1
|
Li X, Zhu D. Role of disulfide death in cancer (Review). Oncol Lett 2025; 29:55. [PMID: 39606569 PMCID: PMC11600708 DOI: 10.3892/ol.2024.14801] [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: 07/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The research field of regulated cell death is growing extensively. Following the recognition of ferroptosis, other unique and distinct forms of regulated cell death, including cuproptosis and disulfide death, have been identified. Disulfide death occurs due to the abnormal accumulation of disulfides within cells in environments lacking glucose, leading to contraction of the actin cytoskeleton, which ultimately triggers various signaling pathways and cell death. The induction of disulfide death in the treatment of cancer may exhibit significant therapeutic potential. Therefore, in the present review, a comprehensive and critical analysis of the current understanding of the molecular mechanisms and regulatory networks of disulfide death is presented. In addition, the potential physiological functions of disulfide death in tumor suppression and immune surveillance as well as its pathological roles and therapeutic potential are described. The core focus areas for future research into this form of cell death are also explored. Given the current lack of extensive clinical findings and well-defined key concepts, these may be regarded as pivotal points of interest in future studies.
Collapse
Affiliation(s)
- Xue Li
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Danxia Zhu
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
2
|
Chang K, Yue Q, Jin L, Fan P, Liu Y, Cao F, Zhang Y. Comprehensive Molecular Analyses of an M2-Like Tumor-Associated Macrophage for Predicting the Prognosis and Immunotherapy in Breast Cancer. J Immunother 2024; 47:205-215. [PMID: 38686904 DOI: 10.1097/cji.0000000000000517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 05/02/2024]
Abstract
The involvement of M2-like tumor-associated macrophages (TAMs) in the advancement and treatment of cancer has been widely documented. This study aimed to develop a new signature associated with M2-like TAMs to predict the prognosis and treatment response in individuals diagnosed with breast cancer (BC). Weighted gene co-expression network analysis (WGCNA) was used to identity for M2-like TAM-related modular genes. The M2-like TAM-related modular subtype was identified using unsupervised clustering. WGCNA identified 722 M2-like TAM genes, 204 of which were associated with recurrence-free survival (RFS). Patients in cluster 1 exhibited upregulated cancer-related pathways, a higher proportion of triple-negative breast cancer (TNBC) subtypes, lower expression of immune checkpoints, and worse prognosis. Cluster 2 was characterized by upregulated immune-related pathways, a higher proportion of luminal A subtypes, and higher expression of immune checkpoints. A prognostic signature was created and confirmed using an independent dataset. A well-built nomogram can accurately forecast the survival outcomes for every individual. Furthermore, patients classified as low-risk exhibited a more favorable outlook, elevated tumor microenvironment (TME) score, and superior reaction to immunotherapy. In conclusion, we discovered 2 different types of M2-like TAMs and developed a prognostic signature revealing the diversity of M2-like TAMs in BC and their correlation with immune status and prognosis. This feature can predict the prognosis and immunotherapeutic effects of BC and offer novel concepts and approaches for tailoring BC treatment.
Collapse
Affiliation(s)
- Kexin Chang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - QingFang Yue
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Long Jin
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Pengyu Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, China
| | - Yi Liu
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Fei Cao
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Yuan Zhang
- Department of Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
3
|
Wang Y, Sun Y, Lu F, Zhao X, Nie Z, Zhu F, He B. Efficacy and safety of a combination treatment of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Clin Transl Oncol 2024; 26:1725-1737. [PMID: 38587602 DOI: 10.1007/s12094-024-03396-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/22/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) in combination with chemotherapy have showed its benefits in clinical studies, and here we conducted a further evaluation on the safety and efficacy of this treatment strategy. METHODS A systematic literature review was conducted in PubMed, Embase and Cochrane Library to identify clinical studies on ICIs and chemotherapy for metastatic breast cancer. The primary efficacy endpoints were progression-free survival (PFS) and overall survival (OS), and adverse events (AEs) were analyzed. Random or fixed effects models were used to estimate pooled Hazard ratio (HR), odds ratio (OR) and the data of 95% confidence interval (CI) depend on the Heterogeneity. Cochrane risk assessment tool was used to assess risk of bias. We also drew forest plots and funnel plots, respectively. RESULTS Seven studies with intend-to-treat (ITT) population for 3255 patients were analyzed. ICIs pooled therapy showed clinical benefits compared with chemotherapy alone, improving PFS (HR = 0.81, 95% CI: 0.74-0.90) of patients with metastatic triple negative breast cancer (mTNBC), especially in patients with PD-L1-positive tumors. However, it had no effect on OS (HR = 0.92, 95% CI 0.85-1.01). Besides, mTNBC patients received pooled therapy were less frequently to have AEs (OR = 1.30, 95% CI: 1.09-1.54). In patients with metastatic Human Epidermal Growth Factor Receptor 2 (HER2) negative breast cancer, pooled therapy showed no benefit for PFS (HR = 0.80, 95% CI: 0.50-1.28) and OS (HR = 0.87, 95% CI: 0.48-1.58). CONCLUSION Pooled therapy had improved PFS in mTNBC patients, especially in patients with PD-L1-positive tumors, and it was less likely to cause grade ≥ 3 AEs.
Collapse
Affiliation(s)
- Ying Wang
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Yalan Sun
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Fang Lu
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xianghong Zhao
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Zhenlin Nie
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Feng Zhu
- Department of Laboratory Medicine, Nanjing Jiangning People's Hospital, 68 Gushan Road, Jiangning District, Nanjing, Jiangsu, 211100, China.
| | - Bangshun He
- School of Basic-Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
- Deparment of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
4
|
Hu Y, Hu Q, Liu Z, Huang C, Xia L. Histogram analysis comparison of readout-segmented and single-shot echo-planar imaging for differentiating luminal from non-luminal breast cancer. Sci Rep 2024; 14:12135. [PMID: 38802446 PMCID: PMC11130195 DOI: 10.1038/s41598-024-62514-0] [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: 02/01/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
To compare diffusion-kurtosis imaging (DKI) and diffusion-weighted imaging (DWI) parameters of single-shot echo-planar imaging (ss-EPI) and readout-segmented echo-planar imaging (rs-EPI) in the differentiation of luminal vs. non-luminal breast cancer using histogram analysis. One hundred and sixty women with 111 luminal and 49 non-luminal breast lesions were enrolled in this study. All patients underwent ss-EPI and rs-EPI sequences on a 3.0T scanner. Histogram metrics were derived from mean kurtosis (MK), mean diffusion (MD) and the apparent diffusion coefficient (ADC) maps of two DWI sequences respectively. Student's t test or Mann-Whitney U test was performed for differentiating luminal subtype from non-luminal subtype. The ROC curves were plotted for evaluating the diagnostic performances of significant histogram metrics in differentiating luminal from non-luminal BC. The histogram metrics MKmean, MK50th, MK75th of luminal BC were significantly higher than those of non-luminal BC for both two DWI sequences (all P<0.05). Histogram metrics from rs-EPI sequence had better diagnostic performance in differentiating luminal from non-Luminal breast cancer compared to those from ss-EPI sequence. MK75th derived from rs-EPI sequence was the most valuable single metric (AUC, 0.891; sensitivity, 78.4%; specificity, 87.8%) for differentiating luminal from non-luminal BC among all the histogram metrics. Histogram metrics of MK derived from rs-EPI yielded better diagnostic performance for distinguishing luminal from non-luminal BC than that from ss-EPI. MK75th was the most valuable metric among all the histogram metrics.
Collapse
Affiliation(s)
- Yiqi Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Qilan Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhiqiang Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Cicheng Huang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Liming Xia
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
5
|
Zuo D, Yang L, Jin Y, Qi H, Liu Y, Ren L. Machine learning-based models for the prediction of breast cancer recurrence risk. BMC Med Inform Decis Mak 2023; 23:276. [PMID: 38031071 PMCID: PMC10688055 DOI: 10.1186/s12911-023-02377-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Breast cancer is the most common malignancy diagnosed in women worldwide. The prevalence and incidence of breast cancer is increasing every year; therefore, early diagnosis along with suitable relapse detection is an important strategy for prognosis improvement. This study aimed to compare different machine algorithms to select the best model for predicting breast cancer recurrence. The prediction model was developed by using eleven different machine learning (ML) algorithms, including logistic regression (LR), random forest (RF), support vector classification (SVC), extreme gradient boosting (XGBoost), gradient boosting decision tree (GBDT), decision tree, multilayer perceptron (MLP), linear discriminant analysis (LDA), adaptive boosting (AdaBoost), Gaussian naive Bayes (GaussianNB), and light gradient boosting machine (LightGBM), to predict breast cancer recurrence. The area under the curve (AUC), accuracy, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and F1 score were used to evaluate the performance of the prognostic model. Based on performance, the optimal ML was selected, and feature importance was ranked by Shapley Additive Explanation (SHAP) values. Compared to the other 10 algorithms, the results showed that the AdaBoost algorithm had the best prediction performance for successfully predicting breast cancer recurrence and was adopted in the establishment of the prediction model. Moreover, CA125, CEA, Fbg, and tumor diameter were found to be the most important features in our dataset to predict breast cancer recurrence. More importantly, our study is the first to use the SHAP method to improve the interpretability of clinicians to predict the recurrence model of breast cancer based on the AdaBoost algorithm. The AdaBoost algorithm offers a clinical decision support model and successfully identifies the recurrence of breast cancer.
Collapse
Affiliation(s)
- Duo Zuo
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Lexin Yang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Yu Jin
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Huan Qi
- China Mobile Group Tianjin Company Limited, Tianjin, 300308, China
| | - Yahui Liu
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
6
|
Bai S, Song D, Chen M, Lai X, Xu J, Dong F. The association between mammographic density and breast cancer molecular subtypes: a systematic review and meta-analysis. Clin Radiol 2023; 78:622-632. [PMID: 37230842 DOI: 10.1016/j.crad.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023]
Abstract
AIM To conduct a systematic review and meta-analysis to evaluate the whether high mammographic density (MD) is differentially associated with all subtypes of breast cancer. MATERIALS AND METHODS The PubMed, Cochrane Library, and Embase databases were searched systematically in October 2022 to include all studies that investigated the association between MD and breast cancer subtype. Aggregate data of 17,193 breast cancer cases from 23 studies were selected, including five cohort/case-control and 18 case-only studies. The relative risk (RR) of MD were combined using random/fixed effects models for case-control studies, and for case-only studies, relative risk ratios (RRRs) were a combination of luminal A, luminal B, and HER2-positive versus triple-negative tumours. RESULTS Women in the highest density category in case-control/cohort studies had a 2.24-fold (95% confidence interval [CI] 1.53, 3.28), 1.81-fold (95% CI 1.15, 2.85), 1.44-fold (95% CI 1.14, 1.81), and 1.59-fold (95% CI 0.89, 2.85) higher risk of triple-negative, HER-2 (human epidermal growth factor receptor 2) positive, luminal A, and luminal B breast cancer compared to women in the lowest density category. RRRs for breast tumours being luminal A, luminal B, and HER-2 positive versus triple-negative in case-only studies were 1.62 (95% CI 1.14, 2.31), 1.81 (95% CI 1.22, 2.71) and 2.58 (95% CI 1.63, 4.08), respectively, for BIRADS 4 versus BIRADS 1. CONCLUSION The evidence indicates MD is a potent risk factor for the majority of breast cancer subtypes to different degrees. Increased MD is more strongly linked to HER-2-positive cancers compared to other breast cancer subtypes. The application of MD as a subtype-specific risk marker may facilitate the creation of personalised risk prediction models and screening procedures.
Collapse
Affiliation(s)
- S Bai
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - D Song
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - M Chen
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - X Lai
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - J Xu
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - F Dong
- Department of Ultrasound, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
7
|
Mao X, Omeogu C, Karanth S, Joshi A, Meernik C, Wilson L, Clark A, Deveaux A, He C, Johnson T, Barton K, Kaplan S, Akinyemiju T. Association of reproductive risk factors and breast cancer molecular subtypes: a systematic review and meta-analysis. BMC Cancer 2023; 23:644. [PMID: 37430191 DOI: 10.1186/s12885-023-11049-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Associations between reproductive factors and breast cancer (BC) risk vary by molecular subtype (i.e., luminal A, luminal B, HER2, and triple negative/basal-like [TNBC]). In this systematic review and meta-analysis, we summarized the associations between reproductive factors and BC subtypes. METHODS Studies from 2000 to 2021 were included if BC subtype was examined in relation to one of 11 reproductive risk factors: age at menarche, age at menopause, age at first birth, menopausal status, parity, breastfeeding, oral contraceptive (OC) use, hormone replacement therapy (HRT), pregnancy, years since last birth and abortion. For each reproductive risk factor, BC subtype, and study design (case-control/cohort or case-case), random-effects models were used to estimate pooled relative risks and 95% confidence intervals. RESULTS A total of 75 studies met the inclusion criteria for systematic review. Among the case-control/cohort studies, later age at menarche and breastfeeding were consistently associated with decreased risk of BC across all subtypes, while later age at menopause, later age of first childbirth, and nulliparity/low parity were associated with increased risk of luminal A, luminal B, and HER2 subtypes. In the case-only analysis, compared to luminal A, postmenopausal status increased the risk of HER2 and TNBC. Associations were less consistent across subtypes for OC and HRT use. CONCLUSION Identifying common risk factors across BC subtypes can enhance the tailoring of prevention strategies, and risk stratification models can benefit from subtype specificity. Adding breastfeeding status to current BC risk prediction models can enhance predictive ability, given the consistency of the associations across subtypes.
Collapse
Affiliation(s)
- Xihua Mao
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Chioma Omeogu
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Shama Karanth
- UF Health Cancer Canter, University of Florida, Gainesville, FL, USA
| | - Ashwini Joshi
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Clare Meernik
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Lauren Wilson
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Amy Clark
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - April Deveaux
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA
| | - Chunyan He
- The Cancer Prevention and Control Research Program, University of Kentucky Markey Cancer Center, Lexington, KY, USA
| | - Tisha Johnson
- Department of Preventive Medicine and Environmental Health, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Karen Barton
- Duke University Medical Center Library & Archives, Duke University School of Medicine, Durham, NC, USA
| | - Samantha Kaplan
- Duke University Medical Center Library & Archives, Duke University School of Medicine, Durham, NC, USA
| | - Tomi Akinyemiju
- Department of Population Health Sciences, School of Medicine, Duke University, Durham, NC, USA.
