Winham
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Christine Mehner
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ethan P Heinzen
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Brendan T Broderick
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Aziza Nassar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Robert A Vierkant
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Tanya L Hoskin
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ryan D Frank
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Chen Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lori A Denison
- Department of Information Technology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Celine M Vachon
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Marlene H Frost
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lynn C Hartmann
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Mark E Sherman
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
16
|
Kochel TJ, Goloubeva OG, Fulton AM. Upregulation of Cyclooxygenase-2/Prostaglandin E2 (COX-2/PGE2) Pathway Member Multiple Drug Resistance-Associated Protein 4 (MRP4) and Downregulation of Prostaglandin Transporter (PGT) and 15-Prostaglandin Dehydrogenase (15-PGDH) in Triple-Negative Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:61-70. [PMID: 27257388 PMCID: PMC4881873 DOI: 10.4137/bcbcr.s38529] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
Elevated levels of cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) are indicators of a poor prognosis in breast cancer. Using several independent publicly available breast cancer gene expression databases, we investigated other members of the PGE2 pathway. PGE2 is produced by COX-2 and actively exported by multiple drug resistance-associated protein 4 (MRP4) into the extracellular microenvironment, where PGE2 can bind four cognate EP receptors (EP1–EP4) and initiate diverse biological signaling pathways. Alternatively, PGE2 is imported via the prostaglandin transporter (PGT) and metabolized by 15-prostaglandin dehydrogenase (15-PGDH/HPGD). We made the novel observation that MRP4, PGT, and 15-PGDH are differentially expressed among distinct breast cancer molecular subtypes; this finding was confirmed in independent datasets. In triple-negative breast cancer, the observed gene expression pattern (high COX-2, high MRP4, low PGT, and low 15-PGDH) would favor high levels of tumor-promoting PGE2 in the tumor microenvironment that may contribute to the overall poor prognosis of triple-negative breast cancer.
Collapse
Affiliation(s)
- Tyler J Kochel
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| | - Olga G Goloubeva
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA.; Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amy M Fulton
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA.; Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.; Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| |
Collapse
|
17
|
Lee O, Khan SA. Novel routes for administering chemoprevention: local transdermal therapy to the breasts. Semin Oncol 2016; 43:107-115. [DOI: 10.1053/j.seminoncol.2015.09.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
18
|
Incorporating Biomarkers in Studies of Chemoprevention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 882:69-94. [PMID: 26987531 DOI: 10.1007/978-3-319-22909-6_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite Food and Drug Administration approval of tamoxifen and raloxifene for breast cancer risk reduction and endorsement by multiple agencies, uptake of these drugs for primary prevention in the United States is only 4% for risk eligible women likely to benefit from their use. Side effects coupled with incomplete efficacy and lack of a survival advantage are the likely reasons. This disappointing uptake, after the considerable effort and expense of large Phase III cancer incidence trials required for approval, suggests that a new paradigm is required. Current prevention research is focused on (1) refining risk prediction, (2) exploring behavioral and natural product interventions, and (3) utilizing novel translational trial designs for efficacy. Risk biomarkers will play a central role in refining risk estimates from traditional models and selecting cohorts for prevention trials. Modifiable risk markers called surrogate endpoint or response biomarkers will continue to be used in Phase I and II prevention trials to determine optimal dose or exposure and likely effectiveness from an intervention. The majority of Phase II trials will continue to assess benign breast tissue for response and mechanism of action biomarkers. Co-trials are those in which human and animal cohorts receive the same effective dose and the same tissue biomarkers are assessed for modulation due to the intervention, but then additional animals are allowed to progress to cancer development. These collaborations linking biomarker modulation and cancer prevention may obviate the need for cancer incidence trials for non-prescription interventions.
Collapse
|
19
|
Nicolopoulou-Stamati P, Tsipis A, Chelidonis G, Patsouris E, Athanassiadou P, Gonidi M, Athanassiadou AM. Prognostic value of COX-2, P53, and EZH-2 evaluated by quantitative image analysis in premalignant and malignant breast lesions. Diagn Cytopathol 2014; 43:294-300. [PMID: 25355039 DOI: 10.1002/dc.23217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 04/09/2014] [Accepted: 10/06/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND Cytological differential diagnosis of atypical hyperplasia and well differentiated breast carcinoma may be challenging, because sometimes there is an overlap between the cytomorphological features of these lesions. The aim of the study was to investigate COX-2, EZH-2, p53 expression in carcinomas and the gray zone of breast cytology categories of atypical hyperplastic lesions with regard to biological behavior of the tumor. METHODS FNA speciments from 100 patients with breast hyperplastic lesions and cancer were investigated by immunocytochemistry and a quantitative analysis for COX-2, p53, and EZH-2. RESULTS Extent of staining for COX-2 correlated with percentage of positive for EZH-2 (P < 0.0001) and p53 nuclei (P < 0.001). The intensity of COX-2 was lower in the carcinoma group (118.57 ± 12.43) than in the hyperplastic (127.16 ± 11.71) group (P = 0.006). On the contrary the mean value of staining extent was greater in the adenocarcinoma cases (15.96 ± 13.03) than in hyperplastic (4.04 ± 1.94) cases (P < 0.0001). The percentage of EZH-2 and p53 positive cells correlated with the histological type of the lesions (P = 0.001 and P = 0.011, respectively). There was also a statistically significant relation between tumor size and expression of COX-2 (P = 0.007) and EZH-2 (P = 0.010). CONCLUSION Our study showed that the expression of COX-2, EZH-2, and p53 as determined by immunocytochemistry at quantitative level may be a predictor for distinguishing cytologically atypical hyperplastic from malignant breast lesions and may be regarded as potential prognostic factor in breast cancer patients.
