101
|
Naimi A, Soltan M, Amjadi E, Goli P, Kefayat A. Androgen Receptor Expression and Its Correlation with Clinicopathological Parameters in Iranian Patients with Triple Negative Breast Cancer. IRANIAN JOURNAL OF PATHOLOGY 2020; 15:239-244. [PMID: 32754220 PMCID: PMC7354074 DOI: 10.30699/ijp.2020.112819.2224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/11/2020] [Indexed: 11/12/2022]
Abstract
BACKGROUND & OBJECTIVE Our knowledge about correlation of androgen receptor expression and clinicopathological properties of triple-negative breast cancer (TNBC) patients is inadequate, particularly in the Iranian population. The main aim of the present study was to assess the AR expression in TNBC Iranian patients and evaluate its correlation with their clinicopathological parameters. METHODS Herein, 76 TNBC patients were evaluated for the AR expression by immunohistochemistry. The slides' staining intensity was investigated according to the average degree of nuclear staining and sub-classified into negative (0), weak (1), moderate (2), or strong (3). Subsequently, the positive cells percentage for each slide was assessed and sub-classified into <25% (1), 25-50% (2), 50-75% (3), and >75% (4). The aggregation of these two scores was used as the final score ranging from 0 to 7. While 4-7 scores were selected as positive, the others were included in the AR-negative expression group. Fisher's exact test was used to analyze the AR expression correlation with the clinicopathological parameters. RESULTS Positive immunoreactivity for AR was observed in 8 out of 76 (11%) specimens. No-correlation (P>0.05) was observed between the AR expression and grade, stage, lymph node status, and Ki-67 level. The AR-positive patients exhibited older age at the time of diagnosis (P=0.0339) and larger tumor size (P=0.0224) in comparison with the AR-negative patients. Low percentage of TNBC patients expressed AR and no significant correlation was observed between its expression and most of the clinicopathological parameters. CONCLUSION AR may not be a suitable biomarker and treatment target for the Iranian patients with TNBC.
Collapse
Affiliation(s)
- Azar Naimi
- Department. of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Soltan
- Department. of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Amjadi
- Department. of Pathology, Poursina Hakim Digestive Diseases Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Goli
- Department. of Pathology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
102
|
Hadgu E, Seifu D, Tigneh W, Bokretsion Y, Bekele A, Abebe M, Sollie T, Karlsson C, Karlsson MG. Distribution and characteristics of androgen receptor (AR) in breast cancer among women in Addis Ababa, Ethiopia: A cross sectional study. PLoS One 2020; 15:e0232519. [PMID: 32374753 PMCID: PMC7202607 DOI: 10.1371/journal.pone.0232519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
Evaluation of the role of androgen receptor (AR) in the biology of breast cancer is an emerging area of research. There are compelling evidences that AR expression may be used to further refine breast cancer molecular subtyping with prognostic and therapeutic implications. Many studies indicated co-expression of AR with the hormonal receptors in breast cancer has a favorable prognosis. AR is also investigated by many researchers as a potential therapeutic target in treatment of breast cancer. Studies on the frequency and distribution of AR in breast cancer among Africans is barely available. Given the heightened interest to understand its role in breast cancer, we determined AR expression and assessed its association with clinicopathological parameters among Ethiopian women. In this study, 112 newly diagnosed patient with invasive breast cancer at Tikur Anbessa Specialized Hospital were enrolled. Immunohistochemical assessment of AR, ER, PR, Ki67 and HER2 were performed using tissue microarrays (TMA) constructed from their primary tumor block. Out of the 112 participants, 91 (81%) were positive for AR expression and the remaining 21 participants (19%) were negative for AR expression. Expression of AR in ER+, HER2+ and TNBC cases were 93%, 83% and 48% respectively. Our study reveals AR is expressed in a significant number of breast cancers patients and this may indicate that breast cancers cases in Ethiopia have favorable prognosis and could benefit from progresses in AR targeted treatments. Since AR expression has important consequences on the prognosis and treatment of breast cancer, further studies with an increased number of participants is necessary to confirm our reports.
Collapse
Affiliation(s)
- Endale Hadgu
- Department of Biochemistry, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
- * E-mail:
| | - Daniel Seifu
- Department of Biochemistry, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Wondemagegnhu Tigneh
- Department of Oncology, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Yonas Bokretsion
- Department of Pathology, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abebe Bekele
- Department of Surgery, School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Markos Abebe
- Armauer Hansen research Institute (AHRI), Addis Ababa, Ethiopia
| | - Thomas Sollie
- School of Medical Sciences, Orebro University, Orebro, Sweden
| | | | | |
Collapse
|
103
|
Yoon S, Won HS, Kang K, Qiu K, Park WJ, Ko YH. Hormone Receptor-Status Prediction in Breast Cancer Using Gene Expression Profiles and Their Macroscopic Landscape. Cancers (Basel) 2020; 12:cancers12051165. [PMID: 32380759 PMCID: PMC7281553 DOI: 10.3390/cancers12051165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/26/2020] [Accepted: 05/03/2020] [Indexed: 11/24/2022] Open
Abstract
The cost of next-generation sequencing technologies is rapidly declining, making RNA-seq-based gene expression profiling (GEP) an affordable technique for predicting receptor expression status and intrinsic subtypes in breast cancer patients. Based on the expression levels of co-expressed genes, GEP-based receptor-status prediction can classify clinical subtypes more accurately than can immunohistochemistry (IHC). Using data from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA BRCA) and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets, we identified common predictor genes found in both datasets and performed receptor-status prediction based on these genes. By assessing the survival outcomes of patients classified using GEP- or IHC-based receptor status, we compared the prognostic value of the two methods. We found that GEP-based HR prediction provided higher concordance with the intrinsic subtypes and a stronger association with treatment outcomes than did IHC-based hormone receptor (HR) status. GEP-based prediction improved the identification of patients who could benefit from hormone therapy, even in patients with non-luminal breast cancer. We also confirmed that non-matching subgroup classification affected the survival of breast cancer patients and that this could be largely overcome by GEP-based receptor-status prediction. In conclusion, GEP-based prediction provides more reliable classification of HR status, improving therapeutic decision making for breast cancer patients.
Collapse
Affiliation(s)
- Seokhyun Yoon
- Department of Electronics Eng., College of Engineering, Dankook University, Yongin-si 16890, Korea; (S.Y.); (K.Q.)
| | - Hye Sung Won
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan-si 31116, Korea;
| | - Kexin Qiu
- Department of Electronics Eng., College of Engineering, Dankook University, Yongin-si 16890, Korea; (S.Y.); (K.Q.)
| | - Woong June Park
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan-si 31116, Korea;
| | - Yoon Ho Ko
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
104
|
Association between Lymph Node Status and Expression Levels of Androgen Receptor, miR-185, miR-205, and miR-21 in Breast Cancer Subtypes. Int J Breast Cancer 2020; 2020:3259393. [PMID: 32373367 PMCID: PMC7195641 DOI: 10.1155/2020/3259393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/25/2020] [Indexed: 01/04/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer among women. Difficulties in treating breast cancer are associated with the occurrence of metastases at early stages of disease, leading to its further progression. Recent studies have shown that changes in androgen receptor (AR) and microRNAs' expressions are associated with mammary gland carcinogenesis, in particular, with the formation of metastases. Thus, to identify novel metastatic markers, we evaluated the expression levels of AR; miR-185 and miR-205, both of which have been confirmed to target AR; and miR-21, transcription of which is regulated by AR, in breast cancer samples (n = 89). Here, we show that the molecular subtypes of breast cancer differ in the expression profiles of AR and AR-associated microRNAs. In addition, the expression of AR and these microRNAs may depend on the expression of PR, ER, and HER2 receptors. Our results show that the possibility of using AR and microRNAs as markers depends on the tumor subtype: a decrease in AR expression may be the marker for the presence of lymph node metastases in patients with HER2-positive subtypes of breast cancer, and disturbance of miR-205, miR-185, and miR-21 expressions may be the marker in patients with a luminal B HER2-positive subtype. Cases with metastases in this type of breast cancer are characterized by a higher level of miR-205 and a lower level of miR-185 and miR-21 in tumor tissues compared to nonmetastatic cases. A decrease in the miR-185 level is also associated with lymph node metastasis in luminal B HER2-negative breast cancer. Thus, the expression levels of AR, miR-185, miR-205, and miR-21 can serve as markers to predict cancer spread to the lymph node in luminal B- and HER2-positive subtypes of breast cancer.
