1
|
Li A, Ruan M, Fei X, Xu H, Deng S, Bi R, Yang W, Dong L. Altered cytokeratin 5 expression in breast lobular myoepithelial cells. J Clin Pathol 2024; 77:536-543. [PMID: 37116947 DOI: 10.1136/jcp-2023-208835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/11/2023] [Indexed: 04/30/2023]
Abstract
AIMS Cytokeratin 5 (CK5) is a surrogate maker of progenitor cells and early glandular and myoepithelial cells (MECs) in the breast, and CK5 expression in breast MECs varies from ducts to lobules, and from normal to diseased tissue. However, the mechanisms underlying immunophenotypic alterations of CK5 expression in MECs remain unclear. METHODS CK5 expression in MECs of 20 normal breast samples, 58 ductal carcinoma in situ (DCIS; including 21 DCIS with extensive lobular involvement), 11 atypical ductal hyperplasia (ADH), 18 non-invasive lobular neoplasia consisting of 11 atypical lobular hyperplasia (ALH) and 7 lobular carcinoma in situ (LCIS), 20 cystic lobules and 10 usual ductal hyperplasia (UDH) involving lobules were observed to evaluate the effects of contact with benign hyperplastic or cancerous luminal cells and pressure of dilated glands on CK5 expression. RESULTS CK5 expression in normal ductal MECs was exclusively positive, whereas most normal lobular MECs were negative. In DCIS, cancerous ducts were primarily surrounded by CK5-positive MECs (91.0%), as were lobular acini involved by DCIS (89.2%), while the remaining normal acini maintained CK5-negative. CK5-positive MECs were found in 57.5% of acini in ALH and were more prevalent in LCIS (70.7%). CK5 expression was occasionally positive in both cystic lobules (16.7%) and lobules involved by UDH (14.3%), while an increase of CK5-positive MECs was found in ADH (38.2%). CONCLUSIONS These results suggest that CK5 expression in lobular MECs may be altered by contact with cancerous luminal cells rather than benign hyperplastic luminal cells or pressure from dilated glands.
Collapse
Affiliation(s)
- Anqi Li
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Miao Ruan
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Xiaochun Fei
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Shijie Deng
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Rui Bi
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei Dong
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| |
Collapse
|
2
|
Han X, Liu Y, Zhang S, Li L, Zheng L, Qiu L, Chen J, Zhan Z, Wang S, Ma J, Kang D, Chen J. Improving the diagnosis of ductal carcinoma in situ with microinvasion without immunohistochemistry: An innovative method with H&E-stained and multiphoton microscopy images. Int J Cancer 2024; 154:1802-1813. [PMID: 38268429 DOI: 10.1002/ijc.34855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/26/2024]
Abstract
Ductal carcinoma in situ with microinvasion (DCISM) is a challenging subtype of breast cancer with controversial invasiveness and prognosis. Accurate diagnosis of DCISM from ductal carcinoma in situ (DCIS) is crucial for optimal treatment and improved clinical outcomes. However, there are often some suspicious small cancer nests in DCIS, and it is difficult to diagnose the presence of intact myoepithelium by conventional hematoxylin and eosin (H&E) stained images. Although a variety of biomarkers are available for immunohistochemical (IHC) staining of myoepithelial cells, no single biomarker is consistently sensitive to all tumor lesions. Here, we introduced a new diagnostic method that provides rapid and accurate diagnosis of DCISM using multiphoton microscopy (MPM). Suspicious foci in H&E-stained images were labeled as regions of interest (ROIs), and the nuclei within these ROIs were segmented using a deep learning model. MPM was used to capture images of the ROIs in H&E-stained sections. The intensity of two-photon excitation fluorescence (TPEF) in the myoepithelium was significantly different from that in tumor parenchyma and tumor stroma. Through the use of MPM, the myoepithelium and basement membrane can be easily observed via TPEF and second-harmonic generation (SHG), respectively. By fusing the nuclei in H&E-stained images with MPM images, DCISM can be differentiated from suspicious small cancer clusters in DCIS. The proposed method demonstrated good consistency with the cytokeratin 5/6 (CK5/6) myoepithelial staining method (kappa coefficient = 0.818).
Collapse
Affiliation(s)
- Xiahui Han
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Yulan Liu
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Shichao Zhang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Liqin Zheng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Lida Qiu
- College of Physics and Electronic Information Engineering, Minjiang University, Fuzhou, China
| | - Jianhua Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
- College of Life Science, Fujian Normal University, Fuzhou, China
| | - Zhenlin Zhan
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| | - Shu Wang
- College of Mechanical Engineering and Automation, Fuzhou University, Fuzhou, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Deyong Kang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, China
| |
Collapse
|
3
|
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: 23] [Impact Index Per Article: 23.0] [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
|
4
|
Laokulrath N, Lim SK, Lim HY, Gudi M, Tan PH. Case report: Metastatic ovarian mucinous carcinoma to the breast: diagnostic challenges and pitfalls. Front Oncol 2024; 14:1364011. [PMID: 38562166 PMCID: PMC10982378 DOI: 10.3389/fonc.2024.1364011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Metastases to the breast from extramammary sources are extremely rare, with the ovary, primarily high-grade serous carcinoma, being the most common origin. We report a case of breast metastases from advanced stage ovarian mucinous carcinoma in a 48-year-old female- a case hitherto unreported in the literature. The case is noteworthy for its atypical presentation marked by an areolar rash, clinically suggestive of Paget disease of the nipple. This unique clinical scenario, coupled with histopathological examination revealing in-situ-like carcinoma component, posed a diagnostic challenge in discerning the tumour origin. We emphasize the need for heightened awareness among pathologists to avoid misdiagnosing metastatic carcinomas as primary breast tumours, a potential pitfall with significant clinical implications.