Collapse
Affiliation(s)
- Polyxeni Nicolopoulou-Stamati
- National and Kapodistrian University of Athens, School of Medicine, First Department of Pathology and Cytology Unit, 1st Pathology Laboratory, 11527, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
20
|
Harris RE, Casto BC, Harris ZM. Cyclooxygenase-2 and the inflammogenesis of breast cancer. World J Clin Oncol 2014; 5:677-692. [PMID: 25302170 PMCID: PMC4129532 DOI: 10.5306/wjco.v5.i4.677] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 02/28/2014] [Accepted: 03/14/2014] [Indexed: 02/06/2023] Open
Abstract
Cohesive scientific evidence from molecular, animal, and human investigations supports the hypothesis that constitutive overexpression of cyclooxygenase-2 (COX-2) is a ubiquitous driver of mammary carcinogenesis, and reciprocally, that COX-2 blockade has strong potential for breast cancer prevention and therapy. Key findings include the following: (1) COX-2 is constitutively expressed throughout breast cancer development and expression intensifies with stage at detection, cancer progression and metastasis; (2) essential features of mammary carcinogenesis (mutagenesis, mitogenesis, angiogenesis, reduced apoptosis, metastasis and immunosuppression) are linked to COX-2-driven prostaglandin E2 (PGE-2) biosynthesis; (3) upregulation of COX-2 and PGE-2 expression induces transcription of CYP-19 and aromatase-catalyzed estrogen biosynthesis which stimulates unbridled mitogenesis; (4) extrahepatic CYP-1B1 in mammary adipose tissue converts paracrine estrogen to carcinogenic quinones with mutagenic impact; and (5) agents that inhibit COX-2 reduce the risk of breast cancer in women without disease and reduce recurrence risk and mortality in women with breast cancer. Recent sharp increases in global breast cancer incidence and mortality are likely driven by chronic inflammation of mammary adipose and upregulation of COX-2 associated with the obesity pandemic. The totality of evidence clearly supports the supposition that mammary carcinogenesis often evolves as a progressive series of highly specific cellular and molecular changes in response to induction of constitutive over-expression of COX-2 and the prostaglandin cascade in the “inflammogenesis of breast cancer”.
Collapse
|
21
|
COX2 expression in high-grade breast cancer: evidence for prognostic significance in the subset of triple-negative breast cancer patients. Med Oncol 2014; 31:989. [PMID: 24816739 DOI: 10.1007/s12032-014-0989-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/28/2014] [Indexed: 12/31/2022]
Abstract
COX2 expression correlates with high-grade breast cancer, but the clinical significance and possible prognostic influence in these patients have not been studied in depth. Our goal was to evaluate the significance of COX2 expression in a group of patients with high-grade breast cancer. Three hundred and three patients (median age 55; age range 25-95 years) with high-grade breast cancer entered this retrospective study. Mean follow-up was 65.2 months (4-179 months). COX2 expression was studied by immunohistochemistry. The distribution of patients with high-grade tumors according to staining for COX2 was as follows: score 0-28/303 (9.3 %); score 1-101/303 (33.3 %); score 2-114/303 (37.6 %); score 3-60/303 (19.8 %). Patients with score 2 and 3 were classified as COX2 positive (174 of 303 patients (57.4 %). There was no correlation between any clinicopathological pattern, ER, PR, Her2 status and COX2 expression. In the group of patients with triple-negative breast cancer, the 5-year disease-free survival rate was 58.3 % for patients with COX2 expression compared with 83.9 % for patients without COX2 expression (P = 0.042). COX2 expression did not provide any prognostic significance for the other biological subtypes of breast cancer with high-grade histological features.
Collapse
|
22
|
Degnim AC, Brahmbhatt RD, Radisky DC, Hoskin TL, Stallings-Mann M, Laudenschlager M, Mansfield A, Frost MH, Murphy L, Knutson K, Visscher DW. Immune cell quantitation in normal breast tissue lobules with and without lobulitis. Breast Cancer Res Treat 2014; 144:539-49. [PMID: 24596048 PMCID: PMC3962744 DOI: 10.1007/s10549-014-2896-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/20/2014] [Indexed: 12/26/2022]
Abstract
While the immune microenvironment has been investigated in breast cancers, little is known about its role in non-malignant breast tissues. Here we quantify and localize cellular immune components in normal breast tissue lobules, with and without visible immune infiltrates (lobulitis). Up to ten representative lobules each in eleven normal breast tissue samples were assessed for B cells (CD20), cytotoxic T cells (CD8), helper T cells (CD4), dendritic cells (CD11c), leukocytes (CD45), and monocytes/macrophages (CD68). Using digital image analysis, immune cell densities were measured and compared between lobules with/without lobulitis. 109 lobules in 11 normal breast tissue samples were evaluated; 31 with lobulitis and 78 without. Immune cells showed consistent patterns in all normal samples, predominantly localized to lobules rather than stroma. Regardless of lobulitis status, most lobules demonstrated CD8+, CD11c+, CD45+, and CD68+ cells, with lower densities of CD4+ and CD20+ cells. Both CD11c+ and CD8+ cells were consistently and intimately associated with the basal aspect of lobule epithelium. Significantly higher densities of CD4+, CD8+, CD20+, and CD45+ cells were observed in lobules with lobulitis. In contrast, densities of monocytes/macrophages and dendritic cells did not vary with lobulitis. In normal breast tissue, myeloid and lymphoid cells are present and localized to lobules, with cytotoxic T and dendritic cells directly integrated with epithelium. Lobules with lobulitis have significantly more adaptive immune (B and T) cells, but no increase in dendritic cells or monocytes/macrophages. These findings indicate an active and dynamic mucosal immune system in normal breast tissue.
Collapse
Affiliation(s)
- Amy C Degnim
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fornetti J, Jindal S, Middleton KA, Borges VF, Schedin P. Physiological COX-2 expression in breast epithelium associates with COX-2 levels in ductal carcinoma in situ and invasive breast cancer in young women. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1219-1229. [PMID: 24518566 DOI: 10.1016/j.ajpath.2013.12.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/09/2013] [Accepted: 12/19/2013] [Indexed: 11/19/2022]
Abstract
Cyclooxygenase-2 (COX-2) overexpression is implicated in increased risk and poorer outcomes in breast cancer in young women. We investigated COX-2 regulation in normal premenopausal breast tissue and its relationship to malignancy in young women. Quantitative COX-2 immunohistochemistry was performed on adjacent normal and breast cancer tissues from 96 premenopausal women with known clinical reproductive histories, and on rat mammary glands with distinct ovarian hormone exposures. COX-2 expression in the normal breast epithelium varied more than 40-fold between women and was associated with COX-2 expression levels in ductal carcinoma in situ and invasive cancer. Normal breast COX-2 expression was independent of known breast cancer prognostic indicators, including tumor stage and clinical subtype, indicating that factors regulating physiological COX-2 expression may be the primary drivers of COX-2 expression in breast cancer. Ovarian hormones, particularly at pregnancy levels, were identified as modulators of COX-2 in normal mammary epithelium. However, serial breast biopsy analysis in nonpregnant premenopausal women suggested relatively stable baseline levels of COX-2 expression, which persisted independent of menstrual cycling. These data provide impetus to investigate how baseline COX-2 expression is regulated in premenopausal breast tissue because COX-2 levels in normal breast epithelium may prove to be an indicator of breast cancer risk in young women, and predict the chemopreventive and therapeutic efficacy of COX-2 inhibitors in this population.