Collapse
|
105
|
Integrin αvβ3 in the Mediating Effects of Dihydrotestosterone and Resveratrol on Breast Cancer Cell Proliferation. Int J Mol Sci 2020; 21:ijms21082906. [PMID: 32326308 PMCID: PMC7216104 DOI: 10.3390/ijms21082906] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Hormones and their receptors play an important role in the development and progression of breast cancer. Hormones regulate the proliferation of breast cancer cells through binding between estrogen or progestins and steroid receptors that may reside in the cytoplasm or be transcriptionally activated as steroid–protein nuclear receptor complexes. However, receptors for nonpeptide hormones also exist in the plasma membrane. Via those receptors, hormones are able to stimulate breast cancer cell proliferation when activated. Integrins are heterodimeric structural proteins of the plasma membrane. Their primary functions are to interact with extracellular matrix proteins and growth factors. Recently, integrin αvβ3 has been identified as a receptor for nonpeptide hormones, such as thyroid hormone and dihydrotestosterone (DHT). DHT promotes the proliferation of human breast cancer cells through binding to integrin αvβ3. A receptor for resveratrol, a polyphenol stilbene, also exists on this integrin in breast cancer cells, mediating the anti-proliferative, pro-apoptotic action of the compound in these cells. Unrelated activities of DHT and resveratrol that originate at integrin depend upon downstream stimulation of mitogen-activated protein kinase (MAPK, ERK1/2) activity, suggesting the existence of distinct, function-specific pools of ERK1/2 within the cell. This review will discuss the features of these receptors in breast cancer cells, in turn suggesting clinical applications that are based on the interactions of resveratrol/DHT with integrin αvβ3 and other androgen receptors.
Collapse
|
106
|
Wang R, Huang K. CCL11 increases the proportion of CD4+CD25+Foxp3+ Treg cells and the production of IL‑2 and TGF‑β by CD4+ T cells via the STAT5 signaling pathway. Mol Med Rep 2020; 21:2522-2532. [PMID: 32323817 PMCID: PMC7185287 DOI: 10.3892/mmr.2020.11049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/27/2020] [Indexed: 12/20/2022] Open
Abstract
CD4+ regulatory T (Treg) cells are associated with immune tolerance and antitumor immunosuppression. The aim of the present study was to investigate the role and molecular mechanism of C-C motif chemokine ligand 11 (CCL11) in the regulation of Treg cells from patients with breast cancer (BC) and healthy individuals in vitro, and from tumor-bearing mice in vivo. CD4+ T cells isolated from patients with BC or healthy individuals were incubated with anti-CCL11 neutralizing antibodies or recombinant human CCL11 protein, in the presence or absence of a STAT5 inhibitor. The serum CCL11 level and proportion of Treg cells characterized as CD4+CD25+forkhead box P3+ (Foxp3) among the CD4+ T cells in patients with BC and healthy individuals were analyzed by ELISA and flow cytometry, respectively. CCL11, C-C motif chemokine receptor 3 (CCR3), Foxp3, phosphorylated-STAT5 and STAT5 expression levels were determined by western blotting. The serum CCL11 level and the proportion of CD4+CD25+Foxp3+ Treg cells were significantly increased in patients with BC compared with healthy individuals. CCL11 blockade reduced the proportion of CD4+CD25+Foxp3+ Treg cells, the expression of CCR3 and Foxp3, and the level of STAT5 activation in tumor-associated CD4+ T cells, in a dose-dependent manner. CCL11 blockade also reduced the proportion of CD4+CD25+Foxp3+ Treg cells and the serum levels of interleukin (IL)-2 and transforming growth factor (TGF)-β1 in tumor-bearing mice. The recombinant human CCL11 protein increased the proportion of CD4+CD25+Foxp3+ Treg cells, the expression of CCR3 and Foxp3, and the release of IL-2 and TGF-β1 in non-tumor-associated CD4+ T cells via the STAT5 signaling pathway. The results of the present study may aid in identifying therapeutics that could further modulate the immune system during BC.
Collapse
Affiliation(s)
- Rong Wang
- Department of Clinical Laboratory, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Keliang Huang
- Department of Clinical Laboratory, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
107
|
Testa U, Castelli G, Pelosi E. Breast Cancer: A Molecularly Heterogenous Disease Needing Subtype-Specific Treatments. Med Sci (Basel) 2020; 8:E18. [PMID: 32210163 PMCID: PMC7151639 DOI: 10.3390/medsci8010018] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/23/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly occurring cancer in women. There were over two-million new cases in world in 2018. It is the second leading cause of death from cancer in western countries. At the molecular level, breast cancer is a heterogeneous disease, which is characterized by high genomic instability evidenced by somatic gene mutations, copy number alterations, and chromosome structural rearrangements. The genomic instability is caused by defects in DNA damage repair, transcription, DNA replication, telomere maintenance and mitotic chromosome segregation. According to molecular features, breast cancers are subdivided in subtypes, according to activation of hormone receptors (estrogen receptor and progesterone receptor), of human epidermal growth factors receptor 2 (HER2), and or BRCA mutations. In-depth analyses of the molecular features of primary and metastatic breast cancer have shown the great heterogeneity of genetic alterations and their clonal evolution during disease development. These studies have contributed to identify a repertoire of numerous disease-causing genes that are altered through different mutational processes. While early-stage breast cancer is a curable disease in about 70% of patients, advanced breast cancer is largely incurable. However, molecular studies have contributed to develop new therapeutic approaches targeting HER2, CDK4/6, PI3K, or involving poly(ADP-ribose) polymerase inhibitors for BRCA mutation carriers and immunotherapy.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
108
|
Kalinina TS, Kononchuk VV, Sidorov SV, Gulyaeva LF. [Analysis of prolactin receptor expression in breast cancer subtypes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2020; 66:89-94. [PMID: 32116231 DOI: 10.18097/pbmc20206601089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Breast cancer (BC) is the most common cancer among women. It is known that the prolactin receptor (PRLR) may play a role in breast carcinogenesis, but the available data are often contradictory. To get a more complete picture of the relationship between the receptor and mammary gland carcinogenesis, we examined the association between changes in PRLR expression level and tumor subtype (and its main characteristics). To do this, using real-time PCR, we evaluated the level of PRLR mRNA in BC tissue samples and untransformed adjoining tissue samples (89 pairs). Since the androgen receptor (AR) has begun to be seen as a prognostic marker in breast cancer, we also evaluated the association between mRNA levels of AR and PRLR. We found a significant increase in PRLR expression in luminal subtypes; the highest level of PRLR mRNA was detected in luminal A subtype. In HER2-positive ER-, PR-negative BC, the PRLR mRNA level decreases in tumor tissues compared with untransformed tissues. High PRLR expression is also associated with smaller tumor size in luminal B HER2-negative subtype. In ER-, PR-negative tumors, PRLR expression is associated with AR expression: PRLR mRNA level is increased when AR mRNA level is reduced by more than 8 times in triple-negative tumors; in contrast, in HER2-positive subtype it decreases more significantly when AR expression is reduced by more than 3 times. A tendency towards an increase in PRLR expression with an increase in the AR mRNA level was also discovered in luminal subtypes. The level of PRLR expression depends on the age of patients. In luminal A, PRLR expression is higher in patients under 65 years. In contrast, in luminal B HER2-negative and triple-negative BC, reduced PRLR expression was observed in patients under the age of 40 years and under the age of 50 years, respectively. In this group of patients under the age of 40 years with luminal B HER2-negative BC, ER expression was also reduced (0-4 score according to the IHC assay). Thus, PRLR probably plays a different role in the development and progression of BC: in luminal A and luminal B HER2-positive subtypes PRLR may act as an oncogen, and in luminal B HER2-negative and ER-, PR-negative subtypes can play a tumor suppressor role.
