1
|
Houcine Y, Ben Salem H, El Fekih S, Maaoui A, Driss M. Immunohistochemical expression of epidermal growth factor receptor: prognostic value in HER2 positive breast cancer. J Immunoassay Immunochem 2025; 46:262-273. [PMID: 40059383 DOI: 10.1080/15321819.2025.2475291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Epidermal Growth Factor Receptor (EGFR) expression is not well-studied in Human Epidermal Growth Factor Receptor 2 (HER2) positive breast cancer. We aim to study the prevalence of EGFR immunohistochemical expression in HER2-positive breast carcinomas and to correlate this expression with different anatomo-clinical parameters. METHODS It was a retrospective study involving cases of HER2-positive breast carcinoma collected at the Immuno-Histo-Cytology Department of Salah Azaïez Institute of Tunis between 2018 and 2020. An immunohistochemical study using the anti-human EGFR monoclonal antibody was performed. Cases with an overall score ≥1+ were considered positive. RESULTS Fifty patients were included. EGFR expression in HER2-positive breast carcinomas was more likely to occur in patients under the age of 50 (p = 0.063). It was significantly associated with the absence of lymphovascular invasion (p = 0.047). In multivariate analysis, young age, absence of lympho-vascular invasion, and high Ki67 proliferation index (>60%) were independently associated with positive EGFR expression (p = 0.047, p = 0.040, and p = 0.050, respectively). CONCLUSION Through this first Tunisian study, our data revealed that the immunohistochemical expression of EGFR is associated with young age, absence of lymphovascular invasion, and a high mitotic index (Ki67), which may suggest a potential predictive value for chemotherapy response.
Collapse
Affiliation(s)
- Yoldez Houcine
- Immuno-histo-cytology department, Salah Azaiz Institute, Tunis, Tunisia
- Faculty of Medicine, El Manar University, Tunis, Tunisia
| | - Hend Ben Salem
- Faculty of Medicine, El Manar University, Tunis, Tunisia
| | - Sirine El Fekih
- Immuno-histo-cytology department, Salah Azaiz Institute, Tunis, Tunisia
| | - Amal Maaoui
- Immuno-histo-cytology department, Salah Azaiz Institute, Tunis, Tunisia
| | - Maha Driss
- Immuno-histo-cytology department, Salah Azaiz Institute, Tunis, Tunisia
- Faculty of Medicine, El Manar University, Tunis, Tunisia
| |
Collapse
|
2
|
Long L, Fei X, Chen L, Yao L, Lei X. Potential therapeutic targets of the JAK2/STAT3 signaling pathway in triple-negative breast cancer. Front Oncol 2024; 14:1381251. [PMID: 38699644 PMCID: PMC11063389 DOI: 10.3389/fonc.2024.1381251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its propensity for metastasis and poor prognosis. TNBC evades the body's immune system recognition and attack through various mechanisms, including the Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. This pathway, characterized by heightened activity in numerous solid tumors, exhibits pronounced activation in specific TNBC subtypes. Consequently, targeting the JAK2/STAT3 signaling pathway emerges as a promising and precise therapeutic strategy for TNBC. The signal transduction cascade of the JAK2/STAT3 pathway predominantly involves receptor tyrosine kinases, the tyrosine kinase JAK2, and the transcription factor STAT3. Ongoing preclinical studies and clinical research are actively investigating this pathway as a potential therapeutic target for TNBC treatment. This article comprehensively reviews preclinical and clinical investigations into TNBC treatment by targeting the JAK2/STAT3 signaling pathway using small molecule compounds. The review explores the role of the JAK2/STAT3 pathway in TNBC therapeutics, evaluating the benefits and limitations of active inhibitors and proteolysis-targeting chimeras in TNBC treatment. The aim is to facilitate the development of novel small-molecule compounds that target TNBC effectively. Ultimately, this work seeks to contribute to enhancing therapeutic efficacy for patients with TNBC.
Collapse
Affiliation(s)
- Lin Long
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiangyu Fei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liucui Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang Yao
- Department of Pharmacy, Central Hospital of Hengyang, Hengyang, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
4
|
Hamilton AM, Walens A, Van Alsten SC, Olsson LT, Nsonwu-Farley J, Gao X, Kirk EL, Perou CM, Carey LA, Troester MA, Abdou Y. BIRC5 expression by race, age and clinical factors in breast cancer patients. Breast Cancer Res 2024; 26:50. [PMID: 38515208 PMCID: PMC10956264 DOI: 10.1186/s13058-024-01792-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
PURPOSE Survivin/BIRC5 is a proliferation marker that is associated with poor prognosis in breast cancer and an attractive therapeutic target. However, BIRC5 has not been well studied among racially diverse populations where aggressive breast cancers are prevalent. EXPERIMENTAL DESIGN We studied BIRC5 expression in association with clinical and demographic variables and as a predictor of recurrence in 2174 participants in the Carolina Breast Cancer Study (CBCS), a population-based study that oversampled Black (n = 1113) and younger (< 50 years; n = 1137) participants with breast cancer. For comparison, similar analyses were conducted in The Cancer Genome Atlas [TCGA N = 1094, Black (n = 183), younger (n = 295)]. BIRC5 was evaluated as a continuous and categorical variable (highest quartile vs. lower three quartiles). RESULTS Univariate, continuous BIRC5 expression was higher in breast tumors from Black women relative to non-Black women in both estrogen receptor (ER)-positive and ER-negative tumors and in analyses stratified by stage (i.e., within Stage I, Stage II, and Stage III/IV tumors). Within CBCS and TCGA, BIRC5-high was associated with young age (< 50 years) and Black race, as well as hormone receptor-negative tumors, non-Luminal A PAM50 subtypes, advanced stage, and larger tumors (> 2 cm). Relative to BIRC5-low, BIRC5-high tumors were associated with poor 5-year recurrence-free survival (RFS) among ER-positive tumors, both in unadjusted models [HR (95% CI): 2.7 (1.6, 4.6)] and after adjustment for age and stage [Adjusted HR (95% CI): 1.87 (1.07, 3.25)]. However, this relationship was not observed among ER-negative tumors [Crude HR (95% CI): 0.7 (0.39, 1.2); Adjusted HR (95% CI): 0.67 (0.37, 1.2)]. CONCLUSION Black and younger women with breast cancer have a higher burden of BIRC5-high tumors than older and non-Black women. Emerging anti-survivin treatment strategies may be an important future direction for equitable breast cancer outcomes.
Collapse
Affiliation(s)
- Alina M Hamilton
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrea Walens
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sarah C Van Alsten
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Linnea T Olsson
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joseph Nsonwu-Farley
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiaohua Gao
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Erin L Kirk
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Charles M Perou
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Melissa A Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
- Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, CB# 7305, Chapel Hill, NC, 27514, USA.
| |
Collapse
|
5
|
Facca VJ, Cai Z, Ku A, Georgiou CJ, Reilly RM. Adjuvant Auger Electron-Emitting Radioimmunotherapy with [ 111In]In-DOTA-Panitumumab in a Mouse Model of Local Recurrence and Metastatic Progression of Human Triple-Negative Breast Cancer. Mol Pharm 2023; 20:6407-6419. [PMID: 37983089 DOI: 10.1021/acs.molpharmaceut.3c00780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Triple-negative breast cancer (TNBC) has a high risk for recurrence and metastasis. We studied the effectiveness of Auger electron (AE) radioimmunotherapy (RIT) with antiepidermal growth factor receptor (EGFR) panitumumab conjugated with DOTA complexed to 111In ([111In]In-DOTA-panitumumab) for preventing metastatic progression after local treatment of 231/LM2-4 Luc+ human TNBC tumors in the mammary fat pad of NRG mice. Prior to RIT, the primary tumor was resected, and tumor margins were treated with X-irradiation (XRT; 5 days × 6 Gy/d). RIT was administered 1 day post-XRT by intravenous injection of 26 MBq (15 μg) or 2 × 10 MBq (15 μg each) separated by 7 d. These treatments were compared to tumor resection with or without XRT combined with DOTA-panitumumab (15 μg) or irrelevant [111In]In-DOTA-IgG2 (24 MBq; 15 μg), and efficacy was evaluated by Kaplan-Meier survival curves. The effect of [111In]In-DOTA-panitumumab (23 MBq; 15 μg) after tumor resection without local XRT was also studied. Tumor resection followed by XRT and RIT with 26 MBq [111In]In-DOTA-panitumumab significantly increased the median survival to 35 d compared to tumor resection with or without XRT (23-24 d; P < 0.0001). Local treatment with tumor resection and XRT followed by 2 × 10 MBq of [111In]In-DOTA-panitumumab, DOTA-panitumumab, or [111In]In-DOTA-IgG2 did not significantly improve median survival (26 days for all treatments). RIT alone with [111In]In-DOTA-panitumumab postresection of the tumor without XRT increased median survival to 29 days, though this was not significant. Despite significantly improved survival in mice treated with tumor resection, XRT, and RIT with [111In]In-DOTA-panitumumab, all mice eventually succumbed to advanced metastatic disease by 45 d post-tumor resection. SPECT/CT with [111In]In-DOTA-panitumumab, PET/MRI with [64Cu]Cu-DOTA-panitumumab F(ab')2, and PET/CT with [18F]FDG were used to detect recurrent and metastatic disease. Uptake of [111In]In-DOTA-panitumumab at 4 d p.i. in the MFP tumor was 26.8 ± 9.7% ID/g and in metastatic lymph nodes (LN), lungs, and liver was 34.2 ± 26.9% ID/g, 17.5 ± 6.0% ID/g, and 9.4 ± 2.4%ID/g, respectively, while uptake in the lungs (6.0 ± 0.9% ID/g) and liver (5.2 ± 2.9% ID/g) of non-tumor-bearing NRG was significantly lower (P < 0.05). Radiation-absorbed doses in metastatic LN, lungs, and liver were 9.7 ± 6.1, 6.4 ± 2.1, and 10.9 ± 2.7 Gy, respectively. In conclusion, we demonstrated that RIT with [111In]In-DOTA-panitumumab combined with tumor resection and XRT significantly improved the survival of mice with recurrent TNBC. However, the aggressive nature of 231/LM2-4 Luc+ tumors in NRG mice may have contributed to the tumor recurrence and progression observed.
Collapse
Affiliation(s)
- Valerie J Facca
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Anthony Ku
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Constantine J Georgiou
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada
- Department of Medical Imaging, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Joint Department of Medical Imaging and Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| |
Collapse
|
6
|
Quidute P, Quidute R, Perez MM, Pereira EC, da Veiga GL, Alves BDCA, Fonseca FLA. Survivin expression as a prognostic marker for breast cancer. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230167. [PMID: 37729357 PMCID: PMC10508893 DOI: 10.1590/1806-9282.20230167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 09/22/2023]
Abstract
OBJECTIVE Due to the speed of development observed in breast cancer, several studies aimed at discovering new biomarkers have been carried out in order to arrive at an early diagnosis. As survivin plays a fundamental role in the evasion of apoptosis in tumor cells, the aim of this study was to verify the expression profile of the survivin gene in paraffin-embedded breast tumor samples and associate it with the clinical characteristics of the patients. METHODS This is a cross-sectional study, for which 100 tumor samples were obtained from cancer patients treated throughout the year 2019 at Instituto de Mama do Cariri (Juazeiro do Norte, in the state of Ceará). This study included women over 30 years old who had confirmed breast cancer through anatomopathological examination but excluded those with non-neoplastic breast comorbidities, other neoplasms, or chronic diseases. Survivin gene expression was assessed by quantitative polymerase chain reaction. RESULTS The expression of survivin is associated with the lack of expression of estrogen (p=0.027) and progesterone (p>0.0005) receptors. It means that survivin expression is higher in patients in which labeling was absent for estrogen receptor and progesterone receptor. CONCLUSION Our data reinforce that survivin expression is higher in estrogen receptor-patients, thus representing an additional prognostic tool.