Collapse
Affiliation(s)
- Natthawadee Laokulrath
- Department of Pathology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Pathology and Laboratory Medicine, Kandang Kerbau (KK) Women’s and Children’s Hospital, Singapore, Singapore
| | - Siew Kuan Lim
- Solis Breast Care and Surgery Centre, Singapore, Singapore
| | | | - Mihir Gudi
- Department of Pathology and Laboratory Medicine, Kandang Kerbau (KK) Women’s and Children’s Hospital, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Pathology and Laboratory Medicine, Kandang Kerbau (KK) Women’s and Children’s Hospital, Singapore, Singapore
- Luma Medical Centre, Singapore, Singapore
- Parkway Laboratory Services Ltd, Singapore, Singapore
| |
Collapse
|
5
|
Jenkins TM, Mehr CR. Updates in the Use of Immunohistochemical Stains in Breast and Gynecologic Pathology. Arch Pathol Lab Med 2024; 148:33-47. [PMID: 37406290 DOI: 10.5858/arpa.2022-0467-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 07/07/2023]
Abstract
CONTEXT.— The use of immunohistochemical stains in breast and gynecologic pathology has become increasingly complex, with various diagnostic, prognostic, and predictive applications. OBJECTIVE.— To provide an update and review of immunohistochemical stains used in the practice of breast and gynecologic pathology. Established and new entities are reviewed, with descriptions of histomorphology and immunohistochemical staining patterns and discussion of interpretive pitfalls. DATA SOURCES.— Data were obtained from review of the English-language literature and firsthand experience of the authors in breast and gynecologic pathology. CONCLUSIONS.— Many entities in breast and gynecologic pathology benefit from evaluation with various immunohistochemical stains. These studies not only aid in the diagnosis and staging of tumors but also can provide prognostic and predictive information. Updated guidelines for recommended ancillary studies such as mismatch repair, p53, and human epidermal growth factor receptor 2 (HER2) studies in endometrium, as well as estrogen and progesterone receptors and HER2 in breast, are discussed. Finally, the use and interpretation of established and novel immunohistochemical stains are discussed in various breast and gynecologic malignancies.
Collapse
Affiliation(s)
- Taylor M Jenkins
- From the Department of Pathology, University of Virginia Health System, Charlottesville (Jenkins)
| | - Chelsea R Mehr
- Diagnostic Medicine Institute, Geisinger Health System, Geisinger Wyoming Valley Medical Center, Wilkes-Barre, Pennsylvania (Mehr)
| |
Collapse
|
6
|
Foschini MP, Nishimura R, Fabbri VP, Varga Z, Kaya H, Cserni G. Breast lesions with myoepithelial phenotype. Histopathology 2023; 82:53-69. [PMID: 36482278 DOI: 10.1111/his.14826] [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: 06/29/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 12/13/2022]
Abstract
Myoepithelial cells (MECs) constitute a continuous layer of cells surrounding the breast glands, localised between the epithelial cells (ECs) and the basal membrane. MECs play important roles in normal mammary gland as they produce basal membrane and stimulate secretion. During neoplastic transformation, MECs act as a barrier preventing stromal invasion. MECs themselves can undergo a great variety of changes, ranging from hyperplastic to metaplastic, to neoplastic, and giving rise to a wide spectrum of morphological pictures sometimes difficult to interpret on routine diagnoses. Several benign and malignant breast tumours can present features of MECs differentiation. As these latter tumours are quite infrequent, the purpose of the present study is to offer a review of the morphological spectrum of MECs lesions, with correlations to prognosis.
Collapse
Affiliation(s)
- Maria P Foschini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Section of Anatomic Pathology, Bellaria Hospital, Bologna, Italy
| | - Rieko Nishimura
- Department of Pathology, Nagoya Medical Center, Nagoya, Aichi, Japan
| | - Viscardo Paolo Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Department of Pathological Anatomy, Modena University Hospital, Modena, Italy
| | - Zsuzsanna Varga
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Handan Kaya
- Marmara University Pendik Training Hospital Department of Pathology, Marmara University School of Medicine, Istanbul, Turkey
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical Centre, University of Szeged, Szeged, Hungary.,Department of Pathology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| |
Collapse
|
7
|
Quinn C, Maguire A, Rakha E. Pitfalls in breast pathology. Histopathology 2023; 82:140-161. [PMID: 36482276 PMCID: PMC10107929 DOI: 10.1111/his.14799] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022]
Abstract
Accurate pathological diagnosis is the cornerstone of optimal clinical management for patients with breast disease. As non-operative diagnosis has now become the standard of care, histopathologists encounter the daily challenge of making definitive diagnoses on limited breast core needle biopsy (CNB) material. CNB samples are carefully evaluated using microscopic examination of haematoxylin and eosin (H&E)-stained slides and supportive immunohistochemistry (IHC), providing the necessary information to inform the next steps in the patient care pathway. Some entities may be difficult to distinguish on small tissue samples, and if there is uncertainty a diagnostic excision biopsy should be recommended. This review discusses (1) benign breast lesions that may mimic malignancy, (2) malignant conditions that may be misinterpreted as benign, (3) malignant conditions that may be incorrectly diagnosed as primary breast carcinoma, and (4) some IHC pitfalls. The aim of the review is to raise awareness of potential pitfalls in the interpretation of breast lesions that may lead to underdiagnosis, overdiagnosis, or incorrect classification of malignancy with potential adverse outcomes for individual patients.
