151
|
Yin L, Duan JJ, Bian XW, Yu SC. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res 2020; 22:61. [PMID: 32517735 PMCID: PMC7285581 DOI: 10.1186/s13058-020-01296-5] [Citation(s) in RCA: 1282] [Impact Index Per Article: 256.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC), a specific subtype of breast cancer that does not express estrogen receptor (ER), progesterone receptor (PR), or human epidermal growth factor receptor 2 (HER-2), has clinical features that include high invasiveness, high metastatic potential, proneness to relapse, and poor prognosis. Because TNBC tumors lack ER, PR, and HER2 expression, they are not sensitive to endocrine therapy or HER2 treatment, and standardized TNBC treatment regimens are still lacking. Therefore, development of new TNBC treatment strategies has become an urgent clinical need. By summarizing existing treatment regimens, therapeutic drugs, and their efficacy for different TNBC subtypes and reviewing some new preclinical studies and targeted treatment regimens for TNBC, this paper aims to provide new ideas for TNBC treatment.
Collapse
Affiliation(s)
- Li Yin
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China.,Key Laboratory of Cancer Immunopathology, Ministry of Education, ChongQing, 400038, China
| | - Jiang-Jie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China.,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China.,Key Laboratory of Cancer Immunopathology, Ministry of Education, ChongQing, 400038, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China.,Key Laboratory of Cancer Immunopathology, Ministry of Education, ChongQing, 400038, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China. .,Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), ChongQing, 400038, China. .,Key Laboratory of Cancer Immunopathology, Ministry of Education, ChongQing, 400038, China.
| |
Collapse
|
152
|
Reddy TP, Choi DS, Anselme AC, Qian W, Chen W, Lantto J, Horak ID, Kragh M, Chang JC, Rosato RR. Simultaneous targeting of HER family pro-survival signaling with Pan-HER antibody mixture is highly effective in TNBC: a preclinical trial with PDXs. Breast Cancer Res 2020; 22:48. [PMID: 32414394 PMCID: PMC7227035 DOI: 10.1186/s13058-020-01280-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The human epidermal growth factor receptor (HER) family, notably EGFR, is overexpressed in most triple-negative breast cancer (TNBC) cases and provides cancer cells with compensatory signals that greatly contribute to the survival and development of resistance in response to therapy. This study investigated the effects of Pan-HER (Symphogen, Ballerup, Denmark), a novel mixture of six monoclonal antibodies directed against members of the HER family EGFR, HER2, and HER3, in a preclinical trial of TNBC patient-derived xenografts (PDXs). METHODS Fifteen low passage TNBC PDX tumor samples were transferred into the right mammary fat pad of mice for engraftment. When tumors reached an average size of 100-200 mm3, mice were randomized (n ≥ 6 per group) and treated following three 1-week cycles consisting of three times/week intraperitoneal (IP) injection of either formulation buffer (vehicle control) or Pan-HER (50 mg/kg). At the end of treatment, tumors were collected for Western blot, RNA, and immunohistochemistry analyses. RESULTS All 15 TNBC PDXs were responsive to Pan-HER treatment, showing significant reductions in tumor growth consistent with Pan-HER-mediated tumor downmodulation of EGFR and HER3 protein levels and significantly decreased activation of associated HER family signaling pathways AKT and ERK. Tumor regression was observed in five of the models, which corresponded to those PDX tumor models with the highest level of HER family activation. CONCLUSIONS The marked effect of Pan-HER in numerous HER family-dependent TNBC PDX models justifies further studies of Pan-HER in TNBC clinical trials as a potential therapeutic option.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Cell Proliferation/drug effects
- Disease Models, Animal
- Drug Resistance, Neoplasm
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Female
- Humans
- Mice
- Molecular Targeted Therapy
- Mutation
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Tejaswini P Reddy
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Dong S Choi
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Ann C Anselme
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Wen Chen
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Johan Lantto
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Ivan D Horak
- Symphogen A/S, Pederstrupvej 93, DK-2750, Ballerup, Denmark
| | - Michael Kragh
- Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Jenny C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Roberto R Rosato
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
153
|
Chen J, Liu C, Cen J, Liang T, Xue J, Zeng H, Zhang Z, Xu G, Yu C, Lu Z, Wang Z, Jiang J, Zhan X, Zeng J. KEGG-expressed genes and pathways in triple negative breast cancer: Protocol for a systematic review and data mining. Medicine (Baltimore) 2020; 99:e19986. [PMID: 32358373 PMCID: PMC7440132 DOI: 10.1097/md.0000000000019986] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The incidence of triple negative breast cancer (TNBC) is at a relatively high level, and our study aimed to identify differentially expressed genes (DEGs) in TNBC and explore the key pathways and genes of TNBC. METHODS The gene expression profiling (GSE86945, GSE86946 and GSE102088) data were obtained from Gene Expression Omnibus Datasets, DEGs were identified by using R software, Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of DEGs were performed by the Database for Annotation, Visualization and Integrated Discovery (DAVID) tools, and the protein-protein interaction (PPI) network of the DEGs was constructed by the STRING database and visualized by Cytoscape software. Finally, the survival value of hub DEGs in breast cancer patients were performed by the Kaplan-Meier plotter online tool. RESULTS A total of 2998 DEGs were identified between TNBC and health breast tissue, including 411 up-regulated DEGs and 2587 down-regulated DEGs. GO analysis results showed that down-regulated DEGs were enriched in gene expression (BP), extracellular exosome (CC), and nucleic acid binding, and up-regulated were enriched in chromatin assembly (BP), nucleosome (CC), and DNA binding (MF). KEGG pathway results showed that DEGs were mainly enriched in Pathways in cancer and Systemic lupus erythematosus and so on. Top 10 hub genes were picked out from PPI network by connective degree, and 7 of top 10 hub genes were significantly related with adverse overall survival in breast cancer patients (P < .05). Further analysis found that only EGFR had a significant association with the prognosis of triple-negative breast cancer (P < .05). CONCLUSIONS Our study showed that DEGs were enriched in pathways in cancer, top 10 DEGs belong to up-regulated DEGs, and 7 gene connected with poor prognosis in breast cancer, including HSP90AA1, SRC, HSPA8, ESR1, ACTB, PPP2CA, and RPL4. These can provide some guidance for our research on the diagnosis and prognosis of TNBC, and further research is needed to evaluate their value in the targeted therapy of TNBC.
Collapse
Affiliation(s)
| | - Chong Liu
- Department of Spine and Osteopathy Ward
| | | | - Tuo Liang
- Department of Spine and Osteopathy Ward
| | - Jiang Xue
- Department of Spine and Osteopathy Ward
| | | | | | | | | | | | | | - Jie Jiang
- Department of Spine and Osteopathy Ward
| | | | - Jian Zeng
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
154
|
Rivas J, Díaz N, Silva I, Morales D, Lavanderos B, Álvarez A, Saldías MP, Pulgar E, Cruz P, Maureira D, Flores G, Colombo A, Blanco C, Contreras HR, Jaña F, Gallegos I, Concha ML, Vergara-Jaque A, Poblete H, González W, Varela D, Trimmer JS, Cáceres M, Cerda O. KCTD5, a novel TRPM4-regulatory protein required for cell migration as a new predictor for breast cancer prognosis. FASEB J 2020; 34:7847-7865. [PMID: 32301552 DOI: 10.1096/fj.201901195rrr] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 12/23/2022]
Abstract
Transient receptor potential melastatin 4 (TRPM4) is a Ca2+ -activated nonselective cationic channel that regulates cell migration and contractility. Increased TRPM4 expression has been related to pathologies, in which cytoskeletal rearrangement and cell migration are altered, such as metastatic cancer. Here, we identify the K+ channel tetramerization domain 5 (KCTD5) protein, a putative adaptor of cullin3 E3 ubiquitin ligase, as a novel TRPM4-interacting protein. We demonstrate that KCTD5 is a positive regulator of TRPM4 activity by enhancing its Ca2+ sensitivity. We show that through its effects on TRPM4 that KCTD5 promotes cell migration and contractility. Finally, we observed that both TRPM4 and KCTD5 expression are increased in distinct patterns in different classes of breast cancer tumor samples. Together, these data support that TRPM4 activity can be regulated through expression levels of either TRPM4 or KCTD5, not only contributing to increased understanding of the molecular mechanisms involved on the regulation of these important ion channels, but also providing information that could inform treatments based on targeting these distinct molecules that define TRPM4 activity.
Collapse
Affiliation(s)
- José Rivas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile
| | - Nicolás Díaz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Danna Morales
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Alhejandra Álvarez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Eduardo Pulgar
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Guillermo Flores
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Constanza Blanco
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Héctor R Contreras
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Fabián Jaña
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Ivan Gallegos
- Departamento de Oncología Básico Clínica, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Departamento de Anatomía Patológica, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Miguel L Concha
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Millennium Nucleus on Physics of Active Matter, Santiago, Chile
| | - Ariela Vergara-Jaque
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Horacio Poblete
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Multidisciplinary Scientific Nucleus, Universidad de Talca, Talca, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Wendy González
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Center for Bioinformatics and Molecular Simulations (CBSM), Faculty of Engineering, Universidad de Talca, Talca, Chile
| | - Diego Varela
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,Program of Physiology and Biophysics, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|
155
|
Liu ZB, Zhang L, Bian J, Jian J. Combination Strategies of Checkpoint Immunotherapy in Metastatic Breast Cancer. Onco Targets Ther 2020; 13:2657-2666. [PMID: 32308409 PMCID: PMC7133118 DOI: 10.2147/ott.s240655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Checkpoint immunotherapy is emerging as a new therapeutic approach for metastatic breast cancer. Monotherapy of immunoagents against PD1/PD-L1 or CTLA-4 has shown little efficacy in these patients. Recently, to determine the optimal use of immunotherapy, there has been a rapid expansion in the number of clinical trials developing immunotherapy combinations. These combination therapeutic approaches can enhance various aspects of cancer immunity, such as tumor antigenicity or intratumor T cell infiltration, which provides a theoretical basis for combining them with checkpoint immunotherapy to achieve synergistic effects. Here, we review the available data and ongoing efforts to establish the safety and efficacy of immunoagents in combination with chemotherapy, radiotherapy, HER2-targeted therapy, CDK4/6 inhibitors, PARP inhibitors, and another checkpoint immunoagents.
Collapse
Affiliation(s)
- Zhi Bing Liu
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Luyan Zhang
- Department of Oncology, Binzhou People's Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Jia Bian
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Jinbo Jian
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| |
Collapse
|
156
|
Weihua Z, Guorong Z, Xiaolong C, Weizhan L. MiR-33a functions as a tumor suppressor in triple-negative breast cancer by targeting EZH2. Cancer Cell Int 2020; 20:85. [PMID: 32206036 PMCID: PMC7079399 DOI: 10.1186/s12935-020-1160-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background Increasing reports have confirmed that microRNAs play an important role in breast cancer progression, particularly in triple-negative breast cancer (TNBC). The aim of our study was to investigate the role of miR-33a in TNBC progression. Methods PCR assays were performed to detect miR-33a and EZH2 expression in TNBC tissues, adjacent nontumor tissues and cell lines. Western blot, CCK8, Transwell, cell colony formation and EdU cell proliferation, cell cycle analysis and luciferase reporter assays were used to determine the regulation of miR-33a/EZH2 in TNBC progression. Results MiR-33a was significantly downregulated in TNBC tissues and cell lines. MiR-33a overexpression in TNBC cells significantly inhibited cell growth and mobility and induced G1 cell cycle arrest. The luciferase reporter assay revealed that EZH2 is a direct target of miR-33a and that it was upregulated in TNBC tissues and cell lines. There was a negative correlation between miR-33a and EZH2 expression in TNBC tissues. EZH2 knockdown exerted similar inhibitory effects, while ectopic expression of EZH2 showed suppressive effects on malignant behaviors induced by miR-33a overexpression in TNBC cells. Conclusions These findings revealed that miR-33a is a tumor-suppressive miRNA in TNBC and can inhibit proliferation and mobility and induce G1 cell cycle arrest by directly targeting EZH2.
