1
|
Zhang L, Hu H, Cai W, Chen S, Sheng P, Fu X. CaCO 3-complexed pH-responsive nanoparticles encapsulating mitoxantrone and celastrol enhance tumor chemoimmunotherapy. Int J Pharm 2024; 667:124860. [PMID: 39461678 DOI: 10.1016/j.ijpharm.2024.124860] [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: 08/01/2024] [Revised: 09/26/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Modulating the immunosuppressive tumor microenvironment (TME) while enhancing antitumor immune responses is a promising strategy. In this study, we designed an acid-sensitive nanosystem (MCCaNPs) to demonstrate effective immunotherapy against cancer through the systemic delivery of immune-stimulating chemotherapy combinations. A pH-responsive nanoplatform containing CaCO3 was prepared by the double emulsion method, and mitoxantrone (MIT) and celastrol (CEL) were simultaneously encapsulated as immunogenic cell death (ICD) inducers. Due to the acid responsiveness of CaCO3, the nanoparticles rapidly consume H+ to relieve the acidic tumor microenvironment and explosively release CEL and MIT, showing inherent immunomodulatory activity in collaborative tumor chemoimmunotherapy. MIT and CEL synergistically trigger stronger ICD by inducing tumor cells to release calreticulin (CRT), high mobility group box 1 protein (HMGB1). Following the intravenous administration of MCCaNPs, the local tumor microenvironment(TME) was reprogrammed in mice-bearing tumors. This reprogramming was characterized by a significant increase in the density of tumor-infiltrating cytotoxic T lymphocytes(CTLs), ultimately prolonging survival. Therefore, this research proposes a promising approach to trigger immunogenic cell death collaboratively, aiming to boost the tumor CTLs infiltration for anticancer immunotherapy.
Collapse
Affiliation(s)
- Liang Zhang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Huiqiang Hu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Wan Cai
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Shungen Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| | - Ping Sheng
- College of Life and Health Sciences, Anhui Science and Technology University, Fengyang 233100, China.
| | - Xiaomei Fu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; FAN Cuisheng Studio of National Famous TCM, Nanchang 330006, China.
| |
Collapse
|
2
|
Zheng H, Jian L, Li L, Liu W, Chen W. Prior Clinico-Radiological Features Informed Multi-Modal MR Images Convolution Neural Network: A novel deep learning framework for prediction of lymphovascular invasion in breast cancer. Cancer Med 2024; 13:e6932. [PMID: 38230837 PMCID: PMC10905682 DOI: 10.1002/cam4.6932] [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: 10/18/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Current methods utilizing preoperative magnetic resonance imaging (MRI)-based radiomics for assessing lymphovascular invasion (LVI) in patients with early-stage breast cancer lack precision, limiting the options for surgical planning. PURPOSE This study aimed to develop a sophisticated deep learning framework called "Prior Clinico-Radiological Features Informed Multi-Modal MR Images Convolutional Neural Network (PCMM-Net)" to improve the accuracy of LVI prediction in breast cancer. By incorporating multiparameter MRI and prior clinical knowledge, PCMM-Net should enhance the precision of LVI assessment. METHODS A total of 341 patients with breast cancer were randomly divided into training and validation groups at a ratio of 7:3. Imaging features were extracted from T1-weighted, T2-weighted, and contrast-enhanced T1-weighted MRI sequences. Stepwise univariate and multivariate logistic regression were employed to establish a clinico-radiological model for LVI prediction. The radiomics model was built using redundancy and the least absolute shrinkage and selection operator. Then, two deep learning frameworks were developed: the Multi-Modal MR Images Convolutional Neural Network (MM-Net), which does not consider prior radiological features, and PCMM-Net, which incorporates multiparameter MRI and prior clinical knowledge. Receiver operating characteristic curves were used, and the corresponding areas under the curves (AUCs) were calculated for evaluation. RESULTS PCMM-Net achieved the highest AUC of 0.843. The clinico-radiological features displayed the lowest AUC value of 0.743, followed by MM-Net with an AUC of 0.774, and radiomics with an AUC of 0.795. CONCLUSIONS This study introduces PCMM-Net, an innovative deep learning framework that integrates prior clinico-radiological features for accurate LVI prediction in breast cancer. PCMM-Net demonstrates excellent diagnostic performance and facilitates the application of precision medicine.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Lian Jian
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Li Li
- Department of RadiologyHunan Children's HospitalChangshaHunanChina
| | - Wen Liu
- Department of RadiologyThe Third Xiang Ya HospitalCentral South UniversityChangshaHunanChina
| | - Wei Chen
- Department of RadiologyThe Second People's Hospital of Hunan Province, Brain Hospital of Hunan ProvinceChangshaHunanChina
| |
Collapse
|
3
|
Zhong X, Lin Y, Zhang W, Bi Q. Predicting diagnosis and survival of bone metastasis in breast cancer using machine learning. Sci Rep 2023; 13:18301. [PMID: 37880320 PMCID: PMC10600146 DOI: 10.1038/s41598-023-45438-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
This study aimed at establishing more accurate predictive models based on novel machine learning algorithms, with the overarching goal of providing clinicians with effective decision-making assistance. We retrospectively analyzed the breast cancer patients recorded in the Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2016. Multivariable logistic regression analyses were used to identify risk factors for bone metastases in breast cancer, whereas Cox proportional hazards regression analyses were used to identify prognostic factors for breast cancer with bone metastasis (BCBM). Based on the identified risk and prognostic factors, we developed diagnostic and prognostic models that incorporate six machine learning classifiers. We then used the area under the receiver operating characteristic (ROC) curve (AUC), learning curve, precision curve, calibration plot, and decision curve analysis to evaluate performance of the machine learning models. Univariable and multivariable logistic regression analyses showed that bone metastases were significantly associated with age, race, sex, grade, T stage, N stage, surgery, radiotherapy, chemotherapy, tumor size, brain metastasis, liver metastasis, lung metastasis, breast subtype, and PR. Univariate and multivariate Cox regression analyses revealed that age, race, marital status, grade, surgery, radiotherapy, chemotherapy, brain metastasis, liver metastasis, lung metastasis, breast subtype, ER, and PR were closely associated with the prognosis of BCBM. Among the six machine learning models, the XGBoost algorithm predicted the most accurate results (Diagnostic model AUC = 0.98; Prognostic model AUC = 0.88). According to the Shapley additive explanations (SHAP), the most critical feature of the diagnostic model was surgery, followed by N stage. Interestingly, surgery was also the most critical feature of prognostic model, followed by liver metastasis. Based on the XGBoost algorithm, we could effectively predict the diagnosis and survival of bone metastasis in breast cancer and provide targeted references for the treatment of BCBM patients.
Collapse
Affiliation(s)
- Xugang Zhong
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Yanze Lin
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Wei Zhang
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China.
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People's Republic of China.
| | - Qing Bi
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China.
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China.
| |
Collapse
|
4
|
Momeni M, Shamloo A, Hasani-Gangaraj M, Dezhkam R. An experimental study of centrifugal microfluidic platforms for magnetic-inertial separation of circulating tumor cells using contraction-expansion and zigzag arrays. J Chromatogr A 2023; 1706:464249. [PMID: 37531849 DOI: 10.1016/j.chroma.2023.464249] [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: 05/30/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023]
Abstract
Cancer diagnosis has recently been at the forefront of recent medical research, with ongoing efforts to develop devices and technologies for detecting cancer in patients. One promising approach for cancer diagnosis is the detection of Circulating Tumor Cells (CTCs) in blood samples. Separating these rare cells from the diverse background of blood cells and analyzing them can provide valuable insights into the disease's stage and lethality. Here we present the design and fabrication of a centrifugal microfluidic platform on a polymeric disk that utilizes centrifugal forces for cell isolation. The separation units exploit both active and passive methods. In other words, in addition to introducing novel geometry for channels, an external magnetic field is also employed to separate the target cells from the background cells. In order for the external field to function, the CTCs must first be labeled with antibody-conjugated nanoparticles; the separation process should be then performed. Before the experimental tests, a numerical study was done to determine the optimum parameters; the angular velocity and magnetization investigations showed that 2000 rpm and 868,000 (kA/m) are the optimum conditions for the designed device to reach the efficiency of 100% for both White Blood Cells (WBCs) and CTCs. These results indicate that the passive region of the channels primarily contributes to the focusing of the target cells, and showed that the focusing effect is more pronounced in the expansion-contraction geometry compared to the zigzag geometry. Additionally, the results proved that curved channel geometries performed better than straight ones in terms of separation efficiency. However, if the separation relies solely on channel geometry, the majority of cells would be directed towards the non-target chamber, leading to suboptimal results. This is due to the direction of the forces acting on the cells. However, including an external magnetic field improves the direction of the net force and enhances the separation efficiency. Finally, the numerical and experimental results of the study were compared, and the curved expansion-contraction channel is introduced as the best geometry having 100% and ∼92% CTC separation efficiency, respectively.
Collapse
Affiliation(s)
- Maede Momeni
- Department of Mechanical Engineering Sharif University of Technology Tehran, Iran
| | - Amir Shamloo
- Department of Mechanical Engineering Sharif University of Technology Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran.
| | | | - Rasool Dezhkam
- Department of Mechanical Engineering Sharif University of Technology Tehran, Iran; Stem Cell and Regenerative Medicine Center, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
5
|
Li J, Goh ELK, He J, Li Y, Fan Z, Yu Z, Yuan P, Liu DX. Emerging Intrinsic Therapeutic Targets for Metastatic Breast Cancer. BIOLOGY 2023; 12:697. [PMID: 37237509 PMCID: PMC10215321 DOI: 10.3390/biology12050697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Breast cancer is now the most common cancer worldwide, and it is also the main cause of cancer-related death in women. Survival rates for female breast cancer have significantly improved due to early diagnosis and better treatment. Nevertheless, for patients with advanced or metastatic breast cancer, the survival rate is still low, reflecting a need for the development of new therapies. Mechanistic insights into metastatic breast cancer have provided excellent opportunities for developing novel therapeutic strategies. Although high-throughput approaches have identified several therapeutic targets in metastatic disease, some subtypes such as triple-negative breast cancer do not yet have an apparent tumor-specific receptor or pathway to target. Therefore, exploring new druggable targets in metastatic disease is a high clinical priority. In this review, we summarize the emerging intrinsic therapeutic targets for metastatic breast cancer, including cyclin D-dependent kinases CDK4 and CDK6, the PI3K/AKT/mTOR pathway, the insulin/IGF1R pathway, the EGFR/HER family, the JAK/STAT pathway, poly(ADP-ribose) polymerases (PARP), TROP-2, Src kinases, histone modification enzymes, activated growth factor receptors, androgen receptors, breast cancer stem cells, matrix metalloproteinases, and immune checkpoint proteins. We also review the latest development in breast cancer immunotherapy. Drugs that target these molecules/pathways are either already FDA-approved or currently being tested in clinical trials.
