1
|
Wang J, Li B, Luo M, Huang J, Zhang K, Zheng S, Zhang S, Zhou J. Progression from ductal carcinoma in situ to invasive breast cancer: molecular features and clinical significance. Signal Transduct Target Ther 2024; 9:83. [PMID: 38570490 PMCID: PMC10991592 DOI: 10.1038/s41392-024-01779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024] Open
Abstract
Ductal carcinoma in situ (DCIS) represents pre-invasive breast carcinoma. In untreated cases, 25-60% DCIS progress to invasive ductal carcinoma (IDC). The challenge lies in distinguishing between non-progressive and progressive DCIS, often resulting in over- or under-treatment in many cases. With increasing screen-detected DCIS in these years, the nature of DCIS has aroused worldwide attention. A deeper understanding of the biological nature of DCIS and the molecular journey of the DCIS-IDC transition is crucial for more effective clinical management. Here, we reviewed the key signaling pathways in breast cancer that may contribute to DCIS initiation and progression. We also explored the molecular features of DCIS and IDC, shedding light on the progression of DCIS through both inherent changes within tumor cells and alterations in the tumor microenvironment. In addition, valuable research tools utilized in studying DCIS including preclinical models and newer advanced technologies such as single-cell sequencing, spatial transcriptomics and artificial intelligence, have been systematically summarized. Further, we thoroughly discussed the clinical advancements in DCIS and IDC, including prognostic biomarkers and clinical managements, with the aim of facilitating more personalized treatment strategies in the future. Research on DCIS has already yielded significant insights into breast carcinogenesis and will continue to pave the way for practical clinical applications.
Collapse
Affiliation(s)
- Jing Wang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Meng Luo
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
- Department of Plastic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Huang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China
| | - Suzhan Zhang
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
| | - Jiaojiao Zhou
- The Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Breast Surgery and Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Mitchell E, Jindal S, Chan T, Narasimhan J, Sivagnanam S, Gray E, Chang YH, Weinmann S, Schedin P. Loss of myoepithelial calponin-1 characterizes high-risk ductal carcinoma in situ cases, which are further stratified by T cell composition. Mol Carcinog 2020; 59:701-712. [PMID: 32134153 PMCID: PMC7317523 DOI: 10.1002/mc.23171] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/15/2022]
Abstract
A hallmark of ductal carcinoma in situ (DCIS) progression is a loss of the surrounding ductal myoepithelium. However, whether compromise in myoepithelial differentiation, rather than overt cellular loss, can be used to predict the risk of DCIS progression is unknown. Here we address this question utilizing pure and mixed DCIS cases (N = 30) as surrogates for DCIS at low and high risk for progression, respectively. We used multiplex immunohistochemical staining to evaluate the relationship between myoepithelial cell differentiation and lymphoid immune cell types associated with poor prognostic DCIS. Our results show that myoepithelial calponin-1 discriminates between pure and mixed DCIS lesions better than histological subtype, presence of necrosis, or nuclear grade. Additionally, focal loss of myoepithelial cells associated with increased PD-1+CD8+ T cells, which suggests a link between the myoepithelium and immune surveillance. To identify associations between calponin-1 expression and immune response, we performed unsupervised hierarchical clustering of myoepithelial and immune cell biomarkers on 219 DCIS lesions from 30 cases. Notably, the majority of pure (low-risk) DCIS lesions clustered in a high calponin-1, T cell low group, whereas the majority of mixed (high-risk) DCIS lesions clustered in a low calponin-1, T cell high group, specifically with CD8+ and PD-1+CD8+ T cells. However, a subset of pure DCIS lesions had a similar calponin-1 and immune signature as the majority of mixed DCIS lesions, which have low calponin-1 and T cell enrichment-raising the possibility that these pure DCIS lesions might be at a high risk for progression.
