2701
|
Estrogen Receptor Status Oppositely Modifies Breast Cancer Prognosis in BRCA1/BRCA2 Mutation Carriers Versus Non-Carriers. Cancers (Basel) 2019; 11:cancers11060738. [PMID: 31141992 PMCID: PMC6627684 DOI: 10.3390/cancers11060738] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/16/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) prognosis in BRCA1 and BRCA2 mutation carriers has been reported contradictorily, and the significance of variables influencing prognosis in sporadic BC is not established in BC patients with hereditary BRCA1/BRCA2 mutations. In this retrospective cohort study, we analyzed the effect of clinicopathological characteristics on BC prognosis (disease-free survival [DFS] and disease-specific survival [DSS]) in hereditary BRCA1/BRCA2 mutation carriers. We enrolled 234 BRCA1/BRCA2 mutation carriers and 899 non-carriers, of whom 191 carriers and 680 non-carriers, with complete data, were available for survival analyses. We found that patients with ER-positive tumors developed disease recurrence 2.3-times more likely when they carried a BRCA1/BRCA2 mutation (23/60; 38.3% ER-positive carriers vs. 74/445; 16.6% ER-positive non-carriers; p < 0.001). ER-positive mutation carriers also had a 3.4-times higher risk of death due to BC compared with ER-positive non-carriers (13/60; 21.7% vs. 28/445; 6.3%; p < 0.001). Moreover, prognosis in ER-negative BRCA1/BRCA2 mutation carriers was comparable with that in ER-positive non-carriers. Our study demonstrates that ER-positivity worsens BC prognosis in BRCA1/BRCA2 mutation carriers, while prognosis for carriers with ER-negative tumors (including early-onset) is significantly better and comparable with that in ER-positive, older BC non-carriers. These observations indicate that BRCA1/BRCA2 mutation carriers with ER-positive BC represent high-risk patients.
Collapse
|
2702
|
Takahashi N, Hoshi H, Higa A, Hiyama G, Tamura H, Ogawa M, Takagi K, Goda K, Okabe N, Muto S, Suzuki H, Shimomura K, Watanabe S, Takagi M. An In Vitro System for Evaluating Molecular Targeted Drugs Using Lung Patient-Derived Tumor Organoids. Cells 2019; 8:cells8050481. [PMID: 31137590 PMCID: PMC6562414 DOI: 10.3390/cells8050481] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/14/2019] [Accepted: 05/18/2019] [Indexed: 01/02/2023] Open
Abstract
Patient-derived tumor organoids (PDOs) represent a promising preclinical cancer model that better replicates disease, compared with traditional cell culture models. We have established PDOs from various human tumors to accurately and efficiently recapitulate the tissue architecture and function. Molecular targeted therapies with remarkable efficacy are currently in use against various tumors. Thus, there is a need for in vitro functional-potency assays that can be used to test the efficacy of molecular targeted drugs and model complex interactions between immune cells and tumor cells to evaluate the potential for cancer immunotherapy. This study represents an in vitro evaluation of different classes of molecular targeted drugs, including small-molecule inhibitors, monoclonal antibodies, and an antibody-drug conjugate, using lung PDOs. We evaluated epidermal growth factor receptor and human epidermal growth factor receptor 2 (HER2) inhibitors using a suitable high-throughput assay system. Next, the antibody-dependent cellular cytotoxicity (ADCC) activity of an anti-HER2 monoclonal antibody was evaluated to visualize the interactions of immune cells with PDOs during ADCC responses. Moreover, an evaluation system was developed for the immune checkpoint inhibitors, nivolumab and pembrolizumab, using PDOs. Our results demonstrate that the in vitro assay systems using PDOs were suitable for evaluating molecular targeted drugs under conditions that better reflect pathological conditions.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biopsy
- Carcinoma, Adenosquamous/drug therapy
- Carcinoma, Adenosquamous/pathology
- Carcinoma, Adenosquamous/surgery
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/surgery
- Cell Survival/drug effects
- Cells, Cultured
- Drug Evaluation/methods
- ErbB Receptors/antagonists & inhibitors
- Humans
- L-Lactate Dehydrogenase/analysis
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Molecular Targeted Therapy
- Organoids/drug effects
- Receptor, ErbB-2/antagonists & inhibitors
Collapse
Affiliation(s)
- Nobuhiko Takahashi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Hirotaka Hoshi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Arisa Higa
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Gen Hiyama
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Hirosumi Tamura
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Mayu Ogawa
- Research and Development, Biological Evaluation Technology 2, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan.
| | - Kosuke Takagi
- Research and Development, SSD Technology Innovation 3, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan.
| | - Kazuhito Goda
- Research and Development, Biological Evaluation Technology 2, Olympus Corporation, Hachioji, Tokyo 192-8512, Japan.
