1
|
Araki M, Noguchi S, Kubo Y, Yasuda A, Koh M, Otsuka H, Yokosuka M, Soeta S. Expression of receptor-type tumour endothelial marker 8 in carcinoma cells showing luminal progenitor-like phenotypes in canine mammary gland carcinomas. J Comp Pathol 2023; 200:35-45. [PMID: 36641985 DOI: 10.1016/j.jcpa.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/05/2022] [Accepted: 11/05/2022] [Indexed: 01/15/2023]
Abstract
This study aimed to investigate the expression of receptor-type tumour endothelial marker 8 (TEM8RT) in canine mammary gland carcinomas (CMGCs) using immunohistochemistry and to evaluate the association between carcinoma cell TEM8RT expression and tumour histological features, histological grades and the differentiation status of neoplastic epithelial cells. TEM8RT expression was more frequently detected in simple carcinomas (tubular and tubulopapillary) than in solid carcinomas, and it was significantly correlated with histological grade Ⅰ tumours and a low mitotic index. Additionally, TEM8RT+ carcinoma cells were more frequently found in CMGCs showing luminal progenitor-like phenotypes, such as Notch1+, CK19+/CK5+/CD49f+ and CK19+/CK5-/CD49f+. Double-labelling immunofluorescence detection techniques confirmed that most TEM8RT+ carcinoma cells expressed CD49f, Notch1 and CK19. However, TEM8RT immunoreactivity was not found in carcinoma cells expressing GATA3, which upregulates mature luminal cell differentiation. Furthermore, TEM8RT+ carcinoma cells were detected in a few CMGCs showing basal/stem cell-like phenotypes such as CK19-/CK5+/CD49f+ and CK19-/CK5+/CD49f-. These findings indicate that TEM8RT is expressed in luminal progenitor-like carcinoma cells in CMGCs. Since TEM8 enhances self-renewal in human mammary stem/progenitor cells, it also may be involved in maintenance of luminal progenitor-like carcinoma cells, resulting in prevention of their transition to basal/stem cell-like carcinoma cells and development of less malignant CMGCs. Therefore, TEM8RT may be useful for indicating prognostic outcomes and identifying the possible ontogeny of carcinoma cells in mammary gland tumours.
Collapse
Affiliation(s)
- Mami Araki
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Syunya Noguchi
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Yoshiaki Kubo
- Veterinary Medical Teaching Hospital, Attached Facility, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Akiko Yasuda
- Veterinary Medical Teaching Hospital, Attached Facility, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Miki Koh
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Hirotada Otsuka
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Makoto Yokosuka
- Laboratory of Comparative and Behavioral Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Satoshi Soeta
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan.
| |
Collapse
|
2
|
Sheng DL, Shen XG, Shi ZT, Chang C, Li JW. Survival outcome assessment for triple-negative breast cancer: a nomogram analysis based on integrated clinicopathological, sonographic, and mammographic characteristics. Eur Radiol 2022; 32:6575-6587. [PMID: 35759017 PMCID: PMC9474369 DOI: 10.1007/s00330-022-08910-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/31/2022]
Abstract
Objective This study aimed to incorporate clinicopathological, sonographic, and mammographic characteristics to construct and validate a nomogram model for predicting disease-free survival (DFS) in patients with triple-negative breast cancer (TNBC). Methods Patients diagnosed with TNBC at our institution between 2011 and 2015 were retrospectively evaluated. A nomogram model was generated based on clinicopathological, sonographic, and mammographic variables that were associated with 1-, 3-, and 5-year DFS determined by multivariate logistic regression analysis in the training set. The nomogram model was validated according to the concordance index (C-index) and calibration curves in the validation set. Results A total of 636 TNBC patients were enrolled and divided into training cohort (n = 446) and validation cohort (n = 190). Clinical factors including tumor size > 2 cm, axillary dissection, presence of LVI, and sonographic features such as angular/spiculated margins, posterior acoustic shadows, and presence of suspicious lymph nodes on preoperative US showed a tendency towards worse DFS. The multivariate analysis showed that no adjuvant chemotherapy (HR = 6.7, 95% CI: 2.6, 17.5, p < 0.0005), higher axillary tumor burden (HR = 2.7, 95% CI: 1.0, 7.1, p = 0.045), and ≥ 3 malignant features on ultrasound (HR = 2.4, CI: 1.1, 5.0, p = 0.021) were identified as independent prognostic factors associated with poorer DFS outcomes. In the nomogram, the C-index was 0.693 for the training cohort and 0.694 for the validation cohort. The calibration plots also exhibited excellent consistency between the nomogram-predicted and actual survival probabilities in both the training and validation cohorts. Conclusions Clinical variables and sonographic features were correlated with the prognosis of TNBCs. The nomogram model based on three variables including no adjuvant chemotherapy, higher axillary tumor load, and more malignant sonographic features showed good predictive performance for poor survival outcomes of TNBC. Key Points • The absence of adjuvant chemotherapy, heavy axillary tumor load, and malignant-like sonographic features can predict DFS in patients with TNBC. • Mammographic features of TNBC could not predict the survival outcomes of patients with TNBC. • The nomogram integrating clinicopathological and sonographic characteristics is a reliable predictive model for the prognostic outcome of TNBC. Supplementary Information The online version contains supplementary material available at 10.1007/s00330-022-08910-4.
Collapse
Affiliation(s)
- Dan-Li Sheng
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xi-Gang Shen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Zambelli A, Sgarra R, De Sanctis R, Agostinetto E, Santoro A, Manfioletti G. Heterogeneity of triple-negative breast cancer: understanding the Daedalian labyrinth and how it could reveal new drug targets. Expert Opin Ther Targets 2022; 26:557-573. [PMID: 35638300 DOI: 10.1080/14728222.2022.2084380] [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] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is considered the most aggressive breast cancer subtype with the least favorable outcomes. However, recent research efforts have generated an enhanced knowledge of the biology of the disease and have provided a new, more comprehensive understanding of the multifaceted ecosystem that underpins TNBC. AREAS COVERED In this review, the authors illustrate the principal biological characteristics of TNBC, the molecular driver alterations, targetable genes, and the biomarkers of immune engagement that have been identified across the subgroups of TNBC. Accordingly, the authors summarize the landscape of the innovative and investigative biomarker-driven therapeutic options in TNBC that emerge from the unique biological basis of the disease. EXPERT OPINION The therapeutic setting of TNBC is rapidly evolving. An enriched understanding of the tumor spatial and temporal heterogeneity and the surrounding microenvironment of this complex disease can effectively support the development of novel and tailored opportunities of treatment.
Collapse
Affiliation(s)
- Alberto Zambelli
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Riccardo Sgarra
- Department of Life sciences, University of Trieste, Trieste, Italy
| | - Rita De Sanctis
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elisa Agostinetto
- Department of Biomedical Sciences, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium and Humanitas University, Milan, Italy
| | - Armando Santoro
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy
| | | |
Collapse
|
4
|
Li JW, Cao YC, Zhao ZJ, Shi ZT, Duan XQ, Chang C, Chen JG. Prediction for pathological and immunohistochemical characteristics of triple-negative invasive breast carcinomas: the performance comparison between quantitative and qualitative sonographic feature analysis. Eur Radiol 2022; 32:1590-1600. [PMID: 34519862 DOI: 10.1007/s00330-021-08224-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Sonographic features are associated with pathological and immunohistochemical characteristics of triple-negative breast cancer (TNBC). To predict the biological property of TNBC, the performance using quantitative high-throughput sonographic feature analysis was compared with that using qualitative feature assessment. METHODS We retrospectively reviewed ultrasound images, clinical, pathological, and immunohistochemical (IHC) data of 252 female TNBC patients. All patients were subgrouped according to the histological grade, Ki67 expression level, and human epidermal growth factor receptor 2 (HER2) score. Qualitative sonographic feature assessment included shape, margin, posterior acoustic pattern, and calcification referring to the Breast Imaging Reporting and Data System (BI-RADS). Quantitative sonographic features were acquired based on the computer-aided radiomics analysis. Breast cancer masses were manually segmented from the surrounding breast tissues. For each ultrasound image, 1688 radiomics features of 7 feature classes were extracted. The principal component analysis (PCA), least absolute shrinkage and selection operator (LASSO), and support vector machine (SVM) were used to determine the high-throughput radiomics features that were highly correlated to biological properties. The performance using both quantitative and qualitative sonographic features to predict biological properties of TNBC was represented by the area under the receiver operating characteristic curve (AUC). RESULTS In the qualitative assessment, regular tumor shape, no angular or spiculated margin, posterior acoustic enhancement, and no calcification were used as the independent sonographic features for TNBC. Using the combination of these four features to predict the histological grade, Ki67, HER2, axillary lymph node metastasis (ALNM), and lymphovascular invasion (LVI), the AUC was 0.673, 0.680, 0.651, 0.587, and 0.566, respectively. The number of high-throughput features that closely correlated with biological properties was 34 for histological grade (AUC 0.942), 27 for Ki67 (AUC 0.732), 25 for HER2 (AUC 0.730), 34 for ALNM (AUC 0.804), and 34 for LVI (AUC 0.795). CONCLUSION High-throughput quantitative sonographic features are superior to traditional qualitative ultrasound features in predicting the biological behavior of TNBC. KEY POINTS • Sonographic appearances of TNBCs showed a great variety in accordance with its biological and clinical characteristics. • Both qualitative and quantitative sonographic features of TNBCs are associated with tumor biological characteristics. • The quantitative high-throughput feature analysis is superior to two-dimensional sonographic feature assessment in predicting tumor biological property.