Collapse
Affiliation(s)
- Jiawei Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Eyleen L. K. Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Ji He
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Yan Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Zhimin Fan
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan 250033, China;
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| |
Collapse
|
6
|
Han TY, Hou LS, Li JX, Huan ML, Zhou SY, Zhang BL. Bone targeted miRNA delivery system for miR-34a with enhanced anti-tumor efficacy to bone-associated metastatic breast cancer. Int J Pharm 2023; 635:122755. [PMID: 36801480 DOI: 10.1016/j.ijpharm.2023.122755] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Bone metastatic cancer is the most common occurrence in breast cancer, and the treatment is also facing great challenges. MicroRNA-34a (miRNA-34a) is a promising anti-cancer miRNA for gene therapy to bone metastatic cancer patients. However, the lack of specificity to bone and low accumulation at the site of bone tumor remains the major challenge when used bone-associated tumor. To solve this problem, a bone-targeted vector for delivery of miR-34a to bone metastatic breast cancer was constructed by using the commonly used gene vector branched polyethylenimine 25 k (BPEI 25 k) as the skeleton and linking with alendronate (ALN) moieties for bone targeting group. The constructed gene delivery system PCA/miR-34a can efficiently prevent miR-34a from degradation during blood circulation and enhance the specific bone delivery and distribution. PCA/miR-34a nanoparticles can be uptake into tumor cells through clathrin and caveolae-mediated endocytosis, and directly regulate the expression of oncogenes, thus promoting tumor cell apoptosis and relieving bone tissue erosion. The results of experiments in vitro and in vivo confirmed that the constructed bone-targeted miRNA delivery system PCA/miR-34a can enhance the anti-tumor efficacy in bone metastatic cancer, and provide a potential strategy for gene therapy in bone metastatic cancer.
Collapse
Affiliation(s)
- Tian-Yan Han
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Li-Shuang Hou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Jia-Xin Li
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Meng-Lei Huan
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Si-Yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Pharmacology of the State Administration of Traditional Chinese Medicine, Xi'an 710032, China.
| |
Collapse
|
7
|
La Verde N, Damia G, Garrone O, Santini D, Fabi A, Ciccarese M, Generali DG, Nunzi M, Poletto E, Ferraris E, Cretella E, Scandurra G, Meattini I, Bertolini AS, Cavanna L, Collovà E, Romagnoli E, Rulli E, Legramandi L, Guffanti F, Bramati A, Moretti A, Cassano A, Vici P, Torri V, Farina G. Tolerability of Eribulin and correlation between polymorphisms and neuropathy in an unselected population of female patients with metastatic breast cancer: results of the multicenter, single arm, phase IV PAINTER study. Breast Cancer Res 2022; 24:71. [PMID: 36307826 PMCID: PMC9615373 DOI: 10.1186/s13058-022-01560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metastatic breast cancer (MBC) is an incurable disease and its treatment focuses on prolonging patients' (pts) overall survival (OS) and improving their quality of life. Eribulin is a microtubule inhibitor that increases OS in pre-treated MBC pts. The most common adverse events (AEs) are asthenia, neutropenia and peripheral neuropathy (PN). METHODS PAINTER is a single arm, phase IV study, aimed at evaluating the tolerability of eribulin in MBC pts. Secondary objectives were the description of treatment efficacy and safety, the assessment of the incidence and severity of PN and its association with genetic polymorphisms. Genomic DNA was isolated from blood samples and 15 Single Nucleotide Polymorphisms (SNPs) were genotyped by Taqman specific assays. The association between PN and SNPs were evaluated by Fisher exact test. RESULTS Starting from May 2014 until June 2018 180 pts were enrolled in this study by 20 Italian centers. 170 of these pts could be evaluated for efficacy and toxicity and 159 for polymorphisms analysis. The median age of pts was 60 years old and the biological subtypes were luminal type (64.7%), Her2 positive (18.3%) and triple negative (17%). Pts were pretreated with a median of 5 lines for MBC. The median follow up of this study was 15.4 months with a median number of 4.5 cycles administered (minimum-maximum 1-23). The median overall survival was 12 months. 48.8% of pts experienced a dose reduction, mainly for neutropenia (23.9%) and liver toxicity (12%). 65 pts (38.2%) reported at least one severe toxicity. Neutropenia and neurotoxicity were the most frequent severe AEs (15.3% and 14.7%, respectively); other reported toxicities were osteo-muscular, abdominal or tumor site pain (19.4%), liver toxicity (6.6%), pulmonary toxicity (6.5%) and dermatological toxicity (3.6%). Among the 15 evaluated SNPs, an association with PN was found for rs2233335 and rs7214723. CONCLUSIONS Eribulin is a well-tolerated treatment option in MBC. Schedule and dosage modifications were common, but toxicity rarely led to treatment discontinuation. SNPs rs2233335 (G/T and T/T) in the NDRG1 gene and rs7214723 (CC and CT) in the CAMKK1 gene were associated with PN. These findings, if validated, could allow a tailored treatment with eribulin in cancer patients. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT02864030.
Collapse
Affiliation(s)
- Nicla La Verde
- Department of Oncology, Luigi Sacco Hospital, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ornella Garrone
- Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Santini
- Oncologia Medica Università Campus Biomedico, Rome, Italy
- UOC Oncologia Universitaria, Sapienza University of Rome Polo Pontino, Latina, Italy
| | - Alessandra Fabi
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | | | - Daniele Giulio Generali
- U.O. Multidisciplinare Di Patologia Mammaria E Ricerca, Traslazionale Azienda Socio-Sanitaria Territoriale Di Cremona, Cremona, Italy
| | - Martina Nunzi
- Dept. of Oncology Medical and Translational Oncology Unit, Azienda Ospedaliera S.Maria, Terni, Italy
| | - Elena Poletto
- ASUFC Presidio Ospedaliero Universitario S.M. Misericordia, Udine, Italy
| | - Elisa Ferraris
- Division of Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy
| | | | | | - Icro Meattini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | | | - Luigi Cavanna
- Oncology Haematology Department, ASL Piacenza, Piacenza, Italy
| | - Elena Collovà
- ASST Ovest Milanese, Ospedale Di Legnano, Legnano, MI, Italy
| | | | - Eliana Rulli
- Laboratory of Methodology for Clinical Research, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lorenzo Legramandi
- Laboratory of Methodology for Clinical Research, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Federica Guffanti
- Laboratory of Molecular Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Annalisa Bramati
- Department of Oncology, Fatebenefratelli Hospital, ASST Fatebenefratelli-Sacco, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| | - Anna Moretti
- Department of Oncology, Fatebenefratelli Hospital, ASST Fatebenefratelli-Sacco, Piazza Principessa Clotilde 3, 20121, Milan, Italy.
| | - Alessandra Cassano
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Patrizia Vici
- Phase IV Clinical Studies Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Valter Torri
- Department of Oncology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gabriella Farina
- Department of Oncology, Fatebenefratelli Hospital, ASST Fatebenefratelli-Sacco, Piazza Principessa Clotilde 3, 20121, Milan, Italy
| |
Collapse
|
8
|
Fan TD, Bei DK, Li SW. Nomogram Models Based on the Gene Expression in Prediction of Breast Cancer Bone Metastasis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8431946. [PMID: 36046013 PMCID: PMC9424032 DOI: 10.1155/2022/8431946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/01/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022]
Abstract
Objective The aim of this study is to design a weighted co-expression network and build gene expression signature-based nomogram (GESBN) models for predicting the likelihood of bone metastasis in breast cancer (BC) patients. Methods Dataset GSE124647 was used as a training set, while GSE16446, GSE45255, and GSE14020 were taken as validation sets. In the training cohort, the limma package in R was adopted to obtain differentially expressed genes (DEGs) between BC nonbone metastasis and bone metastasis patients, which were used for functional enrichment analysis. After weighted co-expression network analysis (WGCNA), univariate Cox regression and Kaplan-Meier plotter analyses were performed to screen potential prognosis-related genes. Then, GESBN models were constructed and evaluated. The prognostic value of the GESBN models was investigated in the GSE124647 dataset, which was validated in GSE16446 and GSE45255 datasets. Further, the expression levels of genes in the models were explored in the training set, which was validated in GSE14020. Finally, the expression and prognostic value of hub genes in BC were explored. Results A total of 1858 DEGs were obtained. The WGCNA result showed that the blue module was most significantly related to bone metastasis and prognosis. After survival analyses, GAJ1, SLC24A3, ITGBL1, and SLC44A1 were subjected to construct a GESBN model for overall survival (OS). While GJA1, IGFBP6, MDFI, TGFBI, ANXA2, and SLC24A3 were subjected to build a GESBN model for progression-free survival (PFS). Kaplan-Meier plotter and receiver operating characteristic analyses presented the reliable prediction ability of the models. Cox regression analysis further revealed that GESBN models were independent prognostic predictors for OS and PFS in BC patients. Besides, GJA1, IGFBP6, ITGBL1, SLC44A1, and TGFBI expressions were significantly different between the two groups in GSE124647 and GSE14020. The hub genes had a significant impact on patient prognosis. Conclusion Both the four-gene signature and six-gene signature could accurately predict patient prognosis, which may provide novel treatment insights for BC bone metastasis.
Collapse
Affiliation(s)
- Teng-di Fan
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, Ningbo 315040, Zhejiang, China
| | - Di-kai Bei
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, Ningbo 315040, Zhejiang, China
| | - Song-wei Li
- Department of Orthopedics, Ningbo Medical Center Lihuili Hospital, Ningbo 315040, Zhejiang, China
| |
Collapse
|
9
|
Liu Q, Qiu J, Lu Q, Ma Y, Fang S, Bu B, Song L. Comparison of endocrine therapy and chemotherapy as different systemic treatment modes for metastatic luminal HER2-negative breast cancer patients —A retrospective study. Front Oncol 2022; 12:873570. [PMID: 35957911 PMCID: PMC9360505 DOI: 10.3389/fonc.2022.873570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThe purpose of this study was to evaluate endocrine therapy and chemotherapy for first-line, maintenance, and second-line treatment of hormone receptor-positive HER-2-negative metastatic breast cancer (HR+HER-2-MBC) and the relationship between different treatment options and survival.Patients and methodsThe patients included in this study were all diagnosed with metastatic breast cancer (MBC) at Shandong Cancer Hospital from January 2013 to June 2017. Of the 951 patients with MBC, 307 patients with HR+HER-2-MBC were included in the analysis. The progression-free survival (PFS) and overall survival (OS) of the various treatment modes were evaluated using Kaplan–Meier analysis and the log-rank test. Because of the imbalance in data, we used the synthetic minority oversampling technique (SMOTE) algorithm to oversample the data to increase the balanced amount of data.ResultsThis retrospective study included 307 patients with HR+HER-2-MBC; 246 patients (80.13%) and 61 patients (19.87%) were treated with first-line chemotherapy and first-line endocrine therapy, respectively. First-line endocrine therapy was better than first-line chemotherapy in terms of PFS and OS. After adjusting for known prognostic factors, patients receiving first-line chemotherapy had poorer PFS and OS outcomes than patients receiving first-line endocrine therapy. In terms of maintenance treatment, the endocrine therapy-endocrine therapy maintenance mode achieved the best prognosis, followed by the chemotherapy-endocrine therapy maintenance mode and chemotherapy-chemotherapy maintenance mode, and the no-maintenance mode has resulted in the worst prognosis. In terms of first-line/second-line treatment, the endocrine therapy/endocrine therapy mode achieved the best prognosis, while the chemotherapy/chemotherapy mode resulted in the worst prognosis. The chemotherapy/endocrine therapy mode achieved a better prognosis than the endocrine therapy/chemotherapy mode. There were no significant differences in the KI-67 index (<15%/15-30%/≥30%) among the patients receiving first-line treatment modes, maintenance treatment modes, and first-line/second-line treatment modes. There was no statistical evidence in this study to support that the KI-67 index affected survival. However, in the first-line/second-line model, after SMOTE, we could see that KI-67 ≥ 30% had a poor prognosis.ConclusionsDifferent treatment modes for HR+HER-2-MBC were analyzed. Endocrine therapy achieved better PFS and OS outcomes than chemotherapy. Endocrine therapy should be the first choice for first-line, maintenance, and second-line treatment of HR+HER-2-MBC.