Collapse
Affiliation(s)
- T S Kalinina
- Institute of Molecular Biology and Biophysics - Subdivision of Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - V V Kononchuk
- Institute of Molecular Biology and Biophysics - Subdivision of Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - S V Sidorov
- Novosibirsk State University, Novosibirsk, Russia; Municipal Clinical Hospital No.1, Novosibirsk, Russia
| | - L F Gulyaeva
- Institute of Molecular Biology and Biophysics - Subdivision of Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia; Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
109
|
Wahdan-Alaswad R, Liu B, Thor AD. Targeted lapatinib anti-HER2/ErbB2 therapy resistance in breast cancer: opportunities to overcome a difficult problem. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:179-198. [PMID: 35582612 PMCID: PMC9090587 DOI: 10.20517/cdr.2019.92] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/11/2022]
Abstract
Approximately 20% of invasive breast cancers have upregulation/gene amplification of the oncogene human epidermal growth factor receptor-2 (HER2/ErbB2). Of these, some also express steroid receptors (the so-called Luminal B subtype), whereas others do not (the HER2 subtype). HER2 abnormal breast cancers are associated with a worse prognosis, chemotherapy resistance, and sensitivity to selected anti-HER2 targeted therapeutics. Transcriptional data from over 3000 invasive breast cancers suggest that this approach is overly simplistic; rather, the upregulation of HER2 expression resulting from gene amplification is a driver event that causes major transcriptional changes involving numerous genes and pathways in breast cancer cells. Most notably, this includes a shift from estrogenic dependence to regulatory controls driven by other nuclear receptors, particularly the androgen receptor. We discuss members of the HER receptor tyrosine kinase family, heterodimer formation, and downstream signaling, with a focus on HER2 associated pathology in breast carcinogenesis. The development and application of anti-HER2 drugs, including selected clinical trials, are discussed. In light of the many excellent reviews in the clinical literature, our emphasis is on recently developed and successful strategies to overcome targeted therapy resistance. These include combining anti-HER2 agents with programmed cell death-1 ligand or cyclin-dependent kinase 4/6 inhibitors, targeting crosstalk between HER2 and other nuclear receptors, lipid/cholesterol synthesis to inhibit receptor tyrosine kinase activation, and metformin, a broadly inhibitory drug. We seek to facilitate a better understanding of new approaches to overcome anti-HER2 drug resistance and encourage exploration of two other therapeutic interventions that may be clinically useful for HER+ invasive breast cancer patients.
Collapse
Affiliation(s)
- Reema Wahdan-Alaswad
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO 80014, USA
| | - Bolin Liu
- Department of Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ann D Thor
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora CO 80014, USA
| |
Collapse
|
110
|
Genetic Variants Detected Using Cell-Free DNA from Blood and Tumor Samples in Patients with Inflammatory Breast Cancer. Int J Mol Sci 2020; 21:ijms21041290. [PMID: 32075053 PMCID: PMC7072950 DOI: 10.3390/ijms21041290] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/27/2022] Open
Abstract
We studied genomic alterations in 19 inflammatory breast cancer (IBC) patients with advanced disease using samples of tissue and paired blood serum or plasma (cell-free DNA, cfDNA) by targeted next generation sequencing (NGS). At diagnosis, the disease was triple negative (TN) in eleven patients (57.8%), ER+ Her2- IBC in six patients (31.6%), ER+ Her2+ IBC in one patient (5.3%), and ER- Her2+ IBC in one other patient (5.3%). Pathogenic or likely pathogenic variants were frequently detected in TP53 (47.3%), PMS2 (26.3%), MRE11 (26.3%), RB1 (10.5%), BRCA1 (10.5%), PTEN (10.5%) and AR (10.5%); other affected genes included PMS1, KMT2C, BRCA2, PALB2, MUTYH, MEN1, MSH2, CHEK2, NCOR1, PIK3CA, ESR1 and MAP2K4. In 15 of the 19 patients in which tissue and paired blood were collected at the same time point, 80% of the variants detected in tissue were also detected in the paired cfDNA. Higher concordance between tissue and cfDNA was found for variants with higher allele fraction in tissue (AFtissue ≥ 5%). Furthermore, 86% of the variants detected in cfDNA were also detected in paired tissue. Our study suggests that the genetic profile measured in blood cfDNA is complementary to that of tumor tissue in IBC patients.
Collapse
|
111
|
Salvi S, Bonafè M, Bravaccini S. Androgen receptor in breast cancer: A wolf in sheep’s clothing? A lesson from prostate cancer. Semin Cancer Biol 2020; 60:132-137. [DOI: 10.1016/j.semcancer.2019.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
|
112
|
Wardhani BW, Puteri MU, Watanabe Y, Louisa M, Setiabudy R, Kato M. TGF-β-Induced TMEPAI Attenuates the Response of Triple-Negative Breast Cancer Cells to Doxorubicin and Paclitaxel. J Exp Pharmacol 2020; 12:17-26. [PMID: 32158279 PMCID: PMC6986256 DOI: 10.2147/jep.s235233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Triple-negative breast cancer (TNBC) is a refractory type of breast cancer with poor prognosis and limited choice for treatment. Previous studies had shown that TNBC has high expressions of transmembrane prostate androgen-induced protein (TMEPAI). TMEPAI was known to be induced by TGF-β/Smad signaling and have tumorigenic functions that converting TGF-β from tumor suppressor to tumor promoter and inducing epithelial–mesenchymal transition (EMT). Therefore, we aimed to define the role of TMEPAI in triple-negative breast cancer cells treatment using several anti-cancers in the presence of TGF-β. Methods TMEPAI-knock out (KO) was carried out in a triple-negative breast cancer cell, BT549. TMEPAI editing was developed using the CRISPR-Cas9 system using two combinations of sgRNA to remove exon 4 of the TMEPAI gene entirely. Genotyping and proteomic analysis were performed to check the establishment of the TMEPAI-KO cells. Wild type (WT) and KO cells were used to determine inhibitory concentration 50% (IC50) of several anti-cancers: doxorubicin, cisplatin, paclitaxel, and bicalutamide in the presence of TGF-β treatment. Results KO cells were successfully established by completely removing the TMEPAI gene, which was proven in genomic and proteomic analysis. Further, in TMEPAI-KO cells, we found a significant reduction of IC50 for doxorubicin and paclitaxel, and minimal effects were seen for cisplatin and bicalutamide. Our findings suggest that TGF-β-induced TMEPAI attenuates the response of TNBC to doxorubicin and paclitaxel, but not to cisplatin and bicalutamide. Conclusion TGF-β induced TMEPAI contributes to the reduced response of TNBC treatment to doxorubicin and paclitaxel, but minimal on cisplatin and bicalutamide. Further study is needed to confirm our findings in other growth factor-induced cells, as well as in in vivo model.