Collapse
Affiliation(s)
- Patricia Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Ricardo Quidute
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | - Matheus Moreira Perez
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Glaucia Luciano da Veiga
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
| | | | - Fernando Luiz Affonso Fonseca
- Centro Universitário Faculdade de Medicina do ABC, Laboratório de Análises Clínicas – Santo André (SP), Brazil
- Universidade Federal de São Paulo, Instituto de Ciências Farmacêuticas – Diadema (SP), Brazil
| |
Collapse
|
7
|
Ipinloju N, Ibrahim A, da Costa RA, Adigun TB, Olubode SO, Abayomi KJ, Aiyelabegan AO, Esan TO, Muhammad SA, Oyeneyin OE. Quantum evaluation and therapeutic activity of (E)-N-(4-methoxyphenyl)-2-(4-(3-oxo-3-phenylprop-1-en-1-yl) phenoxy)acetamide and its modified derivatives against EGFR and VEGFR-2 in the treatment of triple-negative cancer via in silico approach. J Mol Model 2023; 29:159. [PMID: 37099048 DOI: 10.1007/s00894-023-05543-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 04/03/2023] [Indexed: 04/27/2023]
Abstract
The most dangerous subtype of breast cancer, triple-negative breast cancer (TNBC), accounts for 25% of all breast cancer-related deaths and 15% of all breast cancer cases. TNBC is distinguished by the lack of immunohistochemical expression of HER2, progesterone receptors, or oestrogen receptors. Although it has been reported that upregulation of EGFR and VEGFR-2 is associated with TNBC progression, no proven effective targeted therapy exists at this time. We used structural bioinformatics methods, including density functional theory, molecular docking, molecular dynamic simulation, pharmacokinetic and drug-likeness models, to identify promising EGFR/VEGFR-2 inhibitors from N-(4-methoxyphenyl)-2-[4-(3-oxo-3-phenylprop-1-en-1-yl) phenoxy] acetamide and six of its modified derivatives in light of the lack of effective targets inhibitor Version 14 of Spartan software was used to analyse density functional theory. The Schrodinger software suite 2018's Maestro interface was used for the molecular docking analysis, and the admetSAR and swissADME servers were used for drug-likeness and absorption, distribution, metabolism, excretion, and toxicity. All of the compounds showed strong electronic characteristics. Additionally, all of the tested compounds met the ADMET and drug-likeness requirements without a single instance of Lipinski's rule of five violations. Additionally, the molecules' levels of affinity for the target proteins varied. The highest binding affinities were demonstrated by the MOLb-VEGFR-2 complex (- 9.925 kcal/mol) and the MOLg-EGFR complex (- 5.032 kcal/mol). The interaction of the molecules in the domain of the EGFR and VEGFR-2 receptors was also better understood through molecular dynamic simulation of the complex.
Collapse
Affiliation(s)
- Nureni Ipinloju
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.
| | - Abdulwasiu Ibrahim
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria.
| | - Renato Araujo da Costa
- Institute of Education, Science and Technology of Para (IFPA), Campus Abaetetuba, Abaetetuba, Para, Brazil
| | | | - Samuel Olawale Olubode
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Kehinde Joan Abayomi
- Faculty of Medical Laboratory Science, Usmanu DanFodiyo University, Sokoto, Nigeria
| | | | - Timothy Oluwaseun Esan
- Department of Chemical Sciences, Bamidele Olumilua University of Education Science and Technology, Ikere-Ekiti, Ekiti State, Nigeria
| | | | - Oluwatoba Emmanuel Oyeneyin
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.
| |
Collapse
|
8
|
Zhang Q, Kanyomse Q, Luo C, Mo Q, Zhao X, Wang L, Peng W, Ren G. The Prognostic Value of ADAMTS8 and Its Role as a Tumor Suppressor in Breast Cancer. Cancer Invest 2023; 41:119-132. [PMID: 36346393 DOI: 10.1080/07357907.2022.2128367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A disintegrin-like and metalloprotease with therombospondin type1 motif 8 (ADAMTS8) plays an important role in many malignancies. However, the clinical and biological significance of ADAMTS8 in breast cancer remain unknown. In this study, the clinical data from 1066 breast cancer patients were analyzed by The Cancer Genome Atlas (TCGA) database, and were analyzed using the correlation between ADAMTS8 expression and the clinicopathological features and prognoses. The CCK-8 assay, clone formation assay, flow cytometry and Transwell assay were used to characterize the effects of ADAMTS8 on proliferation, migration and invasion of breast cancer cells. Gene set enrichment analysis (GSEA) and western blotting were used to identify the potential molecular mechanism on how ADAMTS8 exert its biological function. ADAMTS8 overexpression correlated longer overall survival (OS) and progression-free survival (PFS). ADAMTS8 was considered as an independent prognostic factor for OS. ADAMTS8 overexpression inhibited breast cancer cell proliferation, migration and invasion in vitro, and induced G2/M cell cycle arrest. ADAMTS8 was also involved in cell cycle regulation and was associated with the EGFR/Akt signaling pathway. ADAMTS8 knockdown showed the reverse effect. Together, the results showed that ADAMTS8 functioned as a tumor suppressor gene (TGS) and could be a prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Qia Zhang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quist Kanyomse
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenghao Luo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingfan Mo
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - XunPing Zhao
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Long Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Garlapati C, Joshi S, Bhattarai S, Krishnamurthy J, Turaga RC, Nguyen T, Li X, Aneja R. PLK1 and AURKB phosphorylate survivin differentially to affect proliferation in racially distinct triple-negative breast cancer. Cell Death Dis 2023; 14:12. [PMID: 36627281 PMCID: PMC9832024 DOI: 10.1038/s41419-022-05539-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023]
Abstract
Protein diversity due to alternative mRNA splicing or post-translational modifications (PTMs) plays a vital role in various cellular functions. The mitotic kinases polo-like kinase 1 (PLK1) and Aurora B (AURKB) phosphorylate survivin, an inhibitor of apoptosis (IAP) family member, thereby regulating cell proliferation. PLK1, AURKB, and survivin are overexpressed in triple-negative breast cancer (TNBC), an aggressive breast cancer subtype. TNBC is associated with high proliferative capacity, high rates of distant metastasis, and treatment resistance. The proliferation-promoting protein survivin and its activating kinases, PLK1 and AURKB, are overexpressed in TNBC. In this study, we investigated the role of survivin phosphorylation in racial disparities in TNBC cell proliferation. Analysis of TCGA TNBC data revealed higher expression levels of PLK1 (P = 0.026) and AURKB (P = 0.045) in African Americans (AAs; n = 41) than in European Americans (EAs; n = 86). In contrast, no significant racial differences in survivin mRNA or protein levels were observed. AA TNBC cells exhibited higher p-survivin levels than EA TNBC cells. Survivin silencing using small interfering RNAs significantly attenuated cell proliferation and cell cycle progression in AA TNBC cells, but not in EA TNBC cells. In addition, PLK1 and AURKB inhibition with volasertib and barasertib significantly inhibited the growth of AA TNBC xenografts, but not of EA TNBC tumors. These data suggest that inhibition of PLK1 and AURKB suppresses cell proliferation and tumor growth, specifically in AA TNBC. These findings suggest that targeting survivin phosphorylation may be a viable therapeutic option for AA patients with TNBC.
Collapse
Affiliation(s)
- Chakravarthy Garlapati
- Alkermes Inc, Waltham, MA, 02451, USA
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Shristi Bhattarai
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | | | | | - Thi Nguyen
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xiaoxian Li
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
10
|
Adinew GM, Messeha S, Taka E, Soliman KFA. The Prognostic and Therapeutic Implications of the Chemoresistance Gene BIRC5 in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:cancers14215180. [PMID: 36358602 PMCID: PMC9659000 DOI: 10.3390/cancers14215180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
Chemoresistance affects TNBC patient treatment responses. Therefore, identifying the chemoresistant gene provides a new approach to understanding chemoresistance in TNBC. BIRC5 was examined in the current study as a tool for predicting the prognosis of TNBC patients and assisting in developing alternative therapies using online database tools. According to the examined studies, BIRC5 was highly expressed in 45 to 90% of TNBC patients. BIRC5 is not only abundantly expressed but also contributes to resistance to chemotherapy, anti-HER2 therapy, and radiotherapy. Patients with increased expression of BIRC5 had a median survival of 31.2 months compared to 85.8 months in low-expression counterparts (HR, 1.73; CI, 1.4−2.13; p = 2.5 × 10−7). The overall survival, disease-free survival, relapse-free survival, distant metastasis-free survival, and the complete pathological response of TNBC patients with high expression of BIRC5 who received any chemotherapy (Taxane, Ixabepilone, FAC, CMF, FEC, Anthracycline) and anti-HER2 therapy (Trastuzumab, Lapatinib) did not differ significantly from those patients receiving any other treatment. Data obtained indicate that the BIRC5 promoter region was substantially methylated, and hypermethylation was associated with higher BIRC5 mRNA expression (p < 0.05). The findings of this study outline the role of BIRC5 in chemotherapy-induced resistance of TNBC, further indicating that BIRC5 may serve as a promising prognostic biomarker that contributes to chemoresistance and could be a possible therapeutic target. Meanwhile, several in vitro studies show that flavonoids were highly effective in inhibiting BIRC5 in genetically diverse TNBC cells. Therefore, flavonoids would be a promising strategy for preventing and treating TNBC patients with the BIRC5 molecule.
Collapse
|
11
|
Facca VJ, Cai Z, Gopal NEK, Reilly RM. Panitumumab-DOTA- 111In: An Epidermal Growth Factor Receptor Targeted Theranostic for SPECT/CT Imaging and Meitner-Auger Electron Radioimmunotherapy of Triple-Negative Breast Cancer. Mol Pharm 2022; 19:3652-3663. [PMID: 35926098 DOI: 10.1021/acs.molpharmaceut.2c00457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Epidermal growth factor receptors (EGFR) are overexpressed in triple-negative breast cancer (TNBC) and are an attractive target for the development of theranostic radiopharmaceuticals. We studied anti-EGFR panitumumab labeled with 111In (panitumumab-DOTA-111In) for SPECT/CT imaging and Meitner-Auger electron (MAE) radioimmunotherapy (RIT) of TNBC. Panitumumab-DOTA-111In was bound, internalized, and routed to the nucleus in MCF7, MDA-MB-231/Luc, and MDA-MB-468 human breast cancer (BC) cells dependent on the EGFR expression level (1.5 × 104, 1.7 × 105, or 1.3 × 106 EGFR/cell, respectively). The absorbed dose in the nuclei of MCF7, MDA-MB-231/Luc, and MDA-MB-468 cells incubated with 4.4 MBq of panitumumab-DOTA-111In (20 nM) was 1.20 ± 0.02, 2.2 ± 0.1, and 25 ± 2 Gy, respectively. The surviving fraction (SF) of MDA-MB-231/Luc cells treated with panitumumab-DOTA-111In (10-300 nM; 1.5 MBq/μg) was reduced as the absorbed dose in the cell increased, with clonogenic survival reduced to an SF = 0.12 ± 0.05 at 300 nM corresponding to 12.7 Gy. The SFs of MDA-MB-468, MDA-MB-231/Luc, and MCF7 cells treated with panitumumab-DOTA-111In (20 nM; 1.7 MBq/μg) were <0.01, 0.56 ± 0.05, and 0.67 ± 0.04, respectively. Unlabeled panitumumab had no effect on SF, and irrelevant IgG-DOTA-111In only modestly reduced the SF of MDA-MB-231/Luc cells but not MCF7 or MDA-MB-468 cells. The cytotoxicity of panitumumab-DOTA-111In was mediated by increased DNA double-strand breaks (DSB), cell cycle arrest at G2/M-phase and apoptosis measured by immunofluorescence detection by flow cytometry. MDA-MB-231/Luc tumors in the mammary fat pad (MFP) of NRG mice were clearly imaged with panitumumab-DOTA-111In by microSPECT/CT at 4 days postinjection (p.i.), and biodistribution studies revealed high tumor uptake [18 ± 2% injected dose/g (% ID/g] and lower normal tissue uptake (<10% ID/g). Administration of up to 24 MBq (15 μg) of panitumumab-DOTA-111In to healthy NRG mice caused no major hematological, renal, or hepatic toxicity with no decrease in body weight. Treatment of NOD SCID mice with MDA-MB-231 tumors with panitumumab-DOTA-111In (22 MBq; 15 μg) slowed tumor growth. The mean time for tumors to reach a volume of ≥500 mm3 was 61 ± 5 days for RIT with panitumumab-DOTA-111In compared to 42 ± 6 days for mice treated with irrelevant IgG2-DOTA-111In (P < 0.0001) and 35 ± 3 days for mice receiving 0.9% NaCl (P < 0.0001). However, tumors regrew at later time points. The median survival of mice treated with panitumumab-DOTA-111In was 70 days versus 46 days for IgG2-DOTA-111In (P < 0.0001) or 40 days for 0.9% NaCl (P < 0.0001). We conclude that panitumumab-DOTA-111In is a promising theranostic agent for TNBC. Increasing the administered amount of panitumumab-DOTA-111In and/or combination with radiosensitizing PARP inhibitors used for treatment of patients with TNBC may provide a more durable response to RIT.