Collapse
Affiliation(s)
- Qiuyue Liu
- Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Juan Qiu
- Oncology Department, The Fourth People’s Hospital of Jinan, Jinan, China
| | - Qianrun Lu
- Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Yujin Ma
- Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Shu Fang
- Department of Breast Medicine, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Bing Bu
- Department of Breast Medicine, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Lihua Song
- Department of Breast Medicine, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
- *Correspondence: Lihua Song,
| |
Collapse
|
10
|
Xu J, Ma L, Wang D, Yang J. Uncarboxylated osteocalcin promotes proliferation and metastasis of MDA-MB-231 cells through TGF-β/SMAD3 signaling pathway. BMC Mol Cell Biol 2022; 23:18. [PMID: 35413833 PMCID: PMC9003967 DOI: 10.1186/s12860-022-00416-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/23/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most severe type of breast cancer owing to its high heterogeneity, aggressiveness and lack of treatment. Studies have reported that uncarboxylated osteocalcin (GluOC) promotes the development of prostate and other cancers. Studies have also found elevated levels of serum osteocalcin in breast cancer patients with bone metastasis, and serum osteocalcin can be a marker of bone metastasis. However, whether GluOC promotes the development of TNBC and the related mechanisms need to be further clarified. RESULTS Our results revealed that GluOC is associated with the proliferation and metastasis of MDA-MB-231 cells. GluOC increased the viability and proliferation of MDA-MB-231 cells. In addition, GluOC enhanced the metastatic ability of MDA-MB-231 cells by promoting the expression of matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-13 (MMP13), and vascular endothelial growth factor (VEGF) and inducing epithelial-mesenchymal transition (EMT). We also found that GluOC upregulated the expression of interleukin-8 (IL-8) and parathyroid hormone-related protein (PTHrP) genes in MDA-MB-231 breast cancer cells. Moreover, the promoting effect of GluOC was reversed in MDA-MB-231 breast cancer cells treated with specific inhibitor of SMAD3 (SIS3), a SMAD3 phosphorylation inhibitor. CONCLUSION Our research proved for the first time that GluOC facilitates the proliferation and metastasis of MDA-MB-231 cells by accelerating the transforming growth factor-β (TGF-β)/SMAD3 signaling pathway. Moreover, GluOC also promotes the gene expression of IL-8 and PTHrP. Both IL-8 and PTHrP can act as osteolytic factors in breast cancer cells. This study indicates that GluOC may be a useful target for preventing TNBC bone metastasis.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Medical School, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, People's Republic of China
| | - Luyao Ma
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Danqing Wang
- Medical School, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, People's Republic of China
| | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, People's Republic of China.
| |
Collapse
|
11
|
Feng Y, Zhang Y, Xiang Y, Guo K, Jin H, Ruan S, Guan Z. Nomograms for Predicting Specific Distant Metastatic Sites and Overall Survival of Breast Invasive Ductal Carcinoma Patients After Surgery: A Large Population-Based Study. Front Surg 2022; 9:779220. [PMID: 35402478 PMCID: PMC8986992 DOI: 10.3389/fsurg.2022.779220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 02/25/2022] [Indexed: 11/14/2022] Open
Abstract
Background Breast cancer (BC) has become the most common malignancy worldwide, accounting for 11.7% of newly diagnosed cancer cases last year. Invasive ductal carcinoma (IDC) is the most common pathological type of BC. However, there were few studies to predict distant metastatic sites and overall survival (OS) of IDC patients. Methods Post-operative IDC patients from 2010 to 2016 in the Surveillance, Epidemiology, and End Results (SEER) database were reviewed. Nomograms were developed to predict the specific distant metastatic sites and OS of IDC patients. The performance of nomograms was evaluated with the calibration curves, area under the curve (AUC), and decision curve analysis (DCA). Kaplan-Meier analysis and log-rank tests were used to estimate the survival times of IDC patients with distant metastases. Results A total of 171,967 post-operative IDC patients were enrolled in our study. Univariate and multivariate analyses were used to establish the nomograms of significant variables. The AUC of the nomograms for the prediction of liver, lung, bone, and brain metastases was 0.903, 0.877, 0.863, and 0.811, respectively. In addition, the AUC of the nomogram for the prediction of 1-, 3-, and 5-year OS was 0.809, 0.813, 0.787, respectively. Calibration curves and DCA showed good consistency and clinical benefits, respectively. Conclusions We constructed new predictive models for liver, lung, brain, bone metastases and 1-, 3-, and 5-year OS in IDC patients. These can help clinicians to individualize the treatment of IDC patients, so that patients can get the more appropriate treatment options.
Collapse
Affiliation(s)
- Yuqian Feng
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiting Zhang
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuying Xiang
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kaibo Guo
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huimin Jin
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Shanming Ruan
| | - Zhuoya Guan
- Department of Mammary Gland, Medical Community of Taizhou Luqiao Traditional Chinese Medicine Hospital, Taizhou, China
- *Correspondence: Zhuoya Guan
| |
Collapse
|
12
|
Kitayama K, Kawamoto T, Kawakami Y, Hara H, Takemori T, Fujiwara S, Yahiro S, Miyamoto T, Mifune Y, Hoshino Y, Kakutani K, Matsumoto T, Matsushita T, Niikura T, Kuroda R, Akisue T. Regulatory roles of miRNAs 16, 133a, and 223 on osteoclastic bone destruction caused by breast cancer metastasis. Int J Oncol 2021; 59:97. [PMID: 34713296 PMCID: PMC8562387 DOI: 10.3892/ijo.2021.5277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022] Open
Abstract
Osteolytic bone metastasis leads to skeletal-related events, resulting in a decline in the patient activities and survival; therefore, it is important to understand the mechanism underlying bone metastasis. Recent studies have suggested that microRNAs (miRNAs or miRs) are involved in osteoclast differentiation and/or osteolytic bone metastasis; however, the roles of miRNAs have not been elucidated. In the present study, the roles of miRNAs in bone destruction caused by breast cancer metastasis were investigated in vitro and in vivo. miR-16, miR-133a and miR-223 were transfected into a human breast cancer cell line, MDA-MB-231. The expression of osteolytic factors in conditioned medium (miR-CM) collected from the culture of transfected cells was assessed. To evaluate the effects of miRNAs on osteoclast differentiation and activities, tartrate-resistant acid phosphatase (TRAP) staining and bone resorptive assays were performed in osteoclasts following miR-CM treatment. To create in vivo bone metastasis models for histological and morphometric evaluation, miRNA-transfected MDA-MB-231 cells were transplanted into the proximal tibia of nude mice. Expression of osteolytic factors, including receptor activator for nuclear factor-κB ligand (RANKL), interleukin (IL)-1β, IL-6, parathyroid hormone-related protein (PTHrP), and tumor necrosis factor (TNF), was increased in miR-16-CM, whereas it was decreased in both miR-133a-CM and miR-223-CM. TRAP staining and bone resorptive assays revealed that osteoclast function and activities were promoted by miR-16-CM treatment, whereas they were suppressed by miR-133a-CM and miR-223-CM. Consistent with in vitro findings, in vivo experiments revealed that the overexpression of miR-16 increased osteoclast activities and bone destruction in MDA-MB-231 cells, whereas the opposite results were observed in both miR-133a- and miR-223-transfected MDA-MB-231 cells. Our results indicated that miR-16 promoted osteoclast activities and bone destruction caused by breast cancer metastasis in the bone microenvironment, whereas miR-133a and miR-223 suppressed them. These miRNAs could be potential biomarkers and therapeutic targets for breast cancer bone metastasis.
Collapse
Affiliation(s)
- Kazumichi Kitayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Teruya Kawamoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Yohei Kawakami
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Hitomi Hara
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Toshiyuki Takemori
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Shuichi Fujiwara
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Shunsuke Yahiro
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Tomohiro Miyamoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Yutaka Mifune
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Yuichi Hoshino
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Kenichiro Kakutani
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| | - Toshihiro Akisue
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650‑0017, Japan
| |
Collapse
|
13
|
Lopes C, Piairo P, Chícharo A, Abalde-Cela S, Pires LR, Corredeira P, Alves P, Muinelo-Romay L, Costa L, Diéguez L. HER2 Expression in Circulating Tumour Cells Isolated from Metastatic Breast Cancer Patients Using a Size-Based Microfluidic Device. Cancers (Basel) 2021; 13:4446. [PMID: 34503260 PMCID: PMC8431641 DOI: 10.3390/cancers13174446] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
HER2 is a prognostic and predictive biomarker in breast cancer, normally assessed in tumour biopsy and used to guide treatment choices. Circulating tumour cells (CTCs) escape the primary tumour and enter the bloodstream, exhibiting great metastatic potential and representing a real-time snapshot of the tumour burden. Liquid biopsy offers the unique opportunity for low invasive sampling in cancer patients and holds the potential to provide valuable information for the clinical management of cancer patients. This study assesses the performance of the RUBYchip™, a microfluidic system for CTC capture based on cell size and deformability, and compares it with the only FDA-approved technology for CTC enumeration, CellSearch®. After optimising device performance, 30 whole blood samples from metastatic breast cancer patients were processed with both technologies. The expression of HER2 was assessed in isolated CTCs and compared to tissue biopsy. Results show that the RUBYchipTM was able to isolate CTCs with higher efficiency than CellSearch®, up to 10 times more, averaging all samples. An accurate evaluation of different CTC subpopulations, including HER2+ CTCs, was provided. Liquid biopsy through the use of the RUBYchipTM in the clinic can overcome the limitations of histological testing and evaluate HER2 status in patients in real-time, helping to tailor treatment during disease evolution.
Collapse
Affiliation(s)
- Cláudia Lopes
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Paulina Piairo
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Alexandre Chícharo
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Sara Abalde-Cela
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| | - Liliana R. Pires
- RUBYnanomed Lda, Praça Conde de Agrolongo 123, 4700-312 Braga, Portugal;
| | - Patrícia Corredeira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
| | - Patrícia Alves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago (IDIS), Complejo Hospitalario de Santiago de Compostela, Trav. Choupana s/n, 15706 Santiago de Compostela, Spain;
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, Calle de Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Luís Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (P.C.); (P.A.); (L.C.)
- Oncology Division, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Av Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal; (C.L.); (A.C.); (S.A.-C.)
| |
Collapse
|
14
|
Tanni KA, Truong CB, Johnson BS, Qian J. Comparative effectiveness and safety of eribulin in advanced or metastatic breast cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 163:103375. [PMID: 34087344 DOI: 10.1016/j.critrevonc.2021.103375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/08/2021] [Accepted: 05/29/2021] [Indexed: 12/24/2022] Open
Abstract
Eribulin is one of the few recommended chemotherapies for locally advanced breast cancer (LABC) or metastatic breast cancer (MBC). We systematically searched MEDLINE Ovid, Cochrane Library, IPA, CINAHL, Web of Science and ProQuest Dissertations for studies evaluating eribulin versus non-eribulin regimens in LABC/MBC till January 15, 2021. Primary effectiveness and safety outcomes were overall survival (OS) and adverse events (AE), respectively. Hazard ratios (HR) and relative risks (RR) with 95 % confidence intervals (CIs) were calculated using fixed or random-effects meta-analyses. Of 1183 publications identified, 13 studies were included in this review. Eribulin based therapy showed significantly increased OS [HR (95 % CI) = 0.77 (0.67-0.88)] compared to non-eribulin in both main and sensitivity analyses, as well as subgroup analyses according to receptor expression and line of therapy. Incidence of all-grade neutropenia was the only significant AE in eribulin than non-eribulin groups. Eribulin has a manageable toxicity profile and provides significant survival benefit in LABC/MBC patients.
