1
|
Wang X, Zhao S, Xin Q, Zhang Y, Wang K, Li M. Recent progress of CDK4/6 inhibitors' current practice in breast cancer. Cancer Gene Ther 2024; 31:1283-1291. [PMID: 38409585 PMCID: PMC11405274 DOI: 10.1038/s41417-024-00747-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/28/2024]
Abstract
Dysregulated cellular proliferation represents a hallmark feature across all cancers. Aberrant activation of the cyclin-dependent kinase 4 and 6 (CDK4/6) pathway, independent of mitogenic signaling, engenders uncontrolled breast cancer cell proliferation. Consequently, the advent of CDK4/6 inhibition has constituted a pivotal milestone in the realm of targeted breast cancer therapy. The combination of CDK4/6 inhibitors (CDK4/6i) with endocrine therapy (ET) has emerged as the foremost therapeutic modality for patients afflicted with hormone receptor-positive (HR + )/HER2-negative (HER2-) advanced breast cancer. At present, the Food and Drug Administration (FDA) has sanctioned various CDK4/6i for employment as the primary treatment regimen in HR + /HER2- breast cancer. This therapeutic approach has demonstrated a substantial extension of progression-free survival (PFS), often amounting to several months, when administered alongside endocrine therapy. Within this comprehensive review, we systematically evaluate the utilization strategies of CDK4/6i across various subpopulations of breast cancer and explore potential therapeutic avenues following disease progression during application of CDK4/6i therapy.
Collapse
Affiliation(s)
- Xueqing Wang
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Shanshan Zhao
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Qinghan Xin
- Department of Breast Surgery, Dalian Municipal Central Hospital, Dalian, China
| | - Yunkun Zhang
- Department of Pathology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Kainan Wang
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, China.
| | - Man Li
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
2
|
Curcio F, Sanguedolce M, Filice L, Testa F, Catapano G, Giordano F, Trombino S, Cassano R. Hybrid Nanoparticles Based on Mesoporous Silica and Functionalized Biopolymers as Drug Carriers for Chemotherapeutic Agents. MATERIALS (BASEL, SWITZERLAND) 2024; 17:3877. [PMID: 39124540 PMCID: PMC11313671 DOI: 10.3390/ma17153877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) are promising drug carriers for cancer therapy. Their functionalization with ligands for specific tissue/cell targeting and stimuli-responsive cap materials for sealing drugs within the pores of MSNs is extensively studied for biomedical and pharmaceutical applications. The objective of the present work was to establish MSNs as ideal nanocarriers of anticancer drugs such as 5-FU and silymarin by exploiting characteristics such as their large surface area, pore size, and biocompatibility. Furthermore, coating with various biopolymeric materials such as carboxymethyl chitosan-dopamine and hyaluronic acid-folic acid on their surface would allow them to play the role of ligands in the process of active targeting to tumor cells in which there is an overexpression of specific receptors for them. From the results obtained, it emerged, in fact, that these hybrid nanoparticles not only inhibit the growth of glioblastoma and breast cancer cells, but also act as pH-responsive release systems potentially useful as release vectors in tumor environments.
Collapse
Affiliation(s)
- Federica Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende, 87036 Cosenza, Italy; (F.C.); (F.G.)
| | - Michela Sanguedolce
- Department of Mechanical, Energy and Management Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (M.S.); (L.F.); (G.C.)
| | - Luigino Filice
- Department of Mechanical, Energy and Management Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (M.S.); (L.F.); (G.C.)
| | - Flaviano Testa
- Department of Computer Engineering, Modeling, Electronics and Systems Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy;
| | - Gerardo Catapano
- Department of Mechanical, Energy and Management Engineering, University of Calabria, Via P. Bucci, 87036 Rende, Italy; (M.S.); (L.F.); (G.C.)
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende, 87036 Cosenza, Italy; (F.C.); (F.G.)
| | - Sonia Trombino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende, 87036 Cosenza, Italy; (F.C.); (F.G.)
| | - Roberta Cassano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende, 87036 Cosenza, Italy; (F.C.); (F.G.)
| |
Collapse
|
3
|
Wu J, Ge L, Guo Y, Zhao A, Yao J, Wang Z, Xu D. Predicting hormone receptor status in invasive breast cancer through radiomics analysis of long-axis and short-axis ultrasound planes. Sci Rep 2024; 14:16503. [PMID: 39080346 PMCID: PMC11289262 DOI: 10.1038/s41598-024-67145-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
The hormone receptor (HR) status plays a significant role in breast cancer, serving as the primary guide for treatment decisions and closely correlating with prognosis. This study aims to investigate the predictive value of radiomics analysis in long-axis and short-axis ultrasound planes for distinguishing between HR-positive and HR-negative breast cancers. A cohort of 505 patients from two hospitals was stratified into discovery (Institute 1, 416 patients) and validation (Institute 2, 89 patients) cohorts. A comprehensive set of 788 ultrasound radiomics features was extracted from both long-axis and short-axis ultrasound planes, respectively. Utilizing least absolute shrinkage and selection operator (LASSO) regression analysis, distinct models were constructed for the long-axis and short-axis data. Subsequently, radiomics scores (Rad-scores) were computed for each patient. Additionally, a combined model was formulated by integrating data from long-axis and short-axis Rad-scores along with clinical factors. The diagnostic efficacy of all models was evaluated by the area under the receiver operating characteristic (ROC) curve (AUC). The long-axis and short-axis models, consisting of 11 features and 15 features, respectively, were established, yielding AUCs of 0.743 and 0.751 in the discovery cohort, and 0.795 and 0.744 in the validation cohort. The calculated long-axis and short-axis Rad-scores exhibited significant differences between HR-positive and HR-negative groups across all cohorts (all p < 0.001). Univariate analysis identified ultrasound-reported tumor size as an independent predictor. The combined model, incorporating long-axis and short-axis Rad-scores along with tumor size, achieved superior AUCs of 0.788 and 0.822 in the discovery and validation cohorts, respectively. The combined model effectively distinguishes between HR-positive and HR-negative breast cancers based on ultrasound radiomics features and tumor size, which may offer a valuable tool to facilitate treatment decision making and prognostic assessment.
Collapse
Affiliation(s)
- Jiangfeng Wu
- Department of Ultrasonography, Dongyang People's Hospital, No. 60 Wuning West Road, Dongyang, Zhejiang, China.
| | - Lifang Ge
- Department of Ultrasonography, Dongyang People's Hospital, No. 60 Wuning West Road, Dongyang, Zhejiang, China
| | - Yinghong Guo
- Department of Ultrasonography, Dongyang People's Hospital, No. 60 Wuning West Road, Dongyang, Zhejiang, China
| | - Anli Zhao
- Department of Ultrasonography, Dongyang People's Hospital, No. 60 Wuning West Road, Dongyang, Zhejiang, China
| | - Jincao Yao
- Department of Ultrasonography, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Zhengping Wang
- Department of Ultrasonography, Dongyang People's Hospital, No. 60 Wuning West Road, Dongyang, Zhejiang, China
| | - Dong Xu
- Department of Ultrasonography, Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.
| |
Collapse
|
4
|
De Marchi T, Lai CF, Simmons GM, Goldsbrough I, Harrod A, Lam T, Buluwela L, Kjellström S, Brueffer C, Saal LH, Malmström J, Ali S, Niméus E. Proteomic profiling reveals that ESR1 mutations enhance cyclin-dependent kinase signaling. Sci Rep 2024; 14:6873. [PMID: 38519482 PMCID: PMC10959978 DOI: 10.1038/s41598-024-56412-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
Three quarters of all breast cancers express the estrogen receptor (ER, ESR1 gene), which promotes tumor growth and constitutes a direct target for endocrine therapies. ESR1 mutations have been implicated in therapy resistance in metastatic breast cancer, in particular to aromatase inhibitors. ESR1 mutations promote constitutive ER activity and affect other signaling pathways, allowing cancer cells to proliferate by employing mechanisms within and without direct regulation by the ER. Although subjected to extensive genetic and transcriptomic analyses, understanding of protein alterations remains poorly investigated. Towards this, we employed an integrated mass spectrometry based proteomic approach to profile the protein and phosphoprotein differences in breast cancer cell lines expressing the frequent Y537N and Y537S ER mutations. Global proteome analysis revealed enrichment of mitotic and immune signaling pathways in ER mutant cells, while phosphoprotein analysis evidenced enriched activity of proliferation associated kinases, in particular CDKs and mTOR. Integration of protein expression and phosphorylation data revealed pathway-dependent discrepancies (motility vs proliferation) that were observed at varying degrees across mutant and wt ER cells. Additionally, protein expression and phosphorylation patterns, while under different regulation, still recapitulated the estrogen-independent phenotype of ER mutant cells. Our study is the first proteome-centric characterization of ESR1 mutant models, out of which we confirm estrogen independence of ER mutants and reveal the enrichment of immune signaling pathways at the proteomic level.
Collapse
Affiliation(s)
- Tommaso De Marchi
- Division of Surgery, Oncology, and Pathology, Department of Clinical Sciences, Lund University, Solvegatan 19, 22362, Lund, Sweden.
| | - Chun-Fui Lai
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Georgia M Simmons
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Isabella Goldsbrough
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Alison Harrod
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Thai Lam
- Division of Surgery, Oncology, and Pathology, Department of Clinical Sciences, Lund University, Solvegatan 19, 22362, Lund, Sweden
| | - Lakjaya Buluwela
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Sven Kjellström
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, Solvegatan 19, 22362, Lund, Sweden
- Swedish National Infrastructure for Biological Mass Spectrometry - BioMS, Lund, Sweden
| | - Christian Brueffer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, 22381, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, 22381, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Klinikgatan 32, 22184, Lund, Sweden
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| | - Emma Niméus
- Division of Surgery, Oncology, and Pathology, Department of Clinical Sciences, Lund University, Solvegatan 19, 22362, Lund, Sweden.
- Department of Surgery, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
5
|
Fard SS, Holz MK. Regulation of mRNA translation by estrogen receptor in breast cancer. Steroids 2023; 200:109316. [PMID: 37806603 PMCID: PMC10841406 DOI: 10.1016/j.steroids.2023.109316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023]
Abstract
Breast cancer is one of the leading causes of cancer-related fatalities and the most often diagnosed malignancy in women globally. Dysregulation of sex hormone signaling pathways mediated by the estrogen receptor (ER) in breast cancer is well characterized. Although ER is known to promote cell growth and survival by altering gene transcription, recent research suggests that its effects in cancers are also mediated through dysregulation of protein synthesis. This implies that ER can coordinately affect gene expression through both translational and transcriptional pathways, leading to the development of malignancy. In this review, we will cover the current understanding of how the ER controls mRNA translation in breast cancer and discuss any potential clinical implications of this phenomenon.