- Duke Cancer Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Wang Z, Du X, Lian W, Chen J, Hong C, Li L, Chen D. A novel disulfidptosis-associated expression pattern in breast cancer based on machine learning. Front Genet 2023; 14:1193944. [PMID: 37456667 PMCID: PMC10343428 DOI: 10.3389/fgene.2023.1193944] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Background: Breast cancer (BC), the leading cause of cancer-related deaths among women, remains a serious threat to human health worldwide. The biological function and prognostic value of disulfidptosis as a novel strategy for BC treatment via induction of cell death remain unknown. Methods: Gene mutations and copy number variations (CNVs) in 10 disulfidptosis genes were evaluated. Differential expression, prognostic, and univariate Cox analyses were then performed for 10 genes, and BC-specific disulfidptosis-related genes (DRGs) were screened. Unsupervised consensus clustering was used to identify different expression clusters. In addition, we screened the differentially expressed genes (DEGs) among different expression clusters and identified hub genes. Moreover, the expression level of DEGs was detected by RT-qPCR in cellular level. Finally, we used the least absolute shrinkage and selection operator (LASSO) regression algorithm to establish a prognostic feature based on DEGs, and verified the accuracy and sensitivity of its prediction through prognostic analysis and subject operating characteristic curve analysis. The correlation of the signature with the tumor immune microenvironment and tumor stemness was analyzed. Results: Disulfidptosis genes showed significant CNVs. Two clusters were identified based on three DRGs (DNUFS1, LRPPRC, SLC7A11). Cluster A was found to be associated with better survival outcomes(p < 0.05) and higher levels of immune cell infiltration(p < 0.05). A prognostic signature of four disulfidptosis-related DEGs (KIF21A, APOD, ALOX15B, ELOVL2) was developed by LASSO regression analysis. The signature showed a good prediction ability. In addition, the prognostic signature in this study were strongly related to the tumor microenvironment (TME), tumor immune cell infiltration, tumor mutation burden (TMB), tumor stemness, and drug sensitivity. Conclusion: The prognostic signature we constructed based on disulfidptosis-DEGs is a good predictor of prognosis in patients with BC. This prognostic signature is closely related to TME, and its potential correlation provides clues for further studies.
Collapse
|
9
|
Assunção Ribeiro da Costa RE, Rocha de Oliveira FT, Nascimento Araújo AL, Vieira SC. Impact of Pathologic Complete Response on the Prognosis of Triple-Negative Breast Cancer Patients: A Cohort Study. Cureus 2023; 15:e37396. [PMID: 37182056 PMCID: PMC10171840 DOI: 10.7759/cureus.37396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is a molecular subtype in which estrogen (ER)/progesterone receptor (PR) and human epidermal growth receptor 2 (HER2) expression does not occur. The objective of this study was to analyze the impact of pathologic complete response (pCR) after neoadjuvant chemotherapy on the prognosis of triple-negative breast cancer (TNBC) patients. Methods This cohort study was conducted in a private-sector oncology clinic located in the city of Teresina, Brazil. Medical charts of 532 breast cancer patients treated from 2007 to 2020 were analyzed. Of these patients, 83 women with TNBC were selected (10 patients were excluded from the study). Univariate and multivariate analyses (Cox regression) were performed to evaluate the impact on patient survival, comparing patients with or without pCR. A significance level of 5% was set. Overall survival (OS) and disease-free survival (DFS) curves were constructed according to the Kaplan-Meier model. Results Angiolymphatic invasion and positive sentinel lymph node were associated with a lower OS and/or DFS in TNBC (p<0.05). The 10-year OS was 78% and 49%, and the 10-year DFS was 97% and 32% in patients with or without pCR, respectively. Conclusion pCR after neoadjuvant chemotherapy was associated with improvement in OS and DFS in TNBC patients.
Collapse
|
10
|
Zavala VA, Casavilca-Zambrano S, Navarro-Vásquez J, Tamayo LI, Castañeda CA, Valencia G, Morante Z, Calderón M, Abugattas JE, Gómez HL, Fuentes HA, Liendo-Picoaga R, Cotrina JM, Neciosup SP, Roque K, Vásquez J, Mas L, Gálvez-Nino M, Fejerman L, Vidaurre T. Breast cancer subtype and clinical characteristics in women from Peru. Front Oncol 2023; 13:938042. [PMID: 36925912 PMCID: PMC10013058 DOI: 10.3389/fonc.2023.938042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Breast cancer is a heterogeneous disease, and the distribution of the different subtypes varies by race/ethnic category in the United States and by country. Established breast cancer-associated factors impact subtype-specific risk; however, these included limited or no representation of Latin American diversity. To address this gap in knowledge, we report a description of demographic, reproductive, and lifestyle breast cancer-associated factors by age at diagnosis and disease subtype for The Peruvian Genetics and Genomics of Breast Cancer (PEGEN-BC) study. Methods The PEGEN-BC study is a hospital-based breast cancer cohort that includes 1943 patients diagnosed at the Instituto Nacional de Enfermedades Neoplásicas in Lima, Peru. Demographic and reproductive information, as well as lifestyle exposures, were collected with a questionnaire. Clinical data, including tumor Hormone Receptor (HR) status and Human Epidermal Growth Factor Receptor 2 (HER2) status, were abstracted from electronic medical records. Differences in proportions and mean values were tested using Chi-squared and one-way ANOVA tests, respectively. Multinomial logistic regression models were used for multivariate association analyses. Results The distribution of subtypes was 52% HR+HER2-, 19% HR+HER2+, 16% HR-HER2-, and 13% HR-HER2+. Indigenous American (IA) genetic ancestry was higher, and height was lower among individuals with the HR-HER2+ subtype (80% IA vs. 76% overall, p=0.007; 152 cm vs. 153 cm overall, p=0.032, respectively). In multivariate models, IA ancestry was associated with HR-HER2+ subtype (OR=1.38,95%CI=1.06-1.79, p=0.017) and parous women showed increased risk for HR-HER2+ (OR=2.7,95%CI=1.5-4.8, p<0.001) and HR-HER2- tumors (OR=2.4,95%CI=1.5-4.0, p<0.001) compared to nulliparous women. Multiple patient and tumor characteristics differed by age at diagnosis (<50 vs. >=50), including ancestry, region of residence, family history, height, BMI, breastfeeding, parity, and stage at diagnosis (p<0.02 for all variables). Discussion The characteristics of the PEGEN-BC study participants do not suggest heterogeneity by tumor subtype except for IA genetic ancestry proportion, which has been previously reported. Differences by age at diagnosis were apparent and concordant with what is known about pre- and post-menopausal-specific disease risk factors. Additional studies in Peru should be developed to further understand the main contributors to the specific age of onset and molecular disease subtypes in this population and develop population-appropriate predictive models for prevention.
Collapse
Affiliation(s)
- Valentina A. Zavala
- Department of Public Health Sciences, University of California, Davis, Davis, CA, United States
| | | | | | - Lizeth I. Tamayo
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, United States
| | - Carlos A. Castañeda
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Guillermo Valencia
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Zaida Morante
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Mónica Calderón
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Julio E. Abugattas
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Cirugía de Mamas y tumores Blandos, Lima, Peru
| | - Henry L. Gómez
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Hugo A. Fuentes
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | | | - Jose M. Cotrina
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Cirugía de Mamas y tumores Blandos, Lima, Peru
| | - Silvia P. Neciosup
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Katia Roque
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Jule Vásquez
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Luis Mas
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Marco Gálvez-Nino
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| | - Laura Fejerman
- Department of Public Health Sciences, University of California, Davis, Davis, CA, United States
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA, United States
| | - Tatiana Vidaurre
- Instituto Nacional de Enfermedades Neoplásicas, Departamento de Oncología Médica, Lima, Peru
| |
Collapse
|
11
|
Stordal B. Breastfeeding reduces the risk of breast cancer: A call for action in high-income countries with low rates of breastfeeding. Cancer Med 2023; 12:4616-4625. [PMID: 36164270 PMCID: PMC9972148 DOI: 10.1002/cam4.5288] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 11/11/2022] Open
Abstract
Women in the UK have a 15% lifetime risk of developing breast cancer. Like other high-income countries, women in the UK are having children later in life which increases their risk. The risk of breast cancer is reduced by 4.3% for every 12 months of breastfeeding, this is in addition to the 7.0% decrease in risk observed for each birth. Breastfeeding reduces the risk of Triple-Negative Breast Cancer (20%) and in carriers of BRCA1 mutations (22-55%). The mechanisms of reduced risk as a result of pregnancy are related to changes in RNA processing and cellular differentiation. The UK has a low rate of breastfeeding (81%) and this is contrasted to countries with higher (Sweden, Australia) and lower rates (Ireland). The low UK rate is in part due to a lack of experience in the population, todays grandmothers have less experience with breastfeeding (62%) than their daughters. An estimated 4.7% of breast cancer cases in the UK are caused by not breastfeeding. The UK only has 43% of maternity services with full Baby-Friendly accreditation which promotes compliance with the WHO 'Ten Steps to Successful Breast Feeding'. Legislation in the UK and Europe is far short of the WHO Guidance on restricting the advertising of formula milk. Expansion of the Baby-Friendly Hospital Initiative, stricter laws on the advertising of formula milk and legislation to support nursing mothers in the workplace have the potential to increase breastfeeding in the UK. Women with a family history of breast cancer should particularly be supported to breastfeed as a way of reducing their risk.
Collapse
Affiliation(s)
- Britta Stordal
- Department of Natural Sciences, Middlesex University London, London, UK
| |
Collapse
|
12
|
Xie C, Hu J, Hu Q, Jiang L, Chen W. Classification of the mitochondrial ribosomal protein-associated molecular subtypes and identified a serological diagnostic biomarker in hepatocellular carcinoma. Front Surg 2023; 9:1062659. [PMID: 36684217 PMCID: PMC9853988 DOI: 10.3389/fsurg.2022.1062659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Purpose The objective of this study was to sort out innovative molecular subtypes associated with mitochondrial ribosomal proteins (MRPs) to predict clinical therapy response and determine the presence of circulating markers in hepatocellular carcinoma (HCC) patients. Methods Using an unsupervised clustering method, we categorized the relative molecular subtypes of MRPs in HCC patients. The prognosis, biological properties, immune checkpoint inhibitor and chemotherapy response of the patients were clarified. A signature and nomogram were developed to evaluate the prognosis. Enzyme-linked immunosorbent assay (ELISA) measured serum mitochondrial ribosomal protein L9 (MRPL9) levels in liver disease patients and normal individuals. Receiver operating characteristic (ROC) curves were conducted to calculate the diagnostic effect. The Cell Counting Kit 8 was carried out to examine cell proliferation, and flow cytometry was used to investigate the cell cycle. Transwell assay was applied to investigate the potential of cell migration and invasion. Western blot detected corresponding changes of biological markers. Results Participants were classified into two subtypes according to MRPs expression levels, which were characterized by different prognoses, biological features, and marked differences in response to chemotherapy and immune checkpoint inhibitors. Serum MRPL9 was significantly higher in HCC patients than in normal individuals and the benign liver disease group. ROC curve analysis showed that MRPL9 was superior to AFP and Ferritin in differentiating HCC from healthy and benign patients, or alone. Overexpressed MRPL9 could enhance aggressiveness and facilitate the G1/S progression in HCC cells. Conclusion We constructed novel molecular subtypes based on MRPs expression in HCC patients, which provided valuable strategies for the prediction of prognosis and clinical personalized treatment. MRPL9 might act as a reliable circulating diagnostic biomarker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
| | | | | | | | - Weixian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Wang Z, Deng H, Jin Y, Luo M, Huang J, Wang J, Zhang K, Wang L, Zhou J. Circular RNAs: biology and clinical significance of breast cancer. RNA Biol 2023; 20:859-874. [PMID: 37882644 PMCID: PMC10730165 DOI: 10.1080/15476286.2023.2272468] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 10/27/2023] Open
Abstract
Circular RNAs (circRNAs) are novel noncoding RNAs with covalently closed-loop structures that can regulate eukaryotic gene expression. Due to their stable structure, circRNAs are widely distributed in the cytoplasm and have important biological functions, including as microRNA sponges, RNA-binding protein conjugates, transcription regulators, and translation templates. Breast cancer is among the most common malignant cancers diagnosed in women worldwide. Despite the development of comprehensive treatments, breast cancer still has high mortality rates. Recent studies have unmasked critical roles for circRNAs in breast cancer as regulators of tumour initiation, progression, and metastasis. Further, research has revealed that some circRNAs have the potential for use as diagnostic and prognostic biomarkers in clinical practice. Herein, we review the biogenesis and biological functions of circRNAs, as well as their roles in different breast cancer subtypes. Moreover, we provide a comprehensive summary of the clinical significance of circRNAs in breast cancer. CircRNAs are believed to be a hot focus in basic and clinical research of breast cancer, and innovative future research directions of circRNAs could be used as biomarkers, therapeutic targets, or novel drugs.Abbreviations: CeRNA: Competitive endogenous RNA; ciRNA: Circular intronic RNA; circRNA: Circular RNA; EIciRNA: Exon-intron circRNA; EMT: Epithelial-mesenchymal transition; IRES: Internal ribosome entry site; lncRNA: Long non-coding RNA; miRNA: MicroRNA; MRE: MiRNA response element; ncRNA: Non-coding RNA; RBP: RNA-binding protein; RNA-seq: RNA sequencing; RT-PCR: Reverse transcription-polymerase chain reaction.
Collapse
Affiliation(s)
- Zhanwei Wang
- Department of Breast Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Hao Deng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Jin
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Wang
- Department of Emergency, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Jung AY, Ahearn TU, Behrens S, Middha P, Bolla MK, Wang Q, Arndt V, Aronson KJ, Augustinsson A, Beane Freeman LE, Becher H, Brenner H, Canzian F, Carey LA, Czene K, Eliassen AH, Eriksson M, Evans DG, Figueroa JD, Fritschi L, Gabrielson M, Giles GG, Guénel P, Hadjisavvas A, Haiman CA, Håkansson N, Hall P, Hamann U, Hoppe R, Hopper JL, Howell A, Hunter DJ, Hüsing A, Kaaks R, Kosma VM, Koutros S, Kraft P, Lacey JV, Le Marchand L, Lissowska J, Loizidou MA, Mannermaa A, Maurer T, Murphy RA, Olshan AF, Olsson H, Patel AV, Perou CM, Rennert G, Shibli R, Shu XO, Southey MC, Stone J, Tamimi RM, Teras LR, Troester MA, Truong T, Vachon CM, Wang SS, Wolk A, Wu AH, Yang XR, Zheng W, Dunning AM, Pharoah PDP, Easton DF, Milne RL, Chatterjee N, Schmidt MK, García-Closas M, Chang-Claude J. Distinct Reproductive Risk Profiles for Intrinsic-Like Breast Cancer Subtypes: Pooled Analysis of Population-Based Studies. J Natl Cancer Inst 2022; 114:1706-1719. [PMID: 35723569 PMCID: PMC9949579 DOI: 10.1093/jnci/djac117] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/22/2022] [Accepted: 05/03/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Reproductive factors have been shown to be differentially associated with risk of estrogen receptor (ER)-positive and ER-negative breast cancer. However, their associations with intrinsic-like subtypes are less clear. METHODS Analyses included up to 23 353 cases and 71 072 controls pooled from 31 population-based case-control or cohort studies in the Breast Cancer Association Consortium across 16 countries on 4 continents. Polytomous logistic regression was used to estimate the association between reproductive factors and risk of breast cancer by intrinsic-like subtypes (luminal A-like, luminal B-like, luminal B-HER2-like, HER2-enriched-like, and triple-negative breast cancer) and by invasiveness. All statistical tests were 2-sided. RESULTS Compared with nulliparous women, parous women had a lower risk of luminal A-like, luminal B-like, luminal B-HER2-like, and HER2-enriched-like disease. This association was apparent only after approximately 10 years since last birth and became stronger with increasing time (odds ratio [OR] = 0.59, 95% confidence interval [CI] = 0.49 to 0.71; and OR = 0.36, 95% CI = 0.28 to 0.46 for multiparous women with luminal A-like tumors 20 to less than 25 years after last birth and 45 to less than 50 years after last birth, respectively). In contrast, parous women had a higher risk of triple-negative breast cancer right after their last birth (for multiparous women: OR = 3.12, 95% CI = 2.02 to 4.83) that was attenuated with time but persisted for decades (OR = 1.03, 95% CI = 0.79 to 1.34, for multiparous women 25 to less than 30 years after last birth). Older age at first birth (Pheterogeneity < .001 for triple-negative compared with luminal A-like breast cancer) and breastfeeding (Pheterogeneity < .001 for triple-negative compared with luminal A-like breast cancer) were associated with lower risk of triple-negative breast cancer but not with other disease subtypes. Younger age at menarche was associated with higher risk of all subtypes; older age at menopause was associated with higher risk of luminal A-like but not triple-negative breast cancer. Associations for in situ tumors were similar to luminal A-like. CONCLUSIONS This large and comprehensive study demonstrates a distinct reproductive risk factor profile for triple-negative breast cancer compared with other subtypes, with implications for the understanding of disease etiology and risk prediction.