Collapse
Affiliation(s)
- Kaniz Afroz Tanni
- Department of Health Outcomes Research and Policy, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA.
| | - Cong Bang Truong
- Department of Health Outcomes Research and Policy, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA.
| | - Brandon S Johnson
- Spencer Cancer Center, East Alabama Medical Center, Opelika, AL 36801, USA.
| | - Jingjing Qian
- Department of Health Outcomes Research and Policy, Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA.
| |
Collapse
|
15
|
Zhang Q, Wang W, Shen H, Tao H, Wu Y, Ma L, Yang G, Chang R, Wang J, Zhang H, Wang C, Zhang F, Qi J, Mi C. Low-Intensity Focused Ultrasound-Augmented Multifunctional Nanoparticles for Integrating Ultrasound Imaging and Synergistic Therapy of Metastatic Breast Cancer. NANOSCALE RESEARCH LETTERS 2021; 16:73. [PMID: 33928450 PMCID: PMC8085141 DOI: 10.1186/s11671-021-03532-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/19/2021] [Indexed: 05/06/2023]
Abstract
The metastasis of breast cancer is believed to have a negative effect on its prognosis. Benefiting from the remarkable deep-penetrating and noninvasive characteristics, sonodynamic therapy (SDT) demonstrates a whole series of potential leading to cancer treatment. To relieve the limitation of monotherapy, a multifunctional nanoplatform has been explored to realize the synergistic treatment efficiency. Herein, we establish a novel multifunctional nano-system which encapsulates chlorin e6 (Ce6, for SDT), perfluoropentane (PFP, for ultrasound imaging), and docetaxel (DTX, for chemotherapy) in a well-designed PLGA core-shell structure. The synergistic Ce6/PFP/DTX/PLGA nanoparticles (CPDP NPs) featured with excellent biocompatibility and stability primarily enable its further application. Upon low-intensity focused ultrasound (LIFU) irradiation, the enhanced ultrasound imaging could be revealed both in vitro and in vivo. More importantly, combined with LIFU, the nanoparticles exhibit intriguing antitumor capability through Ce6-induced cytotoxic reactive oxygen species as well as DTX releasing to generate a concerted therapeutic efficiency. Furthermore, this treating strategy actives a strong anti-metastasis capability by which lung metastatic nodules have been significantly reduced. The results indicate that the SDT-oriented nanoplatform combined with chemotherapy could be provided as a promising approach in elevating effective synergistic therapy and suppressing lung metastasis of breast cancer.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Wen Wang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Hongyuan Shen
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Hongyu Tao
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Yating Wu
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Liyuan Ma
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Guangfei Yang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ruijiao Chang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jiaxing Wang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Hanfei Zhang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Chenyu Wang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Furong Zhang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Jiaojiao Qi
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Chengrong Mi
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
16
|
Alzain AA, Brisson L, Delaye PO, Pénichon M, Chadet S, Besson P, Chevalier S, Allouchi H, Mohamed MA, Roger S, Enguehard-Gueiffier C. Bioinspired imidazo[1,2-a:4,5-c']dipyridines with dual antiproliferative and anti-migrative properties in human cancer cells: The SAR investigation. Eur J Med Chem 2021; 218:113258. [PMID: 33813152 DOI: 10.1016/j.ejmech.2021.113258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 12/24/2022]
Abstract
Herein, we report the design, synthesis and evaluation of novel bioinspired imidazo[1,2-a:4,5c']dipyridines. The structural optimization identified four anti-proliferative compounds. Compounds 11, 18, 19 and 20 exhibited excellent anticancer activities in vitro with IC50 of 0.4-5 μM against three human cancer cell lines (MDA-MB-468, MDA-MB-435s and MDA-MB-231). These four compounds induced apoptosis in MDA-MB-231 cells in a dose-dependent manner, targeting different apoptotic proteins expression: 11 increased the expression of pro-apoptotic Bax protein while 18-20 reduced the level of anti-apoptotic Bcl-2 protein. Compounds 18 and 19 also reduced MDA-MB-231 cells proliferation as measured by Ki-67 staining. Furthermore, compounds were also tested for the ability to inhibit cell migration in the highly aggressive human MDA-MB-435s cell line. Six compounds of this series (8, 15, 18, 22, 23, 24) inhibited cell migration by 41-50% while four compounds (20, 25, 27, 30) inhibited the migration by 53-62% in wound-healing experiments. Interestingly, compound 20 presented both antiproliferative and anti-migration activities and might be a promising anti-metastatic agent for cancer treatment.
Collapse
Affiliation(s)
- Abdulrahim A Alzain
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France; University of Gezira, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, P.O box 20, Gezira, Sudan
| | - Lucie Brisson
- University of Tours, INSERM, UMR 1069 N2C, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Pierre-Olivier Delaye
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Mélanie Pénichon
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Stéphanie Chadet
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Pierre Besson
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Stéphan Chevalier
- University of Tours, INSERM, UMR 1069 N2C, 10 boulevard Tonnellé, 37032, Tours Cedex, France
| | - Hassan Allouchi
- University of Tours, Faculty of Pharmacy, EA 7502 SIMBA, 31 Avenue Monge, 37200, Tours, France
| | - Magdi A Mohamed
- University of Khartoum, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Khartoum, Sudan; Jouf University, College of Pharmacy, Department of Pharmaceutical Chemistry, Saudi Arabia
| | - Sébastien Roger
- University of Tours, EA 4245 T2I, 10 boulevard Tonnellé, 37032, Tours Cedex, France; Institut Universitaire de France, 75006, Paris, France.
| | | |
Collapse
|
17
|
Peng Y, Xi X, Li J, Ni J, Yang H, Wen C, Wen M. miR-301b and NR3C2 co-regulate cells malignant properties and have the potential to be independent prognostic factors in breast cancer. J Biochem Mol Toxicol 2021; 35:e22650. [PMID: 33063403 DOI: 10.1002/jbt.22650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/19/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
This study intends to address the function of miR-301b/nuclear receptor subfamily 3 group C member 2 (NR3C2) in breast cancer. The Cancer Genome Atlas database was processed to investigate the expression of miR-301b/NR3C2 in breast cancer samples, as well as the relationship between their expression and the prognosis of the patients. Cox regression analysis was performed to determine whether miR-301b/NR3C2 was an independent predictor of the patient's prognosis. Associations between miR-301b and NR3C2 were analyzed by prediction website, dual-luciferase assay, and Pearson correlation coefficient. Quantitative polymerase chain reaction and Western blot analyses were implemented to detect gene expression. The relevant biological characteristics of MCF7 and BCAP-37 cells were tested by cell counting kit-8, colony formation, and transwell assays. Lower expression of NR3C2, which was closely related to the bad prognosis of breast cancer patients, was presented in breast cancer samples and can be used as an independent predictor. miR-301b, as an upstream regulator of NR3C2, was highly expressed in breast cancer samples and can be used as an independent predictor as well. Notably, a higher level of miR-301b and lower level of NR3C2 were related to the reduced overall survival in patients with breast cancer. The proliferative and migratory behaviors of cells were elevated or blocked after overexpression of miR-301b or NR3C2, respectively. However, the above situation was attenuated after together upregulation of miR-301b and NR3C2. The present data afforded evidence that miR-301b may be a tumor-promoting miRNA in breast cancer, and that miR-301b/NR3C2 axis mediated tumor development from cell proliferation and migration.
Collapse
Affiliation(s)
- Yun Peng
- Department of Breast and Thyroid Surgery, People's Hospital of Ganzhou City, Ganzhou, China
| | - Xun Xi
- Department of Breast and Thyroid Surgery, People's Hospital of Ganzhou City, Ganzhou, China
| | - Juntao Li
- Department of Breast and Thyroid Surgery, People's Hospital of Ganzhou City, Ganzhou, China
| | - Jun Ni
- Department of Breast and Thyroid Surgery, People's Hospital of Ganzhou City, Ganzhou, China
| | - Hongbiao Yang
- Department of Anesthesiology, People's Hospital of Ganzhou City, Ganzhou, China
| | - Changyong Wen
- Ganzhou Inteligent Industry Innovation Research Institute, Ganzhou, China
| | - Meiling Wen
- Department of Anesthesiology, People's Hospital of Ganzhou City, Ganzhou, China
| |
Collapse
|
18
|
Kim S, Shin D, Min A, Kim M, Na D, Lee HB, Ryu HS, Yang Y, Woo GU, Lee KH, Lee DW, Kim TY, Lee C, Im SA, Kim JI. Genomic profile of metastatic breast cancer patient-derived xenografts established using percutaneous biopsy. J Transl Med 2021; 19:7. [PMID: 33407601 PMCID: PMC7789010 DOI: 10.1186/s12967-020-02607-2] [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: 05/04/2020] [Accepted: 11/05/2020] [Indexed: 11/16/2022] Open
Abstract
Background Metastatic breast cancer (mBC) is a complex and life-threatening disease and although it is difficult to cure, patients can benefit from sequential anticancer treatment, including endocrine therapy, targeted therapy and cytotoxic chemotherapy. The patient-derived xenograft (PDX) model is suggested as a practical tool to predict the clinical outcome of this disease as well as to screen novel drugs. This study aimed to establish PDX models in Korean patients and analyze their genomic profiles and utility for translational research. Methods Percutaneous core needle biopsy or punch biopsy samples were used for xenotransplantation. Whole exome sequencing and transcriptome analysis were performed to assess the genomic and RNA expression profiles, respectively. Copy number variation and mutational burden were analyzed and compared with other metastatic breast cancer genomic results. Mutational signatures were also analyzed. The antitumor effect of an ATR inhibitor was tested in the relevant PDX model. Results Of the 151 cases studied, 40 (26%) PDX models were established. Notably, the take rate of all subtypes, including the hormone receptor-positive (HR +) subtype, exceeded 20%. The PDX model had genomic fidelity and copy number variation that represented the pattern of its donor sample. TP53, PIK3CA, ESR1, and GATA3 mutations were frequently found in our samples, with TP53 being the most frequently mutated, and the somatic mutations in these genes strengthened their frequency in the PDX model. The ESR1 mutation, CCND1 amplification, and the APOBEC signature were significant features in our HR + HER2- PDX model. Fulvestrant in combination with palbociclib showed a partial response to the relevant patient’s tumor harboring the ESR1 mutation, and CCND1 amplification was found in the PDX model. AZD6738, an ATR inhibitor, delayed tumor growth in a relevant PDX model. Conclusions Our PDX model was established using core needle biopsy samples from primary and metastatic tissues. Genomic profiles of the samples reflected their original tissue characteristics and could be used for the interpretation of clinical outcomes.
Collapse
Affiliation(s)
- Seongyeong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Dongjin Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Minjung Kim
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Deukchae Na
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Han-Byeol Lee
- Department of General Surgery, Seoul National University Hospital, Seoul, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Yaewon Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Translational Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Chungbuk University Hospital, Cheong-Ju, Korea
| | - Go-Un Woo
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Dae-Won Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Charles Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea. .,Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| | - Jong-Il Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea. .,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
19
|
Huang Z, Hu C, Liu K, Yuan L, Li Y, Zhao C, Hu C. Risk factors, prognostic factors, and nomograms for bone metastasis in patients with newly diagnosed infiltrating duct carcinoma of the breast: a population-based study. BMC Cancer 2020; 20:1145. [PMID: 33238981 PMCID: PMC7687803 DOI: 10.1186/s12885-020-07635-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 11/13/2020] [Indexed: 12/29/2022] Open
Abstract
Background Breast cancer is the most common malignancy in women, and it is also the leading cause of death in female patients; the most common pathological type of BC is infiltrating duct carcinoma (IDC). Some nomograms have been developed to predict bone metastasis (BM) in patients with breast cancer. However, there are no studies on diagnostic and prognostic nomograms for BM in newly diagnosed IDC patients. Methods IDC patients with newly diagnosed BM from 2010 to 2016 in the Surveillance, Epidemiology and End Results (SEER) database were reviewed. Multivariate logistic regression analysis was used to identify risk factors for BM in patients with IDC. Univariate and multivariate Cox proportional hazards regression analysis were used to explore the prognostic factors of BM in patients with IDC. We then constructed nomograms to predict the risk and prognosis of BM for patients with IDC. The results were validated using bootstrap resampling and retrospective research on 113 IDC patients with BM from 2015 to 2018 at the Affiliated Hospital of Chengde Medical University. Results This study included 141,959 patients diagnosed with IDC in the SEER database, of whom 2383 cases were IDC patients with BM. The risk factors for BM in patients with IDC included sex, primary site, grade, T stage, N stage, liver metastasis, race, brain metastasis, breast cancer subtype, lung metastasis, insurance status, and marital status. The independent prognostic factors were brain metastases, race, grade, surgery, chemotherapy, age, liver metastases, breast cancer subtype, insurance status, and marital status. Through calibration, receiver operating characteristic curve and decision curve analyses, we found that the nomogram for predicting the prognosis of IDC patients with BM displayed great performance both internally and externally. Conclusion These nomograms are expected to be a precise and personalized tool for predicting the risk and prognosis for BM in patients with IDC. This will help clinicians develop more rational and effective treatment strategies.
