1
|
Weng B, Li M, Zhu W, Peng J, Mao X, Zheng Y, Zhang C, Pan S, Mao H, Zhao J. Distinguished biomimetic dECM system facilitates early detection of metastatic breast cancer cells. Bioeng Transl Med 2024; 9:e10597. [PMID: 38193110 PMCID: PMC10771560 DOI: 10.1002/btm2.10597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 01/10/2024] Open
Abstract
Breast cancer is the most prevalent malignant tumor affecting women's health. Bone is the most common distant metastatic organ, worsening the quality of life and increasing the mortality of patients. Early detection of breast cancer bone metastasis is urgent for halting disease progression and improving tumor prognosis. Recently, extracellular matrix (ECM) with biomimetic tissue niches opened a new avenue for tumor models in vitro. Here, we developed a biomimetic decellularized ECM (dECM) system to recapitulate bone niches at different situations, bone mimetic dECM from osteoblasts (BM-ECM) and bone tumor mimetic dECM from osteosarcoma cells (OS-ECM). The two kinds of dECMs exhibited distinct morphology, protein composition, and distribution. Interestingly, highly metastatic breast cancer cells tended to adhere and migrate on BM-ECM, while lowly metastatic breast cancer cells preferred the OS-ECM niche. Epithelial-to-mesenchymal transition was a potential mechanism to initiate the breast cancer cell migration on different biomimetic dECMs. Importantly, in the nude mice model, the dECM system captured metastatic breast cancer cells as early as 10 days after orthotopic transplantation in mammary gland pads, with higher signal on BM-ECM than that on OS-ECM. Collectively, the biomimetic dECM system might be a promising tumor model to distinguish the metastatic ability of breast cancer cells in vitro and to facilitate early detection of metastatic breast cancer cells in vivo, contributing to the diagnosis of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Bowen Weng
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| | - Mei Li
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Weilai Zhu
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| | - Jing Peng
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| | - Xufeng Mao
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
- Department of Orthopaedic SurgeryThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Yanan Zheng
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| | - Chi Zhang
- Department of Orthopaedic SurgeryThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Senhao Pan
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| | - Haijiao Mao
- Department of Orthopaedic SurgeryThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Jiyuan Zhao
- Zhejiang Key Laboratory of PathophysiologySchool of Medicine, Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
2
|
Zhan Q, Liu B, Situ X, Luo Y, Fu T, Wang Y, Xie Z, Ren L, Zhu Y, He W, Ke Z. New insights into the correlations between circulating tumor cells and target organ metastasis. Signal Transduct Target Ther 2023; 8:465. [PMID: 38129401 PMCID: PMC10739776 DOI: 10.1038/s41392-023-01725-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Organ-specific metastasis is the primary cause of cancer patient death. The distant metastasis of tumor cells to specific organs depends on both the intrinsic characteristics of the tumor cells and extrinsic factors in their microenvironment. During an intermediate stage of metastasis, circulating tumor cells (CTCs) are released into the bloodstream from primary and metastatic tumors. CTCs harboring aggressive or metastatic features can extravasate to remote sites for continuous colonizing growth, leading to further lesions. In the past decade, numerous studies demonstrated that CTCs exhibited huge clinical value including predicting distant metastasis, assessing prognosis and monitoring treatment response et al. Furthermore, increasingly numerous experiments are dedicated to identifying the key molecules on or inside CTCs and exploring how they mediate CTC-related organ-specific metastasis. Based on the above molecules, more and more inhibitors are being developed to target CTCs and being utilized to completely clean CTCs, which should provide promising prospects to administer advanced tumor. Recently, the application of various nanomaterials and microfluidic technologies in CTCs enrichment technology has assisted to improve our deep insights into the phenotypic characteristics and biological functions of CTCs as a potential therapy target, which may pave the way for us to make practical clinical strategies. In the present review, we mainly focus on the role of CTCs being involved in targeted organ metastasis, especially the latest molecular mechanism research and clinical intervention strategies related to CTCs.
Collapse
Affiliation(s)
- Qinru Zhan
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Bixia Liu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Xiaohua Situ
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yuting Luo
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Tongze Fu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yanxia Wang
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Zhongpeng Xie
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Lijuan Ren
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| | - Weiling He
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, 10065, USA.
- School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, 361000, Xiamen, Fujian, P.R. China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
3
|
Alvarez-Frutos L, Barriuso D, Duran M, Infante M, Kroemer G, Palacios-Ramirez R, Senovilla L. Multiomics insights on the onset, progression, and metastatic evolution of breast cancer. Front Oncol 2023; 13:1292046. [PMID: 38169859 PMCID: PMC10758476 DOI: 10.3389/fonc.2023.1292046] [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: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is the most common malignant neoplasm in women. Despite progress to date, 700,000 women worldwide died of this disease in 2020. Apparently, the prognostic markers currently used in the clinic are not sufficient to determine the most appropriate treatment. For this reason, great efforts have been made in recent years to identify new molecular biomarkers that will allow more precise and personalized therapeutic decisions in both primary and recurrent breast cancers. These molecular biomarkers include genetic and post-transcriptional alterations, changes in protein expression, as well as metabolic, immunological or microbial changes identified by multiple omics technologies (e.g., genomics, epigenomics, transcriptomics, proteomics, glycomics, metabolomics, lipidomics, immunomics and microbiomics). This review summarizes studies based on omics analysis that have identified new biomarkers for diagnosis, patient stratification, differentiation between stages of tumor development (initiation, progression, and metastasis/recurrence), and their relevance for treatment selection. Furthermore, this review highlights the importance of clinical trials based on multiomics studies and the need to advance in this direction in order to establish personalized therapies and prolong disease-free survival of these patients in the future.
Collapse
Affiliation(s)
- Lucia Alvarez-Frutos
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Daniel Barriuso
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mercedes Duran
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mar Infante
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, Paris, France
| | - Roberto Palacios-Ramirez
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Laura Senovilla
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
4
|
Wang S, Wu W, Lin X, Zhang KM, Wu Q, Luo M, Zhou J. Predictive and prognostic biomarkers of bone metastasis in breast cancer: current status and future directions. Cell Biosci 2023; 13:224. [PMID: 38041134 PMCID: PMC10693103 DOI: 10.1186/s13578-023-01171-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
The most common site of metastasis in breast cancer is the bone, where the balance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation is disrupted. This imbalance causes osteolytic bone metastasis in breast cancer, which leads to bone pain, pathological fractures, spinal cord compression, and other skeletal-related events (SREs). These complications reduce patients' quality of life significantly and have a profound impact on prognosis. In this review, we begin by providing a brief overview of the epidemiology of bone metastasis in breast cancer, including current diagnostic tools, treatment approaches, and existing challenges. Then, we will introduce the pathophysiology of breast cancer bone metastasis (BCBM) and the animal models involved in the study of BCBM. We then come to the focus of this paper: a discussion of several biomarkers that have the potential to provide predictive and prognostic value in the context of BCBM-some of which may be particularly compatible with more comprehensive liquid biopsies. Beyond that, we briefly explore the potential of new technologies such as single-cell sequencing and organoid models, which will improve our understanding of tumor heterogeneity and aid in the development of improved biomarkers. The emerging biomarkers discussed hold promise for future clinical application, aiding in the prevention of BCBM, improving the prognosis of patients, and guiding the implementation of personalized medicine.
Collapse
Affiliation(s)
- Shenkangle Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Wenxin Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Xixi Lin
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | | | - QingLiang Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Hangzhou Ninth People's Hospital, Hangzhou, 310014, China
| | - Mingpeng Luo
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
5
|
Cheng TC, Wu JH, Zhu B, Gao HY, Zheng L, Chen WX. Identification of a novel five ferroptosis-related gene signature as a promising prognostic model for breast cancer. J Cancer Res Clin Oncol 2023; 149:16779-16795. [PMID: 37728703 PMCID: PMC10645672 DOI: 10.1007/s00432-023-05423-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Breast cancer (BCa) is a major challenge for women's health worldwide. Ferroptosis is closely related to tumorigenesis and cancer progression. However, the prognostic value of ferroptosis-related genes in BCa remains unclear, and more accurate prognostic models are urgently needed. METHODS Gene expression profiles and clinical information of BCa patients were collected from public databases. LASSO and multivariate Cox regression analysis were utilized to construct the prognostic gene signature. Kaplan-Meier plotter, receiver operating characteristic (ROC) curves, and nomogram were used to validate the prognostic value of the gene signature. Gene set enrichment analysis was performed to explore the molecular functions and signaling pathways. RESULTS Differentially expressed ferroptosis-related genes between BCa samples and normal tissues were obtained. A novel five-gene signature including BCL2, SLC40A1, TFF1, APOOL, and PRAME was established for prognosis prediction. Patients stratified into high-risk or low-risk group displayed significantly different survival. Kaplan-Meier and ROC curves showed a good performance for survival prediction in different cohorts. Biological function analysis revealed that the five-gene signature was associated with cancer progression, immune infiltration, immune response, and drug resistance. Nomogram including the five-gene signature was established. CONCLUSION A novel five ferroptosis-related gene signature and nomogram could be used for prognostic prediction in BCa.
Collapse
Affiliation(s)
- Tian- Cheng Cheng
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglongxiang, Changzhou, 213000, Jiangsu Province, China
- Graduate School, Bengbu Medical College, Bengbu, 233000, Anhui Province, China
| | - Jia-Hao Wu
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglongxiang, Changzhou, 213000, Jiangsu Province, China
- Graduate School, Dalian Medical University, Dalian, 116000, Liaoning Province, China
| | - Bei Zhu
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglongxiang, Changzhou, 213000, Jiangsu Province, China
| | - Hai-Yan Gao
- Department of Breast Surgery, The Affiliated Changzhou Tumor Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, China
| | - Lin Zheng
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglongxiang, Changzhou, 213000, Jiangsu Province, China.
| | - Wei-Xian Chen
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglongxiang, Changzhou, 213000, Jiangsu Province, China.
- Post-Doctoral Working Station, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, ChangzhouJiangsu Province, 213000, China.
| |
Collapse
|
6
|
Lu C, Zhan Y, Jiang Y, Liao J, Qiu Z. Exosome-derived ANXA9 functions as an oncogene in breast cancer. J Pathol Clin Res 2023; 9:378-390. [PMID: 37294149 PMCID: PMC10397375 DOI: 10.1002/cjp2.334] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023]
Abstract
Breast cancer (BCA) is one of the most prevalent cancers among women. Emerging evidence has revealed that Annexin A-9 (ANXA9) plays a crucial function in the development of some cancers. Notably, ANXA9 has been reported to be a new prognostic biomarker for gastric and colorectal cancers. However, its expression and biological function in BCA have not yet been investigated. Using online bioinformatics tools such as TIMER, GEPIA, HPA, and UALCAN, we predicted ANXA9 expression and its correlation with the clinicopathological characteristics of BCA patients. RT-qPCR and western blot were utilized to measure ANXA9 mRNA and ANXA9 protein expression in BCA patient tissues and cells. BCA-derived exosomes were identified by transmission electron microscopy. Functional assays were employed to evaluate the biological role of ANXA9 in BCA cell proliferation, migration, invasion, and apoptosis. A tumor xenograft in vivo model was utilized to assess the role of ANXA9 in tumor growth in mice. Bioinformatics and functional screening analysis revealed that ANXA9 was highly expressed in BCA patient tissues, with median ANXA9 expression 1.5- to 2-fold higher than in normal tissues (p < 0.05). RT-qPCR confirmed that ANXA9 expression in BCA tissues was around 1.5-fold higher than the adjacent normal tissues (p < 0.001). ANXA9 expression in different subtypes of BCA also showed a difference, and ANXA9 was found to be mostly significantly upregulated in luminal BCA relative to normal tissues or other histological subtypes (p < 0.001). Moreover, ANXA9 expression was elevated in different races, ages, clinical stages, node metastasis status, and menopause status groups relative to the normal group (p < 0.001). Furthermore, ANXA9 was found to be secreted by BCA tissue-derived exosomes and its expression was upregulated 1- to 7-fold in BCA cells treated with exosomes (p < 0.001), while its expression in MCF10A cells was not significantly altered by treatment with exosomes (p > 0.05). ANXA9 silencing induced a significant decrease of around 30% in the colony number of BCA cells (p < 0.01). The number of migrated and invaded BCA cells also decreased by around 65 and 68%, respectively, after silencing ANXA9 (p < 0.01). Tumor size was significantly reduced (nearly half) in the LV-sh-ANXA9 group relative to the LV-NC group in the xenograft model (p < 0.01), suggesting that ANXA9 silencing repressed tumor progression in BCA progression in vitro and in vivo. In conclusion, exosome-derived ANXA9 functions as an oncogene that facilitates the proliferation, migration, and invasiveness of BCA cells and enhances tumor growth in BCA development, which may provide a new prognostic and therapeutic biomarker for BCA patients.
Collapse
Affiliation(s)
- Cuiping Lu
- Department of Medical OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPR China
| | - Ying Zhan
- Department of Medical OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPR China
| | - Yunshan Jiang
- Department of Medical OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPR China
| | - Jianrong Liao
- Department of Medical OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPR China
| | - Zidan Qiu
- Department of Medical OncologyLongyan First Affiliated Hospital of Fujian Medical UniversityLongyanFujianPR China
| |
Collapse
|
7
|
Wu Q, Tian P, He D, Jia Z, He Y, Luo W, Lv X, Wang Y, Zhang P, Liang Y, Zhao W, Qin J, Su P, Jiang YZ, Shao ZM, Yang Q, Hu G. SCUBE2 mediates bone metastasis of luminal breast cancer by modulating immune-suppressive osteoblastic niches. Cell Res 2023; 33:464-478. [PMID: 37142671 PMCID: PMC10235122 DOI: 10.1038/s41422-023-00810-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/02/2023] [Indexed: 05/06/2023] Open
Abstract
Estrogen receptor (ER)-positive luminal breast cancer is a subtype with generally lower risk of metastasis to most distant organs. However, bone recurrence occurs preferentially in luminal breast cancer. The mechanisms of this subtype-specific organotropism remain elusive. Here we show that an ER-regulated secretory protein SCUBE2 contributes to bone tropism of luminal breast cancer. Single-cell RNA sequencing analysis reveals osteoblastic enrichment by SCUBE2 in early bone-metastatic niches. SCUBE2 facilitates release of tumor membrane-anchored SHH to activate Hedgehog signaling in mesenchymal stem cells, thus promoting osteoblast differentiation. Osteoblasts deposit collagens to suppress NK cells via the inhibitory LAIR1 signaling and promote tumor colonization. SCUBE2 expression and secretion are associated with osteoblast differentiation and bone metastasis in human tumors. Targeting Hedgehog signaling with Sonidegib and targeting SCUBE2 with a neutralizing antibody both effectively suppress bone metastasis in multiple metastasis models. Overall, our findings provide a mechanistic explanation for bone preference in luminal breast cancer metastasis and new approaches for metastasis treatment.
