1
|
Sidhu S, Kwatra KS, Kinsley PA. Androgen Receptor Expression in ER and PR Negative Breast Cancer-A Study from a Tertiary Hospital in Northern India. South Asian J Cancer 2023; 12:319-325. [PMID: 38130277 PMCID: PMC10733066 DOI: 10.1055/s-0043-1768920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Sumeet SidhuObjectives Breast cancer is the leading cause of cancer-related deaths in women. Estrogen (ER) and progesterone receptor (PR) status and Her2 overexpression are major determinants in therapeutic decision making. Triple-negative breast cancers (TNBCs) have limited treatment options. Androgen receptor (AR) expression opens up therapeutic avenues for these patients. The aim of this article was to study the immunohistochemical expression of ARs in ER and PR Negative breast carcinomas and to correlate AR expression with various clinical, histopathological, and other immunohistochemical parameters. Materials and Methods It is a cross-sectional study including 105 ER and PR Negative cases of breast carcinoma. Clinical parameters, histopathology, and immunohistochemical expression of AR, Her2, and Ki67 were analyzed in all cases. Results AR expression was observed in 63.8% of ER and PR Negative breast cancers. In this group, AR expression was strongly associated with Her2 co-expression (89.2%) as compared to TNBCs (45.8%); p -value = 0.0002. Significant correlation was also observed between AR expression and tumor necrosis ( p -value = 0.034) and postmenopausal status ( p = 0.007). Conclusion Our study shows that significant proportion of ER and PR Negative breast carcinomas (ER- PR- Her2+ and TNBCs) show AR expression. We strongly recommend routine evaluation of all hormone receptor-negative breast carcinomas for AR status by immunohistochemistry.
Collapse
Affiliation(s)
- Sumeet Sidhu
- Christian Medical College and Hospital Ludhiana, Ludhiana, Punjab, India
| | | | | |
Collapse
|
2
|
Li L, Zheng S, Chen M, Chi W, Xue J, Wu J. The Prognostic Values of Androgen Receptor in Breast Cancer. Arch Pathol Lab Med 2023; 147:1075-1085. [PMID: 36508355 DOI: 10.5858/arpa.2021-0590-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 09/01/2023]
Abstract
CONTEXT.— Whether androgen receptor (AR) expression can predict prognosis in breast cancer is under debate. OBJECTIVE.— To analyze, retrospectively, the prognostic and treatment-predictive ability of AR status in breast cancer. DESIGN.— A total of 5765 patients diagnosed with primary invasive breast cancer without distant metastasis in the adjuvant setting were analyzed. The propensity score-matching method was used to develop a new cohort of 3978 patients (1989 patients each) in which important prognostic factors were balanced. RESULTS.— Positive AR expression is an independent prognostic factor for disease-free survival and overall survival. Estrogen receptor (ER)+ and progesterone receptor (PR)+ AR+ breast cancer patients had the longest survival, whereas ER-PR-AR- breast cancer patients had the shortest survival. The ER/PR/AR combinations could not predict the treatment effects for adjuvant trastuzumab but could be used for adjuvant chemotherapy and endocrine therapy selection. The worst survival was found in ER+PR-AR- patients receiving toremifene, ER+PR-AR+ patients receiving exemestane, ER+PR+AR- patients receiving anthracycline, and ER-PR-AR+ patients receiving taxanes. ER+PR-AR-, ER-PR-AR+, and ER-PR-AR- patients were associated with the worst survival among those who received radiotherapy and anthracycline plus taxanes. CONCLUSIONS.— AR in combination with ER and PR could predict the prognosis and treatment effects of chemotherapy, endocrine therapy, and radiotherapy in the adjuvant setting.
Collapse
Affiliation(s)
- Lun Li
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China (Li)
| | - Shuyue Zheng
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Ming Chen
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Weiru Chi
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Jingyan Xue
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
| | - Jiong Wu
- From the Department of Breast Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China (Li, Zheng, Chen, Chi, Xue, Wu)
- The Collaborative Innovation Center for Cancer Medicine, Shanghai, China (Wu)
| |
Collapse
|
3
|
Habiburrahman M, Sutopo S, Wardoyo MP. Role of DEK in carcinogenesis, diagnosis, prognosis, and therapeutic outcome of breast cancer: An evidence-based clinical review. Crit Rev Oncol Hematol 2023; 181:103897. [PMID: 36535490 DOI: 10.1016/j.critrevonc.2022.103897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is a significantly burdening women's cancer with limited diagnostic modalities. DEK is a novel biomarker overexpressed in breast cancers, currently exhaustively researched for its diagnosis and prognosis. Search for relevant meta-analyses, cohorts, and experimental studies in the last fifteen years was done in five large scientific databases. Non-English, non-full text articles or unrelated studies were excluded. Thirteen articles discussed the potential of DEK to estimate breast cancer characteristics, treatment outcomes, and prognosis. This proto-oncogene plays a role in breast carcinogenesis, increasing tumour proliferation and invasion, preventing apoptosis, and creating an immunodeficient tumour milieu with M2 tumour-associated macrophages. DEK is also associated with worse clinicopathological features and survival in breast cancer patients. Using a Kaplan-Meier plotter data analysis, DEK expression predicts worse overall survival (HR 1.24, 95%CI: 1.01-1.52, p = 0.039), comparable to other biomarkers. DEK is a promising novel biomarker requiring further research to determine its bedside applications.