Collapse
Affiliation(s)
- Zhangheng Huang
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China
| | - Chuan Hu
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China.,Department of Orthopedic, The Affiliated Hospital of Qingdao University, Shinan District, Qingdao, Shandong Province, China
| | - Kewen Liu
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China
| | - Luolin Yuan
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China
| | - Yinglun Li
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China
| | - Chengliang Zhao
- Department of Spine Surgery, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China.
| | - Chanchan Hu
- Department of Oncology, Affiliated Hospital of Chengde Medical University, No.36 Nanyingzi St, Shuangqiao District, Chengde, Hebei Province, China.
| |
Collapse
|
20
|
Luyendijk M, Vernooij RWM, Blommestein HM, Siesling S, Uyl-de Groot CA. Assessment of Studies Evaluating Incremental Costs, Effectiveness, or Cost-Effectiveness of Systemic Therapies in Breast Cancer Based on Claims Data: A Systematic Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:1497-1508. [PMID: 33127021 DOI: 10.1016/j.jval.2020.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES Large secondary databases, such as those containing insurance claims data, are increasingly being used to compare the effects and costs of treatments in routine clinical practice. Despite their appeal, however, caution must be exercised when using these data. In this study, we aimed to identify and assess the methodological quality of studies that used claims data to compare the effectiveness, costs, or cost-effectiveness of systemic therapies for breast cancer. METHODS We searched Embase, the Cochrane Library, Medline, Web of Science, and Google Scholar for English-language publications and assessed the methodological quality using the Good Research for Comparative Effectiveness principles. This study was registered with the International Prospective Register of Systematic Reviews (PROSPERO) under number CRD42018103992. RESULTS We identified 1251 articles, of which 106 met the inclusion criteria. Most studies were conducted in the United States (74%) and Taiwan (9%) and were based on claims data sets (35%) or claims data linked to cancer registries (58%). Furthermore, most included large samples (mean 17 130 patients) and elderly patients, and they covered various outcomes (eg, survival, adverse events, resource use, and costs). Key methodological shortcomings were the lack of information on relevant confounders, the risk of immortal time bias, and the lack of information on the validity of outcomes. Only a few studies performed sensitivity analyses. CONCLUSIONS Many comparative studies of cost, effectiveness, and cost-effectiveness have been published in recent decades based on claims data, and the number of publications has increased over time. Despite the availability of guidelines to improve quality, methodological issues persist and are often inappropriately addressed or reported.
Collapse
Affiliation(s)
- Marianne Luyendijk
- Department of Research and Development, Netherlands Comprehensive Cancer Center, Utrecht, The Netherlands; Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands.
| | - Robin W M Vernooij
- Department of Research and Development, Netherlands Comprehensive Cancer Center, Utrecht, The Netherlands
| | - Hedwig M Blommestein
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| | - Sabine Siesling
- Department of Research and Development, Netherlands Comprehensive Cancer Center, Utrecht, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management, Erasmus University, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Zhao C, Cai X, Wang Y, Wang D, Wang T, Gong H, Sun H, Jia Q, Zhou W, Wu Z, Li Z, Xiao J. NAT1 promotes osteolytic metastasis in luminal breast cancer by regulating the bone metastatic niche via NF-κB/IL-1B signaling pathway. Am J Cancer Res 2020; 10:2464-2479. [PMID: 32905535 PMCID: PMC7471372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 06/11/2023] Open
Abstract
Breast cancer is a molecularly heterogeneous disease that can be subdivided into different subtypes. Compared with the other subtypes, luminal breast cancer (LBC) is considered more susceptible to bone metastasis. However, the intrinsic mechanisms remain elusive. Bioinformatics analysis of the preset study showed that N-acetyltransferase 1 (NAT1) was specifically expressed in LBC and closely correlated with bone metastasis. In addition, NAT1 could promote LBC cell migration and clonal formation, induce osteoclast differentiation and raise the Rankl/Opg ratio in osteoblasts. Our in vivo experiment demonstrated that NAT1 promoted LBC bone metastasis and bone destruction, which could be reversed by NAT1 inhibitor treatment. The result of cytokine array showed that NAT1 could significantly over activate the NF-κB signaling pathway and up-regulate the expression of IL-1B, which further worked as downstream factors in these processes. All these results demonstrated NAT1 was up-regulated in LBC and promoted the formation of bone metastatic niche and osteolytic bone metastasis through the NAT1/NF-κB/IL-1B axis. This finding may provide a new pathway to help understand the mechanisms of LBC bone metastasis and suggest a novel therapeutic and diagnostic target for its treatment.
Collapse
Affiliation(s)
- Chenglong Zhao
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Xiaopan Cai
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Yao Wang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Dongsheng Wang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Ting Wang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Haiyi Gong
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Haitao Sun
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Qi Jia
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Wang Zhou
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Zhipeng Wu
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Zhenxi Li
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| | - Jianru Xiao
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Navy Medical University (Second Military Medical University) Shanghai, China
| |
Collapse
|
22
|
Gong C, Yu X, You B, Wu Y, Wang R, Han L, Wang Y, Gao S, Yuan Y. Macrophage-cancer hybrid membrane-coated nanoparticles for targeting lung metastasis in breast cancer therapy. J Nanobiotechnology 2020; 18:92. [PMID: 32546174 PMCID: PMC7298843 DOI: 10.1186/s12951-020-00649-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/11/2020] [Indexed: 12/27/2022] Open
Abstract
Cell membrane- covered drug-delivery nanoplatforms have been garnering attention because of their enhanced bio-interfacing capabilities that originate from source cells. In this top-down technique, nanoparticles (NPs) are covered by various membrane coatings, including membranes from specialized cells or hybrid membranes that combine the capacities of different types of cell membranes. Here, hybrid membrane-coated doxorubicin (Dox)-loaded poly(lactic-co-glycolic acid) (PLGA) NPs (DPLGA@[RAW-4T1] NPs) were fabricated by fusing membrane components derived from RAW264.7(RAW) and 4T1 cells (4T1). These NPs were used to treat lung metastases originating from breast cancer. This study indicates that the coupling of NPs with a hybrid membrane derived from macrophage and cancer cells has several advantages, such as the tendency to accumulate at sites of inflammation, ability to target specific metastasis, homogenous tumor targeting abilities in vitro, and markedly enhanced multi-target capability in a lung metastasis model in vivo. The DPLGA@[RAW-4T1] NPs exhibited excellent chemotherapeutic potential with approximately 88.9% anti-metastasis efficacy following treatment of breast cancer-derived lung metastases. These NPs were robust and displayed the multi-targeting abilities of hybrid membranes. This study provides a promising biomimetic nanoplatform for effective treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Chunai Gong
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Xiaoyan Yu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Benming You
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yan Wu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Rong Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Lu Han
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Yujie Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
23
|
Sinha S, Sharma S, Vora J, Shrivastava N. Emerging role of sirtuins in breast cancer metastasis and multidrug resistance: Implication for novel therapeutic strategies targeting sirtuins. Pharmacol Res 2020; 158:104880. [PMID: 32442721 DOI: 10.1016/j.phrs.2020.104880] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Sirtuins (SIRTs), a class III histone deacetylases (HDACs) that require NAD+ as a cofactor and include SIRT1-7 proteins in mammals. Accumulative evidence has established that every sirtuin possesses exclusive and poised biology, implicating their role in the regulation of multifaceted biological functions leading to breast cancer initiation, progression, and metastasis. This article provides an outline of recent developments in the role of sirtuins in breast cancer metastasis and development of multidrug resistance (MDR). In addition, we have also highlighted the impending prospects of targeting SIRTs to overcome MDR to bring advancement in breast cancer management. Further, this review will focus on strategies for improving the activity and efficacy of existing cancer therapeutics by combining (adjuvant treatment/therapy) them with sirtuin inhibitors/modulators. All available as well as newly discovered synthetic and dietary sirtuin inhibitors, activators/modulators have been extensively reviewed and compiled to provide a rationale for targeting sirtuins. Further, we discuss their potential in developing future therapeutics against sirtuins proposing their use along with conventional chemotherapeutics to overcome the problem of breast cancer metastasis and MDR.
Collapse
Affiliation(s)
- Sonam Sinha
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Sonal Sharma
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India
| | - Jaykant Vora
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Neeta Shrivastava
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.
| |
Collapse
|
24
|
Cavaco M, Gaspar D, ARB Castanho M, Neves V. Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Pharmaceutics 2020; 12:E62. [PMID: 31940974 PMCID: PMC7023012 DOI: 10.3390/pharmaceutics12010062] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/13/2019] [Accepted: 12/28/2019] [Indexed: 12/25/2022] Open
Abstract
The incidence of brain metastases (BM) in cancer patients is increasing. After diagnosis, overall survival (OS) is poor, elicited by the lack of an effective treatment. Monoclonal antibody (mAb)-based therapy has achieved remarkable success in treating both hematologic and non-central-nervous system (CNS) tumors due to their inherent targeting specificity. However, the use of mAbs in the treatment of CNS tumors is restricted by the blood-brain barrier (BBB) that hinders the delivery of either small-molecules drugs (sMDs) or therapeutic proteins (TPs). To overcome this limitation, active research is focused on the development of strategies to deliver TPs and increase their concentration in the brain. Yet, their molecular weight and hydrophilic nature turn this task into a challenge. The use of BBB peptide shuttles is an elegant strategy. They explore either receptor-mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT) to cross the BBB. The latter is preferable since it avoids enzymatic degradation, receptor saturation, and competition with natural receptor substrates, which reduces adverse events. Therefore, the combination of mAbs properties (e.g., selectivity and long half-life) with BBB peptide shuttles (e.g., BBB translocation and delivery into the brain) turns the therapeutic conjugate in a valid approach to safely overcome the BBB and efficiently eliminate metastatic brain cells.
Collapse
Affiliation(s)
| | | | - Miguel ARB Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (M.C.); (D.G.)