Collapse
Affiliation(s)
- Zeng Weihua
- Department of Oncology, Panyu District Cancer Institute, Guangzhou Panyu Central Hospital, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511486 People's Republic of China
| | - Zou Guorong
- Department of Oncology, Panyu District Cancer Institute, Guangzhou Panyu Central Hospital, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511486 People's Republic of China
| | - Cao Xiaolong
- Department of Oncology, Panyu District Cancer Institute, Guangzhou Panyu Central Hospital, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511486 People's Republic of China
| | - Li Weizhan
- Department of Oncology, Panyu District Cancer Institute, Guangzhou Panyu Central Hospital, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511486 People's Republic of China
| |
Collapse
|
157
|
Dees S, Pontiggia L, Jasmin JF, Mercier I. Phosphorylated STAT3 (Tyr705) as a biomarker of response to pimozide treatment in triple-negative breast cancer. Cancer Biol Ther 2020; 21:506-521. [PMID: 32164483 PMCID: PMC7515519 DOI: 10.1080/15384047.2020.1726718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) displays an aggressive clinical course, heightened metastatic potential, and is linked to poor survival rates. Through its lack of expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), this subtype remains unresponsive to traditional targeted therapies. Undesirable and sometimes life-threatening side effects associated with current chemotherapeutic agents warrant the development of more targeted treatment options. Targeting signal transducer and activator of transcription 3 (STAT3), a transcription factor implicated in breast cancer (BCa) progression, has proven to be an efficient approach to halt cancer growth in vitro and in vivo. Currently, there are no FDA-approved STAT3 inhibitors for TNBC. Although pimozide, a FDA-approved antipsychotic drug, has been attributed a role as a STAT3 inhibitor in several cancers, its role on this pathway remains unexplored in TNBC. As a "one size fits all" approach cannot be applied to TNBC therapies due to the heterogeneous nature of this aggressive cancer, we hypothesized that STAT3 could be a novel biomarker of response to guide pimozide therapy. Using human cell lines representative of four TNBC subtypes (basal-like 1, basal-like 2, mesenchymal-like, mesenchymal stem-like), our current report demonstrates that pimozide significantly reduced their invasion and migration, an effect that was predicted by STAT3 phosphorylation on tyrosine residue 705 (Tyr705). Mechanistically, phosphorylated STAT3 (Tyr705) inhibition resulting from pimozide treatment caused a downregulation of downstream transcriptional targets such as matrix metalloproteinase-9 (MMP-9) and vimentin, both implicated in invasion and migration. The identification of biomarkers of response to TNBC treatments is an active area of research in the field of precision medicine and our results propose phosphorylated STAT3 (Tyr705) as a novel biomarker to guide pimozide treatment as an inhibitor of invasion and migration.
Collapse
Affiliation(s)
- Sundee Dees
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Laura Pontiggia
- Department of Mathematics, Physics and Statistics, Misher College of Arts and Sciences, University of the Sciences, Philadelphia, PA, USA
| | - Jean-Francois Jasmin
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Isabelle Mercier
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA.,Program in Personalized Medicine and Targeted Therapeutics, University of the Sciences, Philadelphia, PA, USA
| |
Collapse
|
158
|
Grellety T. [Androgen receptor-positive triple negative breast cancer: From biology to therapy]. Bull Cancer 2020; 107:506-516. [PMID: 32145961 DOI: 10.1016/j.bulcan.2019.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 10/24/2022]
Abstract
A subgroup of androgen receptor-expressing tumors represents approximately 30 % of all triple negative tumors. The androgen receptor and its signaling pathways have a central biological role in this tumor entity. These triple negative androgen receptor-positive tumors occur in older patients and do not appear to have a better prognosis compared to other triple negative tumors. In addition to androgen receptor-expression, these tumors are genomically characterized by a high frequency of PIK3CA activating mutation. Three clinical trials reported efficacy data for anti-androgens (bicalutamide, abiraterone acetate and enzalutamide) based on strong preclinical rationale. These trials report clinical benefit rates in about one in five patients. These encouraging but still limited results make a case for the identification of predictive response factors and therapeutic combinations to improve response rates. This review will provide an update on the biological and clinical knowledge of this tumoral subgroup that opens the way to non-cytotoxic anti-androgen therapies.
Collapse
Affiliation(s)
- Thomas Grellety
- Centre hospitalier de la Côte Basque, service d'oncologie médicale, 13, avenue de l'Interne Jacques-Loeb, 64100 Bayonne, France; Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux, France.
| |
Collapse
|
159
|
Shen Y, Schmidt BUS, Kubitschke H, Morawetz EW, Wolf B, Käs JA, Losert W. Detecting heterogeneity in and between breast cancer cell lines. CANCER CONVERGENCE 2020; 4:1. [PMID: 32090168 PMCID: PMC6997265 DOI: 10.1186/s41236-020-0010-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 01/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cellular heterogeneity in tumor cells is a well-established phenomenon. Genetic and phenotypic cell-to-cell variability have been observed in numerous studies both within the same type of cancer cells and across different types of cancers. Another known fact for metastatic tumor cells is that they tend to be softer than their normal or non-metastatic counterparts. However, the heterogeneity of mechanical properties in tumor cells are not widely studied. Results Here we analyzed single-cell optical stretcher data with machine learning algorithms on three different breast tumor cell lines and show that similar heterogeneity can also be seen in mechanical properties of cells both within and between breast tumor cell lines. We identified two clusters within MDA-MB-231 cells, with cells in one cluster being softer than in the other. In addition, we show that MDA-MB-231 cells and MDA-MB-436 cells which are both epithelial breast cancer cell lines with a mesenchymal-like phenotype derived from metastatic cancers are mechanically more different from each other than from non-malignant epithelial MCF-10A cells. Conclusion Since stiffness of tumor cells can be an indicator of metastatic potential, this result suggests that metastatic abilities could vary within the same monoclonal tumor cell line.
Collapse
Affiliation(s)
- Yang Shen
- 1Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742 USA
| | - B U Sebastian Schmidt
- 1Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742 USA
| | - Hans Kubitschke
- 2Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstr. 5, 04103 Leipzig, Germany
| | - Erik W Morawetz
- 2Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstr. 5, 04103 Leipzig, Germany
| | - Benjamin Wolf
- Leipzig University Medical Center, Department of Obstetrics and Gynecology, Liebigstr. 20a, 04103 Leipzig, Germany
| | - Josef A Käs
- 2Peter Debye Institute for Soft Matter Physics, Leipzig University, Linnéstr. 5, 04103 Leipzig, Germany
| | - Wolfgang Losert
- 1Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
160
|
Costea T, Vlad OC, Miclea LC, Ganea C, Szöllősi J, Mocanu MM. Alleviation of Multidrug Resistance by Flavonoid and Non-Flavonoid Compounds in Breast, Lung, Colorectal and Prostate Cancer. Int J Mol Sci 2020; 21:E401. [PMID: 31936346 PMCID: PMC7013436 DOI: 10.3390/ijms21020401] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the manuscript is to discuss the influence of plant polyphenols in overcoming multidrug resistance in four types of solid cancers (breast, colorectal, lung and prostate cancer). Effective treatment requires the use of multiple toxic chemotherapeutic drugs with different properties and targets. However, a major cause of cancer treatment failure and metastasis is the development of multidrug resistance. Potential mechanisms of multidrug resistance include increase of drug efflux, drug inactivation, detoxification mechanisms, modification of drug target, inhibition of cell death, involvement of cancer stem cells, dysregulation of miRNAs activity, epigenetic variations, imbalance of DNA damage/repair processes, tumor heterogeneity, tumor microenvironment, epithelial to mesenchymal transition and modulation of reactive oxygen species. Taking into consideration that synthetic multidrug resistance agents have failed to demonstrate significant survival benefits in patients with different types of cancer, recent research have focused on beneficial effects of natural compounds. Several phenolic compounds (flavones, phenolcarboxylic acids, ellagitannins, stilbens, lignans, curcumin, etc.) act as chemopreventive agents due to their antioxidant capacity, inhibition of proliferation, survival, angiogenesis, and metastasis, modulation of immune and inflammatory responses or inactivation of pro-carcinogens. Moreover, preclinical and clinical studies revealed that these compounds prevent multidrug resistance in cancer by modulating different pathways. Additional research is needed regarding the role of phenolic compounds in the prevention of multidrug resistance in different types of cancer.
Collapse
Affiliation(s)
- Teodora Costea
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Oana Cezara Vlad
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - Luminita-Claudia Miclea
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Research Excellence Center in Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Constanta Ganea
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Maria-Magdalena Mocanu
- Department of Biophysics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (O.C.V.); (C.G.)
| |
Collapse
|
161
|
Wu J, Mamidi TKK, Zhang L, Hicks C. Deconvolution of the Genomic and Epigenomic Interaction Landscape of Triple-Negative Breast Cancer. Cancers (Basel) 2019; 11:cancers11111692. [PMID: 31683572 PMCID: PMC6896043 DOI: 10.3390/cancers11111692] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/07/2019] [Accepted: 10/19/2019] [Indexed: 12/26/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive form of breast cancer. Emerging evidenced suggests that both genetics and epigenetic factors play a role in the pathogenesis of TNBC. However, oncogenic interactions and cooperation between genomic and epigenomic variation have not been characterized. The objective of this study was to deconvolute the genomic and epigenomic interaction landscape in TNBC using an integrative genomics approach, which integrates information on germline, somatic, epigenomic and gene expression variation. We hypothesized that TNBC originates from a complex interplay between genomic (both germline and somatic variation) and epigenomic variation. We further hypothesized that these complex arrays of interacting genomic and epigenomic factors affect entire molecular networks and signaling pathways which, in turn, drive TNBC. We addressed these hypotheses using germline variation from genome-wide association studies and somatic, epigenomic and gene expression variation from The Cancer Genome Atlas (TCGA). The investigation revealed signatures of functionally related genes containing germline, somatic and epigenetic variations. DNA methylation had an effect on gene expression. Network and pathway analysis revealed molecule networks and signaling pathways enriched for germline, somatic and epigenomic variation, among them: Role of BRCA1 in DNA Damage Response, Hereditary Breast Cancer Signaling, Molecular Mechanisms of Cancer, Estrogen-Dependent Breast Cancer, p53, MYC Mediated Apoptosis, and PTEN Signaling pathways. The investigation revealed that integrative genomics is a powerful approach for deconvoluting the genomic-epigenomic interaction landscape in TNBC. Further studies are needed to understand the biological mechanisms underlying oncogenic interactions between genomic and epigenomic factors in TNBC.
Collapse
Affiliation(s)
- Jiande Wu
- Department of Genetics, Louisiana State University Health Sciences Center, School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA.
| | - Tarun Karthik Kumar Mamidi
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35233, USA.
| | - Lu Zhang
- Department of Public Health Sciences, Clemson University, 513 Edwards Hall, Clemson, SC 29634, USA.
| | - Chindo Hicks
- Department of Genetics, Louisiana State University Health Sciences Center, School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA.
| |
Collapse
|
162
|
Attia YM, El-Kersh DM, Ammar RA, Adel A, Khalil A, Walid H, Eskander K, Hamdy M, Reda N, Mohsen NE, Al-Toukhy GM, Mansour MT, Elmazar MM. Inhibition of aldehyde dehydrogenase-1 and p-glycoprotein-mediated multidrug resistance by curcumin and vitamin D3 increases sensitivity to paclitaxel in breast cancer. Chem Biol Interact 2019; 315:108865. [PMID: 31628941 DOI: 10.1016/j.cbi.2019.108865] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/19/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
Treatment of breast cancer by paclitaxel (PAX) often encounters therapeutic failure most likely caused by innate/acquired resistance. Cancer stem cells (CSCs) and multidrug resistance complex (MDR-1 or P-glycoprotein) overexpression are main mechanisms implicated in chemoresistance. Increased aldehyde dehrogenase-1 (ALDH-1) was previously correlated with the stemness features of CSCs and hence is used as a marker for identification and CSCs targeting. The present study, therefore, aimed at investigating the effect of both curcumin (CUR) and vitamin D3 (D3) on MDR-1 and ALDH-1 expression and consequently the resistance to PAX both in vitro and in vivo. CUR was isolated from Turmeric rhizomes and identified using UPLC-ESI-MS/MS. For in vitro studies, the antiproliferative effect of PAX, CUR, 1,25(OH)2D3 (the active form of D3, also known as calcitriol) was determined, each alone and combined (PAX+CUR, PAX+1,25(OH)2D3, and PAX+CUR+1,25(OH)2D3) on MCF-7 breast cancer cells. Ehrlich ascites carcinoma solid tumor animal model was also used for in vivo studies. Combining CUR and/or 1,25(OH)2D3 to PAX showed synergistic cytotoxic interaction on MCF-7 cells. The apoptotic potential was also enhanced, as evidenced by a significant increase in caspase-7 and -9 as well as the pro-apoptotic Bax whereas a decrease in Bcl-2 levels was reported. Combining CUR and 1,25(OH)2D3 to PAX caused a downregulation in both MDR-1 and ALDH-1 gene expression in MCF-7 besides a decrease in their protein levels. In vivo, the triple therapy group (PAX+CUR+D3) showed the least tumor size. It also showed the lowest levels of MDR-1 and ALDH-1. PAX alone, however, showed increased levels of MDR-1 and ALDH-1 compared to control. Overall, the present study showed that PAX, as a monotherapy, demonstrated acquired resistance possibly by increasing MDR-1 expression and enriching CSCs population, as evidenced by increased ALDH-1. However, using CUR and D3 enhanced tumor response to PAX.