Collapse
Affiliation(s)
- Muhammad Habiburrahman
- Faculty of Medicine Universitas Indonesia, Central Jakarta, DKI Jakarta, Indonesia; Dr. Cipto Mangunkusumo Hospital, Central Jakarta, DKI Jakarta, Indonesia.
| | - Stefanus Sutopo
- Faculty of Medicine Universitas Indonesia, Central Jakarta, DKI Jakarta, Indonesia
| | - Muhammad Prasetio Wardoyo
- Faculty of Medicine Universitas Indonesia, Central Jakarta, DKI Jakarta, Indonesia; Dr. Cipto Mangunkusumo Hospital, Central Jakarta, DKI Jakarta, Indonesia
| |
Collapse
|
4
|
Yang Y, Min A, Lee KH, Ryu HS, Kim TY, Woo GU, Suh KJ, Lee DW, Lee HB, Moon HG, Han W, Park IA, Noh DY, Im SA. Prognostic Role of Androgen Receptor Expression in Surgically Resected Early Breast Cancer Patients. J Breast Cancer 2020; 23:182-193. [PMID: 32395377 PMCID: PMC7192742 DOI: 10.4048/jbc.2020.23.e28] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/11/2020] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Endocrine therapy is a standard treatment for hormone receptor-positive breast cancer, which accounts for 60%-75% of all breast cancer. Hormone receptor positivity is a prognostic and predictive biomarker in breast cancer. Approximately 50%-80% of breast cancer is also positive for androgen receptor (AR), but the prognostic and predictive value of AR expression in breast cancer is controversial. Here, we investigated AR expression and its prognostic value in patients with surgically resected breast cancer in Korea. METHODS We retrospectively reviewed the medical records of patients who had surgically resected breast cancer to collect AR expression data and other clinicopathological data. The optimal cut-off for AR positivity was determined using a receiver operating characteristic curve analysis. RESULTS We reviewed 957 patients with surgically resected breast cancer from June 2012 to April 2013. The median follow-up was 62 months, and relapse events occurred in 101 (10.6%) patients. Unlike the cut-off value of 1% or 10% in previous reports, 35% was determined to be best for predicting relapse-free survival (RFS) in this study. At the cut-off value of 35%, 654 (68.4%) patients were AR-positive. AR expression was more prevalent in luminal A (87.6%) and luminal B (73.1%) types than in human epidermal growth factor receptor 2-positive (56.2%) or triple-negative (20.6%) types. AR expression of ≥ 35% was significantly related to longer RFS in a multivariate analysis (hazard ratio, 0.430; 95% confidence interval, 0.260-0.709; p = 0.001). CONCLUSION We propose a cut-off value of 35% to best predict RFS in patients with surgically resected breast cancer. AR expression was positive in 68.4% of patients, and AR positivity was found to be an independent prognostic factor for longer RFS.
Collapse
Affiliation(s)
- Yaewon Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Han Suk Ryu
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Go-un Woo
- Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Koung Jin Suh
- Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dae-Won Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Han-Byoel Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyeong-Gon Moon
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Wonshik Han
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - In Ae Park
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Young Noh
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
The estrogen receptor coactivator AIB1 is a new putative prognostic biomarker in ER-positive/HER2-negative invasive lobular carcinoma of the breast. Breast Cancer Res Treat 2019; 175:305-316. [PMID: 30796653 PMCID: PMC6533234 DOI: 10.1007/s10549-019-05138-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
Purpose According to the 2017 St Gallen surrogate definitions of the intrinsic subtypes, Ki67, progesterone receptor (PR) and Nottingham histological grade (NHG) are used for prognostic classification of estrogen receptor (ER) positive/HER2-negative breast cancer into luminal A- or luminal B-like. The aim of the present study was to investigate if additional biomarkers, related to endocrine signaling pathways, e.g., amplified in breast cancer 1 (AIB1), androgen receptor (AR), and G protein-coupled estrogen receptor (GPER), can provide complementary prognostic information in a subset of ER-positive/HER-negative invasive lobular carcinoma (ILC). Methods Biomarkers from 224 patients were analyzed immunohistochemically on tissue microarray. The primary endpoint was breast cancer mortality (BCM), analyzed with 10- and 25-year follow-up (FU). In addition, the prognostic value of gene expression data for these biomarkers was analyzed in three publicly available ILC datasets. Results AIB1 (high vs. low) was associated to BCM in multivariable analysis (adjusted for age, tumor size, nodal status, NHG, Ki67, luminal-like classification, and adjuvant systemic therapy) with 10-year FU (HR 6.8, 95% CI 2.3–20, P = 0.001) and 25-year FU (HR 3.0, 95% CI 1.1–7.8, P = 0.03). The evidence of a prognostic effect of AIB1 could be confirmed by linking gene expression data to outcome in independent publicly available ILC datasets. AR and GPER were neither associated to BCM with 10-year nor with 25-year FU (P > 0.33). Furthermore, Ki67 and NHG were prognostic for BCM at both 10-year and 25-year FU, whereas PR was not. Conclusions AIB1 is a new putative prognostic biomarker in ER-positive/HER2-negative ILC. Electronic supplementary material The online version of this article (10.1007/s10549-019-05138-7) contains supplementary material, which is available to authorized users.
Collapse
|
6
|
Feasibility of Classification of Triple Negative Breast Cancer by Immunohistochemical Surrogate Markers. Clin Breast Cancer 2018; 18:e1123-e1132. [DOI: 10.1016/j.clbc.2018.03.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/03/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023]
|
7
|
Min A, Jang H, Kim S, Lee KH, Kim DK, Suh KJ, Yang Y, Elvin P, O'Connor MJ, Im SA. Androgen Receptor Inhibitor Enhances the Antitumor Effect of PARP Inhibitor in Breast Cancer Cells by Modulating DNA Damage Response. Mol Cancer Ther 2018; 17:2507-2518. [PMID: 30232143 DOI: 10.1158/1535-7163.mct-18-0234] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/24/2018] [Accepted: 09/13/2018] [Indexed: 11/16/2022]
Abstract
The androgen receptor (AR) is expressed in 60%-70% of breast cancers regardless of estrogen receptor status, and has been proposed as a therapeutic target in breast cancers that retain AR. In this study, the authors aimed to investigate a new treatment strategy using a novel AR inhibitor AZD3514 in breast cancer. AZD3514 alone had a minimal antiproliferative effect on most breast cancer cell lines irrespective of AR expression level, but it downregulated the expressions of DNA damage response (DDR) molecules, including ATM and chk2, which resulted in the accumulation of damaged DNA in some breast cancer cells. Furthermore, AZD3514 enhanced cellular sensitivity to a PARP inhibitor olaparib by blocking the DDR pathway in breast cancer cells. Furthermore, the downregulation of NKX3.1 expression in MDA-MB-468 cells by AZD3514 occurred in parallel with the suppression of ATM-chk2 axis activation, and the suppression of NKX3.1 by AZD3514 was found to result from AZD3514-induced TOPORS upregulation and a resultant increase in NKX3.1 degradation. The study shows posttranslational regulation of NKX3.1 via TOPORS upregulation by AZD3514-induced ATM inactivation-increased olaparib sensitivity in AR-positive and TOPORS-expressing breast cancer cells, and suggests the antitumor effect of AZD3514/olaparib cotreatment is caused by compromised DDR activity in breast cancer cell lines and in a xenograft model. These results provide a rationale for future clinical trials of olaparib/AR inhibitor combination treatment in breast cancer.