Collapse
Affiliation(s)
- Cecily Quinn
- Irish National Breast Screening Programme and Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Aoife Maguire
- Irish National Breast Screening Programme and Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland
| | - Emad Rakha
- Department of Histopathology, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| |
Collapse
|
8
|
Tabár L, Dean PB, Lee Tucker F, Yen AMF, Chang RWJ, Hsu CY, Smith RA, Duffy SW, Chen THH. Breast cancers originating from the major lactiferous ducts and the process of neoductgenesis: Ductal Adenocarcinoma of the Breast, DAB. Eur J Radiol 2022; 153:110363. [DOI: 10.1016/j.ejrad.2022.110363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/30/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
|
9
|
Valdivia G, Alonso-Diez Á, Alonso-Miguel D, Suárez M, García P, Ortiz-Díez G, Pérez-Alenza MD, Peña L. Epitheliosis is a histopathological finding associated with malignancy and poor prognosis in dogs with mammary tumors. Vet Pathol 2022; 59:747-758. [PMID: 35451346 DOI: 10.1177/03009858221092013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Canine mammary epitheliosis (ME) is a poorly studied dysplasia that may have premalignant potential. In this study, the clinicopathological relevance of ME was prospectively studied in 90 female dogs with mammary tumors (MTs) that underwent radical mastectomy. ME distribution, extent, and coexistence with benign and malignant MTs were evaluated for each case (505 mammary glands). ME was macroscopically undetectable and was present in 47/90 (52%) cases, frequently bilateral. In dogs with malignant MTs and ME, diffuse ME throughout the mammary chain was present in 10/39 (26%) cases. A histological ME-carcinoma transition was evident in certain histotypes. By immunohistochemistry (AE1/AE3, cytokeratin 14 [CK-14], CK-8/18, vimentin, calponin, p63, Ki-67, estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2), ME was a slow-growing, triple-negative process with a strong predominance of basal-like nonmyoepithelial cells. ME was associated with older dogs (P = .016), malignant tumors (P = .044), worse clinical stages (P = .013), lymph node metastasis (LNM, P = .021), higher histological grade tumors (P = .035), and shorter overall survival (OS) in univariate analysis (P = .012). Interestingly, ME was distantly located to the malignant tumor in most cases (P = .007). In multivariate analyses, LNM (P = .005), histological grade (P = .006), and tumor size (P = .006) were independent predictors of OS. For the pathologist, the observation of ME should be clearly stated in the MT biopsy report to alert the surgeon/oncologist. Given the differences between canine ME and its human histopathological counterpart (atypical ductal hyperplasia), "epitheliosis" should remain the preferred term for the dog.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Laura Peña
- Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
10
|
Wilson GM, Dinh P, Pathmanathan N, Graham JD. Ductal Carcinoma in Situ: Molecular Changes Accompanying Disease Progression. J Mammary Gland Biol Neoplasia 2022; 27:101-131. [PMID: 35567670 PMCID: PMC9135892 DOI: 10.1007/s10911-022-09517-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/13/2022] [Indexed: 10/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive ductal carcinoma (IDC), whereby if left untreated, approximately 12% of patients develop invasive disease. The current standard of care is surgical removal of the lesion, to prevent potential progression, and radiotherapy to reduce risk of recurrence. There is substantial overtreatment of DCIS patients, considering not all DCIS lesions progress to invasive disease. Hence, there is a critical imperative to better predict which DCIS lesions are destined for poor outcome and which are not, allowing for tailored treatment. Active surveillance is currently being trialed as an alternative management practice, but this approach relies on accurately identifying cases that are at low risk of progression to invasive disease. Two DCIS-specific genomic profiling assays that attempt to distinguish low and high-risk patients have emerged, but imperfections in risk stratification coupled with a high price tag warrant the continued search for more robust and accessible prognostic biomarkers. This search has largely turned researchers toward the tumor microenvironment. Recent evidence suggests that a spectrum of cell types within the DCIS microenvironment are genetically and phenotypically altered compared to normal tissue and play critical roles in disease progression. Uncovering the molecular mechanisms contributing to DCIS progression has provided optimism for the search for well-validated prognostic biomarkers that can accurately predict the risk for a patient developing IDC. The discovery of such markers would modernize DCIS management and allow tailored treatment plans. This review will summarize the current literature regarding DCIS diagnosis, treatment, and pathology.