Collapse
Affiliation(s)
- Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Yu-Cheng Cao
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China
| | - Zhi-Jin Zhao
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Xiao-Qian Duan
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, No 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.
| | - Jian-Gang Chen
- Shanghai Key Laboratory of Multidimensional Information Processing, East China Normal University, #500 Dongchuan Rd., Shanghai, 200241, China.
| |
Collapse
|
5
|
Xie F, Liu L, Yang H, Liu M, Wang S, Guo J, Yu L, Zhou F, Wang F, Xiang Y, Yu Z, Wang S. OUP accepted manuscript. Oncologist 2022; 27:e1-e8. [PMID: 35305101 PMCID: PMC8842323 DOI: 10.1093/oncolo/oyab018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Fei Xie
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Liyuan Liu
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Houpu Yang
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Miao Liu
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Siyuan Wang
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Jiajia Guo
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Lixiang Yu
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Fei Zhou
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Fei Wang
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yujuan Xiang
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Zhigang Yu, Department of Breast Surgery, the Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, Shandong 250033, People’s Republic of China. Tel: +86-531-8587-5048;
| | - Shu Wang
- Department of Breast Center, Peking University People’s Hospital, Beijing, People’s Republic of China
- Corresponding author: Shu Wang, Department of Breast Center, Peking University People’s Hospital, 11 Xizhimen South Street, Xicheng, Beijing 100044, People’s Republic of China. Tel: +86-10-8832-4010;
| |
Collapse
|
6
|
Ensenyat-Mendez M, Llinàs-Arias P, Orozco JIJ, Íñiguez-Muñoz S, Salomon MP, Sesé B, DiNome ML, Marzese DM. Current Triple-Negative Breast Cancer Subtypes: Dissecting the Most Aggressive Form of Breast Cancer. Front Oncol 2021; 11:681476. [PMID: 34221999 PMCID: PMC8242253 DOI: 10.3389/fonc.2021.681476] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous disease defined by the absence of estrogen receptor (ER) and progesterone receptor (PR) expression, and human epidermal growth factor receptor 2 (HER2) overexpression that lacks targeted treatments, leading to dismal clinical outcomes. Thus, better stratification systems that reflect intrinsic and clinically useful differences between TNBC tumors will sharpen the treatment approaches and improve clinical outcomes. The lack of a rational classification system for TNBC also impacts current and emerging therapeutic alternatives. In the past years, several new methodologies to stratify TNBC have arisen thanks to the implementation of microarray technology, high-throughput sequencing, and bioinformatic methods, exponentially increasing the amount of genomic, epigenomic, transcriptomic, and proteomic information available. Thus, new TNBC subtypes are being characterized with the promise to advance the treatment of this challenging disease. However, the diverse nature of the molecular data, the poor integration between the various methods, and the lack of cost-effective methods for systematic classification have hampered the widespread implementation of these promising developments. However, the advent of artificial intelligence applied to translational oncology promises to bring light into definitive TNBC subtypes. This review provides a comprehensive summary of the available classification strategies. It includes evaluating the overlap between the molecular, immunohistochemical, and clinical characteristics between these approaches and a perspective about the increasing applications of artificial intelligence to identify definitive and clinically relevant TNBC subtypes.
Collapse
Affiliation(s)
- Miquel Ensenyat-Mendez
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Pere Llinàs-Arias
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Javier I J Orozco
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Sandra Íñiguez-Muñoz
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Matthew P Salomon
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Borja Sesé
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Maggie L DiNome
- Department of Surgery, David Geffen School of Medicine, University California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Diego M Marzese
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| |
Collapse
|
7
|
Li JW, Zhou J, Shi ZT, Li N, Zhou SC, Chang C. Sonographic Features of Triple-Negative Breast Carcinomas Are Correlated With mRNA-lncRNA Signatures and Risk of Tumor Recurrence. Front Oncol 2021; 10:587422. [PMID: 33542899 PMCID: PMC7851073 DOI: 10.3389/fonc.2020.587422] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023] Open
Abstract
Background To determine a correlation between mRNA and lncRNA signatures, sonographic features, and risk of recurrence in triple-negative breast cancers (TNBC). Methods We retrospectively reviewed the data from 114 TNBC patients having undergone transcriptome analysis. The risk of tumor recurrence was determined based on the correlation between transcriptome profiles and recurrence-free survival. Ultrasound (US) features were described according to the Breast Imaging Reporting and Data System. Multivariate logistic regression analysis determined the correlation between US features and risk of recurrence. The predictive value of sonographic features in determining tumor recurrence was analyzed using receiver operating characteristic curves. Results Three mRNAs (CHRDL1, FCGR1A, and RSAD2) and two lncRNAs (HIF1A-AS2 and AK124454) were correlated with recurrence-free survival in patients with TNBC. Among the three mRNAs, two were upregulated (FCGR1A and RSAD2) and one was downregulated (CHRDL1) in TNBCs. LncRNAs HIF1A-AS2 and AK124454 were upregulated in TNBCs. Based on these signatures, an integrated mRNA–lncRNA model was established using Cox regression analysis to determine the risk of tumor recurrence. Benign-like sonographic features, such as regular shape, circumscribed margin, posterior acoustic enhancement, and no calcifications, were associated with HIF1A-AS2 expression and high risk of tumor recurrence (P<0.05). Malignant-like features, such as irregular shape, uncircumscribed margin, no posterior acoustic enhancement, and calcifications, were correlated with CHRDL1 expression and low risk of tumor recurrence (P<0.05). Conclusions Sonographic features and mRNA–lncRNA signatures in TNBCs represent the risk of tumor recurrence. Taken together, US may be a promising technique in determining the prognosis of patients with TNBC.
Collapse
Affiliation(s)
- Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Zhou
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Na Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Chong Zhou
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Kriegsmann K, Flechtenmacher C, Heil J, Kriegsmann J, Mechtersheimer G, Aulmann S, Weichert W, Sinn HP, Kriegsmann M. Immunohistological Expression of SOX-10 in Triple-Negative Breast Cancer: A Descriptive Analysis of 113 Samples. Int J Mol Sci 2020; 21:ijms21176407. [PMID: 32899175 PMCID: PMC7503807 DOI: 10.3390/ijms21176407] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/13/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022] Open
Abstract
Background: SRY-related HMG-box 10 (SOX-10) is commonly expressed in triple negative breast cancer (TNBC). However, data on the biological significance of SOX-10 expression is limited. Therefore, we investigated immunhistological SOX-10 expression in TNBC and correlated the results with genetic alterations and clinical data. Methods: A tissue microarray including 113 TNBC cases was stained by SOX-10. Immunohistological data of AR, BCL2, CD117, p53 and Vimentin was available from a previous study. Semiconductor-based panel sequencing data including commonly altered breast cancer genes was also available from a previous investigation. SOX-10 expression was correlated with clinicopathological, immunohistochemical and genetic data. Results: SOX-10 was significantly associated with CD117 and Vimentin, but not with AR expression. An association of SOX-10 with BCL2, EGFR or p53 staining was not observed. SOX-10-positive tumors harbored more often TP53 mutations but less frequent mutations of PIK3CA or alterations of the PIK3K pathway. SOX-10 expression had no prognostic impact either on disease-free, distant disease-free, or overall survival. Conclusions: While there might be a value of SOX-10 as a differential diagnostic marker to identify metastases of TNBC, its biological role remains to be investigated.