Collapse
Affiliation(s)
- Audrey Y Jung
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Medical Center Hamburg-Eppendorf, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Thomas U Ahearn
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sabine Behrens
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pooja Middha
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manjeet K Bolla
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Qin Wang
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kristan J Aronson
- Department of Public Health Sciences, and Cancer Research Institute, Queen’s University, Kingston, ON, Canada
| | | | - Laura E Beane Freeman
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heiko Becher
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - CTS Consortium
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - D Gareth Evans
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Academic Health Science Centre, North West Genomics Laboratory Hub, Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jonine D Figueroa
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Usher Institute of Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Lin Fritschi
- School of Population Health, Curtin University, Perth, Western Australia, Australia
| | - Marike Gabrielson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Pascal Guénel
- Institut national de la santé et de la recherche médicale (INSERM), University Paris-Saclay, Center for Research in Epidemiology and Population Health (CESP), Team Exposome and Heredity, Villejuif, France
| | - Andreas Hadjisavvas
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Niclas Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Reiner Hoppe
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony Howell
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Anika Hüsing
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Veli-Matti Kosma
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Stella Koutros
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - James V Lacey
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie National Research Oncology Institute, Warsaw, Poland
| | - Maria A Loizidou
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- The Cyprus Institute of Neurology and Genetics, Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Arto Mannermaa
- Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Tabea Maurer
- Cancer Epidemiology Group, University Medical Center Hamburg-Eppendorf, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| | - Rachel A Murphy
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- BC Cancer Agency, Cancer Control Research, Vancouver, BC, Canada
| | - Andrew F Olshan
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Håkan Olsson
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alpa V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Charles M Perou
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gad Rennert
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Rana Shibli
- Carmel Medical Center and Technion Faculty of Medicine, Clalit National Cancer Control Center, Haifa, Israel
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer Stone
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Genetic Epidemiology Group, School of Population and Global Health, University of Western Australia, Perth, Western Australia, Australia
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Lauren R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Melissa A Troester
- Department of Epidemiology, Gillings School of Global Public Health and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thérèse Truong
- Institut national de la santé et de la recherche médicale (INSERM), University Paris-Saclay, Center for Research in Epidemiology and Population Health (CESP), Team Exposome and Heredity, Villejuif, France
| | - Celine M Vachon
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Sophia S Wang
- Department of Computational and Quantitative Medicine, City of Hope, Duarte, CA, USA
- City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA, USA
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiaohong R Yang
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Paul D P Pharoah
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Nilanjan Chatterjee
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Biostatistics, Bloomberg School of Public Health, John Hopkins University, Baltimore, MD, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Montserrat García-Closas
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer Epidemiology Group, University Medical Center Hamburg-Eppendorf, University Cancer Center Hamburg (UCCH), Hamburg, Germany
| |
Collapse
|
15
|
Urso L, Manco L, Castello A, Evangelista L, Guidi G, Castellani M, Florimonte L, Cittanti C, Turra A, Panareo S. PET-Derived Radiomics and Artificial Intelligence in Breast Cancer: A Systematic Review. Int J Mol Sci 2022; 23:13409. [PMID: 36362190 PMCID: PMC9653918 DOI: 10.3390/ijms232113409] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 08/13/2023] Open
Abstract
Breast cancer (BC) is a heterogeneous malignancy that still represents the second cause of cancer-related death among women worldwide. Due to the heterogeneity of BC, the correct identification of valuable biomarkers able to predict tumor biology and the best treatment approaches are still far from clear. Although molecular imaging with positron emission tomography/computed tomography (PET/CT) has improved the characterization of BC, these methods are not free from drawbacks. In recent years, radiomics and artificial intelligence (AI) have been playing an important role in the detection of several features normally unseen by the human eye in medical images. The present review provides a summary of the current status of radiomics and AI in different clinical settings of BC. A systematic search of PubMed, Web of Science and Scopus was conducted, including all articles published in English that explored radiomics and AI analyses of PET/CT images in BC. Several studies have demonstrated the potential role of such new features for the staging and prognosis as well as the assessment of biological characteristics. Radiomics and AI features appear to be promising in different clinical settings of BC, although larger prospective trials are needed to confirm and to standardize this evidence.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy
| | - Luigi Manco
- Medical Physics Unit, Azienda USL of Ferrara, 44124 Ferrara, Italy
- Medical Physics Unit, University Hospital of Ferrara, 44124 Cona, Italy
| | - Angelo Castello
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Laura Evangelista
- Department of Medicine DIMED, University of Padua, 35128 Padua, Italy
| | - Gabriele Guidi
- Medical Physics Unit, University Hospital of Modena, 41125 Modena, Italy
| | - Massimo Castellani
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Luigia Florimonte
- Nuclear Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Ferrara, Italy
- Nuclear Medicine Unit, Oncological Medical and Specialist Department, University Hospital of Ferrara, 44124 Cona, Italy
| | - Alessandro Turra
- Medical Physics Unit, University Hospital of Ferrara, 44124 Cona, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, 41125 Modena, Italy
| |
Collapse
|
16
|
An R, Yu H, Wang Y, Lu J, Gao Y, Xie X, Zhang J. Integrative analysis of plasma metabolomics and proteomics reveals the metabolic landscape of breast cancer. Cancer Metab 2022; 10:13. [PMID: 35978348 PMCID: PMC9382832 DOI: 10.1186/s40170-022-00289-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most commonly diagnosed cancer. Currently, mammography and breast ultrasonography are the main clinical screening methods for BC. Our study aimed to reveal the specific metabolic profiles of BC patients and explore the specific metabolic signatures in human plasma for BC diagnosis. METHODS This study enrolled 216 participants, including BC patients, benign patients, and healthy controls (HC) and formed two cohorts, one training cohort and one testing cohort. Plasma samples were collected from each participant and subjected to perform nontargeted metabolomics and proteomics. The metabolic signatures for BC diagnosis were identified through machine learning. RESULTS Metabolomics analysis revealed that BC patients showed a significant change of metabolic profiles compared to HC individuals. The alanine, aspartate and glutamate pathways, glutamine and glutamate metabolic pathways, and arginine biosynthesis pathways were the critical biological metabolic pathways in BC. Proteomics identified 29 upregulated and 2 downregulated proteins in BC. Our integrative analysis found that aspartate aminotransferase (GOT1), L-lactate dehydrogenase B chain (LDHB), glutathione synthetase (GSS), and glutathione peroxidase 3 (GPX3) were closely involved in these metabolic pathways. Support vector machine (SVM) demonstrated a predictive model with 47 metabolites, and this model achieved a high accuracy in BC prediction (AUC = 1). Besides, this panel of metabolites also showed a fairly high predictive power in the testing cohort between BC vs HC (AUC = 0.794), and benign vs HC (AUC = 0.879). CONCLUSIONS This study uncovered specific changes in the metabolic and proteomic profiling of breast cancer patients and identified a panel of 47 plasma metabolites, including sphingomyelins, glutamate, and cysteine could be potential diagnostic biomarkers for breast cancer.
Collapse
Affiliation(s)
- Rui An
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Haitao Yu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Jie Lu
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Xinyou Xie
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China
| | - Jun Zhang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China. .,Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, People's Republic of China.
| |
Collapse
|
17
|
Wang C, Qu Z, Chen L, Pan Y, Tang Y, Hu G, Gao R, Niu R, Liu Q, Gao X, Fang Y. Characterization of Lactate Metabolism Score in Breast and Thyroid Cancers to Assist Immunotherapy via Large-Scale Transcriptomic Data Analysis. Front Pharmacol 2022; 13:928419. [PMID: 35873566 PMCID: PMC9301074 DOI: 10.3389/fphar.2022.928419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2022] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) and thyroid cancer (TC) have the highest rate of incidence, especially in women. Previous studies have revealed that lactate provides energetic and anabolic support to cancer cells, thus serving as an important oncometabolite with both extracellular and intracellular signaling functions. However, the correlation of lactate metabolism scores with thyroid and breast cancer immune characteristics remains to be systematically analyzed. To investigate the role of lactate at the transcriptome level and its correlation with the clinical outcome of BC and TC, transcriptome data of 1,217 patients with breast cancer (BC) and 568 patients with thyroid cancer (TC) were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets with their corresponding clinical and somatic mutation data. The lactate metabolism score was calculated based on a single-sample gene set enrichment analysis (ssGSEA). The results showed that lactate metabolism-related genes and lactate metabolism scores was significantly associated with the survival of patients with BRCA and THCA. Notably, the lactate metabolism scores were strongly correlated with human leukocyte antigen (HLA) expression, tumor-infiltrating lymphocyte (TIL) infiltration, and interferon (IFN) response in BC and TC. Furthermore, the lactate metabolism score was an independent prognostic factor and could serve as a reliable predictor of overall survival, clinical characteristics, and immune cell infiltration, with the potential to be applied in immunotherapy or precise chemotherapy of BC and TC.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Cheng Wang, ; Yi Fang,
| | - Zheng Qu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Chen
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yunhao Pan
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing Tang
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guangfu Hu
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruijie Niu
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingyan Gao
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Cheng Wang, ; Yi Fang,
| |
Collapse
|
18
|
Zhao R, He W, Guan X. Benefits of post-mastectomy radiation for T4N0M0 breast cancer patients: A SEER Database study. Cancer Treat Res Commun 2022; 32:100586. [PMID: 35691256 DOI: 10.1016/j.ctarc.2022.100586] [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: 04/21/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE To evaluate the impact of post-mastectomy radiation therapy (PMRT) in breast cancer patients with stage T4N0M0. METHOD Patients diagnosed with breast cancer of T4N0M0 from Jan 2010 to Dec 2015 were extracted from SEER database. Multivariable logistic regression was used to analyze the factors associated with PMRT. Univariate and multivariate COX regression were used to analyze factors that might be associated with breast cancer specific survival (BCSS) and overall survival (OS) of patients. Kaplan-Meier analysis was performed to evaluate BCSS and OS in different subtypes of patients. RESULT Multivariable logistic regression showed that patients ≥ 71 years were less intend to have PMRT. Multivariate Cox analysis showed that married statue, HR+/Her- and HR+/Her+ subtypes, were independent predictors of improved BCSS and OS, and use of PMRT could improve BCSS and OS . PMRT was beneficial for OS in all subtypes breast cancer patients, but BCSS benefit was observed only in TNBC patients. CONCLUSION The use of PMRT improves OS in all T4N0M0 patients, but in terms of BCSS, it only beneficial for TNBC patients.
Collapse
Affiliation(s)
- Ruipeng Zhao
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing Jiangsu, China; Department of Thyroid and Breast Surgery,The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Weiwei He
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing Jiangsu, China
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Clinical College of Nanjing Medical University, Nanjing Jiangsu, China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Jiangsu, China.
| |
Collapse
|
19
|
Arias-Calvachi C, Blanco R, Calaf GM, Aguayo F. Epstein-Barr Virus Association with Breast Cancer: Evidence and Perspectives. BIOLOGY 2022; 11:799. [PMID: 35741320 PMCID: PMC9220417 DOI: 10.3390/biology11060799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/16/2022]
Abstract
Epstein-Barr virus (EBV) is an enveloped DNA virus that belongs to the gamma Herpesviridae family. The virus establishes a latent/lytic persistent infection, though it can be involved in cancer development in some subjects. Indeed, evidence supports an etiological role of EBV in undifferentiated nasopharyngeal carcinoma (NPC), a subset of gastric carcinomas and lymphomas. Additionally, EBV has been detected in breast carcinomas (BCs) although its role has not been established. In this review, we summarize epidemiological information regarding the presence of EBV in BC and we propose mechanistic models. However, additional epidemiological and experimental evidence is warranted to confirm these models.
Collapse
Affiliation(s)
- Claudia Arias-Calvachi
- Programa de Virología, Laboratorio de Oncovirología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (C.A.-C.); (R.B.)
| | - Rancés Blanco
- Programa de Virología, Laboratorio de Oncovirología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile; (C.A.-C.); (R.B.)
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | | |
Collapse
|
20
|
de Almeida LM, Cortés S, Vilensky M, Valenzuela O, Cortes-Sanabria L, de Souza M, Barbeito RA, Abdelhay E, Artagaveytia N, Daneri-Navarro A, Llera AS, Müller B, Podhajcer OL, Velazquez C, Alcoba E, Alonso I, Bravo AI, Camejo N, Carraro DM, Castro M, Cataldi S, Cayota A, Cerda M, Colombo A, Crocamo S, Del Toro-Arreola A, Delgadillo-Cristerna R, Delgado L, Breitenbach MD, Fernández E, Fernández J, Fernández W, Franco-Topete RA, Gaete F, Gómez J, Gonzalez-Ramirez LP, Guerrero M, Gutierrez-Rubio SA, Jalfin B, Lopez-Vazquez A, Loria D, Míguez S, Moran-Mendoza ADJ, Morgan-Villela G, Mussetti C, Nagai MA, Oceguera-Villanueva A, Reis RM, Retamales J, Rodriguez R, Rosales C, Salas-Gonzalez E, Segovia L, Sendoya JM, Silva-Garcia AA, Viña S, Zagame L, Jones B, Szklo M. Socioeconomic, Clinical, and Molecular Features of Breast Cancer Influence Overall Survival of Latin American Women. Front Oncol 2022; 12:845527. [PMID: 35530311 PMCID: PMC9071365 DOI: 10.3389/fonc.2022.845527] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular profile of breast cancer in Latin-American women was studied in five countries: Argentina, Brazil, Chile, Mexico, and Uruguay. Data about socioeconomic characteristics, risk factors, prognostic factors, and molecular subtypes were described, and the 60-month overall cumulative survival probabilities (OS) were estimated. From 2011 to 2013, 1,300 eligible Latin-American women 18 years or older, with a diagnosis of breast cancer in clinical stage II or III, and performance status ≦̸1 were invited to participate in a prospective cohort study. Face-to-face interviews were conducted, and clinical and outcome data, including death, were extracted from medical records. Unadjusted associations were evaluated by Chi-squared and Fisher's exact tests and the OS by Kaplan-Meier method. Log-rank test was used to determine differences between cumulative probability curves. Multivariable adjustment was carried out by entering potential confounders in the Cox regression model. The OS at 60 months was 83.9%. Multivariable-adjusted death hazard differences were found for women living in Argentina (2.27), Chile (1.95), and Uruguay (2.42) compared with Mexican women, for older (≥60 years) (1.84) compared with younger (≤40 years) women, for basal-like subtype (5.8), luminal B (2.43), and HER2-enriched (2.52) compared with luminal A subtype, and for tumor clinical stages IIB (1.91), IIIA (3.54), and IIIB (3.94) compared with stage IIA women. OS was associated with country of residence, PAM50 intrinsic subtype, age, and tumor stage at diagnosis. While the latter is known to be influenced by access to care, including cancer screening, timely diagnosis and treatment, including access to more effective treatment protocols, it may also influence epigenetic changes that, potentially, impact molecular subtypes. Data derived from heretofore understudied populations with unique geographic ancestry and sociocultural experiences are critical to furthering our understanding of this complexity.