Collapse
Affiliation(s)
- Yasmeen M Attia
- Pharmacology Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Dina M El-Kersh
- Pharmacognosy Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Reham A Ammar
- Pharmacology Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Aliaa Adel
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Aya Khalil
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Hoda Walid
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Kirullos Eskander
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Mohamed Hamdy
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Nada Reda
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Nour Elhoda Mohsen
- Senior Students, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Ghada M Al-Toukhy
- Virology and Immunology Department, Children's Cancer Hospital, 57357, Cairo, Egypt
| | - Mohamed Tarek Mansour
- Virology and Immunology Department, Children's Cancer Hospital, 57357, Cairo, Egypt; Virology and Immunology Department, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed M Elmazar
- Pharmacology Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
163
|
Sridharan S, Howard CM, Tilley AMC, Subramaniyan B, Tiwari AK, Ruch RJ, Raman D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front Oncol 2019; 9:1003. [PMID: 31681564 PMCID: PMC6805781 DOI: 10.3389/fonc.2019.01003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/18/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer stem cells (BCSCs) play a vital role in tumor progression and metastasis. They are heterogeneous and inherently radio- and chemoresistant. They have the ability to self-renew and differentiate into non-BCSCs. These determinants of BCSCs including the plasticity between the mesenchymal and epithelial phenotypes often leads to minimal residual disease (MRD), tumor relapse, and therapy failure. By studying the resistance mechanisms in BCSCs, a combinatorial therapy can be formulated to co-target BCSCs and bulk tumor cells. This review addresses breast cancer stemness and molecular underpinnings of how the cancer stemness can lead to pharmacological resistance. This might occur through rewiring of signaling pathways and modulated expression of various targets that support survival and self-renewal, clonogenicity, and multi-lineage differentiation into heterogeneous bulk tumor cells following chemotherapy. We explore emerging novel and alternative molecular targets against BC stemness and chemoresistance involving survival, drug efflux, metabolism, proliferation, cell migration, invasion, and metastasis. Strategic targeting of such vulnerabilities in BCSCs may overcome the chemoresistance and increase the longevity of the metastatic breast cancer patients.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Cory M. Howard
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | | | | | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
| | - Randall J. Ruch
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
164
|
Knight JF, Sung VYC, Kuzmin E, Couzens AL, de Verteuil DA, Ratcliffe CDH, Coelho PP, Johnson RM, Samavarchi-Tehrani P, Gruosso T, Smith HW, Lee W, Saleh SM, Zuo D, Zhao H, Guiot MC, Davis RR, Gregg JP, Moraes C, Gingras AC, Park M. KIBRA (WWC1) Is a Metastasis Suppressor Gene Affected by Chromosome 5q Loss in Triple-Negative Breast Cancer. Cell Rep 2019; 22:3191-3205. [PMID: 29562176 PMCID: PMC5873529 DOI: 10.1016/j.celrep.2018.02.095] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 01/15/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) display a complex spectrum of mutations and chromosomal aberrations. Chromosome 5q (5q) loss is detected in up to 70% of TNBCs, but little is known regarding the genetic drivers associated with this event. Here, we show somatic deletion of a region syntenic with human 5q33.2–35.3 in a mouse model of TNBC. Mechanistically, we identify KIBRA as a major factor contributing to the effects of 5q loss on tumor growth and metastatic progression. Re-expression of KIBRA impairs metastasis in vivo and inhibits tumorsphere formation by TNBC cells in vitro. KIBRA functions co-operatively with the protein tyrosine phosphatase PTPN14 to trigger mechanotransduction-regulated signals that inhibit the nuclear localization of oncogenic transcriptional co-activators YAP/TAZ. Our results argue that the selective advantage produced by 5q loss involves reduced dosage of KIBRA, promoting oncogenic functioning of YAP/TAZ in TNBC. Reduced KIBRA expression is associated with chr 5q loss in breast cancer Restoring Kibra expression inhibits metastatic dissemination in mice KIBRA impairs the self-renewal capacity of triple-negative breast cancer cells KIBRA blocks mechanotransduction signals required for YAP/TAZ activation
Collapse
Affiliation(s)
- Jennifer F Knight
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | - Vanessa Y C Sung
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H2W 1S6, Canada
| | - Elena Kuzmin
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H2W 1S6, Canada
| | - Amber L Couzens
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | | | - Colin D H Ratcliffe
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H2W 1S6, Canada
| | - Paula P Coelho
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H2W 1S6, Canada
| | - Radia M Johnson
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | | | - Tina Gruosso
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Oncology, McGill University, Montreal, QC H2W 1S6, Canada
| | - Harvey W Smith
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | - Wontae Lee
- Department of Biomedical Engineering, McGill University, Montreal, QC H3A 2B4, Canada
| | - Sadiq M Saleh
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | - Dongmei Zuo
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | - Hong Zhao
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada
| | - Marie-Christine Guiot
- Montreal Neurological Institute, Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ryan R Davis
- Department of Pathology and Laboratory Medicine, University of California at Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jeffrey P Gregg
- Department of Pathology and Laboratory Medicine, University of California at Davis School of Medicine, Sacramento, CA 95817, USA
| | - Christopher Moraes
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biomedical Engineering, McGill University, Montreal, QC H3A 2B4, Canada; Department of Chemical Engineering, McGill University, Montreal, QC H3A 0C5, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 0B1, Canada; Department of Biochemistry, McGill University, Montreal, QC H2W 1S6, Canada; Department of Oncology, McGill University, Montreal, QC H2W 1S6, Canada.
| |
Collapse
|
165
|
Net JM, Whitman GJ, Morris E, Brandt KR, Burnside ES, Giger ML, Ganott M, Sutton EJ, Zuley ML, Rao A. Relationships Between Human-Extracted MRI Tumor Phenotypes of Breast Cancer and Clinical Prognostic Indicators Including Receptor Status and Molecular Subtype. Curr Probl Diagn Radiol 2019; 48:467-472. [PMID: 30270031 PMCID: PMC6387644 DOI: 10.1067/j.cpradiol.2018.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE The purpose of this study was to investigate if human-extracted MRI tumor phenotypes of breast cancer could predict receptor status and tumor molecular subtype using MRIs from The Cancer Genome Atlas project. MATERIALS AND METHODS Our retrospective interpretation study utilized the analysis of HIPAA-compliant breast MRI data from The Cancer Imaging Archive. One hundred and seven preoperative breast MRIs of biopsy proven invasive breast cancers were analyzed by 3 fellowship-trained breast-imaging radiologists. Each study was scored according to the Breast Imaging Reporting and Data System lexicon for mass and nonmass features. The Spearman rank correlation was used for association analysis of continuous variables; the Kruskal-Wallis test was used for associating continuous outcomes with categorical variables. The Fisher-exact test was used to assess correlations between categorical image-derived features and receptor status. Prediction of estrogen receptor (ER), progesterone receptor, human epidermal growth factor receptor, and molecular subtype were performed using random forest classifiers. RESULTS ER+ tumors were associated with the absence of rim enhancement (P = 0.019, odds ratio [OR] 5.5), heterogeneous internal enhancement (P = 0.02, OR 6.5), peritumoral edema (P = 0.0001, OR 10.0), and axillary adenopathy (P = 0.04, OR 4.4). ER+ tumors were smaller than ER- tumors (23.7 mm vs 29.2 mm, P = 0.02, OR 8.2). All of these variables except the lack of axillary adenopathy were also associated with progesterone receptor+ status. Luminal A tumors (n = 57) were smaller compared to nonLuminal A (21.8 mm vs 27.5 mm, P = 0.035, OR 7.3) and lacked peritumoral edema (P = 0.001, OR 6.8). Basal like tumors were associated with heterogeneous internal enhancement (P = 0.05, OR 10.1), rim enhancement (P = 0.05, OR6.9), and perituomral edema (P = 0.0001, OR 13.8). CONCLUSIONS Human extracted MRI tumor phenotypes may be able to differentiate those tumors with a more favorable clinical prognosis from their more aggressive counterparts.
Collapse
Affiliation(s)
- Jose M Net
- Department of Radiology, University of Miami, Miller School of Medicine, Miami, FL.
| | - Gary J Whitman
- Department of Diagnostic Imaging, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Elizabteh Morris
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | - Marie Ganott
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA
| | - Elizabeth J Sutton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Arvind Rao
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
166
|
Huang Y, Fang J, Lu W, Wang Z, Wang Q, Hou Y, Jiang X, Reizes O, Lathia J, Nussinov R, Eng C, Cheng F. A Systems Pharmacology Approach Uncovers Wogonoside as an Angiogenesis Inhibitor of Triple-Negative Breast Cancer by Targeting Hedgehog Signaling. Cell Chem Biol 2019; 26:1143-1158.e6. [PMID: 31178408 PMCID: PMC6697584 DOI: 10.1016/j.chembiol.2019.05.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 03/29/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous disease that lacks clinically actionable genetic alterations that limit targeted therapies. Here we explore a systems pharmacology approach that integrates drug-target networks and large-scale genomic profiles of TNBC and identify wogonoside, one of the major active flavonoids, as a potent angiogenesis inhibitor. We validate that wogonoside attenuates cell migration, tube formation, and rat aorta microvessel outgrowth, and reduces formation of blood vessels in chicken chorioallantoic membrane and TNBC cell-induced Matrigel plugs. In addition, wogonoside inhibits growth and angiogenesis in TNBC cell xenograft models. This network-based approach predicts, and we empirically validate, wogonoside's antiangiogenic effects resulting from vascular endothelial growth factor secretion. Mechanistically, wogonoside inhibits Gli1 nuclear translocation and transcriptional activities associated with Hedgehog signaling, by promoting Smoothened degradation in a proteasome-dependent mechanism. This study offers a powerful, integrated, systems pharmacology-based strategy for oncological drug discovery and identifies wogonoside as a potential TNBC angiogenesis inhibitor.
Collapse
MESH Headings
- Angiogenesis Inhibitors/chemistry
- Angiogenesis Inhibitors/isolation & purification
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Biological Products/chemistry
- Biological Products/isolation & purification
- Biological Products/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Female
- Flavanones/chemistry
- Flavanones/isolation & purification
- Flavanones/pharmacology
- Glucosides/chemistry
- Glucosides/isolation & purification
- Glucosides/pharmacology
- Hedgehog Proteins/antagonists & inhibitors
- Hedgehog Proteins/metabolism
- Humans
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Scutellaria baicalensis/chemistry
- Signal Transduction/drug effects
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Yujie Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jiansong Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Zihao Wang
- Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Xingwu Jiang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ofer Reizes
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Justin Lathia
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44915, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
167
|
Schilling J, Busch S, Heinrich G, Heinig K, Martin Kurbacher C, Klare P, Pöllinger B. Treatment patterns, health care resource use and outcomes in metastatic triple-negative breast cancer in Germany: retrospective chart review study (OBTAIN). Curr Med Res Opin 2019; 35:1405-1414. [PMID: 30880482 DOI: 10.1080/03007995.2019.1595551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background: There is limited data on the real-life situation and outcomes of patients with metastatic triple-negative breast cancer (mTNBC) in Germany. The aim of this chart review was to describe the current treatment patterns, resource use and outcomes in this patient group. Methods: Retrospective data collection in 30 gyneco-oncological sites (hospitals and office-based) across Germany between January and April 2017. Index date was defined as initiation of treatment with gemcitabine, vinorelbin, capecitabine or eribulin therapy following discontinuation of taxane and/or anthracycline therapy. Results: In the 91 evaluable patients, median time between primary diagnosis and index date was 20.9 months (range 0-187 months). Ten percent of patients had no distant metastases, while 57% had newly diagnosed metastases. Cancer stage at index date was mostly IV (82 patients). A number of 135 different regimens (monotherapy or combination therapy) were used. For first-line chemo treatment, 29 patients received monotherapy and 54 patients combination therapy. Bevacizumab and paclitaxel were also the most frequently used single substances among all therapy lines together and for first-line therapy. While taxanes were at least occasionally administered for second-line therapy, no patient received taxanes for third-line therapy. Chemotherapy modifications in terms of dose reduction or treatment interruption were rare. However, the therapy was terminated in more than two thirds of all cases. Fifty-nine patients were hospitalized at least once. For first-, second- and third-line therapy, median overall survival was 19.1/10.8/14.6 months, and median progression-free survival was 7.7/2.5/5.6 months. Conclusion: In clinical routine, a wide variety of treatment approaches is applied, while outcomes in terms of survival are poor. New treatment options are needed for this challenging tumor type.