Collapse
Affiliation(s)
- Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Christa Flechtenmacher
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.F.); (G.M.); (H.-P.S.)
| | - Jörg Heil
- Breast Unit, Women’s Hospital, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Jörg Kriegsmann
- Institute of Pathology, Cytology and Molecular Pathology, 54296 Trier, Germany;
- Danube Private University Krems, 3500 Krems, Austria
| | - Gunhild Mechtersheimer
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.F.); (G.M.); (H.-P.S.)
| | | | - Wilko Weichert
- Institute of Pathology, TU Munich, 81675 Munich, Germany;
| | - Hans-Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.F.); (G.M.); (H.-P.S.)
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (C.F.); (G.M.); (H.-P.S.)
- Correspondence: ; Tel.: +49-6221-56-36930
| |
Collapse
|
9
|
Li JW, Li N, Jiang YZ, Liu YR, Shi ZT, Chang C, Shao ZM. Ultrasonographic appearance of triple-negative invasive breast carcinoma is associated with novel molecular subtypes based on transcriptomic analysis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:435. [PMID: 32395479 PMCID: PMC7210204 DOI: 10.21037/atm.2020.03.204] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Various sonographic features of triple-negative invasive breast carcinomas (TNBC) expected to be associated with the molecular subtypes based on transcriptomic analysis were examined. The effects of clinical, sonographic, pathological, and molecular features on survival outcome was also studied. Methods One hundred and fourteen patients with breast cancer with negative expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal receptor 2 (HER2) were included in our retrospective study. Based on the transcriptomic profiles, four stable clusters named immunomodulatory (IM), luminal androgen receptor (LAR), mesenchymal-like (MES), and basal-like and immune-suppressed (BLIS) were identified. Ultrasound (US) images were reviewed by two US physicians according to Breast Imaging Reporting and Data System (BI-RADS). Multivariate Cox regression was used to determine the variables associated with recurrence-free survival (RFS) and overall survival (OS). Results There were 21 IM, 18 LAR, 36 MES, and 39 BLIS cases. The four molecular subtypes showed significant differences in terms of tumor shape (P=0.008) and posterior acoustic pattern (P=0.028). Compared with the subtypes LAR and MES, the IM and BLIS subtypes had higher probability of presenting benign-like sonographic features, such as regular shape, no angular/spiculated margin, and posterior acoustic enhancement (P<0.05). The independent risk factors for RFS events and death were axillary lymph node metastasis (P<0.05) and BLIS subtype (P<0.05). BLIS subtype showed worse OS than other subtypes (log rank P=0.05). TNBCs with benign sonographic features tended to have less death events (3.3% vs. 15.2%, P=0.088). Conclusions Sonographic appearance of TNBCs is associated with transcriptome-based molecular subtypes, and tends to correlate with the survival outcome.
Collapse
Affiliation(s)
- Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Na Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi-Rong Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhi-Ming Shao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| |
Collapse
|
10
|
Cui J, Jiang H. Prediction of postoperative survival of triple-negative breast cancer based on nomogram model combined with expression of HIF-1α and c-myc. Medicine (Baltimore) 2019; 98:e17370. [PMID: 31577739 PMCID: PMC6783179 DOI: 10.1097/md.0000000000017370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aims of this study were to explore the expression of hypoxia inducible factor-1α (HIF-1α) and c-myc protein in triple-negative breast cancer (TNBC) and its clinical prognostic significance, and to establish a prediction model for postoperative survival of TNBC based on nomogram.A total of 87 patients with TNBC at the Department of Breast Surgery, Beijing Chaoyang Hospital, Capital Medical University from January 2012 to December 2015 were enrolled in this study. Immunohistochemistry was performed to detect the expression of HIF-1α and c-myc protein in breast cancer tissues. Cox regression analyses were performed to explore the correlation between HIF-1α/c-myc expression and clinical pathological parameters as well as prognosis. Receiver-operating characteristic curve was generated for cox multivariate analysis. A nomogram was generated based on the cox multivariate analysis, and a calibration curve was prepared for the nomogram to evaluate the consistency between the predicted probability of the nomogram and the actual observed probability. The stability of nomogram model was validated with an external cohort including 39 TNBC patients.The positive expression rates of HIF-1α and c-myc protein in breast cancer tissues were 41.4% (36/87) and 55.2% (48/87), respectively. HIF-1α expression was significantly correlated with age, tumor diameter, histological grade, lymph node status, and tumor TNM stage; c-myc expression was significantly associated with tumor diameter, histological grade, lymph node status, and tumor TNM stage. Cox univariate and multivariate analyses showed that HIF-1α and c-myc protein expression, histological grade, lymph node status, and tumor TNM stage were the independent risk factors for postoperative survival in TNBC patients. The AUC of prediction model was 0.843 (0.809-0.887). The nomogram could predict the probability of 3-year disease-free survival according to each patient's condition. The calibration curve displayed good agreement of the predicted probability with the actual observed probability, indicating that the nomogram model had great value of prediction. The external validation indicated the prediction model had good stability.HIF-1α-positive expression, c-myc positive expression, histological grade III, lymph node positive, and TNM stage III tumors suggested that TNBC patients had a poor prognosis. This prediction model can be used to predict postoperative survival of TNBC.
Collapse
|
11
|
Role of Hedgehog Signaling in Breast Cancer: Pathogenesis and Therapeutics. Cells 2019; 8:cells8040375. [PMID: 31027259 PMCID: PMC6523618 DOI: 10.3390/cells8040375] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality in women, only followed by lung cancer. Given the importance of BC in public health, it is essential to identify biomarkers to predict prognosis, predetermine drug resistance and provide treatment guidelines that include personalized targeted therapies. The Hedgehog (Hh) signaling pathway plays an essential role in embryonic development, tissue regeneration, and stem cell renewal. Several lines of evidence endorse the important role of canonical and non-canonical Hh signaling in BC. In this comprehensive review we discuss the role of Hh signaling in breast development and homeostasis and its contribution to tumorigenesis and progression of different subtypes of BC. We also examine the efficacy of agents targeting different components of the Hh pathway both in preclinical models and in clinical trials. The contribution of the Hh pathway in BC tumorigenesis and progression, its prognostic role, and its value as a therapeutic target vary according to the molecular, clinical, and histopathological characteristics of the BC patients. The evidence presented here highlights the relevance of the Hh signaling in BC, and suggest that this pathway is key for BC progression and metastasis.
Collapse
|
12
|
Coussy F, de Koning L, Lavigne M, Bernard V, Ouine B, Boulai A, El Botty R, Dahmani A, Montaudon E, Assayag F, Morisset L, Huguet L, Sourd L, Painsec P, Callens C, Chateau-Joubert S, Servely JL, Larcher T, Reyes C, Girard E, Pierron G, Laurent C, Vacher S, Baulande S, Melaabi S, Vincent-Salomon A, Gentien D, Dieras V, Bieche I, Marangoni E. A large collection of integrated genomically characterized patient-derived xenografts highlighting the heterogeneity of triple-negative breast cancer. Int J Cancer 2019; 145:1902-1912. [PMID: 30859564 DOI: 10.1002/ijc.32266] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/26/2018] [Accepted: 02/19/2019] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) represents 10% of all breast cancers and is a very heterogeneous disease. Globally, women with TNBC have a poor prognosis, and the development of effective targeted therapies remains a real challenge. Patient-derived xenografts (PDX) are clinically relevant models that have emerged as important tools for the analysis of drug activity and predictive biomarker discovery. The purpose of this work was to analyze the molecular heterogeneity of a large panel of TNBC PDX (n = 61) in order to test targeted therapies and identify biomarkers of response. At the gene expression level, TNBC PDX represent all of the various TNBC subtypes identified by the Lehmann classification except for immunomodulatory subtype, which is underrepresented in PDX. NGS and copy number data showed a similar diversity of significantly mutated gene and somatic copy number alteration in PDX and the Cancer Genome Atlas TNBC patients. The genes most commonly altered were TP53 and oncogenes and tumor suppressors of the PI3K/AKT/mTOR and MAPK pathways. PDX showed similar morphology and immunohistochemistry markers to those of the original tumors. Efficacy experiments with PI3K and MAPK inhibitor monotherapy or combination therapy showed an antitumor activity in PDX carrying genomic mutations of PIK3CA and NRAS genes. TNBC PDX reproduce the molecular heterogeneity of TNBC patients. This large collection of PDX is a clinically relevant platform for drug testing, biomarker discovery and translational research.
