1
|
Xie L, Wang Y, Wan A, Huang L, Wang Q, Tang W, Qi X, Hu X. Research trends of neoadjuvant therapy for breast cancer: A bibliometric analysis. Hum Vaccin Immunother 2025; 21:2460272. [PMID: 39904891 PMCID: PMC11801352 DOI: 10.1080/21645515.2025.2460272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
The approach of neoadjuvant therapy for breast cancer, which involves administering systemic treatment prior to primary surgery, has undergone substantial advancements in recent decades. This strategy is intended to reduce tumor size, thereby enabling less invasive surgical procedures and enhancing patient outcomes. This study presents a comprehensive bibliometric analysis of research trends in neoadjuvant therapy for breast cancer from 2009 to 2024. Using data extracted from the Web of Science Core Collection, a total of 3,674 articles were analyzed to map the research landscape in this field. The analysis reveals a steady increase in publication output, peaking in 2022, with the United States and China identified as the leading contributors. Key institutions, such as the University of Texas System and MD Anderson Cancer Center, have been instrumental in advancing the research on neoadjuvant therapy. The study also highlights the contributions of influential authors like Sibylle Loibl and Gunter von Minckwitz, as well as major journals such as the Journal of Clinical Oncology. Emerging research topics, including immunotherapy, liquid biopsy, and artificial intelligence, are gaining prominence and represent potential future directions for clinical applications. This bibliometric analysis provides critical insights into global research trends, key contributors, and future developments in the field of neoadjuvant therapy for breast cancer, offering a foundation for future research and clinical practice advancements.
Collapse
Affiliation(s)
- Laiping Xie
- Department of Nuclear Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuhang Wang
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Andi Wan
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis, Chongqing, China
| | - Lin Huang
- Department of Radiology, People’s Hospital of Xingyi, Guizhou, China
| | - Qing Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wanyan Tang
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
- Key Laboratory of Chongqing Health Commission for Minimally Invasive and Precise Diagnosis, Chongqing, China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Hu W, Ding X, Wu X, Xi X, Xu J, Dai S, Chen J, Hu S, Zhao Q, Chen F. A Comprehensive Analysis of Epoxide Hydrolase 2 (EPHX2) in Pan-Cancer. Cancer Rep (Hoboken) 2025; 8:e70188. [PMID: 40129060 PMCID: PMC11932960 DOI: 10.1002/cnr2.70188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND AND AIMS Epoxide hydrolase 2 (EPHX2) regulates lipid signaling across various metabolites by encoding soluble epoxide hydrolase. However, its mechanisms and implications in human malignancies remain unknown. This research aimed to detail the prognostic landscape of EPHX2 in pan-cancer and explore its potential relationship with immune infiltration in the tumor microenvironment. METHODS Herein, multiple bioinformatics tools were used to comprehensively evaluate the expression, diagnostic, and prognostic significance of EPHX2 and its roles in the tumor immune microenvironment in human cancers. The underlying EPHX2-associated signaling pathways in cancers were investigated by gene set variation analysis (GSVA). TIDE, GDSC, and CTRP databases were applied to predict the response of EPHX2 to immunotherapy and sensitivity to small molecule drugs. Furthermore, EPHX2 expression was also validated by qPCR experiments in various cancer cell lines. RESULTS Overall results revealed significant down-regulation of EPHX2 mRNA expression in most tumors. Despite its high predictive significance across cancers, EPHX2 played a protective or detrimental effect in distinct types of cancers. EPHX2 proved to be a valuable diagnostic biomarker in a range of tumor types, particularly in kidney renal clear cell carcinoma, cervical squamous cell carcinoma, and endocervical adenocarcinoma. Genetic alterations of EPHX2 in 33 tumors were also investigated. EPHX2 expression was significantly linked to immune cell infiltrations (particularly tumor-associated macrophages), tumor mutation burden, microsatellite instability, immune modulators, and immunotherapeutic biomarkers. Single-cell sequencing and GSVA highlighted the relevance of EPHX2 in regulating various cancer-related biological processes, including cell cycle and apoptosis. In this view, targeting EPHX2-dependent signaling could be a promising therapeutic strategy for tumor immunotherapy. CONCLUSION EPHX2 may serve as a potential molecular biomarker for diagnosis and prognosis in pan-cancer and could become a novel therapeutic target for various cancers.
Collapse
Affiliation(s)
- Weiquan Hu
- Department of Joint SurgeryGanzhou People's HospitalGanzhouJiangxiChina
| | - Xiaoli Ding
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Xiangsheng Wu
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Xuxiang Xi
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Jing Xu
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Shengyun Dai
- National Institutes for Food and Drug ControlBeijingChina
| | - Jing Chen
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Suping Hu
- Department of EmergencyFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Qinfei Zhao
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Fangfang Chen
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouGuangdongChina
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingJiangsuChina
| |
Collapse
|
3
|
Witkiewicz AK, Wang J, Schultz E, O'Connor TN, O'Connor T, Levine E, Knudsen ES. Using prognostic signatures and machine learning to identify core features associated with response to CDK4/6 inhibitor-based therapy in metastatic breast cancer. Oncogene 2025:10.1038/s41388-025-03308-0. [PMID: 40011574 DOI: 10.1038/s41388-025-03308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
CDK4/6 inhibitors in combination with endocrine therapy are widely used to treat HR+/HER2- metastatic breast cancer leading to improved progression-free survival (PFS) compared to single agent endocrine therapy. Over 300 patients receiving standard-of-care CDK4/6 inhibitor combination therapy for metastatic disease were enrolled at a single institution. Clinical, pathological, and gene expression data were employed to define determinants for PFS duration. Visceral disease (HR 1.55, p = 0.0013), prior endocrine therapy (HR 2.34, p < 0.001), and the type of endocrine therapy (HR 2.16, p < 0.001) were highly associated with PFS duration. Multiple pre-defined gene expression signatures were employed to determine association with response to CDK4/6 inhibitor-based therapy. Random survival forest was applied to define key gene expression and clinical features associated with PFS and develop a predictive model. The time to progression predicted by this model was related to the median PFS observed in PALOMA-2/3 and PEARL studies. Interrogating genes identified as highly significant across all studies indicated common enrichment of gene networks associated with cell cycle and estrogen receptor signaling. These findings indicate that there are common features from real-world use of CDK4/6 inhibitors that could be used to infer time to progression and better inform treatment.
Collapse
Affiliation(s)
- Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Emily Schultz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Thomas N O'Connor
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Tracey O'Connor
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Ellis Levine
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| |
Collapse
|
4
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
5
|
Deldadeh N, Shahbazi S, Ghiasvand S, Shahriari F, Javidi MA. COVID-19 vaccination anti-cancer impact on the PI3K/AKT signaling pathway in MC4L2 mice models. Microb Pathog 2024; 196:106955. [PMID: 39303961 DOI: 10.1016/j.micpath.2024.106955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
The most promising method of containing the COVID-19 pandemic is considered to be vaccination against SARS-CoV-2 infection. However, research on the relationship between vaccination against COVID-19 and cancer has primarily examined induced immunity rather than the disease itself. Considering that breast cancer is the most common cancer among women, the main goal of this study was to examine the impact of the Sinopharm and AstraZeneca vaccination on tumor characteristics such as tumor size, important tumor markers, tumor-infiltrating lymphocytes, metastasis to vital organs, and investigation of the PI3K/AKT signaling pathway, and the expression levels of relevant genes (PTEN, mTOR, AKT, PI3K, GSK3, and FoxO1) of the luminal B (MC4L2) mouse model. The tumor size of the mice was measured and monitored every two days, and after thirty days, the mice were euthanized. Remarkably, after vaccination, all vaccinated mice showed a decrease in the size of their tumor and an increase in the number of lymphocytes that had invaded the tumors. Tumor marker levels (VEGF, Ki-67, MMP-2/9), CD4/CD8 ratio, metastasis to vital organs, hormone receptors (ER, PR, and HER-2), and expression of genes related to the advancement of the PI3K/AKT signaling pathway were lower in vaccinated mice. Our research showed that the COVID-19 vaccine can have an anti-cancer effect by slowing the tumor progression and metastasis.
Collapse
Affiliation(s)
- Negar Deldadeh
- Department of Integrative Oncology, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Sahba Shahbazi
- Protein Biotechnology Research Lab (PBRL), Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Saeedeh Ghiasvand
- Department of Biology, Faculty of Science, Malayer University, Malayer, Iran.
| | - Fatemeh Shahriari
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Amin Javidi
- Department of Integrative Oncology, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
6
|
Wu W, Wu MY, Dai T, Ke LN, Shi Y, Hu J, Wang Q. Terphenyllin induces CASP3-dependent apoptosis and pyroptosis in A375 cells through upregulation of p53. Cell Commun Signal 2024; 22:409. [PMID: 39169379 PMCID: PMC11337594 DOI: 10.1186/s12964-024-01784-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Melanoma, one of the most lethal forms of skin cancer, has the potential to develop in any area where melanocytes are present. Currently, postoperative recurrence due to the emergence of systemic drug resistance represents a significant challenge in the treatment of melanoma. In this study, terphenyllin (TER), a distinctive inhibitory impact on melanoma cells was identified from the natural p-terphenyl metabolite. This study aimed to elucidate the intrinsic mechanism of this inhibitory effect, which may facilitate the discovery of novel chemotherapeutic agents. METHODS A transcriptome sequencing and metabolomic analysis of TER-treated A375 cells was conducted to identify potential pathways of action. The key proteins were knocked out and backfilled using CRISPR-Cas9 technology and molecular cloning. Subsequently, the results of cytosolic viability, LDH release, immunofluorescence and flow cytometry were employed to demonstrate the cell death status of the drug-treated cells. RESULTS The p53 signalling pathway was markedly upregulated following TER treatment, leading to the activation of CASP3 via the intrinsic apoptotic pathway. The activated CASP3 initiated apoptosis, while simultaneously continuing to cleave the GSDME, thereby triggering pyroptosis. The knockout of p53, a key protein situated upstream of this pathway, resulted in a significant rescue of TER-induced cell death, as well as an alleviation of the decrease in cell viability. However, the knockout of key proteins situated downstream of the pathway (CASP3 and GSDME) did not result in a rescue of TER-induced cell death, but rather a transformation of the cells from apoptosis and pyroptosis. CONCLUSIONS The induction of apoptosis and pyroptosis in A375 cells by TER is mediated via the p53-BAX/FAS-CASP3-GSDME signalling pathway. This lays the foundation for TER as a potential anti-melanoma drug in the future. It should be noted that CASP3 and GSDME in this pathway solely regulate the mode of cell death, rather than determine whether cell death occurs. This distinction may prove valuable in future studies of apoptosis and pyroptosis.