Collapse
Affiliation(s)
- Elizabeth Mitchell
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Sonali Jindal
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
- Cancer Prevention and Control, Knight Cancer InstituteOregon Health and Science UniversityPortlandOregon
| | - Tiffany Chan
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Jayasri Narasimhan
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Shamilene Sivagnanam
- Computational Biology Program, Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
| | - Elliot Gray
- Department of Biomedical Engineering, Oregon Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandOregon
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Center for Spatial Systems BiomedicineOregon Health and Science UniversityPortlandOregon
| | - Sheila Weinmann
- Center for Health ResearchKaiser Permanente NorthwestPortlandOregon
| | - Pepper Schedin
- Department of Cell, Developmental, and Cancer BiologyOregon Health and Science UniversityPortlandOregon
- Cancer Prevention and Control, Knight Cancer InstituteOregon Health and Science UniversityPortlandOregon
| |
Collapse
|
3
|
Rakha EA, Miligy IM, Gorringe KL, Toss MS, Green AR, Fox SB, Schmitt FC, Tan PH, Tse GM, Badve S, Decker T, Vincent-Salomon A, Dabbs DJ, Foschini MP, Moreno F, Wentao Y, Geyer FC, Reis-Filho JS, Pinder SE, Lakhani SR, Ellis IO. Invasion in breast lesions: the role of the epithelial-stroma barrier. Histopathology 2018; 72:1075-1083. [DOI: 10.1111/his.13446] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Emad A Rakha
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Islam M Miligy
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Kylie L Gorringe
- Cancer Genomics Program; Peter MacCallum Cancer Centre; Melbourne Vic. Australia
- The Sir Peter MacCallum Department of Oncology; University of Melbourne; Melbourne Vic. Australia
| | - Michael S Toss
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Andrew R Green
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| | - Stephen B Fox
- Pathology Department; Peter MacCallum Cancer Centre; Melbourne Vic. Australia
| | - Fernando C Schmitt
- Institute of Molecular Pathology and Immunology (IPATIMUP) and Medical Faculty; University of Porto; Porto Portugal
| | - Puay-Hoon Tan
- Department of Pathology; Singapore General Hospital; Singapore
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology; Prince of Wales Hospital; Hong Kong
| | - Sunil Badve
- Departments of Pathology and Internal Medicine; Clarian Pathology Laboratory of Indiana University; Indianapolis IN USA
| | - Thomas Decker
- Breast-Screening-Pathology; Reference Centre Munster; Gerhard Domagk-Institute of Pathology; University Hospital Münster; Münster Germany
| | | | - David J Dabbs
- University of Pittsburgh Medical Centre; Pittsburgh PA USA
| | - Maria P Foschini
- Department of Biomedical and Neuromotor Sciences; Section of Anatomic Pathology at Bellaria Hospital; University of Bologna; Bologna Italy
| | - Filipa Moreno
- Anatomic Pathology Department; Centro Hospitalar do Porto; Porto Portugal
| | - Yang Wentao
- Pathology; Fudan University Shanghai Cancer Center; Shanghai China
| | - Felipe C Geyer
- Department of Pathology; Memorial Sloan Kettering Cancer Centre; New York NY USA
| | - Jorge S Reis-Filho
- Department of Pathology; Memorial Sloan Kettering Cancer Centre; New York NY USA
| | - Sarah E Pinder
- Division of Cancer Studies; King's College London; Guy's Hospital; London UK
| | - Sunil R Lakhani
- Discipline of Molecular & Cellular Pathology; Faculty of Medicine; University of Queensland; The Royal Brisbane & Women's Hospital; Brisbane QLD Australia
| | - Ian O Ellis
- Department of Histopathology; Nottingham City Hospital NHS Trust; Nottingham University; Nottingham UK
| |
Collapse
|
4
|
Kumar S, Desmedt C, Larsimont D, Sotiriou C, Goormaghtigh E. Change in the microenvironment of breast cancer studied by FTIR imaging. Analyst 2013; 138:4058-65. [PMID: 23662300 DOI: 10.1039/c3an00241a] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Fourier transform infrared (FTIR) imaging was applied on histopathological specimens of breast cancer of different tumor histological grades. Focus was given to the extracellular matrix. FTIR spectral changes were observed when examining the extracellular matrix close to and far from carcinoma. Major changes were observed, in particular in the relative intensities of the collagen bands at 1640 and 1630 cm(-1). PCA analysis and global fitting indicate a continuous progression in collagen spectral features when moving away from the tumor. These preliminary results suggest FTIR spectral features present in the 1700-1600 cm(-1) spectral range could be used as spectral markers to identify cancer-induced modifications in collagen. This chemical imaging approach to analyze the breast cancer microenvironment could be used in the future for improving diagnostics of breast cancer.