Collapse
MESH Headings
- Adult
- Animals
- Biomarkers, Tumor/analysis
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/enzymology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/enzymology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cyclooxygenase 2/biosynthesis
- Female
- Humans
- Immunoblotting
- Immunohistochemistry
- Middle Aged
- Rats
- Rats, Sprague-Dawley
- Young Adult
Collapse
Affiliation(s)
- Jaime Fornetti
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sonali Jindal
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kara A Middleton
- Laboratory of Genetics, National Institutes of Aging, Baltimore, Maryland
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Pepper Schedin
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Program in Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado; AMC Cancer Research Foundation, Denver, Colorado.
| |
Collapse
|
24
|
Sclerosing adenosis and risk of breast cancer. Breast Cancer Res Treat 2014; 144:205-12. [PMID: 24510013 PMCID: PMC3924024 DOI: 10.1007/s10549-014-2862-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 01/24/2014] [Indexed: 01/16/2023]
Abstract
Over one million American women have a benign breast biopsy annually. Sclerosing adenosis (SA) is a common, but poorly understood benign breast lesion demonstrating increased numbers of distorted lobules accompanied by stromal fibrosis. Few studies of its association with breast cancer have been conducted, with contradictory results. We studied SA in the Mayo Benign Breast Disease (BBD) Cohort, which includes women who had benign biopsies at Mayo-Rochester 1967–2001. Breast cancer risk in defined subsets was assessed using standardized incidence ratios (SIRs), relative to the Iowa Surveillance, Epidemiology, and End Results registry. This BBD cohort of 13,434 women was followed for a median of 15.7 years. SA was present in 3,733 women (27.8 %) who demonstrated an SIR for breast cancer of 2.10 (95 % CI 1.91–2.30) versus an SIR of 1.52 (95 % CI 1.42–1.63) for the 9,701 women without SA. SA was present in 62.4 % of biopsies with proliferative disease without atypia and 55.1 % of biopsies with atypical hyperplasia. The presence of SA stratified risk in subsets of women defined by age, involution status, and family history. However, SA does not further stratify risk in women diagnosed with other forms of proliferative breast disease, either with or without atypia. SA is a common proliferative lesion of the breast which, as a single feature, conveys an approximate doubling of breast cancer risk. Its role in breast carcinogenesis remains undefined; its presence may aid in risk prediction for women after a breast biopsy.
Collapse
|
25
|
Expression of COX-2, CD44v6 and CD147 and relationship with invasion and lymph node metastasis in hypopharyngeal squamous cell carcinoma. PLoS One 2013; 8:e71048. [PMID: 24019861 PMCID: PMC3760833 DOI: 10.1371/journal.pone.0071048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/26/2013] [Indexed: 12/11/2022] Open
Abstract
To assess the expression of COX-2,CD44v6 and CD147 in hypopharyngeal squamous cell carcinomas and the three biomarkers correlation with tumor invasion and lymph node metastasis of Chinese people. 101 cases of surgically excised primary tumor were included in this study, and 40 tissues of epithelium adjacent to carcinoma were used as controls. We characterized the immunohistochemical expression of COX-2, CD44v6, and CD147 in141 formalin-fixed, paraffin-embedded tissues, and measured the mean optical density (OD) of the positive area to identify the expression of the three bio-markers and relationship with tumor invasion and lymph node metastasis. Our study demonstrates that the expression of the COX-2 and CD147 were significantly increased in carcinoma tissues compared to the epithelium adjacent to carcinoma. We also observed that the expression of COX-2, CD44v6, and CD147 were significantly associated with T classification, lymph node metastasis and clinical stage. There was strong significant correlation among the three biomarkers as well. Additionally, we indicated that recurrence and ≥P50 level of COX-2 expression had an independent prognostic effect on prognosis. In conclusion, the three biomarkers play important roles in tumor invasion and lymph node metastases and might be valuable indicators of tumor metastasis in hypopharyngeal squamous cell carcinoma.
Collapse
|
26
|
Lin F, Luo J, Gao W, Wu J, Shao Z, Wang Z, Meng J, Ou Z, Yang G. COX-2 promotes breast cancer cell radioresistance via p38/MAPK-mediated cellular anti-apoptosis and invasiveness. Tumour Biol 2013; 34:2817-26. [PMID: 23771849 DOI: 10.1007/s13277-013-0840-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/01/2013] [Indexed: 12/21/2022] Open
Abstract
Radioresistance is one of the major barriers to improve the survival rate of breast cancer patients. Cyclooxygenase 2 (COX-2) is usually overexpressed in highly invasive and metastatic breast cancer, which may indicate an association with breast cancer radioresistance. The function role of COX-2 was investigated by using a radioresistant breast cancer cell line MDA-MB-231/RR10 and its parental cell line MDA-MB-231 cells before or after COX-2 was silenced by a specific small hairpin RNA (shRNA). The cell proliferation, migration, invasion, colony formation, and apoptosis were measured by CCK-8, scratch-wound, transwell, clone formation assay, and flow cytometry. Protein and mRNA expression were analyzed by Western blot and quantitative reverse transcriptase-polymerase chain reaction. COX-2 is upregulated in MDA-MB-231/RR10 cells compared with in MDA-MB-231 cells, and silencing of COX-2 expression by shRNA in MDA-MB-231/RR10 cells decreases the expression of Bcl-2 and Bcl-XL, but increases the proapoptotic protein BAK, leading to the increased apoptosis following treatment with γ-irradiation in comparison with those in control cells. Silencing of COX-2 also increases the expression of β-catenin and E-cadherin, two anti-invasion proteins, resulting in reduced cell migration and invasion tested by transwell chambers and wound-healing assays. Further study demonstrated that COX-2-induced radioresistance is negatively regulated through the phosphorylation of p38 at Tyr182, and that the phosphorylation of p38 induced by TNF-alpha reduces the expression of Bcl-2, BCL-XL, but increases β-catenin and E-cadherin, leading to the decreased invasiveness of cells. Our data suggest that COX-2, p38, Bcl-2, Bcl-XL, β-catenin, and E-cadherin may be considered as potential therapeutic targets against radioresistant breast cancer.
