1
|
Zhang R, Jiang Q, Zhuang Z, Zeng H, Li Y. A bibliometric analysis of drug resistance in immunotherapy for breast cancer: trends, themes, and research focus. Front Immunol 2024; 15:1452303. [PMID: 39188717 PMCID: PMC11345160 DOI: 10.3389/fimmu.2024.1452303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
While breast cancer treatments have advanced significantly nowadays, yet metastatic, especially triple-negative breast cancer (TNBC), remains challenging with low survival. Cancer immunotherapy, a promising approach for HER2-positive and TNBC, still faces resistance hurdles. Recently, numerous studies have set their sights on the resistance of immunotherapy for breast cancer. Our study provides a thorough comprehension of the current research landscape, hotspots, and emerging breakthroughs in this critical area through a meticulous bibliometric analysis. As of March 26, 2024, a total of 1341 articles on immunology resistance in breast cancer have been gathered from Web of Science Core Collection, including 765 articles and 576 reviews. Bibliometrix, CiteSpace and VOSviewer software were utilized to examine publications and citations per year, prolific countries, contributive institutions, high-level journals and scholars, as well as highly cited articles, references and keywords. The research of immunotherapy resistance in breast cancer has witnessed a remarkable surge over the past seven years. The United States and China have made significant contributions, with Harvard Medical School being the most prolific institution and actively engaging in collaborations. The most contributive author is Curigliano, G from the European Institute of Oncology in Italy, while Wucherpfennig, K. W. from the Dana-Farber Cancer Institute in the USA, had the highest citations. Journals highly productive primarily focus on clinical, immunology and oncology research. Common keywords include "resistance", "expression", "tumor microenvironment", "cancer", "T cell", "therapy", "chemotherapy" and "cell". Current research endeavors to unravel the mechanisms of immune resistance in breast cancer through the integration of bioinformatics, basic experiments, and clinical trials. Efforts are underway to develop strategies that improve the effectiveness of immunotherapy, including the exploration of combination therapies and advancements in drug delivery systems. Additionally, there is a strong focus on identifying novel biomarkers that can predict patient response to immunology. This study will provide researchers with an up-to-date overview of the present knowledge in drug resistance of immunology for breast cancer, serving as a valuable resource for informed decision-making and further research on innovative approaches to address immunotherapy resistance.
Collapse
Affiliation(s)
- Rendong Zhang
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Qiongzhi Jiang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zhemin Zhuang
- Engineering College, Shantou University, Shantou, Guangdong, China
| | - Huancheng Zeng
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yaochen Li
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
2
|
Akça KT, Demirel MA, Süntar I. The Role of Aromatase Enzyme in Hormone Related Diseases and Plant-Based Aromatase Inhibitors as Therapeutic Regimens. Curr Top Med Chem 2021; 22:229-246. [PMID: 34844542 DOI: 10.2174/1568026621666211129141631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/17/2021] [Accepted: 10/31/2021] [Indexed: 11/22/2022]
Abstract
Medicinal plants have a long history of use as food and remedy in traditional and modern societies, as well as have been used as herbal drugs and sources of novel bioactive compounds. They provide a wide array of chemical compounds, many of which can not be synthesized via current synthesis methods. Natural products may provide aromatase inhibitory activity through various pathways and may act clinically effective for treating pathologies associated with excessive aromatase secretion including breast, ovarian and endometrial cancers, endometriosis, uterine fibroid, benign prostatic hyperplasia (BPH), prostate cancer, infertility, and gynecomastia. Recent studies have shown that natural products with aromatase inhibitory activity, could also be good options against secondary recurrence of breast cancer by exhibiting chemopreventive effects. Therefore, screening for new plant-based aromatase inhibitors may provide novel leads for drug discovery and development, particularly with increased clinical efficacy and decreased side effects.
