1
|
Yang H, Wang W, Cheng Z, Zheng T, Cheng C, Cheng M, Wang Z. Radiomic Machine Learning in Invasive Ductal Breast Cancer: Prediction of Ki-67 Expression Level Based on Radiomics of DCE-MRI. Technol Cancer Res Treat 2024; 23:15330338241288751. [PMID: 39431304 PMCID: PMC11504335 DOI: 10.1177/15330338241288751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
PURPOSE Our study aimed to investigate the potential of radiomics with DCE-MRI for predicting Ki-67 expression in invasive ductal breast cancer. METHOD We conducted a retrospective study including 223 patients diagnosed with invasive ductal breast cancer. Radiomics features were extracted from DCE-MRI using 3D-Slicer software. Two Ki-67 expression cutoff values (20% and 29%) were examined. Patients were divided into training (70%) and test (30%) sets. The Elastic Net method selected relevant features, and five machine-learning models were established. Radiomics models were created from intratumoral, peritumoral, and combined regions. Performance was assessed using ROC curves, accuracy, sensitivity, and specificity. RESULT For a Ki-67 cutoff value of 20%, the combined model exhibited the highest performance, with area under the curve (AUC) values of 0.838 (95% confidence interval (CI): 0.774-0.897) for the training set and 0.863 (95% CI: 0.764-0.949) for the test set. The AUC values for the tumor model were 0.816 (95% CI: 0.745-0.880) and 0.830 (95% CI: 0.724-0.916), and for the peritumor model were 0.790 (95% CI: 0.711-0.857) and 0.808 (95% CI: 0.682-0.910). When the Ki-67 cutoff value was set at 29%, the combined model also demonstrated superior predictive ability in both training set (AUC: 0.796; 95% CI: 0.724-0.862) and the test set (AUC: 0.823; 95% CI: 0.723-0.911). The AUC values for the tumor model were 0.785 (95% CI: 0.708-0.861) and 0.784 (95% CI: 0.663-0.882), and for the peritumor model were 0.773 (95% CI: 0.690-0.844) and 0.729 (95% CI: 0.603-0.847). CONCLUSION Radiomics with DCE-MRI can predict Ki-67 expression in invasive ductal breast cancer. Integrating radiomics features from intratumoral and peritumoral regions yields a dependable prognostic model, facilitating pre-surgical detection and treatment decisions. This holds potential for commercial diagnostic tools.
Collapse
Affiliation(s)
- Huan Yang
- Department of Emergency, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Wenxi Wang
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhiyong Cheng
- Department of Education, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Cheng Cheng
- Department of Emergency, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Mengyu Cheng
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhanqiu Wang
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
2
|
Fan Y, Zhao D, Su J, Yuan W, Niu S, Guo W, Jiang W. Radiomic Signatures Based on Mammography and Magnetic Resonance Imaging as New Markers for Estimation of Ki-67 and HER-2 Status in Breast Cancer. J Comput Assist Tomogr 2023; 47:890-897. [PMID: 37948363 DOI: 10.1097/rct.0000000000001502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
OBJECTIVE The aim of the study is to investigate the values of intratumoral and peritumoral regions based on mammography and magnetic resonance imaging for the prediction of Ki-67 and human epidermal growth factor (HER-2) status in breast cancer (BC). METHODS Two hundred BC patients were consecutively enrolled between January 2017 and March 2021 and divided into training (n = 133) and validation (n = 67) groups. All the patients underwent breast mammography and magnetic resonance imaging screening. Features were derived from intratumoral and peritumoral regions of the tumor and selected using the least absolute shrinkage and selection operator regression to build radiomic signatures (RSs). Receiver operating characteristic curve analysis and the DeLong test were performed to assess and compare each RS. RESULTS For each modality, the combined RSs integrating features from intratumoral and peritumoral regions always showed better prediction performance for predicting Ki-67 and HER-2 status compared with the RSs derived from intratumoral or peritumoral regions separately. The multimodality and multiregional combined RSs achieved the best prediction performance for predicting the Ki-67 and HER-2 status with an area under the receiver operating characteristic curve of 0.888 and 0.868 in the training cohort and 0.800 and 0.848 in the validation cohort, respectively. CONCLUSIONS Peritumoral areas provide complementary information to intratumoral regions of BC. The developed multimodality and multiregional combined RSs have good potential for noninvasive evaluation of Ki-67 and HER-2 status in BC.
Collapse
Affiliation(s)
- Ying Fan
- From the School of Intelligent Medicine, China Medical University, Shenyang
| | - Dan Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning
| | - Juan Su
- From the School of Intelligent Medicine, China Medical University, Shenyang
| | - Wendi Yuan
- From the School of Intelligent Medicine, China Medical University, Shenyang
| | - Shuxian Niu
- From the School of Intelligent Medicine, China Medical University, Shenyang
| | - Wei Guo
- College of Computer Science, Shenyang Aerospace University, Shenyang
| | - Wenyan Jiang
- Department of Scientific Research and Academic, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Liaoning, People's Republic. China
| |
Collapse
|
3
|
Zhu Y, Dou Y, Qin L, Wang H, Wen Z. Prediction of Ki-67 of Invasive Ductal Breast Cancer Based on Ultrasound Radiomics Nomogram. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:649-664. [PMID: 35851691 DOI: 10.1002/jum.16061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE The objective of this research was to develop and validate an ultrasound-based radiomics nomogram for the pre-operative assessment of Ki-67 in breast cancer (BC). MATERIALS AND METHODS From December 2016 to December 2018, 515 patients with invasive ductal breast cancer who received two-dimensional (2D) ultrasound and Ki-67 examination were studied and analyzed retrospectively. The dataset was distributed at random into a training cohort (n = 360) and a test cohort (n = 155) in the ratio of 7:3. Each tumor region of interest was defined based on 2D ultrasound images and radiomics features were extracted. ANOVA, maximum correlation minimum redundancy (mRMR) algorithm, and minimum absolute shrinkage and selection operator (LASSO) were performed to pick features, and independent clinical predictors were integrated with radscore to construct the nomogram for predicting Ki-67 index by univariate and multivariate logistic regression analysis. The performance and utility of the models were evaluated by plotting receiver operating characteristic (ROC) curves, decision curve analysis (DCA), and calibration curves. RESULTS In the testing cohort, the area under the receiver characteristic curve (AUC) of the nomogram was 0.770 (95% confidence interval, 0.690-0.860). In both cohorts, the nomogram outperformed both the clinical model and the radiomics model (P < .05 according to the DeLong test). The analysis of DCA proved that the model has clinical utility. CONCLUSIONS The nomogram based on 2D ultrasound images offered an approach for predicting Ki-67 in BC.
Collapse
Affiliation(s)
- Yunpei Zhu
- Ultrasound Department, First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Yanping Dou
- Ultrasound Department, First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Ling Qin
- Ultrasound Department, First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Hui Wang
- Ultrasound Department, First Affiliated Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Zhihong Wen
- Radiology Department, Dalian Fifth People's Hospital, Dalian City, Liaoning Province, China
| |
Collapse
|
4
|
Haruna M, Daramola AO, Awolola NA, Badr NM, Banjo AAF, Shaaban A. Clinicopathological features and androgen receptor expression in triple negative breast cancer at Lagos, Nigeria. Ecancermedicalscience 2022; 16:1452. [PMID: 36405944 PMCID: PMC9666286 DOI: 10.3332/ecancer.2022.1452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Androgen receptor (AR) is one of the predominant nuclear hormone receptors in invasive breast cancer and can be explored as a biomarker of response for targeted anti-androgen therapy, especially in the setting of triple negative breast cancer (TNBC). Luminal AR is a distinct subtype amongst TNBC cases following gene expression studies. TNBC is higher in Africans (23%-82%) and African-Americans (29.8%) compared to Caucasian (10%-15%) breast cancer patients; however, there is a paucity of data on AR expression in this population. The aim of this study is to determine the expression of AR and the proportion of AR positive cancers in TNBCs at the Lagos University Teaching Hospital, Lagos, Nigeria. METHODOLOGY Out of 99 reviewed cases, 78 formalin fixed, paraffin embedded TNBC cases were assembled into a tissue microarray, stained and analysed for AR expression using immunohistochemistry. RESULTS The mean age of the TNBC patients was 49.3 years (range: 20-80 years). The histologic types in this study were invasive carcinoma (no special type) 75.4%; metaplastic carcinoma 21.4%; lobular carcinoma and mucinous carcinoma 1.6% each. Of 61 TNBC cases analysed, 37.7% were AR positive and 62.3% were AR negative, making the latter to become quadruple negative breast cancers. There was a significant association between age and AR expression (p = 0.02). In the subjects that expressed AR positivity, patients below 50 years accounted for 34.8% (8 of 23) while 65.2% (15 of 23) were above 50 years. There was no significant association between AR expression and histologic type or tumour grade. CONCLUSION Over a third of this Nigerian TNBC cohort study is AR+. This warrants further exploration of the predictive and prognostic significance of its expression amongst TNBC and the potential for targeted therapy, specifically androgen antagonists to improve the outcome of this disease with limited therapeutic options.