Collapse
Affiliation(s)
| | | | | | - Karin Heinig
- d MVZ Poliklinik Spremberg GmbH , Spremberg , Germany
| | | | - Peter Klare
- f Praxisklinik Krebsheilkunde für Frauen/Brustzentrum , Berlin , Germany
| | | |
Collapse
|
168
|
Notch Signaling Activation as a Hallmark for Triple-Negative Breast Cancer Subtype. JOURNAL OF ONCOLOGY 2019; 2019:8707053. [PMID: 31379945 PMCID: PMC6657611 DOI: 10.1155/2019/8707053] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/19/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subgroup of 15%-20% of diagnosed breast cancer patients. It is generally considered to be the most difficult breast cancer subtype to deal with, due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), which usually direct targeted therapies. In this scenario, the current treatments of TNBC-affected patients rely on tumor excision and conventional chemotherapy. As a result, the prognosis is overall poor. Thus, the identification and characterization of targets for novel therapies are urgently required. The Notch signaling pathway has emerged to act in the pathogenesis and tumor progression of TNBCs. Firstly, Notch receptors are associated with the regulation of tumor-initiating cells (TICs) behavior, as well as with the aetiology of TNBCs. Secondly, there is a strong evidence that Notch pathway is a relevant player in mammary cancer stem cells maintenance and expansion. Finally, Notch receptors expression and activation strongly correlate with the aggressive clinicopathological and biological phenotypes of breast cancer (e.g., invasiveness and chemoresistance), which are relevant characteristics of TNBC subtype. The purpose of this up-to-date review is to provide a detailed overview of the specific role of all four Notch receptors (Notch1, Notch2, Notch3, and Notch4) in TNBCs, thus identifying the Notch signaling pathway deregulation/activation as a pathognomonic feature of this breast cancer subtype. Furthermore, this review will also discuss recent information associated with different therapeutic options related to the four Notch receptors, which may be useful to evaluate prognostic or predictive indicators as well as to develop new therapies aimed at improving the clinical outcome of TNBC patients.
Collapse
|
169
|
Hu XL, Cai Q, Gao J, Field RA, Chen GR, Jia N, Zang Y, Li J, He XP. Self-Assembled 2D Glycoclusters for the Targeted Delivery of Theranostic Agents to Triple-Negative Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:22181-22187. [PMID: 31150201 DOI: 10.1021/acsami.9b06016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Triple-negative breast cancer (TNBC) is a devastating disease worldwide, for which targeted imaging and therapeutic agents remain elusive. There has been growing awareness that carbohydrates are valuable as drug candidates and targeting agents for a variety of human diseases, including cancers that overexpress carbohydrate receptors on the cell surface. Here, we develop a two-dimensional (2D) glycocluster by means of simple, stepwise self-assembly for the targeted delivery of theranostic agents to TNBC cells that express mannose receptors (MRs) on the cell surface. Human serum albumin, which contains a variety of hydrophobic pockets capable of accommodating small molecules, was used to simultaneously encapsulate a mannose-based glycoprobe and a commercial photosensitizer (i.e., Ce6). The multicomponent "neoglycoprotein" formed was used to self-assemble with 2D MnO2, producing 2D glycoclusters, which could be selectively internalized by a TNBC cell line (MDA-MB-231) as facilitated by binding to the transmembrane MR. The intracellular degradation of the 2D MnO2 backbone by biothiols then released Ce6 for cell imaging and, subsequently, photodynamic therapy. This study provides insights into the development of carbohydrate-based materials for targeted, stimuli-responsive theranostics of TNBC.
Collapse
Affiliation(s)
- Xi-Le Hu
- Key Laboratory for Advanced Materials & Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , P. R. China
| | - Quanyu Cai
- Department of Radiology , Eastern Hepatobiliary Surgery Hospital , Shanghai 200438 , P. R. China
| | - Jie Gao
- Key Laboratory for Advanced Materials & Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , P. R. China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Road , Shanghai 201203 , P. R. China
| | - Robert A Field
- Department of Biological Chemistry , John Innes Centre, Norwich Research Park , Norwich NR4 7UH , U.K
| | - Guo-Rong Chen
- Key Laboratory for Advanced Materials & Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , P. R. China
| | - Ningyang Jia
- Department of Radiology , Eastern Hepatobiliary Surgery Hospital , Shanghai 200438 , P. R. China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Road , Shanghai 201203 , P. R. China
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica , Chinese Academy of Sciences , 189 Guo Shoujing Road , Shanghai 201203 , P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials & Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering , East China University of Science and Technology , 130 Meilong Road , Shanghai 200237 , P. R. China
| |
Collapse
|
170
|
Gluz O, Kolberg-Liedtke C, Prat A, Christgen M, Gebauer D, Kates R, Paré L, Grischke EM, Forstbauer H, Braun M, Warm M, Hackmann J, Uleer C, Aktas B, Schumacher C, Kuemmel S, Wuerstlein R, Pelz E, Nitz U, Kreipe HH, Harbeck N. Efficacy of deescalated chemotherapy according to PAM50 subtypes, immune and proliferation genes in triple-negative early breast cancer: Primary translational analysis of the WSG-ADAPT-TN trial. Int J Cancer 2019; 146:262-271. [PMID: 31162838 DOI: 10.1002/ijc.32488] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022]
Abstract
In the neoadjuvant WSG-ADAPT-TN trial, 12-week nab-paclitaxel + carboplatin (nab-pac/carbo) was highly effective and superior to nab-paclitaxel + gemcitabine (nab-pac/gem) in triple-negative breast cancer regarding pathological complete response (pCR). Predictive markers for deescalated taxane/carbo use in TNBC need to be identified. Patients received 4 × nab-pac 125 mg/m2 (plus carbo AUC2 or gem 1,000 mg/m2 d1,8 q21). Expression of 119 genes and PAM50 scores by nCounter were available in 306/336 pretherapeutic samples. Interim survival analysis was planned after 36 months median follow-up. Basal-like (83.3%) compared to other subtypes was positively associated with pCR (38% vs. 20%, p = 0.015), as was lower HER2 score (p < 0.001). Proliferation biomarkers were positively associated with pCR, that is, PAM50 proliferation, ROR scores (all p < 0.004), higher Ki-67 (IHC; p < 0.001). For nab-pac/carbo, expression of immunological (CD8, PD1 and PFDL1) genes and proliferation markers (proliferation and ROR scores, MKI67, CDC20, NUF2, KIF2C, CENPF, EMP3 and TYMS) were positively associated with pCR (p < 0.05 for all). For nab-pac/gem, angiogenesis genes were negatively associated with pCR (ANGPTL4: p = 0.05; FGFR4: p = 0.02; VEGFA: p = 0.03). pCR after 12 weeks was strongly associated with favorable outcome (3y event-free survival: 92% vs. 71%, p < 0.001). In early TNBC, basal-like subtype, higher Ki-67 (IHC) and lower HER2 score were, associated with chemosensitivity. Chemoresistance pathways differed between the two taxane based combinations. Combination of proliferation/immune markers and PAM50 subtype could allow patient selection for further deescalated chemotherapy and/or immune treatment approaches.
Collapse
Affiliation(s)
- Oleg Gluz
- West German Study Group, Mönchengladbach, Germany.,Ev. Hospital Bethesda, Breast Center Niederrhein, Mönchengladbach, Germany.,University Clinics Cologne, Cologne, Germany
| | | | - Aleix Prat
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | | | - Ronald Kates
- West German Study Group, Mönchengladbach, Germany
| | - Laia Paré
- Department of Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | | | - Michael Braun
- Rotkreuz Clinics Munich, Breast Center, Munich, Germany
| | - Mathias Warm
- City Hospital Holweide, Breast Center, Cologne, Germany
| | | | | | - Bahriye Aktas
- University Clinics Essen, Women's Clinic, Essen, Germany.,University Clinics Leipzig, Women's Clinic, Leipzig, Germany
| | | | | | - Rachel Wuerstlein
- West German Study Group, Mönchengladbach, Germany.,Breast Center, Dept. OB&GYN, University of Munich (LMU) and CCCLMU, Munich, Germany
| | - Enrico Pelz
- Institute of Pathology Viersen, Viersen, Germany
| | - Ulrike Nitz
- West German Study Group, Mönchengladbach, Germany.,Ev. Hospital Bethesda, Breast Center Niederrhein, Mönchengladbach, Germany
| | | | - Nadia Harbeck
- West German Study Group, Mönchengladbach, Germany.,Breast Center, Dept. OB&GYN, University of Munich (LMU) and CCCLMU, Munich, Germany
| |
Collapse
|
171
|
Yildirim N, Simsek M, Aldemir MN, Bilici M, Tekin SB. Relationship between 18-FDG-PET/CT and Clinicopathological Features and Pathological Responses in Patients with Locally Advanced Breast Cancers. Eurasian J Med 2019; 51:154-159. [PMID: 31258356 DOI: 10.5152/eurasianjmed.2018.18036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective We investigated the relationship between the maximum standardized uptake value (SUVmax) of whole-body positron emission tomography/computed tomography (PET/CT) performed before treatment and the demographical and histopathological features in locally advanced breast cancer (LABC), as well as the role of PET/CT in the evaluation of pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). Materials and Methods Fifty-one LABC patients who received NAC in our center between 2011 and 2015 were retrospectively analyzed. Basal PET/CT was performed in all the patients before NAC. The SUVmax levels and demographical and histopathological results were compared. The relationship between the SUVmax values after NAC and pathological responses were evaluated. Results The mean age of the patients was 49 (32-69) years. PET/CT performed after NAC showed complete response in 20 patients (39.2%), partial response in 28 patients (54.9%), stable disease in 2 patients (3.9%), and progressive disease in 1 patient (2%). There was no significant difference between the mean SUVmax values of the patients according to age (>50 and ≤50 years), menopausal status, tumor localization, clinical stage, and grade. The mean SUVmax value was higher in the triple-negative group than those in the HER2 positive and luminal groups. There was a significant difference in the SUVmax values between the group that achieved pCR after NAC and the group that could not achieve pCR (SUVmax value for breast 2.92 vs. 0.30; p=0.01; SUVmax value for axilla 1.5 vs. 0.0, p=0.02). Conclusion The SUVmax values are independent of demographical features. There was a significant relationship between the pCR and SUVmax values after NAC. PET/CT could be useful in the evaluation of patients to predict the biological characteristics of tumors.
Collapse
Affiliation(s)
- Nilgun Yildirim
- Department of Medical Oncology, Dr. Ersin Arslan Training and Research Hospital, Gaziantep, Turkey
| | - Melih Simsek
- Department of Medical Oncology, Atatürk University School of Medicine, Erzurum, Turkey
| | - Mehmet Naci Aldemir
- Department of Medical Oncology, Erzincan University School of Medicine, Erzincan, Turkey
| | - Mehmet Bilici
- Department of Medical Oncology, Atatürk University School of Medicine, Erzurum, Turkey
| | - Salim Basol Tekin
- Department of Medical Oncology, Atatürk University School of Medicine, Erzurum, Turkey
| |
Collapse
|
172
|
Chen J, Qian X, He Y, Han X, Pan Y. Novel key genes in triple‐negative breast cancer identified by weighted gene co‐expression network analysis. J Cell Biochem 2019; 120:16900-16912. [PMID: 31081967 DOI: 10.1002/jcb.28948] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Jian Chen
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Xiaojun Qian
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Yifu He
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Xinghua Han
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| | - Yueyin Pan
- Department of Oncology The First Affiliated Hospital of University of Science and Technology of China Hefei China
| |
Collapse
|
173
|
Collina F, Aquino G, Brogna M, Cipolletta S, Buonfanti G, De Laurentiis M, Di Bonito M, Cantile M, Botti G. LncRNA HOTAIR up-regulation is strongly related with lymph nodes metastasis and LAR subtype of Triple Negative Breast Cancer. J Cancer 2019; 10:2018-2024. [PMID: 31205562 PMCID: PMC6548158 DOI: 10.7150/jca.29670] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/05/2019] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancers (TNBCs) represent a heterogeneous disease characterized by several molecular subtypes with different prognoses and responses to therapy. For a correct clinical management of TNBC patients the knowledge of the gene regulation mechanisms related to tumor progression and drug response has become fundamental. LncRNAs regulate gene expression through various processes, including chromatin modification, transcription and post-transcription and they are emerging as important cancer biomarkers being involved in tumor pathogenesis, metastatic progression and drug resistance. In this study we aimed to analyze the expression of the lncRNA HOTAIR, mainly involved in breast cancer disease, in a large case series of TNBC patients. We used ISH methods by a RNA probe to better define its staining in tumor tissues and its relation with clinical-pathological parameters and outcomes of patients. Our results show that high HOTAIR expression in tumor tissues is strongly correlated with lymph nodes metastasis (LNM) (p=0.039), as reported also for other tumor types, and has a direct strong association with Androgen Receptor (AR) expression (p= 0.019). These data confirm the prognostic role of HOTAIR in TNBC, and, its involvement in the regulation of AR pathway, suggests the possibility to establish new therapeutic strategies for AR+TNBC patients.