Collapse
Affiliation(s)
- Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyemin Jang
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Seongyeong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | - Koung Jin Suh
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Translational Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul, Korea
| | - Yaewon Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Translational Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Chungbuk University Hospital, Cheong-Ju, Korea
| | - Paul Elvin
- Oncology IMED, AstraZeneca UK Ltd., Cambridge, United Kingdom
| | - Mark J O'Connor
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca UK Ltd., Cambridge, United Kingdom
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Liu X, Feng C, Liu J, Cao L, Xiang G, Liu F, Wang S, Jiao J, Niu Y. Androgen receptor and heat shock protein 27 co-regulate the malignant potential of molecular apocrine breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:90. [PMID: 29699584 PMCID: PMC5921986 DOI: 10.1186/s13046-018-0762-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/17/2018] [Indexed: 01/20/2023]
Abstract
Background The most striking feature of molecular apocrine breast cancer (MABC) is the expression of androgen receptor (AR). We report here the mechanism of the AR in regulating the behavior of MABC. Methods The MABC cell line, MDA-MB-453, and the nonMABC cell line, MCF7, were used in this study. The effect of dihydrotestosterone (DHT) and heat shock protein 27 (HSP27) on cell proliferation was quantified using the cell counter kit-8 (CCK8) and clonogenic assays in vitro and by a xenograft tumor model in vivo. The expression of the AR and HSP27 was analyzed using western blot, qPCR, and immunofluorescence assays. Complexes of the AR and HSP27 were detected by co-immunoprecipitation (Co-IP). Results In MDA-MB-453 cells, DHT promoted cell proliferation and stimulated AR and HSP27 translocation from the cytoplasm to the nucleus, whereas, it inhibited MCF7 cell growth, and only the AR translocated into the nucleus. HSP27 knock-down decreased the proliferative ability of MDA-MB-453 cells, which could be rescued by DHT, while HSP27 and DHT had synergistic effects on MCF7 cells. HSP27 phosphorylation was a prerequisite for AR translocation into the nucleus, especially phosphorylation on serine 82. In addition, DHT stimulated the tumorigenic and metastatic capacities of MDA-MB-453 cells, while HSP27 knock-down decreased the rate of tumor formation and induced apoptosis in cells. Conclusions The results suggest that HSP27 assists the AR in regulating the malignant behavior of MABC, and these findings might be helpful in the treatment of MABC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0762-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Changyun Feng
- Department of Maternal and Child Health Hospital of Linyi, Qinghe-South Road, Luozhuang District, Linyi, 276016, China
| | - Junjun Liu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Lu Cao
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Guomin Xiang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Fang Liu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Shuling Wang
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Jiao Jiao
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China
| | - Yun Niu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, China.
| |
Collapse
|
9
|
Ricciardelli C, Bianco-Miotto T, Jindal S, Butler LM, Leung S, McNeil CM, O'Toole SA, Ebrahimie E, Millar EKA, Sakko AJ, Ruiz AI, Vowler SL, Huntsman DG, Birrell SN, Sutherland RL, Palmieri C, Hickey TE, Tilley WD. The Magnitude of Androgen Receptor Positivity in Breast Cancer Is Critical for Reliable Prediction of Disease Outcome. Clin Cancer Res 2018. [PMID: 29514843 DOI: 10.1158/1078-0432.ccr-17-1199] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Consensus is lacking regarding the androgen receptor (AR) as a prognostic marker in breast cancer. The objectives of this study were to comprehensively review the literature on AR prognostication and determine optimal criteria for AR as an independent predictor of breast cancer survival.Experimental Design: AR positivity was assessed by immunostaining in two clinically validated primary breast cancer cohorts [training cohort, n = 219; validation cohort, n = 418; 77% and 79% estrogen receptor alpha (ERα) positive, respectively]. The optimal AR cut-point was determined by ROC analysis in the training cohort and applied to both cohorts.Results: AR was an independent prognostic marker of breast cancer outcome in 22 of 46 (48%) previous studies that performed multivariate analyses. Most studies used cut-points of 1% or 10% nuclear positivity. Herein, neither 1% nor 10% cut-points were robustly prognostic. ROC analysis revealed that a higher AR cut-point (78% positivity) provided optimal sensitivity and specificity to predict breast cancer survival in the training (HR, 0.41; P = 0.015) and validation (HR, 0.50; P = 0.014) cohorts. Tenfold cross-validation confirmed the robustness of this AR cut-point. Patients with ERα-positive tumors and AR positivity ≥78% had the best survival in both cohorts (P < 0.0001). Among the combined ERα-positive cases, those with comparable or higher levels of AR (AR:ERα-positivity ratio >0.87) had the best outcomes (P < 0.0001).Conclusions: This study defines an optimal AR cut-point to reliably predict breast cancer survival. Testing this cut-point in prospective cohorts is warranted for implementation of AR as a prognostic factor in the clinical management of breast cancer. Clin Cancer Res; 24(10); 2328-41. ©2018 AACR.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Tina Bianco-Miotto
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,School of Agriculture, Food and Wine, University of Adelaide, Adelaide, South Australia, Australia
| | - Shalini Jindal
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Lisa M Butler
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catriona M McNeil
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Sandra A O'Toole
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Esmaeil Ebrahimie
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Ewan K A Millar
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Andrew J Sakko
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Alexandra I Ruiz
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah L Vowler
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, United Kingdom
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Stephen N Birrell
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert L Sutherland
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Carlo Palmieri
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.,Academic Department of Medical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
10
|