Collapse
Affiliation(s)
- Shahrzad S Fard
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA
| | - Marina K Holz
- Department of Cell Biology and Anatomy, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA; Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
6
|
Abstract
Proteolysis-targeting chimera (PROTAC) is an emerging technique for degrading disease-related proteins. However, the current PROTACs suffer from inadequate solubility and lack of organ targeting, which has hampered their druggability. Herein, we report direct and sustained delivery of PROTACs using microneedle patches to the diseased tissues. In this study, we use an estrogen receptor alpha (ERα)-degrading PROTAC, ERD308, to treat ER-positive breast cancer. A pH-sensitive micelle, MPEG-poly(β-amino ester) (MPEG-PAE), is used to encapsulate ERD308 along with an FDA-approved CDK4/6 inhibitor, Palbociclib (Pal), before loading into biodegradable microneedle patches. These patches enable prolonged drug release into deep tumors, maintaining therapeutic levels for at least 4 days, with an excellent drug retention rate of over 87% in tumors. ERD308 released from the microneedle patches can sufficiently degrade ERα in MCF7 cells. Co-administration of ERD308 and Palbociclib exhibits excellent efficacy by over 80% tumor reduction as well as a good safety profile. Our work demonstrates the feasibility and proof-of-concept therapeutic potential of using microneedle patches to directly deliver PROTACs into tumors.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Shiqi Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Ke Cheng
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
7
|
Musheyev D, Miller E, Birnbaum N, Miller E, Erblich S, Schuck A, Alayev A. Inhibition of ERK signaling for treatment of ERRα positive TNBC. PLoS One 2023; 18:e0283047. [PMID: 37163498 PMCID: PMC10171695 DOI: 10.1371/journal.pone.0283047] [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: 08/08/2022] [Accepted: 02/28/2023] [Indexed: 05/12/2023] Open
Abstract
Breast cancer is the second leading cause of cancer-related deaths in women and triple-negative breast cancer (TNBC), in particular, is an aggressive and highly metastatic type of breast cancer that does not respond to established targeted therapies and is associated with poor prognosis and worse survival. Previous studies identified a subgroup of triple-negative breast cancer patients with high expression of estrogen related receptor alpha (ERRα) that has better prognosis when treated with tamoxifen. We therefore set out to identify common targets of tamoxifen and ERRα in the context of TNBC using phosphoproteomic analysis. In this study, we discovered that phosphorylation of mitogen-activated protein kinase 1 (MAPK1) is regulated by tamoxifen as well as ERRα. Additionally, we showed that inhibition of MAPK signaling together with the use of a selective ERRα inverse agonist, XCT-790, leads to a significant upregulation of apoptosis and paves way for the therapeutic use of MAPK inhibitors for treatment of ERRα expressing TNBC.
Collapse
Affiliation(s)
- David Musheyev
- Department of Internal Medicine, Stony Brook Southampton Hospital, Southampton, New York, United States of America
| | - Esther Miller
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Natania Birnbaum
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Elisheva Miller
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Shoshana Erblich
- Department of Mechanical Engineering, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Alyssa Schuck
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Anya Alayev
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| |
Collapse
|
8
|
Giordano F, Paolì A, Forastiero M, Marsico S, De Amicis F, Marrelli M, Naimo GD, Mauro L, Panno ML. Valproic acid inhibits cell growth in both MCF-7 and MDA-MB231 cells by triggering different responses in a cell type-specific manner. J Transl Med 2023; 21:165. [PMID: 36864445 PMCID: PMC9983172 DOI: 10.1186/s12967-023-04015-8] [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: 01/03/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Breast cancer is the second leading cause of death among women after lung cancer. Despite the improvement in prevention and in therapy, breast cancer still remains a threat, both for pre- and postmenopausal women, due to the development of drug resistance. To counteract that, novel agents regulating gene expression have been studied in both hematologic and solid tumors. The Histone Deacetylase (HDAC) inhibitor Valproic Acid (VA), used for epilepsy and other neuropsychiatric diseases, has been demonstrated a strong antitumoral and cytostatic activity. In this study, we tested the effects of Valproic Acid on the signaling pathways involved in breast cancer cells viability, apoptosis and in Reactive Oxygen Species (ROS) production using ER-α positive MCF-7 and triple negative MDA-MB-231 cells. METHODS Cell proliferation assay was performed by MTT Cell cycle, ROS levels and apoptosis were analyzed by flow cytometry, protein levels were detected by Western Blotting. RESULTS Cell treatment with Valproic Acid reduced cell proliferation and induced G0/G1 cell cycle arrest in MCF-7 and G2/M block in MDA-MB-231 cells. In addition, in both cells the drug enhanced the generation of ROS by the mitochondria. In MCF-7 treated cells, it has been observed a reduction in mitochondrial membrane potential, a down regulation of the anti-apoptotic marker Bcl-2 and an increase of Bax and Bad, leading to release of cytochrome C and PARP cleavage. Less consistent effects are recorded in MDA-MB-231 cells, in which the greater production of ROS, compared to MCF-7cells, involves an inflammatory response (activation of p-STAT3, increased levels of COX2). CONCLUSIONS Our results have demonstrated that in MCF-7 cells the Valproic Acid is a suitable drug to arrest cell growth, to address apoptosis and mitochondrial perturbations, all factors that are important in determining cell fate and health. In a triple negative MDA-MB 231 cells, valproate directs the cells towards the inflammatory response with a sustained expression of antioxidant enzymes. Overall, the not always unequivocal data between the two cellular phenotypes indicate that further studies are needed to better define the use of the drug, also in combination with other chemotherapy, in the treatment of breast tumors.
Collapse
Affiliation(s)
- Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| | - Alessandro Paolì
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Martina Forastiero
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Mariangela Marrelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Loredana Mauro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
9
|
Brodzki A, Łopuszyński W, Brodzki P, Głodkowska K, Knap B, Gawin P. Pharmacological Treatment of Perianal Gland Tumors in Male Dogs. Animals (Basel) 2023; 13:ani13030463. [PMID: 36766353 PMCID: PMC9913509 DOI: 10.3390/ani13030463] [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: 12/13/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The presence of androgen (AR) and estrogen (ER) receptors has been demonstrated both in normal perianal (hepatoid) glands and in perianal tumors. The aim of this study was to demonstrate the relationship between the expression of AR and ER in perianal gland tumors and the effectiveness of antihormonal treatment. The study was performed on 41 male dogs with neoplastic lesions of the anal region. Histopathological evaluation of the lesions revealed 24 adenomas, 12 epitheliomas, and five carcinomas. Treatment was administered orally with tamoxifen at a dose of 1 mg/kg BW and cyproterone acetate at a dose of 5 mg/kg. Tumor diameters were measured regularly with calipers and recorded in millimeters starting with the measurement before treatment, and then after 1, 2, 3, 6, 12, 18, and 24 months of therapy. The results show that hepatoid adenomas that are characterized by high expression of AR and ER receptors respond positively to antihormonal therapy, resulting in complete tumor regression. For locally malignant hepatoid epitheliomas and carcinomas with low expression of AR and ER receptors, antihormonal therapy makes it possible to reduce the size of the tumor, but does not make it possible to cure it completely.
Collapse
Affiliation(s)
- Adam Brodzki
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Wojciech Łopuszyński
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
- Correspondence: ; Tel.: +48-81-445-6162
| | - Piotr Brodzki
- Department of Andrology and Biotechnology of Reproduction, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Katarzyna Głodkowska
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Bartosz Knap
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland
- Chair and Department of Experimental and Clinical Pharmacology, Faculty of Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | | |
Collapse
|
10
|
Scherbakov AM, Basharina AA, Sorokin DV, Mikhaevich EI, Mizaeva IE, Mikhaylova AL, Bogush TA, Krasil’nikov MA. Targeting hormone-resistant breast cancer cells with docetaxel: a look inside the resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:103-115. [PMID: 37065867 PMCID: PMC10099602 DOI: 10.20517/cdr.2022.96] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/01/2022] [Accepted: 01/04/2023] [Indexed: 04/18/2023]
Abstract
Aim: The study aims to analyze the effect of long-term incubation of ERα-positive MCF7 breast cancer cells with 4-hydroxytamoxifen (HT) on their sensitivity to tubulin polymerization inhibitor docetaxel. Methods: The analysis of cell viability was performed by the MTT method. The expression of signaling proteins was analyzed by immunoblotting and flow cytometry. ERα activity was evaluated by gene reporter assay. To establish hormone-resistant subline MCF7, breast cancer cells were treated with 4-hydroxytamoxifen for 12 months. Results: The developed MCF7/HT subline has lost sensitivity to 4-hydroxytamoxifen, and the resistance index was 2. Increased Akt activity (2.2-fold) and decreased ERα expression (1.5-fold) were revealed in MCF7/HT cells. The activity of the estrogen receptor α was reduced (1.5-fold) in MCF7/HT. Evaluation of class III β-tubulin expression (TUBB3), a marker associated with metastasis, revealed the following trends: higher expression of TUBB3 was detected in triple-negative breast cancer MDA-MB-231 cells compared to hormone-responsive MCF7 cells (P < 0.05). The lowest expression of TUBB3 was found in hormone-resistant MCF7/HT cells (MCF7/HT < MCF7 < MDA-MB-231, approximately 1:2:4). High TUBB3 expression strongly correlated with docetaxel resistance: IC50 value of docetaxel for MDA-MB-231 cells was greater than that for MCF7 cells, whereas resistant MCF7/HT cells were the most sensitive to the drug. The accumulation of cleaved PARP (a 1.6-fold increase) and Bcl-2 downregulation (1.8-fold) were more pronounced in docetaxel-treated resistant cells (P < 0.05). The expression of cyclin D1 decreased (2.8-fold) only in resistant cells after 4 nM docetaxel treatment, while this marker was unchanged in parental MCF7 breast cancer cells. Conclusion: Further development of taxane-based chemotherapy for hormone-resistant cancer looks highly promising, especially for cancers with low TUBB3 expression.
Collapse
Affiliation(s)
- Alexander M. Scherbakov
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
- Correspondence to: Dr. Alexander M. Scherbakov, Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, Moscow 115522, Russia. E-mail:
| | - Anna A. Basharina
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Danila V. Sorokin
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Ekaterina I. Mikhaevich
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Iman E. Mizaeva
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Alexandra L. Mikhaylova
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Tatiana A. Bogush
- Group of Molecular Tumor Markers, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| | - Mikhail A. Krasil’nikov
- Department of Experimental Tumor Biology, Blokhin N.N. National Medical Research Center of Oncology, Moscow 115522, Russian Federation
| |
Collapse
|
11
|
Wo G, Zhu Z, Fang Z, Chen X, Liang M, Wang Y, Shao X, Shen H, Tang J. Dihydrotanshinone I: A Target for STAT3 in the Therapy of Tamoxifen‐Resistant Breast Cancer. ChemistrySelect 2022. [DOI: 10.1002/slct.202203082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Guanqun Wo
- Nanjing University of Chinese Medicine Xianlin Road 138 Nanjing 210023 P. R. China
| | - Zhen Zhu
- Nanjing Medical University Nanjing 210029 P. R. China
| | - Zheng Fang
- Nanjing Medical University Nanjing 210029 P. R. China
| | - Xi Chen
- Nanjing University of Chinese Medicine Xianlin Road 138 Nanjing 210023 P. R. China
| | | | - Yalin Wang
- Xuzhou Medical University 209 Tongshan Road Xuzhou 221004 P. R. China
| | - Xinyi Shao
- Nanjing University of Chinese Medicine Xianlin Road 138 Nanjing 210023 P. R. China
| | - Hongyu Shen
- Nanjing Medical University Nanjing 210029 P. R. China
| | - Jin‐Hai Tang
- Nanjing University of Chinese Medicine Xianlin Road 138 Nanjing 210023 P. R. China
| |
Collapse
|
12
|
A kinase inhibitor screen reveals MEK1/2 as a novel therapeutic target to antagonize IGF1R-mediated antiestrogen resistance in ERα-positive luminal breast cancer. Biochem Pharmacol 2022; 204:115233. [PMID: 36041543 DOI: 10.1016/j.bcp.2022.115233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
Antiestrogen resistance of breast cancer has been related to enhanced growth factor receptor expression and activation. We have previously shown that ectopic expression and subsequent activation of the insulin-like growth factor-1 receptor (IGF1R) or the epidermal growth factor receptor (EGFR) in MCF7 or T47D breast cancer cells results in antiestrogen resistance. In order to identify novel therapeutic targets to prevent this antiestrogen resistance, we performed kinase inhibitor screens with 273 different inhibitors in MCF7 cells overexpressing IGF1R or EGFR. Kinase inhibitors that antagonized antiestrogen resistance but are not directly involved in IGF1R or EGFR signaling were prioritized for further analyses. Various ALK (anaplastic lymphoma receptor tyrosine kinase) inhibitors inhibited cell proliferation in IGF1R expressing cells under normal and antiestrogen resistance conditions by preventing IGF1R activation and subsequent downstream signaling; the ALK inhibitors did not affect EGFR signaling. On the other hand, MEK (mitogen-activated protein kinase kinase)1/2 inhibitors, including PD0325901, selumetinib, trametinib and TAK733, selectively antagonized IGF1R signaling-mediated antiestrogen resistance but did not affect cell proliferation under normal growth conditions. RNAseq analysis revealed that MEK inhibitors PD0325901 and selumetinib drastically altered cell cycle progression and cell migration networks under IGF1R signaling-mediated antiestrogen resistance. In a group of 219 patients with metastasized ER+ breast cancer, strong pMEK staining showed a significant correlation with no clinical benefit of first-line tamoxifen treatment. We propose a critical role for MEK activation in IGF1R signaling-mediated antiestrogen resistance and anticipate that dual-targeted therapy with a MEK inhibitor and antiestrogen could improve treatment outcome.