Collapse
Affiliation(s)
- Gemma M Wilson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Phuong Dinh
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Nirmala Pathmanathan
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - J Dinny Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
11
|
Deckwirth V, Rajakylä EK, Cattavarayane S, Acheva A, Schaible N, Krishnan R, Valle-Delgado JJ, Österberg M, Björkenheim P, Sukura A, Tojkander S. Cytokeratin 5 determines maturation of the mammary myoepithelium. iScience 2021; 24:102413. [PMID: 34007958 PMCID: PMC8111680 DOI: 10.1016/j.isci.2021.102413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 12/06/2020] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
At invasion, transformed mammary epithelial cells expand into the stroma through a disrupted myoepithelial (ME) cell layer and basement membrane (BM). The intact ME cell layer has thus been suggested to act as a barrier against invasion. Here, we investigate the mechanisms behind the disruption of ME cell layer. We show that the expression of basal/ME proteins CK5, CK14, and α-SMA altered along increasing grade of malignancy, and their loss affected the maintenance of organotypic 3D mammary architecture. Furthermore, our data suggests that loss of CK5 prior to invasive stage causes decreased levels of Zinc finger protein SNAI2 (SLUG), a key regulator of the mammary epithelial cell lineage determination. Consequently, a differentiation bias toward luminal epithelial cell type was detected with loss of mature, α-SMA-expressing ME cells and reduced deposition of basement membrane protein laminin-5. Therefore, our data discloses the central role of CK5 in mammary epithelial differentiation and maintenance of normal ME layer.
Collapse
Affiliation(s)
- Vivi Deckwirth
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Sandhanakrishnan Cattavarayane
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Niccole Schaible
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - Pia Björkenheim
- Veterinary Teaching Hospital, University of Helsinki, Helsinki 00014, Finland
| | - Antti Sukura
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöberginkatu 2, Helsinki 00014, Finland
| |
Collapse
|
12
|
Nakamura R, Oyama T, Inokuchi M, Ishikawa S, Hirata M, Kawashima H, Ikeda H, Dobashi Y, Ooi A. Neural EGFL like 2 expressed in myoepithelial cells and suppressed breast cancer cell migration. Pathol Int 2021; 71:326-336. [PMID: 33657249 DOI: 10.1111/pin.13087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/13/2021] [Indexed: 11/30/2022]
Abstract
Breast tissue has a branching structure that contains double-layered cells, consisting primarily of luminal epithelial cells inside and myoepithelial cells outside. Ductal carcinoma in situ (DCIS) still has myoepithelial cells surrounding the cancer cells. However, myoepithelial cells disappear in invasive ductal carcinoma. In this study, we detected expression of neural EGFL like (NELL) 2 and one of its receptors, roundabout guidance receptor (ROBO) 3, in myoepithelial and luminal epithelial cells (respectively) in normal breast tissue. NELL2 also was expressed in myoepithelial cells surrounding the non-cancerous intraductal proliferative lesions and DCIS. However, the expression level and proportion of NELL2-positive cells in DCIS were lower than those in normal and non-cancerous intraductal proliferative lesions. ROBO3 expression was decreased in invasive ductal carcinoma compared to that in normal and non-cancerous intraductal proliferative lesions. An evaluation of NELL2's function in breast cancer cell lines demonstrated that full-length NELL2 suppressed cell adhesion and migration in vitro. In contrast, the N-terminal domain of NELL2 increased cell adhesion in the early phase and migration in vitro in some breast cancer cells. These results suggested that full-length NELL2 protein, when expressed in myoepithelial cells, might serve as an inhibitor of breast cancer cell migration.
Collapse
Affiliation(s)
- Ritsuko Nakamura
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takeru Oyama
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Masafumi Inokuchi
- Department of Breast Surgery, Kanazawa University Hospital, Ishikawa, Japan.,Department of Breast and Endocrine Surgery, Kanazawa Medical University, Ishikawa, Japan
| | - Satoko Ishikawa
- Department of Breast Surgery, Kanazawa University Hospital, Ishikawa, Japan
| | - Miki Hirata
- Department of Breast Surgery, Kanazawa University Hospital, Ishikawa, Japan
| | - Hiroko Kawashima
- Radiology Division, Kanazawa University Hospital, Ishikawa, Japan
| | - Hiroko Ikeda
- Division of Diagnostic Pathology, Kanazawa University Hospital, Ishikawa, Japan
| | - Yoh Dobashi
- Department of Pathology, Saitama Medical Center, Jichi Medical University, Saitama, Japan.,Department of Pathology, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Akishi Ooi
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
13
|
Trinh A, Gil Del Alcazar CR, Shukla SA, Chin K, Chang YH, Thibault G, Eng J, Jovanović B, Aldaz CM, Park SY, Jeong J, Wu C, Gray J, Polyak K. Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System. Mol Cancer Res 2020; 19:623-635. [PMID: 33443130 DOI: 10.1158/1541-7786.mcr-20-0949] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 11/16/2022]
Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy-number profiles, mutational profiles, expression, neoantigen load, and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy-number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune "hot" and "cold" tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T cells and CD68 macrophages. Tumor-specific copy-number aberrations in MHC-I presentation machinery and losses in 3p, 4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER)-negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC. IMPLICATIONS: We demonstrate that the in situ to IDC evolutionary bottleneck is shaped by both tumor and immune cells.