Wielsøe M, Bjerregaard-Olesen C, Kern P, Bonefeld-Jørgensen EC. Receptor activities of persistent pollutant serum mixtures and breast cancer risk. Endocr Relat Cancer 2018; 25:201-215. [PMID: 29237712 DOI: 10.1530/erc-17-0366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 11/08/2022]
Abstract
Studies on associations between persistent organic pollutants (POPs) and breast cancer risk are inconclusive. The majority of studies have evaluated the effect of single compounds, without considering multiple exposures to and interactions between different POPs. The present study aimed at evaluating breast cancer risk related to combined effects of serum POP mixtures on cellular receptor functions. Data on breast cancer cases (n = 77) and controls (n = 84) were collected among Greenlandic Inuit women. Serum mixtures of lipophilic POPs (lipPOPs), perfluoroalkyl acids (PFAAs) and dioxin-like POPs were extracted. The effect of the mixture extracts on the estrogen receptor (ER), androgen receptor (AR) and aryl hydrocarbon receptor (AhR) was determined using cell culture reporter gene assays. The serum mixtures were analyzed alone and upon co-exposure with natural receptor ligands to determine agonistic and antagonistic/competitive activity. We found that the frequency of lipPOP mixtures eliciting no, decreasing, or agonizing xenoandrogenic effect differed by breast cancer status. Using lipPOP mixtures with no effect on AR as reference, the mixtures with decreasing effects reduced breast cancer risk (OR: 0.30 (0.12; 0.76)). The AhR-toxic equivalent of serum mixtures was significantly lower in cases than in controls, and a reduced breast cancer risk was found when comparing the third tertile to the first (OR: 0.34 (0.14; 0.83)). We found no association between the xenoestrogenic activities of lipPOPs or PFAAs and breast cancer risk. Serum lipPOP mixtures are hormone disruptive and may influence breast cancer risk, whereas PFAAs seem to influence breast cancer risk through other pathways.
Collapse
Affiliation(s)
- Maria Wielsøe
- Centre for Arctic Health & Molecular EpidemiologyDepartment of Public Health, Aarhus University, Aarhus, Denmark
| | - Christian Bjerregaard-Olesen
- Centre for Arctic Health & Molecular EpidemiologyDepartment of Public Health, Aarhus University, Aarhus, Denmark
| | - Peder Kern
- Department of Gynecology and ObstetricsDronning Ingrid's Hospital, Nuuk, Greenland
| | - Eva Cecilie Bonefeld-Jørgensen
- Centre for Arctic Health & Molecular EpidemiologyDepartment of Public Health, Aarhus University, Aarhus, Denmark
- Institute for Nursing and Health ScienceUniversity of Greenland, Nuuk, Greenland
| |
Collapse
|
11
|
Liu X, Yang Y, Feng X, Shen H, Liu J, Liu X, Niu Y. Early versus late distant metastasis and adjuvant chemotherapy alone versus both radiotherapy and chemotherapy in molecular apocrine breast cancer. Oncotarget 2018; 7:48905-48917. [PMID: 27340922 PMCID: PMC5226479 DOI: 10.18632/oncotarget.10211] [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: 05/02/2016] [Accepted: 06/03/2016] [Indexed: 11/25/2022] Open
Abstract
As a new subtype of breast cancer, molecular apocrine breast cancer (MABC) is estrogen receptor (ER) and progesterone receptor (PR) negative expression, but androgen receptor (AR) positive expression. The prognostic significance and clinical biological behavior of MABC have remained unclear up to now. This study aimed to analysis the distant metastasis behavior and response to adjuvant radiotherapy and chemotherapy of MABC subgroup. The report showed that there were significant differences between early and late distant metastasizing tumors with respect to Ki67, epidermal growth factor receptor 2 (HER2) and vascular endothelial growth factor (VEGF) expressions by a retrospective analysis consisting of 410 invasive breast cancer patients, which included 205 MABC and 205 nonMABC cases. MABC subgroup metastasized earlier than nonMABC subgroup, and MABC showed a tendency for a higher metastasis rate in lung, liver and brain, but lower in bone. HER2-positive or VEGF-positive tumors were more inclined to develop bone metastasis within MABC subgroup. The survival rate was superior for patients undergone both adjuvant radiotherapy and chemotherapy than those undergone chemotherapy alone in nonMABC subgroup, but there was no significant difference in MABC subgroup. Our data suggested that MABC subgroup seemed to develop distant metastasis earlier than nonMABC subgroup, and patients with MABC indicated poor prognosis. This study might also provide a foundation for helping patients receive reasonable treatments according to molecular subtype.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yang Yang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaolong Feng
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Honghong Shen
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jian Liu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xia Liu
- Department of Oncology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yun Niu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
12
|
Dai X, Hua T, Hong T. Integrated diagnostic network construction reveals a 4-gene panel and 5 cancer hallmarks driving breast cancer heterogeneity. Sci Rep 2017; 7:6827. [PMID: 28754978 PMCID: PMC5533795 DOI: 10.1038/s41598-017-07189-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/23/2017] [Indexed: 12/26/2022] Open
Abstract
Breast cancer encompasses a group of heterogeneous diseases, each associated with distinct clinical implications. Dozens of molecular biomarkers capable of categorizing tumors into clinically relevant subgroups have been proposed which, though considerably contribute in precision medicine, complicate our understandings toward breast cancer subtyping and its clinical translation. To decipher the networking of markers with diagnostic roles on breast carcinomas, we constructed the diagnostic networks by incorporating 6 publically available gene expression datasets with protein interaction data retrieved from BioGRID on previously identified 1015 genes with breast cancer subtyping roles. The Greedy algorithm and mutual information were used to construct the integrated diagnostic network, resulting in 37 genes enclosing 43 interactions. Four genes, FAM134B, KIF2C, ALCAM, KIF1A, were identified having comparable subtyping efficacies with the initial 1015 genes evaluated by hierarchical clustering and cross validations that deploy support vector machine and k nearest neighbor algorithms. Pathway, Gene Ontology, and proliferation marker enrichment analyses collectively suggest 5 primary cancer hallmarks driving breast cancer differentiation, with those contributing to uncontrolled proliferation being the most prominent. Our results propose a 37-gene integrated diagnostic network implicating 5 cancer hallmarks that drives breast cancer heterogeneity and, in particular, a 4-gene panel with clinical diagnostic translation potential.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.