Collapse
Affiliation(s)
- Valerie J Facca
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Nakita E K Gopal
- Faculty of Arts and Sciences, University of Toronto, Toronto, Ontario M5S 3G3, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 3M2, Canada.,Department of Medical Imaging, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Joint Department of Medical Imaging and Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| |
Collapse
|
12
|
Yang X, Sun Y, Liu X, Jiang Z. A risk model of 10 aging-related genes for predicting survival and immune response in triple-negative breast cancer. Cancer Med 2022; 11:3182-3193. [PMID: 35297220 PMCID: PMC9385588 DOI: 10.1002/cam4.4674] [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: 11/03/2021] [Revised: 01/05/2022] [Accepted: 02/08/2022] [Indexed: 11/23/2022] Open
Abstract
Accumulated studies showed that the clinical significance of aging on the development and malignancy of tumors, while the relationship between aging and the prognosis, immune response in triple‐negative breast cancer (TNBC) has not been well clarified. Here, we constructed a risk model of 10 prognostic aging‐related genes (ARGs) from METABRIC database. Then, TNBC patients were classified into high‐ and low‐risk groups, the survival diversity, immune response, genomic function, and tumor mutation burden (TMB) between different risk groups were explored in METABRIC, TCGA, and GSE58812 cohorts. Results showed that patients in the high‐risk group had poorer survival outcomes compared to their counterparts (all p < 0.05), and the nomogram we established showed reliable prediction ability for survival in TNBC patients. Besides, TNBC patients with high‐risk scores had a lower expression of immune checkpoint markers and a lower fraction of activated immune cells. Furthermore, GSEA showed that Notch signaling pathway was significantly enriched in the high‐risk group. Thus, a risk model based on the aging‐related genes was developed and validated in this study, which may serve as a potential biomarker for prognosis and personalized treatment in TNBCs.
Collapse
Affiliation(s)
- Xia Yang
- Department of pathology, Sir Run Run Shaw hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Sun
- Department of Pathology, first affiliated hospital of Shenzhen University, Shenzhen, China
| | - Xia Liu
- Department of Pathology, first affiliated hospital of Shenzhen University, Shenzhen, China
| | - Zhinong Jiang
- Department of pathology, Sir Run Run Shaw hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Gialeli C, Tuysuz EC, Staaf J, Guleed S, Paciorek V, Mörgelin M, Papadakos KS, Blom AM. Complement inhibitor CSMD1 modulates epidermal growth factor receptor oncogenic signaling and sensitizes breast cancer cells to chemotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:258. [PMID: 34404439 PMCID: PMC8371905 DOI: 10.1186/s13046-021-02042-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Human CUB and Sushi multiple domains 1 (CSMD1) is a large membrane-bound tumor suppressor in breast cancer. The current study aimed to elucidate the molecular mechanism underlying the effect of CSMD1 in highly invasive triple negative breast cancer (TNBC). METHODS We examined the antitumor action of CSMD1 in three TNBC cell lines overexpressing CSMD1, MDA-MB-231, BT-20 and MDA-MB-486, in vitro using scanning electron microscopy, proteome array, qRT-PCR, immunoblotting, proximity ligation assay, ELISA, co-immunoprecipitation, immunofluorescence, tumorsphere formation assays and flow cytometric analysis. The mRNA expression pattern and clinical relevance of CSMD1 were evaluated in 3520 breast cancers from a modern population-based cohort. RESULTS CSMD1-expressing cells had distinct morphology, with reduced deposition of extracellular matrix components. We found altered expression of several cancer-related molecules, as well as diminished expression of signaling receptors including Epidermal Growth Factor Receptor (EGFR), in CSMD1-expressing cells compared to control cells. A direct interaction of CSMD1 and EGFR was identified, with the EGF-EGFR induced signaling cascade impeded in the presence of CSMD1. Accordingly, we detected increased ubiquitination levels of EGFR upon activation in CSMD1-expressing cells, as well as increased degradation kinetics and chemosensitivity. Accordingly, CSMD1 expression rendered tumorspheres pretreated with gefitinib more sensitive to chemotherapy. In addition, higher mRNA levels of CSMD1 tend to be associated with better outcome of triple negative breast cancer patients treated with chemotherapy. CONCLUSIONS Our results indicate that CSMD1 cross-talks with the EGFR endosomal trafficking cascade in a way that renders highly invasive breast cancer cells sensitive to chemotherapy. Our study unravels one possible underlying molecular mechanism of CSMD1 tumor suppressor function and may provide novel avenues for design of better treatment.
Collapse
Affiliation(s)
- Chrysostomi Gialeli
- Department of Translational Medicine, Lund University, Malmö, Sweden.,Experimental Cardiovascular Research Group, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Emre Can Tuysuz
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Lund, Sweden
| | - Safia Guleed
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Veronika Paciorek
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | | | | | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden.
| |
Collapse
|
14
|
Chen W, Ru R, Wang F, Li M. Automated breast volume scanning combined with shear wave elastography for diagnosis of triple-negative breast cancer and human epidermal growth factor receptor 2-positive breast cancer. Rev Assoc Med Bras (1992) 2021; 67:1167-1171. [PMID: 34669864 DOI: 10.1590/1806-9282.20210586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To explore the values of automated breast volume scanning (ABVS) combined with shear wave elastography (SWE) in the differential diagnosis of triple-negative breast cancer (TNBC) and human epidermal growth factor receptor 2-positive breast cancers (HER2+BC). METHODS In this study, 28 patients with TNBC and 32 patients with HER2+BC were enrolled. The characteristics of ABVS and virtual touch quantification (VTQ) in SWE of all patients were reviewed. The multivariate logistic regression analysis was carried out and the receiver operating characteristic curves of ABVS and ABVS+VTQ were drawn. RESULTS In ABVS imaging, the microcalcification, posterior echo, internal echo, shape, and edge had significant difference between TNBC and HER2+BC groups (p<0.05). The regular shape was the independent factor for TNBC (p=0.04, odds ratio [OR]=4.479), and the microcalcification in mass was the independent factor for HER2+BC (p=0.01, OR=2.997). In VTQ imaging, the shear wave velocity (SWV)max, SWVmin, and SWVmean in TNBC group were significantly lower than those in HER2+BC group (p<0.001). The sensitivity, specificity, and accuracy of ABVS+VTQ in diagnosing TNBC were higher than those of ABVS alone. CONCLUSIONS ABVS combined with SWE has certain advantages in differentiating TNBC from HER2+BC, which is helpful for the treatment planning and prognosis judgment.
Collapse
Affiliation(s)
- Weiping Chen
- Hangzhou Normal University, Xiaoshan Hospital, Department of Ultrasound - Hangzhou, China
| | - Rongrong Ru
- Hangzhou Normal University, Xiaoshan Hospital, Department of Ultrasound - Hangzhou, China
| | - Fang Wang
- Hangzhou Normal University, Xiaoshan Hospital, Department of Ultrasound - Hangzhou, China
| | - Mingkui Li
- Hangzhou Normal University, Xiaoshan Hospital, Department of Ultrasound - Hangzhou, China
| |
Collapse
|
15
|
Li Y, Lu W, Yang J, Edwards M, Jiang S. Survivin as a biological biomarker for diagnosis and therapy. Expert Opin Biol Ther 2021; 21:1429-1441. [PMID: 33877952 DOI: 10.1080/14712598.2021.1918672] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Survivin (SVN) is a member of the inhibitor of apoptosis (IAP) protein family that promotes cellular proliferation and inhibits apoptosis. Overexpression of SVN is associated with autoimmune disease, hyperplasia, and tumors and can be used as a biomarker in these diseases. SVN is widely recognized as a tumor-associated antigen (TAA) and has become an important target for cancer diagnosis and treatment.Areas covered: We reviewed SVN research progress from the PubMed and clinical trials focused on SVN from https://clinicaltrials.gov since 2000 and anticipate future developments in the field. The trials reviewed cover various modalities including diagnostics for early detection and disease progression, small molecule inhibitors of the SVN pathway and immunotherapy targeting SVN epitopes.Expert opinion: The most promising developments involve anti-SVN immunotherapy, with several therapeutic SVN vaccines under evaluation in phase I/II trials. SVN is an important new immune-oncology target that expands the repertoire of individualized combination treatments for cancer.
Collapse
Affiliation(s)
- Yuming Li
- Department of Oncology, University of Oxford, Oxford, UK.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Wenshu Lu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Jiarun Yang
- Department of Oncology, University of Oxford, Oxford, UK
| | - Mark Edwards
- Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| | - Shisong Jiang
- Department of Oncology, University of Oxford, Oxford, UK.,Department of Research and Development, Oxford Vacmedix UK Ltd, Oxford, UK
| |
Collapse
|
16
|
Differential reprogramming of breast cancer subtypes in 3D cultures and implications for sensitivity to targeted therapy. Sci Rep 2021; 11:7259. [PMID: 33790333 PMCID: PMC8012355 DOI: 10.1038/s41598-021-86664-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Screening for effective candidate drugs for breast cancer has shifted from two-dimensional (2D) to three-dimensional (3D) cultures. Here we systematically compared the transcriptomes of these different culture conditions by RNAseq of 14 BC cell lines cultured in both 2D and 3D conditions. All 3D BC cell cultures demonstrated increased mitochondrial metabolism and downregulated cell cycle programs. Luminal BC cells in 3D demonstrated overall limited reprogramming. 3D basal B BC cells showed increased expression of extracellular matrix (ECM) interaction genes, which coincides with an invasive phenotype not observed in other BC cells. Genes downregulated in 3D were associated with metastatic disease progression in BC patients, including cyclin dependent kinases and aurora kinases. Furthermore, the overall correlation of the cell line transcriptome to the BC patient transcriptome was increased in 3D cultures for all TNBC cell lines. To define the most optimal culture conditions to study the oncogenic pathway of interest, an open source bioinformatics strategy was established.
Collapse
|
17
|
Liao M, Zhang J, Wang G, Wang L, Liu J, Ouyang L, Liu B. Small-Molecule Drug Discovery in Triple Negative Breast Cancer: Current Situation and Future Directions. J Med Chem 2021; 64:2382-2418. [PMID: 33650861 DOI: 10.1021/acs.jmedchem.0c01180] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, but an effective targeted therapy has not been well-established so far. Considering the lack of effective targets, where do we go next in the current TNBC drug development? A promising intervention for TNBC might lie in de novo small-molecule drugs that precisely target different molecular characteristics of TNBC. However, an ideal single-target drug discovery still faces a huge challenge. Alternatively, other new emerging strategies, such as dual-target drug, drug repurposing, and combination strategies, may provide new insight into the improvement of TNBC therapeutics. In this review, we focus on summarizing the current situation of a series of candidate small-molecule drugs in TNBC therapy, including single-target drugs, dual-target drugs, as well as drug repurposing and combination strategies that will together shed new light on the future directions targeting TNBC vulnerabilities with small-molecule drugs for future therapeutic purposes.