Collapse
Affiliation(s)
- Fengjuan Lin
- Cancer Research Laboratory, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang J, Scholtens D, Holko M, Ivancic D, Lee O, Hu H, Chatterton RT, Sullivan ME, Hansen N, Bethke K, Zalles CM, Khan SA. Lipid metabolism genes in contralateral unaffected breast and estrogen receptor status of breast cancer. Cancer Prev Res (Phila) 2013; 6:321-30. [PMID: 23512947 DOI: 10.1158/1940-6207.capr-12-0304] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Risk biomarkers that are specific to estrogen receptor (ER) subtypes of breast cancer would aid the development and implementation of distinct prevention strategies. The contralateral unaffected breast of women with unilateral breast cancer (cases) is a good model for defining subtype-specific risk because women with ER-negative (ER-) index primaries are at high risk for subsequent ER-negative primary cancers. We conducted random fine needle aspiration of the unaffected breasts of cases. Samples from 30 subjects [15 ER-positive (ER+) and 15 ER- cases matched for age, race and menopausal status] were used for Illumina expression array analysis. Findings were confirmed using quantitative real-time PCR (qRT-PCR) in the same samples. A validation set consisting of 36 subjects (12 ER+, 12 ER- and 12 standard-risk healthy controls) was used to compare gene expression across groups. ER- case samples displayed significantly higher expression of 18 genes/transcripts, 8 of which were associated with lipid metabolism on gene ontology analysis (GO: 0006629). This pattern was confirmed by qRT-PCR in the same samples, and in the 24 cases of the validation set. When compared to the healthy controls in the validation set, significant overexpression of 4 genes (DHRS2, HMGCS2, HPGD and ACSL3) was observed in ER- cases, with significantly lower expression of UGT2B11 and APOD in ER+ cases, and decreased expression of UGT2B7 in both subtypes. These data suggest that differential expression of lipid metabolism genes may be involved in the risk for subtypes of breast cancer, and are potential biomarkers of ER-specific breast cancer risk.
Collapse
Affiliation(s)
- Jun Wang
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yee LD, Agarwal D, Rosol TJ, Lehman A, Tian M, Hatton J, Heestand J, Belury MA, Clinton SK. The inhibition of early stages of HER-2/neu-mediated mammary carcinogenesis by dietary n-3 PUFAs. Mol Nutr Food Res 2012; 57:320-7. [PMID: 23213007 DOI: 10.1002/mnfr.201200445] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/08/2012] [Accepted: 10/22/2012] [Indexed: 12/18/2022]
Abstract
SCOPE We previously demonstrated that lifelong feeding of diets enriched in n-3 fatty acids such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) significantly inhibits HER-2/neu-mediated mammary tumorigenesis in mice. Of interest is whether dietary n-3 fatty acids exert effects at early stages of mammary carcinogenesis. METHODS AND RESULTS Seven-week-old female MMTV-HER-2/neu transgenic mice were randomized to AIN-based semipurified diets containing either fish or corn oil at 25% energy. Mice were evaluated at 25, 30, and 35 weeks with analysis of mammary glands for atypical ductal hyperplasia (hematoxylin and eosin), cell proliferation (Ki67 immunostaining), and fatty acid synthase and cyclooxygenase-2 gene expression (qRT-PCR). Tissue fatty acid profiles were quantitated by GC. Atypia grade decreased significantly in mice fed fish oil (p = 0.002). Mammary epithelial cells in mammary glands from mice fed fish oil also had an eightfold lower percentage of Ki67 expression. COX-2 expression in mammary fat-pads significantly decreased in mice fed fish versus corn oil enriched diets. CONCLUSION Dietary fish oil inhibits atypical ductal hyperplasia at early stages of HER-2/neu-mediated mammary carcinogenesis relative to corn oil diets. This histologic change is associated with suppression of mammary epithelial cell proliferation and decreased COX-2 expression in mammary tissue.
Collapse
Affiliation(s)
- Lisa D Yee
- Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Borges VF, Schedin P. Could NSAIDs become a preventative therapy in pregnancy-associated breast cancer? BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Pregnancy-associated breast cancer (PABC) is a unique type of young women’s breast cancer that includes two biologically distinct conditions: those diagnosed during pregnancy and those diagnosed postpartum. It is the dominant subset of postpartum PABC that is more consistently associated with higher breast cancer mortality. Preclinical work has identified the normal event of postpartum involution as a wound-healing milieu rich in immune cells. We have shown that the involution environment drives tumor growth, proliferation and metastasis. Moreover, we have demonstrated in animal models that this ‘involution effect’ can be abrogated with drug therapy, namely NSAIDs, which target normal involution pathways implicated in PABC tumor promotion. In this perspective, we review the contemporary understanding of PABC, our preclinical modeling and its implications and the unmet research needs required for future translation of these preclinical studies into rational and safe human clinical trials.
Collapse
Affiliation(s)
- Virginia F Borges
- University of Colorado Denver Anschutz Medical Campus, 12801 E. 17th Avenue, Room 8112, Aurora, CO 80045, USA
| | - Pepper Schedin
- AMC Cancer Research Center, 3401 Quebec Street, Suite 3200, Denver, CO 80207, USA
| |
Collapse
|
30
|
Degnim AC, Visscher DW, Hoskin TL, Frost MH, Vierkant RA, Vachon CM, Shane Pankratz V, Radisky DC, Hartmann LC. Histologic findings in normal breast tissues: comparison to reduction mammaplasty and benign breast disease tissues. Breast Cancer Res Treat 2012; 133:169-77. [PMID: 21881938 PMCID: PMC3242875 DOI: 10.1007/s10549-011-1746-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 08/16/2011] [Indexed: 11/30/2022]
Abstract
Investigations of breast carcinogenesis often rely upon comparisons between cancer tissue and nonmalignant breast tissue. It is unclear how well common reference sources of nonmalignant breast tissues reflect normal breast tissue. Breast tissue samples were evaluated from three sources: (1) normal donor tissues in the Susan G. Komen for the Cure Tissue Bank at Indiana University Simon Cancer Center (KTB), (2) women who underwent reduction mammaplasty (RM) at Mayo Clinic Rochester, and (3) the Mayo Clinic Benign Breast Disease Cohort Study (BBD). Samples were examined histologically and assessed for proliferative disease and degree of lobular involution. Univariate comparisons were performed among the study groups, and multivariate analyses were performed with logistic regression to assess the association between study group and the presence of epithelial proliferative disease and complete lobular involution. Histologic data were collected for 455 KTB samples, 259 RM samples, and 319 BBD samples. Histologic findings and the frequency of epithelial proliferation were significantly different among the groups. Histologic abnormalities were seen in a minority of the KTB samples (35%), whereas an abnormality was present in 88% of RM tissues and 97.5% of BBD samples. The presence of proliferative disease (with or without atypical hyperplasia) was present in 3.3% of normal donors (3.3%), 17% of RM samples, and 34.9% of BBD samples (P < 0.0001 for each comparison). Multivariate analyses confirmed that these differences remained significant and also showed higher likelihood of complete lobular involution in the normal donor samples compared to RM and BBD tissues. Compared to benign breast disease tissues and reduction mammaplasty tissues, breast tissue samples from normal donors have significantly fewer histologic abnormalities and a higher frequency of more complete lobular involution. Breast tissue samples from normal donors represent a unique tissue resource with histologic features consistent with lower breast cancer risk.