Collapse
Affiliation(s)
- Anne Trinh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Carlos R Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Koei Chin
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Guillaume Thibault
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Jennifer Eng
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - C Marcelo Aldaz
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University Medical College, Seoul, Korea
| | - Catherine Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Joe Gray
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
14
|
Gil Del Alcazar CR, Alečković M, Polyak K. Immune Escape during Breast Tumor Progression. Cancer Immunol Res 2020; 8:422-427. [PMID: 32238387 DOI: 10.1158/2326-6066.cir-19-0786] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy using checkpoint inhibitors is one of the most promising current cancer treatment strategies. However, in breast cancer, its success has been limited to a subset of patients with triple-negative disease, whose durability of observed responses remain unclear. The lack of detailed understanding of breast tumor immune evasion mechanisms and the treatment of patients with highly heterogeneous metastatic disease contribute to these disappointing results. Here we discuss the current knowledge about immune-related changes during breast tumor progression, with special emphasis on the in situ-to-invasive breast carcinoma transition that may represent a key step of immunoediting in breast cancer. Comprehensive characterization of early-stage disease and better understanding of immunologic drivers of disease progression will likely expand the tools available for immunotherapy and improve patient stratification.
Collapse
Affiliation(s)
- Carlos R Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Shan NL, Minden A, Furmanski P, Bak MJ, Cai L, Wernyj R, Sargsyan D, Cheng D, Wu R, Kuo HCD, Li SN, Fang M, Maehr H, Kong AN, Suh N. Analysis of the Transcriptome: Regulation of Cancer Stemness in Breast Ductal Carcinoma In Situ by Vitamin D Compounds. Cancer Prev Res (Phila) 2020; 13:673-686. [PMID: 32467291 DOI: 10.1158/1940-6207.capr-19-0566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/01/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS), which accounts for one out of every five new breast cancer diagnoses, will progress to potentially lethal invasive ductal carcinoma (IDC) in about 50% of cases. Vitamin D compounds have been shown to inhibit progression to IDC in the MCF10DCIS model. This inhibition appears to involve a reduction in the cancer stem cell-like population in MCF10DCIS tumors. To identify genes that are involved in the vitamin D effects, a global transcriptomic analysis was undertaken of MCF10DCIS cells grown in mammosphere cultures, in which cancer stem-like cells grow preferentially and produce colonies by self-renewal and maturation, in the presence and absence of 1α25(OH)2D3 and a vitamin D analog, BXL0124. Using next-generation RNA-sequencing, we found that vitamin D compounds downregulated genes involved in maintenance of breast cancer stem-like cells (e.g., GDF15), epithelial-mesenchymal transition, invasion, and metastasis (e.g., LCN2 and S100A4), and chemoresistance (e.g., NGFR, PPP1R1B, and AGR2), while upregulating genes associated with a basal-like phenotype (e.g., KRT6A and KRT5) and negative regulators of breast tumorigenesis (e.g., EMP1). Gene methylation status was analyzed to determine whether the changes in expression induced by vitamin D compounds occurred via this mechanism. Ingenuity pathway analysis was performed to identify upstream regulators and downstream signaling pathway genes differentially regulated by vitamin D, including TP63 and vitamin D receptor -mediated canonical pathways in particular. This study provides a global profiling of changes in the gene signature of DCIS regulated by vitamin D compounds and possible targets for chemoprevention of DCIS progression to IDC in patients.
Collapse
Affiliation(s)
- Naing Lin Shan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Audrey Minden
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Philip Furmanski
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Min Ji Bak
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Li Cai
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Biomedical Engineering, School of Engineering, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Roman Wernyj
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - David Cheng
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Hsiao-Chen D Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Shanyi N Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Mingzhu Fang
- Environmental and Occupational Health Sciences Institute and School of Public Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Hubert Maehr
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Ah-Ng Kong
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, New Brunswick, New Jersey. .,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
16
|
Masood S. Is it ductal carcinoma in situ with microinvasion or "Ductogenesis"? The role of myoepithelial cell markers. Breast J 2020; 26:1138-1147. [PMID: 32447817 DOI: 10.1111/tbj.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/01/2020] [Indexed: 11/29/2022]
Abstract
Mammary myoepithelial cells have been under-recognized for many years since they were considered less important in breast cancer tumorigenesis compared to luminal epithelial cells. However, in recent years with advances in genomics, cell biology, and research in breast cancer microenvironment, more emphasis has been placed on better understanding of the role that myoepithelial cells play in breast cancer progression. As the result, it has been recognized that the presence or absence of myoepithelial cells play a critical role in the assessment of tumor invasion in diagnostic breast pathology. In addition, advances in screening mammography and breast imaging has resulted in increased detection of ductal carcinoma in situ and consequently more diagnosis of ductal carcinoma in situ with microinvasion. In the present review, we discuss the characteristics of myoepithelial cells, their genomic markers and their role in the accurate diagnosis of ductal carcinoma in situ with microinvasion. We also share our experience with reporting of various morphologic features of ductal carcinoma in situ that may mimic microinvasion and introduce the term of ductogenesis.