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan.
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Shinya Watanabe
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Motoki Takagi
- Medical-Industrial Translational Research Center, Fukushima Medical University, Fukushima 960-1295, Japan.
| |
Collapse
|
2703
|
|
2704
|
Qin JJ, Yan L, Zhang J, Zhang WD. STAT3 as a potential therapeutic target in triple negative breast cancer: a systematic review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:195. [PMID: 31088482 PMCID: PMC6518732 DOI: 10.1186/s13046-019-1206-z] [Citation(s) in RCA: 261] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022]
Abstract
Triple negative breast cancer (TNBC), which is typically lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), represents the most aggressive and mortal subtype of breast cancer. Currently, only a few treatment options are available for TNBC due to the absence of molecular targets, which underscores the need for developing novel therapeutic and preventive approaches for this disease. Recent evidence from clinical trials and preclinical studies has demonstrated a pivotal role of signal transducer and activator of transcription 3 (STAT3) in the initiation, progression, metastasis, and immune evasion of TNBC. STAT3 is overexpressed and constitutively activated in TNBC cells and contributes to cell survival, proliferation, cell cycle progression, anti-apoptosis, migration, invasion, angiogenesis, chemoresistance, immunosuppression, and stem cells self-renewal and differentiation by regulating the expression of its downstream target genes. STAT3 small molecule inhibitors have been developed and shown excellent anticancer activities in in vitro and in vivo models of TNBC. This review discusses the recent advances in the understanding of STAT3, with a focus on STAT3’s oncogenic role in TNBC. The current targeting strategies and representative small molecule inhibitors of STAT3 are highlighted. We also propose potential strategies that can be further examined for developing more specific and effective inhibitors for TNBC prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China.
| | - Li Yan
- School of Pharmacy, Naval Medical University, 325 Guohe Road, Yangpu District, Shanghai, 200433, China
| | - Jia Zhang
- Shanxi Institute of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, 325 Guohe Road, Yangpu District, Shanghai, 200433, China. .,Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
2705
|
Affiliation(s)
- Kathryn J Ruddy
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Patricia A Ganz
- Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, California
- Department of Medicine, Division of Hematology Oncology, David Geffen School of Medicine, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
| |
Collapse
|
2706
|
Chen X, Lv Y, Sun Y, Zhang H, Xie W, Zhong L, Chen Q, Li M, Li L, Feng J, Yao A, Zhang Q, Huang X, Yu Z, Yao P. PGC1β Regulates Breast Tumor Growth and Metastasis by SREBP1-Mediated HKDC1 Expression. Front Oncol 2019; 9:290. [PMID: 31058090 PMCID: PMC6478765 DOI: 10.3389/fonc.2019.00290] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Breast cancer is a very common cancer with significant premature mortality in women. In this study, we show that HKDC1 expression in breast cancer cells is increased significantly. We aim to investigate the detailed mechanism for the regulation of HKDC1 expression and its potential contribution to tumorigenesis. Methods: Gene expression was evaluated by real time PCR, western blotting, and immunohistochemistry. The mechanism for PGC1β/SREBP1-mediated HKDC1 expression was investigated using luciferase reporter assay, chromatin immunoprecipitation, and siRNA techniques. In addition, HKDC1 was overexpressed or knocked down by lentivirus to evaluate the potential effect on in vitro cell proliferation, glucose uptake, mitochondrial function, apoptosis, and reactive oxygen species (ROS) formation. Furthermore, an in vivo xenograft tumor development study was employed to investigate the effect of HKDC1 on tumor growth and mouse survival. Results: HKDC1 is highly expressed in both breast cancer cells and clinical tumor tissues. HKDC1 expression is upregulated and co-activated by PGC1β through SREBP1 binding motif on the HKDC1 promoter. HKDC1 is located on the mitochondrial membrane and regulates the permeability transition pore opening by binding with VDAC1, subsequently modulating glucose uptake and cell proliferation. Overexpression of HKDC1 increases while knockdown of HKDC1 decreases in vitro breast cancer cell proliferation and in vivo tumor growth, metastasis, and mouse survival. Conclusions: PGC1β regulates breast cancer tumor growth and metastasis by SREBP1-mediated HKDC1 expression. This provides a novel therapeutic strategy through targeting the PGC1β/HKDC1 signaling pathway for breast cancer treatment.