Collapse
Affiliation(s)
- Bantari Wk Wardhani
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Meidi Utami Puteri
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukihide Watanabe
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Rianto Setiabudy
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences and Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
113
|
Huang Z, Wang SL, Huang QS, Li XD, Chen H, Lin JH. Clinicopathological value of ErbB2 gene and protein expression in osteochondroma. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2020; 54:34-41. [PMID: 32175895 DOI: 10.5152/j.aott.2020.01.484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The aim of this study was to investigate ErbB2 expression in osteochondroma and its relationship with clinicopathologic features of osteochondroma, so as to identify a new biomarker for the malignant transformation potential of osteochondroma. METHODS Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) were used to investigate the expression status of ErbB2 protein and gene in 30 osteochondroma tissues and 20 non-neoplastic bone tissues. The association of ErbB2 gene and protein expression with clinicopathological parameters of osteochondroma was analyzed by using the χ2 test and Fishers exact test. RESULTS ErbB2 protein was found to be over-expressed in 4 of 30 (13.3%) osteochondromas and 1 of 20 (5%) non-neoplastic bone samples, which were not statistically significant (p=0.336). However, 13 of the 30 (43.3%) osteochondromas showed ErbB2 gene amplification, which was failed to be observed in any of the non-neoplastic bone tissue. ErbB2 gene amplification in osteochondroma was significantly higher compared with that in non-neoplastic bone tissue (p=0.001). In addition, the ErbB2 gene amplification was closely associated with clinical pathological parameters of osteochondroma, including high expression of cellularity (p=0.001), presence of binucleated cells (p=0.001), nuclear pleomorphism (p=0.003), calcification (p=0.002), nodularity (p=0.002), necrosis (p=0.009) and cartilage thickness (p=0.026). The association of the gene amplification with other clinicopathological parameters of osteochondroma, including permeation of trabecular bone, cystic/mucoid changes, mitosis, radiographic appearance, cap volume and subtype of osteochondroma was not observed. The over-expression of ErbB2 protein was not found to be associated with the above stated clinical pathological parameters of osteochondroma. CONCLUSION ErbB2 gene amplification was associated with adverse clinicopathological status of osteochondroma and could serve as an index for malignant conversion of osteochondroma. Further research is required to verify the predictive values of ErbB2 for osteochondroma. LEVEL OF EVIDENCE Level IV, Diagnostic Study.
Collapse
Affiliation(s)
- Zhen Huang
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Sheng-Lin Wang
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qing-Shan Huang
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiao-Dong Li
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Hui Chen
- Fujian Provincial Institute of Orthopedic, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jian-Hua Lin
- Department of Orthopedics, First Affiliated Hospital of Fujian Medical University, Fuzhou, China;Fujian Provincial Institute of Orthopedic, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
114
|
Farrington CC, Yuan E, Mazhar S, Izadmehr S, Hurst L, Allen-Petersen BL, Janghorban M, Chung E, Wolczanski G, Galsky M, Sears R, Sangodkar J, Narla G. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49933-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
115
|
Michmerhuizen AR, Chandler B, Olsen E, Wilder-Romans K, Moubadder L, Liu M, Pesch AM, Zhang A, Ritter C, Ward ST, Santola A, Nyati S, Rae JM, Hayes D, Feng FY, Spratt D, Wahl D, Eisner J, Pierce LJ, Speers C. Seviteronel, a Novel CYP17 Lyase Inhibitor and Androgen Receptor Antagonist, Radiosensitizes AR-Positive Triple Negative Breast Cancer Cells. Front Endocrinol (Lausanne) 2020; 11:35. [PMID: 32117061 PMCID: PMC7027396 DOI: 10.3389/fendo.2020.00035] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/17/2020] [Indexed: 12/21/2022] Open
Abstract
Increased rates of locoregional recurrence (LR) have been observed in triple negative breast cancer (TNBC) despite multimodality therapy, including radiation (RT). Recent data suggest inhibiting the androgen receptor (AR) may be an effective radiosensitizing strategy, and AR is expressed in 15-35% of TNBC tumors. The aim of this study was to determine whether seviteronel (INO-464), a novel CYP17 lyase inhibitor and AR antagonist, is able to radiosensitize AR-positive (AR+) TNBC models. In cell viability assays, seviteronel and enzalutamide exhibited limited effect as a single agent (IC50 > 10 μM). Using clonogenic survival assays, however, AR knockdown and AR inhibition with seviteronel were effective at radiosensitizing cells with radiation enhancement ratios of 1.20-1.89 in models of TNBC with high AR expression. AR-negative (AR-) models, regardless of their estrogen receptor expression, were not radiosensitized with seviteronel treatment at concentrations up to 5 μM. Radiosensitization of AR+ TNBC models was at least partially dependent on impaired dsDNA break repair with significant delays in repair at 6, 16, and 24 h as measured by immunofluorescent staining of γH2AX foci. Similar effects were observed in an in vivo AR+ TNBC xenograft model where there was a significant reduction in tumor volume and a delay to tumor doubling and tripling times in mice treated with seviteronel and radiation. Following combination treatment with seviteronel and radiation, increased binding of AR occurred at DNA damage response genes, including genes involved both in homologous recombination and non-homologous end joining. This trend was not observed with combination treatment of enzalutamide and RT, suggesting that seviteronel may have a different mechanism of radiosensitization compared to other AR inhibitors. Enzalutamide and seviteronel treatment also had different effects on AR and AR target genes as measured by immunoblot and qPCR. These results implicate AR as a mediator of radioresistance in AR+ TNBC models and support the use of seviteronel as a radiosensitizing agent in AR+ TNBC.
Collapse
Affiliation(s)
- Anna R. Michmerhuizen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin Chandler
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Cancer Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Eric Olsen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Leah Moubadder
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Meilan Liu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Andrea M. Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Amanda Zhang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Cassandra Ritter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - S. Tanner Ward
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Alyssa Santola
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Shyam Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - James M. Rae
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Daniel Hayes
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Felix Y. Feng
- Department of Urology, Medicine and Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States
| | - Daniel Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Daniel Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Joel Eisner
- Innocrin Pharmaceuticals Inc., Durham, NC, United States
| | - Lori J. Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Corey Speers
| |
Collapse
|
116
|
Farrington CC, Yuan E, Mazhar S, Izadmehr S, Hurst L, Allen-Petersen BL, Janghorban M, Chung E, Wolczanski G, Galsky M, Sears R, Sangodkar J, Narla G. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers. J Biol Chem 2019; 295:757-770. [PMID: 31822503 DOI: 10.1074/jbc.ra119.011443] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor suppressor protein phosphatase 2A (PP2A) is a serine/threonine phosphatase whose activity is inhibited in most human cancers. One of the best-characterized PP2A substrates is MYC proto-oncogene basic helix-loop-helix transcription factor (MYC), whose overexpression is commonly associated with aggressive forms of this disease. PP2A directly dephosphorylates MYC, resulting in its degradation. To explore the therapeutic potential of direct PP2A activation in a diverse set of MYC-driven cancers, here we used biochemical assays, recombinant cell lines, gene expression analyses, and immunohistochemistry to evaluate a series of first-in-class small-molecule activators of PP2A (SMAPs) in Burkitt lymphoma, KRAS-driven non-small cell lung cancer, and triple-negative breast cancer. In all tested models of MYC-driven cancer, the SMAP treatment rapidly and persistently inhibited MYC expression through proteasome-mediated degradation, inhibition of MYC transcriptional activity, decreased cancer cell proliferation, and tumor growth inhibition. Importantly, we generated a series of cell lines expressing PP2A-dependent phosphodegron variants of MYC and demonstrated that the antitumorigenic activity of SMAPs depends on MYC degradation. Collectively, the findings presented here indicate a pharmacologically tractable approach to drive MYC degradation by using SMAPs for the management of a broad range of MYC-driven cancers.