Collapse
Affiliation(s)
- Kevser Taban Akça
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Etiler, Ankara. Turkey
| | - Murside Ayşe Demirel
- Department of Basic Pharmaceutical Sciences, Laboratory Animals Breeding and Experimental Research Center, Gazi University, Faculty of Pharmacy, 06330, Etiler, Ankara. Turkey
| | - Ipek Süntar
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Etiler, Ankara. Turkey
| |
Collapse
|
3
|
Bondhopadhyay B, Sisodiya S, Kasherwal V, Nazir SU, Khan A, Tanwar P, Dil-Afroze, Singh N, Rasool I, Agrawal U, Rath G, Mehrotra R, Hussain S. The differential expression of Promyelocytic Leukemia (PML) and retinoblastoma (RB1) genes in breast cancer. Meta Gene 2021; 28:100852. [DOI: 10.1016/j.mgene.2021.100852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
4
|
Vazquez N, Lopez A, Cuello V, Persans M, Schuenzel E, Innis-Whitehouse W, Keniry M. NVP-BEZ235 or JAKi Treatment leads to decreased survival of examined GBM and BBC cells. Cancer Treat Res Commun 2021; 27:100340. [PMID: 33636591 DOI: 10.1016/j.ctarc.2021.100340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/04/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Cancer cells almost universally harbor constitutively active Phosphatidylinositol-3 Kinase (PI3K) Pathway activity via mutation of key signaling components and/or epigenetic mechanisms. Scores of PI3K Pathway inhibitors are currently under investigation as putative chemotherapeutics. However, feedback and stem cell mechanisms induced by PI3K Pathway inhibition can lead to reduced treatment efficacy. To address therapeutic barriers, we examined whether JAKi would reduce stem gene expression in a setting of PI3K Pathway inhibition in order to improve treatment efficacy. We targeted the PI3K Pathway with NVP-BEZ235 (dual PI3K and mTOR inhibitor) in combination with the Janus Kinase inhibitor JAKi in glioblastoma (GBM) and basal-like breast cancer (BBC) cell lines. We examined growth, gene expression, and apoptosis in cells treated with NVP-BEZ235 and/or JAKi. Growth and recovery assays showed no significant impact of dual treatment with NVP-BEZ235/JAKi compared to NVP-BEZ235 treatment alone. Gene expression and flow cytometry revealed that single and dual treatments induced apoptosis. Stem gene expression was retained in dual NVP-BEZ235/JAKi treatment samples. Future in vivo studies may give further insight into the impact of combined NVP-BEZ235/JAKi treatment in GBM and BBC.
Collapse
Affiliation(s)
- Neftali Vazquez
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Alma Lopez
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Victoria Cuello
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Michael Persans
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Erin Schuenzel
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Wendy Innis-Whitehouse
- School of Medicine, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States
| | - Megan Keniry
- Department of Biology, University of Texas- Rio Grande Valley, 1201 W. University Dr., Edinburg, TX 78539, United States.
| |
Collapse
|
5
|
Patel S, Sangeeta S. Pesticides as the drivers of neuropsychotic diseases, cancers, and teratogenicity among agro-workers as well as general public. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:91-100. [PMID: 30411285 DOI: 10.1007/s11356-018-3642-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/30/2018] [Indexed: 06/08/2023]
Abstract
The need to maximize agricultural productivity has made pesticides an indispensable part of current times. Farmers are unaware of the lurking consequences of the pesticide exposure, which endanger their health. It also puts the unsuspecting consumers in peril. The pesticides (from organophosphates, organochlorine, and carbamate class) disrupt the immune and hormonal signaling, causing recurrent inflammation, which leads to a wide array pathologies, including teratogenicity. Numerous farmers have fallen victim to neural disorders-driven suicides and lungs, prostate/breast cancer-caused untimely deaths. Green revolution which significantly escalated agricultural productivity is backfiring now. It is high time that environmental and agricultural authorities act to restrain the excessive usage of the detrimental chemicals and educate farmers regarding the crisis. This review discusses the biological mechanisms of pesticide-driven pathogenesis (such as the activation or inhibition of caspase, serine protease, acetylcholinesterase) and presents the pesticide-exposure-caused health deterioration in USA, India, and Africa. This holistic and critical review should be an eye-opener for general public, and a guide for researchers.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, 5500 Campanile Dr, San Diego, CA, 92182, USA.
| | - Sushree Sangeeta
- Department of Ecology and Environmental Sciences, Pondicherry University, Puducherry, 605014, India.
| |
Collapse
|
6
|
Patel S, Homaei A, Raju AB, Meher BR. Estrogen: The necessary evil for human health, and ways to tame it. Biomed Pharmacother 2018; 102:403-411. [PMID: 29573619 DOI: 10.1016/j.biopha.2018.03.078] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Estrogen is a pivotal enzyme for survival and health in both genders, though their quantum, tropism, tissue-specific distribution, and receptor affinity varies with different phases of life. Converted from androgen via aromatase enzyme, this hormone is indispensable to glucose homeostasis, immune robustness, bone health, cardiovascular health, fertility, and neural functions. However, estrogen is at the center of almost all human pathologies as well-infectious, autoimmune, metabolic to degenerative. Both hypo and hyper level of estrogen has been linked to chronic and acute diseases. While normal aging is supposed to lower its level, leading to tissue degeneration (bone, muscle, neural etc.), and metabolite imbalance (glucose, lipid etc.), the increment in inflammatory agents in day-to-day life are enhancing the estrogen (or estrogen mimic) level, fueling 'estrogen dominance'. The resultant excess estrogen is inducing an overexpression of estrogen receptors (ERα and ERβ), harming tissues, leading to autoimmune diseases, and neoplasms. The unprecedented escalation in the polycystic ovary syndrome, infertility, breast cancer, ovary cancer, and gynecomastia cases are indicating that this sensitive hormone is getting exacerbated. This critical review is an effort to analyze the dual, and opposing facets of estrogen, via understanding its crosstalk with other hormones, enzymes, metabolites, and drugs. Why estrogen level correction is no trivial task, and how it can be restored to normalcy by a disciplined lifestyle with wise dietary and selective chemical usage choices has been discussed. Overall, our current state of knowledge does not disclose the full picture of estrogen's pleiotropic importance. Hence, this review should be a resource for general public as well as researchers to work in that direction.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, 92182, San Diego, CA, USA.