Collapse
Affiliation(s)
- Muibat Haruna
- Lagos University Teaching Hospital, Lagos 100254, Nigeria
| | - Adetola Olubunmi Daramola
- Lagos University Teaching Hospital, Lagos 100254, Nigeria
- Department of Anatomic and Molecular Pathology, College of Medicine, University of Lagos, Lagos 101014, Nigeria
| | - Nicholas Awodele Awolola
- Lagos University Teaching Hospital, Lagos 100254, Nigeria
- Department of Anatomic and Molecular Pathology, College of Medicine, University of Lagos, Lagos 101014, Nigeria
| | - Nahla Mustafa Badr
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin Elkom 32511, Egypt
- Institute of Cancer Science and Genomics, University of Birmingham, Birmingham B15 2WB, UK
| | - Adekunbiola Aina Fehintola Banjo
- Lagos University Teaching Hospital, Lagos 100254, Nigeria
- Department of Anatomic and Molecular Pathology, College of Medicine, University of Lagos, Lagos 101014, Nigeria
| | - Abeer Shaaban
- Institute of Cancer Science and Genomics, University of Birmingham, Birmingham B15 2WB, UK
- Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham B15 2WB, UK
| |
Collapse
|
5
|
The Autophagic Route of E-Cadherin and Cell Adhesion Molecules in Cancer Progression. Cancers (Basel) 2021; 13:cancers13246328. [PMID: 34944948 PMCID: PMC8699259 DOI: 10.3390/cancers13246328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary A hallmark of carcinoma progression is the loss of epithelial integrity. In this context, the deregulation of adhesion molecules, such as E-cadherin, affects epithelial structures and associates with epithelial to mesenchymal transition (EMT). This, in turn, fosters cancer progression. Autophagy endows cancer cells with the ability to overcome intracellular and environmental stress stimuli, such as anoikis, nutrient deprivation, hypoxia, and drugs. Furthermore, it plays an important role in the degradation of cell adhesion proteins and in EMT. This review focuses on the interplay between the turnover of adhesion molecules, primarily E-cadherin, and autophagy in cancer progression. Abstract Cell-to-cell adhesion is a key element in epithelial tissue integrity and homeostasis during embryogenesis, response to damage, and differentiation. Loss of cell adhesion and gain of mesenchymal features, a phenomenon known as epithelial to mesenchymal transition (EMT), are essential steps in cancer progression. Interestingly, downregulation or degradation by endocytosis of epithelial adhesion molecules (e.g., E-cadherin) associates with EMT and promotes cell migration. Autophagy is a physiological intracellular degradation and recycling process. In cancer, it is thought to exert a tumor suppressive role in the early phases of cell transformation but, once cells have gained a fully transformed phenotype, autophagy may fuel malignant progression by promoting EMT and conferring drug resistance. In this review, we discuss the crosstalk between autophagy, EMT, and turnover of epithelial cell adhesion molecules, with particular attention to E-cadherin.
Collapse
|
6
|
Koca B, Yildirim M, Kuru B. Prognostic Factors Affecting Disease-Free Survival in Triple-Negative Breast Cancer and Impact of Ki-67. Indian J Surg 2021. [DOI: 10.1007/s12262-021-03066-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
7
|
Hossain F, Majumder S, David J, Miele L. Precision Medicine and Triple-Negative Breast Cancer: Current Landscape and Future Directions. Cancers (Basel) 2021; 13:cancers13153739. [PMID: 34359640 PMCID: PMC8345034 DOI: 10.3390/cancers13153739] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The implementation of precision medicine will revolutionize cancer treatment paradigms. Notably, this goal is not far from reality: genetically similar cancers can be treated similarly. The heterogeneous nature of triple-negative breast cancer (TNBC) made it a suitable candidate to practice precision medicine. Using TNBC molecular subtyping and genomic profiling, a precision medicine-based clinical trial is ongoing. This review summarizes the current landscape and future directions of precision medicine and TNBC. Abstract Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous subtype of breast cancer associated with a high recurrence and metastasis rate that affects African-American women disproportionately. The recent approval of targeted therapies for small subgroups of TNBC patients by the US ‘Food and Drug Administration’ is a promising development. The advancement of next-generation sequencing, particularly somatic exome panels, has raised hopes for more individualized treatment plans. However, the use of precision medicine for TNBC is a work in progress. This review will discuss the potential benefits and challenges of precision medicine for TNBC. A recent clinical trial designed to target TNBC patients based on their subtype-specific classification shows promise. Yet, tumor heterogeneity and sub-clonal evolution in primary and metastatic TNBC remain a challenge for oncologists to design adaptive precision medicine-based treatment plans.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
- Correspondence:
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
| | - Justin David
- School of Medicine, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA;
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA; (S.M.); (L.M.)
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA
- School of Medicine, Louisiana State University Health Sciences Center (LSUHSC), New Orleans, LA 70112, USA;
| |
Collapse
|
8
|
Wu Q, Siddharth S, Sharma D. Triple Negative Breast Cancer: A Mountain Yet to Be Scaled Despite the Triumphs. Cancers (Basel) 2021; 13:3697. [PMID: 34359598 PMCID: PMC8345029 DOI: 10.3390/cancers13153697] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 12/12/2022] Open
Abstract
Metastatic progression and tumor recurrence pertaining to TNBC are certainly the leading cause of breast cancer-related mortality; however, the mechanisms underlying TNBC chemoresistance, metastasis, and tumor relapse remain somewhat ambiguous. TNBCs show 77% of the overall 4-year survival rate compared to other breast cancer subtypes (82.7 to 92.5%). TNBC is the most aggressive subtype of breast cancer, with chemotherapy being the major approved treatment strategy. Activation of ABC transporters and DNA damage response genes alongside an enrichment of cancer stem cells and metabolic reprogramming upon chemotherapy contribute to the selection of chemoresistant cells, majorly responsible for the failure of anti-chemotherapeutic regime. These selected chemoresistant cells further lead to distant metastasis and tumor relapse. The present review discusses the approved standard of care and targetable molecular mechanisms in chemoresistance and provides a comprehensive update regarding the recent advances in TNBC management.
Collapse
Affiliation(s)
| | - Sumit Siddharth
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA;
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231, USA;
| |
Collapse
|
9
|
Korpal M, Yelamanchi R, Durga CK, Bharadwaj M, Kambalimath C. Immunohistochemical E-cadherin Expression and Response to Chemotherapy in Breast Cancer Subjects. Indian J Surg 2021. [DOI: 10.1007/s12262-020-02576-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
10
|
Li C, Song L, Yin J. Intratumoral and Peritumoral Radiomics Based on Functional Parametric Maps from Breast DCE-MRI for Prediction of HER-2 and Ki-67 Status. J Magn Reson Imaging 2021; 54:703-714. [PMID: 33955619 DOI: 10.1002/jmri.27651] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Radiomics has been applied to breast magnetic resonance imaging (MRI) for gene status prediction. However, the features of peritumoral regions were not thoroughly investigated. PURPOSE To evaluate the use of intratumoral and peritumoral regions from functional parametric maps based on breast dynamic contrast-enhanced MRI (DCE-MRI) for prediction of HER-2 and Ki-67 status. STUDY TYPE Retrospective. POPULATION A total of 351 female patients (average age, 51 years) with pathologically confirmed breast cancer were assigned to the training (n = 243) and validation (n = 108) cohorts. FIELD STRENGTH/SEQUENCE 3.0T, T1 gradient echo. ASSESSMENT Radiomic features were extracted from intratumoral and peritumoral regions on six functional parametric maps calculated using time-intensity curves of DCE-MRI. The intraclass correlation coefficients (ICCs) were used to determine the reproducibility of feature extraction. Based on the intratumoral, peritumoral, and combined intra- and peritumoral regions, three radiomics signatures (RSs) were built using the least absolute shrinkage and selection operator (LASSO) logistic regression model, respectively. STATISTICAL TESTS Wilcoxon rank-sum test, minimum redundancy maximum relevance, LASSO, receiver operating characteristic curve (ROC) analysis, and DeLong test. RESULTS The intratumoral and peritumoral RSs for prediction of HER-2 and Ki-67 status achieved areas under the ROC (AUCs) of 0.683 (95% confidence interval [CI], 0.574-0.793) and 0.690 (95% CI, 0.577-0.804), and 0.714 (95% CI, 0.616-0.812) and 0.692 (95% CI, 0.590-0.794) in the validation cohort, respectively. The combined RSs yielded AUCs of 0.713 (95% CI, 0.604-0.823) and 0.749 (95% CI, 0.656-0.841), respectively. There were no significant differences in prediction performance among intratumoral, peritumoral, and combined RSs. Most (69.7%) of the features had good agreement (ICCs >0.8). DATA CONCLUSION Radiomic features of intratumoral and peritumoral regions on functional parametric maps based on breast DCE-MRI had the potential to identify HER-2 and Ki-67 status. LEVEL OF EVIDENCE 3 Technical Efficacy Stage: 2.
Collapse
Affiliation(s)
- Chunli Li
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lirong Song
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Brumec M, Sobočan M, Takač I, Arko D. Clinical Implications of Androgen-Positive Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:1642. [PMID: 33915941 PMCID: PMC8037213 DOI: 10.3390/cancers13071642] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 12/22/2022] Open
Abstract
This review summarizes the recent findings of a vast array of studies conducted on androgen receptor-positive triple-negative breast cancer (AR-positive TNBC) to provide a better understanding of this specific breast cancer subgroup. AR expression is correlated with higher age, lower histological grade, lower proliferation index Ki-67, spiculated masses, and calcifications on mammography. Studies investigating the correlation between AR expression and lymph node metastasis are highly discordant. In addition, results regarding prognosis are highly contradictory. AR antagonists are a promising novel therapeutic approach in AR-positive TNBC. However, AR signaling pathways should be more investigated in order to understand the influence of AR expression on TNBC more thoroughly.
Collapse
Affiliation(s)
- Maša Brumec
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (M.B.); (I.T.); (D.A.)
| | - Monika Sobočan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (M.B.); (I.T.); (D.A.)
- Department of Pharmacology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Divison of Gynecology and Perinatology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Iztok Takač
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (M.B.); (I.T.); (D.A.)
- Divison of Gynecology and Perinatology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Darja Arko
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (M.B.); (I.T.); (D.A.)
- Divison of Gynecology and Perinatology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
12
|
Cao W, Jiang Y, Ji X, Guan X, Lin Q, Ma L. Identification of novel prognostic genes of triple-negative breast cancer using meta-analysis and weighted gene co-expressed network analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:205. [PMID: 33708832 PMCID: PMC7940929 DOI: 10.21037/atm-20-5989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with high rates of metastasis and recurrence. Conventional clinical treatments are ineffective for it as it lacks therapeutic biomarkers. Figuring out the biomarkers related to TNBC will be beneficial for its clinical treatment and prognosis. Methods Five independent datasets downloaded from the Gene Expression Omnibus database were merged to identify differentially expressed genes between TNBC and non-TNBC samples by using the MetaDE.ES method followed by mapping the differentially expressed genes into a protein-protein interaction network. Meanwhile, the weighted gene co-expressed network analysis (WGCNA) of The Cancer Genome Atlas data was performed to screen the hub genes. The gene functional analyses were conducted by Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The correlation between gene expression level and patient overall survival was evaluated by survival analysis. Results A total of 11 differentially expressed genes (CDH1, SP1, MYC, FAF2, IFI16, MDM2, AR, DBN1, HSPB1, FLNA, YWHAB) were obtained from the protein-protein interaction network with degree >10. WGCNA revealed 5 hub genes (TPX2, CTPS1, KIF2C, MELK, CDCA8) that were significantly associated with TNBC. Cell cycle, oocyte meiosis, spliceosome were the pathways significantly enriched in these genes according to GO functionally annotated terms and KEGG pathways analysis. The Kaplan-Meier curves showed that the expression levels of HSPB1, IFI16, TPX2 were significantly associated with the survival time of TNBC patients (P<0.05). Conclusions A total of 16 genes significantly associated with TNBC were identified by bioinformatic analyses. Among these 16 genes, HSPB1, IFI16, TPX2 might be able to be used as biomarkers of TNBC.