Collapse
Affiliation(s)
- Francesca Collina
- Pathology Unit, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| | - Gabriella Aquino
- Pathology Unit, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| | - Marianna Brogna
- Università degli Studi di Napoli "Federico II", Naples, Italy
| | | | | | - Michelino De Laurentiis
- Department of Breast Surgery and Cancer Prevention, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| | - Maurizio Di Bonito
- Pathology Unit, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| | - Monica Cantile
- Pathology Unit, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| | - Gerardo Botti
- Department of Breast Surgery and Cancer Prevention, ISTITUTO NAZIONALE TUMORI-IRCCS-FONDAZIONE G.PASCALE, Naples, ITALY
| |
Collapse
|
174
|
Wang Q, Xu M, Sun Y, Chen J, Chen C, Qian C, Chen Y, Cao L, Xu Q, Du X, Yang W. Gene Expression Profiling for Diagnosis of Triple-Negative Breast Cancer: A Multicenter, Retrospective Cohort Study. Front Oncol 2019; 9:354. [PMID: 31134153 PMCID: PMC6513966 DOI: 10.3389/fonc.2019.00354] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) accounts for 12–20% of all breast cancers. Diagnosis of TNBC is sometimes quite difficult based on morphological assessment and immunohistochemistry alone, particularly in the metastatic setting with no prior history of breast cancer. Methods: Molecular profiling is a promising diagnostic approach that has the potential to provide an objective classification of metastatic tumors with unknown primary. In this study, performance of a novel 90-gene expression signature for determination of the site of tumor origin was evaluated in 115 TNBC samples. For each specimen, expression profiles of the 90 tumor-specific genes were analyzed, and similarity scores were obtained for each of the 21 tumor types on the test panel. Predicted tumor type was compared to the reference diagnosis to calculate accuracy. Furthermore, rank product analysis was performed to identify genes that were differentially expressed between TNBC and other tumor types. Results: Analysis of the 90-gene expression signature resulted in an overall 97.4% (112/115, 95% CI: 0.92–0.99) agreement with the reference diagnosis. Among all specimens, the signature correctly classified 97.6% of TNBC from the primary site (41/42) and lymph node metastasis (41/42) and 96.8% of distant metastatic tumors (30/31). Furthermore, a list of genes, including AZGP1, KRT19, and PIGR, was identified as differentially expressed between TNBC and other tumor types, suggesting their potential use as discriminatory markers. Conclusion: Our results demonstrate excellent performance of a 90-gene expression signature for identification of tumor origin in a cohort of both primary and metastatic TNBC samples. These findings show promise for use of this novel molecular assay to aid in differential diagnosis of TNBC, particularly in the metastatic setting.
Collapse
Affiliation(s)
- Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | - Yizuo Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liyu Cao
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Qinghua Xu
- Canhelp Genomics, Hangzhou, China.,Institute of Machine Learning and Systems Biology, College of Electronics and Information Engineering, Tongji University, Shanghai, China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
175
|
Calcitriol Inhibits the Proliferation of Triple-Negative Breast Cancer Cells through a Mechanism Involving the Proinflammatory Cytokines IL-1 β and TNF- α. J Immunol Res 2019; 2019:6384278. [PMID: 31093512 PMCID: PMC6481021 DOI: 10.1155/2019/6384278] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/06/2019] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive tumors, with poor prognosis and high metastatic capacity. The aggressive behavior may involve inflammatory processes characterized by deregulation of molecules related to the immunological responses in which interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) are involved. It is known that calcitriol, the active vitamin D metabolite, modulates the synthesis of immunological mediators; however, its role in the regulation of IL-1β and TNF-α in TNBC has been scarcely studied. In the present study, we showed that TNBC cell lines SUM-229PE and HCC1806 expressed vitamin D, IL-1β, and TNF-α receptors. Moreover, calcitriol, its analogue EB1089, IL-1β, and TNF-α inhibited cell proliferation. In addition, we showed that synthesis of both IL-1β and TNF-α was stimulated by calcitriol and its analogue. Interestingly, the antiproliferative activity of calcitriol was significantly abrogated when the cells were treated with anti-IL-1β receptor 1 (IL-1R1) and anti-TNF-α receptor type 1 (TNFR1) antibodies. Furthermore, the combination of calcitriol with TNF-α resulted in a greater antiproliferative effect than either agent alone, in the two TNBC cell lines and an estrogen receptor-positive cell line. In summary, this study demonstrated that calcitriol exerted its antiproliferative effects in part by inducing the synthesis of IL-1β and TNF-α through IL-1R1 and TNFR1, respectively, in TNBC cells, highlighting immunomodulatory and antiproliferative functions of calcitriol in TNBC tumors.
Collapse
|
176
|
Caramelo O, Silva C, Caramelo F, Frutuoso C, Almeida-Santos T. The effect of neoadjuvant platinum-based chemotherapy in BRCA mutated triple negative breast cancers -systematic review and meta-analysis. Hered Cancer Clin Pract 2019; 17:11. [PMID: 30962858 PMCID: PMC6434625 DOI: 10.1186/s13053-019-0111-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Background Triple negative breast cancers (TNBC) are associated with an aggressive clinical course, earlier recurrence and short survival. BRCA – mutated tumours represent up to 25% of all TNBC. BRCA status is being studied as a predictive biomarker of response to platinum agents. However, the predictive role of BRCA status is still uncertain in this setting. Since TNBC is a very heterogeneous group of diseases, it is important to identify subsets of TNBC patients that may benefit from platinum-based therapy. This study aims to establish if the presence of a germline BRCA mutation in women with TNBC improves the pathologic complete response (pCR) after neoadjuvant chemotherapy with platinum compounds. Methods An extensive literature search was performed in MEDLINE, EMBASE and LILACS databases, WHO (WHO International Clinical Trials Registry Platform) and the Cochrane Controlled Trials Register Database, for online trial registries and conference proceedings. The measurement of pCR was assessed by pathology review of breast specimen and lymph nodes. Results The overall OR was computed using random effects models. Seven studies were included, comprising a total of 808 TNBC patients, among which 159 were BRCA mutated. Among mutated TNBC patients, 93 (93/159; 58.4%) achieved pCR, while 410 wildtype patients (410/808; 50.7%) showed pCR (OR 1.459 CI 95% [0.953–2.34] p = 0.082) although this result did not reach statistical significance. Conclusions This meta-analysis shows that the addition of platinum to chemotherapy regimens in the neoadjuvant setting increases pCR rate in BRCA – mutated as compared to wild-type TNBC patients. However, this trend did not achieve statistical significance. Trial registration CRD42018092341
Collapse
Affiliation(s)
- Olga Caramelo
- 1Gynecology Department, Coimbra Hospital and University Centre (CHUC), EPE, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Cristina Silva
- 2Faculty of Pharmacy of the University of Coimbra, Rua Filipe Simões n° 33, 3000-186 Coimbra, Portugal
| | - Francisco Caramelo
- 3Laboratory of Biostatistics and Medical Informatics, iCBR - Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Cristina Frutuoso
- 1Gynecology Department, Coimbra Hospital and University Centre (CHUC), EPE, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Teresa Almeida-Santos
- 4Centre for Fertility Preservation, Human Reproduction Department, Coimbra Hospital and University Centre (CHUC), EPE, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal.,5Faculty of Medicine of the University of Coimbra, Azinhaga de Santa Comba - Celas, 3000-548 Coimbra, Portugal
| |
Collapse
|
177
|
Wu J, Mamidi TKK, Zhang L, Hicks C. Integrating Germline and Somatic Mutation Information for the Discovery of Biomarkers in Triple-Negative Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16061055. [PMID: 30909550 PMCID: PMC6466377 DOI: 10.3390/ijerph16061055] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/22/2022]
Abstract
Recent advances in high-throughput genotyping and the recent surge of next generation sequencing of the cancer genomes have enabled discovery of germline mutations associated with an increased risk of developing breast cancer and acquired somatic mutations driving the disease. Emerging evidence indicates that germline mutations may interact with somatic mutations to drive carcinogenesis. However, the possible oncogenic interactions and cooperation between germline and somatic alterations in triple-negative breast cancer (TNBC) have not been characterized. The objective of this study was to investigate the possible oncogenic interactions and cooperation between genes containing germline and somatic mutations in TNBC. Our working hypothesis was that genes containing germline mutations associated with an increased risk developing breast cancer also harbor somatic mutations acquired during tumorigenesis, and that these genes are functionally related. We further hypothesized that TNBC originates from a complex interplay among and between genes containing germline and somatic mutations, and that these complex array of interacting genetic factors affect entire molecular networks and biological pathways which in turn drive the disease. We tested this hypothesis by integrating germline mutation information from genome-wide association studies (GWAS) with somatic mutation information on TNBC from The Cancer Genome Atlas (TCGA) using gene expression data from 110 patients with TNBC and 113 controls. We discovered a signature of 237 functionally related genes containing both germline and somatic mutations. We discovered molecular networks and biological pathways enriched for germline and somatic mutations. The top pathways included the hereditary breast cancer and role of BRCA1 in DNA damage response signaling pathways. In conclusion, this is the first large-scale and comprehensive analysis delineating possible oncogenic interactions and cooperation among and between genes containing germline and somatic mutations in TNBC. Genetic and somatic mutations, along with the genes discovered in this study, will require experimental functional validation in different ethnic populations. Functionally validated genetic and somatic variants will have important implications for the development of novel precision prevention strategies and discovery of prognostic markers in TNBC.
Collapse
Affiliation(s)
- Jiande Wu
- Department of Genetics and the Bioinformatics and Genomics Program, Louisiana State University Health Sciences Center, School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA.
| | - Tarun Karthik Kumar Mamidi
- Department of Genetics and the Bioinformatics and Genomics Program, Louisiana State University Health Sciences Center, School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA.
| | - Lu Zhang
- Louisiana Tumor Registry, Louisiana State University Health Sciences Center, School of Public Health, 2020 Gravier Street, New Orleans, LA 70112, USA.
| | - Chindo Hicks
- Department of Genetics and the Bioinformatics and Genomics Program, Louisiana State University Health Sciences Center, School of Medicine, 533 Bolivar Street, New Orleans, LA 70112, USA.
| |
Collapse
|
178
|
Gupta I, Sareyeldin RM, Al-Hashimi I, Al-Thawadi HA, Al Farsi H, Vranic S, Al Moustafa AE. Triple Negative Breast Cancer Profile, from Gene to microRNA, in Relation to Ethnicity. Cancers (Basel) 2019; 11:cancers11030363. [PMID: 30871273 PMCID: PMC6468678 DOI: 10.3390/cancers11030363] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most frequent cause of cancer-related deaths among women worldwide. It is classified into four major molecular subtypes. Triple-negative breast cancers (TNBCs), a subgroup of breast cancer, are defined by the absence of estrogen and progesterone receptors and the lack of HER-2 expression; this subgroup accounts for ~15% of all breast cancers and exhibits the most aggressive metastatic behavior. Currently, very limited targeted therapies exist for the treatment of patients with TNBCs. On the other hand, it is important to highlight that knowledge of the molecular biology of breast cancer has recently changed the decision-making process regarding the course of cancer therapies. Thus, a number of new techniques, such as gene profiling and sequencing, proteomics, and microRNA analysis have been used to explore human breast carcinogenesis and metastasis including TNBC, which consequently could lead to new therapies. Nevertheless, based on evidence thus far, genomics profiles (gene and miRNA) can differ from one geographic location to another as well as in different ethnic groups. This review provides a comprehensive and updated information on the genomics profile alterations associated with TNBC pathogenesis associated with different ethnic backgrounds.
Collapse
Affiliation(s)
- Ishita Gupta
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | | | - Israa Al-Hashimi
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | | | - Halema Al Farsi
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | - Semir Vranic
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
| | - Ala-Eddin Al Moustafa
- College of Medicine, Qatar University, Doha P. O. Box:2713, Qatar.