Collapse
Affiliation(s)
- Qiuyao Wu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pu Tian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dasa He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenchang Jia
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yunfei He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenqian Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xianzhe Lv
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peiyuan Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yajun Liang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjin Zhao
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | - Jun Qin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Ji'nan, Shandong, China.
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
Qureshi R, Picon-Ruiz M, Sho M, Van Booven D, Nunes de Paiva V, Diaz-Ruano AB, Ince TA, Slingerland J. Estrone, the major postmenopausal estrogen, binds ERa to induce SNAI2, epithelial-to-mesenchymal transition, and ER+ breast cancer metastasis. Cell Rep 2022; 41:111672. [PMID: 36384125 PMCID: PMC9798480 DOI: 10.1016/j.celrep.2022.111672] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 09/22/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Recent work showed that the dominant post-menopausal estrogen, estrone, cooperates with nuclear factor κB (NF-κB) to stimulate inflammation, while pre-menopausal 17β-estradiol opposes NF-κB. Here, we show that post-menopausal estrone, but not 17β-estradiol, activates epithelial-to-mesenchymal transition (EMT) genes to stimulate breast cancer metastasis. HSD17B14, which converts 17β-estradiol to estrone, is higher in cancer than normal breast tissue and in metastatic than primary cancers and associates with earlier metastasis. Treatment with estrone, but not 17β-estradiol, and HSD17B14 overexpression both stimulate an EMT, matrigel invasion, and lung, bone, and liver metastasis in estrogen-receptor-positive (ER+) breast cancer models, while HSD17B14 knockdown reverses the EMT. Estrone:ERα recruits CBP/p300 to the SNAI2 promoter to induce SNAI2 and stimulate an EMT, while 17β-estradiol:ERα recruits co-repressors HDAC1 and NCOR1 to this site. Present work reveals novel differences in gene regulation by these estrogens and the importance of estrone to ER+ breast cancer progression. Upon loss of 17β-estradiol at menopause, estrone-liganded ERα would promote ER+ breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Rehana Qureshi
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA; Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA; John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, 18071 Granada, Spain
| | - Maiko Sho
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA
| | - Derek Van Booven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vanessa Nunes de Paiva
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anna B Diaz-Ruano
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Tan A Ince
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Joyce Slingerland
- Breast Cancer Program, Lombardi Comprehensive Cancer Centre, Department of Oncology, Georgetown University, Washington, DC 20007, USA; Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
9
|
Li JJ, Wang S, Guan ZN, Zhang JX, Zhan RX, Zhu JL. Anterior Gradient 2 is a Significant Prognostic Biomarker in Bone Metastasis of Breast Cancer. Pathol Oncol Res 2022; 28:1610538. [PMID: 36405393 PMCID: PMC9668893 DOI: 10.3389/pore.2022.1610538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022]
Abstract
Background: The study aimed to detect DEGs associated with BRCA bone metastasis, filter prognosis biomarkers, and explore possible pathways. Methods: GSE175692 dataset was used to detect DEGs between BRCA bone metastatic cases and non-bone metastatic cases, followed by the construction of a PPI network among DEGs. The main module among the PPI network was then determined and pathway analysis on genes within the module was performed. Through performing Cox regression, Kaplan-Meier, nomogram, and ROC curve analyses using GSE175692 and GSE124647 datasets at the same time, the most significant prognostic biomarker was gradually filtered. Finally, important pathways associated with prognostic biomarkers were explored by GSEA analysis. Results: The 74 DEGs were detected between bone metastasis and non-bone metastasis groups. A total of 15 nodes were included in the main module among the whole PPI network and they mainly correlated with the IL-17 signaling pathway. We then performed Cox analysis on 15 genes using two datasets and only enrolled the genes with p < 0.05 in Cox analysis into the further analyses. Kaplan-Meier analyses using two datasets showed that the common biomarker AGR2 expression was related to the survival time of BRCA metastatic cases. Further, the nomogram determined the greatest contribution of AGR2 on the survival probability and the ROC curve revealed its optimal prognostic performance. More importantly, high expression of AGR2 prolonged the survival time of BRCA bone metastatic patients. These results all suggested the importance of AGR2 in metastatic BRCA. Finally, we performed the GSEA analysis and found that AGR2 was negatively related to IL-17 and NF-kβ signaling pathways. Conclusion: AGR2 was finally determined as the most important prognostic biomarker in BRCA bone metastasis, and it may play a vital role in cancer progression by regulating IL-17 and NF-kB signaling pathways.
Collapse
Affiliation(s)
- Jin-Jin Li
- Department of Orthopaedics, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Shuai Wang
- Department of Pathology, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Zhong-Ning Guan
- Department of Orthopaedics, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Jin-Xi Zhang
- Department of Orthopaedics, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Ri-Xin Zhan
- Department of Medical Record Management, Hangzhou Ninth People’s Hospital, Hangzhou, China
| | - Jian-Long Zhu
- Department of Orthopaedics, Hangzhou Ninth People’s Hospital, Hangzhou, China
- *Correspondence: Jian-Long Zhu,
| |
Collapse
|
10
|
Bennett C, Carroll C, Wright C, Awad B, Park JM, Farmer M, Brown E(B, Heatherly A, Woodard S. Breast Cancer Genomics: Primary and Most Common Metastases. Cancers (Basel) 2022; 14:3046. [PMID: 35804819 PMCID: PMC9265113 DOI: 10.3390/cancers14133046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Specific genomic alterations have been found in primary breast cancer involving driver mutations that result in tumorigenesis. Metastatic breast cancer, which is uncommon at the time of disease onset, variably impacts patients throughout the course of their disease. Both the molecular profiles and diverse genomic pathways vary in the development and progression of metastatic breast cancer. From the most common metastatic site (bone), to the rare sites such as orbital, gynecologic, or pancreatic metastases, different levels of gene expression indicate the potential involvement of numerous genes in the development and spread of breast cancer. Knowledge of these alterations can, not only help predict future disease, but also lead to advancement in breast cancer treatments. This review discusses the somatic landscape of breast primary and metastatic tumors.
Collapse
Affiliation(s)
- Caroline Bennett
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Caleb Carroll
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Cooper Wright
- Birmingham Marnix E. Heersink School of Medicine, The University of Alabama, 1670 University Blvd, Birmingham, AL 35233, USA; (C.B.); (C.C.); (C.W.)
| | - Barbara Awad
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, 6965 Cumberland Gap Pkwy, Harrogate, TN 37752, USA;
| | - Jeong Mi Park
- Department of Radiology, The University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA;
| | - Meagan Farmer
- Department of Genetics, Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL 35233, USA; (M.F.); (A.H.)
| | - Elizabeth (Bryce) Brown
- Laboratory Genetics Counselor, UAB Medical Genomics Laboratory, Kaul Human Genetics Building, 720 20th Street South, Suite 332, Birmingham, AL 35294, USA;
| | - Alexis Heatherly
- Department of Genetics, Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL 35233, USA; (M.F.); (A.H.)
| | - Stefanie Woodard
- Department of Radiology, The University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL 35249, USA;
| |
Collapse
|
11
|
Fina E, Cleris L, Dugo M, Lecchi M, Ciniselli CM, Lecis D, Bianchi GV, Verderio P, Daidone MG, Cappelletti V. Gene signatures of circulating breast cancer cell models are a source of novel molecular determinants of metastasis and improve circulating tumor cell detection in patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:78. [PMID: 35216615 PMCID: PMC8876758 DOI: 10.1186/s13046-022-02259-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
Abstract
Background Progression to stage IV disease remains the main cause of breast cancer-related deaths. Increasing knowledge on the hematogenous phase of metastasis is key for exploiting the entire window of opportunity to interfere with early dissemination and to achieve a more effective disease control. Recent evidence suggests that circulating tumor cells (CTCs) possess diverse adaptive mechanisms to survive in blood and eventually metastasize, encouraging research into CTC-directed therapies. Methods On the hypothesis that the distinguishing molecular features of CTCs reveal useful information on metastasis biology and disease outcome, we compared the transcriptome of CTCs, primary tumors, lymph-node and lung metastases of the MDA-MB-231 xenograft model, and assessed the biological role of a panel of selected genes, by in vitro and in vivo functional assays, and their clinical significance in M0 and M+ breast cancer patients. Results We found that hematogenous dissemination is governed by a transcriptional program and identified a CTC signature that includes 192 up-regulated genes, mainly related to cell plasticity and adaptation, and 282 down-regulated genes, involved in chromatin remodeling and transcription. Among genes up-regulated in CTCs, FADS3 was found to increases cell membrane fluidity and promote hematogenous diffusion and lung metastasis formation. TFF3 was observed to be associated with a subset of CTCs with epithelial-like features in the experimental model and in a cohort of 44 breast cancer patients, and to play a role in cell migration, invasion and blood-borne dissemination. The analysis of clinical samples with a panel of CTC-specific genes (ADPRHL1, ELF3, FCF1, TFF1 and TFF3) considerably improved CTC detection as compared with epithelial and tumor-associated markers both in M0 and stage IV patients, and CTC kinetics informed disease relapse in the neoadjuvant setting. Conclusions Our findings provide evidence on the potential of a CTC-specific molecular profile as source of metastasis-relevant genes in breast cancer experimental models and in patients. Thanks to transcriptome analysis we generated a novel CTC signature in the MDA-MB-231 xenograft model, adding a new piece to the current knowledge on the key players that orchestrate tumor cell hematogenous dissemination and breast cancer metastasis, and expanding the list of CTC-related biomarkers for future validation studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02259-8.
Collapse
Affiliation(s)
- Emanuela Fina
- Biomarkers Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy. .,Current affiliation: Humanitas Research Center, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy.
| | - Loredana Cleris
- Biomarkers Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy
| | - Matteo Dugo
- Platform of Integrated Biology, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy.,Current affiliation: Department of Medical Oncology, IRCCS Ospedale San Raffaele, via Olgettina 60, 20132, Milan, Italy
| | - Mara Lecchi
- Bioinformatics and Biostatistics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Chiara Maura Ciniselli
- Bioinformatics and Biostatistics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Daniele Lecis
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Giulia Valeria Bianchi
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Paolo Verderio
- Bioinformatics and Biostatistics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Maria Grazia Daidone
- Biomarkers Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy
| | - Vera Cappelletti
- Biomarkers Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Venezian 1, 20133, Milan, Italy.
| |
Collapse
|
12
|
Carlisle SM, Trainor PJ, Doll MA, Hein DW. Human Arylamine N-Acetyltransferase 1 (NAT1) Knockout in MDA-MB-231 Breast Cancer Cell Lines Leads to Transcription of NAT2. Front Pharmacol 2022; 12:803254. [PMID: 35046826 PMCID: PMC8762260 DOI: 10.3389/fphar.2021.803254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Many cancers, including breast cancer, have shown differential expression of human arylamine N-acetyltransferase 1 (NAT1). The exact effect this differential expression has on disease risk and progression remains unclear. While NAT1 is classically defined as a xenobiotic metabolizing enzyme, other functions and roles in endogenous metabolism have recently been described providing additional impetus for investigating the effects of varying levels of NAT1 on global gene expression. Our objective is to further evaluate the role of NAT1 in breast cancer by determining the effect of NAT1 overexpression, knockdown, and knockout on global gene expression in MDA-MB-231 cell lines. RNA-seq was utilized to interrogate differential gene expression (genes correlated with NAT1 activity) across three biological replicates of previously constructed and characterized MDA-MB-231 breast cancer cell lines expressing parental (Scrambled), increased (Up), decreased (Down, CRISPR 2–12), or knockout (CRISPR 2–19, CRISPR 5–50) levels of NAT1. 3,889 genes were significantly associated with the NAT1 N-acetylation activity of the cell lines (adjusted p ≤ 0.05); of those 3,889 genes, 1,756 were positively associated with NAT1 N-acetylation activity and 2,133 were negatively associated with NAT1 N-acetylation activity. An enrichment of genes involved in cell adhesion was observed. Additionally, human arylamine N-acetyltransferase 2 (NAT2) transcripts were observed in the complete NAT1 knockout cell lines (CRISPR 2–19 and CRISPR 5–50). This study provides further evidence that NAT1 functions as more than just a drug metabolizing enzyme given the observation that differences in NAT1 activity have significant impacts on global gene expression. Additionally, our data suggests the knockout of NAT1 results in transcription of its isozyme NAT2.