Collapse
Affiliation(s)
- Amy C Degnim
- Department of Surgery, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Development of imaging agents that can be used broadly for early detection of neoplasia at various tissue sites and at various stages of disease and that also can assess states of minimal residual disease would have tremendous utility in the diagnosis and management of cancer. In a series of articles culminating with a report in this issue of the journal (beginning on page 1536), Uddin and colleagues show their ability to systemically target the enzyme COX-2 with imaging probes that will serve as agents for early detection, risk assessment, prognosis, and intervention outcome measures. These probes will enable the detection and localization of regions of inflammation and a wide variety of premalignant lesions and cancers, with utility in monitoring the effects of cancer prevention and therapy.
Collapse
Affiliation(s)
- David A Ostrov
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, USA
| | | |
Collapse
|
32
|
Allred DC. Molecular Biomarkers of Risk in Premalignancy and Breast Cancer Prevention: Figure 1. Cancer Prev Res (Phila) 2011; 4:1947-52. [DOI: 10.1158/1940-6207.capr-11-0478] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Radisky DC, Santisteban M, Berman HK, Gauthier ML, Frost MH, Reynolds CA, Vierkant RA, Pankratz VS, Visscher DW, Tlsty TD, Hartmann LC. p16(INK4a) expression and breast cancer risk in women with atypical hyperplasia. Cancer Prev Res (Phila) 2011; 4:1953-60. [PMID: 21920875 DOI: 10.1158/1940-6207.capr-11-0282] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
p16, a nuclear protein encoded by the p16(INK4a) gene, is a regulator of cell-cycle regulation. Previous studies have shown that expression of p16 in tissue biopsies of patients with ductal carcinoma in situ (DCIS) is associated with increased risk of breast cancer, particularly when considered in combination with other markers such as Ki-67 and COX-2. Here, we evaluated how expression of p16 in breast tissue biopsies of women with atypical hyperplasia (AH), a putative precursor lesion to DCIS, is associated with subsequent development of cancer. p16 expression was assessed by immunohistochemistry in archival sections from 233 women with AH diagnosed at the Mayo Clinic. p16 expression in the atypical lesions was scored by percentage of positive cells and intensity of staining. We also studied coexpression of p16, with Ki-67 and COX-2, biomarkers of progression in AH. Risk factor and follow-up data were obtained via study questionnaire and medical records. Forty-seven patients (20%) developed breast cancer with a median follow-up of 14.5 years. Staining of p16 was increased in older patients relative to younger patients (P = 0.0025). Although risk of developing breast cancer was not associated with increased p16 expression, joint overexpression of Ki-67 and COX-2 was found to convey stronger risk of breast cancer in the first 10 years after diagnosis as compared with one negative marker (P < 0.01). However, the addition of p16 levels did not strengthen this association. p16 overexpression, either alone or in combination with COX-2 and Ki-67, does not significantly stratify breast cancer risk in women with AH.
Collapse
Affiliation(s)
- Derek C Radisky
- Division of Biochemistry/Molecular Biology, Mayo Clinic in Jacksonville, Jacksonville, Florida 32224, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lyons TR, O’Brien J, Borges V, Conklin MW, Keely PJ, Eliceiri KW, Marusyk A, Tan AC, Schedin P. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nat Med 2011; 17:1109-15. [PMID: 21822285 PMCID: PMC3888478 DOI: 10.1038/nm.2416] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 06/10/2011] [Indexed: 11/08/2022]
Abstract
The prognosis of breast cancer in young women is influenced by reproductive history. Women diagnosed within 5 years postpartum have worse prognosis than nulliparous women or women diagnosed during pregnancy. Here we describe a mouse model of postpartum breast cancer that identifies mammary gland involution as a driving force of tumor progression. In this model, human breast cancer cells exposed to the involuting mammary microenvironment form large tumors that are characterized by abundant fibrillar collagen, high cyclooxygenase-2 (COX-2) expression and an invasive phenotype. In culture, tumor cells are invasive in a fibrillar collagen and COX-2-dependent manner. In the involuting mammary gland, inhibition of COX-2 reduces the collagen fibrillogenesis associated with involution, as well as tumor growth and tumor cell infiltration to the lung. These data support further research to determine whether women at high risk for postpartum breast cancer would benefit from treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) during postpartum involution.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/physiopathology
- Carcinoma, Ductal/drug therapy
- Carcinoma, Ductal/physiopathology
- Celecoxib
- Cell Line, Tumor
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Female
- Fibrillar Collagens/metabolism
- Humans
- Ibuprofen/pharmacology
- Ibuprofen/therapeutic use
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/physiology
- Mice
- Mice, SCID
- Neoplasm Invasiveness/physiopathology
- Postpartum Period/drug effects
- Postpartum Period/physiology
- Pregnancy
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
Collapse
Affiliation(s)
- Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, MS8117, RC-1S, 8401K, 12801 E 17 Ave, Aurora, CO, USA 80045
| | - Jenean O’Brien
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, MS8117, RC-1S, 8401K, 12801 E 17 Ave, Aurora, CO, USA 80045
- Program in Cancer Biology, University of Colorado Denver, MS8104, RC-1S, 5117, 12801 E 17 Ave, Aurora, CO, USA 80045
| | - Virginia Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, MS8117, RC-1S, 8401K, 12801 E 17 Ave, Aurora, CO, USA 80045
- University of Colorado Cancer Center, Bldg 500, Suite 6004C, 13001 E 17 Place, Aurora, CO, USA 0045
| | - Matthew W Conklin
- Department of Cell and Regenerative Biology and UW Carbone Cancer Center, 1525 Linden Drive, University of Wisconsin, Madison, WI, USA 53706
- Laboratory of Cell and Molecular Biology, Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA 53706
| | - Patricia J Keely
- Department of Cell and Regenerative Biology and UW Carbone Cancer Center, 1525 Linden Drive, University of Wisconsin, Madison, WI, USA 53706
- Laboratory of Cell and Molecular Biology, Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA 53706
| | - Kevin W Eliceiri
- Laboratory of Cell and Molecular Biology, Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA 53706
| | - Andriy Marusyk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA 02115
| | - Aik-Choon Tan
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, MS8117, RC-1S, 8401K, 12801 E 17 Ave, Aurora, CO, USA 80045
- University of Colorado Cancer Center, Bldg 500, Suite 6004C, 13001 E 17 Place, Aurora, CO, USA 0045
| | - Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, MS8117, RC-1S, 8401K, 12801 E 17 Ave, Aurora, CO, USA 80045
- Program in Cancer Biology, University of Colorado Denver, MS8104, RC-1S, 5117, 12801 E 17 Ave, Aurora, CO, USA 80045
- University of Colorado Cancer Center, Bldg 500, Suite 6004C, 13001 E 17 Place, Aurora, CO, USA 0045
- AMC Cancer Research Center, 3401 Quebec Street, Suite 3200, Denver, CO 80207
| |
Collapse
|
35
|