Collapse
Affiliation(s)
- Wei Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Meng-Yuan Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Ting Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Li-Na Ke
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Yan Shi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jin Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
7
|
Tufail M, Hu JJ, Liang J, He CY, Wan WD, Huang YQ, Jiang CH, Wu H, Li N. Predictive, preventive, and personalized medicine in breast cancer: targeting the PI3K pathway. J Transl Med 2024; 22:15. [PMID: 38172946 PMCID: PMC10765967 DOI: 10.1186/s12967-023-04841-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer (BC) is a multifaceted disease characterized by distinct molecular subtypes and varying responses to treatment. In BC, the phosphatidylinositol 3-kinase (PI3K) pathway has emerged as a crucial contributor to the development, advancement, and resistance to treatment. This review article explores the implications of the PI3K pathway in predictive, preventive, and personalized medicine for BC. It emphasizes the identification of predictive biomarkers, such as PIK3CA mutations, and the utility of molecular profiling in guiding treatment decisions. The review also discusses the potential of targeting the PI3K pathway for preventive strategies and the customization of therapy based on tumor stage, molecular subtypes, and genetic alterations. Overcoming resistance to PI3K inhibitors and exploring combination therapies are addressed as important considerations. While this field holds promise in improving patient outcomes, further research and clinical trials are needed to validate these approaches and translate them into clinical practice.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Ju Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cai-Yun He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Qi Huang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
8
|
Gutu N, Binish N, Keilholz U, Herzel H, Granada AE. p53 and p21 dynamics encode single-cell DNA damage levels, fine-tuning proliferation and shaping population heterogeneity. Commun Biol 2023; 6:1196. [PMID: 38001355 PMCID: PMC10673849 DOI: 10.1038/s42003-023-05585-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Cells must accurately and quickly detect DNA damage through a set of checkpoint mechanisms that enable repair and control proliferation. Heterogeneous levels of cellular stress and noisy signaling processes can lead to phenotypic variability but little is known about their role in underlying proliferation heterogeneity. Here we study two previously published single cell datasets and find that cells encode heterogeneous levels of endogenous and exogenous DNA damage to shape proliferation heterogeneity at the population level. Using a comprehensive time series analysis of short- and long-term signaling dynamics of p53 and p21, we show that DNA damage levels are quantitatively translated into p53 and p21 signal parameters in a gradual manner. Analyzing instantaneous proliferation and signaling differences among equally-radiated cells, we identify time-localized changes in the period of p53 pulses that drive cells out of a low proliferative state. Our findings suggest a novel role of the p53-p21 network in quantitatively encoding DNA damage strength and fine-tuning proliferation trajectories.
Collapse
Affiliation(s)
- Nica Gutu
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Humboldt-Universität zu Berlin, Berlin, Germany
| | - Neha Binish
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Hertie Institute for Clinical Brain Research, Center for Neurology, University Medical Center Tübingen, Tübingen, Germany
| | - Ulrich Keilholz
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- German Cancer Consortium, Deutschen Konsortiums für Translationale Krebsforschung (DKTK), Berlin, Germany
| | - Hanspeter Herzel
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Theoretical Biology, Berlin, Germany
| | - Adrián E Granada
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany.
- German Cancer Consortium, Deutschen Konsortiums für Translationale Krebsforschung (DKTK), Berlin, Germany.
| |
Collapse
|
9
|
Demircan K, Bengtsson Y, Chillon TS, Vallon-Christersson J, Sun Q, Larsson C, Malmberg M, Saal LH, Rydén L, Borg Å, Manjer J, Schomburg L. Matched analysis of circulating selenium with the breast cancer selenotranscriptome: a multicentre prospective study. J Transl Med 2023; 21:658. [PMID: 37741974 PMCID: PMC10517476 DOI: 10.1186/s12967-023-04502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/03/2023] [Indexed: 09/25/2023] Open
Abstract
INTRODUCTION Low serum selenium and altered tumour RNA expression of certain selenoproteins are associated with a poor breast cancer prognosis. Selenoprotein expression stringently depends on selenium availability, hence circulating selenium may interact with tumour selenoprotein expression. However, there is no matched analysis to date. METHODS This study included 1453 patients with newly diagnosed breast cancer from the multicentric prospective Sweden Cancerome Analysis Network - Breast study. Total serum selenium, selenoprotein P and glutathione peroxidase 3 were analysed at time of diagnosis. Bulk RNA-sequencing was conducted in matched tumour tissues. Fully adjusted Cox regression models with an interaction term were employed to detect dose-dependent interactions of circulating selenium with the associations of tumour selenoprotein mRNA expression and mortality. RESULTS 237 deaths were recorded within ~ 9 years follow-up. All three serum selenium biomarkers correlated positively (p < 0.001). All selenoproteins except for GPX6 were expressed in tumour tissues. Single cell RNA-sequencing revealed a heterogeneous expression pattern in the tumour microenvironment. Circulating selenium correlated positively with tumour SELENOW and SELENON expression (p < 0.001). In fully adjusted models, the associations of DIO1, DIO3 and SELENOM with mortality were dose-dependently modified by serum selenium (p < 0.001, p = 0.020, p = 0.038, respectively). With increasing selenium, DIO1 and SELENOM associated with lower, whereas DIO3 expression associated with higher mortality. Association of DIO1 with lower mortality was only apparent in patients with high selenium [above median (70.36 µg/L)], and the HR (95%CI) for one-unit increase in log(FPKM + 1) was 0.70 (0.50-0.98). CONCLUSIONS This first unbiased analysis of serum selenium with the breast cancer selenotranscriptome identified an effect-modification of selenium on the associations of DIO1, SELENOM, and DIO3 with prognosis. Selenium substitution in patients with DIO1-expressing tumours merits consideration to improve survival.
Collapse
Affiliation(s)
- Kamil Demircan
- Institute for Experimental Endocrinology, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH), Biomedical Innovation Academy (BIA), Berlin, Germany
| | - Ylva Bengtsson
- Department of Surgery, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Thilo Samson Chillon
- Institute for Experimental Endocrinology, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Qian Sun
- Institute for Experimental Endocrinology, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christer Larsson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Martin Malmberg
- Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lisa Rydén
- Department of Surgery, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jonas Manjer
- Department of Surgery, Skåne University Hospital Malmö, Lund University, Malmö, Sweden.
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
10
|
Witkiewicz AK, Schultz E, Wang J, Hamilton D, Levine E, O'Connor T, Knudsen ES. Determinants of response to CDK4/6 inhibitors in the real-world setting. NPJ Precis Oncol 2023; 7:90. [PMID: 37704753 PMCID: PMC10499925 DOI: 10.1038/s41698-023-00438-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
Despite widespread use and a known mechanism of action for CDK4/6 inhibitors in combination with endocrine therapy, features of disease evolution and determinants of therapeutic response in the real-world setting remain unclear. Here, a cohort of patients treated with standard-of-care combination regimens was utilized to explore features of disease and determinants of progression-free survival (PFS) and overall survival (OS). In this cohort of 280 patients, >90% of patients were treated with palbociclib in combination with either an aromatase inhibitor (AI) or fulvestrant (FUL). Most of these patients had modified Scarff-Bloom-Richardson (SBR) scores, and ER, HER2, and PR immunohistochemistry. Both the SBR score and lack of PR expression were associated with shorter PFS in patients treated with AI combinations and remained significant in multivariate analyses (HR = 3.86, p = 0.008). Gene expression analyses indicated substantial changes in cell cycle and estrogen receptor signaling during the course of treatment. Furthermore, gene expression-based subtyping indicated that predominant subtypes changed with treatment and progression. The luminal B, HER2, and basal subtypes exhibited shorter PFS in CDK4/6 inhibitor combinations when assessed in the pretreatment biopsies; however, they were not associated with OS. Using unbiased approaches, cell cycle-associated gene sets were strongly associated with shorter PFS in pretreatment biopsies irrespective of endocrine therapy. Estrogen receptor signaling gene sets were associated with longer PFS particularly in the AI-treated cohort. Together, these data suggest that there are distinct pathological and biological features of HR+/HER2- breast cancer associated with response to CDK4/6 inhibitors. Clinical trial registration number: NCT04526587.
Collapse
Affiliation(s)
- Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA.
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA.
| | - Emily Schultz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Deanna Hamilton
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Ellis Levine
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Tracey O'Connor
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA.
| |
Collapse
|
11
|
El-Sadoni M, Shboul SA, Alhesa A, Shahin NA, Alsharaiah E, Ismail MA, Ababneh NA, Alotaibi MR, Azab B, Saleh T. A three-marker signature identifies senescence in human breast cancer exposed to neoadjuvant chemotherapy. Cancer Chemother Pharmacol 2023; 91:345-360. [PMID: 36964435 DOI: 10.1007/s00280-023-04523-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/13/2023] [Indexed: 03/26/2023]
Abstract
PURPOSE Despite the beneficial effects of chemotherapy, therapy-induced senescence (TIS) manifests itself as an undesirable byproduct. Preclinical evidence suggests that tumor cells undergoing TIS can re-emerge as more aggressive divergents and contribute to recurrence, and thus, senolytics were proposed as adjuvant treatment to eliminate senescent tumor cells. However, the identification of TIS in clinical samples is essential for the optimal use of senolytics in cancer therapy. In this study, we aimed to detect and quantify TIS using matched breast cancer samples collected pre- and post-exposure to neoadjuvant chemotherapy (NAC). METHODS Detection of TIS was based on the change in gene and protein expression levels of three senescence-associated markers (downregulation of Lamin B1 and Ki-67 and upregulation of p16INK4a). RESULTS Our analysis revealed that 23 of 72 (31%) of tumors had a shift in the protein expression of the three markers after exposure to NAC suggestive of TIS. Gene expression sets of two independent NAC-treated breast cancer samples showed consistent changes in the expression levels of LMNB1, MKI67 and CDKN2A. CONCLUSIONS Collectively, our study shows a more individualized approach to measure TIS hallmarks in matched breast cancer samples and provides an estimation of the extent of TIS in breast cancer clinically. Results from this work should be complemented with more comprehensive identification approaches of TIS in clinical samples in order to adopt a more careful implementation of senolytics in cancer treatment.
Collapse
Affiliation(s)
- Mohammed El-Sadoni
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Sofian Al Shboul
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan
| | - Ahmad Alhesa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Nisreen Abu Shahin
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Elham Alsharaiah
- Department of Pathology, Royal Medical Services, King Hussein Medical Center, Amman, 11942, Jordan
| | | | - Nidaa A Ababneh
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bilal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, The Hashemite University, Zarqa, 13133, Jordan.