Collapse
Affiliation(s)
| | - Eric Yuan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106
| | - Sahar Mazhar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Sudeh Izadmehr
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Lauren Hurst
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Brittany L Allen-Petersen
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Mahnaz Janghorban
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Eric Chung
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106
| | - Grace Wolczanski
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Matthew Galsky
- Department of Medicine, Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Rosalie Sears
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon 97239
| | - Jaya Sangodkar
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48105
| |
Collapse
|
117
|
Stromal Cell Signature Associated with Response to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer. Cells 2019; 8:cells8121566. [PMID: 31817155 PMCID: PMC6953077 DOI: 10.3390/cells8121566] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 01/26/2023] Open
Abstract
Breast cancer stromal compartment, may influence responsiveness to chemotherapy. Our aim was to detect a stromal cell signature (using a direct approach of microdissected stromal cells) associated with response to neoadjuvant chemotherapy (neoCT) in locally advanced breast cancer (LABC). The tumor samples were collected from 44 patients with LABC (29 estrogen receptor (ER) positive and 15 ER negative) before the start of any treatment. Neoadjuvant chemotherapy consisted of doxorubicin and cyclophosphamide, followed by paclitaxel. Response was defined as downstaging to maximum ypT1a-b/ypN0. The stromal cells, mainly composed of fibroblast and immune cells, were microdissected from fresh frozen tumor samples and gene expression profile was determined using Agilent SurePrint G3 Human Gene Expression microarrays. Expression levels were compared using MeV (MultiExperiment Viewer) software, applying SAM (significance analysis of microarrays). To classify samples according to tumor response, the order of median based on confidence statements (MedOr) was used, and to identify gene sets correlated with the phenotype downstaging, gene set enrichment analysis (GSEA). Nine patients presented disease downstaging. Eleven sequences (FDR 17) were differentially expressed, all of which (except H2AFJ) more expressed in responsive tumors, including PTCHD1 and genes involved in abnormal cytotoxic T cell physiology, TOX, LY75, and SH2D1A. The following four pairs of markers could correctly classify all tumor samples according to response: PTCHD1/PDXDC2P, LOC100506731/NEURL4, SH2D1A/ENST00000478672, and TOX/H2AFJ. Gene sets correlated with tumor downstaging (FDR < 0.01) were mainly involved in immune response or lymphocyte activation, including CD47, LCK, NCK1, CD24, CD3E, ZAP70, FOXP3, and CD74, among others. In locally advanced breast cancer, stromal cells may present specific features of immune response that may be associated with chemotherapy response.
Collapse
|
118
|
Fundytus A, Saad N, Logie N, Roldan Urgoiti G. Breast cancer in transgender female‐to‐male individuals: A case report of androgen receptor‐positive breast cancer. Breast J 2019; 26:1007-1012. [DOI: 10.1111/tbj.13655] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Adam Fundytus
- Department of Oncology University of Calgary/Tom Baker Cancer Calgary AB Canada
| | - Nathalie Saad
- Department of Endocrinology University of Calgary Calgary AB Canada
| | - Natalie Logie
- Department of Radiation Oncology University of Calgary/Tom Baker Cancer Calgary AB Canada
| | | |
Collapse
|
119
|
Suzuki A, Shim J, Ogata K, Yoshioka H, Iwata J. Cholesterol metabolism plays a crucial role in the regulation of autophagy for cell differentiation of granular convoluted tubules in male mouse submandibular glands. Development 2019; 146:dev.178335. [PMID: 31558435 DOI: 10.1242/dev.178335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022]
Abstract
It has been long appreciated that sex hormone receptors are expressed in various non-gonadal organs. However, it remains unclear how sex hormones regulate the morphogenesis of these non-gonadal organs. To address this issue, we used a male mouse model of androgen-dependent salivary gland morphogenesis. Mice with excessive cholesterol synthesis in the salivary glands exhibited defects in the maturation of granular convoluted tubules (GCTs), which is regulated through sex hormone-dependent cascades. We found that excessive cholesterol synthesis resulted in autophagy failure specifically in the duct cells of salivary glands, followed by the accumulation of NRF2, a transcription factor known as one of the specific substrates for autophagy. The accumulated NRF2 suppressed the expression of Foxa1, which forms a transcriptional complex with the androgen receptor to regulate target genes. Taken together, our results indicate that cholesterol metabolism plays a crucial role in GCT differentiation through autophagy.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Junbo Shim
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Kenichi Ogata
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Hiroki Yoshioka
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA .,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA.,MD Anderson Cancer Center University of Texas Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
120
|
Olivares-Urbano MA, Griñán-Lisón C, Zurita M, Del Moral R, Ríos-Arrabal S, Artacho-Cordón F, Arrebola JP, González AR, León J, Antonio Marchal J, Núñez MI. Matrix metalloproteases and TIMPs as prognostic biomarkers in breast cancer patients treated with radiotherapy: A pilot study. J Cell Mol Med 2019; 24:139-148. [PMID: 31568637 PMCID: PMC6933337 DOI: 10.1111/jcmm.14671] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/14/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022] Open
Abstract
Breast cancer (BC) is the most common tumour in women and one of the most important causes of cancer death worldwide. Radiation therapy (RT) is widely used for BC treatment. Some proteins have been identified as prognostic factors for BC (Ki67, p53, E‐cadherin, HER2). In the last years, it has been shown that variations in the expression of MMPs and TIMPs may contribute to the development of BC. The aim of this pilot work was to study the effects of RT on different MMPs (‐1, ‐2, ‐3, ‐7, ‐8, ‐9, ‐10, ‐12 and ‐13) and TIMPs (‐1 to ‐4), as well as their relationship with other variables related to patient characteristics and tumour biology. A group of 20 BC patients treated with RT were recruited. MMP and TIMP serum levels were analysed by immunoassay before, during and after RT. Our pilot study showed a slight increase in the levels of most MMP and TIMP with RT. However, RT produced a significantly decrease in TIMP‐1 and TIMP‐3 levels. Significant correlations were found between MMP‐3 and TIMP‐4 levels, and some of the variables studied related to patient characteristics and tumour biology. Moreover, MMP‐9 and TIMP‐3 levels could be predictive of RT toxicity. For this reason, MMP‐3, MMP‐9, TIMP‐3 and TIMP‐4 could be used as potential prognostic and predictive biomarkers for BC patients treated with RT.