Rizzo MT. Cyclooxygenase-2 in oncogenesis. Clin Chim Acta 2010; 412:671-87. [PMID: 21187081 DOI: 10.1016/j.cca.2010.12.026] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/20/2010] [Accepted: 12/21/2010] [Indexed: 02/07/2023]
Abstract
Compelling experimental and clinical evidence supports the notion that cyclooxygenase-2, the inducible isoform of cyclooxygenase, plays a crucial role in oncogenesis. Clinical and epidemiological data indicate that aberrant regulation of cyclooxygenase-2 in certain solid tumors and hematological malignancies is associated with adverse clinical outcome. Moreover, findings extrapolated from experimental studies in cultured tumor cells and animal tumor models indicate that cyclooxygenase-2 critically influences all stages of tumor development from tumor initiation to tumor progression. Cyclooxygenase-2 elicits cell-autonomous effects on tumor cells resulting in stimulation of growth, increased cell survival, enhanced tumor cell invasiveness, stimulation of neovascularization, and tumor evasion from the host immune system. Additionally, the oncogenic effects of cyclooxygenase-2 stem from its unique ability to impact tumor cell surroundings and create a proinflammatory environment conducive for tumor development, growth and progression. The initial enthusiasm generated by the availability of cyclooxygenase-2 selective inhibitors for cancer prevention and therapy has been lessened by the severe cardiovascular adverse side effects associated with their long-term use, as well as by the mixed results of recent clinical trials evaluating the efficacy of cyclooxygenase-2 inhibitors in adjuvant chemotherapy. Therefore, our ability to efficiently target the oncogenic effects of cyclooxygenase-2 for therapeutic and preventive purposes strictly depends on a better understanding of the spatial and temporal aspects of its activation in tumor cells along with a clearer elucidation of the signaling networks whereby cyclooxygenase-2 affects tumor cells and their interactions with the tumor microenvironment. This knowledge has the potential of leading to the identification of novel cyclooxygenase-2-dependent molecular and signaling networks that can be exploited to improve cancer prevention and therapy.
Collapse
Affiliation(s)
- Maria Teresa Rizzo
- Signal Transduction Laboratory, Methodist Research Institute, Clarian Health and Department of Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
36
|
Cunha POR, Ornstein M, Jones JL. Progression of Ductal Carcinoma in Situ from the Pathological Perspective. ACTA ACUST UNITED AC 2010; 5:233-239. [PMID: 22590443 DOI: 10.1159/000319625] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS) now represents up to 20% of breast cancer cases, yet its behaviour is still poorly understood. Morphological classifications go some way to predicting prognosis, but more sophisticated approaches are required to better tailor therapy to the individual. A number of biological molecules have been identified that appear to relate to prognosis and, in model systems, promote progression to invasive disease. Some of these, such as COX-2, provide real therapeutic opportunities, whilst other marker combinations are showing promise in categorising women according to risk. Gene expression studies have led to an emerging molecular classification of invasive breast cancer, and it is now evident that at least some of these molecular subtypes can be identified at the pre-invasive stage. The difference in frequency of these subtypes between DCIS and invasive cancer may hold clues as to the biological mechanisms underpinning disease transition. It is increasingly clear that the host microenvironment can have a major impact on disease behaviour, and as well as acting as potential predictive factors, the altered microenvironment phenotype also offers novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pedro Oscar R Cunha
- Centre for Tumour Biology, Barts Institute of Cancer, John Vane Science Building, Barts and the London School of Medicine and Dentistry, London, UK
| | | | | |
Collapse
|
37
|
Expression of cyclooxygenase-2 (COX-2) in an advanced metastasized hypopharyngeal carcinoma and cultured tumor cells. Oral Maxillofac Surg 2010; 14:53-7. [PMID: 19821125 DOI: 10.1007/s10006-009-0181-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE The inducible enzyme cyclooxygenase-2 (COX-2) catalyzes PGE(2) production and plays an important role in the progression of many solid cancers. However, the role of COX-2 expression in cervical lymph node metastases of head and neck cancer has not been clarified yet. PATIENT AND METHODS We comment on a male patient aged 53 who was admitted to an ENT-department with acute bleeding from an advanced hypopharyngeal carcinoma and a frontotemporal mass. Prior to palliative intended radiotherapy, the metastasis was resected. During the procedure, a small amount of tumor tissue was harvested for primary tumor cell culture. RESULTS COX-2 overexpression was demonstrated in the primary tumor tissue, the metastasis, in the cultured tumor cells by standard immunohistochemistry, as well as cytochemistry. CONCLUSIONS A simultaneous expression of COX-2 in head and neck carcinoma was presented for the first time. Besides the prognostic impact in oral carcinogenesis, this COX-2 role of biomarker for aggressive head and neck squamous cell carcinomas should be further evaluated. Additionally, treatment of hypopharyngeal carcinomas with selective COX-2 inhibitors could be beneficial when administered in combination with radiochemotherapy.
Collapse
|
38
|
Huang CC, Lo CP, Chiu CY, Shyur LF. Deoxyelephantopin, a novel multifunctional agent, suppresses mammary tumour growth and lung metastasis and doubles survival time in mice. Br J Pharmacol 2010; 159:856-71. [PMID: 20105176 DOI: 10.1111/j.1476-5381.2009.00581.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Elephantopus scaber L. (Asteraceae) is a traditional herbal medicine with anti-cancer effects. We evaluated the in vitro and in vivo efficacy of a major sesquiterpene lactone constituent of E. scaber, deoxyelephantopin (DET), against mammary adenocarcinoma and the underlying molecular mechanism. EXPERIMENTAL APPROACH A variety of cellular assays, immunoblotting and immunohistochemistry, as well as both orthotopic and metastatic TS/A tumour models in BALB/c mice, were used. Test mice were pretreated and post-treated with DET or paclitaxel and mammary tumour growth evaluated. KEY RESULTS DET (< or =2 microg x mL(-1)) significantly inhibited colony formation, cell proliferation, migration and invasion of TS/A cells and induced G(2)/M arrest and apoptosis in TS/A cells. c-Jun N-terminal kinase-mediated p21(Waf1/Cip1) expression and caspase activation cascades were up-regulated by DET, effects suppressed by N-acetyl-L-cysteine. Moreover, tumour necrosis factor alpha-induced matrix metalloproteinase-9 enzyme activity and expression and nuclear factor-kappa B activation were abolished by DET. Pretreatment with DET was more effective than paclitaxel, for profound suppression of orthotopic tumour growth (99% vs. 68% reduction in tumour size) and lung metastasis of TS/A cells (82% vs. 63% reduction in metastatic pulmonary foci) and prolonged median survival time (56 vs. 37 days, P < 0.01) in mice. The levels of cyclooxygenase-2 and vascular endothelial growth factor in metastatic lung tissues of TS/A-bearing mice were attenuated by DET. CONCLUSIONS AND IMPLICATIONS Our data provide evidence for the suppression of mammary adenocarcinoma by DET with several mechanisms and suggest that DET has potential as a chemopreventive agent for breast cancer.