| |
Collapse
|
12
|
Liu ZM, Bao Y, Li TK, Di YB, Song WJ. MKI67 an potential oncogene of oral squamous cell carcinoma via the high throughput technology. Medicine (Baltimore) 2022; 101:e32595. [PMID: 36596059 PMCID: PMC9803484 DOI: 10.1097/md.0000000000032595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Oral squamous cell carcinoma is a malignant tumor that occurs in the oral cavity, with poor prognosis and easy recurrence. However, the relationship between MKI67 and oral squamous cell carcinoma remains unclear. The oral squamous cell carcinoma datasets GSE138206, GSE146483 and GSE184616 were downloaded from the gene expression omnibus database, and the differentially expressed genes (DEGs) were screened. The protein-protein interaction network was constructed and analyzed by search tool for the retrieval of interacting genes database and Cytoscape software. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) were used for functional enrichment analysis. GO and KEGG analyses were performed on the whole genome, as formulated by gene set enrichment analysis. comparative toxicogenomics database was used to identify the diseases most associated with the core genes. TargetScan was used to screen miRNA regulating central DEGs. A total of 1472 DEGs were identified. GO analysis showed that the differentially expressed genes were mainly enriched in the tissues of extracellular matrix, type i interferon signaling pathway, human papillomavirus infection, adhesion spot, hepatitis C and ECM-receptor interaction. Enrichment items were similar to GO and KEGG enrichment items of differentially expressed genes. 10 core genes were obtained, and their expression was different between oral squamous cell carcinoma and normal tissue samples. MKI67 is highly expressed in oral squamous cell carcinoma and may be an oncogene in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Zhe-Min Liu
- Department of Stomatology, Shijiazhuang Xingye Shengrui Stomatological Hospital, Shijlazhuang, Hebei Province, PR China
| | - Yang Bao
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijlazhuang,Hebei Province, PR China
- * Correspondence: Yang Bao, Department of Stomatology, The Fourth Hospital of Hebei Medical University, Changan District Health Road 12, Shijlazhuang, Hebei Province 050011, PR China (e-mail: )
| | - Tian-Ke Li
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijlazhuang,Hebei Province, PR China
| | - Yong-Bin Di
- Department of Stomatology, The First Hospital of Hebei Medical University, Shijlazhuang,Hebei Province, PR China
| | - Wei-Jing Song
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijlazhuang,Hebei Province, PR China
| |
Collapse
|
13
|
Thakran S, Cohen E, Jahani N, Weinstein SP, Pantalone L, Hylton N, Newitt D, DeMichele A, Davatzikos C, Kontos D. Impact of deformable registration methods for prediction of recurrence free survival response to neoadjuvant chemotherapy in breast cancer: Results from the ISPY 1/ACRIN 6657 trial. Transl Oncol 2022; 20:101411. [PMID: 35395604 PMCID: PMC8990167 DOI: 10.1016/j.tranon.2022.101411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/09/2022] [Accepted: 03/27/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Image registration plays a vital role in spatially aligning multiple MRI scans for better longitudinal assessment of tumor morphological features. The objective was to evaluate the effect of registration accuracy of six established deformable registration methods(ANTs, DRAMMS, ART, NiftyReg, SSD-FFD, and NMI-FFD) on the predictive value of extracted radiomic features when modeling recurrence-free-survival(RFS) for women after neoadjuvant chemotherapy(NAC) for locally advanced breast cancer. METHODS 130 women had DCE-MRI scans available from the first two visits in the ISPY1/ACRIN-6657 cohort. We calculated the transformation field from each of the different deformable registration methods, and used it to compute voxel-wise parametric-response-maps(PRM) for established four kinetic features.104-radiomic features were computed from each PRM map to characterize intra-tumor heterogeneity. We evaluated performance for RFS using Cox-regression, C-statistic, and Kaplan-Meier(KM) plots. RESULTS A baseline model(F1:Age, Race, and Hormone-receptor-status) had a 0.54 C-statistic, and model F2(baseline + functional-tumor-volume at early treatment visit(FTV2)) had 0.63. The F2+ANTs had the highest C-statistic(0.72) with the smallest landmark differences(5.40±4.40mm) as compared to other models. The KM curve for model F2 gave p=0.004 for separation between women above and below the median hazard compared to the model F1(p=0.31). A models augmented with radiomic features, also achieved significant KM curve separation(p<0.001) except the F2+ART model. CONCLUSION Incorporating image registration in quantifying changes in tumor heterogeneity during NAC can improve prediction of RFS. Radiomic features of PRM maps derived from warping the DCE-MRI kinetic maps using ANTs registration method further improved the early prediction of RFS as compared to other methods.
Collapse
Affiliation(s)
- Snekha Thakran
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Eric Cohen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Nariman Jahani
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Susan P Weinstein
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Lauren Pantalone
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Nola Hylton
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94115 United States
| | - David Newitt
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94115 United States
| | - Angela DeMichele
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Christos Davatzikos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States
| | - Despina Kontos
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 United States.
| |
Collapse
|
14
|
Shen M, Yang L, Lei T, Zhang P, Xiao L, Cao S, Chen F, Li L, Ye F, Bu H. Correlation between CA12 and TFF3 and their prediction value of neoadjuvant chemotherapy response in breast cancer. J Clin Pharm Ther 2022; 47:609-618. [PMID: 35229335 DOI: 10.1111/jcpt.13580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/01/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Compared with other molecular subtypes, hormone receptor-positive breast cancer often shows worse neoadjuvant chemotherapy efficacy. This study aims to explore the relationship between the oestrogen receptor (ER)-related genes carbonic anhydrase 12 (CA12) and trefoil factor 3 (TFF3) and their predictive value of neoadjuvant chemotherapy for breast cancer. METHODS We investigated the relationships between CA12, TFF3 and ER status and their predictive value of anthracycline-taxane neoadjuvant chemotherapy in 115 female breast cancer patients via real-time polymerase chain reaction (RT-PCR) and 4 GEO datasets: GSE41998, GSE25065, GSE20194 and GSE20271. Then, the effects of CA12 and TFF3 on the chemotherapy drugs doxorubicin and docetaxel were verified in vitro in the breast cancer cell lines MCF-7 and BT474. RESULTS AND DISCUSSION The GEO datasets and RT-PCR results showed that the relative expression of both CA12 and TFF3 was higher in oestrogen receptor-positive samples compared with the other samples (p < 0.05). CA12 was significantly correlated with TFF3 (p < 0.05). In MCF-7 cells, inhibition of TFF3 induced downregulation of CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 also downregulated TFF3 and ESR1 (p < 0.05). In BT474 cells, inhibition of TFF3 downregulated CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 led to slight upregulation of TFF3 and ESR1 (p > 0.05). Moreover, GEO datasets and RT-PCR results showed that CA12 and TFF3 were more highly expressed in nonpathological complete response (non-pCR) samples than in pCR samples (p < 0.05). Cell viability assays of MCF-7 and BT474 cells showed that inhibiting CA12 and TFF3 could enhance sensitivity to doxorubicin and docetaxel (p < 0.05). WHAT IS NEW AND CONCLUSION CA12 and TFF3 were correlated with each other, and their high expression might explain the worse efficacy of neoadjuvant chemotherapy in oestrogen receptor-positive breast cancer patients.
Collapse
Affiliation(s)
- Mengjia Shen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Libo Yang
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Lei
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Peichuan Zhang
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Xiao
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyu Cao
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fei Chen
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Li
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ye
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Bu
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Lab of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Knudsen ES, Schultz E, Hamilton D, Attwood K, Edge S, O’Connor T, Levine E, Witkiewicz AK. OUP accepted manuscript. Oncologist 2022; 27:646-654. [PMID: 35666660 PMCID: PMC9355808 DOI: 10.1093/oncolo/oyac089] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
Background A study was initiated at Roswell Park Comprehensive Cancer Center to capture the real-world experience related to the use of CDK4/6 inhibitors (Ciclibs) for the treatment of metastatic hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-). Patients and Methods A total of 222 patients were evaluated who received CDK4/6 inhibitors in the period from 2015 to 2021. Detailed clinical and demographic information was obtained on each patient and used to define clinical and demographic features associated with progression-free survival on CDK4/6 inhibitor-based therapies. Results In this real-world analysis, the majority of patients received palbociclib as the CDK4/6 inhibitor with letrozole or fulvestrant as the predominant endocrine therapies. The median progression-free survival (PFS) in the letrozole (27.6 months) and fulvestrant (17.2 months) groups were comparable to that observed in clinical trials. As expected, age at start of the treatment and menopausal status influenced endocrine therapy utilization but were not associated with PFS. Patients with recurrent disease had shorter PFS (P = .0024) than those presenting with de novo metastasis. The presence of visceral metastasis trended toward shorter PFS (P = .051). Similarly, prior endocrine therapy (P = .003) or chemotherapy (P = .036) was associated with shorter PFS. Body mass index was not associated with PFS or with dose interruption and/or modification. While the number of minorities in this analysis is limited (n = 26), these patients as a group had statistically shorter PFS on treatment (P = .002). Conclusions The real-world progression-free survival with CDK4/6 inhibitors mimics that observed in the clinical trial. A number of clinical and demographic features were associated with PFS on CDK4/6 inhibitor-based therapy. Further studies are ongoing to validate these findings incorporating additional cancer centers.
Collapse
Affiliation(s)
- Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Emily Schultz
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Deanna Hamilton
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kris Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Stephen Edge
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tracey O’Connor
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellis Levine
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Agnieszka K Witkiewicz
- Corresponding author: Agnieszka K. Witkiewicz, MD, Roswell Park Comprehensive Cancer Center, Elm and Carlton St, Buffalo, NY 14203, USA.
| |
Collapse
|
16
|
Uxa S, Castillo-Binder P, Kohler R, Stangner K, Müller GA, Engeland K. Ki-67 gene expression. Cell Death Differ 2021; 28:3357-3370. [PMID: 34183782 PMCID: PMC8629999 DOI: 10.1038/s41418-021-00823-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Ki-67 serves as a prominent cancer marker. We describe how expression of the MKI67 gene coding for Ki-67 is controlled during the cell cycle. MKI67 mRNA and Ki-67 protein are maximally expressed in G2 phase and mitosis. Expression is dependent on two CHR elements and one CDE site in the MKI67 promoter. DREAM transcriptional repressor complexes bind to both CHR sites and downregulate the expression in G0/G1 cells. Upregulation of MKI67 transcription coincides with binding of B-MYB-MuvB and FOXM1-MuvB complexes from S phase into G2/M. Importantly, binding of B-MYB to the two CHR elements correlates with loss of CHR-dependent MKI67 promoter activation in B-MYB-knockdown experiments. In knockout cell models, we find that DREAM/MuvB-dependent transcriptional control cooperates with the RB Retinoblastoma tumor suppressor. Furthermore, the p53 tumor suppressor indirectly downregulates transcription of the MKI67 gene. This repression by p53 requires p21/CDKN1A. These results are consistent with a model in which DREAM, B-MYB-MuvB, and FOXM1-MuvB together with RB cooperate in cell cycle-dependent transcription and in transcriptional repression following p53 activation. In conclusion, we present mechanisms how MKI67 gene expression followed by Ki-67 protein synthesis is controlled during the cell cycle and upon induction of DNA damage, as well as upon p53 activation.
Collapse
Affiliation(s)
- Sigrid Uxa
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Paola Castillo-Binder
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Robin Kohler
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| | - Konstanze Stangner
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany ,grid.5252.00000 0004 1936 973XPresent Address: Ludwig-Maximilians-Universität München, Anatomische Anstalt, Munich, Germany
| | - Gerd A. Müller
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany ,grid.205975.c0000 0001 0740 6917Present Address: Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA USA
| | - Kurt Engeland
- grid.9647.c0000 0004 7669 9786Molecular Oncology, Medical School, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Forouhari S, Beygi Z, Mansoori Z, Hajsharifi S, Heshmatnia F, Gheibihayat SM. Liposomes: Ideal drug delivery systems in breast cancer. Biotechnol Appl Biochem 2021; 69:1867-1884. [PMID: 34505736 DOI: 10.1002/bab.2253] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer (BC) has been recognized as the most common type of cancer in females across the world, accounting for 12% of each cancer case. In this sense, better diagnosis and screening have been thus far proven to contribute to higher survival rates. Moreover, traditional (or standard) chemotherapy is still known as one of the several prominent therapeutic options available, though it suffers from unsuitable cell selectivity, severe consequences, as well as resistance. In this regard, nanobased drug delivery systems (DDSs) are likely to provide promising grounds for BC treatment. Liposomes are accordingly effective nanosystems, having the benefits of multiple formulations verified to treat different diseases. Such systems possess specific features, including smaller size, biodegradability, hydrophobic/hydrophilic characteristics, biocompatibility, lower toxicity, as well as immunogenicity, which can all lead to considerable efficacy in treating various types of cancer. As chemotherapy uses drugs to target tumors, generates higher drug concentrations in tumors, which can provide for their slow release, and enhances drug stability, it can be improved via liposomes in DDSs for BC treatment. Therefore, the present study aims to review the existing issues regarding BC treatment and discuss liposome-based targeting in order to overcome barriers to conventional drug therapy.