Collapse
Affiliation(s)
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada, Spain
| | - Mercedes Zurita
- Department of Radiation Oncology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Rosario Del Moral
- Department of Radiation Oncology, Virgen de las Nieves University Hospital, Granada, Spain
| | - Sandra Ríos-Arrabal
- Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, Spain
| | - Francisco Artacho-Cordón
- Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs.Granada, Granada, Spain
| | - Juan Pedro Arrebola
- Biosanitary Research Institute, ibs.Granada, Granada, Spain.,Department of Preventive Medicine and Public Health, School of Medicine, University of Granada, Granada, Spain
| | - Amanda Rocío González
- Bio-Health Research Foundation of Eastern Andalusia - Alejandro Otero (FIBAO), Granada, Spain
| | - Josefa León
- Biosanitary Research Institute, ibs.Granada, Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs.Granada, Granada, Spain
| | - María Isabel Núñez
- Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, Spain.,Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs.Granada, Granada, Spain
| |
Collapse
|
121
|
Giovannelli P, Di Donato M, Galasso G, Di Zazzo E, Medici N, Bilancio A, Migliaccio A, Castoria G. Breast cancer stem cells: The role of sex steroid receptors. World J Stem Cells 2019; 11:594-603. [PMID: 31616537 PMCID: PMC6789191 DOI: 10.4252/wjsc.v11.i9.594] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/06/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women, and current available therapies often have high success rates. Nevertheless, BC might acquire drug resistance and sometimes relapse. Current knowledge about the most aggressive forms of BC points to the role of specific cells with stem properties located within BC, the so-called “BC stem cells” (BCSCs). The role of BCSCs in cancer formation, growth, invasiveness, therapy resistance and tumor recurrence is becoming increasingly clear. The growth and metastatic properties of BCSCs are regulated by different pathways, which are only partially known. Sex steroid receptors (SSRs), which are involved in BC etiology and progression, promote BCSC proliferation, dedifferentiation and migration. However, in the literature, there is incomplete information about their roles. Particularly, there are contrasting conclusions about the expression and role of the classical BC hormonal biomarkers, such as estrogen receptor alpha (ERα), together with scant, albeit promising information concerning ER beta (ERβ) and androgen receptor (AR) properties that control different transduction pathways in BCSCs. In this review, we will discuss the role that SRs expressed in BCSCs play to BC progression and recurrence and how these findings have opened new therapeutic possibilities.
Collapse
Affiliation(s)
- Pia Giovannelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Marzia Di Donato
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Giovanni Galasso
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Erika Di Zazzo
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Nicola Medici
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Antonio Bilancio
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Antimo Migliaccio
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| | - Gabriella Castoria
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Naples 80138, Italy
| |
Collapse
|
122
|
Evaluations of Biomarker Status Changes between Primary and Recurrent Tumor Tissue Samples in Breast Cancer Patients. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7391237. [PMID: 31583246 PMCID: PMC6754865 DOI: 10.1155/2019/7391237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/14/2019] [Accepted: 07/30/2019] [Indexed: 01/01/2023]
Abstract
Background Obtaining tumor specimens and re-evaluating targeted markers is recommended, if possible, in breast cancer patients who relapsed after curative treatment. The biomarker status changes in rebiopsied tumors have been demonstrated to have considerable clinical implications. Objectives To identify the changes of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status between the primary and recurrent lesions. Materials and Methods We conducted a study among 67 patients with recurrent breast cancer, recruited from January 2014 to September 2018 in the Vietnam National Cancer Hospital to compare ER, PR, and HER2 status between the primary and recurrent lesions. For each patient, a specimen of their primary tumor and another specimen of recurrent lesions underwent pathological assessment. Immunohistochemistry (IHC) was performed to determine ER, PR, and HER2 status in both specimens. Results Biomarker status conversion rates (in both directions) between primary and recurrent tumors were 26.9% for ER, 38.8% for PR, and 22.4% for HER2. Overall, IHC subtypes (hormone receptor positive, HER2 amplified, and triple-negative) changed in 25 out of 67 (37.3%) cases. Conversion rates were not statistically significantly different between patients with different recurrent sites and times of recurrence. Eight out of 13 initially triple-negative patients (61.5%) had a change to positive status of either ER, PR, or HER2. Conclusion A substantial discordance in ER, PR, and HER2 status were observed between primary breast cancer tissues and recurrent lesions. Rebiopsy could bring new therapeutic opportunities in the management of patients with recurrent breast cancer.
Collapse
|
123
|
Cohn BA, Cirillo PM. In utero and postnatal programing of dehydroepiandrosterone sulfate (DHEAS) in young adult women. Reprod Toxicol 2019; 92:148-154. [PMID: 31173873 DOI: 10.1016/j.reprotox.2019.05.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/02/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
Abstract
Fetal adrenal-derived OH-DHEAS is the primary precursor for maternal estriol, an abundant, human, placental estrogen. We measured maternal pregnancy estriol as a marker of fetal adrenal function + placenta capacity to synthesize estriol. We hypothesized that maternal estriol is directly correlated with the adrenal hormone, DHEAS, in young adult women. We tested this hypothesis in a subset of women in the Child Health and Development Studies (351 of 470 eligible). 176 of these had serum samples collected at ages 27-30 for DHEAS assays, archived maternal pregnancy serum for estriol assays, and childhood growth data. In regression analyses, both maternal estriol and accelerated growth in middle childhood were independently, directly associated with DHEAS (+19% for quartile 4 versus quartile 1 of estriol, 95%CI=+ 2%, +36% and +12% for quartile 4 versus quartile 1 for middle childhood growth, 95%CI= +3%, +21%). Adrenal function may be programmed in utero and middle childhood with long-term consequences.
Collapse
Affiliation(s)
- Barbara A Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley CA 94708, United States.
| | - Piera M Cirillo
- Child Health and Development Studies, Public Health Institute, Berkeley CA 94708, United States
| |
Collapse
|
124
|
Pizato N, Kiffer LFMV, Luzete BC, Assumpção JAF, Correa LH, Melo HABD, Sant'Ana LPD, Ito MK, Magalhães KG. Omega 3-DHA and Delta-Tocotrienol Modulate Lipid Droplet Biogenesis and Lipophagy in Breast Cancer Cells: the Impact in Cancer Aggressiveness. Nutrients 2019; 11:E1199. [PMID: 31141912 PMCID: PMC6627337 DOI: 10.3390/nu11061199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Omega 3-docosahexaenoic acid (DHA) and vitamin E Delta-tocotrienol (Delta-T3) are extensively studied as protective nutrients against cancer development. Little is known about the biological mechanisms targeted by these bioactive molecules on lipid droplet (LD) biogenesis, an important breast cancer aggressiveness marker, and the occurrence of lipophagy in breast cancer cells. The aim of this study was to investigate the effect of DHA, Delta-T3 and DHA plus Delta-T3 co-treatment in LD biogenesis and lipophagy process in triple negative breast cancer cell line MDA-MB-231. Cells were treated with 50 μM DHA and/or 5 μM Delta-T3. Our results demonstrated that DHA can trigger an increase in LD biogenesis and co-treatment with Delta-T3 was able to reduce this LD biogenesis. In addition, we showed that a higher cytoplasmic LD content is associated with a higher breast cancer cells malignance and proliferation. Reduction of cytoplasmic LD content by silencing ADRP (adipose differentiation-related protein), a structural LD protein, also decreased cell proliferation in MDA-MB-231 cells. Treatment with DHA and Delta-T3 alone or co-treatment did not reduce cell viability. Moreover, we showed here that DHA can trigger lipophagy in MDA-MB-231 cells and DHA plus Delta-T3 co-treatment was able to enhance this lipophagy process. Our findings demonstrated that co-treatment with DHA plus Delta-T3 in MDA-MB-231 cells could reduce LD biogenesis and potentiate lipophagy in these cells, possibly having a positive impact to inhibit breast cancer malignancy. Therefore, suitable doses of DHA and Delta-T3 vitamin E isoform supplementation can be a prominent tool in therapeutic treatments against breast cancer.