Collapse
Affiliation(s)
- S Kumar
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics, Université Libre de Bruxelles, Campus Plaine CP 206/2 Bâtiment BC, Blvd. du Triomphe, B-1050 Bruxelles, Belgium
| | | | | | | | | |
Collapse
|
5
|
Khamis ZI, Sahab ZJ, Sang QXA. Active roles of tumor stroma in breast cancer metastasis. Int J Breast Cancer 2012; 2012:574025. [PMID: 22482059 PMCID: PMC3296264 DOI: 10.1155/2012/574025] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 11/04/2011] [Accepted: 11/11/2011] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the major cause of death for breast cancer patients. Tumors are heterogenous cellular entities composed of cancer cells and cells of the microenvironment in which they reside. A reciprocal dynamic interaction occurs between the tumor cells and their surrounding stroma under physiological and pathological conditions. This tumor-host communication interface mediates the escape of tumor cells at the primary site, survival of circulating cancer cells in the vasculature, and growth of metastatic cancer at secondary site. Each step of the metastatic process is accompanied by recruitment of stromal cells from the microenvironment and production of unique array of growth factors and chemokines. Stromal microenvironment may play active roles in breast cancer metastasis. Elucidating the types of cells recruited and signal pathways involved in the crosstalk between tumor cells and stromal cells will help identify novel strategies for cotargeting cancer cells and tumor stromal cells to suppress metastasis and improve patient outcome.
Collapse
Affiliation(s)
- Zahraa I. Khamis
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4390, USA
| | - Ziad J. Sahab
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4390, USA
| |
Collapse
|
6
|
Novel stromal biomarkers in human breast cancer tissues provide evidence for the more malignant phenotype of estrogen receptor-negative tumors. J Biomed Biotechnol 2011; 2011:723650. [PMID: 21976967 PMCID: PMC3185288 DOI: 10.1155/2011/723650] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 07/19/2011] [Indexed: 01/25/2023] Open
Abstract
Research efforts were focused on genetic alterations in epithelial cancer cells. Epithelial-stromal interactions play a crucial role in cancer initiation, progression, invasion, angiogenesis, and metastasis; however, the active role of stroma in human breast tumorigenesis in relation to estrogen receptor (ER) status of epithelial cells has not been explored. Using proteomics and biochemical approaches, we identified two stromal proteins in ER-positive and ER-negative human breast cancer tissues that may affect malignant transformation in breast cancer. Two putative biomarkers, T-cell receptor alpha (TCR-α) and zinc finger and BRCA1-interacting protein with a KRAB domain (ZBRK1), were detected in leukocytes of ER-positive and endothelial cells of ER-negative tissues, respectively. Our data suggest an immunosuppressive role of leukocytes in invasive breast tumors, propose a multifunctional nature of ZBRK1 in estrogen receptor regulation and angiogenesis, and demonstrate the aggressiveness of ER-negative human breast carcinomas. This research project may identify new stromal drug targets for the treatment of breast cancer patients.
Collapse
|
7
|
Sahab ZJ, Man YG, Byers SW, Sang QXA. Putative biomarkers and targets of estrogen receptor negative human breast cancer. Int J Mol Sci 2011; 12:4504-21. [PMID: 21845093 PMCID: PMC3155366 DOI: 10.3390/ijms12074504] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 06/27/2011] [Accepted: 07/04/2011] [Indexed: 11/26/2022] Open
Abstract
Breast cancer is a progressive and potentially fatal disease that affects women of all ages. Like all progressive diseases, early and reliable diagnosis is the key for successful treatment and annihilation. Biomarkers serve as indicators of pathological, physiological, or pharmacological processes. Her2/neu, CA15.3, estrogen receptor (ER), progesterone receptor (PR), and cytokeratins are biomarkers that have been approved by the Food and Drug Administration for disease diagnosis, prognosis, and therapy selection. The structural and functional complexity of protein biomarkers and the heterogeneity of the breast cancer pathology present challenges to the scientific community. Here we review estrogen receptor-related putative breast cancer biomarkers, including those of putative breast cancer stem cells, a minor population of estrogen receptor negative tumor cells that retain the stem cell property of self-renewal. We also review a few promising cytoskeleton targets for ER alpha negative breast cancer.