Collapse
Affiliation(s)
| | - Sandra Cortés
- Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marta Vilensky
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | | | | | | | - Nora Artagaveytia
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | - Andrea S Llera
- Fundación Instituto Leloir, CONICET, Buenos Aires, Argentina
| | | | | | | | - Elsa Alcoba
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | - Isabel Alonso
- Centro Hospitalario Pereira Rossell, Montevideo, Uruguay
| | - Alicia I Bravo
- Hospital Regional de Agudos Eva Perón, Buenos Aires, Argentina
| | - Natalia Camejo
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | - Mónica Castro
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | - Lucia Delgado
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | - Elmer Fernández
- Universidad Católica de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunologia-CONICET, Córdoba, Argentina
| | | | | | | | | | - Jorge Gómez
- Texas A&M University, Houston, TX, United States
| | | | | | | | - Beatriz Jalfin
- Hospital Regional de Agudos Eva Perón, Buenos Aires, Argentina
| | | | - Dora Loria
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | - Silvia Míguez
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | | | | | | | | | | | - Rui M Reis
- Hospital de Câncer de Barretos, Barretos, Brazil
| | - Javier Retamales
- Grupo Oncológico Cooperativo Chileno de Investigación, Santiago, Chile
| | | | - Cristina Rosales
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | | | | | - Juan M Sendoya
- Fundación Instituto Leloir, CONICET, Buenos Aires, Argentina
| | - Aida A Silva-Garcia
- OPD Hospital Civil de Guadalajara, Universidad de Guadalajara, Guadalajara, Mexico
| | - Stella Viña
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | - Livia Zagame
- Instituto Jalisciense de Cancerologia, Guadalajara, Mexico
| | - Beth Jones
- Yale School of Public Health, Yale University, New Heaven, CT, United States
| | - Moysés Szklo
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | | |
Collapse
|
21
|
Yang Y, Li Z, Zhong Q, Zhao L, Wang Y, Chi H. Identification and validation of a novel prognostic signature based on transcription factors in breast cancer by bioinformatics analysis. Gland Surg 2022; 11:892-912. [PMID: 35694087 PMCID: PMC9177273 DOI: 10.21037/gs-22-267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/18/2022] [Indexed: 08/20/2023]
Abstract
BACKGROUND Breast cancer (BRCA) is the leading cause of cancer mortality among women, and it is associated with many tumor suppressors and oncogenes. There is increasing evidence that transcription factors (TFs) play vital roles in human malignancies, but TFs-based biomarkers for BRCA prognosis were still rare and necessary. This study sought to develop and validate a prognostic model based on TFs for BRCA patients. METHODS Differentially expressed TFs were screened from 1,109 BRCA and 113 non-tumor samples downloaded from The Cancer Genome Atlas (TCGA). Univariate Cox regression analysis was used to identify TFs associated with overall survival (OS) of BRCA, and multivariate Cox regression analysis was performed to establish the optimal risk model. The predictive value of the TF model was established using TCGA database and validated using a Gene Expression Omnibus (GEO) data set (GSE20685). A gene set enrichment analysis was conducted to identify the enriched signaling pathways in high-risk and low-risk BRCA patients. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the TF target genes were also conducted separately. RESULTS A total of 394 differentially expressed TFs were screened. A 9-TF prognostic model, comprising PAX7, POU3F2, ZIC2, WT1, ALX4, FOXJ1, SPIB, LEF1 and NFE2, was constructed and validated. Compared to those in the low-risk group, patients in the high-risk group had worse clinical outcomes (P<0.001). The areas under the curve of the prognostic model for 5-year OS were 0.722 in the training cohort and 0.651 in the testing cohort. Additionally, the risk score was an independent prediction indicator for BRCA patients both in the training cohort (HR =1.757, P<0.001) and testing cohort (HR =1.401, P=0.001). It was associated with various cancer signaling pathways. Ultimately, 9 overlapping target genes were predicted by 3 prediction nomograms. The GO and KEGG enrichment analyses of these target genes suggested that the TFs in the model may regulate the activation of some classical tumor signaling pathways to control the progression of BRCA through these target genes. CONCLUSIONS Our study developed and validated a novel prognostic TF model that can effectively predict 5-year OS for BRCA patients.
Collapse
Affiliation(s)
- Yingmei Yang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Zhaoyun Li
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Qianyi Zhong
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lei Zhao
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Hongbo Chi
- Department of Clinical Laboratory Medicine, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
22
|
Li R, Ugai T, Xu L, Zucker D, Ogino S, Wang M. Utility of Continuous Disease Subtyping Systems for Improved Evaluation of Etiologic Heterogeneity. Cancers (Basel) 2022; 14:1811. [PMID: 35406583 PMCID: PMC8997600 DOI: 10.3390/cancers14071811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Molecular pathologic diagnosis is important in clinical (oncology) practice. Integration of molecular pathology into epidemiological methods (i.e., molecular pathological epidemiology) allows for investigating the distinct etiology of disease subtypes based on biomarker analyses, thereby contributing to precision medicine and prevention. However, existing approaches for investigating etiological heterogeneity deal with categorical subtypes. We aimed to fully leverage continuous measures available in most biomarker readouts (gene/protein expression levels, signaling pathway activation, immune cell counts, microbiome/microbial abundance in tumor microenvironment, etc.). We present a cause-specific Cox proportional hazards regression model for evaluating how the exposure-disease subtype association changes across continuous subtyping biomarker levels. Utilizing two longitudinal observational prospective cohort studies, we investigated how the association of alcohol intake (a risk factor) with colorectal cancer incidence differed across the continuous values of tumor epigenetic DNA methylation at long interspersed nucleotide element-1 (LINE-1). The heterogeneous alcohol effect was modeled using different functions of the LINE-1 marker to demonstrate the method's flexibility. This real-world proof-of-principle computational application demonstrates how the new method enables visualizing the trend of the exposure effect over continuous marker levels. The utilization of continuous biomarker data without categorization for investigating etiological heterogeneity can advance our understanding of biological and pathogenic mechanisms.
Collapse
Affiliation(s)
- Ruitong Li
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (R.L.); (S.O.)
| | - Tomotaka Ugai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lantian Xu
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - David Zucker
- Department of Statistics and Data Science, Hebrew University, Jerusalem 91905, Israel;
| | - Shuji Ogino
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; (R.L.); (S.O.)
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, MA 02115, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Sun H, Dai J, Chen M, Chen Q, Xie Q, Zhang W, Li G, Yan M. miR-139-5p Was Identified as Biomarker of Different Molecular Subtypes of Breast Carcinoma. Front Oncol 2022; 12:857714. [PMID: 35433464 PMCID: PMC9009410 DOI: 10.3389/fonc.2022.857714] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Located on chromosome 11q13.4, miR-139-5p has been confirmed by several studies as a possible attractive biomarker for cancer, including breast cancer, but its mechanism of correlation in different molecular subtypes of breast cancer has not been reported. In this study, comprehensive bioinformatics analysis was used to evaluate the expression of miR-139-5p in different molecular subtypes of breast cancer (luminal A, luminal B, HER2-enriched, and basal-like). The target genes of miR-139-5p were predicted by using an online database TargetScan and miRDB, and three key genes, FBN2, MEX3A, and TPD52, were screened in combination with differentially expressed genes in different molecular subtypes of breast cancer. The expression of the three genes was verified separately, and the genes were analyzed for pathway and functional enrichment. Bone marrow mesenchymal stem cells (BMSC) are another kind of highly plastic cell population existing in bone marrow besides hematopoietic stem cells. BMSC can affect the proliferation and migration of cancer cells, promote the metastasis and development of cancer, and regulate the tumor microenvironment by secreting exosome mirnas, thus affecting the malignant biological behavior of tumor cells. Finally, human bone marrow mesenchymal stem cells exosomes were obtained by ultracentrifugation, and the morphology of exosomes was observed by transmission electron microscopy. The expression of miR-139-5p in normal breast cells MCF-10A, human breast cancer cell line MDA-MB-231 cells, and BMSCs-derived exosomes were compared; the exosomes and MDA-MB-231 cells were co-cultured to observe their effects on the proliferation of the MDA-MB-231 cells. Human bone marrow mesenchymal stem cell-derived exosomes inhibited the growth of breast cancer cells and promoted the expression of FBN2, MEX3A, and TPD52 by transporting miR-139-5p.
Collapse
Affiliation(s)
- Haohang Sun
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Ji Dai
- Department of General Surgery, Zhenhai District People’s Hospital, Ningbo, China
| | - Mengze Chen
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Qi Chen
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Qiong Xie
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Weijun Zhang
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Guoqing Li
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
| | - Meidi Yan
- General Surgery I (Thyroid, Breast, Vascular, Hernia Surgery), General Hospital of Zhenhai District People’s Hospital Medical Group, Ningbo, China
- *Correspondence: Meidi Yan,
| |
Collapse
|
24
|
Yang X, Weng X, Yang Y, Jiang Z. Pyroptosis-Related lncRNAs Predict the Prognosis and Immune Response in Patients With Breast Cancer. Front Genet 2022; 12:792106. [PMID: 35360412 PMCID: PMC8963933 DOI: 10.3389/fgene.2021.792106] [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: 10/09/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related death in women worldwide. Pyroptosis and long noncoding RNAs (lncRNAs) have been demonstrated to play vital roles in the tumorigenesis and development of BC. However, the clinical significance of pyroptosis-related lncRNAs in BC remains unclear. Methods: Using the mRNA and lncRNA profiles of BC obtained from TCGA dataset, a risk model based on the pyroptosis-related lncRNAs for prognosis was constructed using univariate and multivariate Cox regression model, and least absolute shrinkage and selection operator. Patients were divided into high- and low-risk groups based on the risk model, and the prognosis value and immune response in different risk groups were analyzed. Furthermore, functional enrichment annotation, therapeutic signature, and tumor mutation burden were performed to evaluate the risk model we established. Moreover, the expression level and clinical significance of the selected pyroptosis-related lncRNAs were further validated in BC samples. Results: 3,364 pyroptosis-related lncRNAs were identified using Pearson’s correlation analysis. The risk model we constructed comprised 10 pyroptosis-related lncRNAs, which was identified as an independent predictor of overall survival (OS) in BC. The nomogram we constructed based on the clinicopathologic features and risk model yielded favorable performance for prognosis prediction in BC. In terms of immune response and mutation status, patients in the low-risk group had a higher expression of immune checkpoint markers and exhibited higher fractions of activated immune cells, while the high-risk group had a highly percentage of TMB. Further analyses in our cohort BC samples found that RP11-459E5.1 was significantly upregulated, while RP11-1070N10.3 and RP11-817J15.3 were downregulated and significantly associated with worse OS. Conclusion: The risk model based on the pyroptosis-related lncRNAs we established may be a promising tool for predicting the prognosis and personalized therapeutic response in BC patients.
Collapse
Affiliation(s)
- Xia Yang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Weng
- Department of Pathology, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Yajie Yang
- Department of Pathology, Shenzhen Second People’s Hospital, Shenzhen, China
| | - ZhiNong Jiang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: ZhiNong Jiang,
| |
Collapse
|
25
|
Zhang J, Zhang L, Wang J, Zhao J, Zhao X, Zhang C, Han P, Geng C. Long non-coding RNA linc00921 suppresses tumorigenesis and epithelial-to-mesenchymal transition of triple-negative breast cancer via targeting miR-9-5p/LZTS2 axis. Hum Cell 2022; 35:909-923. [PMID: 35179718 PMCID: PMC9013323 DOI: 10.1007/s13577-022-00685-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 02/05/2022] [Indexed: 11/25/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Dysregulation of long non-coding RNAs (lncRNAs) plays crucial roles in the initiation and progression of TNBC. In this study, we analyzed public GEO profiles to verify the key lncRNAs in TNBC. Linc00921 was selected for further study. Low expression of linc00921 was observed in 49 of 95 TNBC tissues. Low expression of linc00921 was correlated with poor postoperative disease-free survival (DFS) and overall survival (OS) of TNBC patients. Overexpression of linc00921 with lentivirus suppressed the proliferation, migration and invasion of TNBC cells. A luciferase reporter assay showed that linc00921 could sponge miR-9-5p in TNBC. Moreover, linc00921 and miR-9-5p occupied the same Argonaute-2 (Ago2) protein in TNBC cells. Leucine zipper tumor suppressor 2 (LZTS2) was recognized as a target gene of miR-9-5p, and thereby a linc00921/miR-9-5p/LZTS2 axis was identified in TNBC cells. Overexpression of linc00921 promoted nuclear export of β-catenin, neutralized its function, and subsequently promoted epithelial-to-mesenchymal transition (EMT) in TNBC. A xenograft tumor mouse model showed that the miR-9-5p inhibitor upregulates LZTS2 expression and induce nuclear export of β-catenin in TNBC. Thus, linc00921 upregulates LZTS2 by sponging miR-9-5p to suppress tumorigenesis and EMT of TNBC. Linc00921/miR-9-5p/LZTS2 axis may be a novel biomarker and therapeutic target for TNBC patients.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Plastic Surgery, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Lina Zhang
- Breast Disease Diagnostic and Therapeutic Center, Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050035, Hebei, China
| | - Jianlong Wang
- Department of Minimally Invasive Surgery, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Jing Zhao
- Department of Anus and Intestine Surgery, Second Affiliated Hospital of Hebei Medical University, ShijiazhuangHebei, 050000, China
| | - Xuelian Zhao
- Department of Plastic Surgery, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Chunli Zhang
- Department of Plastic Surgery, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Peng Han
- Department of Plastic Surgery, Second Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Cuizhi Geng
- Breast Disease Diagnostic and Therapeutic Center, Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, 050035, Hebei, China.
| |
Collapse
|
26
|
Michaels EK, Canchola AJ, Beyer KMM, Zhou Y, Shariff-Marco S, Gomez SL. Home mortgage discrimination and incidence of triple-negative and Luminal A breast cancer among non-Hispanic Black and non-Hispanic White females in California, 2006-2015. Cancer Causes Control 2022; 33:727-735. [PMID: 35113296 PMCID: PMC9010391 DOI: 10.1007/s10552-022-01557-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 01/24/2022] [Indexed: 11/24/2022]
Abstract
Purpose In the United States, Black females are burdened by more aggressive subtypes and increased mortality from breast cancer compared to non-Hispanic (NH) White females. Institutional racism may contribute to these inequities. We aimed to characterize the association between home mortgage discrimination, a novel measure of institutional racism, and incidence of Luminal A and triple-negative breast cancer (TNBC) subtypes among NH Black and NH White females in California metropolitan areas. Methods We merged data from the California Cancer Registry on females aged 20 + diagnosed with primary invasive breast cancer between 2006 and 2015 with a census tract-level index of home mortgage lending bias measuring the odds of mortgage loan denial for Black versus White applicants, generated from the 2007–2013 Home Mortgage Disclosure Act database. Poisson regression estimated cross-sectional associations of census tract-level racial bias in mortgage lending with race/ethnicity- and Luminal A and TNBC-specific incidence rate ratios, adjusting for neighborhood confounders. Results We identified n = 102,853 cases of Luminal A and n = 15,528 cases of TNBC over the study period. Compared to NH Whites, NH Black females had higher rates of TNBC, lower rates of Luminal A breast cancer, and lived in census tracts with less racial bias in home mortgage lending. There was no evidence of association between neighborhood racial bias in mortgage lending at the time of diagnosis and either subtype among either racial/ethnic group. Conclusion Future research should incorporate residential history data with measures of institutional racism to improve estimation and inform policy interventions.