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; (M.C.); (D.G.)
| |
Collapse
|
25
|
Liu Y, Li H, Liu F, Gao LB, Han R, Chen C, Ding X, Li S, Lu K, Yang L, Tian HM, Chen BB, Li X, Xu DH, Deng XL, Shi SL. Heterogeneous nuclear ribonucleoprotein A2/B1 is a negative regulator of human breast cancer metastasis by maintaining the balance of multiple genes and pathways. EBioMedicine 2020; 51:102583. [PMID: 31901866 PMCID: PMC6948170 DOI: 10.1016/j.ebiom.2019.11.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Background Heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 is an important RNA-binding protein that affects the RNA processing, splicing, transport and stability of many genes. hnRNPA2/B1 is expressed during proliferation and metastasis of various cancer types and promotes such processes. However, the precise role and mechanism of hnRNPA2/B1 in breast cancer remain unclear. Methods The association of hnRNPA2/B1 with breast cancer metastasis was assessed using tissue chips, mouse models and publicly available data. The role and mechanism of hnRNPA2/B1 in breast cancer metastasis were studied in cell lines and mouse models. Findings In contrast to other cancer research findings, hnRNPA2/B1 expression was negatively correlated with breast cancer metastasis. hnRNPA2/B1 inhibited MDA-MB-231 triple-negative breast cancer (TNBC) cell metastasis in vitro and in vivo. hnRNPA2/B1 knockout activated ERK-MAPK/Twist and GR-beta/TCF4 pathways but inhibited STAT3 and WNT/TCF4 signalling pathways. Profilin 2 (PFN2) promoted breast cancer cell migration and invasion, whereas hnRNPA2/B1 bound directly to the UAGGG locus in the 3′-untranslated region of PFN2 mRNA and reduced the stability of PFN2 mRNA. Interpretation Our data supported the role of hnRNPA2/B1 in tumour metastasis risk and survival prediction in patients with breast cancer. The inhibitory role of hnRNPA2/B1 in metastasis was a balance of downstream multiple genes and signalling pathways. PFN2 downregulation by hnRNPA2/B1 might partly explain the inhibitory mechanism of hnRNPA2/B1 in breast cancer metastasis. Therefore, hnRNPA2/B1 might be used as a new prognostic biomarker and valuable molecular target for breast cancer treatments.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China; School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai 200240, PR China
| | - Huan Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Fan Liu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Li-Bin Gao
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Rong Han
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Chen Chen
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xue Ding
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Shuang Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Kun Lu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Ling Yang
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Hui-Min Tian
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Bin-Bin Chen
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xiao Li
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Dong-Hui Xu
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Xiao-Ling Deng
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China
| | - Song-Lin Shi
- Cancer Research Center, School of Medicine, Xiamen University, Room 303, No.4221-122, Xiang'annan Road, Xiang'an District, Xiamen 361102, PR China.
| |
Collapse
|
26
|
He J, Wei X, Li S, Quan X, Li R, Du H, Yuan S, Sun L. DT-13 suppresses breast cancer metastasis by modulating PLOD2 in the adipocytes microenvironment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152778. [PMID: 31005809 DOI: 10.1016/j.phymed.2018.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Metastasis is the main cause of death in breast cancer and previous researches have indicated the pivotal role of adipocytes in breast cancer metastasis. DT-13, the saponin monomer 13 of the Dwarf lilyturf tuber, has been proved to exert potential anti-metastatic effect, the detailed mechanisms have not been well elucidated and the role of DT-13 in modulating adipocyte-breast cancer microenvironment has been given little attention. PURPOSE This study aims to explore the mechanisms of DT-13 in inhibiting breast cancer metastasis and whether DT-13 inhibit breast cancer metastasis via modulating the interactions between adipocytes and breast cancer cells. METHODS The cytotoxic effect of DT-13 on breast cancer cell viability was detected by MTT assay. Migration assays was used to conduct the effect of DT-13 on breast cancer cells migration. Orthotopic xenograft tumor model was used to test the effect of DT-13 on breast cancer metastasis. qRT-PCR and Western blot were used to investigate the mechanisms of DT-13 inhibiting breast cancer metastasis. RESULTS DT-13 inhibited breast cancer cells migration at the concentration without cytotoxicity. Furthermore, DT-13 decreased PLOD2 expression through modulating JAK/STAT3 and PI3K/AKT signaling pathways directly or indirectly in the adipocyte-breast cancer microenvironment. Orthotopic implantation mouse model of breast cancer further confirmed that DT-13 inhibited breast cancer metastasis via downregulating PLOD2 in vivo. CONCLUSION DT-13 suppressed breast cancer metastasis via reducing the expression of PLOD2.
Collapse
Affiliation(s)
- Jinyong He
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaohui Wei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Sijing Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xingping Quan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Ruiming Li
- Tasly Research Institute, Tianjin Tasly Hodling Group Co., Ltd., Tianjin, China
| | - Hongzhi Du
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Shengtao Yuan
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China.
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
27
|
Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C, Parker MI. Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:17-34. [PMID: 29356626 DOI: 10.1089/omi.2017.0174] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Disruptive innovations in medicine are game-changing in nature and bring about radical shifts in the way we understand human diseases, their treatment, and/or prevention. Yet, disruptive innovations in cancer drug design and development are still limited. Therapies that cure all cancer patients are in short supply or do not exist at all. Chief among the causes of this predicament is drug resistance, a mechanism that is much more dynamic than previously understood. Drug resistance has limited the initial success experienced with biomarker-guided targeted therapies as well. A major contributor to drug resistance is intratumor heterogeneity. For example, within solid tumors, there are distinct subclones of cancer cells, presenting profound complexity to cancer treatment. Well-known contributors to intratumor heterogeneity are genomic instability, the microenvironment, cellular genotype, cell plasticity, and stochastic processes. This expert review explains that for oncology drug design and development to be more innovative, we need to take into account intratumor heterogeneity. Initially thought to be the preserve of cancer cells, recent evidence points to the highly heterogeneous nature and diverse locations of stromal cells, such as cancer-associated fibroblasts (CAFs) and cancer-associated macrophages (CAMs). Distinct subpopulations of CAFs and CAMs are now known to be located immediately adjacent and distant from cancer cells, with different subpopulations exerting different effects on cancer cells. Disruptive innovation and precision medicine in clinical oncology do not have to be a distant reality, but can potentially be achieved by targeting these spatially separated and exclusive cancer cell subclones and CAF subtypes. Finally, we emphasize that disruptive innovations in drug discovery and development will likely come from drugs whose effect is not necessarily tumor shrinkage.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Nicholas Ekow Thomford
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa
| | - Collet Dandara
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
28
|
Ghasemi F, Sarabi PZ, Athari SS, Esmaeilzadeh A. Therapeutics strategies against cancer stem cell in breast cancer. Int J Biochem Cell Biol 2019; 109:76-81. [PMID: 30772480 DOI: 10.1016/j.biocel.2019.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 01/06/2023]
Abstract
Breast cancer is known as a most prevalent cancer and second deadly cancer, among women worldwide. Due to the high incidence rate of breast cancer and limitations of conventional therapy it seemed essential to look for new targets in cancer cells and directly target them such as target therapy on breast cancer stem cells. In this review we indicate some of therapeutic uses of cancer stem cells in breast cancer. Some strategies are targeting surface specific markers and activated signaling pathways in their microenvironment such as Notch, Hedgehog, Wnt/b-catenin, PI3K/Akt, NF-kB, BMP and TGF-β and their maintenance and drug resistance, using various miRNAs, enhancement of CSCs apoptosis, differentiation therapy, blocking epithelial to mesenchymal transition and using different natural compounds. Recent studies have shown that cancer stem cells play major roles in target therapy on breast cancer. The new manipulation approaches of cancer stem cells can be used as target therapy of breast cancer that were highlighted for immunotherapy of cancer.
Collapse
Affiliation(s)
- Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Parisa Zia Sarabi
- Molecular and Medicine Research Center, Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
29
|
Zhao C, Lou Y, Wang Y, Wang D, Tang L, Gao X, Zhang K, Xu W, Liu T, Xiao J. A gene expression signature-based nomogram model in prediction of breast cancer bone metastases. Cancer Med 2018; 8:200-208. [PMID: 30575323 PMCID: PMC6346244 DOI: 10.1002/cam4.1932] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is prone to form bone metastases and subsequent skeletal‐related events (SREs) dramatically decrease patients’ quality of life and survival. Prediction and early management of bone lesions are valuable; however, proper prognostic models are inadequate. In the current study, we reviewed a total of 572 breast cancer patients in three microarray data sets including 191 bone metastases and 381 metastases‐free. Gene set enrichment analysis (GSEA) indicated less aggressive and low‐grade features of patients with bone metastases compared with metastases‐free ones, while luminal subtypes are more prone to form bone metastases. Five bone metastases‐related genes (KRT23, REEP1, SPIB, ALDH3B2, and GLDC) were identified and subjected to construct a gene expression signature‐based nomogram (GESBN) model. The model performed well in both training and testing sets for evaluation of breast cancer bone metastases (BCBM). Clinically, the model may help in prediction of early bone metastases, prevention and management of SREs, and even help to prolong survivals for patients with BCBM. The five‐gene GESBN model showed some implications as molecular diagnostic markers and therapeutic targets. Furthermore, our study also provided a way for analysis of tumor organ‐specific metastases. To the best of our knowledge, this is the first published model focused on tumor organ‐specific metastases.
Collapse
Affiliation(s)
- Chenglong Zhao
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yan Lou
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yao Wang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dongsheng Wang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Liang Tang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xin Gao
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Kun Zhang
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei Xu
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tielong Liu
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jianru Xiao
- Spine Tumor Center, Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Cai KT, Feng CX, Zhao JC, He RQ, Ma J, Zhong JC. Upregulated miR‑203a‑3p and its potential molecular mechanism in breast cancer: A study based on bioinformatics analyses and a comprehensive meta‑analysis. Mol Med Rep 2018; 18:4994-5008. [PMID: 30320391 PMCID: PMC6236224 DOI: 10.3892/mmr.2018.9543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 08/31/2018] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) has been identified as the leading malignancy in women worldwide. However, the potential molecular mechanism of microRNA (miR)‑203a‑3p in BC remains to be elucidated. The present study evaluated the expression of miR‑203a‑3p in BC and adjacent normal tissue in several publically available datasets. The distinguishability of precursor miR‑203a and miR‑203a‑3p in BC tissue and adjacent breast tissue was assessed using receiver operating characteristic (ROC) and summarized ROC (sROC) approaches. In addition, gene ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes pathway analysis and protein‑protein interaction analysis were performed to determine the potential molecular mechanism of miR‑203a‑3p in BC. It was identified that the expression of precursor miR‑203a was markedly upregulated in 1,077 BC tissue samples compared to 104 adjacent breast tissue samples from The Cancer Genome Atlas. Additionally, an increasing trend in miR‑203a‑3p expression was observed in 756 BC tissue samples compared with 76 adjacent breast tissue samples from the University of California Santa Cruz Xena project. In addition, a comprehensive meta‑analysis suggested that the expression of miR‑203a‑3p was markedly increased in 2,444 BC tissue samples compared with 559 adjacent breast tissue samples. The area under the curve of the ROC and sROC revealed that miR‑203a‑3p expression was able to distinguish between BC tissue and adjacent breast tissue. However, miR‑203a‑3p exhibited no prognostic value in BC. The results of GO enrichment demonstrated that the miR‑203a target genes were associated with 'plasma membrane integrity', 'cell surface receptor linked signal and transduction' and '3',5'‑cyclic nucleotide phosphodiesterase activity'. 'Purine metabolism' was identified as the pathway with the most enrichment of miR‑203a‑3p target genes in BC. The present study also identified insulin‑like growth factor receptor (IGF1) as a hub gene associated with miR‑203a in BC. In summary, miR‑203a‑3p may enhance the development and oncogenesis of BC, and IGF1 was defined as a hub gene of miR‑203a‑3p in BC.