Collapse
Affiliation(s)
- Ziad J. Sahab
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA; E-Mail:
| | - Yan-Gao Man
- Diagnostic and Translational Research Center, Henry Jackson Foundation for the Advancement of Military Medicine, Gaithersburg, MD 20789, USA; E-Mail:
- Jilin University, Changchun 130012, China
| | - Stephen W. Byers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA; E-Mail:
| | - Qing-Xiang A. Sang
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, 102 Varsity Way, Tallahassee, FL 32306, USA; E-Mail:
| |
Collapse
|
8
|
Khamis ZI, Iczkowski KA, Sang QXA. Metastasis suppressors in human benign prostate, intraepithelial neoplasia, and invasive cancer: their prospects as therapeutic agents. Med Res Rev 2011; 32:1026-77. [PMID: 22886631 DOI: 10.1002/med.20232] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite advances in diagnosis and treatment of prostate cancer, development of metastases remains a major clinical challenge. Research efforts are dedicated to overcome this problem by understanding the molecular basis of the transition from benign cells to prostatic intraepithelial neoplasia (PIN), localized carcinoma, and metastatic cancer. Identification of proteins that inhibit dissemination of cancer cells will provide new perspectives to define novel therapeutics. Development of antimetastatic drugs that trigger or mimic the effect of metastasis suppressors represents new therapeutic approaches to improve patient survival. This review focuses on different biochemical and cellular functions of metastasis suppressors known to play a role in prostate carcinogenesis and progression. Ten putative metastasis suppressors implicated in prostate cancer are discussed. CD44s is decreased in both PIN and cancer; Drg-1, E-cadherin, KAI-1, RKIP, and SSeCKS show similar expression between benign epithelia and PIN, but are downregulated in invasive cancer; whereas, maspin, MKK4, Nm23 and PTEN are upregulated in PIN and downregulated in cancer. Moreover, the potential role of microRNA in prostate cancer progression, the understanding of the cellular distribution and localization of metastasis suppressors, their mechanism of action, their effect on prostate invasion and metastasis, and their potential use as therapeutics are addressed.
Collapse
Affiliation(s)
- Zahraa I Khamis
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306-4390, USA
| | | | | |
Collapse
|
9
|
Hsiao YH, Chou MC, Fowler C, Mason JT, Man YG. Breast cancer heterogeneity: mechanisms, proofs, and implications. J Cancer 2010; 1:6-13. [PMID: 20842218 PMCID: PMC2931354 DOI: 10.7150/jca.1.6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Human breast cancer represents a group of highly heterogeneous lesions consisting of about 20 morphologically distinct subtypes with substantially different molecular and/or biochemical signatures, clinical courses, and prognoses. This study analyzed the possible correlation between the morphological presentations of breast cancer and two hypothesized models of carcinogenesis, in order to identify the intrinsic mechanism(s) and clinical implications of breast cancer heterogeneity.
Collapse
Affiliation(s)
- Yi-Hsuan Hsiao
- 1. Institute of Medicine, Chung Shan Medical University, Taiwan
| | | | | | | | | |
Collapse
|
10
|
Contributions of leukocytes to tumor invasion and metastasis: the "piggy-back" hypothesis. Cancer Epidemiol 2009; 34:3-6. [PMID: 20022838 DOI: 10.1016/j.canep.2009.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 11/18/2009] [Indexed: 11/29/2022]
|
11
|
Zhang X, Hashemi SS, Yousefi M, Gao C, Sheng J, Ni J, Wang W, Mason J, Man YG. Atypical E-cadherin expression in cell clusters overlying focally disrupted mammary myoepithelial cell layers: implications for tumor cell motility and invasion. Pathol Res Pract 2009; 205:375-85. [PMID: 19395181 DOI: 10.1016/j.prp.2008.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 06/17/2008] [Accepted: 08/19/2008] [Indexed: 01/09/2023]
Abstract
Our recent studies showed that cell clusters overlying focal myoepithelial cell layer disruptions (FMCLD) had a significantly higher rate of ER negativity, genetic instabilities, and expression of invasion-related genes than adjacent cells within the same duct. This study attempted to determine if these cells would show aberrant E-cadherin expression, which imparts greater propensity for cell motility and invasion. Consecutive sections from breast tumors with a high frequency of FMCLD were double-immunostained for E-cadherin and a panel of related markers. The E-cadherin mRNA levels in cells overlying FMCLD and adjacent cells within the same duct were compared using real-time PCR. Nearly all the cell clusters overlying FMCLD were strongly immunoreactive for E-cadherin, whereas their adjacent counterparts within the same duct were largely negative. Cell clusters overlying FMCLD were generally arranged as tongue-like projections, "puncturing" deep into the stroma or tube-like structures that often contained red blood cells. The sub-cellular localization of E-cadherin in the above structures, however, was primarily cytoplasmic. The mRNA level of E-cadherin in cell clusters overlying FMCLD was significantly higher than that in adjacent cells within the same duct. These findings suggest that aberrant expression of E-cadherin may contribute to cell motility and invasion.