Collapse
|
13
|
Role of the Mediator Complex and MicroRNAs in Breast Cancer Etiology. Genes (Basel) 2022; 13:genes13020234. [PMID: 35205279 PMCID: PMC8871970 DOI: 10.3390/genes13020234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022] Open
Abstract
Transcriptional coactivators play a key role in RNA polymerase II transcription and gene regulation. One of the most important transcriptional coactivators is the Mediator (MED) complex, which is an evolutionary conserved large multiprotein complex. MED transduces the signal between DNA-bound transcriptional activators (gene-specific transcription factors) to the RNA polymerase II transcription machinery to activate transcription. It is known that MED plays an essential role in ER-mediated gene expression mainly through the MED1 subunit, since estrogen receptor (ER) can interact with MED1 by specific protein–protein interactions; therefore, MED1 plays a fundamental role in ER-positive breast cancer (BC) etiology. Additionally, other MED subunits also play a role in BC etiology. On the other hand, microRNAs (miRNAs) are a family of small non-coding RNAs, which can regulate gene expression at the post-transcriptional level by binding in a sequence-specific fashion at the 3′ UTR of the messenger RNA. The miRNAs are also important factors that influence oncogenic signaling in BC by acting as both tumor suppressors and oncogenes. Moreover, miRNAs are involved in endocrine therapy resistance of BC, specifically to tamoxifen, a drug that is used to target ER signaling. In metazoans, very little is known about the transcriptional regulation of miRNA by the MED complex and less about the transcriptional regulation of miRNAs involved in BC initiation and progression. Recently, it has been shown that MED1 is able to regulate the transcription of the ER-dependent miR-191/425 cluster promoting BC cell proliferation and migration. In this review, we will discuss the role of MED1 transcriptional coactivator in the etiology of BC and in endocrine therapy-resistance of BC and also the contribution of other MED subunits to BC development, progression and metastasis. Lastly, we identified miRNAs that potentially can regulate the expression of MED subunits.
Collapse
|
14
|
Yuan Y, Xiao WW, Xie WH, Li RZ, Gao YH. Prognostic value of ubiquitin E2 UBE2W and its correlation with tumor-infiltrating immune cells in breast cancer. BMC Cancer 2021; 21:479. [PMID: 33931024 PMCID: PMC8086329 DOI: 10.1186/s12885-021-08234-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background Ubiquitin-conjugating enzyme E2W (UBE2W) is a protein-coding gene that has an important role in ubiquitination and may be vital in the repair of DNA damage. However, studies on the prognostic value of UBE2W and its correlation with tumor-infiltrating immune cells in multiple cancers have not been addressed. Methods We investigated UBE2W expression in the Oncomine database, the Tumor Immune Estimation Resource (TIMER), TNMplot database. Then, the clinical prognostic value of UBE2W was analyzed via online public databases. Meanwhile, we explored the correlation between UBE2W and DNA repair associate genes expression and DNA methyltransferase expression by TIMER and Gene Expression Profiling Interactive Analysis (GEPIA). By using the same method, the correlation between UBE2W and tumor-infiltrating immune cells was explored. Genomic Profiles of UBE2W in breast cancer (BRCA) were accessed in cBioPortal (v3.5.0). Functional proteins associated network was analyzed by STRING database (v11.0). Results UBE2W was abnormally expressed and significantly correlated with mismatch repair (MMR) gene mutation levels, DNA methyltransferase, and BRCA1/2 expression in breast cancer. High expression of UBE2W may promote the tumor immunosuppression and metastasis, induce endocrine therapy resistance and deteriorate outcomes of patients with breast cancer. These findings suggest that UBE2W could be a potential biomarker of prognosis and tumor-infiltrating immune cells. Besides, RBX1 may be a new E3 that was regulated by UBE2W. Conclusions Ubiquitin E2 UBE2W is a potential prognostic biomarker and is correlated with immune infiltration in BRCA. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08234-4.
Collapse
Affiliation(s)
- Yan Yuan
- State Key laboratory of Oncology in South China, Collaborative innovation Center for cancer Medicine, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wei-Wei Xiao
- State Key laboratory of Oncology in South China, Collaborative innovation Center for cancer Medicine, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wei-Hao Xie
- State Key laboratory of Oncology in South China, Collaborative innovation Center for cancer Medicine, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Rong-Zhen Li
- State Key laboratory of Oncology in South China, Collaborative innovation Center for cancer Medicine, Guangzhou, P. R. China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yuan-Hong Gao
- State Key laboratory of Oncology in South China, Collaborative innovation Center for cancer Medicine, Guangzhou, P. R. China. .,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| |
Collapse
|
15
|
Leenhardt F, Alexandre M, Jacot W. Alpelisib for the treatment of PIK3CA-mutated, hormone receptor-positive, HER2-negative metastatic breast cancer. Expert Opin Pharmacother 2021; 22:667-675. [PMID: 33622114 DOI: 10.1080/14656566.2021.1873952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Two-thirds of advanced breast cancers are hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-). Gene mutations in PIK3CA, encoding the PI3K catalytic subunit alpha of phosphatidyl-inositol 3-kinase (PI3K), are a frequent event in this population and are implicated in hormone therapy resistance. Alpelisib is a PI3K-alpha inhibitor and is the first PI3K inhibitor approved, in association with fulvestrant, by the FDA and EMA, based on improved progression-free survival (PFS) versus fulvestrant alone in a randomized phase III trial in HR+/HER2-, PIK3CA-mutated tumors following progression on/after HT.Areas covered: The scientific rationale, preclinical development, pharmacokinetics, and clinical efficacy/safety of alpelisib-fulvestrant are summarized. The role of alpelisib in the clinical setting is discussed, referencing current therapeutic options and clinical challenges associated with alpelisib's safety profile.Expert opinion: Alpelisib is an option for patients with HR+/HER2-, PIK3CA-mutated tumors whose disease progressed during/after aromatase inhibitor treatment. The PFS benefit appears clinically significant over fulvestrant alone, with a 7.9 months, non-significant, improvement in overall survival. Its safety profile requires strict patient selection, mainly based on baseline glycemic status, and close monitoring.
Collapse
Affiliation(s)
- Fanny Leenhardt
- Laboratoire de Pharmacocinétique, Faculté de Pharmacie, Université de Montpellier, Montpellier, France.,Service Pharmacie, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France
| | - Marie Alexandre
- Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| | - William Jacot
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France.,Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, Montpellier, France
| |
Collapse
|
16
|
Metcalf S, Petri BJ, Kruer T, Green B, Dougherty S, Wittliff JL, Klinge CM, Clem BF. Serine synthesis influences tamoxifen response in ER+ human breast carcinoma. Endocr Relat Cancer 2021; 28:27-37. [PMID: 33112838 PMCID: PMC7780089 DOI: 10.1530/erc-19-0510] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022]
Abstract
Estrogen receptor-positive breast cancer (ER+ BC) is the most common form of breast carcinoma accounting for approximately 70% of all diagnoses. Although ER-targeted therapies have improved survival outcomes for this BC subtype, a significant proportion of patients will ultimately develop resistance to these clinical interventions, resulting in disease recurrence. Phosphoserine aminotransferase 1 (PSAT1), an enzyme within the serine synthetic pathway (SSP), has been previously implicated in endocrine resistance. Therefore, we determined whether expression of SSP enzymes, PSAT1 or phosphoglycerate dehydrogenase (PHGDH), affects the response of ER+ BC to 4-hydroxytamoxifen (4-OHT) treatment. To investigate a clinical correlation between PSAT1, PHGDH, and endocrine resistance, we examined microarray data from ER+ patients who received tamoxifen as the sole endocrine therapy. We confirmed that higher PSAT1 and PHGDH expression correlates negatively with poorer outcomes in tamoxifen-treated ER+ BC patients. Next, we found that SSP enzyme expression and serine synthesis were elevated in tamoxifen-resistant compared to tamoxifen-sensitive ER+ BC cells in vitro. To determine relevance to endocrine sensitivity, we modified the expression of either PSAT1 or PHGDH in each cell type. Overexpression of PSAT1 in tamoxifen-sensitive MCF-7 cells diminished 4-OHT inhibition on cell proliferation. Conversely, silencing of either PSAT1 or PHGDH resulted in greater sensitivity to 4-OHT treatment in LCC9 tamoxifen-resistant cells. Likewise, the combination of a PHGDH inhibitor with 4-OHT decreased LCC9 cell proliferation. Collectively, these results suggest that overexpression of serine synthetic pathway enzymes contribute to tamoxifen resistance in ER+ BC, which can be targeted as a novel combinatorial treatment option.
Collapse
Affiliation(s)
- Stephanie Metcalf
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - Belinda J. Petri
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - Traci Kruer
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - Benjamin Green
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - Susan Dougherty
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - James L. Wittliff
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
| | - Carolyn M. Klinge
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville,
Louisville, KY, USA
| | - Brian F. Clem
- Department of Biochemistry and Molecular Genetics,
University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville,
Louisville, KY, USA
| |
Collapse
|
17
|
Regulatory mechanisms and clinical significance of vimentin in breast cancer. Biomed Pharmacother 2020; 133:111068. [PMID: 33378968 DOI: 10.1016/j.biopha.2020.111068] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/05/2023] Open
Abstract
Vimentin, a kind of intermediate filament protein III in mesenchymal cells, has become a highly researched topic around the world in recent years, as it holds complex biological functions and plays an important role in the epithelial-mesenchymal transition in the evolution of various tumors. This article reviews the biological function of vimentin and its relationship with breast cancer in order to provide novel ideas about the clinical diagnosis and targeted therapy of breast cancer.