Collapse
Affiliation(s)
- Wenning Cao
- Department of Chemistry, Tsinghua University, Beijing, China.,State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Yike Jiang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China.,Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
| | - Xiang Ji
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,School of Life Science, Tsinghua University, Beijing, China
| | - Xuejiao Guan
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,School of Life Science, Tsinghua University, Beijing, China
| | - Qianyu Lin
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China
| | - Lan Ma
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China.,Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| |
Collapse
|
13
|
Chen Y, Lin Y, Cui Z. Identification of adriamycin resistance genes in breast cancer based on microarray data analysis. Transl Cancer Res 2020; 9:7486-7494. [PMID: 35117349 PMCID: PMC8797850 DOI: 10.21037/tcr-19-2145] [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] [Received: 10/11/2019] [Accepted: 02/05/2020] [Indexed: 11/06/2022]
Abstract
Background Breast cancer is a common malignant tumor with increasing incidence worldwide. This study aimed to investigate the molecular mechanisms of the adriamycin (ADR) resistance in breast cancer. Methods The GSE76540 dataset downloaded from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database was adopted for analysis. Differentially expressed genes (DEGs) in chemo-sensitive cases and chemo-resistant cases were identified using the GEO2R online tool respectively. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of DEGs were carried out by using the DAVID online tool. The protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING) and visualized with Cytoscape software. The impact of key tumor genes on the survival and prognosis were described. Results A total of 1,481 DEGs were excavated, including 549 up-regulated genes and 932 down-regulated genes. According to the GO analysis, the DEGs were significantly enriched in: extracellular matrix organization, positive regulation of transcription from RNA polymerase II promoter, lung development, positive regulation of gene expression, axon guidance and so on. The results of KEGG pathway enrichment analysis showed that the most enriched DEGs can be detected in: pathways in cancer, PI3K/AKT signaling pathway, focal adhesion, Ras signaling pathway and so on. In the PPI network analysis, hub genes of CDH1, ESR1, SOX2, AR, GATA3, FOXA1, KRT19, CLDN7, AGR2, ESRP1, RAB25, CLDN4, IGF1R, CLDN3 and IRS1 were detected. Finally, there is a correlation filter out these hub genes in resistance of ADR. Conclusions Hub genes associated with ADR resistance were identified using bioinformatic techniques. The results of this study may contribute to the development of targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Yan Chen
- Laboratory of Biochemistry and Molecular Biology Research, Fujian Provincial Key Laboratory of Tumor Biotherapy, Department of Clinical Laboratory, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yingfeng Lin
- Laboratory of Biochemistry and Molecular Biology Research, Fujian Provincial Key Laboratory of Tumor Biotherapy, Department of Clinical Laboratory, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Zhaolei Cui
- Laboratory of Biochemistry and Molecular Biology Research, Fujian Provincial Key Laboratory of Tumor Biotherapy, Department of Clinical Laboratory, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
14
|
Gupta GK, Collier AL, Lee D, Hoefer RA, Zheleva V, Siewertsz van Reesema LL, Tang-Tan AM, Guye ML, Chang DZ, Winston JS, Samli B, Jansen RJ, Petricoin EF, Goetz MP, Bear HD, Tang AH. Perspectives on Triple-Negative Breast Cancer: Current Treatment Strategies, Unmet Needs, and Potential Targets for Future Therapies. Cancers (Basel) 2020; 12:E2392. [PMID: 32846967 PMCID: PMC7565566 DOI: 10.3390/cancers12092392] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/10/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC), characterized by the absence or low expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER2), is the most aggressive subtype of breast cancer. TNBC accounts for about 15% of breast cancer cases in the U.S., and is known for high relapse rates and poor overall survival (OS). Chemo-resistant TNBC is a genetically diverse, highly heterogeneous, and rapidly evolving disease that challenges our ability to individualize treatment for incomplete responders and relapsed patients. Currently, the frontline standard chemotherapy, composed of anthracyclines, alkylating agents, and taxanes, is commonly used to treat high-risk and locally advanced TNBC. Several FDA-approved drugs that target programmed cell death protein-1 (Keytruda) and programmed death ligand-1 (Tecentriq), poly ADP-ribose polymerase (PARP), and/or antibody drug conjugates (Trodelvy) have shown promise in improving clinical outcomes for a subset of TNBC. These inhibitors that target key genetic mutations and specific molecular signaling pathways that drive malignant tumor growth have been used as single agents and/or in combination with standard chemotherapy regimens. Here, we review the current TNBC treatment options, unmet clinical needs, and actionable drug targets, including epidermal growth factor (EGFR), vascular endothelial growth factor (VEGF), androgen receptor (AR), estrogen receptor beta (ERβ), phosphoinositide-3 kinase (PI3K), mammalian target of rapamycin (mTOR), and protein kinase B (PKB or AKT) activation in TNBC. Supported by strong evidence in developmental, evolutionary, and cancer biology, we propose that the K-RAS/SIAH pathway activation is a major tumor driver, and SIAH is a new drug target, a therapy-responsive prognostic biomarker, and a major tumor vulnerability in TNBC. Since persistent K-RAS/SIAH/EGFR pathway activation endows TNBC tumor cells with chemo-resistance, aggressive dissemination, and early relapse, we hope to design an anti-SIAH-centered anti-K-RAS/EGFR targeted therapy as a novel therapeutic strategy to control and eradicate incurable TNBC in the future.
Collapse
Affiliation(s)
- Gagan K. Gupta
- Leroy T. Canoles Jr. Cancer Research Center, Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA;
| | - Amber L. Collier
- DeWitt Daughtry Family Department of Surgery, Surgical Oncology, University of Miami/Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33131, USA;
| | - Dasom Lee
- Department of Medicine, Internal Medicine, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL 33620, USA;
| | - Richard A. Hoefer
- Dorothy G. Hoefer Foundation, Sentara CarePlex Hospital, Newport News, VA 23666, USA;
- Sentara Cancer Network, Sentara Healthcare, Norfolk, VA 23507, USA;
| | - Vasilena Zheleva
- Surgical Oncology, Cancer Treatment Centers of America—Comprehensive Care and Research Center Phoenix, 14200 W Celebrate Life Way, Goodyear, AZ 85338, USA;
| | | | - Angela M. Tang-Tan
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley, CA 94720, USA;
| | - Mary L. Guye
- Sentara Cancer Network, Sentara Healthcare, Norfolk, VA 23507, USA;
- Sentara Surgery Specialists, Sentara CarePlex Hospital, Newport News, VA 23666, USA
| | - David Z. Chang
- Virginia Oncology Associates, 1051 Loftis Boulevard, Suite 100, Newport News, VA 23606, USA;
| | - Janet S. Winston
- Breast Pathology Services, Pathology Sciences Medical Group, Department of Pathology, Sentara Norfolk General Hospital (SNGH), Norfolk, VA 23507, USA; (J.S.W.); (B.S.)
| | - Billur Samli
- Breast Pathology Services, Pathology Sciences Medical Group, Department of Pathology, Sentara Norfolk General Hospital (SNGH), Norfolk, VA 23507, USA; (J.S.W.); (B.S.)
| | - Rick J. Jansen
- Department of Public Health, North Dakota State University, Fargo, ND 58102, USA;
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason University, Manassas, VA 20110, USA;
| | - Matthew P. Goetz
- Departments of Oncology and Pharmacology, Mayo Clinic Breast Cancer Specialized Program of Research Excellence (SPORE), Women’s Cancer Program, Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN 55905, USA;
| | - Harry D. Bear
- Departments of Surgery and Microbiology & Immunology, Division of Surgical Oncology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Amy H. Tang
- Leroy T. Canoles Jr. Cancer Research Center, Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA;
| |
Collapse
|
15
|
Govindan S, Siraganahalli Eswaraiah M, Basavaraj C, Adinarayan M, Sankaran S, Bakre M. Androgen Receptor mRNA levels determine the prognosis in triple-negative breast cancer patients. BMC Cancer 2020; 20:745. [PMID: 32778063 PMCID: PMC7419184 DOI: 10.1186/s12885-020-07218-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/26/2020] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Anti-Androgen Receptor (AR) therapy holds promise for a subset of AR expressing triple-negative breast cancer (TNBC) patients. However, current AR assays are suboptimal in detecting the dynamic range of AR expression, contributing to its controversial role in TNBC disease prognosis. This study is aimed at evaluating the feasibility of qRT-PCR to sensitively and robustly detect AR mRNA levels for prognostication. METHODS mRNA expression profiling was performed on FFPE blocks from a retrospective cohort of 101 TNBC patients using qRT-PCR and compared with AR protein expression by immunohistochemistry . Statistical analyses included Spearman's rank correlation, Chi-square and Kaplan-Meier analyses. Distant Metastasis Free Survival was used as the end point in survival analysis. RESULTS AR mRNA expression was observed in 34/101 patients (34%) whereas 12/80 cases (15%) were positive by IHC. qRT-PCR could thus detect more AR positive patients as compared to IHC, with 75% (9/12) concordance between the two methods. Co-expression of GATA3 and FOXA1 mRNA was observed in 85 and 88% of AR mRNA positive tumors, respectively. AR mRNA positivity was significantly correlated with age at disease onset (p = 0.02), high FOXA1/GATA3 (p < 0.05) and distant recurrence. AR mRNA positive patients had poorer DMFS (43%; p = 0.002). DMFS dropped further to 26% (p = 0.006) in AR (+)/high FOXA1/GATA3 patients. AR mRNA expression together with node positivity had the worst DMFS (23%; p < 0.0001) compared to patients who were either positive for any one of these, or negative for both AR and node status. Low Ki67 mRNA with AR mRNA positivity also had poorer DMFS (39%; p = 0.001) compared to patients expressing low Ki67 with no AR mRNA expression. CONCLUSION qRT-PCR was more sensitive and reliable in detecting the dynamic expression levels of AR compared to IHC and this variation could be explained by the higher sensitivity of the former method. High AR mRNA expression was strongly associated with expression of AR protein, high FOXA1/GATA3 mRNA, and with poor prognosis. qRT-PCR was more efficient in detecting the AR positive cases compared to IHC. A distinct signature involving high GATA3/FOXA1, low Ki67, and node positivity in AR mRNA positive tumors correlated with poor prognosis. Thus, AR mRNA screening can serve as an effective prognostic marker along with offering potential targeted therapy options for TNBC.