Collapse
Affiliation(s)
- Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Leiming Wang
- The Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Jie Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Zhu Y, Li K, Zhang J, Wang L, Sheng L, Yan L. The prognostic and predictive significance of cytokeratin 5/6 and epidermal growth factor receptor in metastatic triple-negative breast cancer treated with maintenance capecitabine. Transl Cancer Res 2021; 10:1193-1203. [PMID: 35116447 PMCID: PMC8798347 DOI: 10.21037/tcr-20-1760] [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: 04/03/2020] [Accepted: 01/27/2021] [Indexed: 11/17/2022]
Abstract
Background Capecitabine is the most widely used agent for maintenance chemotherapy in patients with metastatic triple-negative breast cancer (mTNBC). However, there are no biomarkers for identifying mTNBC patients who could benefit from capecitabine maintenance. Methods The prognostic roles of cytokeratin 5/6 (CK5/6), epidermal growth factor receptor (EGFR), and maintenance therapy were evaluated in mTNBC patients. Both CK5/6 and EGFR were detected using immunohistochemistry. Of 115 patients who achieved disease control, 56 received capecitabine maintenance therapy and 59 underwent observation. The progression-free survival (PFS) and overall survival (OS) of the patients were evaluated. Results The median PFS and OS were longer in the maintenance group than that in the observation group (7.3 versus 5.7 months, P=0.0016; 22.4 versus 17.9 months, P=0.0055). Patients with basal-like TNBC had a poorer survival times than in those with non-basal-like TNBC (P=0.0062). Capecitabine maintenance significantly prolonged the OS of non-basal-like TNBC patients (P=0.0257), while in the basal-like TNBC patients, the difference was not significant (P=0.0541). Multivariate analysis revealed that the prolonged OS was related to age >50 years (P=0.005), presence of visceral metastases (P=0.035), response to initial therapy (P=0.017), maintenance therapy (P=0.033), and CK5/6 and EGFR status (P=0.032). Compared with the observation group, toxicities of all grades were more frequently observed in the maintenance group, including neutropenia, 85.71% vs. 25.87%, P<0.001; thrombocytopenia, 55.36% vs. 11.86%, P<0.001; anemia, 82.14% vs. 52.54%, P= 0.001; nausea 83.47% vs. 11.86%, P<0.001; vomiting 69.64% vs. 8.47%, P<0.001; and hand-foot syndrome (HFS) 32.14% vs. 1.69%, P<0.001. Conclusions Our study revealed that patients with non-basal-like TNBC had a better clinical outcome than those with basal-like TNBC, and capecitabine maintenance treatment significantly prolonged PFS and OS in patients with TNBC. Patients with non-basal-like TNBC could benefit from maintenance therapy with capecitabine and CK5/6 and EGFR are biomarkers for TNBC prognosis.
Collapse
Affiliation(s)
- Yiping Zhu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Kai Li
- Wannan Medical College, Wuhu, China
| | | | - Lu Wang
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Lili Sheng
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | | |
Collapse
|
19
|
Wang Z, Liu L, Li Y, Song Z, Jing Y, Fan Z, Zhang S. Analysis of CK5/6 and EGFR and Its Effect on Prognosis of Triple Negative Breast Cancer. Front Oncol 2021; 10:575317. [PMID: 33552956 PMCID: PMC7855982 DOI: 10.3389/fonc.2020.575317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is considered to be higher grade, more aggressive and have a poorer prognosis than other types of breast cancer. Discover biomarkers in TNBC for risk stratification and treatments that improve prognosis are in dire need. Methods Clinical data of 195 patients with triple negative breast cancer confirmed by pathological examination and received neoadjuvant chemotherapy (NAC) were collected. The expression levels of EGFR and CK5/6 were measured before and after NAC, and the relationship between EGFR and CK5/6 expression and its effect on prognosis of chemotherapy was analyzed. Results The overall response rate (ORR) was 86.2% and the pathological complete remission rate (pCR) was 29.2%. Univariate and multivariate logistic regression analysis showed that cT (clinical Tumor stages) stage was an independent factor affecting chemotherapy outcome. Multivariate Cox regression analysis showed pCR, chemotherapy effect, ypT, ypN, histological grades, and post- NAC expression of CK5/6 significantly affected prognosis. The prognosis of CK5/6-positive patients after NAC was worse than that of CK5/6-negative patients (p=0.036). Changes in CK5/6 and EGFR expression did not significantly affect the effect of chemotherapy, but changes from positive to negative expression of these two markers are associated with a tendency to improve prognosis. Conclusion For late-stage triple negative breast cancer patients receiving NAC, patients who achieved pCR had a better prognosis than those with non- pCR. Patients with the change in expression of EGFR and CK5/6 from positive to negative after neoadjuvant chemotherapy predicted a better prognosis than the change from negative to positive group.
Collapse
Affiliation(s)
- Zhen Wang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ying Li
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zi'an Song
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yi Jing
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ziyu Fan
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Sheng Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
20
|
Juliá EP, Mordoh J, Levy EM. Cetuximab and IL-15 Promote NK and Dendritic Cell Activation In Vitro in Triple Negative Breast Cancer. Cells 2020; 9:cells9071573. [PMID: 32605193 PMCID: PMC7408037 DOI: 10.3390/cells9071573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Triple Negative Breast Cancer (TNBC) treatment is still challenging, and immunotherapy is a potential approach in this tumor subtype. Cetuximab is an IgG1 monoclonal antibody (mAb) directed against Epidermic Growth Factor Receptor (EGFR), a protein overexpressed in a subgroup of TNBC patients and associated with poor prognosis. Previously, we demonstrated in vitro that Cetuximab triggers Ab-dependent cell cytotoxicity against TNBC cells. In this study, using co-cultures including TNBC cells, and NK and Dendritic Cells (DCs) from healthy donors, we studied the effect of Cetuximab-activated NK cells on DC function. Given that we already demonstrated that TNBC has an immunosuppressive effect on NK cells, we also tested Cetuximab combination with IL-15. We determined that Cetuximab opsonization of TNBC cells increased IFN-γ and TNF-α production by NK cells co-cultured with DCs. Moreover, we showed that NK cells activated by TNBC cells opsonized with Cetuximab promoted tumor material uptake and maturation of DCs, as well as their ability to produce IL-12. Furthermore, the stimulation with IL-15 increased the activation of NK cells and the maturation of DCs. These results suggest that IL-15 may enhance the efficacy of Cetuximab in the treatment of TNBC by promoting activation of both NK cells and DCs.
Collapse
Affiliation(s)
- Estefanía Paula Juliá
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (E.P.J.); (J.M.)
| | - José Mordoh
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (E.P.J.); (J.M.)
- Fundación Instituto Leloir, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
- Instituto Alexander Fleming, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina
| | - Estrella Mariel Levy
- Centro de Investigaciones Oncológicas CIO-FUCA, Ciudad Autónoma de Buenos Aires C1426AOE, Argentina; (E.P.J.); (J.M.)
- Correspondence: ; Tel.: +54-11-3221-8900
| |
Collapse
|
21
|
Purrington KS, Knight J, Dyson G, Ali-Fehmi R, Schwartz AG, Boerner JL, Bandyopadhyay S. CLCA2 expression is associated with survival among African American women with triple negative breast cancer. PLoS One 2020; 15:e0231712. [PMID: 32298355 PMCID: PMC7161959 DOI: 10.1371/journal.pone.0231712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/30/2020] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Black/African American (AA) women are twice as likely to be diagnosed with triple negative breast cancer (TNBC) compared to whites, an aggressive breast cancer subtype associated with poor prognosis. There are no routinely used targeted clinical therapies for TNBC; thus there is a clear need to identify prognostic markers and potential therapeutic targets. METHODS We evaluated expression of 27,016 genes in 155 treatment-naïve TN tumors from AA women in Detroit. Associations with survival were evaluated using Cox proportional hazards models adjusting for stage and age at diagnosis, and p-values were corrected using a false discovery rate. Our validation sample consisted of 494 TN tumors using four publically available data sets. Meta-analyses were performed using summary statistics from the four validation results. RESULTS In the Detroit AA cohort, CLCA2 [Hazard ratio (HR) = 1.56, 95% confidence interval (CI) 1.31-1.86, nominal p = 5.1x10-7, FDR p = 0.014], SPIC [HR = 1.47, 95%CI 1.26-1.73, nominal p = 1.8x10-6, FDR p = 0.022], and MIR4311 [HR = 1.57, 95% CI 1.31-1.92, nominal p = 2.5x10-5, FDR p = 0.022] expression were associated with overall survival. Further adjustment for treatment and breast cancer specific survival analysis did not substantially alter effect estimates. CLCA2 was also associated with increased risk of death in the validation cohorts [HR = 1.14, 95% CI 1.05-1.24, p = 0.038, p-heterogeneity = 0.88]. CONCLUSIONS We identified CLCA2 as a potential prognostic marker for TNBC in AA women.
Collapse
Affiliation(s)
- Kristen S. Purrington
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Jimmie Knight
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Gregory Dyson
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Ann G. Schwartz
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Julie L. Boerner
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| | - Sudeshna Bandyopadhyay
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, United States of America
| |
Collapse
|
22
|
Sultana R, Kataki AC, Barthakur BB, Sarma A, Bose S. Clinicopathological and immunohistochemical characteristics of breast cancer patients from Northeast India with special reference to triple negative breast cancer: A prospective study. Curr Probl Cancer 2020; 44:100556. [PMID: 32044043 DOI: 10.1016/j.currproblcancer.2020.100556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/11/2020] [Accepted: 01/17/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Molecular pathogenesis of Triple-negative breast cancer (TNBC) is inconclusively documented from resource limited countries and hence there is a lack of available targeted therapy for clinical interventions. Compared to other breast cancer subtypes, TNBC is more aggressive, higher recurrence rate, and higher prevalence in younger premenopausal women. Sporadic literature indicates predominance of TNBC in all reported breast cancer cases from Northeast India. AIM This study was conducted to evaluate the candidature of panel of key molecular markers involved in the development and progression of TNBC for prognosis and futuristic tailored targeted therapy. MATERIALS AND METHODS We analyzed the clinicopathological characterized and immunohistochemically screened the differential expression of key molecular markers involved in the development and progression of in TNBC cases vis-a-vis non-TNBC and autopsy-based control samples. RESULTS TNBC tends to display at an early reproductive age and is more aggressive in nature. Further, the differential expression of 2 specific markers viz., epidermal growth factor receptor (EGFR) and FolR1 was higher in TNBC cases compared to controls and non-TNBC (both in terms of susceptibility and specificity), clinical staging in TNBC cases (severity) and mortality (outcome). Although Ki67 and vascular endothelial growth factor expression also correlated with severity and outcome of the disease but their differences in non-TNBC cases were not significantly differentiable compared to TNBC. CONCLUSIONS The study indicates that EGFR and FolR1 could serve as useful biomarkers to determine TNBC prognosis. Further studies will be needed to evaluate EGFR and Folate pathways in order to screen out the molecular targets which may be meaningfully used for clinical stratification, intervention, and treatment.