Collapse
Affiliation(s)
- Nathalia Pizato
- Department of Nutrition, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Larissa Fernanda Melo Vasconcelos Kiffer
- Department of Nutrition, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Beatriz Christina Luzete
- Department of Nutrition, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - José Antonio Fagundes Assumpção
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Luis Henrique Correa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Heloisa Antoniella Braz de Melo
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Lívia Pimentel de Sant'Ana
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Marina Kiyomi Ito
- Department of Nutrition, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, UnB, Brasilia 70910-900, Brazil.
| |
Collapse
|
125
|
Xia X, Huang C, Liao Y, Liu Y, He J, Guo Z, Jiang L, Wang X, Liu J, Huang H. Inhibition of USP14 enhances the sensitivity of breast cancer to enzalutamide. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:220. [PMID: 31126320 PMCID: PMC6534920 DOI: 10.1186/s13046-019-1227-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Androgen receptor (AR) is expressed in approximately 70% of breast tumors. Recent studies increasingly support AR as a potential therapeutic target of AR-positive breast cancer. We have previously reported that deubiquitinase USP14 stabilizes AR proteins by deubiquitination and USP14 inhibition results in inhibition of cell growth and tumor progression in AR-positive prostate cancer and breast cancer. The current study aims to explore the anticancer effect of a treatment combining AR antagonist enzalutamide with USP14 inhibition on breast cancer cells. METHODS The combining effects of enzalutamide and USP14 inhibition on breast cancer cell proliferation and apoptosis and associated cell signaling were evaluated in vitro and in vivo. RESULTS USP14 inhibition via administration of IU1 or USP14-specific siRNA/shRNA enhanced cell growth inhibition and apoptosis induction by enzalutamide in breast cancer cell lines in vitro and in vivo. Additionally, the combination of enzalutamide with USP14 inhibition/knockdown induced significant downregulation of AR proteins and suppression of AR-related signaling pathways, including Wnt/β-catenin and PI3K/AKT pathways. Moreover, AKT inhibition via MK2206 increased the antiproliferative and proapoptotic effects of enzalutamide+IU1 combined treatment. CONCLUSION Collectively, our data suggest that USP14 inhibition in combination with enzalutamide represents a potentially new therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Xiaohong Xia
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Chuyi Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuning Liao
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Yuan Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Jinchan He
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Zhiqiang Guo
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Lili Jiang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, 57069, USA
| | - Jinbao Liu
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Hongbiao Huang
- Affiliated Cancer Hospital and institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
126
|
Lima ZS, Ghadamzadeh M, Arashloo FT, Amjad G, Ebadi MR, Younesi L. Recent advances of therapeutic targets based on the molecular signature in breast cancer: genetic mutations and implications for current treatment paradigms. J Hematol Oncol 2019; 12:38. [PMID: 30975222 PMCID: PMC6460547 DOI: 10.1186/s13045-019-0725-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common malignancy in women all over the world. Genetic background of women contributes to her risk of having breast cancer. Certain inherited DNA mutations can dramatically increase the risk of developing certain cancers and are responsible for many of the cancers that run in some families. Regarding the widespread multigene panels, whole exome sequencing is capable of providing the evaluation of genetic function mutations for development novel strategy in clinical trials. Targeting the mutant proteins involved in breast cancer can be an effective therapeutic approach for developing novel drugs. This systematic review discusses gene mutations linked to breast cancer, focusing on signaling pathways that are being targeted with investigational therapeutic strategies, where clinical trials could be potentially initiated in the future are being highlighted.
Collapse
Affiliation(s)
- Zeinab Safarpour Lima
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mostafa Ghadamzadeh
- Departement of Radiology, Hasheminejad Kidney Centre (HKC), Iran University of Medical Sciences, Tehran, Iran
| | | | - Ghazaleh Amjad
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohammad Reza Ebadi
- Shohadaye Haft-e-tir Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ladan Younesi
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
127
|
Bandini E, Fanini F. MicroRNAs and Androgen Receptor: Emerging Players in Breast Cancer. Front Genet 2019; 10:203. [PMID: 30941159 PMCID: PMC6433747 DOI: 10.3389/fgene.2019.00203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/26/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is the most common cause of cancer among women, with a high incidence rate occurrence every year worldwide despite advances in its management. BC is characterized by a spectrum of subtypes which respond differently to treatments due to their biological features, representing the main issue in the control of this type of malignancy. Androgen receptor (AR) is emerging as a target to investigate among hormone receptors, since it seems to play a role at various stages of development of specific BC subsets. For this reason, in recent years AR has become very important in the clinical practice, although its role remains controversial. A number of studies have proposed a correlation between microRNAs (miRNAs), a class of gene expression modulators, and AR in prostate cancer (PC), but there are still few evidences about the relationship between miRNAs and AR in BC. The purpose of this review is to present a state of the art scenario with consideration to the most recent discoveries about miRNAs involved in the AR associated pathogenesis of BC, in order to provide new insights into the role of miRNAs as key drivers in the modulation of AR, and possible actors in the development and progression of BC. Moreover, we consider findings about involvement of AR signaling in all stages of BC, highlighting its association with different subsets of breast carcinomas and with pre- and postmenopausal state of patients.
Collapse
Affiliation(s)
| | - Francesca Fanini
- Biosciences Laboratory, Department of Clinical and Experimental Oncology and Hematology, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) S.r.l. IRCCS, Meldola, Italy
| |
Collapse
|
128
|
Androgens Induce Invasiveness of Triple Negative Breast Cancer Cells Through AR/Src/PI3-K Complex Assembly. Sci Rep 2019; 9:4490. [PMID: 30872694 PMCID: PMC6418124 DOI: 10.1038/s41598-019-41016-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 02/25/2019] [Indexed: 12/22/2022] Open
Abstract
Breast cancer (BC) is still characterized by high morbidity and mortality. A specific BC subtype named triple negative BC (TNBC) lacks estrogen and progesterone receptors (ER and PR, respectively) and is characterized by the absence of overexpression/amplification of human epidermal growth factor receptor 2 (HER2). The androgen receptor (AR) is expressed in TNBC, although its function in these cancers is still debated. Moreover, few therapeutic options are currently available for the treatment of TNBC. In this study, we have used TNBC-derived MDA-MB231 and MDA-MB453 cells that, albeit at different extent, both express AR. Androgen challenging induces migration and invasiveness of these cells. Use of the anti-androgen bicalutamide or AR knockdown experiments show that these effects depend on AR. Furthermore, the small peptide, S1, which mimics the AR proline-rich motif responsible for the interaction of AR with SH3-Src, reverses the effects in both cell lines, suggesting that the assembly of a complex made up of AR and Src drives the androgen-induced motility and invasiveness. Co-immunoprecipitation experiments in androgen-treated MDA-MB231 and MDA-MB453 cells show that the AR/Src complex recruits p85α, the regulatory subunit of PI3-K. In such a way, the basic machinery leading to migration and invasiveness is turned-on. The S1 peptide inhibits motility and invasiveness of TNBC cells and disrupts the AR/Src/p85α complex assembly in MDA-MB231 cells. This study shows that the rapid androgen activation of Src/PI3-K signaling drives migration and invasiveness of TNBC cells and suggests that the S1 peptide is a promising therapeutic option for these cancers.