| | - Ahmad Homaei
- Department of Marine Biology, Faculty of Marine Science and Technology, University of Hormozgan, Bandar Abbas, Iran; Department of Biology, Faculty of Sciences, University of Hormozgan, Bandar Abbas, Iran
| | - Akondi Butchi Raju
- Department of Pharmacology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Biswa Ranjan Meher
- Department of Botany, Berhampur University, Berhampur, Odisha, 760007, India
| |
Collapse
|
7
|
Patel S. Breast cancer: Lesser-known facets and hypotheses. Biomed Pharmacother 2017; 98:499-506. [PMID: 29287197 DOI: 10.1016/j.biopha.2017.12.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most common cancer in females. The deteriorating environment, and lifestyle flaws are raising the frequency of this cancer. Existing therapies are not universally-effective, and they cause side effects, relapses, and high mortality rate. Alternative medications may be milder, but are less effective or are inadequate for a complex disease like the breast cancer. So, it requires the understanding that drugs are not the solution of this cancer, but prevention is the sustainable solution. In the past decades, an enormous quantum of insights on this disease has been obtained. A lifestyle based on the template of estrogenic compounds and, the resultant endocrine disruption, and acidosis, is elevating aromatase level, promoting the deleterious forms of estrogen, and inducing epithelial proliferation. This review provids a holistic account of breast cancer as a inflammatory endocrinopathy, and how it can be curbed by discipline, and awareness.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182, USA.
| |
Collapse
|
8
|
John Mary DJS, Manjegowda MC, Kumar A, Dutta S, Limaye AM. The role of cystatin A in breast cancer and its functional link with ERα. J Genet Genomics 2017; 44:593-597. [DOI: 10.1016/j.jgg.2017.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/05/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
|
9
|
Fragrance compounds: The wolves in sheep’s clothings. Med Hypotheses 2017; 102:106-111. [DOI: 10.1016/j.mehy.2017.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/21/2017] [Indexed: 12/31/2022]
|
10
|
Patel S. Disruption of aromatase homeostasis as the cause of a multiplicity of ailments: A comprehensive review. J Steroid Biochem Mol Biol 2017; 168:19-25. [PMID: 28109841 DOI: 10.1016/j.jsbmb.2017.01.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/20/2016] [Accepted: 01/15/2017] [Indexed: 01/15/2023]
Abstract
Human health is beset with a legion of ailments, which is exacerbated by lifestyle errors. Out of the numerous enzymes in human body, aromatase, a cytochrome P450 enzyme is particularly very critical. Occurring at the crossroads of multiple signalling pathways, its homeostasis is vital for optimal health. Unfortunately, medications, hormone therapy, chemical additives in food, and endocrine-disrupting personal care products are oscillating the aromatase concentration beyond the permissible level. As this enzyme converts androgens (C19) into estrogens (C18), its agitation has different outcomes in different genders and age groups. Some common pathologies associated with aromatase disruption include breast cancer, prostate cancer, polycystic ovary syndrome (PCOS), endometriosis, osteoporosis, ovarian cancer, gastric cancer, pituitary cancer, Alzheimer's disease, schizophrenia, male hypogonadism, and transgender issues. Several drugs, cosmetics and pesticides act as the activators and suppressors of this enzyme. This carefully-compiled critical review is expected to increase public awareness regarding the threats resultant of the perturbations of this enzyme and to motivate researchers for further investigation of this field.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, CA 92182, USA.