Collapse
Affiliation(s)
- Sindhu Govindan
- OncoStem Diagnostics Private Limited, # 4, Raja Ram Mohan Roy Road, Aanand Tower, 2nd Floor, Bangalore, Karnataka, 560027, India
| | | | - Chetana Basavaraj
- OncoStem Diagnostics Private Limited, # 4, Raja Ram Mohan Roy Road, Aanand Tower, 2nd Floor, Bangalore, Karnataka, 560027, India
| | - Manjula Adinarayan
- OncoStem Diagnostics Private Limited, # 4, Raja Ram Mohan Roy Road, Aanand Tower, 2nd Floor, Bangalore, Karnataka, 560027, India
| | - Satish Sankaran
- OncoStem Diagnostics Private Limited, # 4, Raja Ram Mohan Roy Road, Aanand Tower, 2nd Floor, Bangalore, Karnataka, 560027, India
| | - Manjiri Bakre
- OncoStem Diagnostics Private Limited, # 4, Raja Ram Mohan Roy Road, Aanand Tower, 2nd Floor, Bangalore, Karnataka, 560027, India.
| |
Collapse
|
16
|
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
|
17
|
Vidula N, Yau C, Wolf D, Rugo HS. Androgen receptor gene expression in primary breast cancer. NPJ Breast Cancer 2019; 5:47. [PMID: 31840050 PMCID: PMC6904475 DOI: 10.1038/s41523-019-0142-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/22/2019] [Indexed: 12/20/2022] Open
Abstract
We studied androgen receptor (AR) gene expression in primary breast cancer (BC) to determine associations with clinical characteristics and outcomes in the I-SPY 1 study. AR was evaluated in I-SPY 1 (n = 149) using expression microarrays. Associations of AR with clinical and tumor features were determined using the Wilcoxon rank sum test (two-level factors) or the Kruskal-Wallis test (multi-level factors). We identified an optimal AR cut-point to maximize recurrence-free survival (RFS) differences between AR biomarker stratified groups, and assessed the association between the AR stratified groups and RFS using the Cox proportional hazard model. Pearson correlations between AR and selected genes were determined in I-SPY 1, METABRIC (n = 1992), and TCGA (n = 817). AR was lower in triple negative BC vs. hormone receptor positive (HR+)/HER2- and HER2+ disease (p < 0.00001), and lower in basal-like BC (p < 0.00001). AR was higher in grade I/II vs. III tumors (p < 0.00001), in patients >age 50 (p = 0.05), and in node negative disease (p = 0.006). Higher AR was associated with better RFS (p = 0.0007), which remained significant after receptor subtype adjustment (p = 0.01). AR correlated with expression of luminal, HER2, and steroid hormone genes. AR expression was related to clinicopathologic features, intrinsic subtype, and correlated with improved outcome.
Collapse
Affiliation(s)
- Neelima Vidula
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Christina Yau
- University of California San Francisco, 1600 Divisadero Street, San Francisco, CA 94115 USA
| | - Denise Wolf
- University of California San Francisco, 1600 Divisadero Street, San Francisco, CA 94115 USA
| | - Hope S. Rugo
- University of California San Francisco, 1600 Divisadero Street, San Francisco, CA 94115 USA
| |
Collapse
|
18
|
Fonseca-Alves CE, Kobayashi PE, Leis-Filho AF, Lainetti PDF, Grieco V, Kuasne H, Rogatto SR, Laufer-Amorim R. E-Cadherin Downregulation is Mediated by Promoter Methylation in Canine Prostate Cancer. Front Genet 2019; 10:1242. [PMID: 31850082 PMCID: PMC6895247 DOI: 10.3389/fgene.2019.01242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
E-cadherin is a transmembrane glycoprotein responsible for cell-to-cell adhesion, and its loss has been associated with metastasis development. Although E-cadherin downregulation was previously reported in canine prostate cancer (PC), the mechanism involved in this process is unclear. It is well established that dogs, besides humans, spontaneously develop PC with high frequency; therefore, canine PC is an interesting model to study human PC. In human PC, CDH1 methylation has been associated with E-cadherin downregulation. However, no previous studies have described the methylation pattern of CDH1 promoter in canine PC. Herein, we evaluated the E-cadherin protein and gene expression in canine PC compared to normal tissues. DNA methylation pattern was investigated as a regulatory mechanism of CDH1 silencing. Our cohort is composed of 20 normal prostates, 20 proliferative inflammatory atrophy (PIA) lesions, 20 PC, and 11 metastases from 60 dogs. The E-cadherin protein expression was assessed by immunohistochemistry and western blotting and gene expression by qPCR. Bisulfite- pyrosequencing assay was performed to investigate the CDH1 promoter methylation pattern. Membranous E-cadherin expression was observed in all prostatic tissues. A higher number of E-cadherin negative cells was detected more frequently in PC compared to normal and PIA samples. High-grade PC showed a diffuse membranous positive immunostaining. Furthermore, PC patients with a higher number of E-cadherin negative cells presented shorter survival time and higher Gleason scores. Western blotting and qPCR assays confirmed the immunohistochemical results, showing lower E-cadherin protein and gene expression levels in PC compared to normal samples. We identified CDH1 promoter hypermethylation in PIA and PC samples. An in vitro assay with two canine prostate cancer cells (PC1 and PC2 cell lines) was performed to confirm the methylation as a regulatory mechanism of E-cadherin expression. PC1 cell line presented CDH1 hypermethylation and after 5-Aza-dC treatment, a decreased CDH1 methylation and increased gene expression levels were observed. Positive E-cadherin cells were massively found in metastases (mean of 90.6%). In conclusion, low levels of E-cadherin protein, gene downregulation and CDH1 hypermethylation was detected in canine PC. However, in metastatic foci occur E-cadherin re-expression confirming its relevance in these processes.
Collapse
Affiliation(s)
- Carlos Eduardo Fonseca-Alves
- Institute of Health Sciences, Paulista University-UNIP, Bauru, Brazil.,Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, Sao Paulo State University-UNESP, Botucatu, Brazil
| | - Priscila Emiko Kobayashi
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Sao Paulo State University-UNESP, Botucatu, Brazil
| | - Antonio Fernando Leis-Filho
- Department of Veterinary Clinic, School of Veterinary Medicine and Animal Science, Sao Paulo State University-UNESP, Botucatu, Brazil
| | - Patricia de Faria Lainetti
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, Sao Paulo State University-UNESP, Botucatu, Brazil
| | - Valeria Grieco
- Department of Veterinary Medicine, Università degli studi di Milano, Milan, Italy
| | - Hellen Kuasne
- International Center for Research (CIPE), AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Renee Laufer-Amorim
- Department of Veterinary Surgery and Anesthesiology, School of Veterinary Medicine and Animal Science, Sao Paulo State University-UNESP, Botucatu, Brazil
| |
Collapse
|
19
|
Fan M, Yuan W, Zhao W, Xu M, Wang S, Gao X, Li L. Joint Prediction of Breast Cancer Histological Grade and Ki-67 Expression Level Based on DCE-MRI and DWI Radiomics. IEEE J Biomed Health Inform 2019; 24:1632-1642. [PMID: 31794406 DOI: 10.1109/jbhi.2019.2956351] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Histologic grade and Ki-67 proliferation status are important clinical indictors for breast cancer prognosis and treatment. The purpose of this study is to improve prediction accuracy of these clinical indicators based on tumor radiomic analysis. METHODS We jointly predicted Ki-67 and tumor grade with a multitask learning framework by separately utilizing radiomics from tumor MRI series. Additionally, we showed how multitask learning models (MTLs) could be extended to combined radiomics from the MRI series for a better prediction based on the assumption that features from different sources of images share common patterns while providing complementary information. Tumor radiomic analysis was performed with morphological, statistical and textural features extracted on the DWI and dynamic contrast-enhanced MRI (DCE-MRI) series of the precontrast and subtraction images, respectively. RESULTS Joint prediction of Ki-67 status and tumor grade on MR images using the MTL achieved performance improvements over that of single-task-based predictive models. Similarly, for the prediction tasks of Ki-67 and tumor grade, the MTL for combined precontrast and apparent diffusion coefficient (ADC) images achieved AUCs of 0.811 and 0.816, which were significantly better than that of the single-task- based model with p values of 0.005 and 0.017, respectively. CONCLUSION Mapping MRI radiomics to two related clinical indicators improves prediction performance for both Ki-67 expression level and tumor grade. SIGNIFICANCE Joint prediction of indicators by multitask learning that combines correlations of MRI radiomics is important for optimal tumor therapy and treatment because clinical decisions are made by integrating multiple clinical indicators.
Collapse
|
20
|
Vasiliou SK, Diamandis EP. Androgen receptor: A promising therapeutic target in breast cancer. Crit Rev Clin Lab Sci 2019; 56:200-223. [PMID: 30821186 DOI: 10.1080/10408363.2019.1575643] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Breast cancer (BCa) is the second most common cancer worldwide and the most prevalent cancer in women. The majority of BCa cases are positive (+) for the estrogen receptor (ER+, 80%) and progesterone receptor (PR+, 65%). Estrogen and progesterone hormones are known to be involved in cancer progression, and thus hormonal deprivation is used as an effective treatment for ER+PR+ BCa subtypes. However, some ER+PR+ BCa patients develop resistance to such therapies. Meanwhile, chemotherapy is the only available treatment for ER-PR- BCa tumors. Another hormone receptor known as the androgen receptor (AR) has also been found to be widely expressed in human breast carcinomas. However, the mechanisms of AR and its endogenous androgen ligands is not well-understood in BCa and its biological role in this hormone-related disease remains unclear. In this review, we aim to address the importance of the AR in BCa diagnosis and prognosis, current AR-targeting approaches in BCa, and the potential for AR-downstream molecules to serve as therapeutic targets.