Collapse
Affiliation(s)
- Samantha M Carlisle
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, United States.,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Patrick J Trainor
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, United States.,Division of Cardiovascular Medicine, School of Medicine, University of Louisville, Louisville, KY, United States
| | - Mark A Doll
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, United States
| | - David W Hein
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
13
|
Han Y, Wu Y, Xu H, Wang J, Xu B. The impact of hormone receptor on the clinical outcomes of HER2-positive breast cancer: a population-based study. Int J Clin Oncol 2022; 27:707-716. [PMID: 35041101 PMCID: PMC8956538 DOI: 10.1007/s10147-022-02115-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/03/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND To investigate the impact of hormone receptor (HR) on the clinicopathological characteristics and prognosis of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. METHODS Using the Surveillance, Epidemiology, and End Results database, we enrolled patients diagnosed with HER2-positive breast cancer between 2010 and 2016, which were successively assessed for eligibility and categorized into HR + /HER2 + and HR-/HER2 + subgroups. Clinicopathological characteristics were undergone comparative analyses with the baseline distinctions calibrated by propensity score matching, while the survival outcomes were compared using Kaplan-Meier method with log-rank tests. RESULTS A total of 46,803 HER2-positive breast cancer patients were identified, of which 32,919 individuals were HR + /HER2 + subtype and 13,884 individuals were HR-/HER2 + subtype, respectively. Comparatively, HR + /HER2 + breast cancer presented a lower histological grade, a smaller tumor size, a lower nodal involvement, and a lower rate of de novo stage IV disease. Substantial heterogeneity was detected in the metastatic patterns of organ-specific involvement between the two subgroups with initial metastasis. Overall, patients with HR + /HER2 + tumors had increasingly favorable prognosis in terms of overall survival and breast cancer-specific survival than patients with the HR-/HER2 + subtype. However, this kind of tendency exhibited disparities associated with HR-specific subtypes based on estrogen receptor (ER) and progesterone receptor (PgR) status, in which ER-/PgR + tended to present the worst prognosis. CONCLUSION This study revealed profound heterogeneity associated with HR status in the clinical outcomes of HER2-positive breast cancer regarding clinicopathological features, metastatic patterns, and prognosis. Prospective studies to optimize therapeutic strategies for HER2-positive subgroups are warranted.
Collapse
Affiliation(s)
- Yiqun Han
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yun Wu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Hangcheng Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
14
|
Jia X, Lei H, Jiang X, Yi Y, Luo X, Li J, Chen Y, Liu S, Yang C. Identification of Crucial lncRNAs for Luminal A Breast Cancer through RNA Sequencing. Int J Endocrinol 2022; 2022:6577942. [PMID: 35692369 PMCID: PMC9184229 DOI: 10.1155/2022/6577942] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/16/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The growing body of evidence indicates aberrant expression of long noncoding RNAs (lncRNAs) in breast cancer. Nevertheless, a few studies have focused on identifying key lncRNAs for patients with luminal A breast cancer. In our study, we tried to find key lncRNAs and mRNAs in luminal A breast cancer. METHODS RNA sequencing was performed to identify differentially expressed mRNAs (DEmRNAs) and differentially expressed lncRNAs (DElncRNAs) in luminal A breast cancer. The protein-protein interaction (PPI), DElncRNA-DEmRNA coexpression, DElncRNA-nearby DEmRNA interaction networks, and functional annotation were performed to uncover the function of DEmRNAs. Online databases were used to validate the RNA sequencing result. The diagnostic value of candidate mRNAs was evaluated by receiver operating characteristic (ROC) curve analysis. RESULTS A total number of 1451 DEmRNAs and 272 DElncRNAs were identified. Several hub proteins were identified in the PPI network, including TUBB3, HIST2H3C, MCM2, MYOC, NEK2, LIPE, FN1, FOXJ1, S100A7, and DLK1. In the DElncRNA-DEmRNA coexpression, some hub lncRNAs were identified, including AP001528.2, LINC00968, LINC02202, TRHDE-AS1, LINC01140, AL354707.1, AC097534.1, MIR222HG, and AL662844.4. The mRNA expression result of TFF1, COL10A1, LEP, PLIN1, PGM5-AS1, and TRHDE-AD1 in the GSE98793 was consistent with the RNA sequencing result. The protein expression results of TUBB3, MCM2, MYOC, FN1, S100A7, and TFF1 were consistent with the mRNA expression result COL10A1, LEP, PLIN1, PGM5-AS1, and TRHDE-AD1 were capable of discriminating luminal A breast cancer and normal controls. Four lncRNA-nearby and coexpressed mRNA pairs of HOXC-AS3-HOXC10, AC020907.2-FXYD1, AC026461.1-MT1X, and AC132217.1-IGF2 were identified. AMPK (involved LIPE and LEP) and PPAR (involved PLIN1) were two significantly enriched pathways in luminal A breast cancer. CONCLUSION This study could be helpful in unraveling the pathogenesis and providing novel therapeutic strategies for luminal A breast cancer.
Collapse
Affiliation(s)
- Xinjian Jia
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Hai Lei
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Xuemei Jiang
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Ying Yi
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Xue Luo
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Junyan Li
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Yu Chen
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Sha Liu
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| | - Chengcheng Yang
- Department of Breast Surgery, Deyang People's Hospital, Deyang, Sichuan Province, China
| |
Collapse
|
15
|
Song D, He H, Indukuri R, Huang Z, Stepanauskaite L, Sinha I, Haldosén LA, Zhao C, Williams C. ERα and ERβ Homodimers in the Same Cellular Context Regulate Distinct Transcriptomes and Functions. Front Endocrinol (Lausanne) 2022; 13:930227. [PMID: 35872983 PMCID: PMC9299245 DOI: 10.3389/fendo.2022.930227] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
The two estrogen receptors ERα and ERβ are nuclear receptors that bind estrogen (E2) and function as ligand-inducible transcription factors. They are homologues and can form dimers with each other and bind to the same estrogen-response element motifs in the DNA. ERα drives breast cancer growth whereas ERβ has been reported to be anti-proliferative. However, they are rarely expressed in the same cells, and it is not fully investigated to which extent their functions are different because of inherent differences or because of different cellular context. To dissect their similarities and differences, we here generated a novel estrogen-dependent cell model where ERα homodimers can be directly compared to ERβ homodimers within the identical cellular context. By using CRISPR-cas9 to delete ERα in breast cancer MCF7 cells with Tet-Off-inducible ERβ expression, we generated MCF7 cells that express ERβ but not ERα. MCF7 (ERβ only) cells exhibited regulation of estrogen-responsive targets in a ligand-dependent manner. We demonstrated that either ER was required for MCF7 proliferation, but while E2 increased proliferation via ERα, it reduced proliferation through a G2/M arrest via ERβ. The two ERs also impacted migration differently. In absence of ligand, ERβ increased migration, but upon E2 treatment, ERβ reduced migration. E2 via ERα, on the other hand, had no significant impact on migration. RNA sequencing revealed that E2 regulated a transcriptome of around 800 genes via each receptor, but over half were specific for either ERα or ERβ (417 and 503 genes, respectively). Functional gene ontology enrichment analysis reinforced that E2 regulated cell proliferation in opposite directions depending on the ER, and that ERβ specifically impacted extracellular matrix organization. We corroborated that ERβ bound to cis-regulatory chromatin of its unique proposed migration-related direct targets ANXA9 and TFAP2C. In conclusion, we demonstrate that within the same cellular context, the two ERs regulate cell proliferation in the opposite manner, impact migration differently, and each receptor also regulates a distinct set of target genes in response to E2. The developed cell model provides a novel and valuable resource to further complement the mechanistic understanding of the two different ER isoforms.
Collapse
Affiliation(s)
- Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Maternal and Child Health Care Hospital of Shandong Province, Jinan, China
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Huan He
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- School of Public Health, Jilin University, Changchun, China
| | - Rajitha Indukuri
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaite
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
| | - Indranil Sinha
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Arne Haldosén
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Solna, Sweden
- *Correspondence: Cecilia Williams,
| |
Collapse
|
16
|
Zhang T, Yu S, Zhao S. ANXA9 as a novel prognostic biomarker associated with immune infiltrates in gastric cancer. PeerJ 2021; 9:e12605. [PMID: 35003923 PMCID: PMC8684324 DOI: 10.7717/peerj.12605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/16/2021] [Indexed: 11/26/2022] Open
Abstract
Background Gastric cancer (GC) is the most prevalent malignancy among the digestive system tumors. Increasing evidence has revealed that lower mRNA expression of ANXA9 is associated with a poor prognosis in colorectal cancer. However, the role of ANXA9 in GC remains largely unknown. Material and Methods The Gene Expression Profiling Interactive Analysis (GEPIA) and Human Protein Atlas databases were used to investigate the expression of ANXA9 in GC, which was then validated in the four Gene Expression Omnibus (GEO) datasets. The diagnostic value of ANXA9 for GC patients was demonstrated using a receiver operating characteristic (ROC) curve. The correlation between ANXA9 expression and clinicopathological parameters was analyzed in The Cancer Genome Atlas (TCGA) and UALCAN databases. The Kaplan-Meier (K-M) survival curve was used to elucidate the relationship between ANXA9 expression and the survival time of GC patients. We then performed a gene set enrichment analysis (GSEA) to explore the biological functions of ANXA9. The relationship of ANXA9 expression and cancer immune infiltrates was analyzed using the Tumor Immune Estimation Resource (TIMER). In addition, the potential mechanism of ANXA9 in GC was investigated by analyzing its related genes. Results ANXA9 was significantly up-regulated in GC tissues and showed obvious diagnostic value. The expression of ANXA9 was related to the age, gender, grade, TP53 mutation, and histological subtype of GC patients. We also found that ANXA9 expression was associated with immune-related biological function. ANXA9 expression was also correlated with the infiltration level of CD8+ T cells, neutrophils, and dendritic cells in GC. Additionally, copy number variation (VNV) of ANXA9 occurred in GC patients. Function enrichment analyses revealed that ANXA9 plays a role in the GC progression by interacting with its related genes. Conclusions Our results provide strong evidence of ANXA9 expression as a prognostic indicator related to immune responses in GC.
Collapse
|
17
|
Albaradei S, Uludag M, Thafar MA, Gojobori T, Essack M, Gao X. Predicting Bone Metastasis Using Gene Expression-Based Machine Learning Models. Front Genet 2021; 12:771092. [PMID: 34858485 PMCID: PMC8631472 DOI: 10.3389/fgene.2021.771092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
Bone is the most common site of distant metastasis from malignant tumors, with the highest prevalence observed in breast and prostate cancers. Such bone metastases (BM) cause many painful skeletal-related events, such as severe bone pain, pathological fractures, spinal cord compression, and hypercalcemia, with adverse effects on life quality. Many bone-targeting agents developed based on the current understanding of BM onset's molecular mechanisms dull these adverse effects. However, only a few studies investigated potential predictors of high risk for developing BM, despite such knowledge being critical for early interventions to prevent or delay BM. This work proposes a computational network-based pipeline that incorporates a ML/DL component to predict BM development. Based on the proposed pipeline we constructed several machine learning models. The deep neural network (DNN) model exhibited the highest prediction accuracy (AUC of 92.11%) using the top 34 featured genes ranked by betweenness centrality scores. We further used an entirely separate, "external" TCGA dataset to evaluate the robustness of this DNN model and achieved sensitivity of 85%, specificity of 80%, positive predictive value of 78.10%, negative predictive value of 80%, and AUC of 85.78%. The result shows the models' way of learning allowed it to zoom in on the featured genes that provide the added benefit of the model displaying generic capabilities, that is, to predict BM for samples from different primary sites. Furthermore, existing experimental evidence provides confidence that about 50% of the 34 hub genes have BM-related functionality, which suggests that these common genetic markers provide vital insight about BM drivers. These findings may prompt the transformation of such a method into an artificial intelligence (AI) diagnostic tool and direct us towards mechanisms that underlie metastasis to bone events.
Collapse
Affiliation(s)
- Somayah Albaradei
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.,Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmut Uludag
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Maha A Thafar
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.,College of Computers and Information Technology, Taif University, Taif, Saudi Arabia
| | - Takashi Gojobori
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Magbubah Essack
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
18
|
Hsa-miR-105-1 Regulates Cisplatin-Resistance in Ovarian Carcinoma Cells by Targeting ANXA9. ACTA ACUST UNITED AC 2021; 2021:6662486. [PMID: 33680718 PMCID: PMC7929659 DOI: 10.1155/2021/6662486] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/02/2021] [Indexed: 01/29/2023]
Abstract
Purpose Cisplatin is one of the most effective drugs for treating ovarian carcinoma (OC), which is among the most lethal types of carcinoma. However, the chemoresistance to cisplatin that develops over time leads to a poor clinical outcome for many OC patients. Therefore, it is necessary to clearly understand the molecular mechanisms of chemoresistance. In this study, we examined how Hsa-miR-105-1 functions in cisplatin-resistant OC cells. Methods The levels of Hsa-miR-105-1 expression in cisplatin-sensitive and resistant OC cell lines were detected by qRT-PCR. The target gene of Hsa-miR-105-1 was predicted by using the TargetScan and Starbase databases and verified by the double luciferase reporter gene assay. The target gene of Hsa-miR-105-1 was identified as ANXA9, and ANXA9 expression was evaluated by qRT-PCR, western blotting, and immunofluorescence. To validate the function of Hsa-miR-105-1 in OC cells, we silenced or overexpressed Hsa-miR-105-1 in cisplatin-sensitive or resistant OC cell lines, respectively. Furthermore, the expression levels of several apoptosis-related proteins, including P53, P21, E2F1, Bcl-2, Bax, and caspase-3, were examined by western blot analysis. Results The levels of Hsa-miR-105-1 expression were abnormally downregulated in cisplatin-resistant OC cells, while ANXA9 expression was significantly upregulated in those cells. Treatment with an Hsa-miR-105-1 inhibitor promoted the expression of ANXA9 mRNA and protein, enhanced the resistance to cisplatin, and attenuated the cell apoptosis induced by cisplatin in cisplatin-sensitive OC cells. Moreover, treatment with Hsa-miR-105-1 mimics inhibited ANXA9 expression, which further increased the levels of P53, P21, and Bax expression and decreased the levels of E2F1 and Bcl-2 expression, finally resulting in an increased sensitivity to cisplatin in cisplatin-resistant OC cells. Conclusion We found that a downregulation of Hsa-miR-105-1 expression enhanced cisplatin-resistance, while an upregulation of Hsa-miR-105-1 restored the sensitivity of OC cells to cisplatin. The Hsa-miR-105-1/ANXA9 axis plays an important role in the cisplatin-resistance of OC cells.