Collapse
Affiliation(s)
- Eli K Michaels
- Division of Epidemiology, Berkeley School of Public Health, University of California, 2121 Berkeley Way #5302, Berkeley, CA, 94720-7360, USA.
| | - Alison J Canchola
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.,Greater Bay Area Cancer Registry, San Francisco, CA, USA
| | - Kirsten M M Beyer
- Division of Epidemiology and Social Sciences, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhong Zhou
- Division of Epidemiology and Social Sciences, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Salma Shariff-Marco
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.,Greater Bay Area Cancer Registry, San Francisco, CA, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Scarlett L Gomez
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.,Greater Bay Area Cancer Registry, San Francisco, CA, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
27
|
Qi TF, Miao W, Wang Y. Targeted Profiling of Epitranscriptomic Reader, Writer, and Eraser Proteins Accompanied with Radioresistance in Breast Cancer Cells. Anal Chem 2022; 94:1525-1530. [PMID: 35021009 PMCID: PMC8792366 DOI: 10.1021/acs.analchem.1c05441] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Epitranscriptomic reader, writer, and eraser (RWE) proteins recognize, install, and remove modified nucleosides in RNA, which are known to play crucial roles in RNA processing, splicing, and stability. Here, we established a liquid chromatography-parallel-reaction monitoring (LC-PRM) method for high-throughput profiling of a total of 152 epitranscriptomic RWE proteins. We also applied the LC-PRM method, in conjunction with stable isotope labeling by amino acids in cell culture (SILAC), to quantify these proteins in two pairs of matched parental/radioresistant breast cancer cells (i.e., MDA-MB-231 and MCF-7 cells and their corresponding radioresistant C5 and C6 clones), with the goal of assessing the roles of these proteins in radioresistance. We found that eight epitranscriptomic RWE proteins were commonly altered by over 1.5-fold in the two pairs of breast cancer cells. Among them, TRMT1 (an m2,2G writer) may play a role in promoting breast cancer radioresistance due to its clinical relevance and its correlation with DNA repair gene sets. To our knowledge, this is the first report of a targeted proteomic method for comprehensive quantifications of epitranscriptomic RWE proteins. We envision that the LC-PRM method is applicable for studying the roles of these proteins in the metastatic transformation of cancer and therapeutic resistance of other types of cancer in the future.
Collapse
|
28
|
Wei X, Deng W, Dong Z, Luo Y, Hu X, Zhang J, Xie Z, Zheng T, Tan Y, Tang Z, Li H, Na N. Redox Metabolism-Associated Molecular Classification of Clear Cell Renal Cell Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5831247. [PMID: 35096270 PMCID: PMC8799361 DOI: 10.1155/2022/5831247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/17/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma. Redox metabolism has been recognized as the hallmark of cancer. But the concrete role of redox-related genes in patient stratification of ccRCC remains unknown. Herein, we aimed to characterize the molecular features of ccRCC based on the redox gene expression profiles from The Cancer Genome Atlas. Differentially expressed redox genes (DERGs) and vital genes in metabolism regulation were identified and analyzed in the ccRCC. Consensus clustering was performed to divide patients into three clusters (C1, C2, and C3) based on 139 redox genes with median FPKM value > 1. We analyzed the correlation of clusters with clinicopathological characteristics, immune infiltration, gene mutation, and response to immunotherapy. Subclass C1 was metabolic active with moderate prognosis and associated with glucose, lipid, and protein metabolism. C2 had intermediate metabolic activity with worse prognosis and correlated with more tumor mutation burden, neoantigen, and aneuploidy, indicating possible drug sensitivities towards immune checkpoint inhibitors. Metabolic exhausted subtype C3 showed high cytolytic activity score, suggesting better prognosis than C1 and C2. Moreover, the qRT-PCR was performed to verify the expression of downregulated DERGs including ALDH6A1, ALDH1L1, GLRX5, ALDH1A3, and GSTM3, and upregulated SHMT1 in ccRCC. Overall, our study provides an insight into the characteristics of molecular classification of ccRCC patients based on redox genes, thereby deepening the understanding of heterogeneity of ccRCC and allowing prediction of prognosis of ccRCC patients.
Collapse
Affiliation(s)
- Xiangling Wei
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Weiming Deng
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- The First Affiliated Hospital, Department of Urology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhanwen Dong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - You Luo
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiao Hu
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jinhua Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Zhenwei Xie
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tong Zheng
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yuqin Tan
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Zuofu Tang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Heng Li
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
29
|
Choi J, Jia G, Wen W, Tao R, Long J, Shu XO, Zheng W. Associations of genetic susceptibility to 16 cancers with risk of breast cancer overall and by intrinsic subtypes. HGG ADVANCES 2022; 3:100077. [PMID: 35047862 PMCID: PMC8756518 DOI: 10.1016/j.xhgg.2021.100077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/06/2021] [Indexed: 11/24/2022] Open
Abstract
Certain genetic variants are associated with risks of multiple cancers. We investigated breast cancer risk with overall genetic susceptibility to each of 16 other cancers. We constructed polygenic risk scores (PRS) for 16 cancers using risk variants identified by genome-wide association studies. We evaluated the associations of these PRSs with breast cancer risk (overall and by subtypes) using Breast Cancer Association Consortium data, including 106,278 cases and 91,477 controls of European ancestry. Odds ratios (OR) and 95% confidence intervals (CIs) were estimated to measure the association of each PRS with breast cancer risk. Data from the UK Biobank, including 4,337 cases and 209,983 non-cases, were used to replicate the findings. A 5%–8% significantly elevated risk of overall breast cancer was associated with per unit increase of the PRS for glioma and cancers of the corpus uteri, stomach, or colorectum. Analyses by subtype revealed that the PRS for corpus uteri cancer (OR = 1.09; 95% CI, 1.03–1.15) and stomach cancer (OR = 1.07; 95% CI, 1.03–1.12) were associated with estrogen receptor-positive breast cancer, while ovarian cancer PRS was associated with triple-negative breast cancer (OR = 1.25; 95% CI, 1.01–1.55). UK Biobank data supported the positive associations of overall breast cancer risk with PRS for melanoma and cancers of the stomach, colorectum, and ovary. Our study provides strong evidence for shared genetic susceptibility of breast cancer with several other cancers. Results from our study help uncover the genetic basis for breast and other cancers and identify individuals at high risk for multiple cancers.
Collapse
Affiliation(s)
- Jungyoon Choi
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| |
Collapse
|
30
|
Hurson AN, Pal Choudhury P, Gao C, Hüsing A, Eriksson M, Shi M, Jones ME, Evans DGR, Milne RL, Gaudet MM, Vachon CM, Chasman DI, Easton DF, Schmidt MK, Kraft P, Garcia-Closas M, Chatterjee N. Prospective evaluation of a breast-cancer risk model integrating classical risk factors and polygenic risk in 15 cohorts from six countries. Int J Epidemiol 2022; 50:1897-1911. [PMID: 34999890 PMCID: PMC8743128 DOI: 10.1093/ije/dyab036] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Rigorous evaluation of the calibration and discrimination of breast-cancer risk-prediction models in prospective cohorts is critical for applications under clinical guidelines. We comprehensively evaluated an integrated model incorporating classical risk factors and a 313-variant polygenic risk score (PRS) to predict breast-cancer risk. METHODS Fifteen prospective cohorts from six countries with 239 340 women (7646 incident breast-cancer cases) of European ancestry aged 19-75 years were included. Calibration of 5-year risk was assessed by comparing expected and observed proportions of cases overall and within risk categories. Risk stratification for women of European ancestry aged 50-70 years in those countries was evaluated by the proportion of women and future cases crossing clinically relevant risk thresholds. RESULTS Among women <50 years old, the median (range) expected-to-observed ratio for the integrated model across 15 cohorts was 0.9 (0.7-1.0) overall and 0.9 (0.7-1.4) at the highest-risk decile; among women ≥50 years old, these were 1.0 (0.7-1.3) and 1.2 (0.7-1.6), respectively. The proportion of women identified above a 3% 5-year risk threshold (used for recommending risk-reducing medications in the USA) ranged from 7.0% in Germany (∼841 000 of 12 million) to 17.7% in the USA (∼5.3 of 30 million). At this threshold, 14.7% of US women were reclassified by adding the PRS to classical risk factors, with identification of 12.2% of additional future cases. CONCLUSION Integrating a 313-variant PRS with classical risk factors can improve the identification of European-ancestry women at elevated risk who could benefit from targeted risk-reducing strategies under current clinical guidelines.
Collapse
Affiliation(s)
- Amber N Hurson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Parichoy Pal Choudhury
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Gao
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anika Hüsing
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska Univ Hospital, Stockholm, Sweden
| | - Min Shi
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Michael E Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - D Gareth R Evans
- Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Manchester NIHR Biomedical Research Centre, Manchester University Hospitals NHS, Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Mia M Gaudet
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA, USA
| | - Celine M Vachon
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Nilanjan Chatterjee
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
31
|
Han BY, Xu XL, Zhu XZ, Han XC, Hu X, Ling H. Clinicopathological Characteristics and Survival Outcomes of Mammary Paget’s Disease: A Retrospective Study Based on a Chinese Population. Cancer Manag Res 2022; 14:237-247. [PMID: 35125891 PMCID: PMC8807864 DOI: 10.2147/cmar.s338788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/28/2021] [Indexed: 11/23/2022] Open
Abstract
Background Mammary Paget’s disease (PD) is a rare type of breast cancer. Most cases of PD are presented with underlying ductal carcinoma in situ (DCIS) or invasive breast carcinoma (IDC). This study aimed to investigate the clinicopathological characteristics and survival outcomes of PD patients. Materials and Methods A total of 406 patients diagnosed with PD with IDC/DCIS at Fudan University Shanghai Cancer Center (FUSCC) were recruited as the PD group, 1218 patients diagnosed with IDC/DCIS alone during the same period were selected as the non-PD group, and the clinicopathological results of these two groups were compared. The Surveillance, Epidemiology, and End Results (SEER) database was used to investigate the clinicopathological features between PD and non-PD patients for validation. Results Compared with the non-PD group, the PD group was much more likely to have larger (≥2 cm: 43.1% vs 35.5%, P < 0.001), less hormone receptor (HR)-positive (68.5% vs 26.6%, P < 0.001), more human epidermal growth factor receptor-2 (HER-2)-positive (70.7% vs 27.5%, P < 0.001) and higher Ki-67 proportion (51.5% vs 42.5%, P < 0.001) tumors. The HER-2 overexpression subtype accounted for the largest proportion in the PD-IDC group and the lowest proportion in the non-PD-IDC group (54% vs 8%, P < 0.01). Moreover, the PD group had significantly worse disease-free survival (DFS) than the non-PD group (5-year DFS: 91.8% vs 97.3%, P = 0.001), and the SEER database showed a similar trend. Univariate and multivariate Cox regression analyses demonstrated that PD was an independent poor-risk factor. Our matched study showed that the PD group had worse survival than the non-PD group after excluding age, HR, HER-2, tumor size and lymph node status. Conclusion PD with IDC/DCIS is associated with more aggressive tumor characteristics and worse survival outcomes. More than half of PD breast cancers are HER-2 overexpression subtype. PD is an independent poor-risk factor for breast cancer survival.
Collapse
Affiliation(s)
- Bo-Yue Han
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiao-Li Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiu-Zhi Zhu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiang-Chen Han
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Correspondence: Xin Hu; Hong Ling, Tel +86 18017317652; +86 18017312656, Email ;
| | - Hong Ling
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
32
|
Bui OT, Tran HT, Nguyen SM, Dao TV, Bui QV, Pham AT, Shrubsole MJ, Cai Q, Ye F, Zheng W, Luu HN, Tran TV, Shu XO. Menstrual and Reproductive Factors in Association With Breast Cancer Risk in Vietnamese Women: A Case-Control Study. Cancer Control 2022; 29:10732748221140206. [PMID: 36373740 PMCID: PMC9663618 DOI: 10.1177/10732748221140206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Though menstrual and reproductive factors have been associated with the risk of breast cancer in many populations, very few studies have been conducted among Vietnamese women. This study aimed to assess the association between menstrual and reproductive factors and the risk of breast cancer in Vietnamese women. METHODS A retrospective case-control study of 490 breast cancer cases and 468 controls was conducted in Northern Vietnam. Unconditional logistic regression models adjusting for confounders were used to estimate odds ratios (ORs) and their 95% confidence intervals (CIs) for the associations of menstrual and reproductive factors with the risk of breast cancer; overall and by cancer subtype. RESULTS Among breast cancer patients, the luminal B subtype was the most frequent (48.6%), followed by HER2-overexpressing (24.5%), luminal A (16.7%), and triple-negative breast cancer (TNBC; 10.2%). Among menopausal women, menopausal age at 50 years or older (OR = 1.71, 95% CI: 1.15-2.57 vs. <50 y) was associated with an increased risk of breast cancer. Earlier age at menarche (<13 y) was associated with a significantly increased risk of breast cancer (OR = 2.66, 95% CI: 1.08-7.51) among premenopausal women only and the luminal A subtype of breast cancer (OR = 3.06, 95% CI: 1.04-8.16). Having more than two children was associated with a reduced risk of premenopausal (OR = .42, 95%CI: .21-.83), luminal B (OR = .43, 95% CI: .24-.79), and TNBC (OR = .34, 95% CI: .14-.89). Later menopause was positively associated with the risk of breast cancer with HER2 overexpression (OR = 2.19, 95% CI: 1.14-4.23). CONCLUSION Associations of menstrual and reproductive factors with breast cancer among Vietnamese women, particularly for among premenopausal women and for the luminal A subtype, are generally consistent with those reported from other countries. These findings suggest that changes in menstrual and reproductive patterns among young Vietnamese women may contribute to the recent rising incidence of breast cancer in Vietnam.