Collapse
Affiliation(s)
- Stella K Vasiliou
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada
| |
Collapse
|
21
|
Kundur S, Prayag A, Selvakumar P, Nguyen H, McKee L, Cruz C, Srinivasan A, Shoyele S, Lakshmikuttyamma A. Synergistic anticancer action of quercetin and curcumin against triple‐negative breast cancer cell lines. J Cell Physiol 2018; 234:11103-11118. [DOI: 10.1002/jcp.27761] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Sai Kundur
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Amrita Prayag
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Priyanga Selvakumar
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Hung Nguyen
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Lloyd McKee
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Clairissa Cruz
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Asha Srinivasan
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Sunday Shoyele
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| | - Ashakumary Lakshmikuttyamma
- Department of Pharmaceutical Sciences Jefferson College of Pharmacy, Thomas Jefferson University Philadelphia Pennsylvania
| |
Collapse
|
22
|
Wang J, Wang Y, Long F, Yan F, Wang N, Wang Y. The expression and clinical significance of GADD45A in breast cancer patients. PeerJ 2018; 6:e5344. [PMID: 30128181 PMCID: PMC6098681 DOI: 10.7717/peerj.5344] [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: 02/21/2018] [Accepted: 07/09/2018] [Indexed: 01/21/2023] Open
Abstract
Background Growth arrest and DNA-damage-inducible protein 45 alpha (GADD45A) was previously found to be associated with risk of several kinds of human tumors. Here, we studied the expression and clinical significance of GADD45A in breast cancer. Methods We performed an immunohistochemical study of GADD45A protein from 419 breast cancer tissues and 116 adjacent non-neoplastic tissues. Results Significantly high GADD45A expression were observed in breast cancer tissues compared with adjacent non-neoplastic tissues (P < 0.001) and were independently correlative with estrogen receptor negative (P = 0.028) and high Ki-67 index (P < 0.001). Kaplan-Meier survival analysis revealed that patients with high GADD45A expression levels had a worse long-term prognosis in triple negative breast cancer (P = 0.041), but it was not an independent prognostic factor in multivariate analysis (P = 0.058). Conclusions GADD45A expression levels are significantly correlative with estrogen receptor status and Ki-67 index in human breast cancer. Patients with triple negative breast cancer might be stratified into high risk and low risk groups based on the GADD45A expression levels.
Collapse
Affiliation(s)
- Junnan Wang
- Basic Medical College, Navy Medical University, Shanghai, China
| | - Yiran Wang
- Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fei Long
- Department of Oncology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Fengshang Yan
- Basic Medical College, Navy Medical University, Shanghai, China
| | - Ning Wang
- Department of Oncology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Yajie Wang
- Department of Oncology, Changhai Hospital, Navy Medical University, Shanghai, China
| |
Collapse
|
23
|
Expression of PD-1 on CD4 + Tumor-Infiltrating Lymphocytes in Tumor Microenvironment Associated with Pathological Characteristics of Breast Cancer. J Immunol Res 2018; 2018:5690258. [PMID: 30069490 PMCID: PMC6057338 DOI: 10.1155/2018/5690258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/16/2018] [Accepted: 05/23/2018] [Indexed: 12/30/2022] Open
Abstract
Objective This study aimed to investigate the correlation of CD4+/PD-1+ or CD4+/PD-1− tumor-infiltrating lymphocytes with pathological characteristics in breast cancer patients. Methods A cross-sectional study consecutively recruited 133 patients with invasive ductal breast cancer. The expression of CD4, programmed cell death protein 1 (PD-1), CK7, CK20, E-cadherin, or Ki-67 was detected by immunohistochemistry. The associations between CD4+/PD-1+ or CD4+/PD-1− tumor-infiltrating lymphocytes and pathological characteristics were evaluated. Results Elderly patients intended to have a lower level of CD4+/PD-1− tumor-infiltrating lymphocytes (p < 0.05). Patients with positive E-cadherin expression had higher median cell counts of CD4+/PD-1− tumor-infiltrating lymphocytes than patients with negative E-cadherin expression (30/HPF versus 10/HPF, p < 0.05). Counts of CD4+/PD-1+ tumor-infiltrating lymphocytes had a significant correlation with Ki-67 index that the correlation coefficient was 0.29 (p = 0.001). Positive CK20 expression was related to a higher level of CD4+/PD-1− tumor-infiltrating lymphocytes than negative CK20 expression (73/HPF versus 30/HPF, p < 0.05). Conclusion CD4+/PD-1+ or CD4+/PD-1− tumor-infiltrating lymphocytes showed diverse association with pathological features of breast cancer. CD4+/PD-1+ tumor-infiltrating lymphocytes had a significant relationship with Ki-67 expression whereas CD4+/PD-1− tumor-infiltrating lymphocytes had a significant relationship with E-cadherin expression. Further studies are warranted to explore the immunomodulatory effects of phenotypes of CD4+ T cell subsets in breast cancer.
Collapse
|
24
|
Liu YX, Zhang KJ, Tang LL. Clinical significance of androgen receptor expression in triple negative breast cancer-an immunohistochemistry study. Oncol Lett 2018; 15:10008-10016. [PMID: 29844843 PMCID: PMC5958876 DOI: 10.3892/ol.2018.8548] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Androgen receptor (AR) is closely associated with the occurrence and progression of breast cancer; however, the clinical significance of it in triple negative breast cancer (TNBC) has been controversial. There is a limited amount of research regarding the effect of neoadjuvant chemotherapy on AR expression. By examining the expression of AR in patients with TNBC, the aim of the present study is to explore the clinical significance of AR and provide evidence for AR-directed treatment in TNBC. A total of 188 patients with primary TNBC with complete medical records were included in this retrospective study. Tumor sections from 41 patients (21.8%) were positive for AR, which was more often detected in small tumors (P=0.042) and cases with no lymph node involvement (P=0.032). Among them, 102 were treated with neoadjuvant chemotherapy (NAC). A total of 17 patients (16.7%) exhibited pathological complete response. However, the patient response was irrelevant to AR expression. Matched pathological tissues before and after NAC were collected for 49 cases, suggesting an enrichment of AR-expressing tumors following chemotherapy (P=0.008). Further analysis indicated that AR expression had no correlation with the disease-free and overall survival of patients with general TNBC; rather, it predicted a poor survival of the patients with stage III TNBC in comparison with those at earlier stages (P=0.035). AR expression occurs more often in small TNBC tumors or in cases with no lymph node metastasis. It is associated with a poor prognosis of the patients with advanced stages of tumors.
Collapse
Affiliation(s)
- Ya-Xuan Liu
- Department of Breast Surgery, Breast Cancer Prevention and Clinical Research Center, Xiangya Hospital Central South University, Changsha, Hunan 410008, P.R. China
| | - Ke-Jing Zhang
- Department of Breast Surgery, Breast Cancer Prevention and Clinical Research Center, Xiangya Hospital Central South University, Changsha, Hunan 410008, P.R. China
| | - Li-Li Tang
- Department of Breast Surgery, Breast Cancer Prevention and Clinical Research Center, Xiangya Hospital Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
25
|
Yang F, Shen Y, Zhang W, Jin J, Huang D, Fang H, Ji W, Shi Y, Tang L, Chen W, Zhou G, Guan X. An androgen receptor negatively induced long non-coding RNA ARNILA binding to miR-204 promotes the invasion and metastasis of triple-negative breast cancer. Cell Death Differ 2018; 25:2209-2220. [PMID: 29844570 DOI: 10.1038/s41418-018-0123-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 12/30/2022] Open
Abstract
Androgen receptor (AR) is emerging as a novel prognostic biomarker in triple-negative breast cancer (TNBC), but the underlying mechanisms remain unknown. As accumulating evidence has shown that long non-coding RNAs (lncRNAs) regulate important cancer hallmarks, we hypothesised that AR-regulated lncRNAs might play roles in TNBC progression. Here, we performed experiments with or without DHT treatment in three TNBC cell lines, and we identified an AR negatively induced lncRNA (ARNILA), which correlated with poor progression-free survival (PFS) in TNBC patients and promoted epithelial-mesenchymal transition (EMT), invasion and metastasis in vitro and in vivo. Subsequently, we demonstrated that ARNILA functioned as a competing endogenous RNA (ceRNA) for miR-204 to facilitate expression of its target gene Sox4, which is known to induce EMT and contribute to breast cancer progression, thereby promoting EMT, invasion and metastasis of TNBC. Our findings not only provide new insights into the mechanisms of lncRNA in regulating AR but also suggest ARNILA as an alternative therapeutic target to suppress metastasis of TNBC patients.
Collapse
Affiliation(s)
- Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Shen
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Zhang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Juan Jin
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Doudou Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hehui Fang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenfei Ji
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing, China
| | - Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lin Tang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiwei Chen
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guohua Zhou
- Department of Pharmacology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China. .,Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing, China. .,Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
26
|
Horne HN, Oh H, Sherman ME, Palakal M, Hewitt SM, Schmidt MK, Milne RL, Hardisson D, Benitez J, Blomqvist C, Bolla MK, Brenner H, Chang-Claude J, Cora R, Couch FJ, Cuk K, Devilee P, Easton DF, Eccles DM, Eilber U, Hartikainen JM, Heikkilä P, Holleczek B, Hooning MJ, Jones M, Keeman R, Mannermaa A, Martens JWM, Muranen TA, Nevanlinna H, Olson JE, Orr N, Perez JIA, Pharoah PDP, Ruddy KJ, Saum KU, Schoemaker MJ, Seynaeve C, Sironen R, Smit VTHBM, Swerdlow AJ, Tengström M, Thomas AS, Timmermans AM, Tollenaar RAEM, Troester MA, van Asperen CJ, van Deurzen CHM, Van Leeuwen FF, Van't Veer LJ, García-Closas M, Figueroa JD. E-cadherin breast tumor expression, risk factors and survival: Pooled analysis of 5,933 cases from 12 studies in the Breast Cancer Association Consortium. Sci Rep 2018; 8:6574. [PMID: 29700408 PMCID: PMC5920115 DOI: 10.1038/s41598-018-23733-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/16/2018] [Indexed: 01/20/2023] Open
Abstract
E-cadherin (CDH1) is a putative tumor suppressor gene implicated in breast carcinogenesis. Yet, whether risk factors or survival differ by E-cadherin tumor expression is unclear. We evaluated E-cadherin tumor immunohistochemistry expression using tissue microarrays of 5,933 female invasive breast cancers from 12 studies from the Breast Cancer Consortium. H-scores were calculated and case-case odds ratios (OR) and 95% confidence intervals (CIs) were estimated using logistic regression. Survival analyses were performed using Cox regression models. All analyses were stratified by estrogen receptor (ER) status and histologic subtype. E-cadherin low cases (N = 1191, 20%) were more frequently of lobular histology, low grade, >2 cm, and HER2-negative. Loss of E-cadherin expression (score < 100) was associated with menopausal hormone use among ER-positive tumors (ever compared to never users, OR = 1.24, 95% CI = 0.97-1.59), which was stronger when we evaluated complete loss of E-cadherin (i.e. H-score = 0), OR = 1.57, 95% CI = 1.06-2.33. Breast cancer specific mortality was unrelated to E-cadherin expression in multivariable models. E-cadherin low expression is associated with lobular histology, tumor characteristics and menopausal hormone use, with no evidence of an association with breast cancer specific survival. These data support loss of E-cadherin expression as an important marker of tumor subtypes.