Collapse
|
18
|
Wang S, Zhong X, Wang C, Luo H, Lin L, Sun H, Sun G, Zeng K, Zou R, Liu W, Sun N, Song H, Liu W, Zhang Q, Liao Z, Teng X, Zhou T, Sun X, Zhao Y. USP22 positively modulates ERα action via its deubiquitinase activity in breast cancer. Cell Death Differ 2020; 27:3131-3145. [PMID: 32494025 DOI: 10.1038/s41418-020-0568-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor α (ERα) is the crucial factor in ERα-positive breast cancer progression. Endocrine therapies targeting ERα signaling is one of the widely used therapeutic strategies for breast cancer. However, a large number of the patients become refractory to therapy. Abnormal expression of ERα co-regulator facilitates breast cancer development and tendency of endocrine resistance. Thus, it is necessary to discover the novel co-regulators modulating ERα action. Here, we demonstrate that histone deubiquitinase USP22 is highly expressed in breast cancer samples compared with that in the benign tissue, and high expression of USP22 was significantly associated with poorer overall survival in BCa samples. Moreover, USP22 associates with ERα to be involved in maintenance of ERα stability. USP22 enhances ERα-induced transactivation. We further provide the evidence that USP22 is recruited together with ERα to cis-regulatory elements of ERα target gene. USP22 promotes cell growth even under hypoxia condition and with the treatment of ERα antagonist in breast cancer cells. Importantly, the deubiquitination activity of USP22 is required for its functions on maintenance of ERα stability, thereby enhancing ERα action and conferring endocrine resistance in breast cancer.
Collapse
Affiliation(s)
- Shengli Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xinping Zhong
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Chunyu Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hao Luo
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Lin Lin
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hongmiao Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Ge Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Kai Zeng
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Renlong Zou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Liu
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Ning Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Huijuan Song
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wensu Liu
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Qiang Zhang
- Department of mammary gland, LiaoNing Tumor Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Zhixuan Liao
- Department of mammary gland, LiaoNing Tumor Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Tingting Zhou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yue Zhao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China. .,Department of Endocrinology and Metabolism, Institute of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
19
|
Zhou J, Xu M, Le K, Ming J, Guo H, Ruan S, Huang T. SRC Promotes Tamoxifen Resistance in Breast Cancer via Up-Regulating SIRT1. Onco Targets Ther 2020; 13:4635-4647. [PMID: 32547094 PMCID: PMC7259490 DOI: 10.2147/ott.s245749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022] Open
Abstract
Background Endocrine therapy plays a key role in estrogen receptor-positive breast cancer patients; but, tamoxifen resistance could be a real difficulty for these patients. Several attempts have been made to explore the mechanism and new therapies for these patients. We intend to clarify the expression change of SRC and SIRT1 in tamoxifen-resistant breast cancer cells and explore their functions on tamoxifen resistance. Methods SRC and SIRT1 expressions were analyzed by RNA sequencing, qPCR and Western blotting. Loss and gain of function of SRC and SIRT1 were utilized to indicate their oncogenic roles in tamoxifen resistance in vitro and in vivo. Kaplan–Meier analysis and receiver operating characteristic curve were used to evaluate the survival and the predicted effects of SRC and SIRT1 on patients’ prognosis. Results High expressions of SRC and/or SIRT1 were found in tamoxifen-resistant cells and related to poor overall survival (p<0.05 for SRC, p<0.001 for SIRT1, p<0.001 for SRC and SIRT1) and cancer-specific survival (p<0.05 for SRC, p<0.01 for SIRT1, p<0.01 for SRC and SIRT1) of tamoxifen-treated breast cancer patients. Down-regulation of SRC (p<0.01) or SIRT1 (p<0.05) separately reversed the resistance to tamoxifen and the minimal concentration of SRC inhibitor KX-01 (p<0.05) or SIRT1 inhibitor EX527 (p<0.001) could also suppress cell proliferation. The expression level of SIRT1 was positively correlated with that of SRC. Overexpression of SRC significantly promotes the cell resistance to tamoxifen inhibited by SIRT1 (p<0.01). In vivo experiments confirmed the effects of SRC on tumor growth by over- or down-regulating SRC expression (p<0.001 and p<0.001, respectively). Conclusion SRC and SIRT1 are both up-regulated in tamoxifen-resistant breast cancer cells and related to a poor prognosis in tamoxifen-treated breast cancer. Moreover, SRC could promote tamoxifen resistance by up-regulating SIRT1. SRC and SIRT1 might be novel therapeutic targets in tamoxifen-resistant breast cancer and the interaction between SRC and SIRT1 needs to be further explored.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Kehao Le
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Hui Guo
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Shengnan Ruan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
20
|
Occhipinti G, Romagnoli E, Santoni M, Cimadamore A, Sorgentoni G, Cecati M, Giulietti M, Battelli N, Maccioni A, Storti N, Cheng L, Principato G, Montironi R, Piva F. Sequential or Concomitant Inhibition of Cyclin-Dependent Kinase 4/6 Before mTOR Pathway in Hormone-Positive HER2 Negative Breast Cancer: Biological Insights and Clinical Implications. Front Genet 2020; 11:349. [PMID: 32351542 PMCID: PMC7174681 DOI: 10.3389/fgene.2020.00349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/23/2020] [Indexed: 12/25/2022] Open
Abstract
About 75% of all breast cancers are hormone receptor-positive (HR+). However, the efficacy of endocrine therapy is limited due to the high rate of either pre-existing or acquired resistance. In this work we reconstructed the pathways around estrogen receptor (ER), mTOR, and cyclin D in order to compare the effects of CDK4/6 and PI3K/AKT/mTOR inhibitors. A positive feedback loop links mTOR and ER that support each other. We subsequently considered whether a combined or sequential inhibition of CDK4/6 and PI3K/AKT/mTOR could ensure better results. Studies indicate that inhibition of CDK4/6 activates mTOR as an escape mechanism to ensure cell proliferation. In literature, the little evidence dealing with this topic suggests that pre-treatment with mTOR pathway inhibitors could prevent or delay the onset of CDK4/6 inhibitor resistance. Additional studies are needed in order to find biomarkers that can identify patients who will develop this resistance and in whom the sensitivity to CDK4/6 inhibitors can be restored.
Collapse
Affiliation(s)
- Giulia Occhipinti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | | | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Monia Cecati
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | | | - Nadia Storti
- Direzione Sanitaria Azienda Sanitaria Unica Regionale, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
21
|
Santoni M, Occhipinti G, Romagnoli E, Miccini F, Scoccia L, Giulietti M, Principato G, Saladino T, Piva F, Battelli N. Different Cardiotoxicity of Palbociclib and Ribociclib in Breast Cancer: Gene Expression and Pharmacological Data Analyses, Biological Basis, and Therapeutic Implications. BioDrugs 2020; 33:613-620. [PMID: 31529317 DOI: 10.1007/s40259-019-00382-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most frequent tumor in women. The recent advent of cyclin-dependent kinase (CDK) 4/6 inhibitors palbociclib and ribociclib has represented a major step forward for patients with hormone receptor-positive breast cancer. These two agents have showed similar efficacy in terms of breast cancer outcome but different cardiotoxic effects. In particular, ribociclib, but not palbociclib, has been associated with QT interval prolongation, and the mechanisms underlying this event are still unclear. In order to clarify such difference, we matched the candidate genes associated with QT interval prolongation with genes whose expression is altered following palbociclib or ribociclib treatment. We also investigated whether pharmacokinetic and pharmacodynamic characteristics, such as IC50 (hERG) [concentration of drug producing 50% inhibition (human ether-à-go-go related gene)] and maximum concentration (Cmax), could justify the different effects on QT interval prolongation. Our results show that ribociclib, but not palbociclib, could act by down-regulating the expression of KCNH2 (encoding for potassium channel hERG) and up-regulating SCN5A and SNTA1 (encoding for sodium channels Nav1.5 and syntrophin-α1, respectively), three genes associated with long QT syndrome. Consistent with the cardiotoxicity induced by ribociclib, its IC50 (hERG)/free concentration (Cmax free) ratio is closer to the safety threshold than that of palbociclib. In summary, we hypothesize that the different cardiotoxicity associated with ribociclib and palbociclib could be due to the alteration of potassium and sodium channels induced by ribociclib. A better comprehension of the mechanisms of cardiac channelopathies and drug-induced QT interval prolongation will be fundamental to avoid serious and potentially lethal adverse events and, as a consequence, optimize the management of breast cancer patients.
Collapse
Affiliation(s)
- Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | - Giulia Occhipinti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | | | - Francesca Miccini
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | | | - Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy
| | - Tiziana Saladino
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Monte d'Ago, 60131, Ancona, Italy.
| | - Nicola Battelli
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| |
Collapse
|
22
|
Endocrine resistant breast cancer cells with loss of ERα expression retain proliferative ability by reducing caspase7-mediated HDAC3 cleavage. Cell Oncol (Dordr) 2019; 43:65-80. [DOI: 10.1007/s13402-019-00439-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
|
23
|
Investigating circulating tumor cells and distant metastases in patient-derived orthotopic xenograft models of triple-negative breast cancer. Breast Cancer Res 2019; 21:98. [PMID: 31462307 PMCID: PMC6714238 DOI: 10.1186/s13058-019-1182-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) represent a temporal "snapshot" of a patient's cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer. METHODS We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice. RESULTS Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency. CONCLUSION PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.
Collapse
|
24
|
Whitman NA, Lin ZW, Kenney RM, Albertini L, Lockett MR. Hypoxia differentially regulates estrogen receptor alpha in 2D and 3D culture formats. Arch Biochem Biophys 2019; 671:8-17. [PMID: 31163125 PMCID: PMC6688900 DOI: 10.1016/j.abb.2019.05.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/24/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Hypoxia is a common feature in solid tumors. Clinical samples show a positive correlation between the expression of the hypoxia-inducible factor HIF-1α and estrogen receptor alpha (ERα) and a negative correlation between HIF-1α and hormone sensitivity. Results from monolayer cultures are in contention with clinical observations, showing that ER (+) cell lines no longer express ERα under hypoxic conditions (1% O2). Here, we compared the impact of hypoxia on the ERα signaling pathway for T47D cells in a 2D and 3D culture format. In the 2D format, the cells were cultured as monolayers. In the 3D format, paper-based scaffolds supported cells suspended in a collagen matrix. Using ELISA, Western blot, and immunofluorescence measurements, we show that hypoxia differentially regulates ERα protein levels in a culture environment-dependent manner. In the 2D format, the protein levels are significantly decreased in hypoxia. In the 3D format, the protein levels are maintained in hypoxia. Hypoxia reduced ERα transcriptional activation in both culture formats. These results highlight the importance of considering tissue dimensionality for in vitro studies. They also show that ERα protein levels in hypoxia are not an accurate indicator of ERα transcriptional activity, and confirm that a positive stain for ERα in a clinical sample may not necessarily indicate hormone sensitivity.