| |
Collapse
|
11
|
Movassaghian S, Xie Y, Hildebrandt C, Rosati R, Li Y, Kim NH, Conti DS, da Rocha SRP, Yang ZQ, Merkel OM. Post-Transcriptional Regulation of the GASC1 Oncogene with Active Tumor-Targeted siRNA-Nanoparticles. Mol Pharm 2016; 13:2605-21. [PMID: 27223606 DOI: 10.1021/acs.molpharmaceut.5b00948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Basal-like breast cancer (BLBC) accounts for the most aggressive types of breast cancer, marked by high rates of relapse and poor prognoses and with no effective clinical therapy yet. Therefore, investigation of new targets and treatment strategies is more than necessary. Here, we identified a receptor that can be targeted in BLBC for efficient and specific siRNA mediated gene knockdown of therapeutically relevant genes such as the histone demethylase GASC1, which is involved in multiple signaling pathways leading to tumorigenesis. Breast cancer and healthy breast cell lines were compared regarding transferrin receptor (TfR) expression via flow cytometry and transferrin binding assays. Nanobioconjugates made of low molecular weight polyethylenimine (LMW-PEI) and transferrin (Tf) were synthesized to contain a bioreducible disulfide bond. siRNA complexation was characterized by condensation assays and dynamic light scattering. Cytotoxicity, transfection efficiency, and the targeting specificity of the conjugates were investigated in TfR positive and negative healthy breast and breast cancer cell lines by flow cytometry, confocal microscopy, RT-PCR, and Western blot. Breast cancer cell lines revealed a significantly higher TfR expression than healthy breast cells. The conjugates efficiently condensed siRNA into particles with 45 nm size at low polymer concentrations, showed no apparent toxicity on different breast cancer cell lines, and had significantly greater transfection and gene knockdown activity on mRNA and protein levels than PEI/siRNA leading to targeted and therapeutic growth inhibition post GASC1 knockdown. The synthesized nanobioconjugates improved the efficiency of gene transfer and targeting specificity in transferrin receptor positive cells but not in cells with basal receptor expression. Therefore, these materials in combination with our newly identified siRNA sequences are promising candidates for therapeutic targeting of hard-to-treat BLBC and are currently further investigated regarding in vivo targeting efficacy and biocompatibility.
Collapse
Affiliation(s)
- Sara Movassaghian
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Yuran Xie
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Claudia Hildebrandt
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmaceutics and Biopharmaceutics, Kiel University , 24118 Kiel, Germany
| | - Rayna Rosati
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Ying Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Na Hyung Kim
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States
| | - Denise S Conti
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , Detroit, Michigan 48202, United States
| | - Sandro R P da Rocha
- Department of Pharmaceutics, College of Pharmacy, Virginia Commonwealth University , Richmond, Virginia 23298, United States.,Department of Chemical and Life Science Engineering, Virginia Commonwealth University , Richmond, Virginia 23284, United States
| | - Zeng-Quan Yang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States
| | - Olivia M Merkel
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Oncology, Karmanos Cancer Institute, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München , 80539 Munich, Germany
| |
Collapse
|
12
|
Spoelstra NS, Cittelly DM, Christenson JL, Gordon MA, Elias A, Jedlicka P, Richer JK. Dicer expression in estrogen receptor-positive versus triple-negative breast cancer: an antibody comparison. Hum Pathol 2016; 56:40-51. [PMID: 27260947 DOI: 10.1016/j.humpath.2016.05.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/10/2016] [Accepted: 05/19/2016] [Indexed: 01/20/2023]
Abstract
Dicer is an RNase III enzyme responsible for cleaving double-stranded RNAs into small interfering RNAs and microRNAs, which either target messenger RNA transcripts for degradation or inhibit translation. Dicer protein levels have been examined in breast cancer with contradictory results. Our goal was to resolve whether Dicer levels differ in breast cancer versus normal breast epithelium and between estrogen receptor-α-positive (ER+) or estrogen receptor-α-negative (ER-) primary breast cancers. We compared 3 different Dicer antibodies: Abcam 4A6, Abcam ab5818, and Sigma HPA000694, using immunohistochemistry and Western blot analyses. All 3 Dicer antibodies detected higher levels of Dicer in ER+ breast cancer cell lines versus ER-, and all 3 recognized exogenous overexpressed Dicer. In clinical specimens, all 3 antibodies detected higher Dicer in ER+ breast cancers versus triple-negative breast cancer (TNBC) but had very different staining patterns by immunohistochemistry on the same tumor samples. Using the optimal antibody, ab5818, selected for its sensitivity and specificity, Dicer protein expression was significantly higher in ER+ versus TNBC clinical specimens of primary tumor (P<.0001, unpaired t test). Dicer was also significantly higher in adjacent normal breast epithelium versus TNBC (P<.0001, paired t test; n=18 pairs). Differences in antibody performance may explain contrasting results observed in the literature regarding Dicer protein in breast cancer. If Dicer becomes more clinically relevant as a prognostic indicator, further antibody optimization and standardization will be critical.