Collapse
|
19
|
Spadazzi C, Mercatali L, Esposito M, Wei Y, Liverani C, De Vita A, Miserocchi G, Carretta E, Zanoni M, Cocchi C, Bongiovanni A, Recine F, Kang Y, Ibrahim T. Trefoil factor-1 upregulation in estrogen-receptor positive breast cancer correlates with an increased risk of bone metastasis. Bone 2021; 144:115775. [PMID: 33249323 DOI: 10.1016/j.bone.2020.115775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Bone is one of the most preferred sites of metastatic spread from different cancer types, including breast cancer. However, different breast cancer subtypes exhibit distinct metastatic behavior in terms of kinetics and anatomic sites of relapse. Despite advances in the diagnosis, the identification of patients at high-risk of bone recurrence is still an unmet clinical need. We conducted a retrospective analysis, by gene expression and immunohistochemical assays, on 90 surgically resected breast cancer samples collected from patients who experienced no evidence of distant metastasis, bone or visceral metastasis in order to identify a primary tumor-derived marker of bone recurrence. We identified trefoil factor-1 (pS2 or TFF1) as strictly correlated to bone metastasis from ER+ breast cancer. In silico analysis was carried out to confirm this observation, linking gene expression data with clinical characteristics available from public clinical datasets. Then, we investigated TFF1 function in ER+ breast cancer tumorigenesis and bone metastasis through xenograft in vivo models of MCF 7 breast cancer with gain and loss of function of TFF1. As a response to microenvironmental features in primary tumors, TFF1 expression could modulate ER+ breast cancer growth, leading to a less proliferative phenotype. Our results showed it may not play a role in late stages of bone metastasis, however further studies are warranted to understand whether it could contribute in the early-stages of the metastatic cascade. In conclusion, TFF1 upregulation in primary ER+ breast cancer could be useful to identify patients at high-risk of bone metastasis. This could help clinicians in the identification of patients who likely can develop bone metastasis and who could benefit from personalized treatments and follow-up strategies to prevent metastatic disease.
Collapse
Affiliation(s)
- Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Laura Mercatali
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | | | - Michele Zanoni
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Claudia Cocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Federica Recine
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy
| |
Collapse
|
20
|
Yu T, Wang C, Xie M, Zhu C, Shu Y, Tang J, Guan X. Heterogeneity of CTC contributes to the organotropism of breast cancer. Biomed Pharmacother 2021; 137:111314. [PMID: 33581649 DOI: 10.1016/j.biopha.2021.111314] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Circulating tumor cells (CTCs) are viewed as pro-metastasis precursors shed from primary tumors or metastatic sites. The phenotypic and molecular heterogeneity of CTCs is associated with breast cancer progression and prognosis. Therefore, we divided CTCs into several subtypes according to their differences in biomarker status, epithelial/mesenchymal phenotype, aggregation status, and other factors to summarize their characteristics. Considering that the organ-specific metastasis is a hallmark of breast cancer, we adopted the "seed and soil" model to further analyze the relationship between the heterogeneity of CTCs and the organotropism of breast cancer. We speculated that CTCs might not only develop their genetic potential but communicate with surroundings, including chemokine systems, hemocytes, and extracellular matrix components, to regulate the organ-specific metastases of breast cancer.
Collapse
Affiliation(s)
- Tao Yu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Cenzhu Wang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Mengyan Xie
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Chengjun Zhu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China.
| | - Xiaoxiang Guan
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, People's Republic of China.
| |
Collapse
|
21
|
Bertho M, Fraisse J, Patsouris A, Cottu P, Arnedos M, Pérol D, Jaffré A, Goncalves A, Lebitasy MP, D’Hondt V, Dalenc F, Ferrero JM, Levy C, Dabakuyo S, Rouzier R, Penault-Llorca F, Uwer L, Eymard JC, Breton M, Chevrot M, Thureau S, Petit T, Simon G, Frénel JS. Real-life prognosis of 5041 bone-only metastatic breast cancer patients in the multicenter national observational ESME program. Ther Adv Med Oncol 2021; 13:1758835920987657. [PMID: 33613700 PMCID: PMC7841864 DOI: 10.1177/1758835920987657] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bone-only (BO) metastatic breast cancer (MBC) is considered a more favorable entity than other MBC presentations. However, only few retrospective series and data from selected randomized controlled trials have been reported so far. METHODS Using the French national multicenter ESME (Epidemiological Strategy and Medico Economics) Data Platform, the primary objective of our study was to compare the overall survival (OS) of patients with BO versus non-BO MBC at diagnosis, with adjustment on main prognostic factors using a propensity score. Secondary objectives were to compare first-line progression-free survival (PFS1), describe treatment patterns, and estimate factors associated with OS. RESULTS Out of 20,095 eligible women, 5041 (22.4%) patients had BO disease [hormone-receptor positive (HR+)/human epidermal growth-factor-receptor-2 negative (HER2-), n = 4 102/13,229 (31%); HER2+, n = 644/3909 (16.5%); HR-/HER2-, n = 295/2 957 (10%)]. BO MBC patients had a better adjusted OS compared with non-BO MBC [52.1 months (95% confidence interval (CI) 50.3-54.1) versus 34.7 months (95% CI 34.0-35.6) respectively]. The 5-year OS rate of BO MBC patients was 43.4% (95% CI 41.7-45.2). They also had a better PFS1 [13.1 months (95% CI 12.6-13.8) versus 8.5 months (95% CI 8.3-8.7), respectively]. This observation could be repeated in all subtypes. BO disease was an independent prognostic factor of OS [hazard ratio 0.68 (95% CI 0.65-0.72), p < 0.0001]. Results were concordant in all analyses. CONCLUSION BO MBC patients have better outcomes compared with non-BO MBC, consistently, through all MBC subtypes.
Collapse
Affiliation(s)
- Marion Bertho
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest – Paul Papin, Angers, France
| | - Julien Fraisse
- Biometrics Unit, Regional Cancer Institute of Montpellier (ICM), Montpellier, France
| | - Anne Patsouris
- Department of Medical Oncology, Institut de Cancérologie de l’Ouest – Paul Papin, Angers, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - David Pérol
- Biostatistic Unit, Clinical Research and Innovation Department, Centre Léon Bérard, Lyon, France
| | - Anne Jaffré
- Department of Medical Information, Institut Bergonié, Bordeaux, France
| | - Anthony Goncalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Marie-Paule Lebitasy
- Clinical Research and Innovation Department, Centre Oscar Lambret, Lille, France
| | - Véronique D’Hondt
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, France
| | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Institut Centre Antoine Lacassagne, Nice, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sandrine Dabakuyo
- National Quality of Life and Cancer Clinical Research Platform, Centre Georges François Leclerc, Dijon, France
| | - Roman Rouzier
- Department of Surgical Oncology, Institut Curie, Saint-Cloud, France
| | | | - Lionel Uwer
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès- Nancy, France
| | | | - Mathias Breton
- Department of Medical Information, Centre Eugène Marquis, Rennes, France
| | | | - Sébastien Thureau
- Department of Radiation Oncology, Centre Henri Becquerel, Rouen, France
| | - Thierry Petit
- Department of Medical Oncology, Centre Paul Strauss, Strasbourg, France
| | | | - Jean-Sébastien Frénel
- Department of Medical Oncology, ICO René Gauducheau, Boulevard Jacques Monod, Saint Herblain, Pays de la Loire 44805, France
| |
Collapse
|
22
|
Chen M, Zou S, He C, Zhou J, Li S, Shen M, Cheng R, Wang D, Zou T, Yan X, Huang Y, Shen J. Transactivation of SOX5 by Brachyury promotes breast cancer bone metastasis. Carcinogenesis 2020; 41:551-560. [PMID: 31713604 PMCID: PMC7350557 DOI: 10.1093/carcin/bgz142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/22/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
The bone marrow has been long known to host a unique environment amenable to colonization by metastasizing tumor cells. Yet, the underlying molecular interactions which give rise to the high incidence of bone metastasis (BM) in breast cancer patients have long remained uncharacterized. In our study, in vitro and in vivo assays demonstrated that Brachyury (Bry) could promote breast cancer BM. Bry drives epithelial–mesenchymal transition (EMT) and promotes breast cancer aggressiveness. As an EMT driver, SOX5 involves in breast cancer metastasis and the specific function in BM. Chromatin immunoprecipitation (ChIP) assays revealed SOX5 is a direct downstream target gene of Bry. ChIP analysis and reporter assays identified two Bry-binding motifs; one consistent with the classic conserved binding sequence and the other a new motif sequence. This study demonstrates for the first time that Bry promotes breast cancer cells BM through activating SOX5. In clinical practice, targeting the Bry-Sox5-EMT pathway is evolving into a promising avenue for the prevention of bone metastatic relapse, therapeutic resistance and other aspects of breast cancer progression. Brachyury directly regulates the expression of SOX5 by binding to two motifs in its promoter region. The Bry-SOX5-EMT pathway may represent a potential target to develop treatments to prevent and treat bone metastasis from breast cancer.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Shitao Zou
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Suoyuan Li
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Minghong Shen
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Rulei Cheng
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Donglai Wang
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Tianming Zou
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Xueqi Yan
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Ying Huang
- Department of Ultrasonography, the Fifth People’s Hospital of Suzhou, Suzhou, Jiangsu, People’s Republic of China
| | - Jun Shen
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
- To whom correspondence should be addressed. Tel: 008618112603158; Fax: 008651262362502,
| |
Collapse
|
23
|
Chintala S, Levan J, Robinson K, Quist K, Katzenellenbogen RA. Genes Regulated by HPV 16 E6 and High Expression of NFX1-123 in Cervical Cancers. Onco Targets Ther 2020; 13:6143-6156. [PMID: 32617009 PMCID: PMC7326398 DOI: 10.2147/ott.s251926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose High-risk human papillomaviruses (HR HPV) cause cervical cancer, and in these cancers, HPV type 16 is the most common HR type. The HR viral oncogenes E6 and E7 partner with cellular proteins to drive cancer and modulate immune pathways; previously, we demonstrated in keratinocytes that HPV 16 E6 and high expression of the endogenous host protein partner NFX1-123 led to the increased expression of multiple genes, including Notch1, secretory leukocyte peptidase inhibitor (SLPI), and retinoic acid early transcript 1G (RAET1G). The present study was conducted to determine if NFX1-123 was highly expressed in cervical cancer and if genes increased by NFX1-123 and 16E6 in keratinocytes were also increased in cervical cancers. Materials and Methods The Cancer Genome Atlas (TCGA) database and The Human Protein Atlas database were used to compare relative mRNA and protein gene expression, respectively, in the normal cervix and cervical cancers. Formalin-fixed paraffin-embedded (FFPE) normal cervix and HPV 16 positive cervical cancer samples were analyzed for relative protein expression by immunohistochemical staining. Protein expression of a subset of regulated genes was quantified by Western blot of HPV positive and negative cell lines. Results Immunohistochemical staining of HPV 16 positive cervical dysplasias and cancers revealed high NFX1-123, Ki67, and Notch1 expression. NFX1 and NFX1L1 mRNA levels were increased in cervical cancers compared to normal cervix in the TCGA database. Fourteen genes previously identified as upregulated in keratinocytes with 16E6 and overexpressed NFX1-123 also had high mRNA expression and selected genes had high protein expression in cervical cancers and cell lines. Conclusion In cervical cancer, NFX1-123 is highly expressed, and 16E6 and NFX1-123 together alter the expression of a wide set of genes. The involvement of these genes in cell proliferation, differentiation, invasion, and metastasis provides further insight into potential ways that HR HPVs promote cancer initiation and maintenance.
Collapse
Affiliation(s)
- Sreenivasulu Chintala
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Justine Levan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristin Robinson
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kevin Quist
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
24
|
Tian Y, Li J, Su S, Cao Y, Wang Z, Zhao S, Zhao H. PCOS-GWAS Susceptibility Variants in THADA, INSR, TOX3, and DENND1A Are Associated With Metabolic Syndrome or Insulin Resistance in Women With PCOS. Front Endocrinol (Lausanne) 2020; 11:274. [PMID: 32425888 PMCID: PMC7203965 DOI: 10.3389/fendo.2020.00274] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/14/2020] [Indexed: 12/20/2022] Open
Abstract
Polycystic ovary syndrome is characterized by reproductive and metabolic disturbances throughout the female lifespan. Therefore, this study aimed to determine whether genome-wide association studies (GWAS)-identified risk variants for PCOS could confer risk of metabolic syndrome (MS) or insulin resistance (IR). Fifteen independent SNPs mapping to 11 GWAS loci genotyped in a total of 2,082 Han Chinese women independent of previous GWAS and phenotype-genotype correlations were assessed. The CC group for rs12478601 in THADA was associated with decreased rate of MS after adjustment for age (23.2 vs. 27%, P = 0.042, OR = 0.81). Using a dominant model, the GG+AG group for rs2059807 in INSR was associated with increased risk of MS after adjustment for age (26.8 vs. 22.5%, P = 0.023, OR = 1.27). The GG + GT group for rs4784165 in TOX3 was found to be associated with an increased rate of IR after adjustment for age and BMI(53.3 vs. 48.5%, P = 0.027, OR = 1.27). The GG+AG group for rs2479106 in DENND1A was associated with a decreased rate of IR (48.3 vs. 53.6%, adjusted P = 0.039, OR = 0.80). After exclusion of PCOS cases with a family history of diabetes, hypertension, or dyslipidemia, the phenotype-genotype correlations between the genes INSR and TOX3 and MS or IR were still significant (P < 0.05). Three SNPs (rs13429458 in THADA, rs10818854 in DENND1A, and rs2059807 in INSR) were significantly associated with IR; however, their association was not significant after adjustment for age and BMI. This genotype-phenotype study thus provides clues that THADA, INSR, TOX3, and DENND1A play a role in PCOS possibly through a metabolic disorder-related pathway.