Collapse
Affiliation(s)
- Nathan A Whitman
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Zhi-Wei Lin
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Rachael M Kenney
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Leonardo Albertini
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA
| | - Matthew R Lockett
- Department of Chemistry, University of North Carolina at Chapel Hill, Kenan and Caudill Laboratories, Chapel Hill, NC, 27599-3290, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill, NC, 27599-7295, USA.
| |
Collapse
|
25
|
Gu L, Saha ST, Thomas J, Kaur M. Targeting cellular cholesterol for anticancer therapy. FEBS J 2019; 286:4192-4208. [DOI: 10.1111/febs.15018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/30/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Liang Gu
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Sourav Taru Saha
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Jodie Thomas
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| |
Collapse
|
26
|
Zhu S, Wei L, Lin G, Tong Y, Zhang J, Jiang X, He Q, Lu X, Zhu DD, Chen YQ. Metabolic Alterations Induced by Kudingcha Lead to Cancer Cell Apoptosis and Metastasis Inhibition. Nutr Cancer 2019; 72:696-707. [DOI: 10.1080/01635581.2019.1645865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Shenglong Zhu
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
- The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lengyun Wei
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Guangxiao Lin
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yuelin Tong
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jingwei Zhang
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Xuan Jiang
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Qingwen He
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Xuyang Lu
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Dou Dou Zhu
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Yong Q. Chen
- School of Wuxi Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| |
Collapse
|
27
|
Discovery of a novel cathepsin inhibitor with dual autophagy-inducing and metastasis-inhibiting effects on breast cancer cells. Bioorg Chem 2019; 84:239-253. [DOI: 10.1016/j.bioorg.2018.11.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/05/2018] [Accepted: 11/17/2018] [Indexed: 11/17/2022]
|
28
|
Hu J, Hu B, Wang M, Xu F, Miao B, Yang CY, Wang M, Liu Z, Hayes DF, Chinnaswamy K, Delproposto J, Stuckey J, Wang S. Discovery of ERD-308 as a Highly Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Estrogen Receptor (ER). J Med Chem 2019; 62:1420-1442. [DOI: 10.1021/acs.jmedchem.8b01572] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Cuesta R, Berman AY, Alayev A, Holz MK. Estrogen receptor α promotes protein synthesis by fine-tuning the expression of the eukaryotic translation initiation factor 3 subunit f (eIF3f). J Biol Chem 2018; 294:2267-2278. [PMID: 30573685 DOI: 10.1074/jbc.ra118.004383] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/18/2018] [Indexed: 01/09/2023] Open
Abstract
Approximately two thirds of all breast cancer cases are estrogen receptor (ER)-positive. The treatment of this breast cancer subtype with endocrine therapies is effective in the adjuvant and recurrent settings. However, their effectiveness is compromised by the emergence of intrinsic or acquired resistance. Thus, identification of new molecular targets can significantly contribute to the development of novel therapeutic strategies. In recent years, many studies have implicated aberrant levels of translation initiation factors in cancer etiology and provided evidence that identifies these factors as promising therapeutic targets. Accordingly, we observed reduced levels of the eIF3 subunit eIF3f in ER-positive breast cancer cells compared with ER-negative cells, and determined that low eIF3f levels are required for proper proliferation and survival of ER-positive MCF7 cells. The expression of eIF3f is tightly controlled by ERα at the transcriptional (genomic pathway) and translational (nongenomic pathway) level. Specifically, estrogen-bound ERα represses transcription of the EIF3F gene, while promoting eIF3f mRNA translation. To regulate translation, estrogen activates the mTORC1 pathway, which enhances the binding of eIF3 to the eIF4F complex and, consequently, the assembly of the 48S preinitiation complexes and protein synthesis. We observed preferential translation of mRNAs with highly structured 5'-UTRs that usually encode factors involved in cell proliferation and survival (e.g. cyclin D1 and survivin). Our results underscore the importance of estrogen-ERα-mediated control of eIF3f expression for the proliferation and survival of ER-positive breast cancer cells. These findings may provide rationale for the development of new therapies to treat ER-positive breast cancer.
Collapse
Affiliation(s)
- Rafael Cuesta
- From the Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595
| | - Adi Y Berman
- the Department of Biology, Yeshiva University, New York, New York 10016, and
| | - Anya Alayev
- the Department of Biology, Yeshiva University, New York, New York 10016, and
| | - Marina K Holz
- From the Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595, .,Albert Einstein Cancer Center, Bronx, New York 10461
| |
Collapse
|
30
|
Chanchan G, Xiangyu S, Fangfang S, Yan C, Xiaoyi G. The efficacy and safety of targeted therapy plus fulvestrant in postmenopausal women with hormone-receptor positive advanced breast cancer: A meta-analysis of randomized-control trials. PLoS One 2018; 13:e0204202. [PMID: 30235292 PMCID: PMC6157814 DOI: 10.1371/journal.pone.0204202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 09/05/2018] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of targeted therapy plus fulvestrant for postmenopausal patients with hormone receptor-positive advanced breast cancer. METHODS Pubmed, Embase and Web of Science databases were systematically searched on February 26, 2018. Eligible studies were screened according to selection criteria, and two reviewers independently extracted outcome data which included progression-free survival, overall survival, objective response rate, clinical benefit rate and toxicities. RevMan 5.3 and STATA 11.0 software were used to conduct meta-analysis. RESULTS Thirteen articles including twelve randomized-control trials fulfilled selection criteria. There was no evidence regarding the existence of publication bias and high-risk bias of quality in the selected studies. In previously endocrine therapy-treated postmenopausal patients with hormone-receptor positive advanced breast cancer, the PFS (HR = 0.77, 95%CI: 0.66-0.91) and ORR (RR = 1.78, 95%CI: 1.35-2.34) of combination therapy group were significantly higher than that from fulvestrant monotherapy group. Besides, a statistically significant difference in PFS was found across the two arms in postmenopausal women with PIK3CA-mutant ctDNA tumor (HR = 0.52, 95% CI: 0.39-0.69). Moreover, the risk of adverse events (RR = 1.09, 95%CI: 1.05-1.13), CTCAE≥3 (RR = 1.97, 95%CI: 1.49-2.60) and discontinuation due to adverse events (RR = 4.91, 95%CI: 3.37-7.15) were also significantly different between two treatment groups. Sensitivity analysis showed PLOMA-3 trial was an important factor of heterogeneity. DISCUSSION Even though the combination of targeted therapy plus fulvestrant improved PFS and increased ORR in advanced breast cancer patients, the toxicities of combination therapy were also higher than fulvestrant monotherapy. Further studies related to inhibitors targeting the specific signaling pathway or receptors are urgently needed, and more efforts concerning precision medicine of targeted therapy plus endocrine therapy should be taken to improve the clinical benefits.
Collapse
Affiliation(s)
- Gao Chanchan
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Su Xiangyu
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Shi Fangfang
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Chen Yan
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| | - Gu Xiaoyi
- Department of Oncology, Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
31
|
Valproic acid as an adjunctive therapeutic agent for the treatment of breast cancer. Eur J Pharmacol 2018; 835:61-74. [DOI: 10.1016/j.ejphar.2018.07.057] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023]
|
32
|
Chen Y, Zhang Y. Application of the CRISPR/Cas9 System to Drug Resistance in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700964. [PMID: 29938175 PMCID: PMC6010891 DOI: 10.1002/advs.201700964] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/14/2018] [Indexed: 05/29/2023]
Abstract
Clinical evidence indicates that drug resistance is a great obstacle in breast cancer therapy. It renders the disease uncontrollable and causes high mortality. Multiple mechanisms contribute to the development of drug resistance, but the underlying cause is usually a shift in the genetic composition of tumor cells. It is increasingly feasible to engineer the genome with the clustered regularly interspaced short palindromic repeats (CRISPR)/associated (Cas)9 technology recently developed, which might be advantageous in overcoming drug resistance. This article discusses how the CRISPR/Cas9 system might revert resistance gene mutations and identify potential resistance targets in drug-resistant breast cancer. In addition, the challenges that impede the clinical applicability of this technology and highlight the CRISPR/Cas9 systems are presented. The CRISPR/Cas9 system is poised to play an important role in preventing drug resistance in breast cancer therapy and will become an essential tool for personalized medicine.
Collapse
Affiliation(s)
- Yinnan Chen
- School of Molecular SciencesArizona State UniversityTempeAZ85287USA
| | - Yanmin Zhang
- School of PharmacyHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxi Province710061P. R. China
- State Key Laboratory of Shaanxi for Natural Medicines Research and EngineeringXi'an710061P. R. China
- Shaanxi Institute of International Trade & CommenceXianyang712046P. R. China
| |
Collapse
|
33
|
Post AEM, Smid M, Nagelkerke A, Martens JWM, Bussink J, Sweep FCGJ, Span PN. Interferon-Stimulated Genes Are Involved in Cross-resistance to Radiotherapy in Tamoxifen-Resistant Breast Cancer. Clin Cancer Res 2018; 24:3397-3408. [PMID: 29661777 DOI: 10.1158/1078-0432.ccr-17-2551] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/07/2018] [Accepted: 04/10/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Treatment resistance is the main cause of adverse disease outcome in breast cancer patients. Here, we aimed to investigate common features in tamoxifen-resistant and radioresistant breast cancer, as tamoxifen-resistant breast cancer cells are cross-resistant to irradiation in vitroExperimental Design: RNA sequencing of tamoxifen-resistant and radioresistant breast cancer cells was performed and validated by quantitative PCR. Pathways were further investigated in vitro and in breast cancer patient cohorts to establish their relation with treatment resistance.Results: Both tamoxifen-resistant and radioresistant breast cancer cells had increased expression levels of genes involved in type I IFN signaling compared with nonresistant cells. IFN-stimulated genes (ISG) were induced in a dose-dependent and time-dependent manner after tamoxifen treatment and irradiation. Tamoxifen treatment also led to ssDNA presence in the cytoplasm, which is known to induce expression of ISGs, a phenomenon that has already been described for irradiation. Moreover, in a breast cancer patient cohort, high expression levels of ISGs were found in the primary tumor in around half of the patients. This was associated with a tumor-infiltrating lymphocyte (TIL) expression signature, although the ISGs were also expressed by the tumor cells themselves. Importantly, the expression of ISGs correlated with outcome in breast cancer patients treated with adjuvant tamoxifen or radiotherapy, but not in systemically untreated patients or chemotherapy-treated patients.Conclusions: Our data indicate that expression of ISGs by tumor cells is involved in acquired, treatment-induced resistance to tamoxifen and radiotherapy, and might play a role in intrinsic resistance via interaction with TILs. Clin Cancer Res; 24(14); 3397-408. ©2018 AACR.
Collapse
Affiliation(s)
- Annemarie E M Post
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands. .,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Anika Nagelkerke
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands.,Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud university medical center, Nijmegen, the Netherlands
| | - Paul N Span
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
34
|
Yang L, Lv Z, Xia W, Zhang W, Xin Y, Yuan H, Chen Y, Hu X, Lv Y, Xu Q, Weng X, Ni C. The effect of aspirin on circulating tumor cells in metastatic colorectal and breast cancer patients: a phase II trial study. Clin Transl Oncol 2017; 20:912-921. [PMID: 29243075 DOI: 10.1007/s12094-017-1806-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE Aspirin could reduce the risk of cancer metastasis. Circulating tumor cells (CTCs) are a key factor of cancer metastasis, but no evidence has revealed how aspirin affects CTCs and its epithelial-mesenchymal transition (EMT). Here, we conducted a clinical trial to investigate how aspirin affects CTCs in metastatic colorectal cancer (MCC) and breast cancer patients (MBC). METHODS The trial is retrospective registered at clinicaltrials.gov (NCT02602938). The eligible patients are given 100 mg aspirin q.d. for 8 weeks, and CTCs are evaluated at baseline, 4 and 8 weeks for absolute number, phenotype (epithelial type, E+, mesenchymal type, M+, and biophenotypic type, B+), and vimentin expression. RESULTS Data on 21 MCC and 19 MBC patients are analyzed, and it revealed that the CTC numbers decreased with aspirin treatment in MCC (p < 0.001) but not MBC (p = 0.0532); besides, ratio of E+ CTCs increased (p = 0.037) and M+ CTCs decreased at 2 months in MCC (p = 0.013), but neither the ratio of E+ or M+ CTCs changes significantly in MBC; vimentin expression of M+ CTCs is higher than E+ and B+ CTCs either in MBC or MCC patients at baseline (p < 0.01); and aspirin suppresses the vimentin expression in M+ (p = 0.002)and B+ (p = 0.006) CTCs of MCC and M+ CTCs of MBC (p = 0.004); besides it find vimentin expression in B+ (p = 0.004) or M+ (p < 0.001), CTCs are markedly decreased in patients with total CTC numbers declined. CONCLUSION Aspirin could decrease CTCs numbers and block EMT transition in MCC patients and part of MBC patients.