Collapse
Affiliation(s)
- Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Diana M Cittelly
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Anthony Elias
- Division of Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.
| |
Collapse
|
13
|
Labidi-Galy SI, Clauss A, Ng V, Duraisamy S, Elias KM, Piao HY, Bilal E, Davidowitz RA, Lu Y, Badalian-Very G, Györffy B, Kang UB, Ficarro SB, Ganesan S, Mills GB, Marto JA, Drapkin R. Elafin drives poor outcome in high-grade serous ovarian cancers and basal-like breast tumors. Oncogene 2015; 34:373-83. [PMID: 24469047 PMCID: PMC4112176 DOI: 10.1038/onc.2013.562] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/08/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022]
Abstract
High-grade serous ovarian carcinoma (HGSOC) and basal-like breast cancer (BLBC) share many features including TP53 mutations, genomic instability and poor prognosis. We recently reported that Elafin is overexpressed by HGSOC and is associated with poor overall survival. Here, we confirm that Elafin overexpression is associated with shorter survival in 1000 HGSOC patients. Elafin confers a proliferative advantage to tumor cells through the activation of the MAP kinase pathway. This mitogenic effect can be neutralized by RNA interference, specific antibodies and a MEK inhibitor. Elafin expression in patient-derived samples was also associated with chemoresistance and strongly correlates with bcl-xL expression. We extended these findings into the examination of 1100 primary breast tumors and six breast cancer cell lines. We observed that Elafin is overexpressed and secreted specifically by BLBC tumors and cell lines, leading to a similar mitogenic effect through activation of the MAP kinase pathway. Here too, Elafin overexpression is associated with poor overall survival, suggesting that it may serve as a biomarker and therapeutic target in this setting.
Collapse
Affiliation(s)
- S. Intidhar Labidi-Galy
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Adam Clauss
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Vivian Ng
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
| | - Sekhar Duraisamy
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kevin M. Elias
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
- Brigham and Women’s Hospital, Division of Gynecologic Oncology, Boston, MA
| | - Hui-Ying Piao
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
| | - Erhan Bilal
- The Cancer Institute of New Jersey, Robert Wood Johnson Medical School-University of Medicine and Dentistry of New Jersey, New Brunswick, NJ
| | | | - Yiling Lu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gayane Badalian-Very
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Balázs Györffy
- Research Laboratory of Pediatrics and Nephrology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Un-Beom Kang
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Blais Proteomics Center, Boston, MA
| | - Scott B. Ficarro
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Blais Proteomics Center, Boston, MA
| | - Shridar Ganesan
- The Cancer Institute of New Jersey, Robert Wood Johnson Medical School-University of Medicine and Dentistry of New Jersey, New Brunswick, NJ
| | - Gordon B. Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jarrod A. Marto
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Blais Proteomics Center, Boston, MA
| | - Ronny Drapkin
- Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
- Brigham and Women’s Hospital, Department of Pathology, Boston, MA
| |
Collapse
|
14
|
Hashmi AA, Edhi MM, Naqvi H, Khurshid A, Faridi N. Molecular subtypes of breast cancer in South Asian population by immunohistochemical profile and Her2neu gene amplification by FISH technique: association with other clinicopathologic parameters. Breast J 2014; 20:578-85. [PMID: 25219294 DOI: 10.1111/tbj.12329] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Molecular breast cancer subtypes were defined by gene expression prolife; however, immunohistochemical (IHC) expression can categorize breast cancers analogous to gene expression profiling. We aimed to evaluate distribution of these molecular breast cancer subtypes in our population and their association with clinocopathologic parameters. We retrospectively analyzed 1,104 cases of primary breast cancers over 3 years duration. ER, PR, Her2neu IHC staining, and subsequent fluorescent in situ hybridization studies (Her2neu gene amplification in cases with 2+ IHC staining) were performed to categorize breast cancer subtypes. Luminal A breast cancers were most frequent (45.8%) followed by triple negative (18.6%), luminal B (17.8%) and Her2neu (17.8%) subtypes. We found a strong association of breast cancer subtypes with tumor grade and Ki67 proliferation index with triple negative cancers being associated with higher grade and proliferation index. Significant association was seen with age groups, luminal A subtype occurring at a slightly older age, whereas triple negative and Her2neu cancers were more frequent in younger age group. We found a higher proportion of triple negative cancers in our set up, and they were found to have high-tumor grade and proliferation index along with presentation at younger age. As these cancers are associated with BRCA 1 mutations and abnormal BRCA 1 pathways, we suggest that large scale studies should be done to evaluate BRCA 1 mutations and abnormal BRCA 1 pathways in our population to establish risk factors for this highly aggressive tumor subtype.