Collapse
MESH Headings
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/epidemiology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/epidemiology
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- ErbB Receptors/metabolism
- Female
- Folate Receptor 1/metabolism
- Follow-Up Studies
- Humans
- India/epidemiology
- Middle Aged
- Prognosis
- Prospective Studies
- Survival Rate
- Triple Negative Breast Neoplasms/epidemiology
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/surgery
Collapse
Affiliation(s)
- Rizwana Sultana
- Department of Bioengineering and Technology, Gauhati University, Guwahati, Assam, India; Multidisciplinary Research Unit, Department of Health Research, ICMR, Fakhruddin Ali Ahmed Medical College, Barpeta, Assam, India
| | - Amal Ch Kataki
- Department of Gynecologic Oncology, Dr B. Borooah Cancer Institute, Guwahati, Assam, India
| | | | - Anupam Sarma
- Department of Pathology, Dr B. Borooah Cancer Institute, Guwahati, Assam, India
| | - Sujoy Bose
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India.
| |
Collapse
|
23
|
Nedeljković M, Damjanović A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge. Cells 2019; 8:E957. [PMID: 31443516 PMCID: PMC6770896 DOI: 10.3390/cells8090957] [Citation(s) in RCA: 509] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Triple-negative (TNBC) is the most lethal subtype of breast cancer owing to high heterogeneity, aggressive nature, and lack of treatment options. Chemotherapy remains the standard of care for TNBC treatment, but unfortunately, patients frequently develop resistance. Accordingly, in recent years, tremendous effort has been made into elucidating the mechanisms of TNBC chemoresistance with the goal of identifying new molecular targets. It has become evident that the development of TNBC chemoresistance is multifaceted and based on the elaborate interplay of the tumor microenvironment, drug efflux, cancer stem cells, and bulk tumor cells. Alterations of multiple signaling pathways govern these interactions. Moreover, TNBC's high heterogeneity, highlighted in the existence of several molecular signatures, presents a significant obstacle to successful treatment. In the present, in-depth review, we explore the contribution of key mechanisms to TNBC chemoresistance as well as emerging strategies to overcome them. We discuss novel anti-tumor agents that target the components of these mechanisms and pay special attention to their current clinical development while emphasizing the challenges still ahead of successful TNBC management. The evidence presented in this review outlines the role of crucial pathways in TNBC survival following chemotherapy treatment and highlights the importance of using combinatorial drug strategies and incorporating biomarkers in clinical studies.
Collapse
Affiliation(s)
- Milica Nedeljković
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia.
| | - Ana Damjanović
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
24
|
Huang Q, Li S, Zhang L, Qiao X, Zhang Y, Zhao X, Xiao G, Li Z. CAPE- pNO 2 Inhibited the Growth and Metastasis of Triple-Negative Breast Cancer via the EGFR/STAT3/Akt/E-Cadherin Signaling Pathway. Front Oncol 2019; 9:461. [PMID: 31214503 PMCID: PMC6558049 DOI: 10.3389/fonc.2019.00461] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
Overexpressed epidermal growth factor receptor (EGFR) and overactivated epithelial-mesenchymal transition (EMT) in triple-negative breast cancer (TNBC) can enhance tumorigenesis and tumor recurrence and metastasis. Caffeic acid p-nitro-phenethyl ester (CAPE-pNO2) has various pharmacological activities in our previous research, but its effect on metastasis and growth of TNBC has not been studied. In this study, Caffeic acid phenethyl ester (CAPE) was as a positive control. in vitro, MTT, Transwell, wound healing, colony formation and cell adhesion assays were performed to examine the effect on viability, invasion, migration, colony formation and adhesion of MDA-MB-231 cells by CAPE-pNO2, the results indicated that CAPE-pNO2 significantly dose-dependently inhibited metastasis of MDA-MB-231 cells (p < 0.05). in vivo, TNBC xenograft mice were established by subcutaneously injected with MDA-MB-231 cells, and they were used to estimate the effect on metastasis and growth of CAPE-pNO2 after 38 days of treatment. HE staining and TUNEL staining were carried out in tumor tissues, results showed that CAPE-pNO2 obviously suppressed the tumor growth, induced cells apoptosis (p < 0.01) and decreased pulmonary and splenic metastatic tumor cells. The results of IHC demonstrated that the VEGFA and Ki-67 proteins expression were downregulated (p < 0.01) in tumor tissues. Furthermore, western blot analysis was used to quantify key metastasis- and growth-associated proteins expression in vitro and in vivo, the results suggested that CAPE-pNO2 downregulated the proteins expression of p-EGFR, p-STAT3, p-Akt, MMP-2, MMP-9, Survivin, and key EMT-related proteins (Vimentin and N-cadherin) (p < 0.01), and increased the expression of E-cadherin (p < 0.01) in vivo and in vitro. Besides, CAPE-pNO2 had a similar effect as erlotinib in regulating the EGFR downstream proteins in EGF-induced MDA-MB-231cells. Collectively, these results indicated that CAPE-pNO2 possessed inhibitory effect on the growth and metastasis of TNBC may via the EGFR/STAT3/Akt/E-cadherin signaling pathway, and CAPE-pNO2 is better than CAPE in inhibiting growth and metastasis.
Collapse
Affiliation(s)
- Qin Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Sai Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Liwen Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xufang Qiao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Yanyan Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xiaoyan Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Guojun Xiao
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Zhubo Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| |
Collapse
|
25
|
Shi W, Xu X, Huang R, Yu Q, Zhang P, Xie S, Zheng H, Lu R. Plasma C-MYC level manifesting as an indicator in progression of breast cancer. Biomark Med 2019; 13:917-929. [PMID: 31144531 DOI: 10.2217/bmm-2019-0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aim: To investigate whether plasma C-MYC level could be an indicator in clinical progression of breast cancer. Materials & methods: Plasma level of C-MYC expression was detected by quantitative real time PCR and the level of c-myc protein in breast cancer tissues was detected by immunohistochemistry. The expression level of C-MYC mRNA in supernatant of cancer cells culture was measured compared with the nonbreast cancer cells. Results: Plasma C-MYC level was significantly higher in patients with breast cancer than that in the controls, which associated with clinical stages, lymph node status, etc. Receiver operating characteristic curve analysis showed the sensitivity and specificity of plasma C-MYC level for diagnosis of breast cancer were 63.6 and 81.8%, respectively. The expression of c-myc protein in breast cancer tissues was associated with plasma C-MYC level, even C-MYC level in supernatant of cancer cells was elevated. Conclusion: Plasma C-MYC level might be a potential indicator in progression of breast cancer.
Collapse
Affiliation(s)
- Weizhong Shi
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Clinical Laboratory, Shanghai Proton & Heavy Ion Center, Shanghai, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Xiaofeng Xu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Clinical Laboratory, Shanghai Proton & Heavy Ion Center, Shanghai, PR China
| | - Ren Huang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Clinical Laboratory, Shanghai Proton & Heavy Ion Center, Shanghai, PR China
| | - Qi Yu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Clinical Laboratory, Shanghai Proton & Heavy Ion Center, Shanghai, PR China
| | - Peiru Zhang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Clinical Laboratory, Shanghai Proton & Heavy Ion Center, Shanghai, PR China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Hui Zheng
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| |
Collapse
|
26
|
Li A, Zhang Y, Wang Z, Dong H, Fu N, Han X. The roles and signaling pathways of prolyl-4-hydroxylase 2 in the tumor microenvironment. Chem Biol Interact 2019; 303:40-49. [PMID: 30817904 DOI: 10.1016/j.cbi.2019.02.019] [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] [Received: 11/21/2018] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 02/06/2023]
Abstract
Tumor hypoxia is a well-known microenvironmental factor that causes cancer progression and resistance to cancer treatment. Proline hydroxylases (PHDs), a small protein family, belong to an evolutionarily conserved superfamily of dioxygenases, considered the central regulator of the molecular hypoxia response. Prolyl-4-hydroxylase 2 (PHD2), one member of PHDs family, regulates the stability of the hypoxia-inducible factor-1 alpha (HIF-1α) in response to oxygen availability. During hypoxia, the inhibition of PHD2 permits the accumulation of HIF-1α, allowing the cellular adaptation to oxygen limitation, causing activation of numerous genes, which enhances the angiogenesis, metastasis and invasiveness. Accurate regulation of oxygen homeostasis is essential, and which implies PHD2 may have a regulatory role in the pathogenesis of cancer. Although ample evidence exists for a positive correlation between HIFs and tumor formation, metastasis and poor prognosis, the function of the PHD2 in carcinogenesis is less well understood. Despite their original role as the oxygen sensors of the cell and many of the its functions are clearly conveyed through the HIF system, PHD2 is currently known to display HIF-independent and hydroxylase-independent functions in cancer cells and stroma in the control of different cellular pathways. In this review, we summarize the recent advances in the structure, regulation and functions of PHD2 in cancer microenvironment.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Zuojun Wang
- Department of Pharmacy, Linqu Country People's Hospital, 438 Shanwang Road, Linqu, 262600, China
| | - Hailing Dong
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Nange Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, China.
| |
Collapse
|
27
|
Noonan MM, Dragan M, Mehta MM, Hess DA, Brackstone M, Tuck AB, Viswakarma N, Rana A, Babwah AV, Wondisford FE, Bhattacharya M. The matrix protein Fibulin-3 promotes KISS1R induced triple negative breast cancer cell invasion. Oncotarget 2018; 9:30034-30052. [PMID: 30046386 PMCID: PMC6059025 DOI: 10.18632/oncotarget.25682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 06/13/2018] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of cancer mortality. In particular, triple negative breast cancer (TNBC) comprise a heterogeneous group of basal-like tumors lacking estrogen receptor (ERα), progesterone receptor (PR) and HER2 (ErbB2). TNBC represents 15-20% of all breast cancers and occurs frequently in women under 50 years of age. Unfortunately, these patients lack targeted therapy, are typically high grade and metastatic at time of diagnosis. The mechanisms regulating metastasis remain poorly understood. We have previously shown that the kisspeptin receptor, KISS1R stimulates invasiveness of TNBC cells. In this report, we demonstrate that KISS1R signals via the secreted extracellular matrix protein, fibulin-3, to regulate TNBC invasion. We found that the fibulin-3 gene is amplified in TNBC primary tumors and that plasma fibulin-3 levels are elevated in TNBC patients compared to healthy subjects. In this study, we show that KISS1R activation increases fibulin-3 expression and secretion. We show that fibulin-3 regulates TNBC metastasis in a mouse experimental metastasis xenograft model and signals downstream of KISS1R to stimulate TNBC invasion, by activating matrix metalloproteinase 9 (MMP-9) and the MAPK pathway. These results identify fibulin-3 as a new downstream mediator of KISS1R signaling and as a potential biomarker for TNBC progression and metastasis, thus revealing KISS1R and fibulin-3 as novel drug targets in TNBC.
Collapse
Affiliation(s)
- Michelle M Noonan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Magdalena Dragan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Michael M Mehta
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - David A Hess
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Krembil Centre for Stem Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, London, ON, Canada
| | - Muriel Brackstone
- Department of Oncology, The University of Western Ontario, London, ON, Canada.,Lawson Health Research Institute, The University of Western Ontario, London, ON, Canada.,Division of Surgical Oncology, The University of Western Ontario, London, ON, Canada
| | - Alan B Tuck
- Department of Oncology, The University of Western Ontario, London, ON, Canada.,Department of Pathology, The University of Western Ontario, London, ON, Canada.,The Pamela Greenaway-Kohlmeier Translational Breast Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA
| | - Andy V Babwah
- Department of Pediatrics, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Frederic E Wondisford
- Department of Medicine, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Moshmi Bhattacharya
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Department of Oncology, The University of Western Ontario, London, ON, Canada.,Lawson Health Research Institute, The University of Western Ontario, London, ON, Canada.,Department of Medicine, Child Health Institute of NJ, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
28
|
Mazzio EA, Lewis CA, Elhag R, Soliman KF. Effects of Sepantronium Bromide (YM-155) on the Whole Transcriptome of MDA-MB-231 Cells: Highlight on Impaired ATR/ATM Fanconi Anemia DNA Damage Response. Cancer Genomics Proteomics 2018; 15:249-264. [PMID: 29976630 PMCID: PMC6070710 DOI: 10.21873/cgp.20083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/16/2018] [Accepted: 05/25/2018] [Indexed: 12/18/2022] Open
Abstract
Sepantronium bromide (YM-155) is believed to elicit apoptosis and mitotic arrest in tumor cells by reducing (BIRC5, survivin) mRNA. In this study, we monitored changes in survivin mRNA and protein after treating MDA-MB-231 cells with YM-155 concurrent with evaluation of whole transcriptomic (WT) mRNA and long intergenic non-coding RNA at 2 time points: 8 h sub-lethal (83 ng/mL) and 20 h at the LC50 (14.6 ng/mL). The data show a tight association between cell death and the precipitating loss of survivin protein and mRNA (-2.67 fold-change (FC), p<0.001) at 20 h, questioning if the decline in survivin is attributed to cell death or drug impact. The meager loss of survivin mRNA was overshadowed by enormous differential change to the WT in both magnitude and significance for over 2000 differentially up/down-regulated transcripts: (+22 FC to -12 FC, p<0.001). The data show YM-155 to up-regulate transcripts in control of circadian rhythm (NOCT, PER, BHLHe40, NFIL3), tumor suppression (SIK1, FOSB), histone methylation (KDM6B) and negative feedback of NF-kappa B signaling (TNFAIP3). Down-regulated transcripts by YM-155 include glucuronidase (GUSBP3), numerous micro-RNAs, DNA damage repair elements (CENPI, POLQ, RAD54B) and the most affected system was the ataxia-telangiectasia mutated (ATM)/Fanconi anemia E3 monoubiquitin ligase core complexes (FANC transcripts - A/B/E/F/G/M), FANC2, FANCI, BRCA1, BRCA2, RAD51, PALB2 gene and ATR (ATM- and Rad3-Related) pathway. In conclusion, these findings suggest that a primary target of YM-155 is the loss of replicative DNA repair systems.