Collapse
Affiliation(s)
- Xichen Zhang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Xu Z, Wang W, Deng CX, Man YG. Aberrant p63 and WT-1 expression in myoepithelial cells of pregnancy-associated breast cancer: implications for tumor aggressiveness and invasiveness. Int J Biol Sci 2009; 5:82-96. [PMID: 19173015 PMCID: PMC2631157 DOI: 10.7150/ijbs.5.82] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 01/08/2009] [Indexed: 11/27/2022] Open
Abstract
Our recent studies revealed that focal alterations in breast myoepithelial cell layers significantly impact the biological presentation of associated epithelial cells. As pregnancy-associated breast cancer (PABC) has a significantly more aggressive clinical course and mortality rate than other forms of breast malignancies, our current study compared tumor suppressor expression in myoepithelial cells of PABC and non-PABC, to determine whether myoepithelial cells of PABC may have aberrant expression of tumor suppressors. Tissue sections from 20 cases of PABC and 20 cases of stage, grade, and age matched non-PABC were subjected to immunohistochemistry, and the expression of tumor suppressor maspin, p63, and Wilms' tumor 1 (WT-1) in calponin positive myoepithelial cells were statistically compared. The expression profiles of maspin, p63, and WT-1 in myoepithelial cells of all ducts encountered were similar between PABC and non-PABC. PABC, however, displayed several unique alterations in terminal duct and lobular units (TDLU), acini, and associated tumor tissues that were not seen in those of non-PABC, which included the absence of p63 and WT-1 expression in a vast majority of the myoepithelial cells, cytoplasmic localization of p63 in the entire epithelial cell population of some lobules, and substantially increasing WT-1 expression in vascular structures of the invasive cancer component. All or nearly all epithelial cells with aberrant p63 and WT-1 expression lacked the expression of estrogen receptor and progesterone receptor, whereas they had a substantially higher proliferation index than their counterparts with p63 and WT-1 expression. Hyperplastic cells with cytoplasmic p63 expression often adjacent to, and share a similar immunohistochemical and cytological profile with, invasive cancer cells. To our best knowledge, our main finings have not been previously reported. Our findings suggest that the functional status of myoepithelial cells may be significantly associated with tumor aggressiveness and invasiveness.
Collapse
Affiliation(s)
- Zheli Xu
- 1. Department of Thyroid and Breast Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Wan Wang
- 1. Department of Thyroid and Breast Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Chu-Xia Deng
- 2. Mammalian Genetics Section, GDDB, NIDDK, National Institutes of Health, Bethesda, MD, USA
| | - Yan-gao Man
- 3. Department of Gynecologic and Breast Pathology, Armed Forces Institute of Pathology and American Registry of Pathology, Washington DC, USA
| |
Collapse
|
13
|
Digital image analysis of breast epithelial cells collected by random periareolar fine-needle aspirates (RPFNA) from women at high risk for breast cancer taking hormone replacement and the aromatase inhibitor, letrozole, for six months. Breast Cancer Res Treat 2009; 115:661-8. [DOI: 10.1007/s10549-008-0274-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 12/09/2008] [Indexed: 11/26/2022]
|
14
|
Zhang X, Hashemi SS, Yousefi M, Ni J, Wang Q, Gao L, Gong P, Gao C, Sheng J, Mason J, Man YG. Aberrant c-erbB2 expression in cell clusters overlying focally disrupted breast myoepithelial cell layers: a trigger or sign for emergence of more aggressive cell clones? Int J Biol Sci 2008; 4:259-69. [PMID: 18726004 PMCID: PMC2519838 DOI: 10.7150/ijbs.4.259] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Accepted: 08/07/2008] [Indexed: 12/26/2022] Open
Abstract
Our recent studies revealed that cell clusters overlying focal myoepithelial cell layer disruption (FMCLD) had a significantly higher frequency of genetic instabilities and expression of invasion-related genes than their adjacent counterparts within the same duct. Our current study attempted to assess whether these cell clusters would also have elevated c-erbB2 expression. Human breast tumors (n=50) with a high frequency of FMCLD were analyzed with double immunohistochemistry, real-time RT-PCR, and chromogenic in situ hybridization for c-erbB2 protein and gene expression. Of 448 FMCLD detected, 404 (90.2%) were associated with cell clusters that had intense c-erbB2 immunoreactivities primarily in their cytoplasm, in contrast to their adjacent counterparts within the same duct, which had no or barely detectable c-erbB2 expression. These c-erbB2 positive cells were arranged as tongue-like projections, "puncturing" into the stroma, and about 20% of them were in direct continuity with tube-like structures that resembled blood vessels. Aberrant c-erbB2 expression was also seen in clusters of architecturally normal-appearing ducts that had distinct cytological abnormalities in both ME and epithelial cells, whereas not in their clear-cut normal counterparts. Molecular assays detected markedly higher c-erbB2 mRNA and gene amplification in cell clusters associated with FMCLD than in those associated with non-disrupted ME cell layers. Our findings suggest that cell clusters overlying FMCLD may represent the precursors of pending invasive lesions, and that aberrant c-erbB2 expression may trigger or signify the emergence of biologically more aggressive cell clones.