Collapse
Affiliation(s)
- Atif Ali Hashmi
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | | | | | | |
Collapse
|
15
|
Yang G, Xue F, Chen X. Prognostic value of different amounts of cancer stem cells in different molecular subtypes of breast cancer. Gland Surg 2014; 1:20-4. [PMID: 25083423 DOI: 10.3978/j.issn.2227-684x.2012.04.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 03/31/2012] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To investigate the differences in the amount of cancer stem cells in different breast cancer subtypes. METHODS THE SPECIMENS OF BREAST CANCER TISSUES WERE DIVIDED INTO FIVE GROUPS: group A (luminal A type), B (luminal B type), C (HER-2 + type), D (basal-like type) and E (normal-like type) according to their molecular subtypes. The cancer stem cell spheres in each group were measured through colony formation method, and then the relations of the amount of cancer stem cells with the subtypes of breast cancer were analyzed. RESULTS The numbers of cancer stem cell spheres in group A and B were (1.1±0.2)/1,000 cells and (1.3±0.1)/1,000 cells, respectively, and this had no significant difference (P>0.05). The number of cancer stem cell spheres in group C was (8.6±1.0)/1,000 cells, which was significantly higher than that in group A and B (both P<0.05). The numbers of cancer stem cell spheres in group D and E were (22.4±1.2)/1,000 cells and (17.7±2.0)/1,000 cells, respectively, and this was statistically similar (P>0.05) but either was significantly higher than that in the other three groups (all P<0.05). CONCLUSIONS Breast cancer tissues of different molecular subtypes contain varying amounts of cancer stem cells, and this may have certain significance for predicting the prognosis of breast cancer.
Collapse
Affiliation(s)
- Guohua Yang
- Department of Oncology Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Fangqing Xue
- Department of Oncology Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Xiaogeng Chen
- Department of Oncology Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| |
Collapse
|
16
|
Overexpression of Raf-1 in basal-like carcinoma of the breast: correlation with clinicopathology and prognosis. Contemp Oncol (Pozn) 2014; 18:391-5. [PMID: 25784836 PMCID: PMC4355656 DOI: 10.5114/wo.2014.47037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 12/21/2013] [Accepted: 02/03/2014] [Indexed: 11/17/2022] Open
Abstract
Aim of the study Increased Raf-1 expression has been associated with an aggressive behaviour in some carcinomas such as pulmonary carcinoma and renal carcinoma. However, its role in breast cancer, especially in basal-like carcinoma of the breast (BLBC), has not been defined. Material and methods The current study attempted to investigate the expression pattern of Raf-1 protein in BLBC, in relation to the biological behaviour and prognosis of the carcinoma. Expression of Raf-1 was detected by immunohistochemistry in carcinoma specimens from 74 cases of BLBC, and associations between their expression and the clinicopathological characteristics were statistically assessed. Results The patients’ age, tumour size, BRCA1, and p53 protein expression was not significantly different between the Raf-1-positive and Raf-1-negative expression groups (p > 0.05). The proportion of histological grade 3 tumours was not significantly higher in the Raf-1 positive group than that of grade 2 tumours (p > 0.05). However, positive cytoplasmic Raf-1 expression was positively correlated to Ki-67 expression (p < 0.05). Also, increased Raf-1 protein was found to exert an unfavourable impact on patients’ axillary lymph node metastasis and overall survival (p < 0.05). Conclusions The study implies that positive Raf-1 expression in BLBC is associated with a more aggressive phenotype and could be considered as a new prognostic biomarker for poor survival in BLBC patients.
Collapse
|
17
|
Exploring novel targets of basal-like breast carcinoma by comparative gene profiling and mechanism analysis. Breast Cancer Res Treat 2013; 141:23-32. [PMID: 23933801 DOI: 10.1007/s10549-013-2664-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/02/2013] [Indexed: 12/30/2022]
Abstract
The heterogeneity of breast cancer makes its diagnosis and treatment far from being optimal. Analysis of traditional pathological and prognostic markers based on immunohistochemistry (IHC) is inadequate in elucidating the inherent heterogeneity of breast cancer, especially basal-like breast carcinoma (BLBC) which displays complex and unique epidemiological, phenotypic, and molecular features with distinctive relapse patterns and poor clinical outcomes. Gene expression profiling opened an avenue in research as independent predictors by classifying breast cancers into discrete groups with prognostic references, but it is not cost-effective in clinical application. It is necessary to develop an effective predictive gene list from gene profiling to optimize the treatment with traditional markers. In this report, we analyzed the correlation between IHC and gene profiling of breast cancer with an emphasis on the BLBC, highlighting the potential discovery of diagnostic markers and cellular mechanisms that may guide the development of BLBC-targeted therapy. Random forest-based classification and PAM50 gene-sets were used in the comparison analysis of traditional prognostic markers including estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and microarray profiles. An intrinsic 40-gene set was developed to classify breast cancer subtypes, and genes expression differentiations were used to explore the different mechanisms between the BLBC and non-BLBC subtypes based on the comparison of clinicopathological markers and microarray profiling. Pathways and DNA repairs were analyzed to evaluate the biological mechanisms in BLBC and other breast cancer subtypes. It is reasonable to define BLBC as those tumors that are negative for ER, PR, and HER2 by IHC for their accordance with gene expression profiles. Focal adhesion kinase, ERBB, and their signaling pathways may play crucial role in BLBC. The intrinsic 40-gene set can be used to classify breast cancer and help to optimize therapeutic management of BLBC.