Collapse
Affiliation(s)
- Elizabeth A Mazzio
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Charles A Lewis
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Rashid Elhag
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A
| | - Karam F Soliman
- College of Pharmacy & Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, U.S.A.
| |
Collapse
|
29
|
Luchsinger C, Aguilar M, Burgos PV, Ehrenfeld P, Mardones GA. Functional disruption of the Golgi apparatus protein ARF1 sensitizes MDA-MB-231 breast cancer cells to the antitumor drugs Actinomycin D and Vinblastine through ERK and AKT signaling. PLoS One 2018; 13:e0195401. [PMID: 29614107 PMCID: PMC5882166 DOI: 10.1371/journal.pone.0195401] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/21/2018] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence indicates that the Golgi apparatus plays active roles in cancer, but a comprehensive understanding of its functions in the oncogenic transformation has not yet emerged. At the same time, the Golgi is becoming well recognized as a hub that integrates its functions of protein and lipid biosynthesis to signal transduction for cell proliferation and migration in cancer cells. Nevertheless, the active function of the Golgi apparatus in cancer cells has not been fully evaluated as a target for combined treatment. Here, we analyzed the effect of perturbing the Golgi apparatus on the sensitivity of the MDA-MB-231 breast cancer cell line to the drugs Actinomycin D and Vinblastine. We disrupted the function of ARF1, a protein necessary for the homeostasis of the Golgi apparatus. We found that the expression of the ARF1-Q71L mutant increased the sensitivity of MDA-MB-231 cells to both Actinomycin D and Vinblastine, resulting in decreased cell proliferation and cell migration, as well as in increased apoptosis. Likewise, the combined treatment of cells with Actinomycin D or Vinblastine and Brefeldin A or Golgicide A, two disrupting agents of the ARF1 function, resulted in similar effects on cell proliferation, cell migration and apoptosis. Interestingly, each combined treatment had distinct effects on ERK1/2 and AKT signaling, as indicated by the decreased levels of either phospho-ERK1/2 or phospho-AKT. Our results suggest that disruption of Golgi function could be used as a strategy for the sensitization of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Charlotte Luchsinger
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Marcelo Aguilar
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Patricia V. Burgos
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Ehrenfeld
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Department of Anatomy, Histology and Pathology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Gonzalo A. Mardones
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- * E-mail:
| |
Collapse
|
30
|
Binding of galectin-1 to integrin β1 potentiates drug resistance by promoting survivin expression in breast cancer cells. Oncotarget 2018; 8:35804-35823. [PMID: 28415760 PMCID: PMC5482619 DOI: 10.18632/oncotarget.16208] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/28/2017] [Indexed: 12/13/2022] Open
Abstract
Galectin-1 is a β-galactoside binding protein secreted by many types of aggressive cancer cells. Although many studies have focused on the role of galectin-1 in cancer progression, relatively little attention has been paid to galectin-1 as an extracellular therapeutic target. To elucidate the molecular mechanisms underlying galectin-1-mediated cancer progression, we established galectin-1 knock-down cells via retroviral delivery of short hairpin RNA (shRNA) against galectin-1 in two triple-negative breast cancer (TNBC) cell lines, MDA-MB-231 and Hs578T. Ablation of galectin-1 expression decreased cell proliferation, migration, invasion, and doxorubicin resistance. We found that these effects were caused by decreased galectin-1-integrin β1 interactions and suppression of the downstream focal adhesion kinase (FAK)/c-Src pathway. We also found that silencing of galectin-1 inhibited extracellular signal-regulated kinase (ERK)/signal transducer and activator of transcription 3 (STAT3) signaling, thereby down-regulating survivin expression. This finding implicates STAT3 as a transcription factor for survivin. Finally, rescue of endogenous galectin-1 knock-down and recombinant galectin-1 treatment both recovered signaling through the FAK/c-Src/ERK/STAT3/survivin pathway. Taken together, these results suggest that extracellular galectin-1 contributes to cancer progression and doxorubicin resistance in TNBC cells. These effects appear to be mediated by galectin-1-induced up-regulation of the integrin β1/FAK/c-Src/ERK/STAT3/survivin pathway. Our results imply that extracellular galectin-1 has potential as a therapeutic target for triple-negative breast cancer.
Collapse
|
31
|
Greenwood E, Maisel S, Ebertz D, Russ A, Pandey R, Schroeder J. Llgl1 prevents metaplastic survival driven by epidermal growth factor dependent migration. Oncotarget 2018; 7:60776-60792. [PMID: 27542214 PMCID: PMC5308616 DOI: 10.18632/oncotarget.11320] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/02/2016] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that Llgl1 loss results in a gain of mesenchymal phenotypes and a loss of apicobasal and planar polarity. We now demonstrate that these changes represent a fundamental shift in cellular phenotype. Llgl1 regulates the expression of multiple cell identity markers, including CD44, CD49f, and CD24, and the nuclear translocation of TAZ and Slug. Cells lacking Llgl1 form mammospheres, where survival and transplantability is dependent upon the Epidermal Growth Factor Receptor (EGFR). Additionally, Llgl1 loss allows cells to grow in soft-agar and maintain prolonged survival as orthotopic transplants in NOD-SCIDmice. Lineage tracing and wound healing experiments demonstrate that mammosphere survival is due to enhanced EGF-dependent migration. The loss of Llgl1 drives EGFR mislocalization and an EGFR mislocalization point mutation (P667A) drives these same phenotypes, including activation of AKT and TAZ nuclear translocation. Together, these data indicate that the loss of Llgl1 results in EGFR mislocalization, promoting pre-neoplastic changes.
Collapse
Affiliation(s)
- Erin Greenwood
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Sabrina Maisel
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| | - David Ebertz
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Atlantis Russ
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona
| | - Ritu Pandey
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Joyce Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Arizona Cancer Center, University of Arizona, Tucson, Arizona.,BIO5 Institute, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| |
Collapse
|
32
|
Hypoxia-inducible factor prolyl hydroxylase 2 (PHD2) is a direct regulator of epidermal growth factor receptor (EGFR) signaling in breast cancer. Oncotarget 2018; 8:9885-9898. [PMID: 28038470 PMCID: PMC5354778 DOI: 10.18632/oncotarget.14241] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/02/2016] [Indexed: 01/09/2023] Open
Abstract
Clinical studies in breast cancer suggest important associations between intratumoral hypoxia, the upregulation of epidermal growth factor receptor (EGFR or HER1), hypoxia-inducible factor 1α (HIF-1α), and reduced patient survival. However, direct molecular links between EGFR and the hypoxia signaling system are not yet established. Since the oxygen sensor hypoxia-inducible factor prolyl hydroxylase 2 (PHD2) is considered to be the main HIF-1α regulator, we hypothesized that PHD2 and EGFR may be interconnected at the molecular level. By analyzing samples from 313 breast cancer patients, we found that EGFR is a first clinicopathological parameter positively correlating with PHD2. Mechanistically, we identified PHD2 as a direct binding partner of EGFR and show that PHD2 regulates EGFR stability as well as its subsequent signaling in breast carcinoma cells. Overall, we introduce for the first time the direct crosstalk between the oxygen sensor PHD2 and EGFR-mediated tumorigenesis in breast cancer.
Collapse
|
33
|
Guo J, Gong G, Zhang B. miR-539 acts as a tumor suppressor by targeting epidermal growth factor receptor in breast cancer. Sci Rep 2018; 8:2073. [PMID: 29391441 PMCID: PMC5794864 DOI: 10.1038/s41598-018-20431-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/03/2017] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most frequently diagnosed malignancy and the leading cause of cancer-associated death in women worldwide. microRNAs (miRNAs) play critical roles in the cellular processes of breast cancer. However, the crucial roles and underlying mechanisms of miR-539 in breast cancer remain unclear. By RT-qPCR, we found that expression of miR-539 was markedly down-regulated in breast cancer tissues and cell lines compared with that in paired adjacent normal tissues and normal cell lines. The low level of miR-539 expression was positively associated with lymph node metastasis. Furthermore, forced expression of miR-539 inhibited proliferation and migration of breast cancer MDA-MB-231 and MCF7 cells in vitro and suppressed tumor growth in vivo. Moreover, bioinformatics analysis and luciferase reporter assays indicated that epidermal growth factor receptor (EGFR) was a direct target of miR-539. Over-expression of miR-539 decreased the EGFR mRNA and protein levels in MDA-MB-231 and MCF7 cells. In addition, ectopic over-expression of EGFR partly reversed miR-539-inhibited proliferation as well as migration of MDA-MB-231 and MCF7 cells. Taken together, our results demonstrate that miR-539 functions as a tumor suppressor in breast cancer by downregulating EGFR, supporting the targeting of the novel miR-539/EGFR axis as a potentially effective therapeutic approach for breast cancer.
Collapse
Affiliation(s)
- Jilong Guo
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, 028000, China.,Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, 028000, China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, 028000, China. .,Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, 028000, China. .,Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinaltreatment, Tongliao, Inner Mongolia, 028007, China.
| | - Bin Zhang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, 028000, China. .,Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, 028000, China. .,Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinaltreatment, Tongliao, Inner Mongolia, 028007, China.
| |
Collapse
|
34
|
Gonzalez-Conchas GA, Rodriguez-Romo L, Hernandez-Barajas D, Gonzalez-Guerrero JF, Rodriguez-Fernandez IA, Verdines-Perez A, Templeton AJ, Ocana A, Seruga B, Tannock IF, Amir E, Vera-Badillo FE. Epidermal growth factor receptor overexpression and outcomes in early breast cancer: A systematic review and a meta-analysis. Cancer Treat Rev 2018; 62:1-8. [DOI: 10.1016/j.ctrv.2017.10.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 01/09/2023]
|
35
|
Wang X, Beitler JJ, Huang W, Chen G, Qian G, Magliocca K, Patel MR, Chen AY, Zhang J, Nannapaneni S, Kim S, Chen Z, Deng X, Saba NF, Chen ZG, Arbiser JL, Shin DM. Honokiol Radiosensitizes Squamous Cell Carcinoma of the Head and Neck by Downregulation of Survivin. Clin Cancer Res 2017; 24:858-869. [PMID: 29180609 DOI: 10.1158/1078-0432.ccr-17-0345] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/16/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
Abstract
Purpose: Previous studies revealed diverging results regarding the role of survivin in squamous cell carcinoma of the head and neck (SCCHN). This study aimed to evaluate the clinical significance of survivin expression in SCCHN; the function of survivin in DNA-damage repair following ionizing radiation therapy (RT) in SCCHN cells; and the potential of honokiol to enhance RT through downregulation of survivin.Experimental Design: Expression of survivin in SCCHN patient primary tumor tissues (n = 100) was analyzed and correlated with clinical parameters. SCCHN cell lines were used to evaluate the function of survivin and the effects of honokiol on survivin expression in vitro and in vivoResults: Overexpression of survivin was significantly associated with lymph nodes' metastatic status (P = 0.025), worse overall survival (OS), and disease-free survival (DFS) in patients receiving RT (n = 65, OS: P = 0.024, DFS: P = 0.006) and in all patients with SCCHN (n = 100, OS: P = 0.002, DFS: P = 0.003). In SCCHN cells, depletion of survivin led to increased DNA damage and cell death following RT, whereas overexpression of survivin increased clonogenic survival. RT induced nuclear accumulation of survivin and its molecular interaction with γ-H2AX and DNA-PKCs. Survivin specifically bound to DNA DSB sites induced by I-SceI endonuclease. Honokiol (which downregulates survivin expression) in combination with RT significantly augmented cytotoxicity in SCCHN cells with acquired radioresistance and inhibited growth in SCCHN xenograft tumors.Conclusions: Survivin is a negative prognostic factor and is involved in DNA-damage repair induced by RT. Targeting survivin using honokiol in combination with RT may provide novel therapeutic opportunities. Clin Cancer Res; 24(4); 858-69. ©2017 AACR.