Collapse
Affiliation(s)
- Xichen Zhang
- 1. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shahreyar Shar Hashemi
- 2. Departments of Surgery and Internal Medicine, Staten Island University Hospital, NY, USA
| | - Morvarid Yousefi
- 2. Departments of Surgery and Internal Medicine, Staten Island University Hospital, NY, USA
| | - Jinsong Ni
- 3. Norman Bethune College of Medical Science, Jilin University, Changchun, Jilin, China
| | - Qiuyue Wang
- 1. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Ling Gao
- 1. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Pengtao Gong
- 1. College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Chunling Gao
- 4. Division of Monoclonal Antibodies, Food and Drug Administration, Bethesda, MD, USA
| | - Joy Sheng
- 5. Real-time PCR Technical Support Department, Applied Biosystems, Foster City, CA, USA
| | - Jeffrey Mason
- 6. Armed Forces Institute of Pathology and American Registry of Pathology, Washington, DC, USA
| | - Yan-gao Man
- 6. Armed Forces Institute of Pathology and American Registry of Pathology, Washington, DC, USA
| |
Collapse
|
15
|
Man YG, Gardner WA. Bad seeds produce bad crops: a single stage-process of prostate tumor invasion. Int J Biol Sci 2008; 4:246-58. [PMID: 18725981 PMCID: PMC2519176 DOI: 10.7150/ijbs.4.246] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Accepted: 08/06/2008] [Indexed: 11/24/2022] Open
Abstract
It is a commonly held belief that prostate carcinogenesis is a multi-stage process and that tumor invasion is triggered by the overproduction of proteolytic enzymes. This belief is consistent with data from cell cultures and animal models, whereas is hard to interpret several critical facts, including the presence of cancer in "healthy" young men and cancer DNA phenotype in morphologically normal prostate tissues. These facts argue that alternative pathways may exist for prostate tumor invasion in some cases. Since degradation of the basal cell layer is the most distinct sign of invasion, our recent studies have attempted to identify pre-invasive lesions with focal basal cell layer alterations. Our studies revealed that about 30% of prostate cancer patients harbored normal appearing duct or acinar clusters with a high frequency of focal basal cell layer disruptions. These focally disrupted basal cell layers had significantly reduced cell proliferation and tumor suppressor expression, whereas significantly elevated degeneration, apoptosis, and infiltration of immunoreactive cells. In sharp contrast, associated epithelial cell had significantly elevated proliferation, expression of malignancy-signature markers, and physical continuity with invasive lesions. Based on these and other findings, we have proposed that these normal appearing duct or acinar clusters are derived from monoclonal proliferation of genetically damaged stem cells and could progress directly to invasion through two pathways: 1) clonal in situ transformation (CIST) and 2) multi-potential progenitor mediated "budding" (MPMB). These pathways may contribute to early onset of prostate cancer at young ages, and to clinically more aggressive prostate tumors.
Collapse
Affiliation(s)
- Yan-gao Man
- American Registry of Pathology and Armed Forces Institute of Pathology, Washington DC 20306-6000, USA.
| | | |
Collapse
|
16
|
Hu M, Polyak K. Microenvironmental regulation of cancer development. Curr Opin Genet Dev 2008; 18:27-34. [PMID: 18282701 DOI: 10.1016/j.gde.2007.12.006] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 12/21/2007] [Accepted: 12/28/2007] [Indexed: 11/30/2022]
Abstract
Numerous studies have demonstrated that the tumor microenvironment not only responds to and supports carcinogenesis, but also actively contributes to tumor initiation, progression, and metastasis. During tumor progression all cells composing the tumor undergo phenotypic and epigenetic changes. Paracrine signaling between epithelial and stromal cells is important for the regulation of the proliferation, invasive, angiogenic, and metastatic behavior of cancer cells. Better understanding the molecular mechanisms by which stromal cells exert these effects may open up new venues for cancer therapeutic and preventative interventions.