| | - Tongyan Hua
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Tingting Hong
- Department of medical oncology, the affiliated hospital of Jiangnan University, the fourth people's hospital of Wuxi, Wuxi, China
| |
Collapse
|
13
|
Pure Apocrine Carcinomas Represent a Clinicopathologically Distinct Androgen Receptor-Positive Subset of Triple-Negative Breast Cancers. Am J Surg Pathol 2017; 40:1109-16. [PMID: 27259012 DOI: 10.1097/pas.0000000000000671] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Apocrine carcinomas comprise ∼1% of all breast cancers and are characterized by large cells bearing abundant eosinophilic granular cytoplasm, round nuclei, and prominent nucleoli. They are typically estrogen receptor/progesterone receptor/HER2 negative, making them unresponsive to typical hormonal or HER2-based chemotherapy. However, this subtype of triple-negative breast cancers expresses androgen receptor (AR), a feature not shared by most nonapocrine triple-negative cancers (NA-TNCs). AR therefore represents a potential diagnostic tool and therapeutic target for apocrine breast carcinoma. All pure apocrine carcinomas diagnosed during a 10-year period were reviewed, and clinicopathologic characteristics were compared with a control group of 26 NA-TNC cases. Twenty apocrine carcinomas were identified (∼0.8% of all breast cancers). The mean age at diagnosis was 69.3 years for apocrine carcinomas and 56.7 years for NA-TNC. All apocrine carcinomas and no NA-TNC were AR positive. The proportions of apocrine carcinoma grades varied, with G1 being seen in 15% of patients, G2 in 55%, and G3 in 30%. In contrast, 100% of NA-TNC cases were G3. The majority of apocrine carcinomas presented at low T stage (T1: 70%; T2: 20%; T3: 10%; T4: 0%), whereas NA-TNC cases more often presented at T2 or higher (T1: 46.2%; T2: 30.8%; T3: 11.5%; T4: 11.5%). Thirty percent of apocrine carcinomas and 30.8% of NA-TNCs had nodal metastases at presentation. Apocrine carcinomas had a favorable clinical prognosis, with 80% of patients showing no evidence of disease-related morbidity or mortality (mean follow-up: 45.2 mo). Pure apocrine carcinomas represent a clinicopathologically distinct subgroup of triple-negative breast cancer characterized by AR positivity. When compared with NA-TNC, apocrine carcinomas more often present in older women with lower grade and T stage, a group in which a more conservative treatment regimen is often desired.
Collapse
|
14
|
Batistatou A, Kotoula V, Bobos M, Kouvatseas G, Zagouri F, Tsolaki E, Gogas H, Koutras A, Pentheroudakis G, Timotheadou E, Pervana S, Goussia A, Petraki K, Sotiropoulou M, Koletsa T, Razis E, Kosmidis P, Aravantinos G, Papadimitriou C, Pectasides D, Fountzilas G. Correlation of MYC Gene and Protein Status With Breast Cancer Subtypes and Outcome of Patients Treated With Anthracycline-Based Adjuvant Chemotherapy. Pooled Analysis of 2 Hellenic Cooperative Group Phase III Trials. Clin Breast Cancer 2017; 18:53-62.e3. [PMID: 28870680 DOI: 10.1016/j.clbc.2017.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/29/2017] [Accepted: 07/07/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The prognostic/predictive value of aberrant MYC gene copies and protein expression is not clear in breast cancer. PATIENTS AND METHODS Early breast cancer patients were treated with anthracycline-containing chemotherapy within 2 randomized adjuvant trials. MYC gene and centromere-8 status, as well as Myc protein expression were investigated on 1060 paraffin tumors with fluorescence in situ hybridization and immunohistochemistry, respectively. RESULTS MYC amplification was present in 45% and polysomy-8 in 23% of the tumors. Cytoplasmic staining was observed in 60% and nuclear staining in 26% of the tumors, strongly correlating with each other but not with MYC gene status. MYC gene amplification in the absence of polysomy-8 was associated with adverse disease-free survival (DFS) and overall survival (OS), and remained as an independent unfavorable prognostic factor in multivariate analysis (Wald P = .022 for DFS; P = .032 for OS), whereas patients with MYC amplification and polysomy-8, with polysomy-8 only, and with normal MYC without polysomy-8 performed significantly better compared with those with MYC gene amplification only. Nuclear Myc protein expression benefitted patients treated with paclitaxel (interaction P = .052 for DFS; P = .049 for OS). This interaction remained independently significant in multivariate analysis for OS (overall P = .028). CONCLUSION The effect of MYC gene status on breast cancer patient outcome seems to depend on the underlying chromosomal instability and appears unfavorable for tumors with MYC amplification without polysomy. Nuclear Myc protein expression seems predictive for benefit from adjuvant paclitaxel. These data might aid in the interpretation of relevant findings from large clinical trials.