Collapse
Affiliation(s)
- Oanh Thi Bui
- Vietnam National Cancer Institute, National Cancer Hospital, Hanoi, Vietnam
| | - Huong Thanh Tran
- Vietnam National Cancer Institute, National Cancer Hospital, Hanoi, Vietnam
- Hanoi Medical University, Hanoi, Vietnam
| | - Sang Minh Nguyen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tu Van Dao
- Vietnam National Cancer Institute, National Cancer Hospital, Hanoi, Vietnam
- Hanoi Medical University, Hanoi, Vietnam
| | | | - Anh Tuan Pham
- Vietnam National Cancer Institute, National Cancer Hospital, Hanoi, Vietnam
- Hanoi Medical University, Hanoi, Vietnam
| | - Martha J. Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Fei Ye
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hung Nguyen Luu
- Hillman Cancer Center, University of Pittsburgh Medical Center and Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thuan Van Tran
- Vietnam National Cancer Institute, National Cancer Hospital, Hanoi, Vietnam
- Ministry of Health, Hanoi, Vietnam
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
33
|
Khaled H, Nada YW, Ramadan KM, Fekry S, Seleam MS, Gaafar R, Lotayef M. Primary therapy of early breast cancer: Egyptian view of 2021 St. Gallen consensus. J Egypt Natl Canc Inst 2022; 34:56. [PMID: 36567400 PMCID: PMC9790763 DOI: 10.1186/s43046-022-00156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/04/2022] [Indexed: 12/27/2022] Open
Abstract
PURPOSE The theme of the St. Gallen International Breast Cancer Conference 2021 held virtually for the first time, due to the COVID-19 pandemic, was on tailoring therapies for patients with early breast cancer. A monkey survey that included an Egyptian Panel voted on most of the questions of the original St. Gallen consensus, and some added new questions most relevant to oncology practice in the country, to be able to compare voting results that reflect differences in breast cancer management and decision making. METHODS The panel included 74 Egyptian scientists from different oncology specialties. Management issues including controversial diagnostic and therapeutic interventions were prepared by a small committee and then projected using the online monkey survey website: https://www.surveymonkey.com . The survey included 130 questions. Results were then analyzed, tabulated, and compared to the voting results of the original St. Gallen consensus. RESULTS AND CONCLUSIONS Voting questions and resulting percentages of answers from the Egyptian panel were summarized. There was no consensus between the Egyptian and the original St. Gallen panels on 28/130 statements. They mostly included genetic and pathologic aspects, specifically the routine use of gene signature assays and a few queries involving surgical, radiotherapeutic, and systemic interventions. Probably, available resources and healthcare system differences in Egypt compared to European and the USA were the cause of these differences. This would also be applicable to other low- and low-middle-income healthcare scenarios present in many countries, especially with the present constraints of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Hussein Khaled
- grid.7776.10000 0004 0639 9286Medical Oncology, National Cancer Institute, Cairo University, El-Khalig Square, Cairo, 11796 Egypt
| | - Yousry Wasef Nada
- Medical Oncology Department, Maadi Armed Forces Medical Compound, Cairo, Egypt
| | | | - Shawkat Fekry
- Medical Oncology Department, Maadi Armed Forces Medical Compound, Cairo, Egypt
| | - Mohamed Samy Seleam
- Medical Oncology Department, Maadi Armed Forces Medical Compound, Cairo, Egypt
| | - Rabab Gaafar
- grid.7776.10000 0004 0639 9286Medical Oncology, National Cancer Institute, Cairo University, El-Khalig Square, Cairo, 11796 Egypt
| | - Mohamed Lotayef
- grid.7776.10000 0004 0639 9286Radiation Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
34
|
Hurson AN, Abubakar M, Hamilton AM, Conway K, Hoadley KA, Love MI, Olshan AF, Perou CM, Garcia-Closas M, Troester MA. TP53 Pathway Function, Estrogen Receptor Status, and Breast Cancer Risk Factors in the Carolina Breast Cancer Study. Cancer Epidemiol Biomarkers Prev 2022; 31:124-131. [PMID: 34737209 PMCID: PMC8755611 DOI: 10.1158/1055-9965.epi-21-0661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/25/2021] [Accepted: 10/26/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND TP53 and estrogen receptor (ER) both play essential roles in breast cancer development and progression, with recent research revealing cross-talk between TP53 and ER signaling pathways. Although many studies have demonstrated heterogeneity of risk factor associations across ER subtypes, associations by TP53 status have been inconsistent. METHODS This case-case analysis included incident breast cancer cases (47% Black) from the Carolina Breast Cancer Study (1993-2013). Formalin-fixed paraffin-embedded tumor samples were classified for TP53 functional status (mutant-like/wild-type-like) using a validated RNA signature. For IHC-based TP53 status, mutant-like was classified as at least 10% positivity. We used two-stage polytomous logistic regression to evaluate risk factor heterogeneity due to RNA-based TP53 and/or ER, adjusting for each other and for PR, HER2, and grade. We then compared this with the results when using IHC-based TP53 classification. RESULTS The RNA-based classifier identified 55% of tumors as TP53 wild-type-like and 45% as mutant-like. Several hormone-related factors (oral contraceptive use, menopausal status, age at menopause, and pre- and postmenopausal body mass index) were associated with TP53 mutant-like status, whereas reproductive factors (age at first birth and parity) and smoking were associated with ER status. Multiparity was associated with both TP53 and ER. When classifying TP53 status using IHC methods, no associations were observed with TP53. Associations observed with RNA-based TP53 remained after accounting for basal-like subtype. CONCLUSIONS This case-case study found breast cancer risk factors associated with RNA-based TP53 and ER. IMPACT RNA-based TP53 and ER represent an emerging etiologic schema of interest in breast cancer prevention research.
Collapse
Affiliation(s)
- Amber N Hurson
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Division of Cancer Epidemiology and Genetics, NCI, Rockville, Maryland
| | - Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, NCI, Rockville, Maryland
| | - Alina M Hamilton
- Department of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen Conway
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew F Olshan
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Melissa A Troester
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
35
|
Tang W, Zhou H, Quan T, Chen X, Zhang H, Lin Y, Wu R. XGboost Prediction Model Based on 3.0T Diffusion Kurtosis Imaging Improves the Diagnostic Accuracy of MRI BiRADS 4 Masses. Front Oncol 2022; 12:833680. [PMID: 35372060 PMCID: PMC8968064 DOI: 10.3389/fonc.2022.833680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/21/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The malignant probability of MRI BiRADS 4 breast lesions ranges from 2% to 95%, leading to unnecessary biopsies. The purpose of this study was to construct an optimal XGboost prediction model through a combination of DKI independently or jointly with other MR imaging features and clinical characterization, which was expected to reduce false positive rate of MRI BiRADS 4 masses and improve the diagnosis efficiency of breast cancer. METHODS 120 patients with 158 breast lesions were enrolled. DKI, Diffusion-weighted Imaging (DWI), Proton Magnetic Resonance Spectroscopy (1H-MRS) and Dynamic Contrast-Enhanced MRI (DCE-MRI) were performed on a 3.0-T scanner. Wilcoxon signed-rank test and χ2 test were used to compare patient's clinical characteristics, mean kurtosis (MK), mean diffusivity (MD), apparent diffusion coefficient (ADC), total choline (tCho) peak, extravascular extracellular volume fraction (Ve), flux rate constant (Kep) and volume transfer constant (Ktrans). ROC curve analysis was used to analyze the diagnostic performances of the imaging parameters. Spearman correlation analysis was performed to evaluate the associations of imaging parameters with prognostic factors and breast cancer molecular subtypes. The Least Absolute Shrinkage and Selectionator operator (lasso) and the area under the curve (AUC) of imaging parameters were used to select discriminative features for differentiating the breast benign lesions from malignant ones. Finally, an XGboost prediction model was constructed based on the discriminative features and its diagnostic efficiency was verified in BiRADS 4 masses. RESULTS MK derived from DKI performed better for differentiating between malignant and benign lesions than ADC, MD, tCho, Kep and Ktrans (p < 0.05). Also, MK was shown to be more strongly correlated with histological grade, Ki-67 expression and lymph node status. MD, MK, age, shape and menstrual status were selected to be the optimized feature subsets to construct an XGboost model, which exhibited superior diagnostic ability for breast cancer characterization and an improved evaluation of suspicious breast tumors in MRI BiRADS 4. CONCLUSIONS DKI is promising for breast cancer diagnosis and prognostic factor assessment. An optimized XGboost model that included DKI, age, shape and menstrual status is effective in improving the diagnostic accuracy of BiRADS 4 masses.
Collapse
Affiliation(s)
- Wan Tang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Institute of Health Monitoring, Inspection and Protection, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Han Zhou
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Tianhong Quan
- Department of Electronic and information Engineering, College of Engineering, Shantou University, Shantou, China
| | - Xiaoyan Chen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Huanian Zhang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yan Lin
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Renhua Wu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
36
|
Rweyemamu LP, Akan G, Adolf IC, Magorosa EP, Mosha IJ, Dharsee N, Namkinga LA, Lyantagaye SL, Nateri AS, Atalar F. The distribution of reproductive risk factors disclosed the heterogeneity of receptor-defined breast cancer subtypes among Tanzanian women. BMC Womens Health 2021; 21:423. [PMID: 34930226 PMCID: PMC8686374 DOI: 10.1186/s12905-021-01536-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 11/08/2021] [Indexed: 12/29/2022] Open
Abstract
Background Recent epidemiological studies suggest that reproductive factors are associated with breast cancer (BC) molecular subtypes. However, these associations have not been thoroughly studied in the African populations. The present study aimed to investigate the prevalence of BC molecular subtypes and assess their association with reproductive factors in Tanzanian BC patients. Methods This hospital-based case-only cross-sectional study consisted of 263 histologically confirmed BC patients in Tanzania. Clinico-pathological data, socio-demographic characteristics, anthropometric measurements, and reproductive risk factors were examined using the Chi-square test and one-way ANOVA. The association among reproductive factors and BC molecular subtypes was analyzed using multinomial logistic regression. The heterogeneity of the associations was assessed using the Wald test. Results We found evident subtype heterogeneity for reproductive factors. We observed that post-menopausal status was more prevalent in luminal-A subtype, while compared to luminal-A subtype, luminal-B and HER-2 enriched subtypes were less likely to be found in post-menopausal women (OR: 0.21, 95%CI 0.10–0.41, p = 0.001; OR: 0.39, 95%CI 0.17–0.89, p = 0.026, respectively). Also, the luminal-B subtype was more likely to be diagnosed in patients aged ≤ 40 years than the luminal-A subtype (OR: 2.80, 95%CI 1.46–5.32, p = 0.002). Women who had their first full-term pregnancy at < 30 years were more likely to be of luminal-B (OR: 2.71, 95%CI 1.18–4.17, p = 0.018), and triple-negative (OR: 2.28, 95%CI 1.02–4.07, p = 0.044) subtypes relative to luminal-A subtype. Furthermore, we observed that breastfeeding might have reduced odds of developing luminal-A, luminal-B and triple-negative subtypes. Women who never breastfed were more likely to be diagnosed with luminal-B and triple-negative subtypes when compared to luminal-A subtype (OR: 0.46, 95%CI 0.22–0.95, p = 0.035; OR: 0.41, 95%CI 0.20–0.85, p = 0.017, respectively). . Conclusion Our results are the first data reporting reproductive factors heterogeneity among BC molecular subtypes in Tanzania. Our findings suggest that breast-feeding may reduce the likelihood of developing luminal-A, luminal-B, and triple-negative subtypes. Meanwhile, the first full-term pregnancy after 30 years of age could increase the chance of developing luminal-A subtype, a highly prevalent subtype in Tanzania. More interventions to promote modifiable risk factors across multiple levels may most successfully reduce BC incidence in Africa. Supplementary Information The online version contains supplementary material available at 10.1186/s12905-021-01536-6.
Collapse
Affiliation(s)
- Linus P Rweyemamu
- Department of Molecular Biology and Biotechnology, University of Dar es Salaam, P.O Box 35179, Dar es Salaam, Tanzania.,Mbeya College of Health and Allied Sciences, University of Dar es Salaam, P.O Box 608, Mbeya, Tanzania
| | - Gokce Akan
- MUHAS Genetic Laboratory, Department of Biochemistry, Muhimbili University of Health and Allied Sciences, P.O Box 65001, Dar es Salaam, Tanzania
| | - Ismael C Adolf
- Mbeya College of Health and Allied Sciences, University of Dar es Salaam, P.O Box 608, Mbeya, Tanzania
| | - Erick P Magorosa
- Department of Anatomical Pathology, Muhimbili National Hospital, P.O Box 65000, Dar es Salaam, Tanzania
| | - Innocent J Mosha
- Department of Anatomical Pathology, Muhimbili National Hospital, P.O Box 65000, Dar es Salaam, Tanzania
| | - Nazima Dharsee
- Academic, Research and Consultancy Unit, Ocean Road Cancer Institute, P.O Box 3592, Dar es Salaam, Tanzania
| | - Lucy A Namkinga
- Department of Molecular Biology and Biotechnology, University of Dar es Salaam, P.O Box 35179, Dar es Salaam, Tanzania
| | - Sylvester L Lyantagaye
- Mbeya College of Health and Allied Sciences, University of Dar es Salaam, P.O Box 608, Mbeya, Tanzania
| | - Abdolrahman S Nateri
- Cancer Genetics and Stem Cell Group, Division of Cancer and Stem Cells, School of Medicine, BioDiscovery Institute, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - Fatmahan Atalar
- MUHAS Genetic Laboratory, Department of Biochemistry, Muhimbili University of Health and Allied Sciences, P.O Box 65001, Dar es Salaam, Tanzania. .,Department of Rare Diseases, Child Health Institute, Istanbul University, Istanbul, 34093, Turkey.
| |
Collapse
|
37
|
Identification of Novel CircRNA-miRNA-mRNA Regulatory Network and Its Prognostic Prediction in Breast Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2916398. [PMID: 34745276 PMCID: PMC8570857 DOI: 10.1155/2021/2916398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022]
Abstract
Aim This study aimed to investigate the expression profiles of circRNAs and candidate circRNA-miRNA-mRNA network in BC. Methods Differentially expressed circRNAs, miRNAs, and mRNAs (DEcircRNAs, DEmiRNAs, and DEmRNAs) between BC and normal breast tissue samples were screened by analyzing raw data of the RNA sequencing profile. The expression levels of hub genes in 48 pairs of cancerous and tumor-free breast tissues surgically resected from BC patients were determined by RT-qPCR analysis. Results A total of 145 DEcircRNAs, 140 DEmiRNAs, and 2451 DEmRNAs between BC and normal breast tissue samples were screened out. There were 5 pairs of upcircRNA-downmiRNA-upmRNA network and 20 pairs of downcircRNA-upmiRNA-downmRNA network. EIF4EBP1, DUSP1, EGR2, EZH1, and CBX7 were found to be correlated with overall survival of the patients with BC. The expression level of EIF4EBP1 was increased and the expression levels of DUSP1, EGR2, EZH1, and CBX7 were decreased in cancerous breast tissues compared to tumor-free breast tissues (p < 0.0001). The RT-qPCR results from 48 BC patients were consistent with the bioinformatics results. Conclusion This study provides a novel perspective to study circRNA-miRNA-mRNA network in BC and assists in the identification of new potential biomarkers to be used for diagnostic and prognostic purposes.