Collapse
|
18
|
Gupta P, Adkins C, Lockman P, Srivastava SK. Metastasis of Breast Tumor Cells to Brain Is Suppressed by Phenethyl Isothiocyanate in a Novel In Vivo Metastasis Model. PLoS One 2013; 8:e67278. [PMID: 23826254 PMCID: PMC3695065 DOI: 10.1371/journal.pone.0067278] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 05/16/2013] [Indexed: 01/28/2023] Open
Abstract
Breast tumor metastasis is a leading cause of cancer-related deaths worldwide. Breast tumor cells frequently metastasize to brain and initiate severe therapeutic complications. The chances of brain metastasis are further elevated in patients with HER2 overexpression. In the current study, we evaluated the anti-metastatic effects of phenethyl isothiocyanate (PEITC) in a novel murine model of breast tumor metastasis. The MDA-MB-231-BR (BR-brain seeking) breast tumor cells stably transfected with luciferase were injected into the left ventricle of mouse heart and the migration of cells to brain was monitored using a non-invasive IVIS bio-luminescent imaging system. In order to study the efficacy of PEITC in preventing the number of tumor cells migrating to brain, mice were given 10 µmol PEITC by oral gavage for ten days prior to intra-cardiac injection of tumor cells labeled with quantum dots. To evaluate the tumor growth suppressive effects, 10 µmol PEITC was given to mice every day starting 14(th) day after intra-cardiac cell injection. Based on the presence of quantum dots in the brain section of control and treated mice, our results reveal that PEITC significantly prevented the metastasis of breast cancer cells to brain. Our results demonstrate that the growth of metastatic brain tumors in PEITC treated mice was about 50% less than that of control. According to Kaplan Meir's curve, median survival of tumor bearing mice treated with PEITC was prolonged by 20.5%. Furthermore as compared to controls, we observed reduced HER2, EGFR and VEGF expression in the brain sections of PEITC treated mice. To the best of our knowledge, our study for the first time demonstrates the anti-metastatic effects of PEITC in vivo in a novel breast tumor metastasis model and provides the rationale for further clinical investigation.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
- Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Chris Adkins
- Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Paul Lockman
- Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Sanjay K. Srivastava
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
- Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| |
Collapse
|
19
|
Davis MB, Liu X, Wang S, Reeves J, Khramtsov A, Huo D, Olopade OI. Expression and sub-cellular localization of an epigenetic regulator, co-activator arginine methyltransferase 1 (CARM1), is associated with specific breast cancer subtypes and ethnicity. Mol Cancer 2013; 12:40. [PMID: 23663560 PMCID: PMC3663705 DOI: 10.1186/1476-4598-12-40] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 04/03/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Co-Activator Arginine Methyltransferase 1(CARM1) is an Estrogen Receptor (ER) cofactor that remodels chromatin for gene regulation via methylation of Histone3. We investigated CARM1 levels and localization across breast cancer tumors in a cohort of patients of either European or African ancestry. METHODS We analyzed CARM1 levels using tissue microarrays with over 800 histological samples from 549 female cancer patients from the US and Nigeria, Africa. We assessed associations between CARM1 expression localized to the nucleus and cytoplasm for 11 distinct variables, including; ER status, Progesterone Receptor status, molecular subtypes, ethnicity, HER2+ status, other clinical variables and survival. RESULTS We found that levels of cytoplasmic CARM1 are distinct among tumor sub-types and increased levels are associated with ER-negative (ER-) status. Higher nuclear CARM1 levels are associated with HER2 receptor status. EGFR expression also correlates with localization of CARM1 into the cytoplasm. This suggests there are distinct functions of CARM1 among molecular tumor types. Our data reveals a basal-like subtype association with CARM1, possibly due to expression of Epidermal Growth Factor Receptor (EGFR). Lastly, increased cytoplasmic CARM1, relative to nuclear levels, appear to be associated with self-identified African ethnicity and this result is being further investigated using quantified genetic ancestry measures. CONCLUSIONS Although it is known to be an ER cofactor in breast cancer, CARM1 expression levels are independent of ER. CARM1 has distinct functions among molecular subtypes, as is indicative of its sub-cellular localization and it may function in subtype etiology. These sub-cellular localization patterns, indicate a novel role beyond its ER cofactor function in breast cancer. Differential localization among ethnic groups may be due to ancestry-specific polymorphisms which alter the gene product.