Collapse
Affiliation(s)
- Florence Coussy
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France.,Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France
| | - Leanne de Koning
- Translational Research Department, RPPA Platform, Institut Curie Research Center, Paris, France
| | - Marion Lavigne
- Department of Biopathology, Institut Curie, Paris, France
| | - Virginie Bernard
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Berengere Ouine
- Translational Research Department, RPPA Platform, Institut Curie Research Center, Paris, France
| | - Anais Boulai
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Franck Assayag
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Ludivine Morisset
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Lea Huguet
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Laura Sourd
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Pierre Painsec
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| | - Celine Callens
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Jean-Luc Servely
- BioPôle Alfort, National Veterinary School of Alfort, Maison Alfort, France
| | | | - Cecile Reyes
- Translational Research Department, Genomics Platform, Institut Curie Research Center, Paris, France
| | | | - Gaelle Pierron
- Unit of Somatic Genomics, Department of Genetics, Institut Curie, Paris, France
| | | | - Sophie Vacher
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | - Sylvain Baulande
- Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, Paris, France
| | - Samia Melaabi
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France
| | | | - David Gentien
- Translational Research Department, Genomics Platform, Institut Curie Research Center, Paris, France
| | | | - Ivan Bieche
- Unit of Pharmacogenomics, Department of Genetics, Institut Curie, Paris, France.,Inserm U1016, Paris Descartes University, Paris, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Department of Translational Research, Institut Curie Research Center, Paris, France
| |
Collapse
|
13
|
Scott DA, Casadonte R, Cardinali B, Spruill L, Mehta AS, Carli F, Simone N, Kriegsmann M, Del Mastro L, Kriegsmann J, Drake RR. Increases in Tumor N-Glycan Polylactosamines Associated with Advanced HER2-Positive and Triple-Negative Breast Cancer Tissues. Proteomics Clin Appl 2019; 13:e1800014. [PMID: 30592377 PMCID: PMC8913074 DOI: 10.1002/prca.201800014] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 12/11/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE Using a recently developed matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS) method, human breast cancer formalin-fixed paraffin-embedded (FFPE) tissue sections and tissue microarrays (TMA) are evaluated for N-linked glycan distribution in the tumor microenvironment. EXPERIMENTAL DESIGN Tissue sections representing multiple human epidermal growth factor receptor 2 (HER2) receptor-positive and triple-negative breast cancers (TNBC) in both TMA and FFPE slide format are processed for high resolution N-glycan MALDI-IMS. An additional FFPE tissue cohort of primary and metastatic breast tumors from the same donors are also evaluated. RESULTS The cumulative N-glycan MALDI-IMS analysis of breast cancer FFPE tissues and TMAs indicate the distribution of specific glycan structural classes to stromal, necrotic, and tumor regions. A series of high-mannose, branched and fucosylated glycans are detected predominantly within tumor regions. Additionally, a series of polylactosamine glycans are detected in advanced HER2+, TNBC, and metastatic breast cancer tissues. Comparison of tumor N-glycan species detected in paired primary and metastatic tissues indicate minimal changes between the two conditions. CONCLUSIONS AND CLINICAL RELEVANCE The prevalence of tumor-associated polylactosamine glycans in primary and metastatic breast cancer tissues indicates new mechanistic insights into the development and progression of breast cancers. The presence of these glycans could be targeted for therapeutic strategies and further evaluation as potential prognostic biomarkers.
Collapse
Affiliation(s)
- Danielle A. Scott
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and MUSC Proteomics Center Medical University of South Carolina Charleston, 29425, SC, USA
| | | | - Barbara Cardinali
- Department of Medical Oncology Ospedale Policlinico San Martino Genova, 16132, GE, Italy
| | - Laura Spruill
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston, 29425, SC, USA
| | - Anand S. Mehta
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and MUSC Proteomics Center Medical University of South Carolina Charleston, 29425, SC, USA
| | - Franca Carli
- Department of Surgical Pathology Ospedale Policlinico San Martino Genova, 16132, GE, Italy
| | - Nicole Simone
- Department of Radiation Oncology Thomas Jefferson University Philadelphia, 19107, PA, USA
| | | | - Lucia Del Mastro
- Department of Internal Medicine University of Genova Genova, 16132, GE, Italy
| | - Joerg Kriegsmann
- Institute of Pathology University of Heidelberg Heidelberg, 69117, Germany
| | - Richard R. Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and MUSC Proteomics Center Medical University of South Carolina Charleston, 29425, SC, USA
| |
Collapse
|
14
|
Li JW, Zhang K, Shi ZT, Zhang X, Xie J, Liu JY, Chang C. Triple-negative invasive breast carcinoma: the association between the sonographic appearances with clinicopathological feature. Sci Rep 2018; 8:9040. [PMID: 29899425 PMCID: PMC5998063 DOI: 10.1038/s41598-018-27222-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/25/2018] [Indexed: 12/31/2022] Open
Abstract
In this study, we aimed to evaluate the clinical and pathological factors that associated with sonographic appearances of triple-negative (TN) invasive breast carcinoma. With the ethical approval, 560 patients who were pathologically confirmed as invasive breast carcinoma were reviewed for ultrasound, clinical, and pathological data. Logistic regression analysis was used to identify the typical sonographic features for TN invasive breast carcinomas. The effect of clinical and pathological factors on the sonographic features of TN invasive breast carcinoma was studied. There were 104 cases of TN invasive breast carcinoma. The independent sonographic features for the TN subgroup included regular shape (odds ratio, OR = 2.14, p = 0.007), no spiculated/angular margin (OR = 1.93, p = 0.035), posterior acoustic enhancement (OR = 2.14, p = 0.004), and no calcifications (OR = 2.10, p = 0.008). Higher pathological grade was significantly associated with regular tumor shape of TN breast cancer (p = 0.012). Higher Ki67 level was significantly associated with regular tumor shape (p = 0.023) and absence of angular/spiculated margin (p = 0.005). Higher human epidermal growth factor receptor 2 (HER2) score was significantly associated with the presence of calcifications (p = 0.033). We conclude that four sonographic features are associated with TN invasive breast carcinoma. Heterogeneity of sonographic features was associated with the pathological grade, Ki67 proliferation level and HER2 score of TN breast cancers.
Collapse
Affiliation(s)
- Jia-Wei Li
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kai Zhang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhao-Ting Shi
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xun Zhang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Juan Xie
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun-Ying Liu
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cai Chang
- Department of Medical Ultrasound, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
15
|
Strietz J, Stepputtis SS, Preca BT, Vannier C, Kim MM, Castro DJ, Au Q, Boerries M, Busch H, Aza-Blanc P, Heynen-Genel S, Bronsert P, Kuster B, Stickeler E, Brabletz T, Oshima RG, Maurer J. ERN1 and ALPK1 inhibit differentiation of bi-potential tumor-initiating cells in human breast cancer. Oncotarget 2018; 7:83278-83293. [PMID: 27829216 PMCID: PMC5347769 DOI: 10.18632/oncotarget.13086] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/21/2016] [Indexed: 12/21/2022] Open
Abstract
Cancers are heterogeneous by nature. While traditional oncology screens commonly use a single endpoint of cell viability, altering the phenotype of tumor-initiating cells may reveal alternative targets that regulate cellular growth by processes other than apoptosis or cell division. We evaluated the impact of knocking down expression of 420 kinases in bi-lineage triple-negative breast cancer (TNBC) cells that express characteristics of both myoepithelial and luminal cells. Knockdown of ERN1 or ALPK1 induces bi-lineage MDA-MB-468 cells to lose the myoepithelial marker keratin 5 but not the luminal markers keratin 8 and GATA3. In addition, these cells exhibit increased β-casein production. These changes are associated with decreased proliferation and clonogenicity in spheroid cultures and anchorage-independent growth assays. Confirmation of these assays was completed in vivo, where ERN1- or ALPK1-deficient TNBC cells are less tumorigenic. Finally, treatment with K252a, a kinase inhibitor active on ERN1, similarly impairs anchorage-independent growth of multiple breast cancer cell lines. This study supports the strategy to identify new molecular targets for types of cancer driven by cells that retain some capacity for normal differentiation to a non-tumorigenic phenotype. ERN1 and ALPK1 are potential targets for therapeutic development.