Collapse
|
38
|
Tian Y, Guida JL, Koka H, Li EN, Zhu B, Sung H, Chan A, Zhang H, Tang E, Guo C, Deng J, Hu N, Lu N, Gierach GL, Li J, Yang XR. Quantitative Mammographic Density Measurements and Molecular Subtypes in Chinese Women With Breast Cancer. JNCI Cancer Spectr 2021; 5:pkaa092. [PMID: 34651101 DOI: 10.1093/jncics/pkaa092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/25/2020] [Accepted: 09/15/2020] [Indexed: 11/14/2022] Open
Abstract
Background Studies investigating associations between mammographic density (MD) and breast cancer subtypes have generated mixed results. We previously showed that having extremely dense breasts was associated with the human epidermal growth factor receptor-2 (HER2)-enriched subtype in Chinese breast cancer patients. Methods In this study, we reevaluated the MD-subtype association in 1549 Chinese breast cancer patients, using VolparaDensity software to obtain quantitative MD measures. All statistical tests were 2-sided. Results Compared with women with luminal A tumors, women with luminal B/HER2- (odds ratio [OR] = 1.20, 95% confidence interval [CI] = 1.04 to 1.38; P = .01), luminal B/HER2+ (OR = 1.22, 95% CI = 1.03 to 1.46; P = .03), and HER2-enriched tumors (OR = 1.30, 95% CI = 1.06 to 1.59; P = .01) had higher fibroglandular dense volume. These associations were stronger in patients with smaller tumors (<2 cm). In contrast, the triple-negative subtype was associated with lower nondense volume (OR = 0.82, 95% CI = 0.68 to 0.99; P = .04), and the association was only seen among older women (age 50 years or older). Conclusion Although biological mechanisms remain to be investigated, the associations for the HER2-enriched and luminal B subtypes with increasing MD may partially explain the higher prevalence of luminal B and HER2+ breast cancers previously reported in Asian women.
Collapse
Affiliation(s)
- Yuan Tian
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jennifer L Guida
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA.,Division of Cancer Control and Population Sciences, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Hela Koka
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Er-Ni Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Zhu
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Hyuna Sung
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA.,Surveillance and Health Services Research, American Cancer Society, Atlanta, GA, USA
| | - Ariane Chan
- Science and Technology, Volpara Health Technologies, Wellington, New Zealand
| | - Han Zhang
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Eric Tang
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Changyuan Guo
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Joseph Deng
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Nan Hu
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Ning Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gretchen L Gierach
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| | - Jing Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong R Yang
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Bethesda, MD, USA
| |
Collapse
|
39
|
Risk factors for breast cancer subtypes among Black women undergoing screening mammography. Breast Cancer Res Treat 2021; 189:827-835. [PMID: 34342765 DOI: 10.1007/s10549-021-06340-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/24/2021] [Indexed: 01/16/2023]
Abstract
PURPOSE Black women are more likely than non-Hispanic White women to be diagnosed with triple negative breast cancer (TNBC), an aggressive subtype with limited treatment options. The study objective was to evaluate the associations of known breast cancer risk factors, including breast density, with TNBC among Black women. METHODS This study included Black women who underwent screening mammography between the ages of 40-84 years at a University of Pennsylvania Health System between 2010 and 2015. Cox proportional hazard models using multiple imputation with chained equations were used to estimate hazard ratios and 95% confidence intervals for risk factors for ER/PR+/HER2- and TNBC. RESULTS Among 25,013 Black women, there were 330 incident breast cancers (1.3%) during a mean follow-up of 5.8 years; 218 (66.1%) ER/PR+ HER- and 61 (18.1%) TNBC. Having dense breasts (heterogeneously dense or extremely dense) vs. non-dense breasts (almost entirely fatty or scattered areas of fibroglandular density) increased risk of ER/PR+/HER2- breast cancer almost 80% (HR 1.79, 95% CI 1.32-2.43) and TNBC more than twofold (HR 2.53, 1.45-4.44). Older age was associated with an increased risk for ER/PR+/HER2- (HR 1.04, 1.03-1.06) and TNBC (HR 1.03, 1.00-1.05). Having a BMI of > 30 kg/m2 was associated with an increased risk (HR 2.77, 1.05-7.30) for TNBC and an increased risk of ERPR+/HER2- breast cancer in postmenopausal but not pre-menopausal women (p-interaction = 0.016). CONCLUSION Our results suggest that breast density and obesity are strong risk factors for TNBC among Black women. Understanding breast cancer subtype specific risk factors among Black women can help improve risk assessment.
Collapse
|
40
|
Kowalczyk W, Waliszczak G, Jach R, Dulińska-Litewka J. Steroid Receptors in Breast Cancer: Understanding of Molecular Function as a Basis for Effective Therapy Development. Cancers (Basel) 2021; 13:4779. [PMID: 34638264 PMCID: PMC8507808 DOI: 10.3390/cancers13194779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer remains one of the most important health problems worldwide. The family of steroid receptors (SRs), which comprise estrogen (ER), progesterone (PR), androgen (AR), glucocorticoid (GR) and mineralocorticoid (MR) receptors, along with a receptor for a secosteroid-vitamin D, play a crucial role in the pathogenesis of the disease. They function predominantly as nuclear receptors to regulate gene expression, however, their full spectrum of action reaches far beyond this basic mechanism. SRs are involved in a vast variety of interactions with other proteins, including extensive crosstalk with each other. How they affect the biology of a breast cell depends on such factors as post-translational modifications, expression of coregulators, or which SR isoform is predominantly synthesized in a given cellular context. Although ER has been successfully utilized as a breast cancer therapy target for years, research on therapeutic application of other SRs is still ongoing. Designing effective hormone therapies requires thorough understanding of the molecular function of the SRs. Over the past decades, huge amount of data was obtained in multiple studies exploring this field, therefore in this review we attempt to summarize the current knowledge in a comprehensive way.
Collapse
Affiliation(s)
- Wojciech Kowalczyk
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| | - Grzegorz Waliszczak
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| | - Robert Jach
- Department of Gynecology and Obstetrics, Jagiellonian University Medical College, 23 Kopernika St., 31-501 Kraków, Poland;
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Kraków, Poland; (W.K.); (G.W.)
| |
Collapse
|
41
|
McCarthy AM, Friebel-Klingner T, Ehsan S, He W, Welch M, Chen J, Kontos D, Domchek SM, Conant EF, Semine A, Hughes K, Bardia A, Lehman C, Armstrong K. Relationship of established risk factors with breast cancer subtypes. Cancer Med 2021; 10:6456-6467. [PMID: 34464510 PMCID: PMC8446564 DOI: 10.1002/cam4.4158] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 01/07/2023] Open
Abstract
Background Breast cancer is a heterogeneous disease, divided into subtypes based on the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Subtypes have different biology and prognosis, with accumulating evidence of different risk factors. The purpose of this study was to compare breast cancer risk factors across tumor subtypes in a large, diverse mammography population. Methods Women aged 40–84 without a history of breast cancer with a screening mammogram at three United States health systems from 2006 to 2015 were included. Risk factor questionnaires were completed at mammogram visit, supplemented by electronic health records. Invasive tumor subtype was defined by immunohistochemistry as ER/PR+HER2−, ER/PR+HER2+, ER, and PR−HER2+, or triple‐negative breast cancer (TNBC). Cox proportional hazards models were run for each subtype. Associations of race, reproductive history, prior breast problems, family history, breast density, and body mass index (BMI) were assessed. The association of tumor subtypes with screen detection and interval cancer was assessed using logistic regression among invasive cases. Results The study population included 198,278 women with a median of 6.5 years of follow‐up (IQR 4.2–9.0 years). There were 4002 invasive cancers, including 3077 (77%) ER/PR+HER2−, 300 (8%) TNBC, 342 (9%) ER/PR+HER2+, and 126 (3%) ER/PR−HER2+ subtype. In multivariate models, Black women had 2.7 times higher risk of TNBC than white women (HR = 2.67, 95% CI 1.99–3.58). Breast density was associated with increased risk of all subtypes. BMI was more strongly associated with ER/PR+HER2− and HER2+ subtypes among postmenopausal women than premenopausal women. Breast density was more strongly associated with ER/PR+HER2− and TNBC among premenopausal than postmenopausal women. TNBC was more likely to be interval cancer than other subtypes. Conclusions These results have implications for risk assessment and understanding of the etiology of breast cancer subtypes. More research is needed to determine what factors explain the higher risk of TNBC for Black women.
Collapse
Affiliation(s)
- Anne Marie McCarthy
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Sarah Ehsan
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Wei He
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Jinbo Chen
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Despina Kontos
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Susan M Domchek
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Emily F Conant
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alan Semine
- Newton Wellesley Hospital, Newton, Massachusetts, USA
| | - Kevin Hughes
- Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Aditya Bardia
- Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Constance Lehman
- Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Katrina Armstrong
- Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Sommer BC, Dhawan D, Ruple A, Ramos-Vara JA, Hahn NM, Utturkar SM, Ostrander EA, Parker HG, Fulkerson CM, Childress MO, Fourez LM, Enstrom AW, Knapp DW. Basal and Luminal Molecular Subtypes in Naturally-Occurring Canine Urothelial Carcinoma are Associated with Tumor Immune Signatures and Dog Breed. Bladder Cancer 2021; 7:317-333. [PMID: 38993617 PMCID: PMC11181872 DOI: 10.3233/blc-201523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/19/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Improved therapies are needed for patients with invasive urothelial carcinoma (InvUC). Tailoring treatment to molecular subtypes holds promise, but requires further study, including studies in pre-clinical animal models. Naturally-occurring canine InvUC harbors luminal and basal subtypes, mimicking those observed in humans, and could offer a relevant model for the disease in people. OBJECTIVE To further validate the canine InvUC model, clinical and tumor characteristics associated with luminal and basal subtypes in dogs were determined, with comparison to findings from humans. METHODS RNA sequencing (RNA-seq) analyses were performed on 56 canine InvUC tissues and bladder mucosa from four normal dogs. Data were aligned to CanFam 3.1, and differentially expressed genes identified. Data were interrogated with panels of genes defining luminal and basal subtypes, immune signatures, and other tumor features. Subject and tumor characteristics, and outcome data were obtained from medical records. RESULTS Twenty-nine tumors were classified as luminal and 27 tumors as basal subtype. Basal tumors were strongly associated with immune infiltration (OR 52.22, 95%CI 4.68-582.38, P = 0.001) and cancer progression signatures in RNA-seq analyses, more advanced clinical stage, and earlier onset of distant metastases in exploratory analyses (P = 0.0113). Luminal tumors were strongly associated with breeds at high risk for InvUC (OR 0.06, 95%CI 0.01 -0.37, P = 0.002), non-immune infiltrative signatures, and less advanced clinical stage. CONCLUSIONS Dogs with InvUC could provide a valuable model for testing new treatment strategies in the context of molecular subtype and immune status, and the search for germline variants impacting InvUC onset and subtype.
Collapse
Affiliation(s)
- Breann C. Sommer
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Deepika Dhawan
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Audrey Ruple
- Department of Public Health, Purdue University, West Lafayette, IN, USA
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - José A. Ramos-Vara
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette IN, USA
| | - Noah M. Hahn
- Department of Oncology and Urology, and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Sagar M. Utturkar
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Elaine A. Ostrander
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heidi G. Parker
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher M. Fulkerson
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Michael O. Childress
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
| | - Lindsey M. Fourez
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Alexander W. Enstrom
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
| | - Deborah W. Knapp
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN, USA
- Purdue University Center for Cancer Research, West Lafayette, IN, USA
| |
Collapse
|
43
|
Zhang X, Shen L, Cai R, Yu X, Yang J, Wu X, Zhu Y, Liu X. Comprehensive Analysis of the Immune-Oncology Targets and Immune Infiltrates of N 6-Methyladenosine-Related Long Noncoding RNA Regulators in Breast Cancer. Front Cell Dev Biol 2021; 9:686675. [PMID: 34277627 PMCID: PMC8283003 DOI: 10.3389/fcell.2021.686675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022] Open
Abstract
Breast cancer (BRCA) has become the highest incidence of cancer due to its heterogeneity. To predict the prognosis of BRCA patients, sensitive biomarkers deserve intensive investigation. Herein, we explored the role of N6-methyladenosine-related long non-coding RNAs (m6A-related lncRNAs) as prognostic biomarkers in BRCA patients acquired from The Cancer Genome Atlas (TCGA; n = 1,089) dataset and RNA sequencing (RNA-seq) data (n = 196). Pearson’s correlation analysis, and univariate and multivariate Cox regression were performed to select m6A-related lncRNAs associated with prognosis. Twelve lncRNAs were identified to construct an m6A-related lncRNA prognostic signature (m6A-LPS) in TCGA training (n = 545) and validation (n = 544) cohorts. Based on the 12 lncRNAs, risk scores were calculated. Then, patients were classified into low- and high-risk groups according to the median value of risk scores. Distinct immune cell infiltration was observed between the two groups. Patients with low-risk score had higher immune score and upregulated expressions of four immune-oncology targets (CTLA4, PDCD1, CD274, and CD19) than patients with high-risk score. On the contrary, the high-risk group was more correlated with overall gene mutations, Wnt/β-catenin signaling, and JAK-STAT signaling pathways. In addition, the stratification analysis verified the ability of m6A-LPS to predict prognosis. Moreover, a nomogram (based on risk score, age, gender, stage, PAM50, T, M, and N stage) was established to evaluate the overall survival (OS) of BRCA patients. Thus, m6A-LPS could serve as a sensitive biomarker in predicting the prognosis of BRCA patients and could exert positive influence in personalized immunotherapy.
Collapse
Affiliation(s)
- Xiaoqiang Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Shen
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruyu Cai
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiafei Yu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junzhe Yang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xian Wu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
Ye T, Li J, Feng J, Guo J, Wan X, Xie D, Liu J. The subtype-specific molecular function of SPDEF in breast cancer and insights into prognostic significance. J Cell Mol Med 2021; 25:7307-7320. [PMID: 34191390 PMCID: PMC8335683 DOI: 10.1111/jcmm.16760] [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: 01/06/2021] [Revised: 05/30/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is a molecular diverse disease which becomes the most common malignancy among women worldwide. There are four BC subtypes (Luminal A, Luminal B, HER2‐enriched and Basal‐like) robustly established following gene expression pattern‐based characterization, behave significant differences in terms of their incidence, risk factors, prognosis and therapeutic sensitivity. Thus, there is an urgent need to provide mechanism research, treatment strategies and/or prognosis evaluation based on the patient stratification of BC subtypes. The prostate‐derived ETS factor SPDEF was first identified as an activator of prostate specific antigen, and then, the involvements in many aspects of BC have been proposed. However, the subtype‐specific molecular function of SPDEF in BC and insights into prognostic significance have not been clearly elucidated. This study demonstrated for the first time that SPDEF may play a diversity role in the expression levels, clinicopathologic importance, biological function and prognostic evaluation in BC via bioinformatics and experimental evidence, which mainly depends on different BC subtyping. In summary, our findings would help to better understand the possible mechanisms of various BC subtypes and to find possible candidate genes for prognostic and therapeutic usage.