Collapse
Affiliation(s)
- Sedighe Forouhari
- Infertility Research Center, Research Center of Quran, Hadith, and Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Beygi
- Department of Nursing and Midwife, Maybod Branch, Islamic Azad University, Maybod, Iran
| | - Zahra Mansoori
- Faculty of Educational Sciences and Psychology, Department of Sports Sciences, Shiraz University, Shiraz, Iran
| | - Sara Hajsharifi
- Student Research Committee, Department of Midwifery, Fatemeh (PBUH) School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Heshmatnia
- Student Research Committee, Department of Midwifery, Fatemeh (PBUH) School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
18
|
A Gene Expression Signature to Predict Nucleotide Excision Repair Defects and Novel Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22095008. [PMID: 34066883 PMCID: PMC8125907 DOI: 10.3390/ijms22095008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
Nucleotide excision repair (NER) resolves DNA adducts, such as those caused by ultraviolet light. Deficient NER (dNER) results in a higher mutation rate that can predispose to cancer development and premature ageing phenotypes. Here, we used isogenic dNER model cell lines to establish a gene expression signature that can accurately predict functional NER capacity in both cell lines and patient samples. Critically, none of the identified NER deficient cell lines harbored mutations in any NER genes, suggesting that the prevalence of NER defects may currently be underestimated. Identification of compounds that induce the dNER gene expression signature led to the discovery that NER can be functionally impaired by GSK3 inhibition, leading to synergy when combined with cisplatin treatment. Furthermore, we predicted and validated multiple novel drugs that are synthetically lethal with NER defects using the dNER gene signature as a drug discovery platform. Taken together, our work provides a dynamic predictor of NER function that may be applied for therapeutic stratification as well as development of novel biological insights in human tumors.
Collapse
|
19
|
PathoNet introduced as a deep neural network backend for evaluation of Ki-67 and tumor-infiltrating lymphocytes in breast cancer. Sci Rep 2021; 11:8489. [PMID: 33875676 PMCID: PMC8055887 DOI: 10.1038/s41598-021-86912-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/16/2021] [Indexed: 12/16/2022] Open
Abstract
The nuclear protein Ki-67 and Tumor infiltrating lymphocytes (TILs) have been introduced as prognostic factors in predicting both tumor progression and probable response to chemotherapy. The value of Ki-67 index and TILs in approach to heterogeneous tumors such as Breast cancer (BC) that is the most common cancer in women worldwide, has been highlighted in literature. Considering that estimation of both factors are dependent on professional pathologists’ observation and inter-individual variations may also exist, automated methods using machine learning, specifically approaches based on deep learning, have attracted attention. Yet, deep learning methods need considerable annotated data. In the absence of publicly available benchmarks for BC Ki-67 cell detection and further annotated classification of cells, In this study we propose SHIDC-BC-Ki-67 as a dataset for the aforementioned purpose. We also introduce a novel pipeline and backend, for estimation of Ki-67 expression and simultaneous determination of intratumoral TILs score in breast cancer cells. Further, we show that despite the challenges that our proposed model has encountered, our proposed backend, PathoNet, outperforms the state of the art methods proposed to date with regard to harmonic mean measure acquired. Dataset is publicly available in http://shiraz-hidc.com and all experiment codes are published in https://github.com/SHIDCenter/PathoNet.
Collapse
|
20
|
Senescence Marker Protein 30 (SMP30): A Novel Pan-Species Diagnostic Marker for the Histopathological Diagnosis of Breast Cancer in Humans and Animals. Int J Mol Sci 2021; 22:ijms22052340. [PMID: 33652881 PMCID: PMC7956281 DOI: 10.3390/ijms22052340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Senescence marker protein 30 (SMP30) is a cell survival factor playing an important role in vitamin C synthesis and antiapoptosis. Moreover, its cytoprotective role suggests a possibility to be related to cancer cell survival. Mammary carcinoma is a common cancer in both humans and animals. Because of its histopathological diversity, especially in the early stage, histopathological diagnosis may be complicated; therefore, a diagnostic marker is helpful for confirmation. The present study analyzed the expression pattern of SMP30 in mammary carcinoma in humans, dogs, and cats. Immunohistochemistry, immunofluorescence, and western blot analysis were used to investigate SMP30 expression patterns. The expression was specifically observed in neoplastic glandular epithelial cells. The expression increased with the malignancy of glandular epithelial cells with a highly proliferative status. However, SMP30 expression was low in normal mammary gland tissues or well-differentiated adenoma tissues. The patterns were consistently reproduced in canine primary mammary carcinoma cells and MCF-7 and MDA-MB-231 human carcinoma cell lines. This study provides useful information to understand SMP30 expression in various stages of mammary carcinoma and to suggest its utility as a pan-species diagnostic marker, thereby helping to establish strategies for diagnosing mammary carcinoma in several species.
Collapse
|
21
|
Molecular Biomarkers for Contemporary Therapies in Hormone Receptor-Positive Breast Cancer. Genes (Basel) 2021; 12:genes12020285. [PMID: 33671468 PMCID: PMC7922594 DOI: 10.3390/genes12020285] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Systemic treatment of hormone receptor-positive (HR+) breast cancer is undergoing a renaissance, with a number of targeted therapies including CDK4/6, mTOR, and PI3K inhibitors now approved for use in combination with endocrine therapies. The increased use of targeted therapies has changed the natural history of HR+ breast cancers, with the emergence of new escape mechanisms leading to the inevitable progression of disease in patients with advanced cancers. The identification of new predictive and pharmacodynamic biomarkers to current standard-of-care therapies and discovery of new therapies is an evolving and urgent clinical challenge in this setting. While traditional, routinely measured biomarkers such as estrogen receptors (ERs), progesterone receptors (PRs), and human epidermal growth factor receptor 2 (HER2) still represent the best prognostic and predictive biomarkers for HR+ breast cancer, a significant proportion of patients either do not respond to endocrine therapy or develop endocrine resistant disease. Genomic tests have emerged as a useful adjunct prognostication tool and guide the addition of chemotherapy to endocrine therapy. In the treatment-resistant setting, mutational profiling has been used to identify ESR1, PIK3CA, and AKT mutations as predictive molecular biomarkers to newer therapies. Additionally, pharmacodynamic biomarkers are being increasingly used and considered in the metastatic setting. In this review, we summarise the current state-of-the-art therapies; prognostic, predictive, and pharmacodynamic molecular biomarkers; and how these are impacted by emerging therapies for HR+ breast cancer.
Collapse
|
22
|
Parry S, Dowsett M, Dodson A. UK NEQAS ICC & ISH Ki-67 Data Reveal Differences in Performance of Primary Antibody Clones. Appl Immunohistochem Mol Morphol 2021; 29:86-94. [PMID: 33337635 PMCID: PMC7993918 DOI: 10.1097/pai.0000000000000899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/16/2020] [Indexed: 01/09/2023]
Abstract
We examined data from 374 laboratories staining for Ki-67 as part of external quality assessment over 8 runs between 2013 and 2017 (total data sets=2601). One of 5 primary antibodies was used for 94.8% of submissions, with MIB-1 (Agilent Dako) comprising 58.8% of the total. Examining assessment score as a continuous variable showed the 30-9 (Ventana) and K2 (Leica Biosystems) clones were associated with the highest mean scores (17.0; 95% confidence interval, 16.8-17.2 and 16.3; 95% confidence interval, 15.9-16.6, respectively). Stain quality was not significantly different between them. Both were associated with significantly better staining compared with MIB-1 (Agilent Dako), MM1 (Leica Biosystems), and SP6 from various suppliers (P<0.05). Similarly, categorical assessment of "Good" versus "Not good" staining quality showed that the 30-9 and K2 clones were both significantly associated with "Good" staining (both P<0.001). Other methodological parameters were examined for significant primary antibody-specific effects; none were seen for 30-9, K2, or SP6. The MM1 clone was more likely to be associated with good quality staining when it was used with Leica Biosystems sourced antigen retrieval, detection, and platform, all statistically significant at P<0.01. MIB-1 was more likely to be associated with good quality staining results when it was used with Agilent Dako antigen retrieval, detection, and staining platforms (P<0.0001), and less likely at the same significance level when used with Leica Biosystems reagents and equipment. The data presented here show the importance of not just primary antibody choice but also matching that choice to other methodological factors.
Collapse
Affiliation(s)
- Suzanne Parry
- UK National External Quality Assessment Scheme for Immunocytochemistry and In-Situ Hybridisation
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Andrew Dodson
- UK National External Quality Assessment Scheme for Immunocytochemistry and In-Situ Hybridisation
| |
Collapse
|
23
|
Lo Iacono L, Bussone S, Andolina D, Tambelli R, Troisi A, Carola V. Dissecting major depression: The role of blood biomarkers and adverse childhood experiences in distinguishing clinical subgroups. J Affect Disord 2020; 276:351-360. [PMID: 32871665 DOI: 10.1016/j.jad.2020.07.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/06/2020] [Accepted: 07/11/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND The syndromic diagnosis of major depressive disorder (MDD) is associated with individual differences in prognosis, course, treatment response, and outcome. There is evidence that patients with a history to adverse childhood experiences (ACEs) may belong to a distinct clinical subgroup. The combination of data on ACEs and blood biomarkers could allow the identification of diagnostic MDD subgroups. METHODS We selected several blood markers (global DNA methylation, and VEGF-a, TOLLIP, SIRT1, miR-34a genes) among factors that contribute to the pathogenetic mechanisms of MDD. We examined their level in 37 MDD patients and 30 healthy subjects. ACEs were measured by the Parental Bonding Instrument and the Childhood Trauma Questionnaire. RESULTS We found significant differences between patients and healthy subjects in three biomarkers (TOLLIP, VEGF-a, and global DNA methylation), independently from the confounding effect of parental care received. By contrast, SIRT1 differences were modulated by quality of parental care. The lowest levels of SIRT1 were recorded in patients with active symptoms and low maternal/paternal care. miR-34a and SIRT1 levels were associated with MDD symptoms especially in early-life stressed patients. LIMITATIONS Small sample size, no information on personality comorbidity and suicidal history, cross-sectional definition of remission, and lack of follow-up. CONCLUSIONS Our findings suggest that the levels of global DNA methylation, TOLLIP, and VEGF-a reflect pathophysiological changes associated with MDD that are independent from the long-term effects of low parental care. This study also suggests that SIRT1 may be an additional variable distinguishing the ecophenotype that includes MDD patients with exposure to ACEs.