Collapse
Affiliation(s)
- Anna Batistatou
- Department of Pathology, Ioannina University Hospital, Ioannina, Greece.
| | - Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece; Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Mattheos Bobos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Eleftheria Tsolaki
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Helen Gogas
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | | | - Eleni Timotheadou
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, Greece
| | - Stavroula Pervana
- Department of Pathology, Papageorgiou Hospital, Thessaloniki, Greece
| | - Anna Goussia
- Department of Pathology, Ioannina University Hospital, Ioannina, Greece
| | | | | | | | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Paris Kosmidis
- Second Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Gerasimos Aravantinos
- Second Department of Medical Oncology, Agii Anargiri Cancer Hospital, Athens, Greece
| | - Christos Papadimitriou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, Greece; Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Severson TM, Zwart W. A review of estrogen receptor/androgen receptor genomics in male breast cancer. Endocr Relat Cancer 2017; 24:R27-R34. [PMID: 28062545 DOI: 10.1530/erc-16-0225] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/06/2017] [Indexed: 01/17/2023]
Abstract
Male breast cancer is a rare disease, of which little is known. In contrast to female breast cancer, the very vast majority of all cases are positive for estrogen receptor alpha (ERα), implicating the function of this steroid hormone receptor in tumor development and progression. Consequently, adjuvant treatment of male breast cancer revolves around inhibition of ERα. In addition, the androgen receptor (AR) gradually receives more attention as a relevant novel target in breast cancer treatment. Importantly, the rationale of treatment decision making is strongly based on parallels with female breast cancer. Yet, prognostic indicators are not necessarily the same in breast cancer between both genders, complicating translatability of knowledge developed in female breast cancer toward male patients. Even though ERα and AR are expressed both in female and male disease, are the genomic functions of both steroid hormone receptors conserved between genders? Recent studies have reported on mutational and epigenetic similarities and differences between male and female breast cancer, further suggesting that some features are strongly conserved between the two diseases, whereas others are not. This review critically discusses the recent developments in the study of male breast cancer in relation to ERα and AR action and highlights the potential future studies to further elucidate the genomic regulation of this rare disease.
Collapse
Affiliation(s)
- Tesa M Severson
- Division of Molecular Pathologythe Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Molecular Carcinogenesisthe Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wilbert Zwart
- Division of Molecular Pathologythe Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
16
|
Tang P, Tse GM. Immunohistochemical Surrogates for Molecular Classification of Breast Carcinoma: A 2015 Update. Arch Pathol Lab Med 2017; 140:806-14. [PMID: 27472239 DOI: 10.5858/arpa.2015-0133-ra] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT -The pioneering works on molecular classification (MC) by Perou and Sorlie et al in the early 2000s using global gene expression profiling identified 5 intrinsic subtypes of invasive breast cancers (IBCs): luminal A, luminal B, normal breast-like, HER2-enriched, and basal-like subtypes, each unique in incidence, survival, and response to therapy. Because the application of gene expression profiling in daily practice is not economical or practical at the present time, many investigators have studied the use of immunohistochemical (IHC) surrogates as a substitute for determining the MC of IBC. OBJECTIVE -To discuss the continuing efforts that have been made to develop clinically significant and readily available IHC surrogates for the MC of IBC. DATA SOURCES -Data were obtained from pertinent peer-reviewed English-language literature. CONCLUSIONS -The most commonly used IHC surrogates are estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), dividing IBC into luminal, HER2, and triple-negative subtypes. The addition of Ki-67, cytokeratin 5, and epidermal growth factor receptor (EGFR) separates luminal B from luminal A subtypes, and basal-like subtype from triple-negative breast cancer. More recently, biomarkers such as androgen receptor and p53 have been shown to further stratify these molecular subtypes. Although many studies of IHC-based MC have shown clinical significance similar to gene expression profiling-defined MC, its critical limitations are: (1) a lack of standardization in terminology, (2) a lack of standardization in biomarkers used for each subtype, and (3) the lack of a uniform cutoff for each biomarker. A panel of IHC surrogates for each subtype of IBC is proposed.
Collapse
Affiliation(s)
| | - Gary M Tse
- From the Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; and the Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
17
|
Abstract
CONTEXT -Molecular diagnostics play a role in the management of many cancers, including breast cancer. OBJECTIVE -To provide an update on molecular testing in current clinical practice, targeted at practicing pathologists who are not breast cancer specialists. DATA SOURCES -This study is a narrative literature review. CONCLUSIONS -In addition to routine hormone (estrogen and progesterone) receptor testing, new and recurrent tumors are tested for HER2 amplification by in situ hybridization or overexpression by immunohistochemistry. Intrinsic subtyping of tumors represents a fundamental advance in our understanding of breast cancer biology, but currently it has an indirect role in patient management. Clinical next-generation sequencing (tumor profiling) is increasingly used to identify potentially actionable mutations in tumor tissue. Multianalyte assays with algorithmic analysis, including MammaPrint, Oncotype DX, and Prosigna, play a larger role in breast cancer than in many other malignancies. Given that a proportion of breast cancers are familial, testing of nontumor tissue for cancer predisposition mutations also plays a role in breast cancer care.