Collapse
Affiliation(s)
- Hisani N Horne
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Division of Molecular Genetics & Pathology, US Food and Drug Administration, Silver Spring, MD, USA
| | - Hannah Oh
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Department of Health Policy and Management, College of Health Science, Korea University, Seoul, Korea
| | - Mark E Sherman
- Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Maya Palakal
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Stephen M Hewitt
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Roger L Milne
- Cancer Epidemiology & Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global health, The University of Melbourne, Melbourne, Victoria, Australia
| | - David Hardisson
- Department of Pathology, Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz IdiPAZ, and Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain
| | - Javier Benitez
- Human Cancer Genetics Program, Spanish National Cancer Research Centre, Madrid, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Carl Blomqvist
- Department of Oncology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Manjeet K Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Research Group Genetic Cancer Epidemiology, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Renata Cora
- Independent contractor, CT(ASCP), MB (ASCP), National Cancer Institute, Bethesda, MD, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Katarina Cuk
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Devilee
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Diana M Eccles
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ursula Eilber
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jaana M Hartikainen
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Heikkilä
- Department of Pathology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | | | - Maartje J Hooning
- Department of Medical Oncology, Family Cancer Clinic, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michael Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Renske Keeman
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Arto Mannermaa
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - John W M Martens
- Department of Medical Oncology, Family Cancer Clinic, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Taru A Muranen
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Nick Orr
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Jose I A Perez
- Servicio de Cirugía General y Especialidades, Hospital Monte Naranco, Oviedo, Spain
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Kai-Uwe Saum
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Caroline Seynaeve
- Department of Medical Oncology, Family Cancer Clinic, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Reijo Sironen
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Maria Tengström
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Cancer Center, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Oncology, University of Eastern Finland, Kuopio, Finland
| | - Abigail S Thomas
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - A Mieke Timmermans
- Department of Medical Oncology, Family Cancer Clinic, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa A Troester
- Department of Pathology and Laboratory Medicin, Gillings School of Global Public Health, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Christi J van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Flora F Van Leeuwen
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Laura J Van't Veer
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh Medical School, Edinburgh, UK.
| |
Collapse
|
27
|
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: 58] [Impact Index Per Article: 8.3] [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
|
28
|
Luo Y, Huang W, Zhang H, Liu G. Prognostic significance of CD117 expression and TP53 missense mutations in triple-negative breast cancer. Oncol Lett 2018; 15:6161-6170. [PMID: 29616097 PMCID: PMC5876428 DOI: 10.3892/ol.2018.8104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/24/2017] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is extremely aggressive and associated with poor prognosis. There are no known predictive or prognostic markers for TNBC. Inhibition of tumor protein P53 (TP53) has been demonstrated to increase the levels of cluster of differentiation 117 (CD117) in human colorectal cancer cells. However, the function of TP53 in the regulation of CD117 in TNBC has, to the best of our knowledge, not been reported. In the present study, the association between the expression of CD117 protein and TP53 mutations was investigated, and their prognostic value in patients with TNBC was assessed. A total of 58 TNBC and 48 non-TNBC breast cancer tissue samples were assessed for the expression of CD117, p53 and TP53 mutations. The marker of proliferation Ki-67 (MKI67) proliferation index and vascular invasion index (obtained by measuring D2-40 and CD34) was investigated via immunohistochemistry, and mutations in exons 4–8 of TP53 were measured using direct sequencing. Associations between CD117 and p53 levels or TP53 mutations and clinical parameters were statistically evaluated. The rates of CD117 or MKI67 positivity, CD117+/TP53 missense mutation+, TP53 missense mutations or recurrence were significantly higher in patients with TNBC than in patients with non-TNBC. In TNBC tissues, the presence of CD117 was associated with TP53 missense mutations (P=0.031), vascular invasion, recurrence and MKI67. CD117+/TP53 missense mutation+ also associated with vascular invasion, recurrence and MKI67. Under univariate analysis, MKI67, vascular invasion, CD117, CD117+/TP53 missense mutation+ and TP53 missense mutations were associated with the overall survival of patients with TNBC. Multivariate analysis revealed that vascular invasion and CD117+/TP53 missense mutation+ in primary tumors were independent prognostic factors in patients with TNBC. In conclusion, CD117+/TP53 missense mutation+ was associated with MKI67, vascular invasion and tumor recurrence in TNBC. The presence of CD117 and TP53 missense mutations together in the primary tumors was an independent prognostic factor for survival of patients with TNBC.
Collapse
Affiliation(s)
- Yanli Luo
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wentao Huang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Guang Liu
- Department of Vascular Surgery, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| |
Collapse
|
29
|
Wang C, Kar S, Lai X, Cai W, Arfuso F, Sethi G, Lobie PE, Goh BC, Lim LHK, Hartman M, Chan CW, Lee SC, Tan SH, Kumar AP. Triple negative breast cancer in Asia: An insider's view. Cancer Treat Rev 2017; 62:29-38. [PMID: 29154023 DOI: 10.1016/j.ctrv.2017.10.014] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/29/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
While tremendous improvement has been made for the treatment of breast cancers, the treatment of triple negative breast cancer (TNBC) still remains a challenge due to its aggressive characteristics and limited treatment options. Most of the studies on TNBC were conducted in Western population and TNBC is reported to be more frequent in the African women. This review encapsulates the studies conducted on TNBC patients in Asian population and elucidates the similarities and differences between these two regions. The current treatment of TNBC includes surgery, radiotherapy and chemotherapy. In addition to the current chemotherapies, which mainly include cytotoxic agents, such as taxanes and anthracyclines, many clinical trials are investigating the potential use of other chemotherapy drugs, targeted therapeutics and combinational therapies to treat TNBC. Moreover, this review also integrates the studies involving novel markers, which will help us to dissect the pathologic process of TNBC and in turn facilitate the development of better treatment strategies to combat TNBC.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shreya Kar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xianning Lai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wanpei Cai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Peter E Lobie
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School at Shenzhen, Shenzhen, China
| | - Boon C Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; NUS Immunology Program, National University of Singapore, Singapore
| | - Mikael Hartman
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; Department of Surgery, National University Cancer Institute, National University Health System, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Ching W Chan
- Department of Surgery, National University Cancer Institute, National University Health System, Singapore
| | - Soo C Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore
| | - Sing H Tan
- Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore; National University Cancer Institute, National University Health System, Singapore; OncoCare Cancer Centre, Gleneagles Medical Centre, Singapore.
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National University Cancer Institute, National University Health System, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
30
|
Li Z, Yin S, Zhang L, Liu W, Chen B. Prognostic value of reduced E-cadherin expression in breast cancer: a meta-analysis. Oncotarget 2017; 8:16445-16455. [PMID: 28147315 PMCID: PMC5369975 DOI: 10.18632/oncotarget.14860] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/16/2017] [Indexed: 01/12/2023] Open
Abstract
The prognostic value of E-cadherin expression in patients with breast cancer has been studied for years, yet results remain controversial. We thus performed a comprehensive evaluation of the association between E-cadherin expression and prognosis through a meta-analysis. The databases PubMed, Embase and Cochrane Library were searched. A total of 7,353 patients from 33 studies were subject to final analysis. The results showed there was a significant association between reduced expression of E-cadherin and overall survival (OS) (HR 1.79, 95% CI 1.41–2.27) and disease-free survival (DFS) (HR 1.62, 95% CI 1.31–1.99) in breast cancer. Downregulated expression of E-cadherin significantly correlated with tumor histological grade (OR 1.44, 95% CI 1.06–1.96), TNM stage (OR 2.44, 95% CI 1.75–3.41), tumor size (OR 1.38, 95% CI 1.18–1.60), lymph node status (OR 1.55, 95% CI 1.15–2.10), and progesterone receptor status (OR 1.44, 95% CI 1.10–1.88).This meta-analysis suggested that reduced E-cadherin expression might be a predictor of a poorer prognosis and could be a potentially new gene therapy target for breast cancer patients.