Collapse
Affiliation(s)
| | - Silvia Bussone
- Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Via degli Apuli, 1, 00185 Rome, Italy
| | - Diego Andolina
- IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Renata Tambelli
- Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Via degli Apuli, 1, 00185 Rome, Italy
| | - Alfonso Troisi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Valeria Carola
- IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Dynamic and Clinical Psychology, Sapienza University of Rome, Via degli Apuli, 1, 00185 Rome, Italy.
| |
Collapse
|
24
|
Van Raemdonck E, Floris G, Berteloot P, Laenen A, Vergote I, Wildiers H, Punie K, Neven P. Efficacy of anti-HER2 therapy in metastatic breast cancer by discordance of HER2 expression between primary and metastatic breast cancer. Breast Cancer Res Treat 2020; 185:183-194. [PMID: 32980945 DOI: 10.1007/s10549-020-05935-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE In stage IV breast cancer, the efficacy of human epidermal growth factor receptor 2 (HER2) targeted therapies in cases with discordance in HER2 expression between primary and metastatic site is not well known. We studied progression free (PFS) and overall survival (OS) by HER2 concordance when treating women with taxane-trastuzumab (± pertuzumab) in first or second line and trastuzumab-emtansine (T-DM1) or capecitabine-lapatinib in later lines. PATIENTS AND METHODS Retrospective monocentric study including all breast cancer patients receiving trastuzumab between January 2002 and September 2017 at the University Hospital in Leuven; we selected metastatic patients with an available HER2 status in primary and metastatic site. The Kaplan-Meier method was used for estimating PFS/OS and log-rank test for analyzing between group differences. A Cox model is used for testing difference between groups while correcting for Pertuzumab. Multivariable Cox regression is used to model overall survival as a function group, correcting for possible confounders. RESULTS We included 74 patients; 46 had an unchanged HER2 status (positive/positive), 9 lost HER2 (positive/negative), while 19 acquired HER2 amplification (negative/positive). 25 out of 28 cases with a discordant HER2 status were positive for ER and/or PgR in the primary site. HER2 positive/negative cases had a significantly lower PFS for taxane-trastuzumab-(pertuzumab) (PFS = 5.5 months), compared to HER2 positive/positive (PFS 9 months, p = 0.01) and HER2 negative/positive (PFS 14 months, p = 0.01) patients. PFS for later line T-DM1 (n = 30) was significantly higher for the HER2 positive/positive group (PFS 6.0 months) than for the discordant groups HER2 negative/positive (PFS 1.0 month, p = 0.04) and HER2 positive/negative (PFS 1.5 month, p = 0.01). After correcting for possible confounders, the HER2 positive/negative group had a significantly worse OS compared to HER2 positive/positive (HR 0.19, 95% CI 0.08-0.44) and negative/positive (HR 0.15, 95% 0.06-0.38). CONCLUSION Conversion of HER2 status was seen in 28 out of 74 cases and was mostly observed in hormone receptor-positive tumors. In contrast to patients with HER2 loss, patients with a positive conversion of HER2 status derived substantial benefit from first line treatment with taxane-trastuzumab-(pertuzumab). This study highlights the importance of re-biopsying the metastatic lesion and changing treatment according to the last HER2 result.
Collapse
Affiliation(s)
- Elisa Van Raemdonck
- Department of Obstetrics and Gynecology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - G Floris
- Laboratory of Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - P Berteloot
- Department of Obstetrics and Gynecology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - A Laenen
- Department of Biostatistics, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - I Vergote
- Department of Obstetrics and Gynecology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - H Wildiers
- Department of General Medical Oncology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - K Punie
- Department of General Medical Oncology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - P Neven
- Department of Obstetrics and Gynecology, KU Leuven-University of Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
25
|
Crown A, Gonen M, Le T, Morrow M. Does Failure to Achieve Pathologic Complete Response with Neoadjuvant Chemotherapy Identify Node-Negative Patients Who Would Benefit from Postmastectomy Radiation or Regional Nodal Irradiation? Ann Surg Oncol 2020; 28:1328-1335. [PMID: 32959140 DOI: 10.1245/s10434-020-09136-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/23/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Postmastectomy radiation (PMRT) and regional nodal irradiation (RNI) improve outcomes for patients at high risk of locoregional recurrence (LRR). Node-negative patients with the triple-negative (TN) subtype and those who do not have a pCR with neoadjuvant chemotherapy (NAC) are at increased risk for LRR, but whether the absolute risk for LRR is high enough to justify PMRT/RNI is uncertain. METHODS Patients with cT1-T3N0 and pN0 disease treated with NAC who had residual disease in the breast were identified from a prospective database. Patients were eligible for the study if they had mastectomy or breast-conserving therapy with negative margins and whole-breast radiation. Those receiving PMRT or RNI were excluded. Actuarial rates were estimated using the cumulative incidence function. RESULTS The 227 patients in this study had a mean age was 51.4 ± 12.6 years, and 82 (36.1%) were TN. During a median follow-up period of 35 months, nine LRR events occurred. The overall crude and actuarial 3-year LRR rates were 4.4% and 5.9%, respectively. The crude LRR rate for the TN patients was 7.4%, and the 3-year actuarial rate was 10.1%. The hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) patients had a crude LRR rate of 2.8% and a 3-year actuarial rate of 3.2%. The HER2+ patients had a crude LRR rate of 2.7% and a 3-year actuarial rate of 3.3%. CONCLUSIONS Locoregional recurrence is uncommon for patients with node-negative HR+/HER2- and HER2+ tumors who have residual disease in the breast; however, TN patients have LRR rates that approach 10% at 3 years, suggesting a possible role for PMRT/RNI for node-negative TN patients. Additional follow-up with more patients is needed for definitive conclusions.
Collapse
Affiliation(s)
- Angelena Crown
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gonen
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tiana Le
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
26
|
Mongre RK, Mishra CB, Jung S, Lee BS, Quynh NTN, Anh NH, Myagmarjav D, Jo T, Lee MS. Exploring the Role of TRIP-Brs in Human Breast Cancer: An Investigation of Expression, Clinicopathological Significance, and Prognosis. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:105-126. [PMID: 33102693 PMCID: PMC7554327 DOI: 10.1016/j.omto.2020.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
TRIP-Brs, a group of transcription factors (TFs) that modulate several mechanisms in higher organisms. However, the novel paradigm to target TRIP-Brs in specific cancer remains to be deciphered. In particular, comprehensive analysis of TRIP-Brs in clinicopathological and patients’ prognosis, especially in breast cancer (BRCA), is being greatly ignored. Therefore, we explored the key roles of TRIP-Br expression, modulatory effects, mutations, immune infiltration, and prognosis in BRCA using multidimensional approaches. We found elevated levels of TRIP-Brs in numerous cancer tissues than normal. Higher expression of TRIP-Br-2/4/5 was shown to be positively associated with lower survival, tumor grade, and malignancy of patients with BRCA. Additionally, higher TRIP-Br-3/4 were also significantly linked with worse/short survival of BRCA patients. TRIP-Br-1/4/5 were significantly overexpressed and enhanced tumorigenesis in large-scale BRCA datasets. The mRNA levels of TRIP-Brs have been also correlated with tumor immune infiltrate in BRCA patients. In addition, TRIP-Brs synergistically play a pivotal role in central carbon metabolism, cancer-associated pathways, cell cycle, and thyroid hormone signaling, which evoke that TRIP-Brs may be a potential target for the therapy of BRCA. Thus, this investigation may lay a foundation for further research on TRIP-Br-mediated management of BRCA.
Collapse
Affiliation(s)
- Raj Kumar Mongre
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Chandra Bhushan Mishra
- College of Pharmacy, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Samil Jung
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Beom Suk Lee
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Nguyen Thi Ngoc Quynh
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Nguyen Hai Anh
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Davaajragal Myagmarjav
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Taeyeon Jo
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| | - Myeong-Sok Lee
- Molecular Cancer Biology Laboratory, Cellular Heterogeneity Research Center, Department of Biosystem, Sookmyung Women's University, Hyochangwon gil-52, Yongsan-Gu, Seoul 140-742, Republic of Korea
| |
Collapse
|
27
|
Tan B, Zhang Y, Zhang T, He J, Luo X, Bian X, Wu J, Zou C, Wang Y, Fu L. Identifying potential serum biomarkers of breast cancer through targeted free fatty acid profiles screening based on a GC-MS platform. Biomed Chromatogr 2020; 34:e4922. [PMID: 32537761 DOI: 10.1002/bmc.4922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/07/2020] [Accepted: 06/10/2020] [Indexed: 12/31/2022]
Abstract
Recent advances suggest that abnormal fatty acid metabolism highly correlates with breast cancer, which provide clues to discover potential biomarkers of breast cancer. This study aims to identify serum free fatty acid (FFA) metabolic profiles and screen potential biomarkers for breast cancer diagnosis. Gas chromatography-mass spectrometry and our in-house fatty acid methyl ester standard substances library were combined to accurately identify FFA profiles in serum samples of breast cancer patients and breast adenosis patients (as controls). Potential biomarkers were screened by applying statistical analysis. A total of 18 FFAs were accurately identified in serum sample. Two groups of patients were correctly discriminated by the orthogonal partial least squares-discriminant analysis model based on FFA profiles. Seven FFA levels were significantly higher in serum from breast cancer patients than that in controls, and exhibited positive correlation with malignant degrees of disease. Furthermore, five candidates (palmitic acid, oleic acid, cis-8,11,14-eicosatrienoic acid, docosanoic acid and the ratio of oleic acid to stearic acid) were selected as potential serum biomarkers for differential diagnosis of breast cancer. Our study will help to reveal the metabolic signature of FFAs in breast cancer patients, and provides valuable information for facilitating clinical noninvasive diagnosis.
Collapse
Affiliation(s)
- Binbin Tan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Ying Zhang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Tiantian Zhang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Jinsong He
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xueying Luo
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiqing Bian
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, China
| | - Jianlin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen People's Hospital, Shenzhen, China
| | - Yangzhi Wang
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
28
|
Wang WK, Tsai CH, Liu YW, Lai CC, Huang CC, Sheen-Chen SM. Afamin expression in breast cancer. Asian J Surg 2020; 43:750-754. [DOI: 10.1016/j.asjsur.2019.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/06/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022] Open
|
29
|
Chu CN, Wang YC, Chang WS, Wang ZH, Liu LC, Wang SC, Lin CC, Liu TY, Chang JG, Tsai CW, Yu CC, Bau DAT. Association of Interleukin-4 Polymorphisms With Breast Cancer in Taiwan. In Vivo 2020; 34:1111-1116. [PMID: 32354899 DOI: 10.21873/invivo.11882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The present study aimed at evaluating the contribution of IL-4 promoter T-1099G (rs2243248), C-589T (rs2243250), C-33T (rs2070874) genotypes to the risk of breast cancer in Taiwanese. MATERIALS AND METHODS A total of 1232 breast cancer patients and 1232 age-matched controls were genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) methodology. RESULTS Genotypic frequencies of IL-4 rs2243248, rs2243250 and rs2070874 were not differentially distributed between case and control groups. Consistently, there was no difference in the distribution of allelic frequencies among patients and controls. CONCLUSION IL-4 rs2243248, rs2243250 and rs2070874 do not confer breast cancer susceptibility in Taiwanese.
Collapse
Affiliation(s)
- Chin-Nan Chu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, R.O.C.,Division of Radiation Oncology, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yun-Chi Wang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Zhi-Hong Wang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan, R.O.C
| | - Liang-Chih Liu
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shao-Chun Wang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Cheng-Chieh Lin
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Ting-Yuan Liu
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jan-Gowth Chang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chien-Chih Yu
- School of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, R.O.C. .,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
30
|
DNA Microsystems for Biodiagnosis. MICROMACHINES 2020; 11:mi11040445. [PMID: 32340280 PMCID: PMC7231314 DOI: 10.3390/mi11040445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022]
Abstract
Researchers are continuously making progress towards diagnosis and treatment of numerous diseases. However, there are still major issues that are presenting many challenges for current medical diagnosis. On the other hand, DNA nanotechnology has evolved significantly over the last three decades and is highly interdisciplinary. With many potential technologies derived from the field, it is natural to begin exploring and incorporating its knowledge to develop DNA microsystems for biodiagnosis in order to help address current obstacles, such as disease detection and drug resistance. Here, current challenges in disease detection are presented along with standard methods for diagnosis. Then, a brief overview of DNA nanotechnology is introduced along with its main attractive features for constructing biodiagnostic microsystems. Lastly, suggested DNA-based microsystems are discussed through proof-of-concept demonstrations with improvement strategies for standard diagnostic approaches.