Collapse
Affiliation(s)
- Kai-Teng Cai
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Cai-Xia Feng
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jin-Che Zhao
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jie Ma
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jin-Cai Zhong
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
31
|
Chen DL, Xu RH. The emerging role of long non-coding RNAs in the drug resistance of colorectal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4735-4743. [PMID: 31949549 PMCID: PMC6962903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/23/2018] [Indexed: 06/10/2023]
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related deaths in the world. Chemotherapy has been used to treat CRC patients in order to improve prognosis. Oxaliplatin and 5-Fluorouracil (5-FU) based chemotherapy is a first line treatment for locally advanced and metastatic CRC. For patients with wild-type KRAS metastatic CRC, cetuximab (an EGFR monoclonal antibody) is a commonly used targeted therapy. CRC is initially sensitive to chemotherapy and targeted therapy. However, drug resistance frequently arises, which significantly affect the treatment outcome in these patients. An increasing number of studies have indicated that lncRNAs are implicated in the drug resistance of CRC. This review aims to gain insights into the role and molecular mechanism of lncRNAs in CRC drug resistance.
Collapse
Affiliation(s)
- Dong-Liang Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center Guangzhou, PR China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center Guangzhou, PR China
| |
Collapse
|
32
|
Stoletov K, Willetts L, Paproski RJ, Bond DJ, Raha S, Jovel J, Adam B, Robertson AE, Wong F, Woolner E, Sosnowski DL, Bismar TA, Wong GKS, Zijlstra A, Lewis JD. Quantitative in vivo whole genome motility screen reveals novel therapeutic targets to block cancer metastasis. Nat Commun 2018; 9:2343. [PMID: 29904055 PMCID: PMC6002534 DOI: 10.1038/s41467-018-04743-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/18/2018] [Indexed: 01/03/2023] Open
Abstract
Metastasis is the most lethal aspect of cancer, yet current therapeutic strategies do not target its key rate-limiting steps. We have previously shown that the entry of cancer cells into the blood stream, or intravasation, is highly dependent upon in vivo cancer cell motility, making it an attractive therapeutic target. To systemically identify genes required for tumor cell motility in an in vivo tumor microenvironment, we established a novel quantitative in vivo screening platform based on intravital imaging of human cancer metastasis in ex ovo avian embryos. Utilizing this platform to screen a genome-wide shRNA library, we identified a panel of novel genes whose function is required for productive cancer cell motility in vivo, and whose expression is closely associated with metastatic risk in human cancers. The RNAi-mediated inhibition of these gene targets resulted in a nearly total (>99.5%) block of spontaneous cancer metastasis in vivo. Tumour metastasis is dependent on tumour cell motility. Here, the authors investigate genes required for tumour cell motility by establishing a quantitative in vivo screening platform based on intravital imaging of human cancer metastasis in ex ovo avian embryos.
Collapse
Affiliation(s)
- Konstantin Stoletov
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Lian Willetts
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Robert J Paproski
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - David J Bond
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Srijan Raha
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Juan Jovel
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Benjamin Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Amy E Robertson
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Francis Wong
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Emma Woolner
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Deborah L Sosnowski
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Tarek A Bismar
- Departments of Pathology and Laboratory Medicine, Oncology, Biochemistry and Molecular Biology, University of Calgary Cumming School of Medicine and Calgary Laboratory Services, Calgary, AB, T2V 1P9, Canada
| | - Gane Ka-Shu Wong
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada.,Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,BGI-Shenzhen, Beishan Industrial Zone, Yantian District, 518083, Shenzhen, China
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, 1161 21st Ave. S., C-2104A MCN, Nashville, TN, 37232-2561, USA
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
33
|
Gallo M, Frezzetti D, Roma C, Chicchinelli N, Barbieri A, Arra C, Scognamiglio G, Botti G, De Luca A, Normanno N. RANTES and IL-6 cooperate in inducing a more aggressive phenotype in breast cancer cells. Oncotarget 2018; 9:17543-17553. [PMID: 29707128 PMCID: PMC5915136 DOI: 10.18632/oncotarget.24784] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/26/2018] [Indexed: 01/04/2023] Open
Abstract
Both the CC chemokine ligand 5 (CCL5/RANTES) and interleukin-6 (IL-6), released by mesenchymal stem cells (MSCs) as well as by neoplastic cells, promote breast cancer cell progression through autocrine and paracrine mechanisms. In order to assess the effects of the simultaneous overexpression of RANTES and IL-6 on the tumor cell phenotype, we overexpressed both proteins in MCF-7 and MDA-MB-231 human breast cancer cell lines. MCF-7 cells co-expressing RANTES and IL-6 had a greater ability to form colonies in soft agar, compared to cells overexpressing RANTES or IL-6. In addition, both MCF-7 and MDA-MB-231 clones co-expressing RANTES and IL-6 showed a significantly higher ability to migrate and to invade. The analysis of phosphorylated ERK1/2, AKT and STAT3 signal transduction proteins revealed that several signaling pathways are simultaneously activated in cells overexpressing both factors. Finally, the overexpression of RANTES and IL-6 in MCF-7 cells significantly increased the in vivo tumor growth. Collectively, our data suggest that the simultaneous expression of IL-6 and RANTES produces a more aggressive phenotype in breast cancer cells and provide evidence that IL-6 and RANTES might represent potential targets for novel therapeutic strategies aimed to block the tumor-stroma interaction.
Collapse
Affiliation(s)
- Marianna Gallo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Nicoletta Chicchinelli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Giosuè Scognamiglio
- Surgical Pathology Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Gerardo Botti
- Surgical Pathology Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| |
Collapse
|
34
|
A tumor-myeloid cell axis, mediated via the cytokines IL-1α and TSLP, promotes the progression of breast cancer. Nat Immunol 2018; 19:366-374. [PMID: 29556001 PMCID: PMC5864553 DOI: 10.1038/s41590-018-0066-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Abstract
Tumors actively manipulate the immune response through the production of factors that attract immune cells and subsequently alter their ability to recognize and effectively remove the tumor. While this immune evasion mechanism is an important aspect of tumor survival, the factors that serve as primary growth factors for the tumor are less understood. Here, we demonstrated a novel mechanism by which breast cancer cells manipulate tumor-infiltrating myeloid cells to maintain their survival. Tumor-derived interleukin 1α (IL-1α), acting on infiltrating myeloid cells, induced the expression of a critical tumor survival factor, the cytokine thymic stromal lymphopoietin (TSLP). TSLP promoted the survival of the tumor cells through induction of Bcl-2 expression. TSLP signaling was also required for metastasis to the lung. These studies define a novel IL-1α–TSLP-mediated crosstalk between tumor-infiltrating myeloid cells and tumor cells in the control of metastatic breast cancer.
Collapse
|
35
|
Xiang D, Yang JY, Zhang CL, Liu D. Research progress on baicalin in treatment of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2018; 26:378-383. [DOI: 10.11569/wcjd.v26.i6.378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors. Traditional Chinese medicine (TCM) has shown special therapeutic effects for HCC. Scutellaria baicalensis Georgi has antipyretic and hepatoprotective effects, and baicalin, the main active compound of Scutellaria baicalensis Georgi, has been widely reported for the treatment of HCC in recent years. The mechanisms of baicalin for inhibition of liver cancer growth involve inducing cancer cell apoptosis and differentiation, inhibiting protein synthesis and cancer cell migration, interfering with intercellular communication, reversing tumor drug resistance and so on. In addition, baicalin combined with traditional chemotherapeutic drugs or other anti-cancer TCMs has been demonstrated to exert good synergistic activity for HCC treatment. Based on the in vivo absorption characteristics and molecular structure of baicalin, researchers have developed solid lipid nanoparticles and metal-ion complexes of baicalin, which provides new insights into the use of baicalin in targeted therapy and improves anti-cancer effects. In this paper, we will review the recent advances in research of baicalin in the treatment of baicalin.
Collapse
|
36
|
Cai WL, Huang WD, Li B, Chen TR, Li ZX, Zhao CL, Li HY, Wu YM, Yan WJ, Xiao JR. microRNA-124 inhibits bone metastasis of breast cancer by repressing Interleukin-11. Mol Cancer 2018; 17:9. [PMID: 29343249 PMCID: PMC5773190 DOI: 10.1186/s12943-017-0746-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/26/2017] [Indexed: 02/06/2023] Open
Abstract
Background Most patients with breast cancer in advanced stages of the disease suffer from bone metastases which lead to fractures and nerve compression syndromes. microRNA dysregulation is an important event in the metastases of breast cancer to bone. microRNA-124 (miR-124) has been proved to inhibit cancer progression, whereas its effect on bone metastases of breast cancer has not been reported. Therefore, this study aimed to investigate the role and underlying mechanism of miR-124 in bone metastases of breast cancer. Methods In situ hybridization (ISH) was used to detect the expression of miR-124 in breast cancer tissues and bone metastatic tissues. Ventricle injection model was constructed to explore the effect of miR-124 on bone metastasis in vivo. The function of cancer cell derived miR-124 in the differentiation of osteoclast progenitor cells was verified in vitro. Dual-luciferase reporter assay was conducted to confirm Interleukin-11 (IL-11) as a miR-124 target. The involvement of miR-124/IL-11 in the prognosis of breast cancer patients with bone metastasis was determined by Kaplan-Meier analysis. Results Herein, we found that miR-124 was significantly reduced in metastatic bone tissues from breast cancers. Down-regulation of miR-124 was associated with aggressive clinical characteristics and shorter bone metastasis-free survival and overall survival. Restoration of miR-124 suppressed, while inhibition of miR-124 promoted the bone metastasis of breast cancer cells in vivo. At the cellular level, gain of function and loss-of function assays indicated that cancer cell-derived miR-124 inhibited the survival and differentiation of osteoclast progenitor cells. At the molecular level, we demonstrated that IL-11 partially mediated osteoclastogenesis suppression by miR-124 using in vitro and in vivo assays. Furthermore, IL-11 levels were inversely correlated with miR-124, and up-regulation IL-11 in bone metastases was associated with a poor prognosis. Conclusions Thus, the identification of a dysregulated miR-124/IL-11 axis helps elucidate mechanisms of breast cancer metastases to bone, uncovers new prognostic markers, and facilitates the development of novel therapeutic targets to treat and even prevent bone metastases of breast cancer. Electronic supplementary material The online version of this article (10.1186/s12943-017-0746-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Luo Cai
- Department of Musculoskeletal Tumor, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai, 200032, People's Republic of China.,Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Wen-Ding Huang
- Department of Musculoskeletal Tumor, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai, 200032, People's Republic of China
| | - Bo Li
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Tian-Rui Chen
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Zhen-Xi Li
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Cheng-Long Zhao
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China
| | - Heng-Yu Li
- Department of Breast and Thyroid Surgery, General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yan-Mei Wu
- Department of Breast and Thyroid Surgery, General Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wang-Jun Yan
- Department of Musculoskeletal Tumor, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong An Road, Shanghai, 200032, People's Republic of China. .,Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China.
| | - Jian-Ru Xiao
- Spine Tumor Center, Changzheng Hospital, Second Military Medical University, 415 Feng Yang Road, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
37
|
Abstract
The mammary epithelium is organized in a hierarchy of mammary stem cells (MaSCs), progenitors, and differentiated cells. The development and homeostasis of mammary gland are tightly controlled by a complex network of cell lineage regulators. These determinants of cellular hierarchy are frequently deregulated in breast tumor cells and closely associated with cancer progression and metastasis. They also contribute to the diversity of breast cancer subtypes and their distinct metastatic patterns. Cell fate regulators that normally promote stem/progenitor activities can serve as drivers for epithelial-mesenchymal transition and metastasis whereas regulators that promote terminal differentiation generally suppress metastasis. In this review, we discuss how some of the key factors function in normal mammary lineage determination and how these processes are hijacked by tumor cells to enhance metastasis. Understanding the molecular connections between normal development and cancer metastasis will enable the development of more specific and effective therapeutic approaches targeting metastatic tumor cells.