Collapse
Affiliation(s)
- Juliane Strietz
- Department of Visceral Surgery, University Hospital Freiburg, German Cancer Consortium (DKTK), Freiburg, Germany
| | - Stella S Stepputtis
- Department of Visceral Surgery, University Hospital Freiburg, German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bogdan-Tiberius Preca
- Department of Visceral Surgery, University Hospital Freiburg, German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Corinne Vannier
- Department of Visceral Surgery, University Hospital Freiburg, German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mihee M Kim
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - David J Castro
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Qingyan Au
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Melanie Boerries
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Systems Biology of the Cellular Microenvironment at The DKFZ Partner Site Freiburg, German Cancer Consortium (DKTK), Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Hauke Busch
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Systems Biology of the Cellular Microenvironment at The DKFZ Partner Site Freiburg, German Cancer Consortium (DKTK), Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Pedro Aza-Blanc
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | | | - Peter Bronsert
- Department of Surgical Pathology, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Bernhard Kuster
- Technische Universitaet Muenchen, Partner Site of the German Cancer Consortium, Freising, Germany
| | - Elmar Stickeler
- Department of OBGYN, University Clinic Aachen (UKA), Aachen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine I, University of Erlangen-Nuernberg, Erlangen, Germany
| | - Robert G Oshima
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Jochen Maurer
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Bissanum R, Lirdprapamongkol K, Svasti J, Navakanitworakul R, Kanokwiroon K. The role of WT1 isoforms in vasculogenic mimicry and metastatic potential of human triple negative breast cancer cells. Biochem Biophys Res Commun 2017; 494:256-262. [PMID: 29024629 DOI: 10.1016/j.bbrc.2017.10.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 01/06/2023]
Abstract
Triple negative breast cancer (TNBC) is highly aggressive and has a few therapeutic treatments, so new targeted therapy and biomarkers are required to provide alternative choices for treating TNBC patients. Recent studies showed that vasculogenic mimicry (VM), the formation of blood channels by aggressive cancer cells that mimic endothelial cells, is a factor contributing to poor prognosis in TNBC. Wilms' tumor 1 (WT1) gene has been found to be highly expressed in TNBC, and has 4 major distinct isoforms; isoform A (-17AA/-KTS; -/-), isoform B (+17AA/-KTS; +/-), isoform C (-17AA/+KTS; -/+) and isoform D (+17AA/+KTS; +/+). The involvement of each WT1 isoform in TNBC progression remains largely unclear. In this study, WT1 isoform-overexpressing cell sublines were established from a TNBC cell line, MDA-MB-231, by stable transfection, and the aggressive behavior of the cell sublines were evaluated. Only the WT1 isoform B- and isoform C-overexpressing cell sublines showed the significant increase in VM forming capability compared to the parental cell line and other isoform cell sublines. qRT-PCR was used to explore the change in expression level of two VM-related genes, EphA2 and VE-cadherin. All WT1 isoform cell sublines showed up-regulation of EphA2 but the levels detected in the isoform B- and isoform C-cell sublines were higher than those observed in other cell sublines. In contrast, significant up-regulation of VE-cadherin was found only in isoform A- and isoform D-cell sublines. Isoform B- and isoform C-cell sublines showed higher rates of cell migration compared to those of other cell sublines, as determined by both wound healing and Transwell assays. Gelatin zymography revealed increased MMP-9 enzyme production in isoform D-cell subline compared to the parental cell line, but this change was not observed in other cell sublines. Western blot analysis showed significantly increased expression of β-catenin in isoform B- and isoform C-cell sublines, compared to parental cell line and other isoform cell sublines. In conclusion, our findings demonstrate that WT1 isoforms play different roles in modulating the VM-forming capacity and metastatic potential of TNBC cells.
Collapse
Affiliation(s)
- Rassanee Bissanum
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | | | - Jisnuson Svasti
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Raphatphorn Navakanitworakul
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | - Kanyanatt Kanokwiroon
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand; The Excellent Research Laboratory of Cancer Molecular Biology, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand.
| |
Collapse
|
17
|
Chen JM, Li Y, Xu J, Gong L, Wang LW, Liu WL, Liu J. Computer-aided prognosis on breast cancer with hematoxylin and eosin histopathology images: A review. Tumour Biol 2017; 39:1010428317694550. [PMID: 28347240 DOI: 10.1177/1010428317694550] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
With the advance of digital pathology, image analysis has begun to show its advantages in information analysis of hematoxylin and eosin histopathology images. Generally, histological features in hematoxylin and eosin images are measured to evaluate tumor grade and prognosis for breast cancer. This review summarized recent works in image analysis of hematoxylin and eosin histopathology images for breast cancer prognosis. First, prognostic factors for breast cancer based on hematoxylin and eosin histopathology images were summarized. Then, usual procedures of image analysis for breast cancer prognosis were systematically reviewed, including image acquisition, image preprocessing, image detection and segmentation, and feature extraction. Finally, the prognostic value of image features and image feature–based prognostic models was evaluated. Moreover, we discussed the issues of current analysis, and some directions for future research.
Collapse
Affiliation(s)
- Jia-Mei Chen
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yan Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, China
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital of Capital Medical University, Beijing, China
| | - Jun Xu
- Jiangsu Key Laboratory of Big Data Analysis Technique, Nanjing University of Information Science and Technology, Nanjing, China
| | - Lei Gong
- Jiangsu Key Laboratory of Big Data Analysis Technique, Nanjing University of Information Science and Technology, Nanjing, China
| | - Lin-Wei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, China
| | - Wen-Lou Liu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Wuhan, China
| | - Juan Liu
- State Key Laboratory of Software Engineering, School of Computer, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Sinn HP, Schneeweiss A, Keller M, Schlombs K, Laible M, Seitz J, Lakis S, Veltrup E, Altevogt P, Eidt S, Wirtz RM, Marmé F. Comparison of immunohistochemistry with PCR for assessment of ER, PR, and Ki-67 and prediction of pathological complete response in breast cancer. BMC Cancer 2017; 17:124. [PMID: 28193205 PMCID: PMC5307758 DOI: 10.1186/s12885-017-3111-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 02/04/2017] [Indexed: 12/23/2022] Open
Abstract
Background Proliferation may predict response to neoadjuvant therapy of breast cancer and is commonly assessed by manual scoring of slides stained by immunohistochemistry (IHC) for Ki-67 similar to ER and PgR. This method carries significant intra- and inter-observer variability. Automatic scoring of Ki-67 with digital image analysis (qIHC) or assessment of MKI67 gene expression with RT-qPCR may improve diagnostic accuracy. Methods Ki-67 IHC visual assessment was compared to the IHC nuclear tool (AperioTM) on core biopsies from a randomized neoadjuvant clinical trial. Expression of ESR1, PGR and MKI67 by RT-qPCR was performed on RNA extracted from the same formalin-fixed paraffin-embedded tissue. Concordance between the three methods (vIHC, qIHC and RT-qPCR) was assessed for all 3 markers. The potential of Ki-67 IHC and RT-qPCR to predict pathological complete response (pCR) was evaluated using ROC analysis and non-parametric Mann-Whitney Test. Results Correlation between methods (qIHC versus RT-qPCR) was high for ER and PgR (spearman´s r = 0.82, p < 0.0001 and r = 0.86, p < 0.0001, respectively) resulting in high levels of concordance using predefined cut-offs. When comparing qIHC of ER and PgR with RT-qPCR of ESR1 and PGR the overall agreement was 96.6 and 91.4%, respectively, while overall agreement of visual IHC with RT-qPCR was slightly lower for ER/ESR1 and PR/PGR (91.2 and 92.9%, respectively). In contrast, only a moderate correlation was observed between qIHC and RT-qPCR continuous data for Ki-67/MKI67 (Spearman’s r = 0.50, p = 0.0001). Up to now no predictive cut-off for Ki-67 assessment by IHC has been established to predict response to neoadjuvant chemotherapy. Setting the desired sensitivity at 100%, specificity for the prediction of pCR (ypT0ypN0) was significantly higher for mRNA than for protein (68.9% vs. 22.2%). Moreover, the proliferation levels in patients achieving a pCR versus not differed significantly using MKI67 RNA expression (Mann-Whitney p = 0.002), but not with qIHC of Ki-67 (Mann-Whitney p = 0.097) or vIHC of Ki-67 (p = 0.131). Conclusion Digital image analysis can successfully be implemented for assessing ER, PR and Ki-67. IHC for ER and PR reveals high concordance with RT-qPCR. However, RT-qPCR displays a broader dynamic range and higher sensitivity than IHC. Moreover, correlation between Ki-67 qIHC and RT-qPCR is only moderate and RT-qPCR with MammaTyper® outperforms qIHC in predicting pCR. Both methods yield improvements to error-prone manual scoring of Ki-67. However, RT-qPCR was significantly more specific. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3111-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 220-221, 69120, Heidelberg, Germany.