Collapse
|
31
|
Hormone receptor, human epidermal growth factor receptor-2, and Ki-67 status in primary breast cancer and corresponding recurrences or synchronous axillary lymph node metastases. Surg Today 2019; 50:657-663. [PMID: 31190183 DOI: 10.1007/s00595-019-01831-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/14/2019] [Indexed: 01/22/2023]
Abstract
The therapeutic strategy for breast cancer is determined by the surrogate subtype, which is defined by biomarkers, such as estrogen receptor (ER), progesterone receptor (PgR), human epidermal growth factor receptor-2 (HER2), and Ki-67. In previous reports, the rate of discordance in ER, PgR, and HER2 between primary breast cancer and recurrent lesions or synchronous axillary lymph node metastasis was 15-25, 25-40, and 5-25 or 7-50, 10-50, and 3-30%, respectively. Overall, hormone receptors tended to weaken during the metastatic process, while patterns of HER2 were not uniform. Regarding the Ki-67 labeling index, an increase in metastatic lesions compared with primary lesions was the dominant pattern, suggesting that aggressive subclones with high proliferative potential form metastases. The loss of expression of hormone receptor or an increase in the Ki-67 labeling index in metastasis seemed to be associated with a poor prognosis. However, most previous studies did not report the background characteristics of patients, or they included subjects with varied characteristics, including those on systemic therapy, and were based on relatively small populations; therefore, definitive conclusions could not be drawn. Future studies should explore how to select therapies according to the biomarkers in primary breast cancer and/or its metastasis.
Collapse
|
32
|
Aman NA, Doukoure B, Koffi KD, Koui BS, Traore ZC, Kouyate M, Effi AB. HER2 overexpression and correlation with other significant clinicopathologic parameters in Ivorian breast cancer women. BMC Clin Pathol 2019; 19:1. [PMID: 30675127 PMCID: PMC6335844 DOI: 10.1186/s12907-018-0081-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022] Open
Abstract
Background The overexpression of HER2 is associated with worse prognosis of breast cancer which responds favourably to anti-HER2 therapy. The objective of this study was to determine the frequency of HER2 and its association with clinicopathologic factors in breast cancer in Ivory Coast. Methods The study included 608 patients who were histologically diagnosed with invasive primary breast carcinoma. The immunohistochemistry testing for ER, PR, and HER2 was performed on the formalin fixed paraffin-embedded blocks of breast tissue of these patients. The analysis of variance and the Chi-Square Test were used to examine the association of the HER2 status with clinicopathologic prognostic features. Results The average age of patients was 47 ± 11 years. Among 608 patients, 355 (58.4%) were premenopausal. Invasive ductal carcinoma of no specific type (511 cases, 84.1%) was the most frequent histologic type. Grade II tumors were 59.8%. The positivity of ER, PR, and ER/PR was 334 cases (54.9%), 252 cases (41.4%), and 356 cases (58.5%), respectively. HER2 was overexpressed in 105 cases (17.3%). The overexpression of HER2 was significantly correlated with Nottingham grade (p = 0.007). No association was observed between HER2 expression and age (p = 0.568), menopausal status (p = 0.929), histologic type (p = 0.666), ER (p = 0.137), PR (p = 0.396), and ER/PR (p = 0.134). Conclusion Breast cancer occurs in young women. HER2 status is closely related to Nottingham grade. The immunohistochemical analysis of HER2 has prognostic and therapeutic implications, and thus, contributing to efficient clinical management of patients.
Collapse
Affiliation(s)
- Nguiessan Alphonse Aman
- Department of Anatomic Pathology, School of Medicine, Alassane Ouattara University, BP V 18 Bouake, Bouake, Ivory Coast
| | - Brahima Doukoure
- Department of Anatomic Pathology, School of Medicine, Felix H Boigny University, 01 BP V 34 Abidjan 01, Abidjan, Ivory Coast
| | - Kouadio Donatien Koffi
- Department of Anatomic Pathology, School of Medicine, Alassane Ouattara University, BP V 18 Bouake, Bouake, Ivory Coast
| | - Baumaney Sylvanus Koui
- Department of Anatomic Pathology, School of Medicine, Felix H Boigny University, 01 BP V 34 Abidjan 01, Abidjan, Ivory Coast
| | - Zie Cheick Traore
- Department of Anatomic Pathology, School of Medicine, Alassane Ouattara University, BP V 18 Bouake, Bouake, Ivory Coast
| | - Mohamed Kouyate
- Department of Anatomic Pathology, School of Medicine, Felix H Boigny University, 01 BP V 34 Abidjan 01, Abidjan, Ivory Coast
| | - Ahoua Benjamin Effi
- Department of Anatomic Pathology, School of Medicine, Alassane Ouattara University, BP V 18 Bouake, Bouake, Ivory Coast
| |
Collapse
|
33
|
Peng JH, Zhang X, Song JL, Ran L, Luo R, Li HY, Wang YH. Neoadjuvant chemotherapy reduces the expression rates of ER, PR, HER2, Ki67, and P53 of invasive ductal carcinoma. Medicine (Baltimore) 2019; 98:e13554. [PMID: 30633152 PMCID: PMC6336639 DOI: 10.1097/md.0000000000013554] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
To analyze whether neoadjuvant chemotherapy (NAC) changes the expression rates of invasive ductal carcinoma (IDC) markers: estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), Ki67, and P53.This was a retrospective study of 112 IDC patients who underwent NAC (docetaxel+epirubicin/pirarubicin+cyclophosphamide) but without pathological complete response (pCR) in 2012 to 2013 at the First Affiliated Hospital of Chongqing Medical University. The IDC subtypes and tumor protein markers were analyzed by immunohistochemistry (IHC). Specific changes in tumor protein markers before/after NAC were compared.The decrease in the positive rate of Ki-67 was the most significant, from 75.9% before NAC to 41.1% after NAC (P < .001). The positive rate of HER2 decreased from 42.0% before NAC to 32.1% after NAC (P = .04). The positive rate of ER decreased from 66.1% before NAC to 56.2% after NAC (P = .04). Increased number of metastatic lymph nodes (P = .006) and body mass index (BMI) (P = .028) seemed to be related to conversion of PR (positive to negative). There was statistical association between the Ki-67 (positive to negative) with the age greater or equal to 50 (P = .015). The BMI greater or equal to 24 (P = .021), age greater or equal to 50 (P = .047), and blood type A (P = .038) were independently associated with conversion of P53 (positive to negative). The BMI greater or equal to 24 (P = .004), number of metastatic lymph nodes greater or equal to 1 (P = .029) and TNM stages I-II (P = .008) were statistically associated with change of HER2 (positive to negative).In patients without pCR, NAC leads to changes in Ki-67, HER2, and hormone receptor (HR) expression. Age, BMI, number of metastatic lymph nodes, and TNM stage are associated with some changes of markers.
Collapse
MESH Headings
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Drug Therapy, Combination
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Ki-67 Antigen/metabolism
- Middle Aged
- Neoadjuvant Therapy
- Neoplasm Invasiveness
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Jian-Heng Peng
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University
| | - Xiang Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing
| | - Jun-Long Song
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liang Ran
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University
| | - Rong Luo
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University
| | - Hong-Yuan Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing
| | - Yong-Hong Wang
- Health Management Center, The First Affiliated Hospital of Chongqing Medical University
| |
Collapse
|
34
|
Sheen-Chen SM, Tsai CH, Liu YW, Huang CC. Netrin-1 expression in breast cancer. JOURNAL OF CANCER RESEARCH AND PRACTICE 2019. [DOI: 10.4103/jcrp.jcrp_8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
35
|
Yuan M, Liao J, Luo J, Cui M, Jin F. Significance of Vesicle-Associated Membrane Protein 8 Expression in Predicting Survival in Breast Cancer. J Breast Cancer 2018; 21:399-405. [PMID: 30607161 PMCID: PMC6310720 DOI: 10.4048/jbc.2018.21.e57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 09/21/2018] [Indexed: 12/23/2022] Open
Abstract
Purpose Vesicle-associated membrane protein 8 (VAMP8) is a soluble N-ethylmaleimide-sensitive factor receptor protein that participates in autophagy by directly regulating autophagosome membrane fusion and has been reported to be involved in tumor progression. Nevertheless, the expression and prognostic value of VAMP8 in breast cancer (BC) remain unknown. This study aimed to evaluate the clinical significance and biological function of VAMP8 in BC. Methods A total of 112 BC samples and 30 normal mammary gland samples were collected. The expression of VAMP8 was assessed in both BC tissues and normal mammary gland tissues via a two-step immunohistochemical detection method. Results The expression of VAMP8 in BC tissues was significantly higher than that in normal breast tissues. Furthermore, increased VAMP8 expression was significantly correlated with tumor size (p=0.007), lymph node metastasis (p=0.024) and recurrence (p=0.001). Patients with high VAMP8 expression had significantly lower cumulative recurrence-free survival and overall survival (p<0.001 for both) than patients with low VAMP8 expression. In multivariate logistic regression and Cox regression analyses, lymph node metastasis and VAMP8 expression were independent prognostic factors for BC. Conclusion VAMP8 is significantly upregulated in human BC tissues and can thus be a practical and potentially effective surrogate marker for survival in BC patients.
Collapse
Affiliation(s)
- Mengci Yuan
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jianhua Liao
- Department of General Surgery, Zhejiang Hospital, Hangzhou, China
| | - Ji Luo
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mengyao Cui
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Division of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Miller I, Min M, Yang C, Tian C, Gookin S, Carter D, Spencer SL. Ki67 is a Graded Rather than a Binary Marker of Proliferation versus Quiescence. Cell Rep 2018; 24:1105-1112.e5. [PMID: 30067968 PMCID: PMC6108547 DOI: 10.1016/j.celrep.2018.06.110] [Citation(s) in RCA: 390] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/11/2018] [Accepted: 06/28/2018] [Indexed: 02/07/2023] Open
Abstract
Ki67 staining is widely used as a proliferation indicator in the clinic, despite poor understanding of this protein's function or dynamics. Here, we track Ki67 levels under endogenous control in single cells over time and find that Ki67 accumulation occurs only during S, G2, and M phases. Ki67 is degraded continuously in G1 and G0 phases, regardless of the cause of entry into G0/quiescence. Consequently, the level of Ki67 during G0 and G1 in individual cells is highly heterogeneous and depends on how long an individual cell has spent in G0. Thus, Ki67 is a graded rather than a binary marker both for cell-cycle progression and time since entry into quiescence.
Collapse
Affiliation(s)
- Iain Miller
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Mingwei Min
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Chen Yang
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA; Department of Molecular, Cellular, and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Chengzhe Tian
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Sara Gookin
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Dylan Carter
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA
| | - Sabrina L Spencer
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303, USA; BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
37
|
Singh KP, Miaskowski C, Dhruva AA, Flowers E, Kober KM. Mechanisms and Measurement of Changes in Gene Expression. Biol Res Nurs 2018; 20:369-382. [PMID: 29706088 PMCID: PMC6346310 DOI: 10.1177/1099800418772161] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Research on gene expression (GE) provides insights into the physiology of a cell or group of cells at a given point in time. Studies of changes in GE can be used to identify patients at higher risk for various medical conditions, a higher symptom burden, and/or the adverse consequences associated with various treatments. The aims of this article are as follows: (1) to describe the different types of RNA transcripts, (2) to describe the processes involved in GE (i.e., RNA transcription, epigenetics, and posttranscriptional modifications), (3) to describe common sources of variation in GE, (4) to describe the most common methods used to measure GE, and (5) to discuss factors to consider when choosing tissue for a GE study. This article begins with an overview of the mechanisms involved in GE. Then, the factors that can influence the findings from GE experiments (e.g., tissue specificity, host age, host gender, and time of sample collection) are described and potential solutions are presented. This article concludes with a discussion of how the types of tissue used in GE studies can affect study findings. Given that the costs associated with the measurement of changes in GE are decreasing and the methods to analyze GE data are becoming easier to use, nurse scientists need to understand the basic principles that underlie any GE study.