Collapse
Affiliation(s)
- L Yang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medicine College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Z Lv
- Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - W Xia
- Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - W Zhang
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People's Republic of China
| | - Y Xin
- Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - H Yuan
- Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Y Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - X Hu
- Department of Anus and Intestine Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Y Lv
- SurExam Bio-Tech, Guangzhou Technology Innovation Base, Science City, Guangzhou, People's Republic of China
| | - Q Xu
- Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - X Weng
- Department of General Surgery, Central Hospital of Haining, Zhejiang, 310000, People's Republic of China
| | - C Ni
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medicine College, Hangzhou, 310014, Zhejiang, People's Republic of China. .,Department of Breast and Thyroid Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China.
| |
Collapse
|
35
|
Fang Z, Yi Y, Shi G, Li S, Chen S, Lin Y, Li Z, He Z, Li W, Zhong S. Role of Brf1 interaction with ERα, and significance of its overexpression, in human breast cancer. Mol Oncol 2017; 11:1752-1767. [PMID: 28972307 PMCID: PMC5709663 DOI: 10.1002/1878-0261.12141] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/06/2017] [Accepted: 09/17/2017] [Indexed: 02/05/2023] Open
Abstract
TFIIB-related factor 1 (Brf1) modulates the transcription of RNA Pol III genes (polymerase-dependent genes). Upregulation of Pol III genes enhances tRNA and 5S RNA production and increases the translational capacity of cells to promote cell transformation and tumor development. However, the significance of Brf1 overexpression in human breast cancer (HBC) remains to be investigated. Here, we investigate whether Brf1 expression is increased in the samples of HBC, and we explore its molecular mechanism and the significance of Brf1 expression in HBC. Two hundred and eighteen samples of HBC were collected to determine Brf1 expression by cytological and molecular biological approaches. We utilized colocalization, coimmunoprecipitation, and chromatin immunoprecipitation methods to explore the interaction of Brf1 with estrogen receptor alpha (ERα). We determined how Brf1 and ERα modulate Pol III genes. The results indicated that Brf1 is overexpressed in most cases of HBC, which is associated with an ER-positive status. The survival period of the cases with high Brf1 expression is significantly longer than those with low levels of Brf1 after hormone treatment. ERα mediates Brf1 expression. Brf1 and ERα are colocalized in the nucleus. These results indicate an interaction between Brf1 and ERα, which synergistically regulates the transcription of Pol III genes. Inhibition of ERα by its siRNA or tamoxifen reduces cellular levels of Brf1 and Pol III gene expression and decreases the rate of colony formation of breast cancer cells. Together, these studies demonstrate that Brf1 is a good biomarker for the diagnosis and prognosis of HBC. This interaction of Brf1 with ERα and Brf1 itself are potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Zeng Fang
- Laboratory of General SurgeryFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Yunfeng Yi
- Department of Cardiothoracic SurgeryXiamen University Affiliated Southeast HospitalZhangzhouChina
| | - Ganggang Shi
- Department of PharmacologyShantou University Medical CollegeChina
| | - Songqi Li
- Laboratory of General SurgeryFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Songlin Chen
- Department of Cardiothoracic SurgeryXiamen University Affiliated Southeast HospitalZhangzhouChina
| | - Ying Lin
- Laboratory of General SurgeryFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Zhi Li
- Laboratory of General SurgeryFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Zhimin He
- Cancer Center of Guangzhou Medical UniversityGuangzhouChina
| | - Wen Li
- Laboratory of General SurgeryFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Shuping Zhong
- Department of PharmacologyShantou University Medical CollegeChina
- Cancer Center of Guangzhou Medical UniversityGuangzhouChina
- Department of Biochemistry and Molecular MedicineKeck School of MedicineUniversity of Southern CaliforniaLos AngeleCAUSA
| |
Collapse
|
36
|
Ito K, Park SH, Katsyv I, Zhang W, De Angelis C, Schiff R, Irie HY. PTK6 regulates growth and survival of endocrine therapy-resistant ER+ breast cancer cells. NPJ Breast Cancer 2017; 3:45. [PMID: 29167821 PMCID: PMC5694002 DOI: 10.1038/s41523-017-0047-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 09/25/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022] Open
Abstract
The non-receptor tyrosine kinase, PTK6/BRK, is highly expressed in multiple tumor types, including prostate, ovarian, and breast cancers, and regulates oncogenic phenotypes such as proliferation, migration, and survival. PTK6 inhibition also overcomes targeted therapy resistance of HER2+ breast cancer. Although PTK6 is highly expressed in ER+ Luminal breast cancers, the role of PTK6 in this subtype has not been elucidated. In this study, we investigated the functions of PTK6 in ER+ Luminal breast cancer cells, including those that are relatively resistant to estrogen deprivation or targeted endocrine therapies used in the treatment of ER+ cancers. Enhanced expression of PTK6 in ER+ breast cancer cells enhances growth of ER+ breast cancer cells, including tamoxifen-treated cells. Downregulation of PTK6 in ER+ breast cancer cells, including those resistant to tamoxifen, fulvestrant, and estrogen deprivation, induces apoptosis, as evidenced by increased levels of cleaved PARP, and an increase in the AnnexinV+ population. PTK6 downregulation impairs growth of these cells in 3D MatrigelTM cultures, and virtually abrogates primary tumor growth of both tamoxifen-sensitive and resistant MCF-7 xenografts. Finally, we show that p38 MAPK activation is critical for PTK6 downregulation-induced apoptosis, a mechanism that we previously reported for survival of HER2+ breast cancer cells, highlighting conserved mechanisms of survival regulation by PTK6 across breast cancer subtypes. In conclusion, our studies elucidate critical functions of PTK6 in ER+ Luminal breast cancers and support PTK6 as an attractive therapeutic target for ER+ breast cancers. Drugs that target a tumor-promoting enzyme called protein tyrosine kinase 6 (PTK6) could help treat hormone-receptor positive breast cancer. A team led by Hanna Irie from the Icahn School of Medicine at Mount Sinai in New York, NY, USA, investigated the role of PTK6, also known as breast tumor kinase, in breast cancer cells that grow in response to the hormone estrogen. They boosted the enzyme’s expression in estrogen receptor-expressing breast cancer cells and saw enhanced growth. Conversely, downregulating PTK6 levels led to cell death, including in tumor cells that were resistant to tamoxifen
and other therapies commonly used to treat estrogen-receptor positive breast cancer. The researchers showed that this effect was dependent on the activation of a signaling pathway previously found to be important in another subtype of breast cancer, highlighting a conserved mechanism of cell survival regulation by PTK6.
Collapse
Affiliation(s)
- Koichi Ito
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY USA
| | - Sun Hee Park
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY USA
| | - Igor Katsyv
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Carmine De Angelis
- Lester & Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX USA.,Department of Medicine, Baylor College of Medicine, Houston, TX USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX USA.,Department of Medicine, Baylor College of Medicine, Houston, TX USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| | - Hanna Y Irie
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY USA.,Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Ave, New York, NY USA
| |
Collapse
|
37
|
Beije N, Sieuwerts AM, Kraan J, Van NM, Onstenk W, Vitale SR, van der Vlugt-Daane M, Dirix LY, Brouwer A, Hamberg P, de Jongh FE, Jager A, Seynaeve CM, Jansen MPHM, Foekens JA, Martens JWM, Sleijfer S. Estrogen receptor mutations and splice variants determined in liquid biopsies from metastatic breast cancer patients. Mol Oncol 2017; 12:48-57. [PMID: 29063679 PMCID: PMC5748489 DOI: 10.1002/1878-0261.12147] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/24/2017] [Accepted: 10/07/2017] [Indexed: 01/09/2023] Open
Abstract
Mutations and splice variants in the estrogen receptor (ER) gene, ESR1, may yield endocrine resistance in metastatic breast cancer (MBC) patients. These putative endocrine resistance markers are likely to emerge during treatment, and therefore, its detection in liquid biopsies, such as circulating tumor cells (CTCs) and cell‐free DNA (cfDNA), is of great interest. This research aimed to determine whether ESR1 mutations and splice variants occur more frequently in CTCs of MBC patients progressing on endocrine treatment. In addition, the presence of ESR1 mutations was evaluated in matched cfDNA and compared to CTCs. CellSearch‐enriched CTC fractions (≥5/7.5 mL) of two MBC cohorts were evaluated, namely (a) patients starting first‐line endocrine therapy (n = 43, baseline cohort) and (b) patients progressing on any line of endocrine therapy (n = 40, progressing cohort). ESR1 hotspot mutations (D538G and Y537S/N/C) were evaluated in CTC‐enriched DNA using digital PCR and compared with matched cfDNA (n = 18 baseline cohort; n = 26 progressing cohort). Expression of ESR1 full‐length and 4 of its splice variants (∆5, ∆7, 36 kDa, and 46 kDa) was evaluated in CTC‐enriched mRNA. It was observed that in the CTCs, the ESR1 mutations were not enriched in the progressing cohort (8%), when compared with the baseline cohort (5%) (P = 0.66). In the cfDNA, however, ESR1 mutations were more prevalent in the progressing cohort (42%) than in the baseline cohort (11%) (P = 0.04). Three of the same mutations were observed in both CTCs and cfDNA, 1 mutation in CTCs only, and 11 in cfDNA only. Only the ∆5 ESR1 splice variant was CTC‐specific expressed, but was not enriched in the progressing cohort. In conclusion, sensitivity for detecting ESR1 mutations in CTC‐enriched fractions was lower than for cfDNA. ESR1 mutations detected in cfDNA, rarely present at the start of first‐line endocrine therapy, were enriched at progression, strongly suggesting a role in conferring endocrine resistance in MBC.