Collapse
|
129
|
Baicalein Suppresses Stem Cell-Like Characteristics in Radio- and Chemoresistant MDA-MB-231 Human Breast Cancer Cells through Up-Regulation of IFIT2. Nutrients 2019; 11:nu11030624. [PMID: 30875792 PMCID: PMC6471144 DOI: 10.3390/nu11030624] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/06/2019] [Accepted: 03/11/2019] [Indexed: 12/22/2022] Open
Abstract
Resistance to both chemotherapy and radiation therapy is frequent in triple-negative breast cancer (TNBC) patients. We established treatment-resistant TNBC MDA-MB-231/IR cells by irradiating the parental MDA-MB-231 cells 25 times with 2 Gy irradiation and investigated the molecular mechanisms of acquired resistance. The resistant MDA-MB-231/IR cells were enhanced in migration, invasion, and stem cell-like characteristics. Pathway analysis by the Database for Annotation, Visualization and Integrated Discovery revealed that the NF-κB pathway, TNF signaling pathway, and Toll-like receptor pathway were enriched in MDA-MB-231/IR cells. Among 77 differentially expressed genes revealed by transcriptome analysis, 12 genes involved in drug and radiation resistance, including interferon-induced protein with tetratricopeptide repeats 2 (IFIT2), were identified. We found that baicalein effectively reversed the expression of IFIT2, which is reported to be associated with metastasis, recurrence, and poor prognosis in TNBC patients. Baicalein sensitized radio- and chemoresistant cells and induced apoptosis, while suppressing stem cell-like characteristics, such as mammosphere formation, side population, expression of Oct3/4 and ABCG2, and CD44highCD24low population in MDA-MB-231/IR cells. These findings improve our understanding of the genes implicated in radio- and chemoresistance in breast cancer, and indicate that baicalein can serve as a sensitizer that overcomes treatment resistance.
Collapse
|
130
|
Licochalcone A Inhibits Cellular Motility by Suppressing E-cadherin and MAPK Signaling in Breast Cancer. Cells 2019; 8:cells8030218. [PMID: 30841634 PMCID: PMC6468539 DOI: 10.3390/cells8030218] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/16/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
A compound isolated from Glycyrrhiza uralensis, licochalcone A (LA) exhibits anti-inflammatory and anti-tumor properties in various cell lines. LA has been found to promote autophagy and suppress specificity protein 1, inducing apoptosis in breast cancer cells. However, the regulation of breast cancer cell invasion and migration by LA is elusive. Thus, the present study investigated whether LA induces apoptosis and cellular motility in MDA-MB-231 breast cells, and investigated the underlying molecular mechanisms. MDA-MB-231 cells treated with LA and cell viability measured by cell counting kit-8 assay. Apoptotic signal proteins checked by flow cytometry, fluorescent staining, and Western blot. LA effectively suppressed cell migration, and modulated E-cadherin and vimentin expression by blocking MAPK and AKT signaling. LA inhibited cell proliferation and cell cycle, modulated mitochondrial membrane potential and DNA damage, and reduced oxidative stress in MDA-MB-231 cells. LA also activated cleaved-caspase 3 and 9, significantly decreased Bcl-2 expression, ultimately causing the release of cytochrome c from the mitochondria into the cytoplasm. Overall, our findings suggest that LA decreases cell proliferation and increases reactive oxygen species production for induced apoptosis, and regulates E-cadherin and vimentin by reducing MAPK and AKT signaling, resulting in suppressed MDA-MB-231 cell migration and invasion.
Collapse
|
131
|
Brunetti A, Manfioletti G. Editorial: Hormone Receptors and Breast Cancer. Front Endocrinol (Lausanne) 2019; 10:205. [PMID: 31001204 PMCID: PMC6454855 DOI: 10.3389/fendo.2019.00205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Affiliation(s)
- Antonio Brunetti
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti
| | | |
Collapse
|
132
|
Schuster S, Biri-Kovács B, Szeder B, Buday L, Gardi J, Szabó Z, Halmos G, Mező G. Enhanced In Vitro Antitumor Activity of GnRH-III-Daunorubicin Bioconjugates Influenced by Sequence Modification. Pharmaceutics 2018; 10:E223. [PMID: 30423956 PMCID: PMC6320914 DOI: 10.3390/pharmaceutics10040223] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 12/21/2022] Open
Abstract
Receptors for gonadotropin releasing hormone (GnRH) are highly expressed in various human cancers including breast, ovarian, endometrial, prostate and colorectal cancer. Ligands like human GnRH-I or the sea lamprey analogue GnRH-III represent a promising approach for the development of efficient drug delivery systems for targeted tumor therapy. Here, we report on the synthesis and cytostatic effect of 14 oxime bond-linked daunorubicin GnRH-III conjugates containing a variety of unnatural amino acids within the peptide sequence. All compounds demonstrated a reduced cell viability in vitro on estrogen receptor α (ERα) positive and ERα negative cancer cells. The best candidate revealed an increased cancer cell growth inhibitory effect compared to our lead-compound GnRH-III-[⁴Lys(Bu),⁸Lys(Dau=Aoa)]. Flow cytometry and fluorescence microscopy studies showed that the cellular uptake of the novel conjugate is substantially improved leading to an accelerated delivery of the drug to its site of action. However, the release of the active drug-metabolite by lysosomal enzymes was not negatively affected by amino acid substitution, while the compound provided a high stability in human blood plasma. Receptor binding studies were carried out to ensure a high binding affinity of the new compound for the GnRH-receptor. It was demonstrated that GnRH-III-[²ΔHis,³d-Tic,⁴Lys(Bu),⁸Lys(Dau=Aoa)] is a highly potent and promising anticancer drug delivery system for targeted tumor therapy.
Collapse
Affiliation(s)
- Sabine Schuster
- Faculty of Science, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary.
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, 1117 Budapest, Hungary.
| | - Beáta Biri-Kovács
- Faculty of Science, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary.
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, 1117 Budapest, Hungary.
| | - Bálint Szeder
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, 1117 Budapest, Hungary.
| | - László Buday
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, 1117 Budapest, Hungary.
| | - János Gardi
- First Department of Internal Medicine, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gábor Mező
- Faculty of Science, Institute of Chemistry, Eötvös Loránd University, 1117 Budapest, Hungary.
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, 1117 Budapest, Hungary.
| |
Collapse
|
133
|
Girgert R, Emons G, Gründker C. Estrogen Signaling in ERα-Negative Breast Cancer: ERβ and GPER. Front Endocrinol (Lausanne) 2018; 9:781. [PMID: 30687231 PMCID: PMC6333678 DOI: 10.3389/fendo.2018.00781] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/12/2018] [Indexed: 01/22/2023] Open
Abstract
Estrogen receptors are important regulators of the growth of breast tumors. Three different receptors for estrogens have been identified in breast tumors, two nuclear receptors, ERα and ERβ, and a G-protein coupled estrogen receptor 1 (GPER) that initiates non-genomic effects of estrogens in the cytosol. Recent findings show that the stimulation of cytoplasmic ERα and ERβ also triggers non-genomic signaling pathways. The treatment of breast cancer with anti-estrogens depends on the presence of ERα. About 40% of all breast cancers, however, do not express ERα. One subgroup of these tumors overexpress Her-2, another important group is designated as triple-negative breast cancer, as they neither express ERα, nor progesterone receptors, nor do they overexpress Her-2. This review addresses the signaling of ERβ and GPER in ERα-negative breast tumors. In addition to the well-established EGF-receptor transactivation pathways of GPER, more recent findings of GPER-dependent activation of FOXO3a, the Hippo-pathway, and HOTAIR-activation are summarized.
Collapse
|