- Biomedical Research Centre, Qatar University, Doha P.O Box: 2713, Qatar.
| |
Collapse
|
179
|
Jin X, Xu XE, Jiang YZ, Liu YR, Sun W, Guo YJ, Ren YX, Zuo WJ, Hu X, Huang SL, Shen HJ, Lan F, He YF, Hu GH, Di GH, He XH, Li DQ, Liu S, Yu KD, Shao ZM. The endogenous retrovirus-derived long noncoding RNA TROJAN promotes triple-negative breast cancer progression via ZMYND8 degradation. SCIENCE ADVANCES 2019; 5:eaat9820. [PMID: 30854423 PMCID: PMC6402854 DOI: 10.1126/sciadv.aat9820] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 01/23/2019] [Indexed: 05/03/2023]
Abstract
Human endogenous retroviruses (HERVs) play pivotal roles in the development of breast cancer. However, the detailed mechanisms of noncoding HERVs remain elusive. Here, our genome-wide transcriptome analysis of HERVs revealed that a primate long noncoding RNA, which we dubbed TROJAN, was highly expressed in human triple-negative breast cancer (TNBC). TROJAN promoted TNBC proliferation and invasion and indicated poor patient outcomes. We further confirmed that TROJAN could bind to ZMYND8, a metastasis-repressing factor, and increase its degradation through the ubiquitin-proteasome pathway by repelling ZNF592. TROJAN also epigenetically up-regulated metastasis-related genes in multiple cell lines. Correlations between TROJAN and ZMYND8 were subsequently confirmed in clinical samples. Furthermore, our study verified that antisense oligonucleotide therapy targeting TROJAN substantially suppressed TNBC progression in vivo. In conclusion, the long noncoding RNA TROJAN promotes TNBC progression and serves as a potential therapeutic target.
Collapse
Affiliation(s)
- Xi Jin
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Xiao-En Xu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Corresponding author. (Z.-M.S.); (X.-E.X.); (K.-D.Y.); (S.L.)
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Yi-Rong Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Wei Sun
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Ya-Jie Guo
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Yi-Xing Ren
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Wen-Jia Zuo
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Xin Hu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Sheng-Lin Huang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Hong-Jie Shen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Key Laboratory of Epigenetics, Department of Cellular and Genetic Medicine, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Fei Lan
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Key Laboratory of Epigenetics, Department of Cellular and Genetic Medicine, School of Basic Medical Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P. R. China
| | - Yun-Fei He
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Guo-Hong Hu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Gen-Hong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
| | - Xiang-Huo He
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Da-Qiang Li
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Suling Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
- Corresponding author. (Z.-M.S.); (X.-E.X.); (K.-D.Y.); (S.L.)
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Corresponding author. (Z.-M.S.); (X.-E.X.); (K.-D.Y.); (S.L.)
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong-An Road, Shanghai 200032, P. R. China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, P. R. China
- Corresponding author. (Z.-M.S.); (X.-E.X.); (K.-D.Y.); (S.L.)
| |
Collapse
|
180
|
Hwang SY, Park S, Kwon Y. Recent therapeutic trends and promising targets in triple negative breast cancer. Pharmacol Ther 2019; 199:30-57. [PMID: 30825473 DOI: 10.1016/j.pharmthera.2019.02.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Breast cancer accounts for 25% of all types of cancer in women, and triple negative breast cancer (TNBC) comprises around 15~20% of breast cancers. Conventional chemotherapy and radiation are the primary systemic therapeutic strategies; no other FDA-approved targeted therapies are yet available as for TNBC. TNBC is generally characterized by a poor prognosis and high rates of proliferation and metastases. Due to these aggressive features and lack of targeted therapies, numerous attempts have been made to discover viable molecular targets for TNBC. Massive cohort studies, clinical trials, and in-depth analyses have revealed diverse molecular alterations in TNBC; however, controversy exists as to whether many of these changes are beneficial or detrimental in caner progression. Here we review the complicated tumorigenic processes and discuss critical findings and therapeutic trends in TNBC with a focus on promising therapeutic approaches, the clinical trials currently underway, and potent experimental compounds under preclinical and evaluation.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
181
|
Moran T, Quiroga V, Cirauqui B, Vila L, Gil-Moreno M, Carcereny E, Margeli M, Muñoz-Marmol A, Mate JL, Velarde JM, Molina MA, Rosell R. A Single-Center Retrospective Study of Patients with Double Primary Cancers: Breast Cancer and EGFR-Mutant Non-Small Cell Lung Cancer. Oncol Res Treat 2019; 42:107-114. [PMID: 30799393 DOI: 10.1159/000495666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/19/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Second primary malignancies (SPM) in the lung are not common in breast cancer (BC) patients. EGFR-mutant lung cancer (LC) is a separate molecular subset, and the co-existence of EGFR-mutant LC and BC has not been explored. We hypothesized that EGFR-mutant LC patients could have higher rates of primary BC than those with EGFR-wild type (WT). METHODS We collected data on clinical and molecular characteristics and outcomes of female patients with LC and a previous or simultaneous history of primary BC treated in our hospital from 2008 to 2014. RESULTS Data on treatment, follow-up, and EGFR mutation status were available for 356 patients. 17.7% (11/62) of patients with EGFR mutations had BC, compared to 1.02% (3/294) of EGFR-WT patients (p < 0.001). Both tumors were metachronous in 81.8%, with LC diagnosed 9 years after the diagnosis of BC. 5 of the 6 (83.3%) BC patients treated with radiotherapy developed LC in an area within the radiation field. No EGFR mutations were detected in BC tissue and no HER2 expression was detected in LC samples. CONCLUSION SPM in the lung and breast occur more frequently among EGFR-mutant compared to EGFR-WT LC patients. Radiotherapy for BC may increase the risk of developing primary LC.
Collapse
|
182
|
Eigeliene N, Saarenheimo J, Jekunen A. Potential of Liquid Biopsies for Breast Cancer Screening, Diagnosis, and Response to Treatment. Oncology 2019; 96:115-124. [PMID: 30654364 DOI: 10.1159/000495615] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/19/2018] [Indexed: 11/19/2022]
Abstract
Cancer therapy decisions are often made according to the histopathological-molecular profile of tumor tissue obtained from surgery or biopsy. It has been shown that tumor profiles change with time and treatment, and that tumor tissue is heterogeneous. Thus, other approaches that are easily accessible and less invasive than surgery or biopsy to monitor responses to treatment and predict relapses are urgently needed. In the last few years, the term "liquid biopsies" has been introduced to represent multifunctional circulating biomarkers in the peripheral blood and other physiological fluids of patients with cancer. Liquid biopsies are a noninvasive alternative to tissue biopsies, but they have not been implemented in routine clinical practice for breast cancer. In addition, liquid biopsies seem to be a promising approach for personalized medicine, which enables the prediction, monitoring, and rational selection of appropriate therapy for individual patients. In this review, we outline recent progress and current challenges with liquid biopsies in clinical practice for breast cancer diagnosis, treatment choices, and responses to therapy from a clinician's perspective.
Collapse
Affiliation(s)
- Natalja Eigeliene
- Department of Oncology, Vaasa Central Hospital, Vaasa, Finland, .,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland,
| | - Jatta Saarenheimo
- Department of Pathology, Vaasa Central Hospital, Vaasa, Finland.,Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | - Antti Jekunen
- Department of Oncology, Vaasa Central Hospital, Vaasa, Finland.,Department of Oncology and Radiotherapy, University of Turku, Turku, Finland
| |
Collapse
|
183
|
Shen M, Jiang YZ, Wei Y, Ell B, Sheng X, Esposito M, Kang J, Hang X, Zheng H, Rowicki M, Zhang L, Shih WJ, Celià-Terrassa T, Liu Y, Cristea II, Shao ZM, Kang Y. Tinagl1 Suppresses Triple-Negative Breast Cancer Progression and Metastasis by Simultaneously Inhibiting Integrin/FAK and EGFR Signaling. Cancer Cell 2019; 35:64-80.e7. [PMID: 30612941 DOI: 10.1016/j.ccell.2018.11.016] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/13/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancer (TNBC) patients have the worst prognosis and distant metastasis-free survival among all major subtypes of breast cancer. The poor clinical outlook is further exacerbated by a lack of effective targeted therapies for TNBC. Here we show that ectopic expression and therapeutic delivery of the secreted protein Tubulointerstitial nephritis antigen-like 1 (Tinagl1) suppresses TNBC progression and metastasis through direct binding to integrin α5β1, αvβ1, and epidermal growth factor receptor (EGFR), and subsequent simultaneous inhibition of focal adhesion kinase (FAK) and EGFR signaling pathways. Moreover, Tinagl1 protein level is associated with good prognosis and reversely correlates with FAK and EGFR activation status in TNBC. Our results suggest Tinagl1 as a candidate therapeutic agent for TNBC by dual inhibition of integrin/FAK and EGFR signaling pathways.
Collapse
Affiliation(s)
- Minhong Shen
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Brian Ell
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Xinlei Sheng
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Mark Esposito
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Jooeun Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Xiang Hang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Hanqiu Zheng
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Michelle Rowicki
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Weichung J Shih
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, 683 Hoes Lane West, Piscataway, NJ 08854, USA; Division of Biometrics, Rutgers Cancer Institute of New Jersey Rutgers, New Brunswick, NJ 08901, USA
| | - Toni Celià-Terrassa
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Yirong Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - IIeana Cristea
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ 08544, USA; Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
184
|
Varghese S, Samuel SM, Varghese E, Kubatka P, Büsselberg D. High Glucose Represses the Anti-Proliferative and Pro-Apoptotic Effect of Metformin in Triple Negative Breast Cancer Cells. Biomolecules 2019; 9:E16. [PMID: 30626087 PMCID: PMC6359242 DOI: 10.3390/biom9010016] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 02/08/2023] Open
Abstract
Metformin, the most widely prescribed anti-diabetic drug, is shown to possess anti-cancer potential in treatment of cancers, including breast cancer; decreases breast cancer risk; and improves overall survival. However, reports suggest that higher glucose concentrations may negatively impact the anti-cancer efficacy of metformin. Therefore, we examined the anti-cancer potential of metformin in triple-negative breast cancer cells (TNBCs) exposed to different glucose (25 mM, 5.5 mM and zero glucose/glucose-starved) conditions. Our data indicates that a high glucose (25 mM) concentration (mimicking diabetes) significantly abrogated the effect of metformin on cell proliferation, cell death and cell cycle arrest in addition to loss of efficacy in inhibition of the mTOR pathway, a key metabolic pathway in TNBC cells. The mTOR pathway is activated in TNBCs compared to other subtypes of breast cancer, regulates the synthesis of proteins that are critical for the growth and survival of cancer cells and its activation is correlated to poor outcomes among TNBC patients, while also contributing to metastatic progression and development of resistance to chemotherapy/radiotherapy. Our studies were performed in two different types of TNBCs, MDA-MB-231 cells (mesenchymal stem cell-like (MSL)) and MDA-MB-468 (basal like-1 (BL-1)). Interestingly, lower concentrations of metformin (50, 100, 250, and 500 μM) significantly increased cell proliferation in 25 mM glucose exposed MDA-MB-231 cells, an effect which was not observed in MDA-MB-468 cells, indicating that the effective concentration of metformin when used as anti-cancer drug in TNBCs may have to be determined based on cell type and blood glucose concentration. Our data indicates that metformin treatment was most effective under zero glucose/glucose-starved conditions in MDA-MB-468 with a significant increase in the apoptotic population (62.3 ± 1.5%; p-value < 0.01). Under 5.5 mM glucose conditions in both MDA-MB-231 and MDA-MB-468 cells our data showed reduced viability of 73.56 ± 2.53%; p-value < 0.05 and 70.49 ± 1.68%; p-value < 0.001, respectively, along with a significant increase in apoptotic populations of both cell types. Furthermore, metformin (2 mM) inhibited the mTOR pathway and its downstream components under zero glucose/glucose-starved conditions indicating that using metformin in combination with agents that inhibit the glycolytic pathway should be more beneficial for the treatment of triple-negative breast cancers in diabetic individuals.