Collapse
Affiliation(s)
- Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Jingyuan Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Jinglan Guo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Xue Wan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Dan Xie
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
45
|
Isheden G, Grassmann F, Czene K, Humphreys K. Lymph node metastases in breast cancer: Investigating associations with tumor characteristics, molecular subtypes and polygenic risk score using a continuous growth model. Int J Cancer 2021; 149:1348-1357. [PMID: 34097750 DOI: 10.1002/ijc.33704] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 11/09/2022]
Abstract
We investigate the association between rate of breast cancer lymph node spread and grade, estrogen receptor (ER) status, progesteron receptor status, decision tree derived PAM50 molecular subtype and a polygenic risk score (PRS), using data on 10 950 women included from two different data sources. Lymph node spread was analyzed using a novel continuous tumor progression model that adjusts for tumor volume in a biologically motivated way and that incorporates covariates of interest. Grades 2 and 3 tumors, respectively, were associated with 1.63 and 2.17 times faster rates of lymph node spread than Grade 1 tumors (P < 10-16 ). ER/PR negative breast cancer was associated with a 1.25/1.19 times faster spread than ER/PR positive breast cancer, respectively (P = .0011 and .0012). Among the molecular subtypes luminal A, luminal B, Her2-enriched and basal-like, Her2-enriched breast cancer was associated with 1.53 times faster spread than luminal A cancer (P = .00072). PRS was not associated with the rate of lymph node spread. Continuous growth models are useful for quantifying associations between lymph node spread and tumor characteristics. These may be useful for building realistic progression models for microsimulation studies used to design individualized screening programs.
Collapse
Affiliation(s)
- Gabriel Isheden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Felix Grassmann
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,The Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Keith Humphreys
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Ghosh S, Das T, Suman SK, Sarma HD, Dash A. Preparation and Preliminary Evaluation of 68Ga-Acridine: An Attempt to Study the Potential of Radiolabeled DNA Intercalator as a PET Radiotracer for Tumor Imaging. Anticancer Agents Med Chem 2021; 20:1538-1547. [PMID: 32357824 DOI: 10.2174/1871520620666200502002609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/13/2019] [Accepted: 02/28/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acridine is a well-known DNA intercalator and thereby gets easily inserted within DNA. As uncontrolled rapid cell division is one of the primary characteristics of the tumors, it is expected that acridine or its suitable derivatives will have preferential accumulation in the tumorous lesions. Therefore, an attempt was made to radiolabel an acridine derivative with 68Ga and study the potential of the 68Ga-acridine complex as a PET agent for tumor imaging. METHODS 9-aminoacridine was coupled with p-NCS-benzyl-DOTA to render it suitable for labeling with 68Ga. The purified acridine-DOTA conjugate was radiolabeled with 68Ga, eluted from a 68Ge/68Ga radionuclide generator. Various radiolabeling parameters were optimized and the stability of the radiolabeled preparation was studied. The biological behavior of the 68Ga-acridine complex was studied both in vitro and in vivo using Raji cell line and fibrosarcoma tumor bearing Swiss mice, respectively. RESULTS 68Ga-acridine complex was obtained with ~100% radiochemical purity under the optimized reaction conditions involving incubation of 2mg/mL of ligand at 100°C for 30 minutes. The complex maintained a radiochemical purity of >95% in normal saline and >65% in human blood serum at 3h post-incubation. In vitro cellular study showed (3.2±0.1)% uptake of the radiotracer in the Raji cells. Biodistribution study revealed significant tumor accumulation [(11.41±0.41)% injected activity in per gram] of the radiotracer within 1h postadministration along with uptake in other non-target organs such as, blood, liver, GIT kidney etc. Conclusion: The present study indicates the potential of 68Ga-acridine as a PET agent for imaging of tumorous lesions. However, further detailed evaluation of the agent is warranted to explore its actual potential.
Collapse
Affiliation(s)
- Subhajit Ghosh
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Tapas Das
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Shishu K Suman
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Haladhar D Sarma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| |
Collapse
|
47
|
Chen Z, Fu H, Wu H, Huang J, Yao L, Zhang X, Li Y. Syntheses and Preliminary Evaluation of Dual Target PET Probe [18F]-NOTA-Gly3- E (2PEG4-RGD-WH701) for PET Imaging of Breast Cancer. Anticancer Agents Med Chem 2021; 20:1548-1557. [PMID: 32329699 DOI: 10.2174/1871520620666200424101936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE Tumor Necrosis Factor Receptor 1 (TNFR1) and integrin αvβ3 receptor are overexpressed in breast cancer. We hypothesized that a peptide ligand recognizing both receptors in a single receptor-binding probe would be advantageous. Here, we developed a novel 18F-labeled fusion peptide probe [18F]-NOTA-Gly3- E(2PEG4-RGD-WH701) targeting dual receptors (TNFR1 and αvβ3) and evaluated the diagnostic efficacy of this radioactive probe in both MDA-MB-231 and MCF-7 xenograft models in mice. METHODS The NOTA-conjugated RGD-WH701 analog was radiolabeled with 18F using NOTA-AlF chelation method. We used two PEG4 molecules and Glutamic acid (Glu) to covalently link c(RGDyK) with WH701. Gly3 was also added to further improve the water solubility and pharmacokinetic properties of the probe. The expression of TNFR1 and Integrin αvβ3 in MCF-7 and MDA-MB-231 cells was detected by western blot analysis and immunofluorescence staining. The tumor-targeting characteristics of [18F]-NOTA-Gly3-E(2PEG4-RGDWH701) were assessed in nude mice bearing MDA-MB-231 and MCF-7 xenografts. RESULTS HPLC analysis of the product NOTA-G3-E (2P4-RGD-WH701) revealed a purity >95%. The yield after attenuation correction was approximately 33.5%±2.8% (n=5), and the radiochemical purity was above 95%. The MDA-MB-231 tumor uptake of [18F]-NOTA-Gly3-E(2PEG4-RGD-WH701) was 1.14±0.14%ID/g, as measured by PET at 40min postinjection (p.i.). In comparison, the tumor uptake of [18F]-NOTA-RGD and [18F]- NOTA-WH701 in MDA-MB-231 xenografts was 0.96±0.13%ID/g and 0.93±0.28%ID/g, respectively. The MCF-7 tumor uptake of [18F]-NOTA-Gly3-E(2PEG4-RGD-WH701) was 1.22±0.11%ID/g, as measured by PET at 40min postinjection (p.i.). In comparison, the tumor uptake of [18F]-NOTA-RGD and [18F]-NOTA-WH701 in MCF-7 xenografts was 0.99±0.18%ID/g and 0.57±0.08%ID/g, respectively. CONCLUSION [18F]AlF-NOTA-Gly3-E(2PEG4-RGD-WH701) was successfully synthesized and labeled with 18F. The results from the microPET/CT and biodistribution studies of [18F]AlF-NOTA-Gly3-E(2PEG4-RGDWH701) showed that the tracer could specifically target TNFR1 and integrin αvβ3 receptors.
Collapse
Affiliation(s)
- Zijun Chen
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Hao Fu
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Hua Wu
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Jinxiong Huang
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Lanlin Yao
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Xianzhong Zhang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| |
Collapse
|
48
|
Figueroa JD, Gierach GL, Duggan MA, Fan S, Pfeiffer RM, Wang Y, Falk RT, Loudig O, Abubakar M, Ginsberg M, Kimes TM, Richert-Boe K, Glass AG, Rohan TE. Risk factors for breast cancer development by tumor characteristics among women with benign breast disease. Breast Cancer Res 2021; 23:34. [PMID: 33736682 PMCID: PMC7977564 DOI: 10.1186/s13058-021-01410-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
Background Among women diagnosed with invasive breast cancer, 30% have a prior diagnosis of benign breast disease (BBD). Thus, it is important to identify factors among BBD patients that elevate invasive cancer risk. In the general population, risk factors differ in their associations by clinical pathologic features; however, whether women with BBD show etiologic heterogeneity in the types of breast cancers they develop remains unknown. Methods Using a nested case-control study of BBD and breast cancer risk conducted in a community healthcare plan (Kaiser Permanente Northwest), we assessed relationships of histologic features in BBD biopsies and patient characteristics with subsequent breast cancer risk and tested for heterogeneity of associations by estrogen receptor (ER) status, tumor grade, and size. The study included 514 invasive breast cancer cases (median follow-up of 9 years post-BBD diagnosis) and 514 matched controls, diagnosed with proliferative or non-proliferative BBD between 1971 and 2006, with follow-up through mid-2015. Odds ratios (ORs) and 95% confidence intervals (CIs) were obtained using multivariable polytomous logistic regression models. Results Breast cancers were predominantly ER-positive (86%), well or moderately differentiated (73%), small (74% < 20 mm), and stage I/II (91%). Compared to patients with non-proliferative BBD, proliferative BBD with atypia conferred increased risk for ER-positive cancer (OR = 5.48, 95% CI = 2.14–14.01) with only one ER-negative case, P-heterogeneity = 0.45. The presence of columnar cell lesions (CCLs) at BBD diagnosis was associated with a 1.5-fold increase in the risk of both ER-positive and ER-negative tumors, with a 2-fold increase (95% CI = 1.21–3.58) observed among postmenopausal women (56%), independent of proliferative BBD status with and without atypia. We did not identify statistically significant differences in risk factor associations by tumor grade or size. Conclusion Most tumors that developed after a BBD diagnosis in this cohort were highly treatable low-stage ER-positive tumors. CCL in BBD biopsies may be associated with moderately increased risk, independent of BBD histology, and irrespective of ER status. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01410-1.
Collapse
Affiliation(s)
- Jonine D Figueroa
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA. .,The Usher Institute, Old Medical School, The University of Edinburgh, Teviot Place, Edinburgh, UK. .,CRUK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK.
| | - Gretchen L Gierach
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Máire A Duggan
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Calgary, Canada
| | - Shaoqi Fan
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Ruth M Pfeiffer
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Yihong Wang
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Roni T Falk
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Olivier Loudig
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, Nutley, NJ, USA
| | - Mustapha Abubakar
- National Cancer Institute, National Institutes of Health, Division of Cancer Epidemiology and Genetics, Bethesda, MD, USA
| | - Mindy Ginsberg
- Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Belfer Building, Room 1301, Bronx, NY, 10461, USA
| | - Teresa M Kimes
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | | | - Andrew G Glass
- Kaiser Permanente Center for Health Research, Portland, OR, USA
| | - Thomas E Rohan
- Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Belfer Building, Room 1301, Bronx, NY, 10461, USA.
| |
Collapse
|
49
|
E2F1 copy number variations in germline and breast cancer: a retrospective study of 222 Italian women. Mol Med 2021; 27:26. [PMID: 33691613 PMCID: PMC7948349 DOI: 10.1186/s10020-021-00287-2] [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: 12/28/2020] [Accepted: 03/04/2021] [Indexed: 11/29/2022] Open
Abstract
Background Breast cancer is the most common neoplasia among women in developed countries. The risk factors of breast cancer can be distinguished in modifiable and unmodifiable factors and, among the latter, genetic factors play a key role. Copy number variations (CNVs) are genetic variants that are classified as rare when present in less than 1% of the healthy population. Since rare CNVs are often cause of diseases, over the last years, their contribution in carcinogenesis has become a relevant matter of study. E2F1 is a transcriptional factor that plays an important role in regulating cell cycle and apoptosis. Its double and conflicting role is the reason why it acts both as oncogene and as tumour suppressor, depending on cell context. Since anomalies in expression or in number of copies of E2F1 have been related to several cancers, we aimed to study number of germline copies of E2F1 in women with breast cancer in order to better elucidate their contribution as predisposing factor to this tumour. Methods We performed, hence, a retrospective study on 222 Italian women with breast cancer recruited from October 2002 to December 2007. TaqMan CNV assay and Real-Time PCR were carried out to analyse, respectively, E2F1 CNV and E2F1 expression in the subjects of the study. Chi square test or Fisher’s exact test and Student's t‐test were used to calculate the frequency of CNVs and differences in continuous variables between groups, respectively. Results Intriguingly, we found that 10/222 (4.5%) women with breast cancer had more copies than controls (0/200, 0%), furthermore, the number of copies positively correlated with E2F1 gene expression in breast cancer tissue, suggesting that the constitutive gain of the gene could translate into an increased risk of genomic instability. Additionally, we found that altered E2F1 copies were present prevalently in the patients with contralateral breast cancer (20%) and all of them had a positive family history, both typically associated with hereditary cancer. Conclusions Our findings suggest that copy number variations of E2F1 might be a susceptibility factor for breast cancer, however, further studies on large cohorts are to be performed in order to better delineate the phenotype linked to the gain of E2F1 copies.
Collapse
|
50
|
Chen C, Pan Y, Bai L, Chen H, Duan Z, Si Q, Zhu R, Chuang TH, Luo Y. MicroRNA-3613-3p functions as a tumor suppressor and represents a novel therapeutic target in breast cancer. Breast Cancer Res 2021; 23:12. [PMID: 33494814 PMCID: PMC7836180 DOI: 10.1186/s13058-021-01389-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/11/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND MicroRNAs have been reported to participate in tumorigenesis, treatment resistance, and tumor metastasis. Novel microRNAs need to be identified and investigated to guide the clinical prognosis or therapy for breast cancer. METHOD The copy number variations (CNVs) of MIR3613 from Cancer Genome Atlas (TCGA) or Cancer Cell Line Encyclopedia (CCLE) were analyzed, and its correlation with breast cancer subtypes or prognosis was investigated. The expression level of miR-3613-3p in tumor tissues or serum of breast cancer patients was detected using in situ hybridization and qPCR. Gain-of-function studies were performed to determine the regulatory role of miR-3613-3p on proliferation, apoptosis, and tumor sphere formation of human breast cancer cells MDA-MB-231 or MCF-7. The effects of miR-3613-3p on tumor growth or metastasis in an immunocompromised mouse model of MDA-MB-231-luciferase were explored by intratumor injection of miR-3613-3p analogue. The target genes, interactive lncRNAs, and related signaling pathways of miR-3613-3p were identified by bioinformatic prediction and 3'-UTR assays. RESULTS We found that MIR3613 was frequently deleted in breast cancer genome and its deletion was correlated with the molecular typing, and an unfavorable prognosis in estrogen receptor-positive patients. MiR-3613-3p level was also dramatically lower in tumor tissues or serum of breast cancer patients. Gain-of-function studies revealed that miR-3613-3p could suppress proliferation and sphere formation and promote apoptosis in vitro and impeded tumor growth and metastasis in vivo. Additionally, miR-3613-3p might regulate cell cycle by targeting SMS, PAFAH1B2, or PDK3 to restrain tumor progression. CONCLUSION Our findings indicate a suppressive role of miR-3613-3p in breast cancer progression, which may provide an innovative marker or treatment for breast cancer patients.
Collapse
Affiliation(s)
- Chong Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Yundi Pan
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Lipeng Bai
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Department of Clinical Laboratory, Jiangxi Cancer Hospital, Nanchang, 330029, China
| | - Huilin Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Zhaojun Duan
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Qin Si
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Ruizhe Zhu
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Yunping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
- Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|