Collapse
Affiliation(s)
- Xu Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jonathan J Beitler
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Wen Huang
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Guo Chen
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Guoqing Qian
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Kelly Magliocca
- Department of Pathology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Mihir R Patel
- Department of Otolaryngology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Amy Y Chen
- Department of Otolaryngology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jun Zhang
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Sreenivas Nannapaneni
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Sungjin Kim
- Department of Biostatistics and Bioinformatics, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Zhengjia Chen
- Department of Biostatistics and Bioinformatics, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Xingming Deng
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Zhuo Georgia Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jack L Arbiser
- Department of Dermatology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.,Veterans Affairs Medical Center, Decatur, Georgia
| | - Dong M Shin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
36
|
Dai X, Cheng H, Bai Z, Li J. Breast Cancer Cell Line Classification and Its Relevance with Breast Tumor Subtyping. J Cancer 2017; 8:3131-3141. [PMID: 29158785 PMCID: PMC5665029 DOI: 10.7150/jca.18457] [Citation(s) in RCA: 712] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
Breast cancer cell lines have been widely used for breast cancer modelling which encompasses a panel of diseases with distinct phenotypical associations. Though cell lines provide unlimited homogenous materials for tumor studies and are relatively easy to culture, they are known to accumulate mutations duringthe initial establishment and subsequent series of cultivations. Thus, whether breast cancer cell line heterogeneity reflects that of carcinoma remains an important issue to resolve before drawing any reliable conclusion at the tumor level using cell lines. Inconsistent nomenclatures used for breast cancer cell line subtyping and the different number of subtypes grouped for cell lines and tumors make their direct matching elusive. By analyzing the molecular features of 92 breast cancer cell lines as documented by different literatures, we categorize 84 cell lines into 5 groups to be consistent with breast tumor classification. After combing through these cell lines, we summarized the molecular features, genetically and epigenetically, of each subtype, and manually documented 10 cell lines lacking explicit information on subtyping. Nine cell lines, either found inconsistent on their primary molecular features from different studies or being contaminated at the origin, are not suggested as the first choice for experimental use. We conclude that breast tumor cell lines, though having a high mutational frequency with many uncertainties and could not fully capture breast cancer heterogeneity, are feasible but crude models for tumors of the same subtype. New cell lines with enriched interferon regulated genes need to be established to enlarge the coverage of cell lines on tumor heterogeneity.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Hongye Cheng
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Zhonghu Bai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jia Li
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
37
|
Yue Y, Astvatsaturyan K, Cui X, Zhang X, Fraass B, Bose S. Stratification of Prognosis of Triple-Negative Breast Cancer Patients Using Combinatorial Biomarkers. PLoS One 2016; 11:e0149661. [PMID: 26930401 PMCID: PMC4773063 DOI: 10.1371/journal.pone.0149661] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/03/2016] [Indexed: 12/13/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is highly diverse group of cancers, and generally considered an aggressive disease associated with poor survival. Stratification of TNBC is highly desired for both prognosis and treatment decisions to identify patients who may benefit from less aggressive therapy. Methods This study retrieved 192 consecutive non-metastasis TNBC patients who had undergone a resection of a primary tumor from 2008 to 2012. All samples were negative for ER, PR, and HER2/neu. Disease-free-survival (DFS) and overall-survival (OS) were evaluated for expression of immunohistochemical biomarkers (P53, Ki-67, CK5/6 and EGFR), as well as clinicopathological variables including age, tumor size, grade, lymph node status, pathologic tumor and nodal stages. The cutoff values of the basal biomarkers, EGFR and CK5/6, were estimated by time-dependent ROC curves. The prognostic values of combinatorial variables were identified by univariate and multivariate Cox analysis. Patients were stratified into different risk groups based on expression status of identified prognostic variables. Results Median age was 57 years (range, 28–92 years). Patients’ tumor stage and nodal stage were significantly associated with OS and DFS. EGFR and CK5/6 were significant prognostic variables at cutoff points of 15% (p = 0.001, AUC = 0.723), and 50% (p = 0.006, AUC = 0.675), respectively. Multivariate Cox analysis identified five significant variables: EGFR (p = 0.016), CK5/6 (p = 0.018), Ki-67 (p = 0.048), tumor stage (p = 0.010), and nodal stage (p = 0.003). Patients were stratified into low basal (EGFR≤15% and CK5/6≤50%) and high basal (EGFR>15% and/or CK5/6>50%) expression groups. In the low basal expression group, patients with low expressions of Ki-67, low tumor and nodal stage had significantly better survival than those with high expressions/stages of three variables, log-rank p = 0.015 (100% vs 68% at 50 months). In the high basal expression group, patient with high basal expression of both biomarkers (EGFR >15% and CK5/6 >50%) had worse survival (mean DFS = 25 months, 41.7% event rate) than those patient with high expression of either one marker (mean DFS = 34 months, 25.5% event rate). Conclusions Immunoexpression of basal biomarkers, EGFR and CK5/6, is useful in predicting survival of TNBC patients. Integrated with Ki-67, tumor and nodal stages, combinatorial biomarker analysis provides a feasible clinical solution to stratify patient risks and help clinical decision-making with respect to selecting the appropriate therapies for individual patients.
Collapse
Affiliation(s)
- Yong Yue
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| | - Kristine Astvatsaturyan
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Xiaojiang Cui
- Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Xiao Zhang
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Benedick Fraass
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Shikha Bose
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
38
|
Wang T, Gantier MP, Xiang D, Bean AG, Bruce M, Zhou SF, Khasraw M, Ward A, Wang L, Wei MQ, AlShamaileh H, Chen L, She X, Lin J, Kong L, Shigdar S, Duan W. EpCAM Aptamer-mediated Survivin Silencing Sensitized Cancer Stem Cells to Doxorubicin in a Breast Cancer Model. Am J Cancer Res 2015; 5:1456-72. [PMID: 26681989 PMCID: PMC4672025 DOI: 10.7150/thno.11692] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 07/30/2015] [Indexed: 12/23/2022] Open
Abstract
Understanding the molecular basis of drug resistance and utilising this information to overcome chemoresistance remains a key challenge in oncology. Here we report that survivin, a key protein implicated in drug resistance, is overexpressed in cancer stem cell pool of doxorubicin-resistant breast cancer cells. Moreover, by utilising an active targeting system consisting of an RNA aptamer targeted against the epithelial cell adhesion molecule and a Dicer substrate survivin siRNA, we could deliver a high dose of the siRNA to cancer stem cells in xenograft tumours. Importantly, silencing of survivin with this aptamer-siRNA chimera in cancer stem cell population led to the reversal of chemoresistance, such that combined treatment with low dose of doxorubicin inhibited stemness, eliminated cancer stem cells via apoptosis, suppressed tumour growth, and prolonged survival in mice bearing chemoresistant tumours. This strategy for in vivo cancer stem cell targeting has wide application for future effective silencing of anti-death genes and in fact any dysregulated genes involved in chemoresistance and tumour relapse.
Collapse
|
39
|
Zheng HM, Chen C, Wu XH, Chen J, Sun S, Sun JZ, Wang MW, Sun SR. Quantum dot-based in situ simultaneous molecular imaging and quantitative analysis of EGFR and collagen IV and identification of their prognostic value in triple-negative breast cancer. Tumour Biol 2015; 37:2509-18. [PMID: 26385773 DOI: 10.1007/s13277-015-4079-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/13/2015] [Indexed: 12/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a unique breast cancer subtype with high heterogeneity and poor prognosis. Currently, the treatment effect of TNBC has reached a bottleneck, rendering new breakthroughs difficult. Cancer invasion is not an entirely cell-autonomous process, requiring the cells to transmigrate across the surrounding extracellular matrix (ECM) barriers. Developing a new system that integrates key constituents in the tumor microenvironment with pivotal cancer cell molecules is essential for the in-depth investigation of the mechanism of invasion in TNBC. We describe a computer-aided algorithm developed using quantum dot (QD)-based multiplex molecular imaging of TNBC tissues. We performed in situ simultaneous imaging and quantitative detection of epidermal growth factor receptor (EGFR), expressed in the TNBC cell membrane, and collagen IV, the major ECM constituent; calculated the EGFR/collagen IV ratio; and investigated the prognostic value of the EGFR/collagen IV ratio in TNBC. We simultaneously imaged and quantitatively detected EGFR and collagen IV in the TNBC samples. In all patients, quantitative determination showed a statistically significant negative correlation between EGFR and collagen IV. The 5-year disease-free survival (5-DFS) of the high and low EGFR/collagen IV ratio subgroups was significantly different. The EGFR/collagen IV ratio was predictive and was an independent prognostic indicator in TNBC. Compared with EGFR expression, the EGFR/collagen IV ratio had a greater prognostic value for 5-DFS. Our findings open up a new avenue for predicting the clinical outcome in TNBC from the perspective of integrating molecules expressed in both cancer cells and the ECM.
Collapse
Affiliation(s)
- Hong-Mei Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Xin-Hong Wu
- Department of Breast Surgery, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Jian Chen
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Jin-Zhong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Ming-Wei Wang
- Department of Pathology, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China.
| |
Collapse
|
40
|
Yue Y, Cui X, Bose S, Audeh W, Zhang X, Fraass B. Stratifying triple-negative breast cancer prognosis using 18F-FDG-PET/CT imaging. Breast Cancer Res Treat 2015; 153:607-16. [PMID: 26346756 PMCID: PMC4589560 DOI: 10.1007/s10549-015-3558-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022]
Abstract
This study aims to stratify prognosis of triple-negative breast cancer (TNBC) patients using pre-treatment 18F-FDG-PET/CT, alone and with correlation to immunohistochemistry biomarkers. 200 consecutive TNBC breast cancer patients treated between 2008 and 2012 were retrieved. Among the full cohort, 79 patients had pre-treatment 18F-FDG-PET/CT scans. Immunostaining status of basal biomarkers (EGFR, CK5/6) and other clinicopathological variables were obtained. Three PET image features were evaluated: maximum uptake values (SUVmax), mean uptake (SUVmean), and metabolic volume (SUVvol) defined by SUV > 2.5. All variables were analyzed versus disease-free survival (DFS) using univariate and multivariate Cox analysis, Kaplan-Meier curves, and log-rank tests. The optimal cutoff points of variables were estimated using time-dependent survival receiver operating characteristic (ROC) analysis. All PET features significantly correlated with proliferation marker Ki-67 (all p < 0.010). SUVmax stratified the prognosis of TNBC patients with optimal cutoff derived by ROC analysis (≤3.5 vs. >3.5, AUC = 0.654, p = 0.006). SUVmax and EGFR were significant prognostic factors in univariate and multivariate Cox analyses. To integrate prognosis of biological and imaging markers, patients were first stratified by EGFR into low (≤15 %) and high (>15 %) risk groups. Further, SUVmax was used as a variable to stratify the two EGFR groups. In the high EGFR group, patients with high FDG uptake (SUVmax > 3.5) had worse survival outcome (median DFS = 7.6 months) than those patients with low FDG uptake (SUVmax ≤ 3.5, median DFS = 11.6 months). In the low EGFR group, high SUVmax also indicated worse survival outcome (17.2 months) than low SUVmax (22.8 months). The risk stratification with integrative EGFR and PET was statistically significant with log-rank p ≪ 0.001. Pre-treatment 18F-FDG-PET/CT imaging has significant prognostic value for predicting survival outcome of TNBC patients. Integrated with basal-biomarker EGFR, PET imaging can further stratify patient risks in the pre-treatment stage and help select appropriate treatment strategies for individual patients.