Collapse
Affiliation(s)
- Min Hu
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney Street, D740C, Boston, MA 02115, USA
| | | |
Collapse
|
17
|
|
18
|
Focal degeneration of basal cells and the resultant auto-immunoreactions: a novel mechanism for prostate tumor progression and invasion. Med Hypotheses 2007; 70:387-408. [PMID: 17658698 DOI: 10.1016/j.mehy.2007.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Accepted: 05/14/2007] [Indexed: 02/01/2023]
Abstract
The development of human prostate cancer is believed to be a multistep process, progressing sequentially from normal, to hyperplasia, to prostatic intraepithelial neoplasia (PIN), and to invasive and metastatic lesions. High grade PIN has been generally considered as the direct precursor of invasive lesions, and the progression of PIN is believed to be triggered primarily, if not solely, by the overproduction of proteolytic enzymes predominately by cancer cells, which result in the degradation of the basement membrane. These theories, however, are hard to reconcile with two main facts: (1) only about 30% untreated PIN progress to invasive stage, while none of the current approaches could accurately identify the specific PIN or individuals at greater risk for progression, and (2) results from recent world-wide clinical trials with a wide variety of proteolytic enzyme inhibitors have been very disappointing, casting doubt on the validity of the proteolytic enzyme theory. Since over 90% of prostate cancer-related deaths result from invasion-related illness and the incidence of PIN could be up to 16.5-25% in routine or ultrasound guided prostate biopsy, there is an urgent need to uncover the intrinsic mechanism of prostate tumor invasion. Promoted by the facts that the basal cell population is the source of several tumor suppressors and the absence of the basal cell layer is the most distinct feature of invasive lesions, our recent studies have intended to identify the early alterations of basal cell layers and their impact on tumor invasion using multidisciplinary approaches. Our studies revealed that a subset of pre-invasive tumors contained focal disruptions (the absence of basal cells resulting in a gap greater than the combined size of at least three epithelial cells) in surrounding basal cell layers. Compared to their non-disrupted counterparts, focally disrupted basal cell layers had several unique features: (1) significantly lower proliferation; (2) significantly lower p63 expression; (3) significantly higher apoptosis; and (4) significantly higher leukocyte infiltration and stromal reactions. Compared to their counterparts distant from focal disruptions or overlying non-disrupted basal cell layers, epithelial cells overlying focal basal cell layer disruptions showed the following unique features: (1) significantly higher proliferation; (2) significantly higher expression of cell cycle control-, cell growth-, and stem cell-related genes; and (3) physical continuity with adjacent invasive lesions. Together, these findings suggest that focal basal cell layer disruptions could substantially impact the molecular profile and biological presentations of the overlying epithelial cells. Based on these and other findings, we have proposed that prostate tumor invasion is triggered by a localized degeneration of aged or injured basal cells and the resultant auto-immunoreactions. Our hypothesized steps for prostate tumor invasion include the following: (1) due to inherited or environmental factors, some patients contained cell cycle control- and renewal-related defects in the basal cell population that cause elevated basal cell degenerations; (2) the degradation products of degenerated basal cells or diffusible molecules of the overlying epithelial cells attract leukocyte infiltration; (3) leukocytes discharge their digestive enzymes upon the direct physical contact, resulting in a focal disruption in the basal cell layer, which leads to several focal alterations: (a) a focal loss of tumor suppressors and paracrine inhibitory function; (b) a focal increase of the permeability for growth-required nutrients and oxygen; (c) a focal increase of growth factors; (d) direct physical contact between epithelial and stromal cells; and (e) the exposure of the overlying epithelial cells directly to the stromal tissue fluid. These alterations individually or collectively stimulate or favor a clonal proliferation and stromal invasion of tumor progenitor or stem cells. Our hypothesis differs from the traditional theories in several aspects, including the triggering factor for the initiation of tumor invasion, the stage of tumor invasion, the cellular origin of invasive lesions, the significance of immunoreactive and stromal cells, and the potential approaches for early detection, treatment, and prevention of invasion. Our hypothesis represents a novel in vivo model as to the cellular mechanism leading to prostate tumor invasion. If confirmed, it could lead to a new direction to search for more effective approaches to combat prostate cancer. It could also have an immediate impact on patient care through improved pathologic evaluation of prostate tumor biopsies. More importantly, our hypothesis might be applicable, and significantly impact the detection, treatment, and prevention of other epithelium-derived tumors.