Collapse
Affiliation(s)
- Melissa B Davis
- The Institute for Genomics and Systems Biology, University of Chicago Biological Sciences Division, Chicago, IL, USA
- Department of Human Genetics, University of Chicago Biological Sciences Division, Chicago, IL, USA
- Georgia Health Sciences University, Athens, GA 30602, USA
- University of Georgia Medical Partnership, Athens, GA 30602, USA
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Xinyu Liu
- Department of Statistics, University of Georgia, Athens, GA 30602, USA
| | - Shiyao Wang
- Department of Statistics, University of Georgia, Athens, GA 30602, USA
| | - Jaxk Reeves
- Department of Statistics, University of Georgia, Athens, GA 30602, USA
| | - Andrey Khramtsov
- Department of Medicine, Center for Clinical Cancer Genetics and Global Health, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Dezheng Huo
- Department of Health Studies, Center for Clinical Cancer Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Olufunmilayo I Olopade
- Department of Medicine, Center for Clinical Cancer Genetics and Global Health, University of Chicago Medicine, Chicago, IL 60637, USA
| |
Collapse
|
20
|
Law ME, Corsino PE, Jahn SC, Davis BJ, Chen S, Patel B, Pham K, Lu J, Sheppard B, Nørgaard P, Hong J, Higgins P, Kim JS, Luesch H, Law BK. Glucocorticoids and histone deacetylase inhibitors cooperate to block the invasiveness of basal-like breast cancer cells through novel mechanisms. Oncogene 2012; 32:1316-29. [PMID: 22543582 DOI: 10.1038/onc.2012.138] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Aggressive cancers often express E-cadherin in cytoplasmic vesicles rather than on the plasma membrane and this may contribute to the invasive phenotype of these tumors. Therapeutic strategies are not currently available that restore the anti-invasive function of E-cadherin in cancers. MDA-MB-231 cells are a frequently used model of invasive triple-negative breast cancer, and these cells express low levels of E-cadherin that is mislocalized to cytoplasmic vesicles. MDA-MB-231 cell lines stably expressing wild-type E-cadherin or E-cadherin fused to glutathione S-transferase or green fluorescent protein were used as experimental systems to probe the mechanisms responsible for cytoplasmic E-cadherin localization in invasive cancers. Although E-cadherin expression partly reduced cell invasion in vitro, E-cadherin was largely localized to the cytoplasm and did not block the invasiveness of the corresponding orthotopic xenograft tumors. Further studies indicated that the glucocorticoid dexamethasone and the highly potent class I histone deacetylase (HDAC) inhibitor largazole cooperated to induce E-cadherin localization to the plasma membrane in triple-negative breast cancers, and to suppress cellular invasion in vitro. Dexamethasone blocked the production of the cleaved form of the CDCP1 (that is, CUB domain-containing protein 1) protein (cCDCP1) previously implicated in the pro-invasive activities of CDCP1 by upregulating the serine protease inhibitor plasminogen activator inhibitor-1. E-cadherin preferentially associated with cCDCP1 compared with the full-length form. In contrast, largazole did not influence CDCP1 cleavage, but increased the association of E-cadherin with γ-catenin. This effect on E-cadherin/γ-catenin complexes was shared with the nonisoform selective HDAC inhibitors trichostatin A (TSA) and vorinostat (suberoylanilide hydroxamic acid, SAHA), although largazole upregulated endogenous E-cadherin levels more strongly than TSA. These results demonstrate that glucocorticoids and HDAC inhibitors, both of which are currently in clinical use, cooperate to suppress the invasiveness of breast cancer cells through novel, complementary mechanisms that converge on E-cadherin.
Collapse
Affiliation(s)
- M E Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville 32610-0267, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Rosmaninho A, Velho G, Caetano M, Selores M. Breast cancer: 2 case reports. An Bras Dermatol 2012; 87:123-6. [PMID: 22481661 DOI: 10.1590/s0365-05962012000100016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 05/14/2011] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is the most frequently diagnosed life-threatening cancer in women and the leading cause of cancer death among them worldwide. It includes a heterogeneous collection of diseases with various histologically defined subsets, clinical presentations, responses to treatment and outcomes. We describe 2 cases of female patients with ductal breast carcinoma. Dermatologists may have an important role in diagnosing such diseases.
Collapse
|