Collapse
Affiliation(s)
- Ye Tian
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Jingyu Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Shizhen Su
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Yongzhi Cao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Zhao Wang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Shigang Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Han Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| |
Collapse
|
25
|
Ahmad M, Shah AA. Predictive role of single nucleotide polymorphism (rs11614913) in the development of breast cancer in Pakistani population. Per Med 2020; 17:213-227. [PMID: 32320336 DOI: 10.2217/pme-2019-0086] [Citation(s) in RCA: 5] [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
Aim: miRNAs play an important role in breast cancer (BC). Variations in miRNAs influence their maturation, expression and consequently regulation of their target genes. Materials & methods: In this study, single nucleotide polymorphism rs11614913 was genotyped in BC patients (n = 300) and 230 controls by employing tetra primer amplification refractory mutation system PCR and Sanger sequencing (Macrogen Korea). Results: A significant difference was observed in the genotypes through co-dominant (χ2.#x00A0;= 42.03; p < 0.0001), additive (odds ratio [OR] = 0.6441 [0.4887-0.8490, 95% confidence interval]; p < 0.0019), dominant (OR = 0.3996 [0.2809-0.5686], p < 0.0001) and recessive (OR = 0.2993 [0.1220-0.7347], p < 0.009) statistical models showed decreased risk association of C allele with BC. Conclusion: Females having CT genotype are at higher risk of BC as compared with those having CC genotype.
Collapse
Affiliation(s)
- Mushtaq Ahmad
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Pakistan
| | - Aftab Ali Shah
- Department of Biotechnology, Faculty of Biological Sciences, University of Malakand, Chakdara, Pakistan
| |
Collapse
|
26
|
N-Acetyltransferase 1 Knockout Elevates Acetyl Coenzyme A Levels and Reduces Anchorage-Independent Growth in Human Breast Cancer Cell Lines. JOURNAL OF ONCOLOGY 2019; 2019:3860426. [PMID: 31531019 PMCID: PMC6720663 DOI: 10.1155/2019/3860426] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Abstract
Elevated expression of N-acetyltransferase 1 (NAT1) is associated with invasive and lobular breast carcinomas as well as with bone metastasis following an epithelial-to-mesenchymal transition. We investigated the effect of NAT1 gene deletion in three different human breast cancer cell lines, MDA-MB-231, MCF-7, and ZR-75-1. Human NAT1 was knocked out using CRISPR/Cas9 technology and two different guide RNAs. None of the NAT1 knockout (KO) cell lines exhibited detectable NAT1 activity when measured using its selective substrate p-aminobenzoic acid (PABA). Endogenous acetyl coenzyme A levels (cofactor for acetylation pathways) in NAT1 KO cell lines were significantly elevated in the MDA-MB-231 (p < 0.001) and MCF-7 (p=0.0127) but not the ZR-75-1 (p > 0.05). Although the effects of NAT1 KO on cell-doubling time were inconsistent across the three breast cancer cell lines, the ability of the NAT1 KO cell lines to form anchorage-independent colonies in soft agar was dramatically and consistently reduced in each of the breast cancer cell lines. The NAT1 KO clones for MDA-MB-231, MCF-7, and ZR-75-1 had a reduction greater than 20-, 6-, and 7- folds in anchorage-independent cell growth, respectively, compared to their parental cell lines (p < 0.0001, p < 0.0001, and p < 0.05, respectively). The results indicate that NAT1 may be an important regulator of cellular acetyl coenzyme A levels and strongly suggest that elevated NAT1 expression in breast cancers contribute to their anchorage-independent growth properties and ultimately metastatic potential.
Collapse
|
27
|
Al-Eitan LN, Rababa'h DM, Alghamdi MA, Khasawneh RH. Correlation between Candidate Single Nucleotide Variants and Several Clinicopathological Risk Factors Related to Breast Cancer in Jordanian Women: A Genotype-Phenotype Study. J Cancer 2019; 10:4647-4654. [PMID: 31528229 PMCID: PMC6746117 DOI: 10.7150/jca.33857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/12/2019] [Indexed: 11/05/2022] Open
Abstract
This study aim to investigate the association of breast cancer risk and prognostic factors with single nucleotide variants of the BRCA1, BRCA2, DAPK1, MMP9, TOX3, and TP53 genes in Jordanian women. Blood samples were collected from 230 Jordanian breast cancer patients for use in DNA extraction followed by genotyping and subsequent statistical analysis. We found that two single nucleotide variants (SNVs) of the BRCA2 gene, namely rs1799944 and rs766173, were significantly associated with breastfeeding status. Likewise, the rs11141901 and rs1041326 SNVs of the DAPK1 gene were linked with co-morbidity (p-value = 0.002) and family history of BC (p-value = 0.015), while the rs1045042 SNV of the same gene was associated with both allergy (p-value = 0.001) and family history of BC (p-value = 0.02). Tumor differentiation was correlated with the DAPK1 SNVs rs11141901 (p-value = 0.041) and rs1041326 (p-value = 0.005). Additionally, the rs2250889 SNV of the MMP9 gene was significantly associated with HER2 status, whereas the TP53 SNVs rs12951053 and rs1042522 were associated with age at menarche (p-value = 0.043) and breastfeeding status (p-value = 0.013), respectively. In contrast, the TP53 SNV rs2287497 was significantly linked to age at first pregnancy (p-value = 0.001), smoking (p-value = 0.041), and axillary lymph node status (p-value = 6e-4). No such association was found for the BRCA1 and TOX3 SNVs. The current findings suggest significant associations between certain SNVs and breast cancer risk and prognosis in Jordanian women.
Collapse
Affiliation(s)
- Laith N Al-Eitan
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan.,Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Doaa M Rababa'h
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | | | - Rame H Khasawneh
- Department of Hematopathology, King Hussein Medical Center (KHMC), Jordan Royal Medical Services (RMS), Amman 11118, Jordan
| |
Collapse
|
28
|
Xiao B, Hang J, Lei T, He Y, Kuang Z, Wang L, Chen L, He J, Zhang W, Liao Y, Sun Z, Li L. Identification of key genes relevant to the prognosis of ER-positive and ER-negative breast cancer based on a prognostic prediction system. Mol Biol Rep 2019; 46:2111-2119. [PMID: 30888555 DOI: 10.1007/s11033-019-04663-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 01/29/2019] [Indexed: 12/24/2022]
Abstract
Few prognostic indicators with differential expression have been reported among the differing ER statuses. We aimed to screen important breast cancer prognostic genes related to ER status and to construct an efficient prognostic prediction system. mRNA expression profiles were downloaded from TCGA and GSE70947 dataset. Two hundred seventy-one overlapping differentially expressed genes (DEGs) between the ER- and ER+ breast cancer samples were identified. Among the 271 DEGs, 109 prognostically relevant mRNAs were screened. mRNAs such as RASEF, ITM2C, CPEB2, ESR1, ANXA9, and VASN correlated strongly with breast cancer prognosis. Three modules, which contained 28, 9 and 8 enriched DEGs, were obtained from the network, and the DEGs in these modules were enriched in response to hormone stimulus, epithelial cell development, and host cell entry. Using bayes discriminant analysis, 48 signature genes were screened. We constructed a prognostic prediction system using the 48 signature genes and validated this system as relatively accurate and reliable. The DEGs might be closely associated with the prognosis in patients with breast cancer. We validated the effectiveness of our prognostic prediction system by GEO database. Therefore, this system might be a useful tool for preliminary screening and validation of potential prognosis indicators for ER+ breast cancer derived from mechanistic research.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Jianfeng Hang
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Ting Lei
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Yongyin He
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Zhenzhan Kuang
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Li Wang
- Department of Medical Research, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong Province, China
| | - Lidan Chen
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Jia He
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Weiyun Zhang
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Yang Liao
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China
| | - Zhaohui Sun
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China.
| | - Linhai Li
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, No. 111, Liuhua Road, Yuexiu district, Guangzhou, 510010, Guangdong Province, China.
| |
Collapse
|
29
|
Zhang L, Zhou R, Zhang W, Yao X, Li W, Xu L, Sun X, Zhao L. Cysteine-rich intestinal protein 1 suppresses apoptosis and chemosensitivity to 5-fluorouracil in colorectal cancer through ubiquitin-mediated Fas degradation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:120. [PMID: 30850009 PMCID: PMC6408822 DOI: 10.1186/s13046-019-1117-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/22/2019] [Indexed: 12/24/2022]
Abstract
Background Cysteine-rich intestinal protein 1 (CRIP1) is highly expressed in human intestine and aberrantly expressed in several types of tumor. However, studies on CRIP1 are limited and its role on tumor development and progression remains controversial and elusive. Methods Immunohistochemistry was performed to evaluate the expression of CRIP1 in paired normal and colorectal tumor specimens, as well as colorectal cell lines. Functional assays, such as CCK8, TUNEL assay and in vivo tumor growth assay, were used to detect the proliferation, apoptosis and response to 5-FU of CRIP1. Western blot was used to analyze Fas-mediated pathway induced by CRIP1. Rescue experiments were performed to evaluate the essential role of CRIP1 for Fas-mediated apoptosis. Results We demonstrated that CRIP1 is overexpressed in CRC tissues compared with adjacent normal mucosa. CRIP1 could dramatically recover the 5-Fluorouracil (5-FU) inhibited CRC cell proliferation in vitro and stimulate the tumor formation of CRC in vivo, probably through inhibiting CRC cell apoptosis. Moreover, CRIP1 also dramatically recovered the 5-Fluorouracil (5-FU) induced tumor cell apoptosis in vitro. Further study demonstrated that CRIP1 down-regulated the expression of Fas protein and proteins related to Fas-mediated apoptosis. CRIP1 could interact with Fas protein and stimulate its ubiquitination and degradation. In addition, a negative correlation was detected between the expression of CRIP1 and Fas protein in most of the clinical human CRC samples. Conclusion The current research reveals a vital role of CRIP1 in CRC progression, which provide a novel target for clinical drug resistance of colorectal cancer and undoubtedly contributing to the therapeutic strategies in CRC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1117-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lanzhi Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Southern Medical University, Guangzhou, China
| | - Rui Zhou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Southern Medical University, Guangzhou, China
| | - Weibin Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Southern Medical University, Guangzhou, China
| | - Xueqing Yao
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Weidong Li
- Department of Medical Oncology, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijun Xu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Southern Medical University, Guangzhou, China
| | - Xuegang Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Salom C, Álvarez-Teijeiro S, Fernández MP, Morgan RO, Allonca E, Vallina A, Lorz C, de Villalaín L, Fernández-García MS, Rodrigo JP, García-Pedrero JM. Frequent Alteration of Annexin A9 and A10 in HPV-Negative Head and Neck Squamous Cell Carcinomas: Correlation with the Histopathological Differentiation Grade. J Clin Med 2019; 8:jcm8020229. [PMID: 30744186 PMCID: PMC6406441 DOI: 10.3390/jcm8020229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023] Open
Abstract
The annexin protein superfamily has been implicated in multiple physiological and pathological processes, including carcinogenesis. Altered expression of various annexins has frequently been observed and linked to the development and progression of various human malignancies. However, information is lacking on the expression and clinical significance of annexin A9 (ANXA9) and A10 (ANXA10) in head and neck squamous cell carcinomas (HNSCC). ANXA9 and ANXA10 expression was evaluated in a large cohort of 372 surgically treated HPV-negative HNSCC patients and correlated with the clinicopathologic parameters and disease outcomes. Down-regulation of ANXA9 expression was found in 42% of HNSCC tissue samples, compared to normal epithelia. ANXA9 expression in tumors was significantly associated with oropharyngeal location and histological differentiation grade (P < 0.001). In marked contrast, ANXA10 expression was absent in normal epithelium, but variably detected in the cytoplasm of cancer cells. Positive ANXA10 expression was found in 64% of tumors, and was significantly associated with differentiation grade (P < 0.001), being also more frequent in oropharyngeal tumors (P = 0.019). These results reveal that the expression of both ANXA9 and ANXA10 is frequently altered in HNSCC and associated to the tumor differentiation grade, suggesting that they could be implicated in the pathogenesis of these cancers.
Collapse
Affiliation(s)
- Cecilia Salom
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
- CIBERONC, Av. Monforte de Lemos, 3-5. 28029, Madrid, Spain.
| | - M Pilar Fernández
- Department of Biochemistry and Molecular Biology and Institute of Biotechnology of Asturias, University of Oviedo, Julian Clavería, 33006, Oviedo, Spain.
| | - Reginald O Morgan
- Department of Biochemistry and Molecular Biology and Institute of Biotechnology of Asturias, University of Oviedo, Julian Clavería, 33006, Oviedo, Spain.
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
- CIBERONC, Av. Monforte de Lemos, 3-5. 28029, Madrid, Spain.
| | - Aitana Vallina
- Department of Pathology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
| | - Corina Lorz
- CIBERONC, Av. Monforte de Lemos, 3-5. 28029, Madrid, Spain.
- Molecular Oncology Unit, CIEMAT (ed 70A), Av. Complutense 40, 28040 Madrid, Spain.
| | - Lucas de Villalaín
- Department of Oral Surgery, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
| | - M Soledad Fernández-García
- Department of Pathology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
- CIBERONC, Av. Monforte de Lemos, 3-5. 28029, Madrid, Spain.
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias, University of Oviedo, Avda. Roma, 33011, Oviedo, Spain.
- CIBERONC, Av. Monforte de Lemos, 3-5. 28029, Madrid, Spain.
| |
Collapse
|
31
|
Zhang W, Bado I, Wang H, Lo HC, Zhang XHF. Bone Metastasis: Find Your Niche and Fit in. Trends Cancer 2019; 5:95-110. [PMID: 30755309 DOI: 10.1016/j.trecan.2018.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023]
Abstract
Metastasis to bones is determined by both intrinsic traits of metastatic tumor cells and properties appertaining to the bone microenvironment. Bone marrow niches are critical for all major steps of metastasis, including the seeding of disseminated tumor cells (DTCs) to bone, the survival of DTCs and microscopic metastases under dormancy, and the eventual outgrowth of overt metastases. In this review, we discuss the role of bone marrow niches in bone colonization. The emphasis is on complicated and dynamic nature of cancer cells-niche interaction, which may underpin the long-standing mystery of metastasis dormancy, and represent a therapeutic target for elimination of minimal residue diseases and prevention of life-taking, overt metastases.