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, University-Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Marius Keller
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 220-221, 69120, Heidelberg, Germany
| | | | - Mark Laible
- BioNTech Diagnostics GmbH, 55131, Mainz, Germany
| | - Julia Seitz
- National Center for Tumor Diseases, University-Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| | - Sotirios Lakis
- STRATIFYER Molecular Pathology GmbH, Werthmannstr. 1c, 50935, Köln, Germany
| | - Elke Veltrup
- STRATIFYER Molecular Pathology GmbH, Werthmannstr. 1c, 50935, Köln, Germany
| | - Peter Altevogt
- German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Sebastian Eidt
- Department of Pathology, St. Elisabeth-Krankenhaus, Werthmannstr. 1c, 50935, Köln, Germany
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, Werthmannstr. 1c, 50935, Köln, Germany.,Department of Pathology, St. Elisabeth-Krankenhaus, Werthmannstr. 1c, 50935, Köln, Germany
| | - Frederik Marmé
- National Center for Tumor Diseases, University-Hospital Heidelberg, Im Neuenheimer Feld 460, 69120, Heidelberg, Germany
| |
Collapse
|
19
|
Masili-Oku SM, Almeida BGLD, Bacchi CE, Filassi JR, Baracat EC, Carvalho FM. Lymphocyte-predominant triple-negative breast carcinomas in premenopausal patients: Lower expression of basal immunohistochemical markers. Breast 2017; 31:34-39. [DOI: 10.1016/j.breast.2016.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 10/20/2022] Open
|
20
|
Çelebi F, Pilancı KN, Ordu Ç, Ağacayak F, Alço G, İlgün S, Sarsenov D, Erdoğan Z, Özmen V. The role of ultrasonographic findings to predict molecular subtype, histologic grade, and hormone receptor status of breast cancer. Diagn Interv Radiol 2016; 21:448-53. [PMID: 26359880 DOI: 10.5152/dir.2015.14515] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The correlation between imaging findings and pathologic characteristics of tumors may provide information for diagnosis and treatment of cancer. The aim of this study is to determine whether ultrasound features of breast cancer are associated with molecular subtype, histologic grade, and hormone receptor status, as well as assess the predictive value of these features. METHODS A total of 201 consecutive invasive breast cancer patients were reviewed from the database according to the Breast Imaging and Reporting Data System (BI-RADS). Tumor margins were classified as circumscribed and noncircumscribed. Noncircumscribed group was divided into indistinct, spiculated, angular, and microlobulated. The posterior acoustic features were divided into four categories: shadowing, enhancement, no change, and mixed pattern. RESULTS Tumors with posterior shadowing were more likely to be of nontriple negative subtype (odds ratio [OR], 7.42; 95% CI, 2.10-24.99; P = 0.002), low histologic grade (grade 1 or 2 vs. grade 3: OR, 2.42; 95% CI, 1.34-4.35; P = 0.003) and having at least one positive receptor (OR, 3.36; 95% CI, 1.55-7.26; P = 0.002). Tumors with circumscribed margins were more often triple-negative subtype (OR, 6.72; 95% CI, 2.56-17.65; P < 0.001), high grade (grade 3 vs. grade 1 or 2: OR, 5.42; 95% CI, 2.66-11.00; P < 0.001) and hormone receptor negative (OR, 4.87; 95% CI, 2.37-9.99; P < 0.001). CONCLUSION Sonographic features are strongly associated with molecular subtype, histologic grade, and hormone receptor status of the tumor. These findings may separate triple-negative breast cancer from other molecular subtypes.
Collapse
Affiliation(s)
- Filiz Çelebi
- Department of Radiology, Florence Nightingale Hospital, İstanbul, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Habib JG, O'Shaughnessy JA. The hedgehog pathway in triple-negative breast cancer. Cancer Med 2016; 5:2989-3006. [PMID: 27539549 PMCID: PMC5083752 DOI: 10.1002/cam4.833] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/26/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Treatment of triple‐negative breast cancer (TNBC) remains challenging due to the underlying heterogeneity of this disease coupled with the lack of predictive biomarkers and effective targeted therapies. Intratumoral heterogeneity, particularly enrichment for breast cancer stem cell‐like subpopulations, has emerged as a leading hypothesis for systemic therapy resistance and clinically aggressive course of poor prognosis TNBC. A growing body of literature supports the role of the stem cell renewal Hedgehog (Hh) pathway in breast cancer. Emerging preclinical data also implicate Hh signaling in TNBC pathogenesis. Herein, we review the evidence for a pathophysiologic role of Hh signaling in TNBC and explore mechanisms of crosstalk between the Hh pathway and other key signaling networks as well as their potential implications for Hh‐targeted interventions in TNBC.
Collapse
Affiliation(s)
- Joyce G Habib
- Baylor Charles A. Sammons Cancer Center, Dallas, Texas
| | - Joyce A O'Shaughnessy
- Baylor Charles A. Sammons Cancer Center, Dallas, Texas.
- Texas Oncology, Dallas, Texas.
| |
Collapse
|
22
|
Gautam P, Karhinen L, Szwajda A, Jha SK, Yadav B, Aittokallio T, Wennerberg K. Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. Mol Cancer 2016; 15:34. [PMID: 27165605 PMCID: PMC4862054 DOI: 10.1186/s12943-016-0517-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/30/2016] [Indexed: 01/23/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines. Method The responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses. Results Our approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines. Conclusions Taken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers. Electronic supplementary material The online version of this article (doi:10.1186/s12943-016-0517-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Prson Gautam
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Leena Karhinen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Agnieszka Szwajda
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sawan Kumar Jha
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Bhagwan Yadav
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
23
|
Fastner G, Hauser-Kronberger C, Moder A, Reitsamer R, Zehentmayr F, Kopp P, Fussl C, Fischer T, Deutschmann H, Sedlmayer F. Survival and local control rates of triple-negative breast cancer patients treated with boost-IOERT during breast-conserving surgery. Strahlenther Onkol 2015; 192:1-7. [PMID: 26403912 DOI: 10.1007/s00066-015-0895-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/27/2015] [Indexed: 11/26/2022]
Abstract
AIM The purpose of this work was to retrospectively evaluate survival and local control rates of triple-negative breast cancer subtypes classified as five marker negative (5NP) and core basal (CB), respectively, after breast-conserving surgery and intraoperative boost radiotherapy with electrons (IOERT) followed by whole breast irradiation. METHODS AND MATERIALS A total of 71 patients with triple-negative breast cancer were enrolled, who were treated with lumpectomy, axillary lymph node dissection, and IOERT with 9.6 Gy (median Dmax) followed by normofractionated whole breast irradiation to median total doses of 54 Gy. Chemotherapy was applied in a neoadjuvant (12 %), adjuvant (75 %), or combinational setting (7 %). RESULTS After a median follow-up of 97 months (range 4-170 months), 5 in-breast recurrences were detected (7.0 %). For all patients, 8-year actuarial rates for local control, metastases-free survival, disease-specific survival, and overall survival amounted to 89, 75, 80, and 69 %, respectively. All local recurrences occurred in grade 3 (G3) tumors irrespective of their specific immunohistochemical phenotype; thus, the local control rate for grades 1/2 (G1/2) was 100 % for both 5NP and CB, while for G3 it was 88 % for 5NP and 90 % for CB (p = 0.65 and 0.82, respectively, n.s.). For disease-specific survival, only the difference of the best-prognosis group 5-NP/G3 vs. the worst-prognosis cohort CB/G1/2 was statistically significant: 90 % vs. 54 % (p = 0.03). CONCLUSION Boost-IOERT provides acceptable long-term in-breast control in triple negative breast cancer. The best subgroup in terms of disease-specific survival was represented by 5NP in combination with tumor grading G3.