Collapse
Affiliation(s)
- Zhan Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Songcheng Yin
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Zhang
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Weiguang Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
31
|
Adamo B, Ricciardi GRR, Ieni A, Franchina T, Fazzari C, Sanò MV, Angelico G, Michele C, Tuccari G, Adamo V. The prognostic significance of combined androgen receptor, E-Cadherin, Ki67 and CK5/6 expression in patients with triple negative breast cancer. Oncotarget 2017; 8:76974-76986. [PMID: 29100362 PMCID: PMC5652756 DOI: 10.18632/oncotarget.20293] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/27/2017] [Indexed: 12/29/2022] Open
Abstract
Background Triple Negative Breast Cancer (TNBC) represents a heterogeneous group of tumors with poor prognosis owing to aggressive tumor biology and lack of targeted therapies. No clear prognostic biomarkers have been identified to date for this subgroup. Materials and Methods In this retrospective study we evaluated the prognostic role of 4 different molecular determinants, including androgen receptor (AR), E-cadherin (CDH1), Ki67 index, and basal cytokeratins (CKs) 5/6, in a cohort of 99 patients with TNBC. All patients received neo/adjuvant chemotherapy (mostly anthracycline/taxane-based). Immunohistochemistry (IHC) was performed in formalin-fixed paraffin-embedded primary tumor samples. CDH1 expression was considered positive as ≥ 30% of the membrane cells staining. AR positivity was defined as > 10% of positive tumor cells. High Ki67 was defined as ≥20% positive tumor cells. CK5/6 expression was judged positive if the score was ≥1. Results The absence of AR expression was significantly associated with highly undifferentiated tumors. Univariate analyses showed that lack of expression of CDH1, tumor size and nodal status were significantly correlated with worse RFS and OS (p< 0.05). AR expression and low Ki67 showed a trend towards better RFS and OS. Patients with absent CK5/6 expression in univariate and multivariate analyses had poorer RFS (p=0.02 and p=0.002, respectively) and OS (p=0.05 and p=0.02, respectively). Multivariate analysis showed an independent association between CDH1 expression and better RFS and OS (p< 0.05) beyond tumor size, nodal status, and grade. The Kaplan-Meier curves showed that patients with AR and CDH1 negative expression and high Ki-67 levels have a significant correlation with poor outcome. Conclusions Our study supports the use of IHC expression of AR, CDH1, Ki67, and CK5/6 as prognostic markers in TNBCs and suggests a link between their expression and prognosis and may help to stratify TNBC patients in different prognostic classes.
Collapse
Affiliation(s)
- Barbara Adamo
- Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain
| | | | - Antonio Ieni
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", Section of Pathology, University of Messina, AOU Policlinico "G. Martino" Messina, Italy
| | - Tindara Franchina
- Medical Oncology Unit A.O. Papardo & Department of Human Pathology University of Messina, Messina, Italy
| | - Carmine Fazzari
- Pathology Unit, Humanitas Center of Oncology, Catania, Italy
| | - Maria Vita Sanò
- Medical Oncology, Humanitas Catania Oncology Center, Catania, Italy
| | - Giuseppe Angelico
- G. F. Ingrassia Department, Section of Anatomic Pathology, University Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| | - Caruso Michele
- Medical Oncology, Humanitas Catania Oncology Center, Catania, Italy
| | - Giovanni Tuccari
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", Section of Pathology, University of Messina, AOU Policlinico "G. Martino" Messina, Italy
| | - Vincenzo Adamo
- Medical Oncology Unit A.O. Papardo & Department of Human Pathology University of Messina, Messina, Italy
| |
Collapse
|
32
|
Chen PN, Yang SF, Yu CC, Lin CY, Huang SH, Chu SC, Hsieh YS. Duchesnea indica extract suppresses the migration of human lung adenocarcinoma cells by inhibiting epithelial-mesenchymal transition. ENVIRONMENTAL TOXICOLOGY 2017; 32:2053-2063. [PMID: 28371048 DOI: 10.1002/tox.22420] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 06/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a process through which epithelial cells are transformed into mesenchymal cells; EMT diminishes cell polarity and cell-cell adhesion in cancer cells, leading to enhanced migratory and invasive properties. In this experiment, zymography, cell invasion, and migration assays were performed. Results indicated that Duchesnea indica extracts (DIE) inhibited highly metastatic A549 and H1299 cells by reducing the secretions of matrix metalloproteinase-2 and urokinase-type plasminogen activator. Cell adhesion assay also demonstrated that DIE reduced the cell adhesion properties. Western blot analysis showed that DIE down-regulated the expression of N-cadherin, fibronectin, and vimentin, which are mesenchymal markers, and enhanced that of E-cadherin, which is an epithelial marker. In vivo study showed that tumor growth was significantly reduced in BALB/c nude mouse xenograft model administered with oral gavage of DIE. Therefore, DIE could be exhibits potential as a phytochemical-based platform for prevention and treatment of lung cancer. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2053-2063, 2017.
Collapse
Affiliation(s)
- Pei-Ni Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Chia Yu
- Institute of Oral Science, School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chin-Yin Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Han Huang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Chen Chu
- Institute and Department of Food Science, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Yih-Shou Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
33
|
Handa T, Katayama A, Yokobori T, Yamane A, Horiguchi J, Kawabata-Iwakawa R, Rokudai S, Bao P, Gombodorj N, Altan B, Kaira K, Asao T, Kuwano H, Nishiyama M, Oyama T. Caspase14 expression is associated with triple negative phenotypes and cancer stem cell marker expression in breast cancer patients. J Surg Oncol 2017; 116:706-715. [PMID: 28570747 DOI: 10.1002/jso.24705] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/06/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND OBJECTIVES The Caspase14 (CASP14) was reported that the low expression of CASP14 in ovarian cancer and colon cancer was associated with cancer progression, on the other hand, that the CASP14 expression in breast cancer was higher than that of non-cancerous tissues. The purpose of this study is to determine the clinical significance of CASP14 in breast cancer. METHODS We performed immunohistochemistry for CASP14, ER, PgR, HER2, Ki67, EGFR, CK5/6, CD44, CD24, ALDH1, claudins, and androgen receptor in 222 breast cancer patients including 55 TNBC cases, and evaluated the relationship of CASP14, above-mentioned markers, and prognosis. Using public microarray database of breast cancer, the prognostic value of CASP14 was calculated. RESULTS High CASP14 expression was significantly associated with TNBC subtype (P = 0.015), nuclear grade (P = 0.006), Ki67, EGFR (P < 0.001, P = 0.016), ALDH1, CD44 and CD24 (P < 0.001, P < 0.001, P = 0.001) in 222 breast cancer cases, and the high expression of claudin1 (P = 0.017), and androgen receptor (P = 0.002) in TNBC cases was related to the high CASP14. According to the public database, survival in the high CASP14 breast cancer patients was shorter than low CASP14 patients. CONCLUSIONS High CASP14 expression is a marker of breast cancer aggressiveness in association with proliferation, TNBC phenotype, and cancer stemness.
Collapse
Affiliation(s)
- Tadashi Handa
- Department of Diagnostic Pathology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Ayaka Katayama
- Department of Diagnostic Pathology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Takehiko Yokobori
- Research Program for Omics-Based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Japan
| | - Arito Yamane
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Jun Horiguchi
- Integrative Center of General Surgery, Gunma University Hospital, Maebashi, Japan
| | - Reika Kawabata-Iwakawa
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Susumu Rokudai
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Pinjie Bao
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Navchaa Gombodorj
- Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Bolag Altan
- Department of Oncology Clinical Development, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Kyoichi Kaira
- Department of Oncology Clinical Development, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Takayuki Asao
- Big Data Center for Integrative Analysis, Gunma University Initiative for Advance Research (GIAR), Maebashi, Japan
| | - Hiroyuki Kuwano
- Integrative Center of General Surgery, Gunma University Hospital, Maebashi, Japan
| | - Masahiko Nishiyama
- Research Program for Omics-Based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research (GIAR), Maebashi, Japan.,Department of Molecular Pharmacology and Oncology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University, Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
34
|
Li X, Oprea-Ilies GM, Krishnamurti U. New Developments in Breast Cancer and Their Impact on Daily Practice in Pathology. Arch Pathol Lab Med 2017; 141:490-498. [DOI: 10.5858/arpa.2016-0288-sa] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Advances in research have transformed our understanding of breast cancers and have altered the daily practice of pathology. Theranostic evaluations performed by pathologists are now critical in triaging the patients into appropriate treatment groups, as are new guidelines that were recently established for the evaluation of HER2/neu gene amplification. Emerging molecular classifications of breast cancers bring novel perspectives to the assessment of individual cases, and opportunities for better treatments. Molecular studies have particularly shed light on distinct biological subsets of triple-negative breast cancers, for which new targeted therapies are being developed. The prognostic and therapeutic utility of new histopathologic parameters, such as tumor-infiltrating lymphocytes, are also being elucidated, and new protocols have been devised for the pathologic evaluation of breast specimens that have undergone neoadjuvant treatment. Novel clinical practices, such as radioactive seed localization, also affect the way breast specimens are processed and evaluated. In this brief review, we highlight the developments that are most relevant to pathology and are changing or could potentially impact our daily practice.
Collapse
Affiliation(s)
| | | | - Uma Krishnamurti
- From the Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Gerogia
| |
Collapse
|
35
|
Luo J, Jin J, Yang F, Sun Z, Zhang W, Shi Y, Xu J, Guan X. The Correlation Between PARP1 and BRCA1 in AR Positive Triple-negative Breast Cancer. Int J Biol Sci 2016; 12:1500-1510. [PMID: 27994514 PMCID: PMC5166491 DOI: 10.7150/ijbs.16176] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) lacks estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER-2) expression and thus cannot benefit from conventional hormonal or anti-HER2 targeted therapies. Anti-androgen therapy has shown a certain effect on androgen receptor (AR) positive TNBC. The emerging researches have proved that poly (ADP-ribose) polymerase (PARP) inhibitor is effective in BRCA1-deficient breast cancers. We demonstrated that combination of AR antagonist (bicalutamide) and PARP inhibitor (ABT-888) could inhibit cell viability and induce cell apoptosis significantly whatever in vitro or in vivo setting in AR-positive TNBC. Previous studies have proved that both BRCA1 and PARP1 have close connections with AR in prostate cancer. We explored the correlation among AR, PARP1 and BRCA1 in TNBC for the first time. After BRCA1 overexpression, the expression of AR and PARP1 were decreased in mRNA and protein levels. Additionally, AR positively regulated PARP1 while PARP1 also up-regulated AR expression in vitro. We also confirmed BRCA1 expression was negatively correlated with AR and PARP1 in TNBC patients using a tissue microarray with TNBC patient samples. These results suggest that the combination of bicalutamide and PARP inhibitor may be a potential strategy for TNBC patients and merits further evaluation.