Collapse
Affiliation(s)
- Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hin-Ching Lo
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Significant association of TOX3/LOC643714 locus-rs3803662 and breast cancer risk in a cohort of Iranian population. Mol Biol Rep 2018; 46:805-811. [PMID: 30515698 DOI: 10.1007/s11033-018-4535-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022]
Abstract
Genome-wide association studies normally focus on low penetrance and moderate to high-frequency single nucleotide polymorphisms (SNPs), which lead to genetic susceptibility to breast cancer. In this regard, the T allele of rs3803662 has been associated with breast cancer risk and with lower expression level of TOX3. We aimed to assess the risk of breast cancer associated with this polymorphism in an Iranian population. Using Tetra Primer ARMS PCR, rs3803662 was analyzed in a total of 943 individuals (430 cases and 513 healthy controls form North East of Iran). Allele frequencies and genotype distribution were analyzed in case and control samples to find out any association using the Chi-squared test and Logistic regression. All cases were pathologically confirmed; all controls were mainly healthy individuals. Genotype frequencies were found to be in agreement with HWE in controls and cases. TOX3-rs3803662 SNP was associated with breast cancer risk in our study (T vs. C allele contrast model: OR 1.36, 95% CI 1.12-1.64, Pvalue = 0.002; TT vs. CT + TT dominant model: OR 0.67, 95% CI 0.51-0.87, Pvalue = 0.003; TT vs. CT + CC recessive model: OR 1.54, 95% CI 1.02-2.30, Pvlue = 0.036). Moreover, after adjusting for age, BMI, history of previous cancer and also family history of cancer, all results, except for the recessive model, were remained significant. TOX3-rs3803662, may confer some degrees of risk of breast cancer in Iranian population. This finding is in line with similar results in other populations. It highlights the importance of TOX3 pathway in tumorigenesis.
Collapse
|
33
|
Li Y, Bai W, Zhang L. The Overexpression of CD80 and ISG15 Are Associated with the Progression and Metastasis of Breast Cancer by a Meta-Analysis Integrating Three Microarray Datasets. Pathol Oncol Res 2018; 26:443-452. [PMID: 30411299 DOI: 10.1007/s12253-018-0478-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
Breast cancer is a common cancer and could result in a substantial mortality. The study aimed to screen gene signatures associated with the development and metastasis of breast cancer and explore their regulation mechanisms. Three datasets of GSE10797, GSE8977 and GSE3744 were downloaded from GEO (Gene Expression Omnibus) database, containing 55 breast cancer samples and 27 normal samples. After data preprocessing using limma software and RMA (robust multi-array average) algorithm, DEGs (differentially expressed genes) between breast tumor and normal tissues in three individual experiments were identified using MADAM package. Function and pathway enrichment analyses were performed for the DEGs. Transcription factors and TAGs (tumor associated genes) among the DEGs were recognized and the PPI (protein-protein-interaction) network for the DEGs was constructed using Cytoscape software. The mRNA expression was analyzed via real-time quantitative PCR and protein expression was measured by western blotting. Totally, 100 DEGs were identified, including 33 up-regulated genes and 67 down-regulated genes. Among them, up-regulated DEGs such as CD80 was enriched in toll-like receptor (TLR) interaction pathway and the TAG, ISG15 was related to RIG-I-like receptor signaling pathway, while CXCL10 was involved in both of the two pathways. Whereas, the down-regulated DEG, CXCL12 was significantly associated with axon guidance pathway. Additionally, these DEGs were also pivotal nodes in the PPI network with high degrees. Besides, CXCL10 and CD80 were both interacted with IFNG. The mRNA expression of ISG15 was obviously enhanced in human breast cancer cells MCF-7, while no significant difference of CXCL10 mRNA level was found between MCF10A and MCF-7 cells. Moreover, the proteins expression levels of CD80 and ISG15 were significantly increased in MCF-7, MDA-MB-468 and MDA-MB-231 breast cancer cells than in normal MCF10A cells. CD80 might be responsible for the breast cancer's progression and metastasis via regulating innate immune system. In addition, ISG15 is identified as a crucial gene signature associated with breast cancer development and metastasis via RIG-I-like receptor signaling pathway.
Collapse
Affiliation(s)
- Yuanhang Li
- Medical Department, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.
| | - Weijun Bai
- Medical Department, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Linlin Zhang
- Medical Department, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| |
Collapse
|
34
|
Kono M, Fujii T, Matsuda N, Harano K, Chen H, Wathoo C, Joon AY, Tripathy D, Meric-Bernstam F, Ueno NT. Somatic mutations, clinicopathologic characteristics, and survival in patients with untreated breast cancer with bone-only and non-bone sites of first metastasis. J Cancer 2018; 9:3640-3646. [PMID: 30310523 PMCID: PMC6171013 DOI: 10.7150/jca.26825] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/07/2018] [Indexed: 02/01/2023] Open
Abstract
Background: Bone is the most common site of metastasis of breast cancer. Biological mechanisms of metastasis to bone may be different from mechanisms of metastasis to non-bone sites, and identification of distinct signaling pathways and somatic mutations may provide insights on biology and rational targets for treatment and prevention of bone metastasis. The aims of this study were to compare and contrast somatic mutations, clinicopathologic characteristics, and survival in breast cancer patients with bone-only versus non-bone sites of first metastasis. Methods: Primary tumor samples were collected before treatment from 389 patients with untreated primary breast cancer and distant metastasis at diagnosis. In each sample, 46 or 50 cancer-related genes were analyzed for mutations by AmpliSeq Ion Torrent next-generation sequencing. Fisher's exact test was used to identify somatic mutations associated with bone-only first metastasis. Logistic regression models were used to identify differences in detected somatic mutations, clinicopathologic characteristics, and survival between patients with bone-only first metastasis and patients with first metastasis in non-bone sites only (“other-only first metastasis”). Results: Among the 389 patients, 72 (18.5%) had bone-only first metastasis, 223 (57.3%) had other-only first metastasis, and 94 (24.2%) had first metastasis in both bone and non-bone sites. The most commonly mutated genes were TP53 (N=103), PIK3CA (N=79), AKT (N=13), and PTEN (N=2). Compared to patients with other-only first metastasis, patients with bone-only first metastasis had higher rates of hormone-receptor-positive disease, non-triple-negative subtype, and lower grade (grade 1 or 2; Nottingham grading system) (all three comparisons, p<0.001); had a lower ratio of cases of invasive ductal carcinoma to cases of invasive lobular carcinoma (p=0.002); and tended to have a higher 5-year overall survival (OS) rate (78.2% [95% confidence interval (CI), 68.6%-89.0%] vs 55.0% [95% CI, 48.1%-62.9%]; p=0.051). However, in the subgroup of patients with TP53 mutation and in the subgroup of patients with PIK3CA mutation, OS did not differ between patients with bone-only and other-only first metastasis (p=0.49 and p=0.68, respectively). In univariate analysis, the rate of TP53 mutation tended to be lower in patients with bone-only first metastasis than in those with other-only first metastasis (15.3% vs 29.1%; p=0.051). In multivariate analysis, TP53 mutation was not significantly associated with site of first metastasis (p=0.54) but was significantly associated with hormone-receptor-negative disease (p<0.001). Conclusions: We did not find associations between somatic mutations and bone-only first metastasis in patients with untreated breast cancer. Patients with bone-only first metastasis tend to have longer OS than patients with other-only first metastasis. More comprehensive molecular analysis may be needed to further understand the factors associated with bone-only metastatic disease in breast cancer.
Collapse
Affiliation(s)
- Miho Kono
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Takeo Fujii
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoko Matsuda
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenichi Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chetna Wathoo
- Sheikh Khalifa Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aron Y Joon
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Funda Meric-Bernstam
- Sheikh Khalifa Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Investigational Cancer Therapeutics (Phase I Trials Department), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
35
|
Gong Y, Zhang J, Ji P, Ling H, Hu X, Shao Z. Incidence proportions and prognosis of breast cancer patients with bone metastases at initial diagnosis. Cancer Med 2018; 7:4156-4169. [PMID: 29984914 PMCID: PMC6089179 DOI: 10.1002/cam4.1668] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Population-based data on the incidence and prognosis of bone metastases at diagnosis of breast cancer are currently limited. Hence, we conducted this study to analyze the incidence proportions and prognostic factors of patients with breast cancer and bone metastases at the time of cancer diagnosis. MATERIALS AND METHODS Patients with primary invasive breast cancer and bone metastases at initial diagnosis between 2010 and 2014 were identified using the Surveillance, Epidemiology, and End Results (SEER) dataset and Fudan University Shanghai Cancer Center (FUSCC) cohort. Multivariable logistic regression was performed to identify predictors of the presence of bone metastases at diagnosis. Univariate and multivariate analyses were performed to determine the effects of each variable on survival. RESULTS Of 229, 195 patients from SEER database included in the analysis, 8295 patients had bone metastases at initial diagnosis, reflecting 3.6% of the entire study population, and 65.1% of the subset with metastatic disease to any distant site. Patients with hormone receptor (HR)-positive human epidermal growth factor receptor 2 (HER2)-negative represented the highest incidence proportions among patients with metastatic disease (73.9%). Among entire cohort, multivariable logistic regression identified eight factors as predictors of the presence of bone metastases at diagnosis. Median OS for the patients with bone metastases in SEER and FUSCC cohorts was 30.0 and 68.2 months, respectively. Patients with HR-positive HER2-positive subtype had the longest median OS, and patients with triple-negative subtype showed the shortest median OS. Multivariable Cox model in SEER cohort confirmed age, histology, grade, tumor subtype, extraosseous metastatic sites, history of primary surgery, insurance status, marital status, and income as independent prognostic factors for both OS and BCSS. CONCLUSIONS The findings of this study provide population-based estimates of the incidence and prognosis for patients with bone metastases at initial diagnosis of breast cancer.
Collapse
Affiliation(s)
- Yue Gong
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jing Zhang
- Department of OrthopedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Peng Ji
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hong Ling
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Xin Hu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
| | - Zhi‐Ming Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Institutes of Biomedical ScienceFudan UniversityShanghaiChina
| |
Collapse
|
36
|
Minchin RF, Butcher NJ. Trimodal distribution of arylamine N-acetyltransferase 1 mRNA in breast cancer tumors: association with overall survival and drug resistance. BMC Genomics 2018; 19:513. [PMID: 29969986 PMCID: PMC6029418 DOI: 10.1186/s12864-018-4894-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 06/25/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Arylamine N-acetyltransferase 1 (NAT1) is a drug metabolizing enzyme that has been associated with cancer cell proliferation in vitro and with survival in vivo. NAT1 expression has been associated with the estrogen receptor and it has been proposed as a prognostic marker for estrogen receptor positive cancers. However, little is known about the distribution of NAT1 mRNA across an entire patient population or its effects on outcomes. To address this, gene expression data from breast cancer patient cohorts were investigated to identify sub-populations based on the level of NAT1 expression. Patient survival and drug response was examined to determine whether NAT1 mRNA levels influenced any of these parameters. RESULTS NAT1 expression showed a trimodal distribution in breast cancer samples (n = 1980) but not in tumor tissue from ovarian, prostate, cervical or colorectal cancers. In breast cancer, NAT1 mRNA in each sub-population correlated with a separate set of genes suggesting different mechanisms of NAT1 gene regulation. Kaplan-Meier plots showed significantly better survival in patients with highest NAT1 mRNA compared to those with intermediate or low expression. While NAT1 expression was elevated in estrogen receptor-positive patients, it did not appear to be dependent on estrogen receptor expression. Overall survival was analyzed in patients receiving no treatment, hormone therapy or chemotherapy. NAT1 expression correlated strongly with survival in the first 5 years in those patients receiving chemotherapy but did not influence survival in the other two groups. This suggests that low NAT1 expression is associated with chemo-resistance. The sensitivity of NAT1 mRNA levels as a single parameter to identify non-responders to chemotherapy was 0.58 at a log(2) < 6.5. CONCLUSIONS NAT1 mRNA can be used to segregate breast cancer patients into sub-populations that demonstrate different overall survival. Moreover, low NAT1 expression shows a distinct poor response to chemotherapy. Analysis of NAT1 expression may be useful for identifying specific individuals who would benefit from alternative therapy or drug combinations. However, additional information is required to increase the sensitivity of identifying non-responders.
Collapse
Affiliation(s)
- Rodney F. Minchin
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, QLD Australia
| | - Neville J. Butcher
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, QLD Australia
| |
Collapse
|
37
|
TFF1 and TFF3 mRNAs Are Higher in Blood from Breast Cancer Patients with Metastatic Disease than Those without. JOURNAL OF ONCOLOGY 2018; 2018:4793498. [PMID: 29977293 PMCID: PMC6011051 DOI: 10.1155/2018/4793498] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 03/03/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022]
Abstract
Introduction Breast cancer metastasis occurs when tumor cells dissociate from the primary tumor and migrate to distant organs through the peripheral bloodstream or lymphatic drainage. Circulating tumor cells (CTCs) originate from primary sites or metastases and circulate in the patients' bloodstream. Molecular assays for the detection and molecular characterization of CTCs can serve as a liquid biopsy and can represent an alternative to invasive biopsies as a source of tumor tissue in the metastatic patients. Patients and Methods We analyzed the presence of CTCs in the peripheral blood of 50 breast cancer patients by quantitative real-time reverse transcriptase polymerase chain reaction (RT-qPCR) to detect trefoil factor family (TFF) 1 and 3 genes. Results We found significant difference in the level of both TFF1 and TFF3 mRNA in the blood of nonmetastatic versus metastatic breast cancer patients (p= 0.001 and p= 0.038, respectively). TFF1 mRNA was detected at higher levels in 34.6% of metastatic breast cancer patients as compared to 0% of nonmetastatic (p= 0.002). As regards TFF3 mRNA, it was detected at higher levels in 46.2% of metastatic breast cancer patients as compared to 4% of nonmetastatic (p= 0.026). Moreover, we found that the high level of both TFF1 and TFF3 mRNA was related to estrogen status of the patients. The detection of high level of TFF1 mRNA in CTCs was associated with bone metastases (77.8%), while that of TFF3 was related to lymph node involvement (75%) and lung metastases (68.8%). Conclusion The combined measurement of both TFF1 and TFF3 mRNA level for differentiation of metastatic from nonmetastatic breast cancer gave 57.69% sensitivity and 83.3% specificity.