Collapse
Affiliation(s)
- Chi-Chang Huang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | | |
Collapse
|
39
|
Fordyce C, Fessenden T, Pickering C, Jung J, Singla V, Berman H, Tlsty T. DNA damage drives an activin a-dependent induction of cyclooxygenase-2 in premalignant cells and lesions. Cancer Prev Res (Phila) 2009; 3:190-201. [PMID: 20028875 DOI: 10.1158/1940-6207.capr-09-0229] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cyclooxygenase-2 (COX-2) catalyzes the rate-limiting step in the synthesis of prostaglandins. Its overexpression induces numerous tumor-promoting phenotypes and is associated with cancer metastasis and poor clinical outcome. Although COX-2 inhibitors are promising chemotherapeutic and chemopreventative agents for cancer, the risk of significant cardiovascular and gastrointestinal complications currently outweighs their potential benefits. Systemic complications of COX-2 inhibition could be avoided by specifically decreasing COX-2 expression in epithelial cells. To that end, we have investigated the signal transduction pathway regulating the COX-2 expression in response to DNA damage in breast epithelial cells. In variant human mammary epithelial cells that have silenced p16 (vHMEC), double-strand DNA damage or telomere malfunction results in a p53- and activin A-dependent induction of COX-2 and continued proliferation. In contrast, telomere malfunction in HMEC with an intact p16/Rb pathway induces cell cycle arrest. Importantly, in ductal carcinoma in situ lesions, high COX-2 expression is associated with high gammaH2AX, TRF2, activin A, and telomere malfunction. These data show that DNA damage and telomere malfunction can have both cell-autonomous and cell-nonautonomous consequences and can provide a novel mechanism for the propagation of tumorigenesis.
Collapse
Affiliation(s)
- Colleen Fordyce
- Department of Pathology, University of California, San Francisco, 94143, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Santisteban M, Reynolds C, Barr Fritcher EG, Frost MH, Vierkant RA, Anderson SS, Degnim AC, Visscher DW, Pankratz VS, Hartmann LC. Ki67: a time-varying biomarker of risk of breast cancer in atypical hyperplasia. Breast Cancer Res Treat 2009; 121:431-7. [PMID: 19774459 DOI: 10.1007/s10549-009-0534-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 08/26/2009] [Indexed: 02/05/2023]
Abstract
Uncontrolled proliferation is a defining feature of the malignant phenotype. Ki67 is a marker for proliferating cells and is overexpressed in many breast cancers. Atypical hyperplasia is a premalignant lesion of the breast (relative risk approximately 4.0). Here, we asked if Ki67 expression could stratify risk in women with atypia. Ki67 expression was assessed immunohistochemically by digital image analysis in archival sections from 192 women with atypia diagnosed at the Mayo Clinic 1/1/67-12/31/91. Risk factor and follow-up data were obtained via study questionnaire and medical records. Observed breast cancer events were compared to population expected rates (Iowa SEER) using standardized incidence ratios (SIRs). We examined two endpoints: risk of breast cancer within 10 years and after 10 years of atypia biopsy. Thirty-two (16.7%) of the 192 women developed breast cancer over a median of 14.6 years. Thirty percent (58) of the atypias had >or=2% cells staining for Ki67. In these women, the risk of breast cancer within 10 years after atypia was increased (SIR 4.42 [2.21-8.84]) but not in those with <2% staining. Specifically, the cumulative incidence for breast cancer at 10 years was 14% in the high Ki67 vs. 3% in the low Ki67 group. Conversely, after 10 years, risk in the low Ki67 group rose significantly (SIR 5.69 [3.63-8.92]) vs. no further increased risk in the high Ki67 group (SIR 0.78 [0.11-5.55]). Ki67 appears to be a time-varying biomarker of risk of breast cancer in women with atypical hyperplasia.
Collapse
Affiliation(s)
- Marta Santisteban
- Department of Oncology, Clinica Universitaia de Navarra, Navarra, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Peralta EA, Murphy LL, Minnis J, Louis S, Dunnington GL. American Ginseng inhibits induced COX-2 and NFKB activation in breast cancer cells. J Surg Res 2009; 157:261-7. [PMID: 19815237 DOI: 10.1016/j.jss.2009.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 04/28/2009] [Accepted: 05/04/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Epidemiologic evidence suggests reduced breast cancer mortality in users of American Ginseng (AG) (Panax quinquefolium). We hypothesized that AG extract decreases proliferation of human breast cancer cells via an anti-inflammatory effect applicable to the prevention of breast and other cancers. MATERIAL AND METHODS A defined lyophilized aqueous extract of AG (LEAG) was dissolved in DMSO 1mg/mL, and serially diluted in saline. The cell lines MDA MB 231 and MCF7 were stimulated with the phorbol ester PDBu and treated with 100-500 mcg/mL LEAG. Proliferation was measured by MDA assay. Induced COX-2 expression was assayed by ELISA. Activation of NFkappaB by phosphorylation of the p65 subunit was quantified by CASE (cellular activation of signaling ELISA). RESULTS Both cell lines had reduced proliferation when treated with LEAG. PDBu stimulation of MDA MB 231 increased expression of the COX-2 protein 20-fold at 48 hours (P<0.005). COX-2 protein expression remained at baseline concentrations in PDBu- treated MDA MB 231 cells exposed to 100 mcg/mL LEAG. The CASE assay showed a 4-fold increase in p65 activation 24 hours after PDBu treatment in normal medium, while phosphorylated p65 dropped below baseline in the cells treated with PDBu plus LEAG. CONCLUSION In MDA MB 231, COX-2 was inducible with PDBu. This induced COX-2 expression was blocked by 100 microgram/mL LEAG in a time course consistent with the decline in the activated p65 subunit of NFkappaB. These results provide an anti-inflammatory mechanism for a possible anti-cancer effect of American Ginseng.