Collapse
Affiliation(s)
- Wei Lu
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Washington Road, LTL 255, Princeton, NJ, 08544, USA.
| |
Collapse
|
38
|
Liu JL, Pan YY, Chen O, Luan Y, Xue X, Zhao JJ, Liu L, Jia HY. Curcumin inhibits MCF-7 cells by modulating the NF-κB signaling pathway. Oncol Lett 2017; 14:5581-5584. [PMID: 29142607 DOI: 10.3892/ol.2017.6860] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
The present study investigated the inhibitory effect of curcumin on human breast cancer MCF-7 cells and investigated the potential underlying molecular mechanisms. MCF-7 cells were cultured with curcumin at different concentrations and time points. The effects of curcumin treatment on breast cancer cell proliferation were studied using a MTT assay. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were used to assess the mRNA and protein expression levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X (Bax), nuclear factor-κ-light-chain-enhancer of activated B cells (NF-κB) and inhibitor of NF-κB-α (IκBα). The proliferation of MCF-7 cells in the group treated with curcumin was markedly decreased compared with the control, with the greatest inhibitory effect at a concentration of 20 µM. The expression of Bax mRNA was increased and Bcl-2 mRNA expression was decreased compared with the control. Additionally, protein expression of NF-κB and IκB was increased. The data indicate that curcumin is able to inhibit breast cancer cell proliferation, possibly by regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jun-Li Liu
- Clinical Molecular Biology Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan-Yan Pan
- Department of Pediatrics, Qilu Children's Hospital of Shandong University, Jinan, Shandong 066600, P.R. China
| | - Ou Chen
- College of Nursing, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Jing-Jie Zhao
- Clinical Molecular Biology Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ling Liu
- Clinical Molecular Biology Laboratory, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Hong-Ying Jia
- Center of Evidence-Based Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
39
|
Chen C, Maimaiti Y, Zhijun S, Zeming L, Yawen G, Pan Y, Tao H. Slingshot-1L, a cofilin phosphatase, induces primary breast cancer metastasis. Oncotarget 2017; 8:66195-66203. [PMID: 29029503 PMCID: PMC5630403 DOI: 10.18632/oncotarget.19855] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/28/2017] [Indexed: 01/06/2023] Open
Abstract
Slingshot (SSH) is a member of the conserved family of cofilin phosphatases that plays a critical role in cell membrane protrusion and migration by transforming inactive phosphorylated cofilin to an active form. SSH-like protein 1 (SSH-1L) expression is detected in various types of tumors; insulin induces the phosphatases activity of SSH-1L in a phosphoinositide 3-kinase-dependent manner. However, little is known about the expression and role of SSH-1L in breast cancer. Here, we analyzed 295 human breast cancer tissue specimens for SSH-1L expression by immunohistochemistry. The correlation between SSH-1L level and patients' clinical characteristics was analyzed with Pearson's χ2 test. The function of SSH-1L was evaluated by gene knockdown and quantitative real-time polymerase chain reaction detection of cofilin expression in MDA-MB-231, MCF-7, and SK-BR-3 human breast cancer cell lines. SSH-1L expression was detected in 88.1% of tissue specimens by immunohistochemistry and was strongly associated with increased metastasis and mortality. Loss of SSH-1L expression decreased the nonphosphorylated, active form of cofilin in SK-BR-3 and MDA-MB-231 cell lines, which was associated with reduced cell motility. Accordingly, SSH-1L/cofilin signaling played a critical role in primary breast cancer metastasis and was a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Chen Chen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yusufu Maimaiti
- Department of General Surgery (Research Institute of Minimally Invasive), People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830000, P.R. China
| | - Shen Zhijun
- Clinical Laboratory, The Third People's Hospital of Hubei Province, Wuhan 430000, P.R. China
| | - Liu Zeming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Guo Yawen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Yu Pan
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Huang Tao
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| |
Collapse
|
40
|
Abstract
The development of intrinsic or acquired resistance to chemotherapeutic agents used in the treatment of various human cancers is a major obstacle for the successful abolishment of cancer. The accumulated efforts in the understanding the exact mechanisms of development of multidrug resistance (MDR) have led to the introduction of several unique and common mechanisms. Recent studies demonstrate the regulatory role of small noncoding RNA or miRNA in the several parts of cancer biology. Practically all aspects of cell physiology under normal and disease conditions are reported to be controlled by miRNAs. In this review, we discuss how the miRNA profile is changed upon MDR development and the pivotal regulatory role played by miRNAs in overcoming resistance to chemotherapeutic agents. It is hoped that further studies will support the use of these differentially expressed miRNAs as prognostic and predictive markers, as well as novel therapeutic targets to overcome resistance in ovarian cancer.
Collapse
Affiliation(s)
- Aynaz Mihanfar
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Biochemistry, Urmia University of Medical Sciences, Urmia, Iran
| | - Amir Fattahi
- Faculty of Advanced Medical Sciences, Department of Reproductive Biology, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Hamid Reza Nejabati
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Shi F, Cai FF, Cai L, Lin XY, Zhang W, Wang QQ, Zhao YJ, Ni QC, Wang H, He ZX. Overexpression of SYF2 promotes cell proliferation and correlates with poor prognosis in human breast cancer. Oncotarget 2017; 8:88453-88463. [PMID: 29179448 PMCID: PMC5687618 DOI: 10.18632/oncotarget.18188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/30/2017] [Indexed: 12/11/2022] Open
Abstract
SYF2, a known cell cycle regulator, is reported to be involved in cell cycle arrest by interacting with cyclin-D-type binding protein 1. In the present study, we investigated the role of SYF2 in human breast cancer (BC) progression. SYF2 was highly upregulated in BC tissues and cell lines, as per Western blot and immunohistochemistry analysis. The SYF2 expression level had a significant correlation with the tumor grade and Ki-67 expression. In vitro starvation-refeeding experiment and SYF2-siRNA transfection assay demonstrated that SYF2 could promote proliferation of BC cells, while SYF2 knockdown resulted in cells cycle arrest at G1/S phase, reducing the cell growth rate of BC cells. These results indicated that SYF2 promotes human BC progression by accelerating the BC cells’ proliferation. SYF2 could be a novel therapeutic target in human BC therapies.
Collapse
Affiliation(s)
- Feng Shi
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Feng-Feng Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Lu Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xiao-Yan Lin
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Wei Zhang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Qin-Qin Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Yu-Jie Zhao
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Qi-Chao Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Hua Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| | - Zhi-Xian He
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong University, Nantong, PR China
| |
Collapse
|
42
|
Roberts LS, Yan P, Bateman LA, Nomura DK. Mapping Novel Metabolic Nodes Targeted by Anti-Cancer Drugs that Impair Triple-Negative Breast Cancer Pathogenicity. ACS Chem Biol 2017; 12:1133-1140. [PMID: 28248089 DOI: 10.1021/acschembio.6b01159] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancers (TNBCs) are estrogen receptor, progesterone receptor, and HER2 receptor-negative subtypes of breast cancers that show the worst prognoses and lack targeted therapies. Here, we have coupled the screening of ∼400 anticancer agents that are under development or in the clinic with chemoproteomic and metabolomic profiling to identify novel metabolic mechanisms for agents that impair TNBC pathogenicity. We identify 20 anticancer compounds that significantly impaired cell survival across multiple types of TNBC cells. Among these 20 leads, the phytoestrogenic natural product licochalcone A was of interest, since TNBCs are unresponsive to estrogenic therapies, indicating that licochalcone A was likely acting through another target. Using chemoproteomic profiling approaches, we reveal that licochalcone A impairs TNBC pathogenicity, not through modulating estrogen receptor activity but rather through inhibiting prostaglandin reductase 1, a metabolic enzyme involved in leukotriene B4 inactivation. We also more broadly performed metabolomic profiling to map additional metabolic mechanisms of compounds that impair TNBC pathogenicity. Overlaying lipidomic profiling with drug responses, we find that deubiquitinase inhibitors cause dramatic elevations in acyl carnitine levels, which impair mitochondrial respiration and contribute to TNBC pathogenic impairments. We thus put forth two unique metabolic nodes that are targeted by drugs or drug candidates that impair TNBC pathogenicity. Our results also showcase the utility of coupling drug screens with chemoproteomic and metabolomic profiling to uncover unique metabolic drivers of TNBC pathogenicity.
Collapse
Affiliation(s)
- Lindsay S. Roberts
- Departments of Chemistry, Molecular and
Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Peter Yan
- Departments of Chemistry, Molecular and
Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Leslie A. Bateman
- Departments of Chemistry, Molecular and
Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Daniel K. Nomura
- Departments of Chemistry, Molecular and
Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
43
|
Olver IN. New initiatives in the treatment of breast cancer. Med J Aust 2017; 205:449-450. [PMID: 27852179 DOI: 10.5694/mja16.01055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/14/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Ian N Olver
- Sansom Institute, University of South Australia, Adelaide, SA
| |
Collapse
|
44
|
The prolyl oligopeptidase inhibitor SUAM-14746 attenuates the proliferation of human breast cancer cell lines in vitro. Breast Cancer 2017; 24:658-666. [DOI: 10.1007/s12282-017-0752-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/03/2017] [Indexed: 02/06/2023]
|
45
|
Chen QN, Wei CC, Wang ZX, Sun M. Long non-coding RNAs in anti-cancer drug resistance. Oncotarget 2017; 8:1925-1936. [PMID: 27713133 PMCID: PMC5352108 DOI: 10.18632/oncotarget.12461] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy is one of the basic treatments for cancers; however, drug resistance is mainly responsible for the failure of clinical treatment. The mechanism of drug resistance is complicated because of interaction among various factors including drug efflux, DNA damage repair, apoptosis and targets mutation. Long non-coding RNAs (lncRNAs) have been a focus of research in the field of bioscience, and the latest studies have revealed that lncRNAs play essential roles in drug resistance in breast cancer, gastric cancer and lung cancer, et al. Dysregulation of multiple targets and pathways by lncRNAs results in the occurrence of chemoresistance. In this review, we will discuss the mechanisms underlying lncRNA-mediated resistance to chemotherapy and the therapeutic potential of lncRNAs in future cancer treatment.
Collapse
Affiliation(s)
- Qin-nan Chen
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chen-chen Wei
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhao-xia Wang
- Department of Oncology, Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ming Sun
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
46
|
The role of liver-directed surgery in patients with hepatic metastasis from primary breast cancer: a multi-institutional analysis. HPB (Oxford) 2016; 18:700-5. [PMID: 27485066 PMCID: PMC4972375 DOI: 10.1016/j.hpb.2016.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/12/2016] [Accepted: 05/30/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Data on surgical management of breast liver metastasis are limited. We sought to determine the safety and long-term outcome of patients undergoing hepatic resection of breast cancer liver metastases (BCLM). METHODS Using a multi-institutional, international database, 131 patients who underwent surgery for BCLM between 1980 and 2014 were identified. Clinicopathologic and outcome data were collected and analyzed. RESULTS Median tumor size of the primary breast cancer was 2.5 cm (IQR: 2.0-3.2); 58 (59.8%) patients had primary tumor nodal metastasis. The median time from diagnosis of breast cancer to metastasectomy was 34 months (IQR: 16.8-61.3). The mean size of the largest liver lesion was 3.0 cm (2.0-5.0); half of patients (52.0%) had a solitary metastasis. An R0 resection was achieved in most cases (90.8%). Postoperative morbidity and mortality were 22.8% and 0%, respectively. Median and 3-year overall-survival was 53.4 months and 75.2%, respectively. On multivariable analysis, positive surgical margin (HR 3.57, 95% CI 1.40-9.16; p = 0.008) and diameter of the BCLM (HR 1.03, 95% CI 1.01-1.06; p = 0.002) remained associated with worse OS. DISCUSSION In selected patients, resection of breast cancer liver metastases can be done safely and a subset of patients may derive a relatively long survival, especially from a margin negative resection.
Collapse
|