Collapse
Affiliation(s)
- Nick Beije
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anieta M Sieuwerts
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jaco Kraan
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ngoc M Van
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wendy Onstenk
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Silvia R Vitale
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Clinical and Molecular Medicine, University of Catania, Italy
| | - Michelle van der Vlugt-Daane
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Luc Y Dirix
- Translational Cancer Research Unit, Department of Medical Oncology, Oncology Center GZA Hospital Sint Augustinus, Antwerp, Belgium
| | - Anja Brouwer
- Translational Cancer Research Unit, Department of Medical Oncology, Oncology Center GZA Hospital Sint Augustinus, Antwerp, Belgium
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - Felix E de Jongh
- Department of Internal Medicine, Ikazia Hospital, Rotterdam, The Netherlands
| | - Agnes Jager
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Caroline M Seynaeve
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maurice P H M Jansen
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John A Foekens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John W M Martens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Beije N, Sieuwerts AM, Kraan J, Van NM, Onstenk W, Vitale SR, van der Vlugt-Daane M, Dirix LY, Brouwer A, Hamberg P, de Jongh FE, Jager A, Seynaeve CM, Jansen MPHM, Foekens JA, Martens JWM, Sleijfer S. Estrogen receptor mutations and splice variants determined in liquid biopsies from metastatic breast cancer patients. Mol Oncol 2017. [PMID: 29063679 DOI: 10.1002/1878‐0261.12147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mutations and splice variants in the estrogen receptor (ER) gene, ESR1, may yield endocrine resistance in metastatic breast cancer (MBC) patients. These putative endocrine resistance markers are likely to emerge during treatment, and therefore, its detection in liquid biopsies, such as circulating tumor cells (CTCs) and cell-free DNA (cfDNA), is of great interest. This research aimed to determine whether ESR1 mutations and splice variants occur more frequently in CTCs of MBC patients progressing on endocrine treatment. In addition, the presence of ESR1 mutations was evaluated in matched cfDNA and compared to CTCs. CellSearch-enriched CTC fractions (≥5/7.5 mL) of two MBC cohorts were evaluated, namely (a) patients starting first-line endocrine therapy (n = 43, baseline cohort) and (b) patients progressing on any line of endocrine therapy (n = 40, progressing cohort). ESR1 hotspot mutations (D538G and Y537S/N/C) were evaluated in CTC-enriched DNA using digital PCR and compared with matched cfDNA (n = 18 baseline cohort; n = 26 progressing cohort). Expression of ESR1 full-length and 4 of its splice variants (∆5, ∆7, 36 kDa, and 46 kDa) was evaluated in CTC-enriched mRNA. It was observed that in the CTCs, the ESR1 mutations were not enriched in the progressing cohort (8%), when compared with the baseline cohort (5%) (P = 0.66). In the cfDNA, however, ESR1 mutations were more prevalent in the progressing cohort (42%) than in the baseline cohort (11%) (P = 0.04). Three of the same mutations were observed in both CTCs and cfDNA, 1 mutation in CTCs only, and 11 in cfDNA only. Only the ∆5 ESR1 splice variant was CTC-specific expressed, but was not enriched in the progressing cohort. In conclusion, sensitivity for detecting ESR1 mutations in CTC-enriched fractions was lower than for cfDNA. ESR1 mutations detected in cfDNA, rarely present at the start of first-line endocrine therapy, were enriched at progression, strongly suggesting a role in conferring endocrine resistance in MBC.
Collapse
Affiliation(s)
- Nick Beije
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anieta M Sieuwerts
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jaco Kraan
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ngoc M Van
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wendy Onstenk
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Silvia R Vitale
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Clinical and Molecular Medicine, University of Catania, Italy
| | - Michelle van der Vlugt-Daane
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Luc Y Dirix
- Translational Cancer Research Unit, Department of Medical Oncology, Oncology Center GZA Hospital Sint Augustinus, Antwerp, Belgium
| | - Anja Brouwer
- Translational Cancer Research Unit, Department of Medical Oncology, Oncology Center GZA Hospital Sint Augustinus, Antwerp, Belgium
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis, Rotterdam, The Netherlands
| | - Felix E de Jongh
- Department of Internal Medicine, Ikazia Hospital, Rotterdam, The Netherlands
| | - Agnes Jager
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Caroline M Seynaeve
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maurice P H M Jansen
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John A Foekens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John W M Martens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Long non-coding RNA00544 serves as a potential novel predictive and prognostic marker for HR+ HER2- subtype breast cancer. Sci Rep 2017; 7:12382. [PMID: 28959047 PMCID: PMC5620366 DOI: 10.1038/s41598-017-11066-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 07/28/2017] [Indexed: 12/31/2022] Open
Abstract
Luminal breast cancers (BC) account for majority of breast cancer. Due to its heterogeneity and the development of treatment resistance, luminal BC patients can vary substantially. Long noncoding RNAs (lncRNAs), as we known, is involved in breast cancer progression. Here, we aim to identify the lncRNAs which are involved in the particular type luminal BC progression. By Gene Chips analysis, we found a novel lncRNA00544, which was highly expressed in the metastatic axillary nodes compared with corresponding luminal BC tissues (fold change = 2.26, P = 0.043). This result was confirmed in luminal BC cell lines (p = 0.0113) and 49 paired breast cancer samples compared with in corresponding controls (p = 0.011). Furthermore, Kaplan-Meier survival curves of 373 breast cancer patients indicated that disease-free survival was significantly poor in breast cancer patients with high lncRNA00544 expression (p < 0.001). Univariate and multivariate Cox regression analyses showed that lncRNA00544 was a significant independent prognostic biomarker in luminal BC patients. Further analysis showed that the prognosis of high lncRNA00544 expression in breast cancer patients was actually related to HR + HER2- subtype. Together, our studies indicate that lncRNA00544 may represent a novel predictive and prognostic indicator in luminal BC patients.
Collapse
|
40
|
López-Tarruella S, Jerez Y, Márquez-Rodas I, Echavarria I, Martin M. Ribociclib for the treatment of advanced hormone receptor-positive, HER2-negative breast cancer. Future Oncol 2017; 13:2137-2149. [PMID: 28758424 DOI: 10.2217/fon-2017-0183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CDK4/6 inhibitors are a promising new class of drugs for hormone-receptor-positive breast cancer and have been shown to overcome and delay hormone resistance in advanced breast cancer. Ribociclib, a selective oral inhibitor of CDK4/6, was approved by the US FDA for first-line treatment of hormone-receptor-positive/HER2-negative metastatic breast cancer. This review summarizes the clinical evidence available for ribociclib, from preclinical data to the pivotal studies, with a special focus on toxicity and its management. In addition, this article reviews potential new combinations under study, as well as ongoing clinical trials both in the metastatic and early setting. Finally, this review compares ribociclib activity and toxicity with those of the drugs of the same class (palbociclib and abemaciclib).
Collapse
Affiliation(s)
- Sara López-Tarruella
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Yolanda Jerez
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Iván Márquez-Rodas
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Isabel Echavarria
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), CiberOnc, Madrid, Spain
| | - Miguel Martin
- Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Universidad Complutense, CiberOnc, GEICAM, Madrid, Spain
| |
Collapse
|
41
|
Singh J, Singh R, Gupta P, Rai S, Ganesher A, Badrinarayan P, Sastry GN, Konwar R, Panda G. Targeting progesterone metabolism in breast cancer with l-proline derived new 14-azasteroids. Bioorg Med Chem 2017; 25:4452-4463. [PMID: 28693914 DOI: 10.1016/j.bmc.2017.06.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/05/2017] [Accepted: 06/17/2017] [Indexed: 02/08/2023]
Abstract
Breast cancer cell proliferation is promoted by a variety of mitogenic signals. Classically estrogen is considered as most predominant mitogenic signal in hormone-dependent breast cancer and progesterone is primarily considered to have protective effect. However, it is suggested that some progesterone metabolite may promote breast cancer and progesterone metabolites like 5α-pregnane and 4-pregnene could serve as regulators of estrogen-responsiveness of breast cancer cells. Here, we estimated the potential of alternate targeting of breast cancer via progesterone signalling. l-Proline derived novel 14-azasteroid compounds were screened against MCF-7 and MDA-MB-231 cell lines using MTT assay. In silico studies, cell cycle, Annexin-V-FITC/PI, JC-1 mitochondrial assay, ROS analysis were performed to analyse the impact of hit compound 3b on breast cancer cells. Further, we analysed the impact of hit 3b on the progesterone, its metabolites and enzymes responsible for the conversion of progesterone and its metabolites using ELISA. Data suggests that compound 3b binds and down regulates of 5α-reductase by specifically inhibiting production of progesterone metabolites that are capable of promoting breast cancer proliferation, epithelial mesenchymal transition and migration. This study establishes the proof of concept and generation of new leads for additional targeting of breast cancer.
Collapse
Affiliation(s)
- Jyotsana Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritesh Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preeti Gupta
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Smita Rai
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Asha Ganesher
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preethi Badrinarayan
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - G Narahari Sastry
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Rituraj Konwar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| | - Gautam Panda
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| |
Collapse
|
42
|
Lin G, Zhu S, Wu Y, Song C, Wang W, Zhang Y, Chen YL, He Z. ω-3 free fatty acids and all-trans retinoic acid synergistically induce growth inhibition of three subtypes of breast cancer cell lines. Sci Rep 2017; 7:2929. [PMID: 28592877 PMCID: PMC5462805 DOI: 10.1038/s41598-017-03231-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
All-trans retinoic acid (ATRA), one of vitamin A derivatives, shows greater growth inhibition of breast cancer cell for ER-positive than ER-negative cells, while triple negative breast cancer cell such as MDA-MB-231 cell is poorly responsive to ATRA treatment. In this study, we found that combination of ω-3 free fatty acids (ω-3 FFAs) and ATRA exhibited synergistic inhibition of cell growth in three subtypes (ER+ MCF7, HER2+ SK-BR-3, Triple negative HCC1806 and MDA-MB-231 cells) of human breast cancer cell lines. The combined treatment of ω-3 FFAs and ATRA resulted in cell cycle arrest. ω-3 FFAs combined with ATRA synergistically provoked cell apoptosis via the caspase signals but not p53. These findings suggest that combined chemotherapy of ω-3 FFAs with ATRA is beneficial for improvement of ATRA sensitivity in breast cancer cells.
Collapse
Affiliation(s)
- Guangxiao Lin
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Shenglong Zhu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yikuan Wu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Ci Song
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Wanjing Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yuan Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China
| | - Yue-Lei Chen
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P.R. China.
| | - Zhao He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China. .,Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, 214122, China. .,School of Medicine, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
43
|
De Marchi T, Timmermans MA, Sieuwerts AM, Smid M, Look MP, Grebenchtchikov N, Sweep FCGJ, Smits JG, Magdolen V, van Deurzen CHM, Foekens JA, Umar A, Martens JW. Phosphoserine aminotransferase 1 is associated to poor outcome on tamoxifen therapy in recurrent breast cancer. Sci Rep 2017; 7:2099. [PMID: 28522855 PMCID: PMC5437008 DOI: 10.1038/s41598-017-02296-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 04/10/2017] [Indexed: 12/22/2022] Open
Abstract
In a previous study, we detected a significant association between phosphoserine aminotransferase 1 (PSAT1) hyper-methylation and mRNA levels to outcome to tamoxifen treatment in recurrent disease. We here aimed to study the association of PSAT1 protein levels to outcome upon tamoxifen treatment and to obtain more insight in its role in tamoxifen resistance. A cohort of ER positive, hormonal therapy naïve primary breast carcinomas was immunohistochemically (IHC) stained for PSAT1. Staining was analyzed for association with patient's time to progression (TTP) and overall response on first-line tamoxifen for recurrent disease. PSAT1 mRNA levels were also assessed by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR; n = 161) and Affymetrix GeneChip (n = 155). Association of PSAT1 to biological pathways on tamoxifen outcome were assessed by global test. PSAT1 protein and mRNA levels were significantly associated to poor outcome to tamoxifen treatment. When comparing PSAT1 protein and mRNA levels, IHC and RT-qPCR data showed a significant association. Global test results showed that cytokine and JAK-STAT signaling were associated to PSAT1 expression. We hereby report that PSAT1 protein and mRNA levels measured in ER positive primary tumors are associated with poor clinical outcome to tamoxifen.