Collapse
Affiliation(s)
- Xiaoli Chen
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Yang Lv
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Ying Sun
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Hongyu Zhang
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Weiguo Xie
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Liyan Zhong
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Qi Chen
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Min Li
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Ling Li
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Jia Feng
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Athena Yao
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Qi Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaodong Huang
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Zhendong Yu
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Paul Yao
- Institute of Rehabilitation Center, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China.,Hainan Maternal and Child Health Hospital, Haikou, China.,Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
2707
|
Xi Y, Zhang X, Yang Z, Zhang X, Guo Q, Zhang Z, Chen S, Zheng H, Hua B. Prognositic significance of P-cadherin expression in breast cancer: Protocol for a meta-analysis. Medicine (Baltimore) 2019; 98:e14924. [PMID: 30896652 PMCID: PMC6709078 DOI: 10.1097/md.0000000000014924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/05/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND P-cadherin is a calcium-dependent cell-cell adhesion glycoprotein. It has been implicated in invasiveness and metastasis. However, the clinical prognostic value of overexpression of P-cadherin in patients with breast cancer (BC) remains unsettled. METHODS A systematic literature search will be performed in all available databases to quantitatively review eligible studies and identify all relevant data, which could be used to detect the relationship between overexpression of P-cadherin and overall survival (OS), disease-free survival (DFS), and clinicopathological parameters. Hazard ratio and 95% confidence intervals (CIs) or P value will be employed as effect measures to estimate the correlation between P-cadherin and the oncologic outcomes including overall survival (OS), disease-free survival (DFS). Odds ratios (ORs) and the 95% CIs will be evaluated for the pooled analysis of the correlation between P-cadherin expression and clinicopathological features. We will use the Review Manager (Revman) 5.3.5 software (Cochrane Community, London, United Kingdom) and STATA 14 software (version 14.0; Stata Corp, College Station, TX) to perform the meta-analysis to calculate the data. RESULTS The review will provide a high-quality synthesis of current evidence of the prognostic role of P-cadherin in BCs. The results will be published in a peer-reviewed journal. CONCLUSION We hope that the results of this study will provide significant evidence to assess whether the expression of P-cadherin is associated with poor prognosis in patients with BC. PROSPERO REGISTRATION NUMBER This meta-analysis protocol has been registered in the PROSPERO network with registration number: CRD42019119880.
Collapse
Affiliation(s)
- Yupeng Xi
- Beijing University of Chinese Medicine
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Xiwen Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Zizhen Yang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
- Shanxi University of Chinese Medicine, Xianyang, Shanxi Province, China
| | - Xing Zhang
- Beijing University of Chinese Medicine
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Qiujun Guo
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Zhenhua Zhang
- Beijing University of Chinese Medicine
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Shuntai Chen
- Beijing University of Chinese Medicine
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Honggang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| | - Baojin Hua
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing
| |
Collapse
|
2708
|
Diaz Bessone MI, Gattas MJ, Laporte T, Tanaka M, Simian M. The Tumor Microenvironment as a Regulator of Endocrine Resistance in Breast Cancer. Front Endocrinol (Lausanne) 2019; 10:547. [PMID: 31440208 PMCID: PMC6694443 DOI: 10.3389/fendo.2019.00547] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
Estrogen receptor positive breast neoplasias represent over 70% of diagnosed breast cancers. Depending on the stage at which the tumor is detected, HER2 status and genomic risk, endocrine therapy is combined with either radio, chemo and/or targeted therapy. A growing amount of evidence supports the notion that components of the tumor microenvironment play specific roles in response to treatment and that strategies targeting these key interactions with tumor cells could pave the way to a new generation of therapies. In this review, we analyze the evidence suggesting different components of the tumor microenvironment play a role in hormone receptor positive breast cancer progression. In particular we focus on the immune system, carcinoma associated fibroblasts and the extracellular matrix. Further insight into the cross talk between these constituents of the microenvironment and the tumor cells may lead to therapies that eliminate disseminated metastatic cells early on, and thus reduce distant disease relapse which is the leading cause of death for patients who are diagnosed with this illness.
Collapse
Affiliation(s)
- María Inés Diaz Bessone
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - María José Gattas
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Tomás Laporte
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Max Tanaka
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
- Amsterdam UMC, VUmc School of Medical Sciences, University of Vrije, Amsterdam, Netherlands
| | - Marina Simian
- Laboratory of NanoBiology, Instituto de Nanosistemas, Universidad Nacional de San Martín, Buenos Aires, Argentina
- *Correspondence: Marina Simian
| |
Collapse
|