Collapse
Affiliation(s)
- Shahla Masood
- Department of Pathology, University of Florida College of Medicine - Jax, Jacksonville, FL, USA
| |
Collapse
|
17
|
Wang L, Xu C, Liu X, Yang Y, Cao L, Xiang G, Liu F, Wang S, Liu J, Meng Q, Jiao J, Niu Y. TGF-β1 stimulates epithelial-mesenchymal transition and cancer-associated myoepithelial cell during the progression from in situ to invasive breast cancer. Cancer Cell Int 2019; 19:343. [PMID: 31889895 PMCID: PMC6923856 DOI: 10.1186/s12935-019-1068-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 12/12/2019] [Indexed: 12/28/2022] Open
Abstract
Background The progression of ductal carcinoma in situ (DCIS) into invasive ductal carcinoma (IDC) is prevented by normal breast myoepithelial cells. Studies have suggested that EMT-associated genes were enriched in IDC in contrast to DCIS. This paper explored the relationship and potential mechanism between myoepithelial cells and EMT-associated genes in facilitating the transformation from DCIS to breast cancer. Methods EMT markers and myoepithelial phenotypic markers in IDC, DCIS, and healthy breast tissue were characterized using immunohistochemical assay. Both in vivo and in vitro models were created to mimic the various cell–cell interactions in the development of invasive breast cancer. Results We found that EMT markers were more abundant in invasive carcinomas than DCIS and adjacent normal breast tissue. Meanwhile, TGF-β1 regulated the morphology of MCF-7 (epithelial cells substitute) migration and EMT markers during the transformation from DCIS to invasive breast cancer. Additionally, TGF-β1 also regulated invasion, migration and cytokines secretion of MDA-MB-231 (myoepithelial cells substitute) and epithelial cells when co-cultured with MCF-7 both in vitro and in vivo. Conclusions In conclusion, these findings demonstrated that both EMT phenotypes and cancer-associated myoepithelial cells may have an impact on the development of invasive breast cancer.
Collapse
Affiliation(s)
- Li Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,2The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Cong Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Xia Liu
- 5Department of Oncology, General Hospital of Tianjin Medical University, 154 Anshan Road, Heping District, Tianjin, 300052 China
| | - Yang Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Lu Cao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Guomin Xiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Fang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Shuling Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,4Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Jing Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Qingxiang Meng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Jiao Jiao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| | - Yun Niu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060 China.,3Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060 China
| |
Collapse
|
18
|
Liu Y, Nekulova M, Nenutil R, Horakova I, Appleyard MV, Murray K, Holcakova J, Galoczova M, Quinlan P, Jordan LB, Purdie CA, Vojtesek B, Thompson AM, Coates PJ. ∆Np63/p40 correlates with the location and phenotype of basal/mesenchymal cancer stem-like cells in human ER + and HER2 + breast cancers. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2019; 6:83-93. [PMID: 31591823 PMCID: PMC6966710 DOI: 10.1002/cjp2.149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022]
Abstract
ΔNp63, also known as p40, regulates stemness of normal mammary gland epithelium and provides stem cell characteristics in basal and HER2‐driven murine breast cancer models. Whilst ΔNp63/p40 is a characteristic feature of normal basal cells and basal‐type triple‐negative breast cancer, some receptor‐positive breast cancers express ΔNp63/p40 and its overexpression imparts cancer stem cell‐like properties in ER+ cell lines. However, the incidence of ER+ and HER2+ tumours that express ΔNp63/p40 is unclear and the phenotype of ΔNp63/p40+ cells in these tumours remains uncertain. Using immunohistochemistry with p63 isoform‐specific antibodies, we identified a ΔNp63/p40+ tumour cell subpopulation in 100 of 173 (58%) non‐triple negative breast cancers and the presence of this population associated with improved survival in patients with ER−/HER2+ tumours (p = 0.006). Furthermore, 41% of ER+/PR+ and/or HER2+ locally metastatic breast cancers expressed ΔNp63/p40, and these cells commonly accounted for <1% of the metastatic tumour cell population that localised to the tumour/stroma interface, exhibited an undifferentiated phenotype and were CD44+/ALDH−. In vitro studies revealed that MCF7 and T47D (ER+) and BT‐474 (HER2+) breast cancer cell lines similarly contained a small subpopulation of ΔNp63/p40+ cells that increased in mammospheres. In vivo, MCF7 xenografts contained ΔNp63/p40+ cells with a similar phenotype to primary ER+ cancers. Consistent with tumour samples, these cells also showed a distinct location at the tumour/stroma interface, suggesting a role for paracrine factors in the induction or maintenance of ΔNp63/p40. Thus, ΔNp63/p40 is commonly present in a small population of tumour cells with a distinct phenotype and location in ER+ and/or HER2+ human breast cancers.