Collapse
Affiliation(s)
- Ian S Hagemann
- From the Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri. Presented at the 2nd Princeton Integrated Pathology Symposium: Breast Pathology; February 8, 2015; Plainsboro, New Jersey
| |
Collapse
|
18
|
Dai X, Xiang L, Li T, Bai Z. Cancer Hallmarks, Biomarkers and Breast Cancer Molecular Subtypes. J Cancer 2016; 7:1281-94. [PMID: 27390604 PMCID: PMC4934037 DOI: 10.7150/jca.13141] [Citation(s) in RCA: 275] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 05/19/2016] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a complex disease encompassing multiple tumor entities, each characterized by distinct morphology, behavior and clinical implications. Besides estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, novel biomarkers have shown their prognostic and predictive values, complicating our understanding towards to the heterogeneity of such cancers. Ten cancer hallmarks have been proposed by Weinberg to characterize cancer and its carcinogenesis. By reviewing biomarkers and breast cancer molecular subtypes, we propose that the divergent outcome observed from patients stratified by hormone status are driven by different cancer hallmarks. 'Sustaining proliferative signaling' further differentiates cancers with positive hormone receptors. 'Activating invasion and metastasis' and 'evading immune destruction' drive the differentiation of triple negative breast cancers. 'Resisting cell death', 'genome instability and mutation' and 'deregulating cellular energetics' refine breast cancer classification with their predictive values. 'Evading growth suppressors', 'enabling replicative immortality', 'inducing angiogenesis' and 'tumor-promoting inflammation' have not been involved in breast cancer classification which need more focus in the future biomarker-related research. This review novels in its global view on breast cancer heterogeneity, which clarifies many confusions in this field and contributes to precision medicine.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Engineering Laboratory for Cereal Fermentation Technology, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, P.R.China
| | - Liangjian Xiang
- National Engineering Laboratory for Cereal Fermentation Technology, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, P.R.China
| | - Ting Li
- National Engineering Laboratory for Cereal Fermentation Technology, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, P.R.China
| | - Zhonghu Bai
- National Engineering Laboratory for Cereal Fermentation Technology, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, P.R.China
| |
Collapse
|
19
|
Hilborn E, Gacic J, Fornander T, Nordenskjöld B, Stål O, Jansson A. Androgen receptor expression predicts beneficial tamoxifen response in oestrogen receptor-α-negative breast cancer. Br J Cancer 2016; 114:248-55. [PMID: 26742006 PMCID: PMC4742586 DOI: 10.1038/bjc.2015.464] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/09/2015] [Accepted: 11/26/2015] [Indexed: 12/29/2022] Open
Abstract
Background: Although the androgen receptor (AR) is frequently expressed in breast cancer, its relevance in the disease is not fully understood. In addition, the relevance of AR in determining tamoxifen treatment efficiency requires evaluation. Purpose: To investigate the tamoxifen predictive relevance of the AR protein expression in breast cancer. Methods Patients were randomised to tamoxifen 40 mg daily for 2 or 5 years or to no endocrine treatment. Mean follow-up was 15 years. Hazard ratios were calculated with recurrence-free survival as end point. Results: In patients with oestrogen receptor (ER)-negative tumours, expression of AR predicted decreased recurrence rate with tamoxifen (hazard ratio (HR)=0.34; 95% confidence interval (CI)=0.14–0.81; P=0.015), whereas the opposite was seen in the AR− group (HR=2.92; 95% CI=1.16–7.31; P=0.022). Interaction test was significant P<0.001. Patients with triple-negative and AR+ tumours benefitted from tamoxifen treatment (HR=0.12; 95% CI=0.014–0.95 P=0.044), whereas patients with AR− tumours had worse outcome when treated with tamoxifen (HR=3.98; 95% CI=1.32–12.03; P=0.014). Interaction test was significant P=0.003. Patients with ER+ tumours showed benefit from tamoxifen treatment regardless of AR expression. Conclusions: AR can predict tamoxifen treatment benefit in patients with ER− tumours and triple-negative breast cancer.
Collapse
Affiliation(s)
- Erik Hilborn
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Jelena Gacic
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Bo Nordenskjöld
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| | - Olle Stål
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden.,Department of Oncology, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Agneta Jansson
- Division of Clinical sciences, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping S-581 85, Sweden
| |
Collapse
|
20
|
Heat shock protein 27 and gross cystic disease fluid protein 15 play critical roles in molecular apocrine breast cancer. Tumour Biol 2015; 37:8027-36. [PMID: 26711786 DOI: 10.1007/s13277-015-4712-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/20/2015] [Indexed: 10/22/2022] Open
Abstract
Molecular apocrine breast cancer (MABC) has a distinct hormonal profile, being estrogen receptor (ER) and progesterone receptor (PR) negative but androgen receptor (AR) positive. The clinical significance of MABC and its relative variables have not been absolutely clarified and remain to be determined. Five hundred cases of invasive breast carcinoma were randomly selected in this study, including 158 MABC cases and 342 nonMABC cases. Expression of ER, PR, epidermal growth factor receptor 2 (HER2), Ki67, AR, gross cystic disease fluid protein 15 (GCDFP15), and heat shock protein 27 (HSP27) were analyzed by immunohistochemistry. Differences of continuous variables between MABC and nonMABC subgroups were evaluated by the chi-square test. The Kaplan-Meier method was performed to evaluate disease-free survival (DFS) and overall survival (OS). The MABC subgroup had higher histological grade, bigger tumor size, more lymph node metastasis, and higher pTNM stage than the nonMABC subgroup (P < 0.05), and patients with MABC had poorer prognosis than those of the nonMABC subgroup (P < 0.05). Both GCDFP15 and HSP27 were expressed differently in the MABC and nonMABC subgroups (P < 0.05). Furthermore, in the MABC subgroup, positive HSP27 expression indicated higher risk of recurrence (P < 0.05) and positive GCDFP15 expression was also a poor marker for patient outcome (P < 0.05). MABC patients with HSP27 and GCDFP15 co-expression had worse outcome (P < 0.05). Our data suggested that MABC had a high risk of recurrence. Positive expression of both GCDFP15 and HSP27 were correlated with MABC malignancy. Targeting AR and HSP27 at the same time might offer a useful strategy to MABC.
Collapse
|
21
|
Papaxoinis G, Kotoula V, Alexopoulou Z, Kalogeras KT, Zagouri F, Timotheadou E, Gogas H, Pentheroudakis G, Christodoulou C, Koutras A, Bafaloukos D, Aravantinos G, Papakostas P, Charalambous E, Papadopoulou K, Varthalitis I, Efstratiou I, Zaramboukas T, Patsea H, Scopa CD, Skondra M, Kosmidis P, Pectasides D, Fountzilas G. Significance of PIK3CA Mutations in Patients with Early Breast Cancer Treated with Adjuvant Chemotherapy: A Hellenic Cooperative Oncology Group (HeCOG) Study. PLoS One 2015; 10:e0140293. [PMID: 26452060 PMCID: PMC4599795 DOI: 10.1371/journal.pone.0140293] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/23/2015] [Indexed: 11/28/2022] Open
Abstract
Background The PI3K-AKT pathway is frequently activated in breast cancer. PIK3CA mutations are most frequently found in the helical (exon 9) and kinase (exon 20) domains of this protein. The aim of the present study was to examine the role of different types of PIK3CA mutations in combination with molecular biomarkers related to PI3K-AKT signaling in patients with early breast cancer. Methods Tumor tissue samples from 1008 early breast cancer patients treated with adjuvant chemotherapy in two similar randomized trials of HeCOG were examined. Tumors were subtyped with immunohistochemistry (IHC) and FISH for ER, PgR, Ki67, HER2 and androgen receptor (AR). PIK3CA mutations were analyzed by Sanger sequencing (exon 20) and qPCR (exon 9) (Sanger/qPCR mutations). In 610 cases, next generation sequencing (NGS) PIK3CA mutation data were also available. PIK3CA mutations and PTEN protein expression (IHC) were analyzed in luminal tumors (ER and/or PgR positive), molecular apocrine carcinomas (MAC; ER/PgR negative / AR positive) and hormone receptor (ER/PgR/AR) negative tumors. Results PIK3CA mutations were detected in 235/1008 tumors (23%) with Sanger/qPCR and in 149/610 tumors (24%) with NGS. Concordance between the two methods was good with a Kappa coefficient of 0.76 (95% CI 0.69–0.82). Lobular histology, low tumor grade and luminal A tumors were associated with helical domain mutations (PIK3CAhel), while luminal B with kinase domain mutations (PIK3CAkin). The overall incidence of PIK3CA mutations was higher in luminal as compared to MAC and hormone receptor negative tumors (p = 0.004). Disease-free and overall survival did not significantly differ with respect to PIK3CA mutation presence and type. However, a statistically significant interaction between PIK3CA mutation status and PTEN low protein expression with regard to prognosis was identified. Conclusions The present study did not show any prognostic significance of specific PIK3CA mutations in a large group of predominantly lymph-node positive breast cancer women treated with adjuvant chemotherapy. Further analyses in larger cohorts are warranted to investigate possible differential effect of distinct PIK3CA mutations in small subgroups of patients.