Collapse
Affiliation(s)
- Sharon Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Bratislava, Slovakia.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar.
| |
Collapse
|
185
|
Sridharan S, Howard CM, Tilley AMC, Subramaniyan B, Tiwari AK, Ruch RJ, Raman D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front Oncol 2019. [PMID: 31681564 DOI: 10.3389/fonc.2019.01003.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Breast cancer stem cells (BCSCs) play a vital role in tumor progression and metastasis. They are heterogeneous and inherently radio- and chemoresistant. They have the ability to self-renew and differentiate into non-BCSCs. These determinants of BCSCs including the plasticity between the mesenchymal and epithelial phenotypes often leads to minimal residual disease (MRD), tumor relapse, and therapy failure. By studying the resistance mechanisms in BCSCs, a combinatorial therapy can be formulated to co-target BCSCs and bulk tumor cells. This review addresses breast cancer stemness and molecular underpinnings of how the cancer stemness can lead to pharmacological resistance. This might occur through rewiring of signaling pathways and modulated expression of various targets that support survival and self-renewal, clonogenicity, and multi-lineage differentiation into heterogeneous bulk tumor cells following chemotherapy. We explore emerging novel and alternative molecular targets against BC stemness and chemoresistance involving survival, drug efflux, metabolism, proliferation, cell migration, invasion, and metastasis. Strategic targeting of such vulnerabilities in BCSCs may overcome the chemoresistance and increase the longevity of the metastatic breast cancer patients.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Cory M Howard
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Augustus M C Tilley
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | | | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
| | - Randall J Ruch
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
186
|
Bai M, Dong Y, Huang H, Fu H, Duan Y, Wang Q, Du L. Tumour targeted contrast enhanced ultrasound imaging dual-modal microbubbles for diagnosis and treatment of triple negative breast cancer. RSC Adv 2019; 9:5682-5691. [PMID: 35515929 PMCID: PMC9060891 DOI: 10.1039/c8ra09737b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/02/2019] [Indexed: 01/17/2023] Open
Abstract
At present, the treatment of triple negative breast cancer (TNBC) is a worldwide problem, urgently requiring early precise diagnosis and effective treatment methods. In our study, we designed a type of tumour targeted dual-modal microbubbles, paclitaxel (PTX)-loaded RGD-lipid microbubbles (PTX@RGD-MBs), combined with ultrasonic targeted microbubble destruction (UTMD) to precisely diagnose TNBC and to improve the curative effect. As the first-line drug, PTX, lacking specific tumour targeting and the ability to be effectively internalized by TNBC cells, is still not effective in killing TNBC cells. For this reason, we used the tumour active targeting peptide RGD to precisely guide MBs around TNBC cells through the high affinity to the integrin αvβ3 receptor, and use UTMD to generate shear stress on cells to open transient pores in the cellular membrane, so as to improve MB penetration into the cellular membrane. Sulfur hexafluoride (SF6) as the internal gas of MBs can greatly improve the resolution and sensitivity of conventional US images through nonlinear harmonics to enhance the comparison between lesions and surrounding normal tissues. The results in our study demonstrated that RGD-MBs with UTMD were internalized by TNBC cells more effectively, leading to significant increase in intercellular drug concentrations of TNBC cells, thus achieving the best inhibitory effect on TNBC cells in vitro. Ultrasonic experiment showed that PTX@RGD-MBs produced high quality contrast enhanced ultrasound (CEUS) images in vitro and in vivo, providing a better method for diagnosis and evaluation of the TNBC. Therefore, we conclude the advantages of excellent anti-tumour effect and CEUS imaging of PTX@RGD-MBs provided a better application for diagnosis and treatment of TNBC. Tumour targeted contrast-enhanced ultrasound imaging dual-modal microbubbles (PTX@RGD-MBs) combined with UTMD for diagnosing and treating triple negative breast cancer.![]()
Collapse
Affiliation(s)
- Min Bai
- Department of Ultrasound
- Shanghai General Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
- China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Qi Wang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals
- Shanghai Key Laboratory of Functional Materials Chemistry
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai 200237
| | - Lianfang Du
- Department of Ultrasound
- Shanghai General Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
- China
| |
Collapse
|
187
|
Varghese E, Samuel SM, Varghese S, Cheema S, Mamtani R, Büsselberg D. Triptolide Decreases Cell Proliferation and Induces Cell Death in Triple Negative MDA-MB-231 Breast Cancer Cells. Biomolecules 2018; 8:biom8040163. [PMID: 30563138 PMCID: PMC6315979 DOI: 10.3390/biom8040163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/25/2018] [Accepted: 11/28/2018] [Indexed: 01/06/2023] Open
Abstract
Triple negative breast cancers (TNBCs) do not respond to conventional estrogen receptor/progesterone receptor/human epidermal growth factor receptor-2 targeted interventions due to the absence of the respective receptor targets. They are aggressive, exhibit early recurrence, metastasize, are more invasive in nature, and develop drug resistance. Some plant-derived substances have been screened and have gained attention as efficient anticancer drugs for TNBCs with few adverse effects. Here, we evaluate triptolide (concentrations in the range of 100 pM to 10 µM), a di-terpene tri-epoxide isolated from thunder god vine for its efficacy as anticancer drug in MDA-MB-231 TNBC cells. Cell proliferation and viability were assessed using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay and trypan blue exclusion assay, respectively. A flow cytometry-based apoptosis assay was performed by using fluorescein isothiocyanate (FITC)-conjugated annexin V and propidium iodide (PI). Western blotting was performed to determine the levels of apoptotic and autophagy proteins such as caspase 3, LC3B and SQSTM1/p62. Results indicate that in 72 h of 1 nM triptolide treatment, the percentage of cell proliferation in MDA-MB-231 cells declined to 49 ± 18.9% (mean ± standard deviation (SD)), whereas the proliferation rate did not drop below 80% in MCF-7 cells (non-TNBC cells which express the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2) even at the highest concentration tested (10 µM). The MDA-MB-468 cells showed a similar trend to MDA-MB-231 cells. Furthermore, triptolide treatment for 72 h significantly decreased cell viability at concentrations above 10 nM. Apoptotic cell death assay in 72 h triptolide-treated MDA-MB-231 cells revealed 29.3 ± 10.57% of early apoptotic cells in comparison to the control group (4.61 ± 2.24%). Cell cycle analysis indicated accumulation of cells in sub G0/G1 phase, indicating apoptosis. Western blot analysis in the 25 nM triptolide treatment group revealed induction of autophagy as shown by a significant decrease in the levels of autophagy marker p62 (by 0.2-fold p < 0.0001) and with an increase in the levels of LC3B-II (by 8-fold p < 0.05). An increase in the levels of the apoptotic marker cleaved caspase 3 (by 4-fold p < 0.05) was also observed in triptolide treated MDA-MB-231 cells. Our data shows that triptolide could be an efficient anticancer agent in the treatment of TNBCs.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| | - Sharon Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| | - Sohaila Cheema
- Institute for Population Health, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| | - Ravinder Mamtani
- Institute for Population Health, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P. O. Box 24144, Doha, Qatar.
| |
Collapse
|
188
|
Walsh EM, Shalaby A, O’Loughlin M, Keane N, Webber MJ, Kerin MJ, Keane MM, Glynn SA, Callagy GM. Outcome for triple negative breast cancer in a retrospective cohort with an emphasis on response to platinum-based neoadjuvant therapy. Breast Cancer Res Treat 2018; 174:1-13. [PMID: 30488345 PMCID: PMC6418073 DOI: 10.1007/s10549-018-5066-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Purpose The rate of pathological complete response (pCR) for patients with triple negative breast cancer (TNBC) is increased when carboplatin is added to neo-adjuvant chemotherapy (NACT). However, while phase III trial data showing a survival benefit are awaited, carboplatin is not yet standard-of-care for TNBC. The aim of this study was to examine the rate of pCR and the outcome for those treated with carboplatin and to examine the predictors of response to therapy. Methods The retrospective series comprised 333 consecutive patients with TNBC (median follow-up time, 43 months). Adjuvant chemotherapy was given to 51% (n = 168) of patients and 29% (n = 97) received anthracycline–taxane NACT with carboplatin given to 9% (n = 31) of patients. Results Overall, 25% (n = 78) of patients experienced a breast cancer recurrence and 22% (n = 68) died from disease. A pCR breast and pCR breast/axilla was more common in those who received carboplatin (n = 18, 58% and n = 17, 55%, respectively) compared those who did not (n = 23, 36% and n = 18, 28%, respectively) (p = 0.041 and p = 0.011, respectively). By multivariable analysis, carboplatin and high tumor grade were independent predictors of pCR breast/axilla (ORnon-pCR = 0.17; 95% CI 0.06–0.54; p = 0.002; and ORnon-pCR = 0.05, 95% CI 0.01–0.27; p < 0.001, respectively). pCR breast/axilla was an independent predictor of DFS (HRnon-pCR=6.23; 95% CI 1.36–28.50; p = 0.018), metastasis-free survival (HRnon-pCR = 5.08; 95% CI 1.09–23.65; p = 0.038) and BCSS (HRnon-pCR = 8.52; 95% CI 1.09–66.64; p = 0.041). Conclusion Carboplatin therapy and high tumor grade are associated with a significant increase in the rate of pCR, which is an independent predictor of outcome. These data support the use of carboplatin in NACT for TNBC, while results from phase III studies are awaited. Electronic supplementary material The online version of this article (10.1007/s10549-018-5066-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elaine M. Walsh
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - Aliaa Shalaby
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Mark O’Loughlin
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Nessa Keane
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Mark J Webber
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Michael J. Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Maccon M. Keane
- Department of Medical Oncology, University Hospital Galway, Galway, Ireland
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| | - Grace M. Callagy
- Discipline of Pathology, Lambe Institute for Translational Research, NUI Galway, Costello Road, Galway, Ireland
| |
Collapse
|
189
|
Pegram MD, Zong Y, Yam C, Goetz MP, Moulder SL. Innovative Strategies: Targeting Subtypes in Metastatic Breast Cancer. Am Soc Clin Oncol Educ Book 2018; 38:65-77. [PMID: 30231328 DOI: 10.1200/edbk_200715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metastatic breast cancer continues to be a life-threatening diagnosis that impacts hundreds of thousands of patients around the world. Targeted therapies are usually associated with less toxicity compared with cytotoxic chemotherapies and often induce response or durable disease control in estrogen receptor (ER) and/or HER2+ breast cancers. Drugs that target CDK 4/6 either alone or in combination with endocrine therapy have demonstrated substantial improvements in progression-free survival (PFS) compared with endocrine monotherapy. Most recently, PARP inhibitors have shown longer PFS compared with physician's choice of chemotherapy in BRCA-associated cancers, leading to the first U.S. Food and Drug Administration (FDA) approval of a targeted therapy with the potential to benefit a subgroup of patients with triple-negative breast cancer (TNBC). Finally, newer drug delivery strategies using antibody drug conjugates have also allowed a "targeted approach" to deliver moderate to extremely potent cytotoxins directly to sites of metastatic disease, with less toxicity.
Collapse
Affiliation(s)
- Mark D Pegram
- From the Stanford Comprehensive Cancer, Stanford, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Mayo Clinic Cancer Center, Rochester, MN
| | - Yu Zong
- From the Stanford Comprehensive Cancer, Stanford, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Mayo Clinic Cancer Center, Rochester, MN
| | - Clinton Yam
- From the Stanford Comprehensive Cancer, Stanford, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Mayo Clinic Cancer Center, Rochester, MN
| | - Matthew P Goetz
- From the Stanford Comprehensive Cancer, Stanford, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Mayo Clinic Cancer Center, Rochester, MN
| | - Stacy L Moulder
- From the Stanford Comprehensive Cancer, Stanford, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Mayo Clinic Cancer Center, Rochester, MN
| |
Collapse
|
190
|
Fan Y, Li M, Ma K, Hu Y, Jing J, Shi Y, Li E, Dong D. Dual-target MDM2/MDMX inhibitor increases the sensitization of doxorubicin and inhibits migration and invasion abilities of triple-negative breast cancer cells through activation of TAB1/TAK1/p38 MAPK pathway. Cancer Biol Ther 2018; 20:617-632. [PMID: 30462562 DOI: 10.1080/15384047.2018.1539290] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a poor prognosis mainly due to insensitivity or resistance to standard anthracycline- and taxane-based chemotherapy, urgently calling for new adjuvants to reverse drug resistance. Dual-target murine double minute 2 (MDM2) and murine double minute X (MDMX) inhibitor has been proved to play a critical part against cancer, particularly focusing on the tremendous potential to enhance the efficacy of doxorubicin (DOX), however little was reported in TNBC. In the present study, we investigated the synergistic antitumor effect of the MDM2/MDMX inhibitor with DOX using three TNBC cell lines, two in situ transplantation tumor models and 214 clinical samples. We observed that the MDM2/MDMX inhibitor combined with DOX could not only inhibit cell vitality and migration and invasion abilities, but also highly inhibit tumor growth in TNBC nude mice. Besides, co-treatment of MDM2/MDMX inhibitor and DOX suppressed epithelial to mesenchymal transition (EMT) through increasing the TAK1-binding protein 1 (TAB1), transforming growth factor β-activated kinase 1 (TAK1) and p38 mitogen-activated protein kinase (MAPK) expression. Small interfering RNA-mediated TAB1 knockdown induced the EMT, desensitized cells to DOX and enhanced the migration and invasion abilities. High MDM2/MDMX expression was positively associated with weak TAB1 expression in 214 TNBC tumor tissues confirmed by immumohistochemical staining and MDM2/MDMX/TAB1 expression was significantly related to TNBC patient survival. These findings indicate that dual-target MDM2/MDMX inhibitor could increase the sensitization of doxorubicin and inhibit migration and invasion abilities in TNBC cells through p38 MAPK pathway activation caused EMT suppression and hence could be useful in TNBC treatments in future.