Collapse
MESH Headings
- Actuarial Analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/radiotherapy
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/mortality
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/radiotherapy
- Carcinoma, Lobular/surgery
- Chemotherapy, Adjuvant
- Cohort Studies
- Combined Modality Therapy
- Dose Fractionation, Radiation
- Electrons/therapeutic use
- Female
- Follow-Up Studies
- Humans
- Intraoperative Period
- Lymph Node Excision
- Mastectomy, Segmental
- Neoplasm Staging
- Radiotherapy Dosage
- Survival Rate
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/mortality
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/radiotherapy
- Triple Negative Breast Neoplasms/surgery
Collapse
Affiliation(s)
- Gerd Fastner
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria.
| | | | - Angelika Moder
- Institute of Inborn Errors in Metabolism, Landeskrankenhaus, Paracelsus Medical University, Salzburg, Austria
| | - Roland Reitsamer
- Department of Special Gynecology, Landeskrankenhaus, Paracelsus Medical University, Salzburg, Austria
- Department of Gynecology, Landeskrankenhaus, Paracelsus Medical University, Salzburg, Austria
| | - Franz Zehentmayr
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Peter Kopp
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Christoph Fussl
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| | - Thorsten Fischer
- Department of Special Gynecology, Landeskrankenhaus, Paracelsus Medical University, Salzburg, Austria
- Department of Gynecology, Landeskrankenhaus, Paracelsus Medical University, Salzburg, Austria
| | - Heinrich Deutschmann
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
- Institute for Research and Development of Advanced Radiation Technologies (radART), Paracelsus Medical University, Salzburg, Austria
| | - Felix Sedlmayer
- Department of Radiotherapy and Radio-Oncology, Landeskrankenhaus, Paracelsus Medical University, Muellner Hauptstrasse 48, 5020, Salzburg, Austria
| |
Collapse
|
24
|
Zheng HM, Chen C, Wu XH, Chen J, Sun S, Sun JZ, Wang MW, Sun SR. Quantum dot-based in situ simultaneous molecular imaging and quantitative analysis of EGFR and collagen IV and identification of their prognostic value in triple-negative breast cancer. Tumour Biol 2015; 37:2509-18. [PMID: 26385773 DOI: 10.1007/s13277-015-4079-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/13/2015] [Indexed: 12/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a unique breast cancer subtype with high heterogeneity and poor prognosis. Currently, the treatment effect of TNBC has reached a bottleneck, rendering new breakthroughs difficult. Cancer invasion is not an entirely cell-autonomous process, requiring the cells to transmigrate across the surrounding extracellular matrix (ECM) barriers. Developing a new system that integrates key constituents in the tumor microenvironment with pivotal cancer cell molecules is essential for the in-depth investigation of the mechanism of invasion in TNBC. We describe a computer-aided algorithm developed using quantum dot (QD)-based multiplex molecular imaging of TNBC tissues. We performed in situ simultaneous imaging and quantitative detection of epidermal growth factor receptor (EGFR), expressed in the TNBC cell membrane, and collagen IV, the major ECM constituent; calculated the EGFR/collagen IV ratio; and investigated the prognostic value of the EGFR/collagen IV ratio in TNBC. We simultaneously imaged and quantitatively detected EGFR and collagen IV in the TNBC samples. In all patients, quantitative determination showed a statistically significant negative correlation between EGFR and collagen IV. The 5-year disease-free survival (5-DFS) of the high and low EGFR/collagen IV ratio subgroups was significantly different. The EGFR/collagen IV ratio was predictive and was an independent prognostic indicator in TNBC. Compared with EGFR expression, the EGFR/collagen IV ratio had a greater prognostic value for 5-DFS. Our findings open up a new avenue for predicting the clinical outcome in TNBC from the perspective of integrating molecules expressed in both cancer cells and the ECM.
Collapse
Affiliation(s)
- Hong-Mei Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Xin-Hong Wu
- Department of Breast Surgery, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Jian Chen
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Jin-Zhong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Ming-Wei Wang
- Department of Pathology, Hubei Cancer Hospital, Wuhan, Hubei, People's Republic of China
| | - Sheng-Rong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Tower 238 Jiefang Road, Wuchang District, Wuhan, Hubei Province, 430060, People's Republic of China.
| |
Collapse
|
25
|
Kriegsmann M, Endris V, Wolf T, Pfarr N, Stenzinger A, Loibl S, Denkert C, Schneeweiss A, Budczies J, Sinn P, Weichert W. Mutational profiles in triple-negative breast cancer defined by ultradeep multigene sequencing show high rates of PI3K pathway alterations and clinically relevant entity subgroup specific differences. Oncotarget 2015; 5:9952-65. [PMID: 25296970 PMCID: PMC4259450 DOI: 10.18632/oncotarget.2481] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mutational profiling of triple-negative breast cancer (TNBC) by whole exome sequencing (WES) yielded a landscape of genomic alterations in this tumor entity. However, the clinical significance of these findings remains enigmatic. Further, integration of WES in routine diagnostics using formalin-fixed paraffin-embedded (FFPE) material is currently not feasible. Therefore, we designed and validated a breast cancer specific gene panel for semiconductor-based sequencing comprising 137 amplicons covering mutational hotspots in 44 genes and applied this panel on a cohort of 104 well-characterized FFPE TNBC with complete clinical follow-up. TP53 mutations were present in more than 80% of cases. PI3K pathway alterations (29.8%) comprising mainly PIK3CA mutations (22.1%) but also mutations and/or amplifications/deletions in other PI3K-associated genes (7.7%) were far more frequently observed, when compared to WES data. Alterations in MAPK signaling genes (8.7%) and cell-cycle regulators (14.4%) were also frequent. Mutational profiles were linked to TNBC subgroups defined by morphology and immunohistochemistry. Alterations in cell-cycle pathway regulators were linked with better overall (p=0.053) but not disease free survival. Taken together, we could demonstrate that breast cancer targeted hotspot sequencing is feasible in a routine setting and yields reliable and clinically meaningful results. Mutational spectra were linked to clinical and immunohistochemically defined parameters.
Collapse
Affiliation(s)
| | - Volker Endris
- Institute of Pathology, University of Heidelberg, Germany. German Cancer Research Center, Heidelberg, Germany
| | - Thomas Wolf
- Institute of Pathology, University of Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, University of Heidelberg, Germany
| | | | | | - Carsten Denkert
- Institute of Pathology, University Hospital Charité Berlin, Germany. German Cancer Consortium (DKTK), Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg, Germany. German Cancer Consortium (DKTK), Germany
| | - Jan Budczies
- Institute of Pathology, University Hospital Charité Berlin, Germany. German Cancer Consortium (DKTK), Germany
| | - Peter Sinn
- Institute of Pathology, University of Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University of Heidelberg, Germany. National Center for Tumor Diseases, Heidelberg, Germany. German Cancer Consortium (DKTK), Germany
| |
Collapse
|
26
|
S100P and HYAL2 as prognostic markers for patients with triple-negative breast cancer. Exp Mol Pathol 2015; 99:180-7. [PMID: 26112095 DOI: 10.1016/j.yexmp.2015.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 06/18/2015] [Indexed: 01/10/2023]
Abstract
Triple-negative breast cancer (TNBC) is a group of very aggressive breast tumours, characterised by lack of expression of oestrogen receptor (ER), progesterone receptor (PR) and erb-b2 receptor tyrosine kinase 2 (ERBB2/HER2). Nevertheless, TNBCs show different clinical characteristics and are very diverse regarding prognostic outcome. So far, only a few prognostic markers for TNBC have been reported that could be helpful for therapeutic stratification. Here we have analysed the expression of S100P and HYAL2 using immunohistochemistry (IHC) in a TNBC cohort of 98 patients with a follow-up for recurrence and death. TNBC patients with high expression of both proteins showed significantly shorter progression-free survival (PFS) (mean PFS=35.9months, P=0.001) compared to TNBC patients with high expression levels of only one of the proteins (mean PFS=69.4months) and to TNBC patients with low expression of both proteins (mean PFS=83.3months). Moreover, multivariate Cox-regression model showed the combined expression of S100P and HYAL2 as independent prognostic factor for PFS (P=0.001). The expression of S100P and HYAL2 indicated similar prognostic effect to the overall survival (OS) of TNBC patients. In addition, high expression levels of both S100P and HYAL2 showed significant association with different clinicopathological characteristics, such as more recurrence events (P=0.004), and higher occurrence of metastasis (P=0.002). Our study proposes S100P and HYAL2 as potential prognostic markers for TNBC.
Collapse
|
27
|
Pennati M, Sbarra S, De Cesare M, Lopergolo A, Locatelli SL, Campi E, Daidone MG, Carlo-Stella C, Gianni AM, Zaffaroni N. YM155 sensitizes triple-negative breast cancer to membrane-bound TRAIL through p38 MAPK- and CHOP-mediated DR5 upregulation. Int J Cancer 2014; 136:299-309. [PMID: 24866585 DOI: 10.1002/ijc.28993] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/12/2014] [Indexed: 12/30/2022]
Abstract
Because available treatments have limited efficacy in triple-negative breast cancer (TNBC), the identification of new therapeutic strategies to improve patients' outcome is urgently needed. In our study, we investigated the effects of the administration of the small molecule selective survivin suppressant YM155, alone or in association with CD34+ cells transduced with a replication-deficient adenovirus encoding the human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene (CD34-TRAIL+ cells), in three TNBC cell models. YM155 exposure significantly impaired TNBC cell growth and selectively modulated survivin expression at both mRNA and protein level. In addition, co-culturing YM155-treated TNBC cells with CD34-TRAIL+ cells resulted in markedly increased cytotoxic effect and apoptotic response in comparison with single treatments. Such a chemosensitizing effect was observed only in TNBC cells inherently expressing DR5 and relied on the ability of YM155 to upregulate DR5 expression through a p38 MAPK- and CHOP-dependent mechanism. YM155/CD34-TRAIL+ combination also showed a significant inhibitory effect on the growth of DR5-expressing TNBC cells following xenotransplantation into NOD/SCID mice, in the absence of toxicity. Overall, our data (i) provide, for the first time, evidence that YM155 sensitizes TNBC cells to CD34-TRAIL+ cells-induced apoptosis by a mechanism involving the downregulation of survivin and the simultaneous p38 MAPK- and CHOP-mediated upregulation of DR5, and (ii) suggest the combination of YM155 with TRAIL-armed CD34+ progenitor cells as a promising therapeutic option for patients with TNBC expressing DR5.