Collapse
|
19
|
Man YG. Focal degeneration of aged or injured myoepithelial cells and the resultant auto-immunoreactions are trigger factors for breast tumor invasion. Med Hypotheses 2007; 69:1340-57. [PMID: 17493765 DOI: 10.1016/j.mehy.2007.02.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 02/15/2007] [Indexed: 10/23/2022]
Abstract
The development of breast cancer is believed to be a multi-step process, sequentially progressing from normal to hyperplastic, to in situ, and to invasive stages. The progression from the in situ to invasive stage is believed to be triggered primarily, if not solely, by the overproduction of proteolytic enzymes by cancer cells, which cause degradation of the basement membrane. This theory is consistent with data derived from studies with cell cultures or animal models, while results from recent worldwide clinical trials with a variety of proteolytic enzyme inhibitors have been very disappointing, casting doubt on the validity of the enzyme theory. Based on our recent studies, we propose that breast tumor invasion is triggered by the following mechanisms and events: (1) the predisposition of genetic abnormalities in ME cell replenishment-related genes or other insults results in elevated focal degeneration of ME cells in some individuals; (2) the degradation products of ME cells or diffusible molecules of epithelial cells attract infiltration of immunoreactive cells (IRC) into the affected sites; (3) the direct physical contact between IRC and degenerated ME cells results in the discharge of digestive enzymes from IRC, causing focal disruptions in the ME cell layer; (4) focal disruptions in a given ME cell layer result in a localized loss of tumor suppressors and paracrine inhibitory function, a focal increase of permeability for oxygen, nutrients, and growth factors, and a localized increase of leukocyte infiltration, which facilitate the monoclonal proliferation of tumor progenitors, forming a biologically more aggressive cell cluster overlying the disrupted ME cell layer; (5) the direct physical contact between the newly formed cell cluster and stromal cells stimulates the production of tenascin and other invasion-associated molecules that facilitate tissue remodeling, angiogenesis, and epithelial-mesenchymal transition, providing a favorable micro-environment for proliferation and invasion. Our hypothesis differs from the enzyme theory in the stage of tumor invasion, the cellular origin of invasive lesions, the significance of IRC and stromal cells, and the potential approaches for treatment and prevention. If confirmed, our hypothesis could facilitate the early detection of specific individuals at increased risk to develop invasive breast cancer. More importantly, our hypothesis may facilitate development of novel approaches, including stimulating ME cell growth, neutralizing ME cell degradation products, manipulating the types and extent of IRC infiltration, and controlling the extent of stromal reactions, to combat tumor invasion.
Collapse
Affiliation(s)
- Yan-gao Man
- Gynecologic and Breast Research Laboratory, Department of Gynecologic and Breast Pathology, Armed Forces Institute of Pathology, Washington, DC 20306-6000, United States.
| |
Collapse
|
20
|
Planas-Silva MD, Waltz PK. Estrogen promotes reversible epithelial-to-mesenchymal-like transition and collective motility in MCF-7 breast cancer cells. J Steroid Biochem Mol Biol 2007; 104:11-21. [PMID: 17197171 DOI: 10.1016/j.jsbmb.2006.09.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 09/25/2006] [Indexed: 12/30/2022]
Abstract
The role of estrogen in the motility and invasion of breast cancer cells is controversial. Although estrogen receptor (ER)-positive breast tumors are considered less aggressive and more differentiated they still undergo metastasis. In many types of epithelial cancers, the ability to undergo metastasis has been associated with a loss of epithelial features and acquisition of mesenchymal properties leading to migration of individual cells, a process known as epithelial-to-mesenchymal transition (EMT). In this report, we show that a subset of ER-positive breast cancer cells can acquire mesenchymal-like features and motility in a reversible manner. In MCF-7 breast cancer cells estrogen-promoted acquisition of mesenchymal-like features while antiestrogens, such as tamoxifen, prevented this transition. Moreover, pharmacological inhibition of Src family kinases decreased the ability of estrogen to promote epithelial-to-mesenchymal-like transition. In addition to mesenchymal-like motility, a subset of estrogen-treated cells also moved as cell clusters (collective motility). While membrane localization of E-cadherin/beta-catenin was decreased in fibroblast-like cells, enhanced levels of E-cadherin/beta-catenin were detected in motile cell clusters. Thus, during tumor progression, estrogen may foster motility and invasion of ER-positive breast cancer by promoting simultaneously reversible EMT-like changes and collective motility. These studies suggest that antiestrogen therapy and Src family kinase inhibitors may decrease development of metastases in ER-positive breast cancer by blocking estrogen-dependent migration of human breast cancer cells.
Collapse
Affiliation(s)
- Maricarmen D Planas-Silva
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, MCH078, Hershey, PA 17033, USA.
| | | |
Collapse
|