Collapse
|
38
|
Tolušić Levak M, Mihalj M, Koprivčić I, Lovrić I, Novak S, Bijelić N, Baus-Lončar M, Belovari T, Kralik K, Pauzar B. Differential Expression of TFF Genes and Proteins in Breast Tumors. Acta Clin Croat 2018; 57:264-277. [PMID: 30431719 PMCID: PMC6532012 DOI: 10.20471/acc.2018.57.02.06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
SUMMARY – The objective of this study was to determine differential expression of TFF1, TFF2 and TFF3 genes and proteins in breast tumor subtypes. In addition, we investigated the correlation between TFF genes within tumor subgroups, and TFF genes with clinical and pathologic characteristics of the tumor. Study group included 122 patients with surgically removed breast tumors. Samples were investigated using qRT-PCR and immunohistochemistry. TFF1 and TFF3 genes and proteins were expressed in breast tumors, while the levels of TFF2 gene and protein expression were very low or undetectable. TFF1 was significantly more expressed in benign tumors, while TFF3 was more expressed in malignant tumors. Gene and protein expression of both TFF1 and TFF3 was greater in lymph node-negative tumors, hormone positive tumors, tumors with moderate levels of Ki67 expression, and in grade II tumors. A strong positive correlation was found between TFF1 and TFF3 genes, and the expression of both negatively correlated with Ki67 and the level of tumor histologic differentiation. Our results suggest that TFF1 and TFF3, but not TFF2, may have a role in breast tumor pathogenesis and could be used in the assessment of tumor differentiation and malignancy.
Collapse
Affiliation(s)
| | - Martina Mihalj
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Ivan Koprivčić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Surgery, Osijek University Hospital Centre, Osijek, Croatia
| | - Ivana Lovrić
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Anatomy, Histology and Embryology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Sanja Novak
- Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Nikola Bijelić
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Mirela Baus-Lončar
- Department of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Tatjana Belovari
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Kristina Kralik
- Department of Medical Statistics and Informatics, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Biljana Pauzar
- Department of Histology and Embryology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.,Department of Clinical Cytology, Osijek University Hospital Centre, Osijek, Croatia
| |
Collapse
|
39
|
Characterizing Molecular Variants and Clinical Utilization of Next-generation Sequencing in Advanced Breast Cancer. Appl Immunohistochem Mol Morphol 2018; 25:392-398. [PMID: 26862949 DOI: 10.1097/pai.0000000000000322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Women with advanced breast carcinomas have few therapeutic options. Recent advances in genomic profiling represent a new paradigm of cancer classification and treatment, but experience with genomic testing in a clinical setting remains limited. We retrospectively determined the genomic variants and correlate these with histology [histomorphologic subtype, nuclear grade, standard immunohistochemistry (IHC)] and clinical utilization (ordering, turnaround time, report review, and targeted therapy). Among 48 patients, 2 showed no genetic alterations, 11 (23%) showed variants of unclear significance only and 35 (73%) showed variant(s) affecting function (VaF) and/or variants of unclear significance. Overall, 119 variants were observed in 20 of 50 tested genes. Each patient had a unique molecular profile, with numerous (n=58) variants not previously reported in breast cancer. VaF detected in more than 2 patients included: TP53 (n=21), PIK3CA (n=20), and FGFR1 (n=3). VaF comprised 46 single nucleotide variants (79%), 7 amplifications (12%), 3 frameshifts (5%), 1 insertion (2%), and 1 deletion (2%). The tested samples had very high Ki67 index (average 57%±23%) and approximately half were hormone receptor and HER2 negative (25/46, 54%). Metastatic breast carcinomas showed a higher average VaF versus breast-localized tumors (1.3±0.99 vs. 0.18±0.60, P<0.05). Next-generation sequencing reports were promptly reported and reviewed (average 1 to 2 d) and 7 (∼25%) of potentially eligible patients received targeted therapy. Advanced breast cancers show unique landscapes of genetic variants. Most testing was done in late disease, often in metastatic and receptor-negative carcinomas. Next-generation sequencing results were promptly reported and reviewed, but the utilization of targeted therapies was limited.
Collapse
|
40
|
Stepp MW, Doll MA, Carlisle SM, States JC, Hein DW. Genetic and small molecule inhibition of arylamine N-acetyltransferase 1 reduces anchorage-independent growth in human breast cancer cell line MDA-MB-231. Mol Carcinog 2018; 57:549-558. [PMID: 29315819 PMCID: PMC5832614 DOI: 10.1002/mc.22779] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 01/07/2023]
Abstract
Arylamine N-acetyltransferase 1 (NAT1) expression is reported to affect proliferation, invasiveness, and growth of cancer cells. MDA-MB-231 breast cancer cells were engineered such that NAT1 expression was elevated or suppressed, or treated with a small molecule inhibitor of NAT1. The MDA-MB-231 human breast cancer cell lines were engineered with a scrambled shRNA, a NAT1 specific shRNA or a NAT1 overexpression cassette stably integrated into a single flippase recognition target (FRT) site facilitating incorporation of these different genetic elements into the same genomic location. NAT1-specific shRNA reduced NAT1 activity in vitro by 39%, increased endogenous acetyl coenzyme A levels by 35%, and reduced anchorage-independent growth (sevenfold) without significant effects on cell morphology, growth rates, anchorage-dependent colony formation, or invasiveness compared to the scrambled shRNA cell line. Despite 12-fold overexpression of NAT1 activity in the NAT1 overexpression cassette transfected MDA-MB-231 cell line, doubling time, anchorage-dependent cell growth, anchorage-independent cell growth, and relative invasiveness were not changed significantly when compared to the scrambled shRNA cell line. A small molecule (5E)-[5-(4-hydroxy-3,5-diiodobenzylidene)-2-thioxo-1,3-thiazolidin-4-one (5-HDST) was 25-fold more selective towards the inhibition of recombinant human NAT1 than N-acetyltransferase 2. Incubation of MDA-MB-231 cell line with 5-HDST resulted in 60% reduction in NAT1 activity and significant decreases in cell growth, anchorage-dependent growth, and anchorage-independent growth. In summary, inhibition of NAT1 activity by either shRNA or 5-HDST reduced anchorage-independent growth in the MDA-MB-231 human breast cancer cell line. These findings suggest that human NAT1 could serve as a target for the prevention and/or treatment of breast cancer.
Collapse
Affiliation(s)
- Marcus W Stepp
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Mark A Doll
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Samantha M Carlisle
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - J Christopher States
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - David W Hein
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
41
|
T lymphocytes facilitate brain metastasis of breast cancer by inducing Guanylate-Binding Protein 1 expression. Acta Neuropathol 2018; 135:581-599. [PMID: 29350274 PMCID: PMC5978929 DOI: 10.1007/s00401-018-1806-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/09/2018] [Accepted: 01/09/2018] [Indexed: 02/01/2023]
Abstract
The discovery of genes and molecular pathways involved in the formation of brain metastasis would direct the development of therapeutic strategies to prevent this deadly complication of cancer. By comparing gene expression profiles of Estrogen Receptor negative (ER-) primary breast tumors between patients who developed metastasis to brain and to organs other than brain, we found that T lymphocytes promote the formation of brain metastases. To functionally test the ability of T cells to promote brain metastasis, we used an in vitro blood–brain barrier (BBB) model. By co-culturing T lymphocytes with breast cancer cells, we confirmed that T cells increase the ability of breast cancer cells to cross the BBB. Proteomics analysis of the tumor cells revealed Guanylate-Binding Protein 1 (GBP1) as a key T lymphocyte-induced protein that enables breast cancer cells to cross the BBB. The GBP1 gene appeared to be up-regulated in breast cancer of patients who developed brain metastasis. Silencing of GBP1 reduced the ability of breast cancer cells to cross the in vitro BBB model. In addition, the findings were confirmed in vivo in an immunocompetent syngeneic mouse model. Co-culturing of ErbB2 tumor cells with activated T cells induced a significant increase in Gbp1 expression by the cancer cells. Intracardial inoculation of the co-cultured tumor cells resulted in preferential seeding to brain. Moreover, intracerebral outgrowth of the tumor cells was demonstrated. The findings point to a role of T cells in the formation of brain metastases in ER- breast cancers, and provide potential targets for intervention to prevent the development of cerebral metastases.
Collapse
|
42
|
Guttery DS, Blighe K, Polymeros K, Symonds RP, Macip S, Moss EL. Racial differences in endometrial cancer molecular portraits in The Cancer Genome Atlas. Oncotarget 2018; 9:17093-17103. [PMID: 29682207 PMCID: PMC5908308 DOI: 10.18632/oncotarget.24907] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/10/2018] [Indexed: 12/11/2022] Open
Abstract
Endometrial cancer (EC) is now the most prevalent gynaecological malignancy in the Western world. Black or African American women (BoAA) have double the mortality of Caucasian women, and their tumours tend to be of higher grade. Despite these disparities, little is known regarding the mutational landscape of EC between races. Hence, we wished to investigate the molecular features of ECs within The Cancer Genome Atlas (TCGA) dataset by racial groupings. In total 374 Caucasian, 109 BoAA and 20 Asian patients were included in the analysis. Asian women were diagnosed at younger age, 54.2 years versus 64.5 years for Caucasian and 64.9 years for BoAA women (OR 3.432; p=0.011); BoAA women were more likely to have serous type tumors (OR 2.061; p=0.008). No difference in overall survival was evident. The most frequently mutated gene in Caucasian and Asian tumours was PTEN (63% and 85%), unlike BoAA cases where it was TP53 (49%). Mutation and somatic copy number alteration (SCNA) analysis revealed an enrichment of TP53 mutations in BoAAs; whereas POLE and RPL22 mutations were more frequent in Caucasians. Major recurrent SCNA racial differences were observed at chromosomes 3p, 8, 10, and 16, which clustered BoAA tumors into 4 distinct groups and Caucasian tumors into 5 groups. There was a significantly higher frequency of somatic mutations in DNA mismatch repair genes in Asian tumours, in particular PMS2 (p=0.0036). In conclusion, inherent racial disparities appear to be present in the molecular profile of EC, which could have potential implications on clinical management.
Collapse
Affiliation(s)
- David S Guttery
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester, UK
| | - Kevin Blighe
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester, UK
| | - Konstantinos Polymeros
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester, UK.,Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, UK.,Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - R Paul Symonds
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester, UK
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Esther L Moss
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester, UK.,Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester, UK
| |
Collapse
|
43
|
Cosphiadi I, Atmakusumah TD, Siregar NC, Muthalib A, Harahap A, Mansyur M. Bone Metastasis in Advanced Breast Cancer: Analysis of Gene Expression Microarray. Clin Breast Cancer 2018; 18:e1117-e1122. [PMID: 29609951 DOI: 10.1016/j.clbc.2018.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/26/2018] [Accepted: 03/05/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Approximately 30% to 40% of breast cancer recurrences involve bone metastasis (BM). Certain genes have been linked to BM; however, none have been able to predict bone involvement. In this study, we analyzed gene expression profiles in advanced breast cancer patients to elucidate genes that can be used to predict BM. PATIENTS AND METHODS A total of 92 advanced breast cancer patients, including 46 patients with BM and 46 patients without BM, were identified for this study. Immunohistochemistry and gene expression analysis was performed on 81 formalin-fixed paraffin-embedded samples. Data were collected through medical records, and gene expression of 200 selected genes compiled from 6 previous studies was performed using NanoString nCounter. RESULTS Genetic expression profiles showed that 22 genes were significantly differentially expressed between breast cancer patients with metastasis in bone and other organs (BM+) and non-BM, whereas subjects with only BM showed 17 significantly differentially expressed genes. The following genes were associated with an increasing incidence of BM in the BM+ group: estrogen receptor 1 (ESR1), GATA binding protein 3 (GATA3), and melanophilin with an area under the curve (AUC) of 0.804. In the BM group, the following genes were associated with an increasing incidence of BM: ESR1, progesterone receptor, B-cell lymphoma 2, Rab escort protein, N-acetyltransferase 1, GATA3, annexin A9, and chromosome 9 open reading frame 116. ESR1 and GATA3 showed an increased strength of association with an AUC of 0.928. CONCLUSION A combination of the identified 3 genes in BM+ and 8 genes in BM showed better prediction than did each individual gene, and this combination can be used as a training set.
Collapse
Affiliation(s)
- Irawan Cosphiadi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.
| | - Tubagus D Atmakusumah
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Nurjati C Siregar
- Department of Pathology Anatomy, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Abdul Muthalib
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Alida Harahap
- Department of Clinical Pathology, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Muchtarruddin Mansyur
- Department of Public Health, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
44
|
Abreu A, Milani C, Katayama M, Barbosa E, da Fonseca LG, Góes J, Brentani M, Folgueira MK. Expression of Heterochromatin Protein 1 in the Primary Tumor of Breast Cancer Patients in the Presence or Absence of Occult Metastatic Cells in the Bone Marrow. Int J Biol Markers 2018; 23:219-24. [DOI: 10.1177/172460080802300404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Gene silencing may occur in breast cancer samples from patients presenting with occult metastatic cells in the bone marrow and one mechanism regulating gene suppression is heterochromatin formation. We have studied whether members of the heterochromatin protein 1 family (HP1Hsα, HP1Hsβ and HP1Hsγ), which take part in chromatin packaging and gene expression regulation, were differentially expressed in tumors from patients with and without occult metastatic cells in their bone marrow. Tumor samples and bone marrow aspirates were obtained from 37 breast cancer patients. Median age was 63 years and 68% of the patients presented with clinical stage I/II disease. Presence of occult metastatic cells in bone marrow was detected through keratin-19 expression by nested RT-PCR in samples from 20 patients (54.1%). The presence of occult metastatic cells in bone marrow was not associated with node involvement, histological grade, estrogen receptor and ERBB2 immunoexpression. Relative gene expression of HP1Hsα, HP1Hsβ and HP1Hsγ was determined by real-time RT-PCR and did not vary according to the presence of occult metastatic cells in bone marrow. In addition, the combined expression of these three transcripts could not be used to classify samples according to the presence of bone marrow micrometastasis. Our work indicates that regulation of heterochromatin formation through HP1 family members may not be the sole mechanism implicated in the metastatic process to the bone marrow.