Collapse
Affiliation(s)
- Marzia Pennati
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gasparini P, Fassan M, Cascione L, Guler G, Balci S, Irkkan C, Paisie C, Lovat F, Morrison C, Zhang J, Scarpa A, Croce CM, Shapiro CL, Huebner K. Androgen receptor status is a prognostic marker in non-basal triple negative breast cancers and determines novel therapeutic options. PLoS One 2014; 9:e88525. [PMID: 24505496 PMCID: PMC3914993 DOI: 10.1371/journal.pone.0088525] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022] Open
Abstract
Triple negative breast cancers are a heterogeneous group of tumors characterized by poor patient survival and lack of targeted therapeutics. Androgen receptor has been associated with triple negative breast cancer pathogenesis, but its role in the different subtypes has not been clearly defined. We examined androgen receptor protein expression by immunohistochemical analysis in 678 breast cancers, including 396 triple negative cancers. Fifty matched lymph node metastases were also examined. Association of expression status with clinical (race, survival) and pathological (basal, non-basal subtype, stage, grade) features was also evaluated. In 160 triple negative breast cancers, mRNA microarray expression profiling was performed, and differences according to androgen receptor status were analyzed. In triple negative cancers the percentage of androgen receptor positive cases was lower (24.8% vs 81.6% of non-triple negative cases), especially in African American women (16.7% vs 25.5% of cancers of white women). No significant difference in androgen receptor expression was observed in primary tumors vs matched metastatic lesions. Positive androgen receptor immunoreactivity was inversely correlated with tumor grade (p<0.01) and associated with better overall patient survival (p = 0.032) in the non-basal triple negative cancer group. In the microarray study, expression of three genes (HER4, TNFSF10, CDK6) showed significant deregulation in association with androgen receptor status; eg CDK6, a novel therapeutic target in triple negative cancers, showed significantly higher expression level in androgen receptor negative cases (p<0.01). These findings confirm the prognostic impact of androgen receptor expression in non-basal triple negative breast cancers, and suggest targeting of new androgen receptor-related molecular pathways in patients with these cancers.
Collapse
Affiliation(s)
- Pierluigi Gasparini
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Matteo Fassan
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona Italy
- * E-mail:
| | - Luciano Cascione
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Gulnur Guler
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Serdar Balci
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Cigdem Irkkan
- Department of Pathology, Hacettepe University, Ankara Turkey
| | - Carolyn Paisie
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Francesca Lovat
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Carl Morrison
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Jianying Zhang
- Bioinformatics Shared Resource, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | - Aldo Scarpa
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona Italy
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Charles L. Shapiro
- Division of Medical Oncology and the Breast Program, James Cancer Hospital and Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, United States of America
| |
Collapse
|
29
|
Reproductive risk factors and breast cancer subtypes: a review of the literature. Breast Cancer Res Treat 2014; 144:1-10. [PMID: 24477977 DOI: 10.1007/s10549-014-2852-7] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 01/20/2014] [Indexed: 12/31/2022]
Abstract
Aside from age, sex, and family history, risk of developing breast cancer is largely linked to reproductive factors, which characterize exposure to sex hormones. Given that, molecular testing at the tumor level is currently possible, clinical characterization of tumor subtypes is routinely conducted to guide treatment decisions. However, despite the vast amount of published data from observational studies on reproductive factor associations and breast cancer risk, relatively fewer reports have been published on associations specific to breast tumor subtypes. We conducted a review of the literature and summarized the results of associations between reproductive factors and risk or odds of three distinct tumor subtypes: estrogen receptor/progesterone receptor positive (hormone receptor positive, HR+ tumors), tumors overexpressing the human epidermal receptor 2 protein (HER2+), and triple negative breast cancer (TNBC), which lacks the three markers. Results show that the most consistent evidence for associations with reproductive risk factors exists for HR+ breast cancers, with nulliparity, current use of menopausal hormone therapy, and prolonged interval between menarche and age at first birth being the strongest risk factors; increased age at first birth and decreased age at menarche were fairly consistently associated with HR+ cancers; and though less consistent, older age at menopause was also positively associated, while lactation was inversely associated with HR+ tumors. Fewer consistent associations have been reported for TNBC. The single protective factor most consistently associated with TNBC was longer duration of breastfeeding. Increased parity, younger age at first birth, older age at menarche, and oral contraceptive use were less consistently shown to be associated with TNBC. No remarkable associations for HER2+ breast cancers were evident, although this was based on relatively scarce data. Findings suggest heterogeneity in reproductive risk factors for the distinct subtypes of breast tumors, which may have implications for recommended prevention strategies.
Collapse
|
30
|
Sinn P, Aulmann S, Wirtz R, Schott S, Marmé F, Varga Z, Lebeau A, Kreipe H, Schneeweiss A. Multigene Assays for Classification, Prognosis, and Prediction in Breast Cancer: a Critical Review on the Background and Clinical Utility. Geburtshilfe Frauenheilkd 2013; 73:932-940. [PMID: 24771945 DOI: 10.1055/s-0033-1350831] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 08/24/2013] [Accepted: 08/25/2013] [Indexed: 12/14/2022] Open
Abstract
Gene signatures which are based on multigene profiling assays have been developed for the purpose to better define the prognosis and prediction of therapy results in early-stage breast cancer. These assays were designed to be more specific than conventional clinico-pathologic parameters in the selection of patients for (neo-)adjuvant treatment and in effect help to avoid unnecessary cytotoxic treatment. In this review we describe molecular risk scores, for which tests are commercially available (PAM50®, MammaTyper®, MammaPrint®, Oncotype DX®, Endopredict®, Genomic Grade Index®) and IHC risk scores (Mammostrat® and IHC4), and discuss the current evidence of their clinical use.
Collapse
Affiliation(s)
- P Sinn
- Department of Pathology, University of Heidelberg, Heidelberg
| | - S Aulmann
- Department of Pathology, University of Heidelberg, Heidelberg
| | - R Wirtz
- Stratifyer Molecular Pathology GmbH, Köln
| | - S Schott
- Department of Gynaecology and Obstetrics, University of Heidelberg, Heidelberg
| | - F Marmé
- Department of Gynaecology and Obstetrics, University of Heidelberg, Heidelberg
| | - Z Varga
- Institute of Surgical Pathology, University Hospital Zürich, Zürich, Switzerland
| | - A Lebeau
- Dept. of Pathology, University Medical Canter Hamburg-Eppendorf, Hamburg
| | - H Kreipe
- Institute of Pathology, Medizinische Hochschule Hannover, Hannover
| | | |
Collapse
|
31
|
Lee KL, Kuo YC, Ho YS, Huang YH. Isolation and characterization of Pseudomonas aeruginosa PAO mutant that produces altered elastase. J Bacteriol 1980; 11:cancers11091334. [PMID: 31505803 PMCID: PMC6769912 DOI: 10.3390/cancers11091334] [Citation(s) in RCA: 144] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is cancer that tested as negative for estrogen receptors (ER), progesterone receptors (PR), and excess human epidermal growth factor receptor 2 (HER2) protein which accounts for 15%–20% of all breast cancer cases. TNBC is considered to be a poorer prognosis than other types of breast cancer, mainly because it involves more aggressive phenotypes that are similar to stem cell–like cancer cells (cancer stem cell, CSC). Thus, targeted treatment of TNBC remains a major challenge in clinical practice. This review article surveys the latest evidence concerning the role of genomic alteration in current TNBC treatment responses, current clinical trials and potential targeting sites, CSC and drug resistance, and potential strategies targeting CSCs in TNBC. Furthermore, the role of insulin-like growth factor 1 receptor (IGF-1R) and nicotinic acetylcholine receptors (nAChR) in stemness expression, chemoresistance, and metastasis in TNBC and their relevance to potential treatments are also discussed and highlighted.
Collapse
Affiliation(s)
- Kha-Liang Lee
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yung-Che Kuo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan.
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|