Collapse
Affiliation(s)
- Yong Yue
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| | - Xiaojiang Cui
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Shikha Bose
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - William Audeh
- Department of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Xiao Zhang
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Benedick Fraass
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| |
Collapse
|
41
|
Lin B, Gao A, Zhang R, Ma H, Shen H, Hu Q, Zhang H, Zhao M, Lan X, Liu K. Use of a Novel Integrase-Deficient Lentivirus for Targeted Anti-Cancer Therapy With Survivin Promoter-Driven Diphtheria Toxin A. Medicine (Baltimore) 2015; 94:e1301. [PMID: 26252309 PMCID: PMC4616595 DOI: 10.1097/md.0000000000001301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
As an immunotoxin, diphtheria toxin has been widely used in gene therapy and gene function assays for its roles in protein synthesis inhibition, and the aim of our study is to set up a nonintegrating lentiviral system for specific expression of diphtheria toxin A (DTA) used in cancer gene therapy.Here, we established a lentiviral system that could coordinately express fluorescent protein and DTA driven by the cytomegalovirus (CMV) promoter, which is convenient for us to precisely trace the expression of DTA and monitor the process of lentivirus packaging. To achieve safer cancer therapy, we replaced the CMV promoter with the Survivin promoter, a specific promoter that is dramatically activated in cancer tissues and cells, but not in normal tissues and cells, and that will impose greater therapeutic potential because a significant expression difference occurred between these 2 groups. Meanwhile, we obtained integrase-deficient lentivirus (IDLV) after packaging with the integrase mutant, which expresses defective integrase RRK262263264AAH, to minimize the side effects that derived from the insertional mutagenesis of the host genome.Our results suggest that the IDLV system that we generated possesses therapeutic potential in cancers in vitro and in vivo.
Collapse
Affiliation(s)
- Baoshun Lin
- From the Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian, P.R. China (BL, AG, RZ, HM, MZ, XL, KL); Dong Fang Hospital, Xiamen University, Fuzhou, Fujian, P.R. China (BL, AG, QH, MZ, XL, KL); Institute for Laboratory Medicine, Fuzhou General Hospital, Second Military Medical University, Fuzhou, Fujian, P.R. China (HM); School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, P.R. China (HS); Agriculture and Animal Husbandry College, Tibet University, Nyingchi, P.R. China (HZ); and Fuzhou General Hospital Clinical Medical School, Fujian Medical University, Fuzhou, P.R. China (RZ, XL, KL)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Robichaux JP, Hallett RM, Fuseler JW, Hassell JA, Ramsdell AF. Mammary glands exhibit molecular laterality and undergo left-right asymmetric ductal epithelial growth in MMTV-cNeu mice. Oncogene 2015; 34:2003-10. [PMID: 24909172 PMCID: PMC4261057 DOI: 10.1038/onc.2014.149] [Citation(s) in RCA: 16] [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: 08/14/2013] [Revised: 04/01/2014] [Accepted: 04/21/2014] [Indexed: 02/07/2023]
Abstract
Significant left-right (L-R) differences in tumor incidence and disease outcome occur for cancers of paired organs, including the breasts; however, the basis for this laterality is unknown. Here, we show that despite their morphologic symmetry, left versus right mammary glands in wild-type mice have baseline differences in gene expression that are L-R independently regulated during pubertal development, including genes that regulate luminal progenitor cell renewal, luminal cell differentiation, mammary tumorigenesis, tamoxifen sensitivity and chemotherapeutic resistance. In MMTV-cNeu(Tg/Tg) mice, which model HER2/Neu-amplified breast cancer, baseline L-R differences in mammary gene expression are amplified, sustained or inverted in a gene-specific manner and the mammary ductal epithelium undergoes L-R asymmetric growth and patterning. Comparative genomic analysis of mouse L-R mammary gene expression profiles with gene expression profiles of human breast tumors revealed significant linkage between right-sided gene expression and decreased breast cancer patient survival. Collectively, these findings are the first to demonstrate that mammary glands are lateralized organs, and, moreover, that mammary glands have L-R differential susceptibility to HER2/Neu oncogene-mediated effects on ductal epithelial growth and differentiation. We propose that intrinsic molecular laterality may have a role in L-R asymmetric breast tumor incidence and, furthermore, that interplay between the L-R molecular landscape and oncogene activity may contribute to the differential disease progression and patient outcome that are associated with tumor situs.
Collapse
Affiliation(s)
- Jacqulyne P. Robichaux
- Department of Regenerative Medicine and Cell Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
| | - Robin M. Hallett
- Department of Biochemistry and Biomedical Sciences, Centre for Functional Genomics, McMaster University, Ontario, Canada
| | - John W. Fuseler
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208
| | - John A. Hassell
- Department of Biochemistry and Biomedical Sciences, Centre for Functional Genomics, McMaster University, Ontario, Canada
| | - Ann F. Ramsdell
- Department of Regenerative Medicine and Cell Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208
- Program In Women’s and Gender Studies, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208
| |
Collapse
|
43
|
Correlation between epidermal growth factor receptor and tumor stem cell markers CD44/CD24 and their relationship with prognosis in breast invasive ductal carcinoma. Med Oncol 2014; 32:275. [PMID: 25429827 PMCID: PMC4246130 DOI: 10.1007/s12032-014-0275-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/29/2014] [Indexed: 12/18/2022]
Abstract
We studied the correlation between epidermal growth factor receptor (EGFR) and the tumor stem cell markers CD44/CD24 in breast invasive ductal carcinoma (BIDC) and their relationship with prognosis. We analyzed the clinical data of 139 BIDC cases retrospectively, detecting EGFR, CD44, and CD24 expressions in tumor tissue using immunohistochemistry. The proportion of EGFR-, CD44-, and CD24-positive cases was 59.0, 62.3, and 30.9 %, respectively. The proportion of CD44-positive [76.9 % (p < 0.05)] and EGFR-positive [67.2 % (p = 0.108)] cases in the triple-negative breast cancer (TNBC) group was higher than that of the non-TNBC group. In the non-TNBC group, 36.5 % was CD24-positive, higher than that in the TNBC group but not statistically significant. The proportion of CD44-positive cases was significantly higher in the EGFR-positive group than in the EGFR-negative group (p = 0.017). EGFR-positive cases were significantly correlated with premenopausal status (p = 0.036), distant metastasis (p = 0.018), and estrogen receptor-negative status (p = 0.020). CD44-positive status was significantly correlated with human epidermal growth receptor 2 (HER2)-negative (p = 0.023), estrogen receptor-negative (p = 0.021), and progesterone receptor-negative status (p = 0.004). CD24-positive status was significantly correlated with HER2-positive status (p = 0.001). Kaplan-Meier survival analysis showed that TNBC patients had shorter survival. EGFR-positive and CD44-positive status were both correlated with shorter survival in the lymph node- and HR-negative groups, while CD24 positive was significantly correlated with poor survival in lymph node-negative and HR-positive patients. EGFR and CD44 expressions have a significantly positive correlation (p = 0.017) in BIDC. Patients both EGFR and CD44 positive had the worst outcome.
Collapse
|
44
|
Jiwa LS, van Diest PJ, Hoefnagel LD, Wesseling J, Wesseling P, Moelans CB. Upregulation of Claudin-4, CAIX and GLUT-1 in distant breast cancer metastases. BMC Cancer 2014; 14:864. [PMID: 25417118 PMCID: PMC4247109 DOI: 10.1186/1471-2407-14-864] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/11/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Several studies have shown that the immunophenotype of distant breast cancer metastases may differ significantly from that of the primary tumor, especially with regard to differences in the level of hormone receptor protein expression, a process known as receptor conversion. This study aimed to compare expression levels of several membrane proteins between primary breast tumors and their corresponding distant metastases in view of their potential applicability for molecular imaging and drug targeting. METHODS Expression of Claudin-4, EGFR, CAIX, GLUT-1 and IGF1R was assessed by immunohistochemistry on tissue microarrays composed of 97 paired primary breast tumors and their distant (non-bone) metastases. RESULTS In both the primary cancers and the metastases, Claudin-4 was most frequently expressed, followed by GLUT-1, CAIX and EGFR.From primary breast cancers to their distant metastases there was positive to negative conversion, e.g. protein expression in the primary tumor with no expression in its paired metastasis, in 6%, 19%, 12%, 38%, and 0% for Claudin-4 (n.s), GLUT-1 (n.s), CAIX (n.s), EGFR (n.s) and IGF1R (n.s) respectively. Negative to positive conversion was seen in 65%, 47%, 43%, 9% and 0% of cases for Claudin-4 (p = 0.049), GLUT-1 (p = 0.024), CAIX (p = 0.002), EGFR (n.s.) and IGF1R (n.s.) respectively. Negative to positive conversion of Claudin-4 in the metastasis was significantly associated with tumor size (p = 0.015), negative to positive conversion of EGFR with negative PR status (p = 0.046) and high MAI (p = 0.047) and GLUT-1 negative to positive conversion with (neo)adjuvant chemotherapy (p = 0.039) and time to metastasis formation (p = 0.034). CAIX and GLUT-1 expression in the primary tumor were significantly associated with high MAI (p = 0.008 and p = 0.038 respectively). CONCLUSION Claudin-4 is frequently expressed in primary breast cancers but especially in their metastases and is thereby an attractive membrane bound molecular imaging and drug target. Conversion in expression of the studied proteins from the primary tumor to metastases was fairly frequent, except for IGF1R, implying that the expression status of metastases cannot always be reliably predicted from the primary tumor, thereby necessitating biopsy for reliable assessment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cathy B Moelans
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, PO Box 85500, Utrecht 3508GA, The Netherlands.
| |
Collapse
|
45
|
Yiqi formula enhances the antitumor effects of erlotinib for treatment of triple-negative breast cancer xenografts. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:628712. [PMID: 25389442 PMCID: PMC4217362 DOI: 10.1155/2014/628712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 08/26/2014] [Accepted: 09/02/2014] [Indexed: 11/17/2022]
Abstract
Yiqi formula (YF), a traditional herbal prescription, has long been used to treat triple-negative breast cancer (TNBC) patients. The present study aims to investigate the effects and the related mechanism of YF for treatment of TNBC xenografts. MDA-MB-231 (human TNBC) cells were subcutaneously injected into the second mammary fat pad of 40 female nude mice, which were divided into four groups: control, erlotinib (an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor), YF, and combination (YF plus erlotinib). All treatments were administered orally for 30 days. Inhibition rate of tumor weight by erlotinib, YF, and the combination was 26.47%, 17.24%, and 39.15%, respectively. Western blotting showed that YF, erlotinib, and the combination downregulated p-EGFR (P < 0.01) and p-Akt1 (pT308) (P < 0.05) and upregulated PTEN compared with control, and the combination was more efficacious than erlotinib alone (P < 0.05). Similar results were detected by immunohistochemistry. Real-time quantitative PCR showed that YF, erlotinib, and the combination increased PTEN mRNA (P < 0.05, P < 0.01) compared with control, and the combination was more efficacious than erlotinib alone (P < 0.05). In conclusion, YF can regulate the main components of the PI3K/Akt pathway in TNBC xenografts. When YF was used in combination with erlotinib, it enhanced the antitumor effects of erlotinib on TNBC xenografts. These findings suggest that YF is suitable to use for the treatment of TNBC patients.
Collapse
|
46
|
The expression of Survivin and NF-κB associated with prognostically worse clinicopathologic variables in hepatocellular carcinoma. Tumour Biol 2014; 35:9905-10. [DOI: 10.1007/s13277-014-2279-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/24/2014] [Indexed: 12/21/2022] Open
|