Collapse
Affiliation(s)
- Tommaso De Marchi
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Mieke A Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anieta M Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maxime P Look
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicolai Grebenchtchikov
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan G Smits
- Department of Pathology, Admiraal de Ruyter Hospital, Goes, The Netherlands
| | - Viktor Magdolen
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | | | - John A Foekens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Arzu Umar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - John W Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Cancer Genomics Center Netherlands, Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Bihani T, Patel HK, Arlt H, Tao N, Jiang H, Brown JL, Purandare DM, Hattersley G, Garner F. Elacestrant (RAD1901), a Selective Estrogen Receptor Degrader (SERD), Has Antitumor Activity in Multiple ER + Breast Cancer Patient-derived Xenograft Models. Clin Cancer Res 2017; 23:4793-4804. [PMID: 28473534 DOI: 10.1158/1078-0432.ccr-16-2561] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/10/2017] [Accepted: 05/01/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Estrogen receptor-positive (ER+) breast cancers are typically treated with endocrine agents, and dependence on the ER pathway is often retained even after multiple rounds of antiestrogen therapy. Selective estrogen receptor degraders (SERD) are being developed as a strategy to more effectively target ER and exploit ER dependence in these cancers, which includes inhibiting both wild-type and mutant forms of ER. The purpose of this study was to evaluate the efficacy of a novel orally bioavailable SERD, elacestrant (RAD1901), in preclinical models of ER+ breast cancer.Experimental Design: Elacestrant was evaluated as a single agent and in combination with palbociclib or everolimus in multiple ER+ breast cancer models, including several patient-derived xenograft models.Results: Elacestrant induces the degradation of ER, inhibits ER-mediated signaling and growth of ER+ breast cancer cell lines in vitro and in vivo, and significantly inhibits tumor growth of multiple PDX models. Furthermore, we demonstrate that elacestrant in combination with palbociclib or everolimus can lead to greater efficacy in certain contexts. Finally, elacestrant exhibits significant antitumor activity both as a single agent and in combination with palbociclib in two patient-derived breast cancer xenograft models harboring ESR1 mutations.Conclusions: These data underscore the potential clinical utility of elacestrant as a single agent and as a combination therapy, for both early- and late-stage ER+ disease. Clin Cancer Res; 23(16); 4793-804. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - Heike Arlt
- Radius Health, Inc, Waltham, Massachusetts
| | | | - Hai Jiang
- Radius Health, Inc, Waltham, Massachusetts
| | | | | | | | | |
Collapse
|
45
|
Review: Receptor Targeted Nuclear Imaging of Breast Cancer. Int J Mol Sci 2017; 18:ijms18020260. [PMID: 28134770 PMCID: PMC5343796 DOI: 10.3390/ijms18020260] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022] Open
Abstract
Receptor targeted nuclear imaging directed against molecular markers overexpressed on breast cancer (BC) cells offers a sensitive and specific method for BC imaging. Currently, a few targets such as estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), somatostatin receptor (SSTR), and the gastrin releasing peptide receptor (GRPR) are being investigated for this purpose. Expression of these targets is BC subtype dependent and information that can be gained from lesion visualization is dependent on the target; ER-targeting radiotracers, e.g., can be used to monitor response to anti-estrogen treatment. Here we give an overview of the studies currently under investigation for receptor targeted nuclear imaging of BC. Main findings of imaging studies are summarized and (potential) purposes of lesion visualization by targeting these molecular markers are discussed. Since BC is a very heterogeneous disease and molecular target expression can vary per subtype, but also during disease progression or under influence of treatment, radiotracers for selected imaging purposes should be chosen carefully.
Collapse
|
46
|
Sammons SL, Topping DL, Blackwell KL. HR+, HER2- Advanced Breast Cancer and CDK4/6 Inhibitors: Mode of Action, Clinical Activity, and Safety Profiles. Curr Cancer Drug Targets 2017; 17:637-649. [PMID: 28359238 PMCID: PMC5652078 DOI: 10.2174/1568009617666170330120452] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cyclin-dependent kinase (CDK) 4/6 inhibitor-based therapies have shown great promise in improving clinical outcomes for patients with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer. OBJECTIVES 1. Discuss the mode of action of the three CDK4/6 inhibitors in late clinical development: palbociclib (PD-0332991; Pfizer), ribociclib (LEE011; Novartis), and abemaciclib (LY2835219; Lilly). 2. Describe the efficacy and safety data relating to their use in HR+, HER2- advanced breast cancer. 3. Discuss the key side effects associated with CDK4/6 inhibitors along with considerations for adverse event management and patient monitoring. METHOD Relevant information and data were assimilated from manuscripts, congress publications, and online sources. RESULTS CDK4/6 inhibitors have demonstrated improved progression-free survival in combination with endocrine therapy compared with endocrine therapy alone. The side-effect profile of each agent is described, along with implications for patient monitoring, and considerations for patient care providers and pharmacists. CONCLUSION Addition of a CDK4/6 inhibitor to endocrine therapy increases efficacy and delays disease progression. Insight into the unique side-effect profiles of this class of agents and effective patient monitoring will facilitate the successful use of CDK4/6 inhibitor-based therapies in the clinic.
Collapse
Affiliation(s)
- Sarah L. Sammons
- Department of Medicine, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Donna L. Topping
- Department of Medicine, Duke University Medical Center, Duke University, Durham, NC, USA
| | - Kimberly L. Blackwell
- Department of Medicine, Duke University Medical Center, Duke University, Durham, NC, USA
| |
Collapse
|
47
|
Spatholobus suberectus Column Extract Inhibits Estrogen Receptor Positive Breast Cancer via Suppressing ER MAPK PI3K/AKT Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:2934340. [PMID: 28096885 PMCID: PMC5209621 DOI: 10.1155/2016/2934340] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/12/2016] [Accepted: 11/09/2016] [Indexed: 12/21/2022]
Abstract
Although Chinese herbal compounds have long been alternatively applied for cancer treatment in China, their treatment effects have not been sufficiently investigated. The Chinese herb Spatholobus suberectus is commonly prescribed to cancer patients. HPLC analysis has shown that the main components of Spatholobus suberectus are flavonoids that can be classified as phytoestrogens, having a structure similar to estrogen. This study was designed to investigate the effects of Spatholobus suberectus column extract (SSCE) on the estrogen receptor-positive (ER+) breast cancer cell line MCF-7 and its possible molecular mechanism. In our study, MTT assay was performed to evaluate cell viability. The results show that SSCE (80, 160, and 320 μg/ml) significantly decreased the viability of MCF-7 cells. SSCE also triggered apoptosis, arrested the cell cycle at the G0/G1 phase, and inhibited cell migration. A dual-luciferase reporter system showed that SSCE suppressed intranuclear p-ER activity; Western blot analysis confirmed the repressed expression of phosphorylated-ER alpha (p-ERα), ERK1/2, p-ERK1/2, AKT, p-AKT, p-mTOR, PI3K, and p-PI3K, indicating that SSCE suppressed the MAPK PI3K/AKT signaling pathway. Collectively, our results suggest that SSCE causes apoptosis, an arrest in the G0/G1 phase, and a decrease in migration in ER+ MCF-7 cells via hypoactivity of the ER and suppression of the MAPK PI3K/AKT pathway.
Collapse
|
48
|
Groner BL, Hynes NE. Breast Cancer Patients Have Greatly Benefited from the Progress in Molecular Oncology. PLoS Biol 2016; 14:e2000314. [PMID: 27684370 PMCID: PMC5042426 DOI: 10.1371/journal.pbio.2000314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cancer research has become a global enterprise, and the number of researchers, as well as the cost for their activities, has skyrocketed. The budget for the National Cancer Institute of the United States National Institutes of Health alone was US$5.2 billion in 2015. Since most of the research is funded by public money, it is perfectly legitimate to ask if these large expenses are worth it. In this brief commentary, we recapitulate some of the breakthroughs that mark the history of breast cancer research over the past decades and emphasize the resulting benefits for afflicted women. In 1971, only 40% of women diagnosed with breast cancer would live another 10 years. Today, nearly 80% of women reach that significant milestone in most developed countries. This dramatic change has afforded breast cancer patients many productive years and a better quality of life. Progress resulted largely from advances in the understanding of the molecular details of the disease and their translation into innovative, rationally designed therapies. These developments are founded on the revolution in molecular and cellular biology, an entirely new array of methods and technologies, the enthusiasm, optimism, and diligence of scientists and clinicians, and the considerable funding efforts from public and private sources. We were lucky to be able to spend our productive years in a period of scientific upheaval in which methods and concepts were revolutionized and that allowed us to contribute, within the global scientific community, to the progress in basic science and clinical practice.
Collapse
Affiliation(s)
- Bernd L. Groner
- Georg Speyer Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- * E-mail: (BLG); (NEH)
| | - Nancy E. Hynes
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- * E-mail: (BLG); (NEH)
| |
Collapse
|
49
|
Muthukaruppan A, Lasham A, Woad KJ, Black MA, Blenkiron C, Miller LD, Harris G, McCarthy N, Findlay MP, Shelling AN, Print CG. Multimodal Assessment of Estrogen Receptor mRNA Profiles to Quantify Estrogen Pathway Activity in Breast Tumors. Clin Breast Cancer 2016; 17:139-153. [PMID: 27756582 DOI: 10.1016/j.clbc.2016.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/25/2016] [Accepted: 09/02/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Molecular markers have transformed our understanding of the heterogeneity of breast cancer and have allowed the identification of genomic profiles of estrogen receptor (ER)-α signaling. However, our understanding of the transcriptional profiles of ER signaling remains inadequate. Therefore, we sought to identify the genomic indicators of ER pathway activity that could supplement traditional immunohistochemical (IHC) assessments of ER status to better understand ER signaling in the breast tumors of individual patients. MATERIALS AND METHODS We reduced ESR1 (gene encoding the ER-α protein) mRNA levels using small interfering RNA in ER+ MCF7 breast cancer cells and assayed for transcriptional changes using Affymetrix HG U133 Plus 2.0 arrays. We also compared 1034 ER+ and ER- breast tumors from publicly available microarray data. The principal components of ER activity generated from these analyses and from other published estrogen signatures were compared with ESR1 expression, ER-α IHC, and patient survival. RESULTS Genes differentially expressed in both analyses were associated with ER-α IHC and ESR1 mRNA expression. They were also significantly enriched for estrogen-driven molecular pathways associated with ESR1, cyclin D1 (CCND1), MYC (v-myc avian myelocytomatosis viral oncogene homolog), and NFKB (nuclear factor kappa B). Despite their differing constituent genes, the principal components generated from these new analyses and from previously published ER-associated gene lists were all associated with each other and with the survival of patients with breast cancer treated with endocrine therapies. CONCLUSION A biomarker of ER-α pathway activity, generated using ESR1-responsive mRNAs in MCF7 cells, when used alongside ER-α IHC and ESR1 mRNA expression, could provide a method for further stratification of patients and add insight into ER pathway activity in these patients.
Collapse
Affiliation(s)
- Anita Muthukaruppan
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Annette Lasham
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Kathryn J Woad
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Michael A Black
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Gavin Harris
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - Nicole McCarthy
- Discipline of Oncology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Michael P Findlay
- Discipline of Oncology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Cristin G Print
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand; New Zealand Bioinformatics Institute, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
50
|
Lin L, Ren L, Wen L, Wang Y, Qi J. Effect of evodiamine on the proliferation and apoptosis of A549 human lung cancer cells. Mol Med Rep 2016; 14:2832-8. [DOI: 10.3892/mmr.2016.5575] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 06/24/2016] [Indexed: 11/05/2022] Open
|