Collapse
Affiliation(s)
- Komal P. Singh
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, USA
| | - Christine Miaskowski
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, USA
| | - Anand A. Dhruva
- School of Medicine, University of California, San Francisco, CA, USA
| | - Elena Flowers
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, USA
| | - Kord M. Kober
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, USA
| |
Collapse
|
38
|
Peripheral biomarkers of major depression and antidepressant treatment response: Current knowledge and future outlooks. J Affect Disord 2018; 233:3-14. [PMID: 28709695 PMCID: PMC5815949 DOI: 10.1016/j.jad.2017.07.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/19/2017] [Accepted: 07/03/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND In recent years, we have accomplished a deeper understanding about the pathophysiology of major depressive disorder (MDD). Nevertheless, this improved comprehension has not translated to improved treatment outcome, as identification of specific biologic markers of disease may still be crucial to facilitate a more rapid, successful treatment. Ongoing research explores the importance of screening biomarkers using neuroimaging, neurophysiology, genomics, proteomics, and metabolomics measures. RESULTS In the present review, we highlight the biomarkers that are differentially expressed in MDD and treatment response and place a particular emphasis on the most recent progress in advancing technology which will continue the search for blood-based biomarkers. LIMITATIONS Due to space constraints, we are unable to detail all biomarker platforms, such as neurophysiological and neuroimaging markers, although their contributions are certainly applicable to a biomarker review and valuable to the field. CONCLUSIONS Although the search for reliable biomarkers of depression and/or treatment outcome is ongoing, the rapidly-expanding field of research along with promising new technologies may provide the foundation for identifying key factors which will ultimately help direct patients toward a quicker and more effective treatment for MDD.
Collapse
|
39
|
Andergassen U, Schlenk K, Jeschke U, Sommer H, K�lbl A. Epithelial‑mesenchymal transition was identified as a potential marker for breast cancer aggressiveness using reverse transcription‑quantitative polymerase chain reaction. Mol Med Rep 2018; 18:1733-1739. [DOI: 10.3892/mmr.2018.9091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/11/2017] [Indexed: 11/06/2022] Open
Affiliation(s)
- Ulrich Andergassen
- Department of Gynecology and Obstetrics, LMU Munich, D‑80337 Munich, Germany
| | - Kristina Schlenk
- Department of Gynecology and Obstetrics, LMU Munich, D‑80337 Munich, Germany
| | - Udo Jeschke
- Department of Gynecology and Obstetrics, LMU Munich, D‑80337 Munich, Germany
| | - Harald Sommer
- Department of Gynecology and Obstetrics, LMU Munich, D‑80337 Munich, Germany
| | - Alexandra K�lbl
- Department of Gynecology and Obstetrics, LMU Munich, D‑80337 Munich, Germany
| |
Collapse
|
40
|
Bonde GV, Yadav SK, Chauhan S, Mittal P, Ajmal G, Thokala S, Mishra B. Lapatinib nano-delivery systems: a promising future for breast cancer treatment. Expert Opin Drug Deliv 2018. [DOI: 10.1080/17425247.2018.1449832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Gunjan Vasant Bonde
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sarita Kumari Yadav
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
- Department of Pharmacy, Moti Lal Nehru Medical College, Allahabad, India
| | - Sheetal Chauhan
- Department of Pharmacology, Melaka Manipal Medical College, Manipal University, Manipal, India
| | - Pooja Mittal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Gufran Ajmal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Sathish Thokala
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
41
|
New Developments in Breast Cancer Prognosis: Molecular Predictors of Treatment Response and Survival. Int J Biol Markers 2018; 28:131-40. [PMID: 23709349 DOI: 10.5301/jbm.5000027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2013] [Indexed: 01/08/2023]
Abstract
Aim This study aimed to assess the molecular subtypes of breast cancer for patients attending a dedicated breast care center and examine the association with clinicopathological features, treatment and survival outcomes. Methods Demographic, clinicopathological and treatment details were collected from women with primary breast cancer. Immunohistochemical subtypes were also collected. The association between breast cancer subtypes and clinicopathological features was assessed using the chi-square or Fisher's exact test. Survival outcomes were compared among subtypes with the log-rank test. Results Immunohistochemical subtypes were not associated with tumor size, lymphovascular invasion or lymph node involvement but differed by histological grade (p=0.014) and nuclear grade of tumors (p=0.001). The 5-year overall survival estimates for luminal A, luminal B, HER-2-positive and triple-negative tumors were 100%, 96.2%, 71.4% and 92.3% respectively. Compared to luminal A tumors (93.4%), luminal B (80.8%), HER-2-positive (71.4%) and triple-negative (76.9%) tumors exhibited a reduced disease-free survival (DFS). Patients with ER-positive tumors had a higher DFS than their ER-negative counterparts (p=0.036). Patients with tumors expressing a low Ki-67 level had a more favorable prognosis (p=0.02). Conclusions The most prevalent luminal A subtype is associated with relatively better prognosis, whereas HER-2-positive and triple-negative tumors are prone to early relapse with diminished survival.
Collapse
|
42
|
Helal DS, El-Guindy DM. Maspin expression and subcellular localization in invasive ductal carcinoma of the breast: Prognostic significance and relation to microvessel density. J Egypt Natl Canc Inst 2017; 29:177-183. [PMID: 29126758 DOI: 10.1016/j.jnci.2017.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 12/20/2022] Open
Abstract
Maspin (Mammary serine protease inhibitor) is a tumor suppressor serine. Its clinical significance and role in breast carcinoma are contradictory and inconclusive. Researches demonstrated that the function of maspin differs according to its subcellular localization. This study was conducted to investigate the expression of maspin in invasive ductal carcinoma (IDC) of the breast with special emphasis on its subcellular localization and to evaluate its prognostic role in relation to clinicopathological parameters and microvessel density (MVD) of the tumor. The expression of maspin was evaluated immunohistochemically in 45 IDC cases. The positive rate of maspin expression was 73.3%. Maspin positivity was significantly related to higher tumor grade (p value = 0.041), nodal metastasis (p value = 0.044), perineural invasion (p value = 0.047), and high CD34+MVD (p value = 0.002). Nuclear maspin was detected in 36.6% whereas cytoplasmic maspin was detected in 63.4% of maspin positive cases. A significant inverse relationship was observed between nuclear maspin and high tumor grade (p value = 0.016), and nodal metastasis (p value = 0.047). These results suggest that maspin expression has a prognostic role in breast cancer. Maspin expression is related to increased angiogenesis. Subcellular localization of maspin can strongly affect cancer prognosis. Cytoplasmic maspin relates to poor prognostic parameters whereas nuclear maspin relates to good prognostic ones.
Collapse
Affiliation(s)
- Duaa S Helal
- Pathology Department, Faculty of Medicine, Tanta University, Egypt
| | - Dina M El-Guindy
- Pathology Department, Faculty of Medicine, Tanta University, Egypt.
| |
Collapse
|
43
|
Presurgical weight loss affects tumour traits and circulating biomarkers in men with prostate cancer. Br J Cancer 2017; 117:1303-1313. [PMID: 28881355 PMCID: PMC5672928 DOI: 10.1038/bjc.2017.303] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/27/2017] [Accepted: 08/09/2017] [Indexed: 01/11/2023] Open
Abstract
Background: Obesity is associated with aggressive prostate cancer. To explore whether weight loss favourably affects tumour biology and other outcomes, we undertook a presurgical trial among overweight and obese men with prostate cancer. Methods: This single-blinded, two-arm randomised controlled trial explored outcomes of a presurgical weight loss intervention (WLI) that promoted ∼1 kg per week loss via caloric restriction and increased physical activity (PA). Forty overweight/obese men with clinically confirmed prostate cancer were randomised to the WLI presurgery or to a control arm; changes in weight, body composition, quality-of-life, circulating biomarkers, gene expression, and immunohistochemical markers in tumour and benign prostatic tissue were evaluated. Results: The study period averaged 50 days. Mean (s.d.) change scores for the WLI vs control arms were as follows: weight: −4.7 (3.1) kg vs −2.2 (4.4) kg (P=0.0508); caloric intake: −500 (636) vs −159 (600) kcal per day (P=0.0034); PA: +0.9 (3.1) vs +1.7 (4.6) MET-hours per day (NS); vitality: +5.3 (7.l4) vs −1.8 (8.1) (P=0.0491); testosterone: +55.1 (86.0) vs −48.3 (203.7) ng dl−1 (P=0.0418); sex hormone-binding globulin: +14.0 (14.6) vs +1.8 (7.6) nmol l−1 (P=0.0023); and leptin: −2.16 (2.6) vs −0.03 (3.75) (P=0.0355). Follow-up Ki67 was significantly higher in WLI vs control arms; median (interquartile range): 5.0 (2.5,10.0) vs 0.0 (0.0,2.5) (P=0.0061) and several genes were upregulated, for example, CTSL, GSK3B, MED12, and LAMC2. Conclusions: Intentional weight loss shows mixed effects on circulating biomarkers, tumour gene expression, and proliferative markers. More study is needed before recommending weight loss, in particular rapid weight loss, among men with prostate cancer.
Collapse
|
44
|
Sadeghi-Naini A, Sannachi L, Tadayyon H, Tran WT, Slodkowska E, Trudeau M, Gandhi S, Pritchard K, Kolios MC, Czarnota GJ. Chemotherapy-Response Monitoring of Breast Cancer Patients Using Quantitative Ultrasound-Based Intra-Tumour Heterogeneities. Sci Rep 2017; 7:10352. [PMID: 28871171 PMCID: PMC5583340 DOI: 10.1038/s41598-017-09678-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer therapies including chemotherapy aim to induce tumour cell death. Cell death introduces alterations in cell morphology and tissue micro-structures that cause measurable changes in tissue echogenicity. This study investigated the effectiveness of quantitative ultrasound (QUS) parametric imaging to characterize intra-tumour heterogeneity and monitor the pathological response of breast cancer to chemotherapy in a large cohort of patients (n = 100). Results demonstrated that QUS imaging can non-invasively monitor pathological response and outcome of breast cancer patients to chemotherapy early following treatment initiation. Specifically, QUS biomarkers quantifying spatial heterogeneities in size, concentration and spacing of acoustic scatterers could predict treatment responses of patients with cross-validated accuracies of 82 ± 0.7%, 86 ± 0.7% and 85 ± 0.9% and areas under the receiver operating characteristic (ROC) curve of 0.75 ± 0.1, 0.80 ± 0.1 and 0.89 ± 0.1 at 1, 4 and 8 weeks after the start of treatment, respectively. The patients classified as responders and non-responders using QUS biomarkers demonstrated significantly different survivals, in good agreement with clinical and pathological endpoints. The results form a basis for using early predictive information on survival-linked patient response to facilitate adapting standard anti-cancer treatments on an individual patient basis.