Collapse
Affiliation(s)
- Elizabeth A Peralta
- Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois 62794-9638, USA.
| | | | | | | | | |
Collapse
|
43
|
Boughey JC, Hartmann LC, Pankratz VS. In Reply. J Clin Oncol 2009. [DOI: 10.1200/jco.2008.21.2498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
44
|
Hu M, Peluffo G, Chen H, Gelman R, Schnitt S, Polyak K. Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast. Proc Natl Acad Sci U S A 2009; 106:3372-7. [PMID: 19218449 PMCID: PMC2642666 DOI: 10.1073/pnas.0813306106] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Indexed: 12/29/2022] Open
Abstract
Epithelial-stromal cell interactions have an important role in breast tumor progression, but the molecular mechanisms underlying these effects are just beginning to be understood. We previously described that fibroblasts promote, whereas normal myoepithelial cells inhibit, the progression of ductal carcinoma in situ (DCIS) to invasive breast carcinomas by using a xenograft model of human DCIS. Here, we report that the tumor growth and progression-promoting effects of fibroblasts are at least in part due to increased COX-2 expression in tumor epithelial cells provoked by their interaction with fibroblasts. Up-regulation of COX-2 in DCIS xenografts resulted in increased VEGF and MMP14 expression, which may contribute to the larger weight and invasive histology of COX-2-expressing tumors. Administration of celecoxib, a selective COX-2 inhibitor, to tumor-bearing mice decreased xenograft tumor weight and inhibited progression to invasion. Coculture of fibroblasts with DCIS epithelial cells enhanced their motility and invasion, and this change was associated with increased MMP14 expression and MMP9 protease activity. We identified the NF-kappaB pathway as one of the mediators of stromal fibroblast-derived signals regulating COX-2 expression in tumor epithelial cells. Inhibition of NF-kappaB and COX-2 activity and down-regulation of MMP9 expression attenuated the invasion-promoting effects of fibroblasts. These findings support a role for COX-2 in promoting the progression of DCIS to invasive breast carcinomas, and suggest that therapeutic targeting of the NF-kappaB and prostaglandin signaling pathways might be used for the treatment and prevention of breast cancer.
Collapse
Affiliation(s)
- Min Hu
- Departments of Medical Oncology and
- Harvard Medical School, Boston, MA 02115
| | - Guillermo Peluffo
- Departments of Medical Oncology and
- Angel Honorio Roffo Oncology Institute, University of Buenos Aires, Buenos Aires, Argentina; and
| | - Haiyan Chen
- Biostatistics and Computational Biology, Dana–Farber Cancer Institute, Boston, MA 02115
- Harvard School of Public Health, Boston, MA 02115
| | - Rebecca Gelman
- Biostatistics and Computational Biology, Dana–Farber Cancer Institute, Boston, MA 02115
- Harvard School of Public Health, Boston, MA 02115
| | - Stuart Schnitt
- Harvard Medical School, Boston, MA 02115
- Department of Pathology, Beth-Israel Deaconess Medical Center, Boston, MA 02115
| | - Kornelia Polyak
- Departments of Medical Oncology and
- Harvard Medical School, Boston, MA 02115
| |
Collapse
|
45
|
Pankratz VS, Hartmann LC, Degnim AC, Vierkant RA, Ghosh K, Vachon CM, Frost MH, Maloney SD, Reynolds C, Boughey JC. Assessment of the accuracy of the Gail model in women with atypical hyperplasia. J Clin Oncol 2008; 26:5374-9. [PMID: 18854574 DOI: 10.1200/jco.2007.14.8833] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE An accurate estimate of a woman's breast cancer risk is essential for optimal patient counseling and management. Women with biopsy-confirmed atypical hyperplasia of the breast (atypia) are at high risk for breast cancer. The Gail model is widely used in these women, but has not been validated in them. PATIENTS AND METHODS Women with atypia were identified from the Mayo Benign Breast Disease (BBD) cohort (1967 to 1991). Their risk factors for breast cancer were obtained, and the Gail model was used to predict 5-year-and follow-up-specific risks for each woman. The predicted and observed numbers of breast cancers were compared, and the concordance between individual risk levels and outcomes was computed. RESULTS Of the 9,376 women in the BBD cohort, 331 women had atypia (3.5%). At a mean follow-up of 13.7 years, 58 of 331 (17.5%) patients had developed invasive breast cancer, 1.66 times more than the 34.9 predicted by the Gail model (95% CI, 1.29 to 2.15; P < .001). For individual women, the concordance between predicted and observed outcomes was low, with a concordance statistic of 0.50 (95% CI, 0.44 to 0.55). CONCLUSION The Gail model significantly underestimates the risk of breast cancer in women with atypia. Its ability to discriminate women with atypia into those who did and did not develop breast cancer is limited. Health care professionals should be cautious when using the Gail model to counsel individual patients with atypia.
Collapse
Affiliation(s)
- V Shane Pankratz
- Division of Biostatistics, Medical Oncology, General Surgery, Internal Medicine, Epidemiology, and Anatomic Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Jacobi CE, de Bock GH, Siegerink B, van Asperen CJ. Differences and similarities in breast cancer risk assessment models in clinical practice: which model to choose? Breast Cancer Res Treat 2008; 28:3591-6. [PMID: 18516672 DOI: 10.1200/jco.2010.28.0784] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To show differences and similarities between risk estimation models for breast cancer in healthy women from BRCA1/2-negative or untested families. After a systematic literature search seven models were selected: Gail-2, Claus Model, Claus Tables, BOADICEA, Jonker Model, Claus-Extended Formula, and Tyrer-Cuzick. Life-time risks (LTRs) for developing breast cancer were estimated for two healthy counsellees, aged 40, with a variety in family histories and personal risk factors. Comparisons were made with guideline thresholds for individual screening. Without a clinically significant family history LTRs varied from 6.7% (Gail-2 Model) to 12.8% (Tyrer-Cuzick Model). Adding more information on personal risk factors increased the LTRs and yearly mammography will be advised in most situations. Older models (i.e. Gail-2 and Claus) are likely to underestimate the LTR for developing breast cancer as their baseline risk for women is too low. When models include personal risk factors, surveillance thresholds have to be reformulated. For current clinical practice, the Tyrer-Cuzick Model and the BOADICEA Model seem good choices.
Collapse
Affiliation(s)
- Catharina E Jacobi
- Department of Medical Decision Making, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|