Collapse
Affiliation(s)
- Yangwei Fan
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| | - Mengya Li
- b Department of Medical Oncology , the First Affiliated Hospital of Henan University , Kaifeng , China
| | - Ke Ma
- c Department of Medical Oncology , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Yuan Hu
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| | - Jiayu Jing
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| | - Yu Shi
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| | - Enxiao Li
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| | - Danfeng Dong
- a Department of Medical Oncology , the First Affiliated Hospital of medical school of Xi'an Jiaotong University , Xi'an , China
| |
Collapse
|
191
|
Guestini F, Ono K, Miyashita M, Ishida T, Ohuchi N, Nakagawa S, Hirakawa H, Tamaki K, Ohi Y, Rai Y, Sagara Y, Sasano H, McNamara KM. Impact of Topoisomerase IIα, PTEN, ABCC1/MRP1, and KI67 on triple-negative breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat 2018; 173:275-288. [PMID: 30306430 DOI: 10.1007/s10549-018-4985-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) patients with residual disease following neoadjuvant chemotherapy (NAC) harbor higher risk of relapse, and eventual demise compared to those who achieve pathologic complete response. Therefore, in this study, we assessed a panel of molecules involved in key pathways of drug resistance and tumor progression before and after NAC in TNBC patients, in order to clarify the underlying mechanisms. METHODS We studied 148 TNBC Japanese patients treated with anthracycline/taxane-based NAC. KI67, Topoisomerase IIα (TopoIIα), PTEN, p53, Bcl2, vimentin, ABCG2/BCRP1, ABCB1/MDR1, and ABCC1/MRP1 were immunolocalized in surgical pathology materials before and after NAC. RESULTS The status of vimentin and increasing labeling index (LI) of TopoIIα and KI67 in biopsy specimens were significantly associated with those who responded to NAC treatment. The abundance of p53 (p = 0.003), ABCC1/MRP1 (p = 0.033), ABCB1/MDR1 (p = 0.022), and a loss of PTEN (p < 0.0001) in surgery specimens following treatment were associated with pathologic parameters. TopoIIα, PTEN, and ABCC1/MRP1 status predicted pathologic response. In addition, the status of PTEN, ABCC1/MRP1, ABCB1/MDR1, Bcl2, and vimentin in surgical specimens was also significantly associated with adverse clinicopathological factors in surgery specimens, suggesting that these alterations could be responsible for tumor relapse in TNBC patients. CONCLUSION KI67, TopoIIα, PTEN, and ABCC1/MRP1 status could predict treatment response and/or eventual clinical outcomes. These results could also provide an insight into the mechanisms of drug resistance and relapse of TNBC patients receiving NAC.
Collapse
|
192
|
Feasibility of Classification of Triple Negative Breast Cancer by Immunohistochemical Surrogate Markers. Clin Breast Cancer 2018; 18:e1123-e1132. [DOI: 10.1016/j.clbc.2018.03.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/03/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023]
|
193
|
Ezenwajiaku N, Ma CX, Ademuyiwa FO. Updates on Molecular Classification of Triple Negative Breast Cancer. CURRENT BREAST CANCER REPORTS 2018. [DOI: 10.1007/s12609-018-0292-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
194
|
Temian DC, Pop LA, Irimie AI, Berindan-Neagoe I. The Epigenetics of Triple-Negative and Basal-Like Breast Cancer: Current Knowledge. J Breast Cancer 2018; 21:233-243. [PMID: 30275851 PMCID: PMC6158152 DOI: 10.4048/jbc.2018.21.e41] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022] Open
Abstract
Breast cancer has the highest incidence among all malignancies diagnosed in women. Therapies have significantly improved over the years due to extensive molecular and clinical research; in a large number of cases, targeted therapies have provided better prognosis. However, one specific subtype remains elusive to targeted therapies–the triple-negative breast cancer. This immunohistochemically defined subtype is resistant to both endocrine and targeted therapies, leading to its poor prognosis. A field that is of great promise in current cancer research is epigenetics. By studying the epigenetic mechanisms underlying tumorigenesis–DNA methylation, histone modifications, and noncoding RNAs–advances in cancer treatment, diagnosis, and prevention are possible. This review aims to synthesize the epigenetic discoveries that have been made related to the triple-negative breast cancer.
Collapse
Affiliation(s)
- Daiana Cosmina Temian
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Laura Ancuta Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Iulia Irimie
- Division of Dental Propaedeutics, Aesthetic, Department of Prosthetic Dentistry and Dental Materials, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,MedFUTURE Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. I Chiricuta", Cluj-Napoca, Romania
| |
Collapse
|
195
|
Lin CC, Lo MC, Moody R, Jiang H, Harouaka R, Stevers N, Tinsley S, Gasparyan M, Wicha M, Sun D. Targeting LRP8 inhibits breast cancer stem cells in triple-negative breast cancer. Cancer Lett 2018; 438:165-173. [PMID: 30227220 DOI: 10.1016/j.canlet.2018.09.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/23/2018] [Accepted: 09/11/2018] [Indexed: 12/30/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most difficult subtype of breast cancer to treat due to a paucity of effective targeted therapies. Many studies have reported that breast cancer stem cells (BCSCs) are enriched in TNBC and are responsible for chemoresistance and metastasis. In this study, we identify LRP8 as a novel positive regulator of BCSCs in TNBC. LRP8 is highly expressed in TNBC compared to other breast cancer subtypes and its genomic locus is amplified in 24% of TNBC tumors. Knockdown of LRP8 in TNBC cell lines inhibits Wnt/β-catenin signaling, decreases BCSCs, and suppresses tumorigenic potential in xenograft models. LRP8 knockdown also induces a more differentiated, luminal-epithelial phenotype and thus sensitizes the TNBC cells to chemotherapy. Together, our study highlights LRP8 as a novel therapeutic target for TNBC as inhibition of LRP8 can attenuate Wnt/β-catenin signaling to suppress BCSCs.
Collapse
Affiliation(s)
- Chang-Ching Lin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Miao-Chia Lo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Rebecca Moody
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Chemical Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ramdane Harouaka
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, 48109, USA
| | - Nicholas Stevers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Samantha Tinsley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mari Gasparyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, 48109, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA; Chemical Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
196
|
Danzinger S, Tan YY, Rudas M, Kastner MT, Weingartshofer S, Muhr D, Singer CF. Differential Claudin 3 and EGFR Expression Predicts BRCA1 Mutation in Triple-Negative Breast Cancer. Cancer Invest 2018; 36:378-388. [DOI: 10.1080/07357907.2018.1499934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Sabine Danzinger
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Yen Yen Tan
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Margaretha Rudas
- Department of Pathology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Marie-Theres Kastner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sigrid Weingartshofer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daniela Muhr
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian F. Singer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
197
|
Triple-Negative Breast Cancers: Systematic Review of the Literature on Molecular and Clinical Features with a Focus on Treatment with Innovative Drugs. Curr Oncol Rep 2018; 20:76. [PMID: 30128845 DOI: 10.1007/s11912-018-0726-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Triple-negative breast cancer (TNBC) accounts for 15-20% of diagnosed breast tumours, with higher incidence in young and African-American women, and it is frequently associated with BRCA germline mutations. Chemotherapy is the only well-established therapeutic option in both early- and advanced-stages of the disease. TNBC tumours relapse earlier after standard anthracycline- and/or taxane-based chemotherapy treatments, generally within 1-3 years after the diagnosis, and often develop visceral metastases, representing the subtype with a worse prognosis among all breast cancers. In the present review, we will provide an updated overview of the available results of recent clinical trials for this disease and we will describe the implications of the known molecular pathways representing novel targets for development of future therapies for TNBC patients. RECENT FINDINGS Over the past decade, the advent of gene expression micro-array technology has led to the identification of different actionable targets including various genomic alterations, androgen receptor, PARP, PI3K, VEGF and other proteins of the angiogenic pathway. Thus, novel targeted drugs have been tested in clinical trials reporting promising results in specific TNBC molecular subgroups. Although cytotoxic chemotherapy remains the mainstay of treatment for TNBC patients, the identification of novel 'drugable' targets and pathways for developing personalized treatments represents a promising investigational approach in the management of the TNBC subtype.
Collapse
|
198
|
Sporikova Z, Koudelakova V, Trojanec R, Hajduch M. Genetic Markers in Triple-Negative Breast Cancer. Clin Breast Cancer 2018; 18:e841-e850. [PMID: 30146351 DOI: 10.1016/j.clbc.2018.07.023] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/22/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for 15% to 20% of breast cancer cases and is characterized by the absence of estrogen, progesterone, and human epidermal growth factor 2 receptors. Though TNBC is a highly heterogenic and aggressive disease, TNBC patients have better response to neoadjuvant therapy compared to other breast cancer subtypes. Nevertheless, patients with residual disease have a very poor prognosis, with higher probability of relapse and lower overall survival in the first years after diagnosis. TNBC has 6 subtypes with distinct molecular signatures with different prognoses and probably different responses to therapy. The precise stratification of TNBC is therefore crucial for the development of potent standardized and targeted therapies. In spite of intensive research into finding new molecular biomarkers and designing personalized therapeutic approaches, BRCA mutational status is the only clinically validated biomarker for personalized therapy in TNBC. Recent studies have reported several promising biomarkers that are currently being validated through clinical trials. The objective of this review was to summarize the clinically relevant genetic markers for TNBC that could serve as diagnostic, prognostic, or predictive or could improve personalized therapeutic strategies.
Collapse
Affiliation(s)
- Zuzana Sporikova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Vladimira Koudelakova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic.
| | - Radek Trojanec
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital in Olomouc, Olomouc, Czech Republic
| |
Collapse
|
199
|
Li J, Gong X, Jiang R, Lin D, Zhou T, Zhang A, Li H, Zhang X, Wan J, Kuang G, Li H. Fisetin Inhibited Growth and Metastasis of Triple-Negative Breast Cancer by Reversing Epithelial-to-Mesenchymal Transition via PTEN/Akt/GSK3β Signal Pathway. Front Pharmacol 2018; 9:772. [PMID: 30108501 PMCID: PMC6080104 DOI: 10.3389/fphar.2018.00772] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Triple negative breast cancer (TNBC), characterized by its highly aggressive and metastatic features, is associated with poor prognosis and high mortality partly due to lack of effective treatment. Fisetin, a natural flavonoid compound, has been demonstrated to possess anti-cancer effects in various cancers. However, the effects and mechanisms of fisetin on metastasis of TNBC remain uncovered. In this study, we found that fisetin dose-dependently inhibited cell proliferation, migration and invasion in TNBC cell lines MDA-MB-231 and BT549 cells. In addition, fisetin reversed epithelial to mesenchymal transition (EMT) as evaluated by cell morphology and EMT markers in MDA-MB-231 and BT549 cells. Furthermore, fisetin suppressed phosphoinositol 3-kinase (PI3K)-Akt-GSK-3β signaling pathway but upregulated the expression of PTEN mRNA and protein in a concentration-dependent manner. Further, silence of PTEN by siRNA abolished these benefits of fisetin on proliferation and metastasis of TNBCs. In vivo, using the metastatic breast cancer xenograft model bearing MDA-MB-231 cells, we found that fisetin dramatically inhibited growth of primary breast tumor and reduced lung metastasis, meanwhile, the expression of EMT molecules and PTEN/Akt/GSK-3β in primary and metastatic tissues changed in the same way as those in vitro experiments. In conclusion, all these results indicated that fisetin could effectively suppress proliferation and metastasis of TNBC and reverse EMT process, which might be mediated by PTEN/Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Dan Lin
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Tao Zhou
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Aijie Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhong Li
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Hongyuan Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
200
|
Moore-Smith L, Forero-Torres A, Stringer-Reasor E. Future Developments in Neoadjuvant Therapy for Triple-Negative Breast Cancer. Surg Clin North Am 2018; 98:773-785. [PMID: 30005773 DOI: 10.1016/j.suc.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Breast cancer is the 2nd leading cause of cancer-related death in women in the United States. In general, advances in targeted treatment for breast cancer have improved over the last twenty years, except in the triple-negative breast cancer (TNBC) subtype. TNBC is an aggressive breast cancer subtype with limited treatment options as compared to hormone positive breast cancers. Recently, genomic profiling of TNBC shows promise in aiding clinicians to develop personalized targeted agents. Prioritizing novel molecular-based therapies in the neoadjuvant setting may help investigators understand mechanisms of resistance and ultimately improve patient outcomes in TNBC.
Collapse
Affiliation(s)
- Lakisha Moore-Smith
- Department of Medicine, Brookwood Baptist Health - Princeton, 833 Princeton Avenue, POB III Suite 200, Birmingham, AL 35211-1311, USA
| | - Andres Forero-Torres
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2517, Birmingham, AL 35294-3300, USA
| | - Erica Stringer-Reasor
- Department of Medicine, Division of Hematology Oncology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL 35294-3300, USA.
| |
Collapse
|