Collapse
Affiliation(s)
- George Papaxoinis
- Oncology Section, Second Department of Internal Medicine, “Hippokration” Hospital, Athens, Greece
- * E-mail:
| | - Vassiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| | - Zoi Alexopoulou
- Health Data Specialists Ltd, Dept of Biostatistics, Athens, Greece
| | - Konstantine T. Kalogeras
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
- Translational Research Section, Hellenic Cooperative Oncology Group, Data Office, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, “Alexandra” Hospital, University of Athens School of Medicine, Athens, Greece
| | - Eleni Timotheadou
- Department of Medical Oncology, “Papageorgiou” Hospital, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| | - Helen Gogas
- First Department of Medicine, “Laiko” General Hospital, University of Athens School of Medicine, Athens, Greece
| | | | | | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | | | - Gerasimos Aravantinos
- Second Department of Medical Oncology, “Agii Anargiri” Cancer Hospital, Athens, Greece
| | | | - Elpida Charalambous
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| | - Kyriaki Papadopoulou
- Department of Pathology, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| | | | | | - Thomas Zaramboukas
- Department of Pathology, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| | - Helen Patsea
- Department of Pathology, IASSO General Hospital, Athens, Greece
| | - Chrisoula D. Scopa
- Department of Pathology, University Hospital, University of Patras Medical School, Patras, Greece
| | - Maria Skondra
- Oncology Section, Second Department of Internal Medicine, “Hippokration” Hospital, Athens, Greece
| | - Paris Kosmidis
- Second Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, “Hippokration” Hospital, Athens, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Faculty of Medicine, Thessaloniki, Greece
| |
Collapse
|
22
|
Gromov P, Espinoza JA, Talman ML, Honma N, Kroman N, Wielenga VT, Moreira JMA, Gromova I. FABP7 and HMGCS2 are novel protein markers for apocrine differentiation categorizing apocrine carcinoma of the breast. PLoS One 2014; 9:e112024. [PMID: 25389781 PMCID: PMC4229141 DOI: 10.1371/journal.pone.0112024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/09/2014] [Indexed: 02/01/2023] Open
Abstract
Apocrine carcinoma of the breast is a distinctive malignancy with unique morphological and molecular features, generally characterized by being negative for estrogen and progesterone receptors, and thus not electable for endocrine therapy. Despite the fact that they are morphologically distinct from other breast lesions, no standard molecular criteria are currently available for their diagnosis. Using gel-based proteomics in combination with mass spectrometry and immunohistochemistry we have identified two novel markers, HMGCS2 and FABP7 that categorize the entire breast apocrine differentiation spectrum from benign metaplasia and cysts to invasive stages. Expression of HMGCS2 and FABP7 is strongly associated with apocrine differentiation; their expression is retained by most invasive apocrine carcinomas (IAC) showing positive immunoreactivity in 100% and 78% of apocrine carcinomas, respectively, as compared to non-apocrine tumors (16.7% and 6.8%). The nuclear localization of FABP7 in tumor cells was shown to be associated with more aggressive stages of apocrine carcinomas. In addition, when added to the panel of apocrine biomarkers previously reported by our group: 15-PGDH, HMGCR and ACSM1, together they provide a signature that may represent a golden molecular standard for defining the apocrine phenotype in the breast. Moreover, we show that combining HMGCS2 to the steroidal profile (HMGCS2+/Androgen Receptor (AR)+/Estrogen Receptor(ER)-/Progesteron Receptor (PR)- identifies IACs with a greater sensitivity (79%) as compared with the steroidal profile (AR+/ER-/PR-) alone (54%). We have also presented a detailed immunohistochemical analysis of breast apocrine lesions with a panel of antibodies against proteins which correspond to 10 genes selected from published transcriptomic signatures that currently characterize molecular apocrine subtype and shown that except for melanophilin that is overexpressed in benign apocrine lesions, these proteins were not specific for morphological apocrine differentiation in breast.
Collapse
Affiliation(s)
- Pavel Gromov
- Danish Cancer Society Research Center, Genome Integrity Unit, Copenhagen, Denmark
- * E-mail:
| | - Jaime A. Espinoza
- Department of Pathology, Center for Investigation in Translational Oncology (CITO), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maj-Lis Talman
- Department of Pathology, the Centre of Diagnostic Investigations, Copenhagen University Hospital, Copenhagen, Denmark
| | - Naoko Honma
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Niels Kroman
- Department of Breast Surgery, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vera Timmermans Wielenga
- Department of Pathology, the Centre of Diagnostic Investigations, Copenhagen University Hospital, Copenhagen, Denmark
| | - José M. A. Moreira
- Section of Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Gromova
- Danish Cancer Society Research Center, Genome Integrity Unit, Copenhagen, Denmark
| |
Collapse
|