Collapse
Affiliation(s)
- Yajing Liu
- NCRC, University of Michigan, Ann Arbor, MI, USA
| | - Marta Nekulova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Rudolf Nenutil
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Iva Horakova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - M Virginia Appleyard
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Karen Murray
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Jitka Holcakova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Michaela Galoczova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Philip Quinlan
- Advanced Data Analysis Centre, University of Nottingham, Nottingham, UK
| | - Lee B Jordan
- Department of Pathology, Ninewells Hospital and Medical School, Dundee, UK
| | - Colin A Purdie
- Department of Pathology, Ninewells Hospital and Medical School, Dundee, UK
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Alastair M Thompson
- Division of Surgical Oncology, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Philip J Coates
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|
19
|
Ohe R, Aung NY, Tamura Y, Kabasawa T, Utsunomiya A, Tamazawa N, Kitaoka T, Meng HX, Shibata K, Yamakawa M. Diagnostic utility of CD205 in breast cancer: Simultaneous detection of myoepithelial cells and dendritic cells in breast tissue by CD205. Histol Histopathol 2019; 35:481-488. [PMID: 31524947 DOI: 10.14670/hh-18-164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND CD205 can be used to detect myoepithelial cells (MECs) and dendritic cells (DCs) in breast tissue. However, the usefulness of CD205 immunostaining in the pathological diagnosis of breast tumors is not fully understood. The objective of this study was to re-evaluate CD205 co-expression with other MEC markers, such as p63 and CD10, in nonneoplastic and neoplastic breast tissue and to evaluate its pathological diagnostic utility in these types of breast cancer. MATERIAL AND METHODS Nonneoplastic breast tissue samples with a terminal duct lobular unit and duct were obtained from fibroadenoma and mastopathy patients. Neoplastic breast tissue samples included ductal carcinoma in situ (DCIS) (n=43) and invasive ductal carcinoma (IDC) (n=60), including the tubule-forming type (n=20). These specimens were investigated by CD205, p63, and CD10 immunostaining. RESULTS In addition to p63 and CD10, CD205 was expressed on MECs in nonneoplastic breast and DCIS tissue samples; CD205 was simultaneously detected on DCs that had infiltrated DCIS and IDC tumor nests. CD205 was expressed on cancer cells themselves in only 7.3% of the breast cancer samples. The number of intratumoral CD205⁺ DCs in tubular IDC was significantly higher than that in DCIS (P<0.01). CONCLUSION Because CD205 was simultaneously detected on MECs and DCs in the same breast tissue sections, it may be useful for distinguishing tubular IDC from DCIS.
Collapse
Affiliation(s)
- Rintaro Ohe
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan.
| | - Naing Ye Aung
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuka Tamura
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Takanobu Kabasawa
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Aya Utsunomiya
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Nobuyuki Tamazawa
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Takumi Kitaoka
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hong-Xue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kenichi Shibata
- First Department of Surgery, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Mitsunori Yamakawa
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
20
|
Ding L, Su Y, Fassl A, Hinohara K, Qiu X, Harper NW, Huh SJ, Bloushtain-Qimron N, Jovanović B, Ekram M, Zi X, Hines WC, Alečković M, Gil Del Alcazar C, Caulfield RJ, Bonal DM, Nguyen QD, Merino VF, Choudhury S, Ethington G, Panos L, Grant M, Herlihy W, Au A, Rosson GD, Argani P, Richardson AL, Dillon D, Allred DC, Babski K, Kim EMH, McDonnell CH, Wagner J, Rowberry R, Bobolis K, Kleer CG, Hwang ES, Blum JL, Cristea S, Sicinski P, Fan R, Long HW, Sukumar S, Park SY, Garber JE, Bissell M, Yao J, Polyak K. Perturbed myoepithelial cell differentiation in BRCA mutation carriers and in ductal carcinoma in situ. Nat Commun 2019; 10:4182. [PMID: 31519911 PMCID: PMC6744561 DOI: 10.1038/s41467-019-12125-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Myoepithelial cells play key roles in normal mammary gland development and in limiting pre-invasive to invasive breast tumor progression, yet their differentiation and perturbation in ductal carcinoma in situ (DCIS) are poorly understood. Here, we investigated myoepithelial cells in normal breast tissues of BRCA1 and BRCA2 germline mutation carriers and in non-carrier controls, and in sporadic DCIS. We found that in the normal breast of non-carriers, myoepithelial cells frequently co-express the p63 and TCF7 transcription factors and that p63 and TCF7 show overlapping chromatin peaks associated with differentiated myoepithelium-specific genes. In contrast, in normal breast tissues of BRCA1 mutation carriers the frequency of p63+TCF7+ myoepithelial cells is significantly decreased and p63 and TCF7 chromatin peaks do not overlap. These myoepithelial perturbations in normal breast tissues of BRCA1 germline mutation carriers may play a role in their higher risk of breast cancer. The fraction of p63+TCF7+ myoepithelial cells is also significantly decreased in DCIS, which may be associated with invasive progression.