Collapse
Affiliation(s)
- Ali Sadeghi-Naini
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Lakshmanan Sannachi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hadi Tadayyon
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - William T Tran
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Centre for Health and Social Care Research, Sheffield Hallam University, Sheffield, UK
| | - Elzbieta Slodkowska
- Division of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maureen Trudeau
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sonal Gandhi
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Kathleen Pritchard
- Division of Medical Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
45
|
Kang TH, Seo JH, Oh H, Yoon G, Chae JI, Shim JH. Licochalcone A Suppresses Specificity Protein 1 as a Novel Target in Human Breast Cancer Cells. J Cell Biochem 2017; 118:4652-4663. [PMID: 28498645 DOI: 10.1002/jcb.26131] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/11/2017] [Indexed: 12/24/2022]
Abstract
Licochalcone A (LCA), isolated from the root of Glycyrrhiza inflata, are known to have medicinal effect such as anti-oxidant, anti-bacterial, anti-viral, and anti-cancer. Though, as a pharmacological mechanism regulator, anti-cancer studies on LCA were not investigated in human breast cancer. We investigated the anti-proliferative and apoptotic effect of LCA in human breast cancer cells MCF-7 and MDA-MB-231 through MTS assay, PI staining, Annexin-V/7-AAD assay, mitochondrial membrane potential assay, multi-caspase assay, RT-PCR, Western blot analysis, and anchorage-independent cell transformation assay. Our results showed the little difference between two cells, as MCF-7 cell is both estrogen/progesterone receptor positive, there were only effect on Sp1 protein level, but not in mRNA level. Adversely, estrogen/progesterone/human epidermal growth factor receptor 2 triple negative, MDA-MB-231 showed decreased Sp1 mRNA, and protein levels. To confirm the participation of Sp1 in breast cancer cell viability, siRNA techniques were introduced. Both cells showed dysfunction of mitochondrial membrane potential and mitochondrial ROS production, which reflects it passed intracellular mitochondrial apoptosis pathway. Additionally, LCA showed the anti-proliferative and apoptotic effect in breast cancer cells through regulating Sp1 and apoptosis-related proteins in a dose- and a time-dependent manner. Consequently, LCA might be a potential anti-breast cancer drug substitute. J. Cell. Biochem. 118: 4652-4663, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tae-Ho Kang
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Hana Oh
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea.,The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| |
Collapse
|
46
|
The role of quantitative estrogen receptor status in predicting tumor response at surgery in breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat 2017; 164:285-294. [DOI: 10.1007/s10549-017-4269-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/27/2017] [Indexed: 11/27/2022]
|
47
|
Inari H, Suganuma N, Kawachi K, Yoshida T, Yamanaka T, Nakamura Y, Yoshihara M, Nakayama H, Yamanaka A, Masudo K, Oshima T, Yokose T, Rino Y, Shimizu S, Miyagi Y, Masuda M. Expression of enhancer of zeste homolog 2 correlates with survival outcome in patients with metastatic breast cancer: exploratory study using primary and paired metastatic lesions. BMC Cancer 2017; 17:160. [PMID: 28241804 PMCID: PMC5330119 DOI: 10.1186/s12885-017-3154-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 02/22/2017] [Indexed: 12/16/2022] Open
Abstract
Background In metastatic breast cancer, the status of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), as well as the Ki-67 index sometimes change between primary and metastatic lesions. However, the change in expression levels of enhancer of zeste homolog 2 (EZH2) between primary and metastatic lesions has not been determined in metastatic breast cancer. Methods Ninety-six metastatic breast cancer patients had biopsies or resections of metastatic lesions between September 1990 and February 2014 at the Kanagawa Cancer Center. We evaluated ER, PR, HER2, Ki-67, and EZH2 in primary lesions and their corresponding metastatic lesions using immunohistochemistry. We examined the change in expression of EZH2 between primary and metastatic lesions, the correlation between the expression of EZH2 and the expression of other biomarkers, and the relationship between EZH2 expression and patient outcome in metastatic breast cancer. Results EZH2 expression was significantly higher in metastatic lesions compared with primary lesions. EZH2 expression was highly correlated with Ki-67 expression in primary and metastatic lesions. High-level expression of EZH2 was associated with poorer disease-free survival (DFS) outcomes in patients with primary lesions (P < 0.001); however, high-level expression of EZH2 was not associated with poorer DFS outcomes in patients with metastatic lesions (P = 0.063). High-level expression of EZH2 was associated with poorer overall survival (OS) postoperatively in patients with primary (P = 0.001) or metastatic lesions (P = 0.005). High-level expression of EZH2 was associated with poorer OS outcomes after recurrence in patients with metastatic lesions (P = 0.014); however, high-level expression of EZH2 was not associated with poorer OS outcomes after recurrence in patients with primary lesions (P = 0.096). High-level expression of EZH2 in metastatic lesions was independently associated with poorer OS outcomes after recurrence. Conclusions EZH2 expression was significantly increased in metastatic lesions compared with primary lesions. High-level expression of EZH2 in metastatic lesions was associated with poorer OS outcomes after primary surgery and recurrence. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3154-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hitoshi Inari
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan.
| | - Nobuyasu Suganuma
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Kae Kawachi
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Tatsuya Yoshida
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Takashi Yamanaka
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Hirotaka Nakayama
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Ayumi Yamanaka
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Katsuhiko Masudo
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takashi Oshima
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Yasushi Rino
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Satoru Shimizu
- Department of Breast and Endocrine Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-0815, Japan.
| | - Munetaka Masuda
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
48
|
De Souza LM, Robertson BM, Robertson GP. Future of circulating tumor cells in the melanoma clinical and research laboratory settings. Cancer Lett 2017; 392:60-70. [PMID: 28163189 DOI: 10.1016/j.canlet.2017.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/24/2022]
Abstract
Circulating tumor cells (CTC) have become a field of interest for oncologists based on the premise that they constitute the underpinning for metastatic dissemination. The lethal nature of cancer is no longer attributed to solid tumor formation, but rather to the process of metastasis; shifting the focus of current studies towards the isolation and identification of metastatic progenitors, such as CTCs. CTCs originate from primary tumor masses that undergo morphologic and genetic alterations, which involve the release of mesenchymal-like cancer cells into the bloodstream, capable of invading nearby tissues for secondary tumor development. Cancerous cells contained in the primary tumor mass acquire the motile mesenchymal phenotype as a result of the Epithelial-to-Mesenchymal Transition, where substantial variations in protein expression and signaling pathways take place. CTCs that migrate from the primary tumor, intravasate into the systemic vasculature, are transported through the bloodstream, and invade tissues and organs suitable for secondary tumor development. While only a limited number of CTCs are viable in the bloodstream, their ability to elude the immune system, evade apoptosis and successfully metastasize at secondary tumor sites, makes CTCs promising candidates for unraveling the triggers that initiates the metastatic process. In this article, these subjects are explored in greater depth to elucidate the potential use of CTCs in the detection, disease staging and management of metastatic melanoma.
Collapse
Affiliation(s)
- Luisa M De Souza
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA.
| | - Bailey M Robertson
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA
| | - Gavin P Robertson
- The Pennsylvania State University College of Medicine, Departments of Pharmacology, 500 University Drive, Hershey, PA 17033, USA; Pathology, 500 University Drive, Hershey, PA 17033, USA; Dermatology, 500 University Drive, Hershey, PA 17033, USA; Surgery, 500 University Drive, Hershey, PA 17033, USA; The Melanoma and Skin Cancer Center, 500 University Drive, Hershey, PA 17033, USA; The Melanoma Therapeutics Program, 500 University Drive, Hershey, PA 17033, USA.
| |
Collapse
|
49
|
Badr HA, AlSadek DMM, El-Houseini ME, Saeui CT, Mathew MP, Yarema KJ, Ahmed H. Harnessing cancer cell metabolism for theranostic applications using metabolic glycoengineering of sialic acid in breast cancer as a pioneering example. Biomaterials 2017; 116:158-173. [PMID: 27926828 PMCID: PMC5193387 DOI: 10.1016/j.biomaterials.2016.11.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/14/2016] [Accepted: 11/24/2016] [Indexed: 12/18/2022]
Abstract
Abnormal cell surface display of sialic acids - a family of unusual 9-carbon sugars - is widely recognized as distinguishing feature of many types of cancer. Sialoglycans, however, typically cannot be identified with sufficiently high reproducibility and sensitivity to serve as clinically accepted biomarkers and similarly, almost all efforts to exploit cancer-specific differences in sialylation signatures for therapy remain in early stage development. In this report we provide an overview of important facets of glycosylation that contribute to cancer in general with a focus on breast cancer as an example of malignant disease characterized by aberrant sialylation. We then describe how cancer cells experience nutrient deprivation during oncogenesis and discuss how the resulting metabolic reprogramming, which endows breast cancer cells with the ability to obtain nutrients during scarcity, constitutes an "Achilles' heel" that we believe can be exploited by metabolic glycoengineering (MGE) strategies to develop new diagnostic methods and therapeutic approaches. In particular, we hypothesize that adaptations made by breast cancer cells that allow them to efficiently scavenge sialic acid during times of nutrient deprivation renders them vulnerable to MGE, which refers to the use of exogenously-supplied, non-natural monosaccharide analogues to modulate targeted aspects of glycosylation in living cells and animals. In specific, once non-natural sialosides are incorporated into the cancer "sialome" they can be exploited as epitopes for immunotherapy or as chemical tags for targeted delivery of imaging or therapeutic agents selectively to tumors.
Collapse
Affiliation(s)
- Haitham A Badr
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Dina M M AlSadek
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Motawa E El-Houseini
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Christopher T Saeui
- Department of Biomedical Engineering and Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Mohit P Mathew
- Department of Biomedical Engineering and Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Kevin J Yarema
- Department of Biomedical Engineering and Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD 21231, USA.
| | - Hafiz Ahmed
- GlycoMantra, Inc., Baltimore, MD 21227, USA.
| |
Collapse
|
50
|
Shi W, Bruce J, Lee M, Yue S, Rowe M, Pintilie M, Kogo R, Bissey PA, Fyles A, Yip KW, Liu FF. MiR-449a promotes breast cancer progression by targeting CRIP2. Oncotarget 2017; 7:18906-18. [PMID: 26934316 PMCID: PMC4951339 DOI: 10.18632/oncotarget.7753] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/14/2016] [Indexed: 12/12/2022] Open
Abstract
The identification of prognostic biomarkers and their underlying mechanisms of action remain of great interest in breast cancer biology. Using global miRNA profiling of 71 lymph node-negative invasive ductal breast cancers and 5 normal mammary epithelial tissues, we identified miR-449a to be highly overexpressed in the malignant breast tissue. Its expression was significantly associated with increased incidence of patient relapse, decreased overall survival, and decreased disease-free survival. In vitro, miR-449a promoted breast cancer cell proliferation, clonogenic survival, migration, and invasion. By utilizing a tri-modal in silico approach for target identification, Cysteine-Rich Protein 2 (CRIP2; a transcription factor) was identified as a direct target of miR-449a, corroborated using qRT-PCR, Western blot, and luciferase reporter assays. MDA-MB-231 cells stably transfected with CRIP2 demonstrated a significant reduction in cell viability, migration, and invasion, as well as decreased tumor growth and angiogenesis in mouse xenograft models. Our data revealed that overexpression of miR-449a suppresses CRIP2, which then affects the tumor vasculature, likely via NF-κB/p65 complex-mediated transcription of VEGF. These finding define an oncogenic function of miR-449a in human breast cancer, and highlight the importance of this pathway in driving aggressive behaviour.
Collapse
Affiliation(s)
- Wei Shi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Jeff Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Matthew Lee
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Shijun Yue
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Matthew Rowe
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Melania Pintilie
- Division of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ryunosuke Kogo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | - Anthony Fyles
- Department of Radiation Oncology, Princess Margaret Hospital, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Kenneth W Yip
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Fei-Fei Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Radiation Oncology, Princess Margaret Hospital, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|