Collapse
Affiliation(s)
- Jiayan Luo
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Juan Jin
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Zijia Sun
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Wenwen Zhang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yaqin Shi
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jing Xu
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| |
Collapse
|
36
|
Shen T, Zhang K, Siegal GP, Wei S. Prognostic Value of E-Cadherin and β-Catenin in Triple-Negative Breast Cancer. Am J Clin Pathol 2016; 146:603-610. [PMID: 27780797 DOI: 10.1093/ajcp/aqw183] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES To analyze the expression of E-cadherin and β-catenin in triple-negative breast cancer (TNBC) to assess their prognostic significance. METHODS The expression of E-cadherin and β-catenin was examined semiquantitatively and correlated with other pathologic factors and survival outcomes. RESULTS Of 72 consecutive TNBCs, 56% showed reduced membranous expression of E-cadherin or β-catenin, with a strong correlation to each other. Of the clinicopathologic factors analyzed, tumor size and nodal status were significantly associated with overall survival and disease-specific survival, while the latter remained an independent factor by multivariate analysis. Reduced E-cadherin and β-catenin were both significantly associated with a poor overall survival and disease-specific survival by univariate and multivariate analyses. CONCLUSIONS E-cadherin and β-catenin expression provides discriminative prognostic power independent of conventional pathologic factors, thus further reinforcing the important role of cell adhesion molecules in the process of tumor metastasis, especially in TNBC.
Collapse
Affiliation(s)
- Tiansheng Shen
- From the Department of Pathology, the University of Alabama at Birmingham
| | - Kui Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton
| | - Gene P Siegal
- From the Department of Pathology, the University of Alabama at Birmingham
| | - Shi Wei
- From the Department of Pathology, the University of Alabama at Birmingham
| |
Collapse
|
37
|
Ieni A, Barresi V, Ricciardi GRR, Adamo B, Adamo V, Tuccari G. Prognostic value of androgen receptor expression in triple negative breast carcinomas: personal experience and comments on a review about "Triple-negative breast cancer: treatment challenges and solutions" by Collignon et al. BREAST CANCER-TARGETS AND THERAPY 2016; 8:157-9. [PMID: 27578997 PMCID: PMC4998015 DOI: 10.2147/bctt.s113577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Antonio Ieni
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", Section of Pathology, University of Messina, AOU "Policlinico G Martino"
| | - Valeria Barresi
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", Section of Pathology, University of Messina, AOU "Policlinico G Martino"
| | - Giuseppina Rosaria Rita Ricciardi
- Medical Oncology Unit AOOR Papardo-Piemonte, Department of Human Pathology of Adult And Evolutive Age "Gaetano Barresi", University of Messina, Messina, Italy
| | | | - Vincenzo Adamo
- Medical Oncology Unit AOOR Papardo-Piemonte, Department of Human Pathology of Adult And Evolutive Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", Section of Pathology, University of Messina, AOU "Policlinico G Martino"
| |
Collapse
|
38
|
Collina F, Cerrone M, Peluso V, Laurentiis MD, Caputo R, Cecio RD, Liguori G, Botti G, Cantile M, Bonito MD. Downregulation of androgen receptor is strongly associated with diabetes in triple negative breast cancer patients. Am J Transl Res 2016; 8:3530-3539. [PMID: 27648143 PMCID: PMC5009405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/03/2016] [Indexed: 06/06/2023]
Abstract
Developing of personalized therapies for Triple Negative Breast Cancer (TNBC) requires a more detailed knowledge of its biology and a correct stratification of molecular subtypes. Androgen Receptor (AR) is expressed in a large part of TNBCs but its prognostic role in this Breast Cancer (BC) subtype is highly debated. In this study, we analyzed AR expression in a series of 238 TNBCs and correlated its expression with clinical-pathological features, survival, and metabolic profile. We showed a consistent association between AR expression and a better prognosis of TNBC patients, while its downregulation appeared strongly associated with diabetic disease. Since a recent prospective study reported a lower BC risk in diabetic women treated with drugs able to reduce circulating levels of glucose compared with non-diabetic woman, and in vitro studies showed that AR level are regulated directly by hyperglycemia, we speculate on the perspective of new integrated therapies for TNBC.
Collapse
Affiliation(s)
- Francesca Collina
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Margherita Cerrone
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Valentina Peluso
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Michelino De Laurentiis
- Department of Breast Surgery and Cancer Prevention, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Roberta Caputo
- Department of Breast Surgery and Cancer Prevention, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Giuseppina Liguori
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Monica Cantile
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| | - Maurizio Di Bonito
- Pathology Unit, Istituto Nazionale Tumori Fondazione “G. Pascale”via Mariano Semmola, 80131 Napoli, Italy
| |
Collapse
|
39
|
Shukla GC, Plaga AR, Shankar E, Gupta S. Androgen receptor-related diseases: what do we know? Andrology 2016; 4:366-81. [PMID: 26991422 DOI: 10.1111/andr.12167] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 01/09/2023]
Abstract
The androgen receptor (AR) and the androgen-AR signaling pathway play a significant role in male sexual differentiation and the development and function of male reproductive and non-reproductive organs. Because of AR's widely varied and important roles, its abnormalities have been identified in various diseases such as androgen insensitivity syndrome, spinal bulbar muscular atrophy, benign prostatic hyperplasia, and prostate cancer. This review provides an overview of the function of androgens and androgen-AR mediated diseases. In addition, the diseases delineated above are discussed with respect to their association with mutations and other post-transcriptional modifications in the AR. Finally, we present an introduction to the potential therapeutic application of most recent pharmaceuticals including miRNAs in prostate cancer that specifically target the transactivation function of the AR at post-transcriptional stages.
Collapse
Affiliation(s)
- G C Shukla
- Center of Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA.,Department of Biological Sciences, Cleveland State University, Cleveland, OH, USA
| | - A R Plaga
- Center of Gene Regulation in Health and Disease, Cleveland State University, Cleveland, OH, USA.,Department of Biological Sciences, Cleveland State University, Cleveland, OH, USA
| | - E Shankar
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, OH, USA
| | - S Gupta
- Department of Urology, Case Western Reserve University & University Hospitals Case Medical Center, Cleveland, OH, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA.,Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH, USA.,Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, USA
| |
Collapse
|
40
|
Abstract
Objective: Ki-67 plays an important function in cell division, but its exact role is still unknown. Moreover, few works regarding its overall function were published. The present study evaluated the clinical significance of Ki-67 index as a prognostic marker and predictor of recurrence in different molecular subtypes of breast cancer. The relationship of Ki-67 index with different clinicopathological factors was also analyzed. Methods: Ki-67 index was measured in 107 cases of primary breast cancer from 2010-2012. These patients were evaluated for estrogen receptor, progesterone receptor, and HER2. Ki-67 was divided according to percentage levels: < 15% and > 15%. Follow-up ranged from 32 months up to 6 years. Results: Approximately 44, 23, 15, and 25 cases were grouped as luminal A, luminal B, HER2 subtype, and triple-negative (TN), respectively. No luminal A patients showed Ki-67 level higher than 15%, and their recurrence was 20%. In luminal B group, Ki-67 level higher than 15% was observed in 69% of patients, and recurrence was 39%. In HER2 subtype, Ki-67 was higher than 15% in 34% of cases, and recurrence was 40%. In triple-negative cases, Ki-67 was higher than 15% in 60% of cases, and recurrence was detected in 32% of patients. Patients with Ki-67 less than 15% displayed better overall survival than those with Ki-67 higher than 15% (P = 0.01). Patients with Ki-67 higher than 15% exhibited higher incidence of metastasis and recurrence than those with Ki-67 less than 15% (P = 0.000).
Conclusions: Ki-67 may be considered as a valuable biomarker in breast cancer patients.
Collapse
Affiliation(s)
- Nahed A Soliman
- Department of Pathology, Faculty of Medicine, University of Helwan, Helwan 11795, Egypt
| | - Shaimaa M Yussif
- Department of Pathology, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt
| |
Collapse
|
41
|
Chen L, Yang L, Qiao F, Hu X, Li S, Yao L, Yang XL, Shao ZM. High Levels of Nucleolar Spindle-Associated Protein and Reduced Levels of BRCA1 Expression Predict Poor Prognosis in Triple-Negative Breast Cancer. PLoS One 2015; 10:e0140572. [PMID: 26485712 PMCID: PMC4618922 DOI: 10.1371/journal.pone.0140572] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/26/2015] [Indexed: 12/13/2022] Open
Abstract
Purpose Nucleolar spindle-associated protein (NuSAP1) is an important mitosis-related protein, and aberrant NuSAP1 expression is associated with abnormal spindles and mitosis. This study investigated the prognostic value of NuSAP1 in breast cancer. Methods Two sets of tissue microarrays (TMAs) that included samples from 450 breast cancer patients were constructed, of which 250 patients were training set and the other 200 patients were validation set. Immunohistochemical staining was performed to determine the NuSAP1 levels. A Kaplan-Meier analysis was used to estimate the prognostic value of NuSAP1 in breast cancer. A stepwise Cox analysis was performed to construct a risk-prediction model for triple-negative breast cancer (TNBC). All statistical analysis was performed with SPSS software. Results There were 108 (43.5%) and 88 (44.0%) patients expressed NuSAP1 in the training set and validation set respectively. High levels of NuSAP1 expression were related to poor disease-free survival (DFS) in both training (P = 0.028) and validation (P = 0.006) cohorts, particularly in TNBC. With combination of two cohorts, both NuSAP1 (HR = 4.136, 95% CI: 1.956–8.747, P < 0.001) and BRCA1 (HR = 0.383, 95% CI: 0.160–0.915, P = 0.031) were independent prognostic indicators of DFS in TNBC. A receiver operating characteristic (ROC) analysis revealed that the combination of NuSAP1 and BRCA1 significantly improved the prognostic power compared with the traditional model (0.778 versus 0.612, P < 0.001). Conclusions Our study confirms the prognostic value of NuSAP1 in breast cancer. The combination of NuSAP1 and BRCA1 could improve the DFS prediction accuracy in TNBC.
Collapse
Affiliation(s)
- Li Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liu Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Feng Qiao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xin Hu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Shan Li
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Ling Yao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- * E-mail: (ZMS); (L Yao)
| | - Xue-Li Yang
- Department of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- * E-mail: (ZMS); (L Yao)
| |
Collapse
|
42
|
Correction: Androgen Receptor (AR), E-Cadherin, and Ki-67 as Emerging Targets and Novel Prognostic Markers in Triple-Negative Breast Cancer (TNBC) Patients. PLoS One 2015; 10:e0132647. [PMID: 26135303 PMCID: PMC4489771 DOI: 10.1371/journal.pone.0132647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|