Collapse
Affiliation(s)
- A.P.S. Abreu
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
- Instituto Brasileiro de Controle do Câncer de São Paulo, São Paulo - Brasil
| | - C. Milani
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
| | - M.L.H. Katayama
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
| | - E.M. Barbosa
- Instituto Brasileiro de Controle do Câncer de São Paulo, São Paulo - Brasil
| | - L. Gomes da Fonseca
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
| | - J.CS. Góes
- Instituto Brasileiro de Controle do Câncer de São Paulo, São Paulo - Brasil
| | - M.M. Brentani
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
| | - M.A.A. Koike Folgueira
- Faculdade de Medicina da Universidade de São Paulo, Departamento de Radiologia, Disciplina de Oncologia, São Paulo
- Instituto Brasileiro de Controle do Câncer de São Paulo, São Paulo - Brasil
| |
Collapse
|
45
|
Mejri N, Benna M, El Benna H, Soumaya L, Afrit M, Zouari B, Boussen H. First site of recurrence after breast cancer adjuvant treatment in the era of multimodality therapy: which imaging for which patient during follow-up? Breast Dis 2017; 37:123-132. [PMID: 29171964 DOI: 10.3233/bd-170290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE We evaluated the relation between first site of recurrence of early breast cancer and disease profile at presentation and reported survival results, suggesting a personalized diagnostic imaging guidance during follow up. METHODS Among 1400 early breast cancer treated from 2000 to 2010, 324 relapses were divided into 4 groups according to first site: A-locoregional, B-bone, C-Brain and D-visceral. We analyzed redictive factors of each group compared to a control group of 100 non relapsing patients and the remaining groups matched. RESULTS In group A, patients were more likely to have histological tumor size above >2 cm, grade 1-2, HR positive and 0-3 involved lymph nodes. In group B, patients had more commonly grade 2-3, 1-3 positive lymph nodes and HR positive tumors. In group C, patients were more frequently young, with large tumor size, grade3, positive lymph nodes and HER2 positive tumors. In group D, patients were more likely to have tumors>2 cm in size, with nodal involvement, grade 3, HR negative and HER2 positive tumors. Annual recurrence rate in group A, was stable ranging between 15%-18%, within the first 3 years and peaked at 19.4% in the interval [1-2]year in group B. Median survival was 46 months in group A, 43 months in group B, with no significant difference. CONCLUSION Outcome of loco-regional and bone relapses was good, suggesting that both systematic mammography and bone-scan/CT scan for high risk patients (N+, gradeIII) during the first 2-3 years may represent a tailored relevant follow-up protocol for breast cancer patients.
Collapse
Affiliation(s)
- Nesrine Mejri
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia
| | - Mehdi Benna
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia
| | - Houda El Benna
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia
| | - Labidi Soumaya
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia
| | - Mehdi Afrit
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia
| | - Bechir Zouari
- University Tunis EI Manar Tunis, Faculty of Medicine, Department of Epidemiology and Statistics, Tunis, Tunisia
| | - Hamouda Boussen
- University Tunis EI Manar Tunis, Faculty of Medicine, Medical Oncology Department, AbderrahmenMami Hospital, Ariana, Tunisia.,"Clinique Taoufik" hospital, Tunis, Tunisia
| |
Collapse
|
46
|
Coleman R, Hall A, Albanell J, Hanby A, Bell R, Cameron D, Dodwell D, Marshall H, Jean-Mairet J, Tercero JC, Rojo F, Gregory W, Gomis RR. Effect of MAF amplification on treatment outcomes with adjuvant zoledronic acid in early breast cancer: a secondary analysis of the international, open-label, randomised, controlled, phase 3 AZURE (BIG 01/04) trial. Lancet Oncol 2017; 18:1543-1552. [PMID: 29037984 DOI: 10.1016/s1470-2045(17)30603-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Adjuvant use of bisphosphonates can reduce the incidence of bone metastases in early breast cancer. Recurrence and survival seem to be improved only in postmenopausal patients, but the underlying mechanisms remain unclear. We investigated whether MAF amplification (a biomarker for bone metastasis) in primary tumours could predict the treatment outcomes of adjuvant zoledronic acid. METHODS The study population included patients enrolled in the international, open-label, randomised, controlled, phase 3 AZURE trial at eligible UK sites who had stage II or III breast cancer and who gave consent for use of their primary tumour samples. Patients were randomly assigned (1:1) to receive standard adjuvant systemic therapy alone (control group) or with zoledronic acid every 3-4 weeks for six doses, then every 3-6 months until the end of 5 years. Minimisation took into account the number of involved axillary lymph nodes, clinical tumour stage, oestrogen-receptor status, type and timing of systemic therapy, menopausal status, statin use, and treating centre. The primary endpoint was disease-free survival; the secondary endpoint, invasive-disease-free survival, was the primary disease endpoint for the analysis in this report. MAF amplification was assessed by fluorescence in-situ hybridisation of two cores of breast tumour tissue in a microarray, done in a central laboratory by technicians unaware of treatment assignment. We used multivariate analyses to assess disease outcomes by intention to treat. We also assessed interactions between MAF-positive status and menopausal status on efficacy of zoledronic acid. The AZURE trial is registered with the International Standard Randomised Controlled Trial Registry, number ISRCTN79831382. FINDINGS 1739 AZURE patients contributed primary tumour samples, of whom 865 (50%) had two assessable cores (445 in the control groups and 420 in the zoledronic acid group). 184 (21%) tumours were MAF positive (85 in the control groups and 99 in the zoledronic acid group) and the remaining tumours were MAF negative. At a median follow-up of 84·6 months (IQR 72·0-95·8), MAF status was not prognostic for invasive-disease-free survival in the control group (MAF-positive vs MAF-negative: hazard ratio [HR] 0·92, 95% CI 0·59-1·41), but was in the zoledronic acid group (0·52, 0·36-0·75). In patients with MAF-negative tumours, zoledronic acid was associated with higher invasive-disease-free survival than was control treatment (HR 0·74, 95% CI 0·56-0·98), but not in patients who had MAF-positive tumours. Additionally, among 121 patients not postmenopausal at randomisation with MAF-positive tumours, zoledronic acid was associated with lower invasive-disease-free survival (HR 2·47, 95% CI 1·23-4·97) and overall survival (2·27, 95% CI 1·04-4·93) than control treatment. INTERPRETATION MAF status can predict likelihood of benefit from adjuvant zoledronic acid and merits further investigation as a potential companion diagnostic. FUNDING Novartis Global and Inbiomotion.
Collapse
Affiliation(s)
- Robert Coleman
- Academic Unit of Clinical Oncology, Weston Park Hospital, Sheffield, UK.
| | - Andrew Hall
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | - Joan Albanell
- IMIM-CIBERONC, Barcelona; Medical Oncology Department, Hospital del Mar, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain
| | - Andrew Hanby
- Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | - David Cameron
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - David Dodwell
- St James Institute of Oncology, St James Hospital, Leeds, UK
| | - Helen Marshall
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | | | | | - Federico Rojo
- Fundación Jiménez Díaz, Madrid, Spain; CIBERONC, Madrid, Spain
| | - Walter Gregory
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | - Roger R Gomis
- IMIM-CIBERONC, Barcelona; Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
47
|
Abstract
Distant metastasis during the advanced stage of malignant tumor progression can cause considerable morbidity in cancer patients. Bone is known to be one of the most common sites of distant metastasis in patients with breast cancer (BC). BC metastases in bone are associated with excessive skeletal complications. These complications can be fatal and reduce quality of life of patients. It is important to understand the metastatic process of BC to bone to improve quality of life and design new therapeutic methods. At present, the molecular mechanisms leading to the BC metastasis to bone are not fully understood. Studying the molecular basis of BC metastasis to bone might improve our insight into this complex process. In addition, it can provide novel approaches for designing advanced and effective targeted therapies. The present article aimed to review the published papers on the molecular basis of the metastatic process of BC to bone, focusing on involved genes and signaling networks. Furthermore, we propose potential therapeutic targets that may be more effective for the inhibition and treatment of BC metastasis to bone.
Collapse
|
48
|
Xu Y, Yuan Q, Zhou J, Chang X, Wang K, Han J. Association of TOX3 polymorphisms with breast cancer: A meta-analysis. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
49
|
Mercatali L, La Manna F, Miserocchi G, Liverani C, De Vita A, Spadazzi C, Bongiovanni A, Recine F, Amadori D, Ghetti M, Ibrahim T. Tumor-Stroma Crosstalk in Bone Tissue: The Osteoclastogenic Potential of a Breast Cancer Cell Line in a Co-Culture System and the Role of EGFR Inhibition. Int J Mol Sci 2017; 18:ijms18081655. [PMID: 28758931 PMCID: PMC5578045 DOI: 10.3390/ijms18081655] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022] Open
Abstract
Although bone metastases represent a major challenge in the natural history of breast cancer (BC), the complex interactions involved have hindered the development of robust in vitro models. The aim of this work is the development of a preclinical model of cancer and bone stromal cells to mimic the bone microenvironment. We studied the effects on osteoclastogenesis of BC cells and Mesenchymal stem cells (MSC) cultured alone or in combination. We also analyzed: (a) whether the blockade of the Epithelial Growth Factor Receptor (EGFR) pathway modified their influence on monocytes towards differentiation, and (b) the efficacy of bone-targeted therapy on osteoclasts. We evaluated the osteoclastogenesis modulation of human peripheral blood monocytes (PBMC) indirectly induced by the conditioned medium (CM) of the human BC cell line SCP2, cultured singly or with MSC. Osteoclastogenesis was evaluated by TRAP analysis. The effect of the EGFR blockade was assessed by treating the cells with gefitinib, and analyzed with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Western Blot (WB). We observed that SCP2 co-cultured with MSC increased the differentiation of PBMC. This effect was underpinned upon pre-treatment of the co-culture with gefitinib. Co-culture of SCP2 with MSC increased the expression of both the bone-related marker Receptor Activator of Nuclear Factor κB (RANK) and EGFR in BC cells. These upregulations were not affected by the EGFR blockade. The effects of the CM obtained by the cells treated with gefitinib in combination with the treatment of the preosteoclasts with the bone-targeted agents and everolimus enhanced the inhibition of the osteoclastogenesis. Finally, we developed a fully human co-culture system of BC cells and bone progenitor cells. We observed that the interaction of MSC with cancer cells induced in the latter molecular changes and a higher power of inducing osteoclastogenesis. We found that blocking EGFR signaling could be an efficacious strategy for breaking the interactions between cancer and bone cells in order to inhibit bone metastasis.
Collapse
Affiliation(s)
- Laura Mercatali
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Federico La Manna
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Giacomo Miserocchi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Chiara Liverani
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Chiara Spadazzi
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Federica Recine
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Dino Amadori
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| | - Martina Ghetti
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
- Biomedical and Neuromotor Sciences Department, University of Bologna, 40123 Bologna, Italy.
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
| |
Collapse
|
50
|
Guo J, Gong G, Zhang B. Identification and prognostic value of anterior gradient protein 2 expression in breast cancer based on tissue microarray. Tumour Biol 2017; 39:1010428317713392. [PMID: 28671019 DOI: 10.1177/1010428317713392] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Breast cancer has attracted substantial attention as one of the major cancers causing death in women. It is crucial to find potential biomarkers of prognostic value in breast cancer. In this study, the expression pattern of anterior gradient protein 2 in breast cancer was identified based on the main molecular subgroups. Through analysis of 69 samples from the Gene Expression Omnibus database, we found that anterior gradient protein 2 expression was significantly higher in non-triple-negative breast cancer tissues compared with normal tissues and triple-negative breast cancer tissues (p < 0.05). The data from a total of 622 patients from The Cancer Genome Atlas were analysed. The data from The Cancer Genome Atlas and results from quantitative reverse transcription polymerase chain reaction also verified the anterior gradient protein 2 expression pattern. Furthermore, we performed immunohistochemical analysis. The quantification results revealed that anterior gradient protein 2 is highly expressed in non-triple-negative breast cancer (grade 3 excluded) and grade 1 + 2 (triple-negative breast cancer excluded) tumours compared with normal tissues. Anterior gradient protein 2 was significantly highly expressed in non-triple-negative breast cancer (grade 3 excluded) and non-triple-negative breast cancer tissues compared with triple-negative breast cancer tissues (p < 0.01). In addition, anterior gradient protein 2 was significantly highly expressed in grade 1 + 2 (triple-negative breast cancer excluded) and grade 1 + 2 tissues compared with grade 3 tissues (p < 0.05). Analysis by Fisher's exact test revealed that anterior gradient protein 2 expression was significantly associated with histologic type, histological grade, oestrogen status and progesterone status. Univariate analysis of clinicopathological variables showed that anterior gradient protein 2 expression, tumour size and lymph node status were significantly correlated with overall survival in patients with grade 1 and 2 tumours. Cox multivariate analysis revealed anterior gradient protein 2 as a putative independent indicator of unfavourable outcomes (p = 0.031). All these data clearly showed that anterior gradient protein 2 is highly expressed in breast cancer and can be regarded as a putative biomarker for breast cancer prognosis.
Collapse
Affiliation(s)
- Jilong Guo
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China
| | - Guohua Gong
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China.,3 Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinal treatment, Tongliao, Inner Mongolia, China
| | - Bin Zhang
- 1 Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China.,2 Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, China.,3 Affiliated Hospital of Inner Mongolia University for Nationalities, Institute of Mongolia and Western Medicinal treatment, Tongliao, Inner Mongolia, China
| |
Collapse
|