Collapse
Affiliation(s)
- Lina Ding
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Su
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Deciphera Pharmaceuticals, Waltham, MA, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Kunihiko Hinohara
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xintao Qiu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas W Harper
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Sung Jin Huh
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- ImmunoGen, Inc, Waltham, MA, USA
| | - Noga Bloushtain-Qimron
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- EMEA Site Intelligence and Activation, Tel Aviv, Israel
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Muhammad Ekram
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- WuXi NextCODE, Cambridge, MA, USA
| | - Xiaoyuan Zi
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Second Military Medical University, Shanghai, 200433, P.R. China
| | - William C Hines
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
- University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Maša Alečković
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Carlos Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ryan J Caulfield
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Dennis M Bonal
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
| | - Vanessa F Merino
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sibgat Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Metamark Genetics Inc, Worcester, MA, USA
| | | | - Laura Panos
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Michael Grant
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - William Herlihy
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Alfred Au
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94143, USA
| | - Gedge D Rosson
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Pedram Argani
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Andrea L Richardson
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
- Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - D Craig Allred
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kirsten Babski
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | - Elizabeth Min Hui Kim
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
- Cancer Treatment Centers of America, Atlanta, GA, USA
| | | | - Jon Wagner
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | - Ron Rowberry
- Sutter Roseville Medical Center, Roseville, CA, 95661, USA
| | | | - Celina G Kleer
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - E Shelley Hwang
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94143, USA
- Duke University, Durham, NC, USA
| | - Joanne L Blum
- Baylor-Charles A. Sammons Cancer Center, Dallas, TX, 75246, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health Boston, Boston, MA, 02215, USA
- Department of Stem Cell and Regenerative Biology, Harvard University Cambridge, Cambridge, MA, 02138, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Saraswati Sukumar
- Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - So Yeon Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Mina Bissell
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Jun Yao
- MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute Boston, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
21
|
Rosa M, Agosto-Arroyo E. Core needle biopsy of benign, borderline and in-situ problematic lesions of the breast: Diagnosis, differential diagnosis and immunohistochemistry. Ann Diagn Pathol 2019; 43:151407. [PMID: 31634810 DOI: 10.1016/j.anndiagpath.2019.151407] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/22/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022]
Abstract
Core needle biopsy (CNB) is the most common sampling technique for the histologic evaluation of breast abnormalities. Diagnosing benign proliferative, borderline and some in-situ lesions in CNB is challenging and subject to a significant degree of interobserver variability. In addition, due to the inherent limitations of CNB, "upgrading" to a more significant pathology at excision is an important consideration for some lesions. Pathologists carry a major responsibility in patient diagnosis, risk stratification and management. Familiarity with the histologic features and the clinical significance of these common and problematic lesions encountered in CNB is necessary for adequate treatment and patient follow-up. This review will focus on benign, atypical and in-situ epithelial proliferations, papillary lesions, radial sclerosing lesions, adenosis and cellular fibroepithelial lesions. Highlights of histologic features, useful strategies for accurate diagnosis, basic immunohistochemistry and management will be presented.
Collapse
Affiliation(s)
- Marilin Rosa
- Department of Anatomic Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America.
| | - Emmanuel Agosto-Arroyo
- Department of Anatomic Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America.
| |
Collapse
|
22
|
Dessources K, Sebastiao APM, Pareja F, Weigelt B, Reis-Filho JS. How Did We Get There? The Progression from Ductal Carcinoma In Situ to Invasive Ductal Carcinoma. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Sirka OK, Shamir ER, Ewald AJ. Myoepithelial cells are a dynamic barrier to epithelial dissemination. J Cell Biol 2018; 217:3368-3381. [PMID: 30061105 PMCID: PMC6168248 DOI: 10.1083/jcb.201802144] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/12/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023] Open
Abstract
Myoepithelial cells function collectively as a dynamic barrier to the invasion and dissemination of Twist1+ luminal epithelial cells and both luminal and basal phenotype breast cancer cells. Barrier function depends on myoepithelial abundance and both smooth muscle contractility and intercellular adhesion within the myoepithelium. The mammary epithelium is composed of an inner luminal and surrounding myoepithelial cell layer. The presence of cancer cells beyond the myoepithelium defines invasive breast cancer, yet the role of the myoepithelium during invasion remains unclear. We developed a 3D organotypic culture assay to model this process through lineage-specific expression of the prometastatic transcription factor Twist1. We sought to distinguish the functional role of the myoepithelium in regulating invasion and local dissemination. Myoepithelial-specific Twist1 expression induced cell-autonomous myoepithelial cell escape. Remarkably, luminal-specific Twist1 expression was rarely sufficient for escape. Time-lapse microscopy revealed that myoepithelial cells collectively restrain and reinternalize invading Twist1+ luminal cells. Barrier function correlated with myoepithelial abundance and required the expression of α-smooth muscle actin and P-cadherin. We next demonstrated that myoepithelial cells can restrain and recapture invasive cancer cells. Our data establish the concept of the myoepithelium as a dynamic barrier to luminal dissemination and implicate both smooth muscle contractility and intercellular adhesion in barrier function.
Collapse
Affiliation(s)
- Orit Katarina Sirka
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eliah R Shamir
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew J Ewald
- Departments of Cell Biology, Oncology, and Biomedical Engineering, Center for Cell Dynamics, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
24
|
Duivenvoorden HM, Spurling A, O’Toole SA, Parker BS. Discriminating the earliest stages of mammary carcinoma using myoepithelial and proliferative markers. PLoS One 2018; 13:e0201370. [PMID: 30044853 PMCID: PMC6059463 DOI: 10.1371/journal.pone.0201370] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/14/2018] [Indexed: 12/17/2022] Open
Abstract
Mammographic screening has led to increased detection of breast cancer at a pre-invasive state, hence modelling the earliest stages of breast cancer invasion is important in defining candidate biomarkers to predict risk of relapse. Discrimination of pre-invasive from invasive lesions is critically important for such studies. Myoepithelial cells are the barrier between epithelial cells and the surrounding stroma in the breast ductal system. A number of myoepithelial immunohistochemistry markers have been identified and validated in human tissue for use by pathologists as diagnostic tools to distinguish in situ carcinoma from invasive breast cancer. However, robust myoepithelial markers for mouse mammary tissue have been largely under-utilised. Here, we investigated the utility of the myoepithelial markers smooth muscle actin (SMA), smooth muscle myosin heavy chain (SMMHC), cytokeratin-14 (CK14) and p63 to discriminate mammary intraepithelial neoplasia (MIN) from invasive disease in the C57BL/6J MMTV-PyMT transgenic model of mammary carcinoma. We identified that SMMHC and CK14 are retained in early in situ neoplasia and are appropriate markers for distinguishing MIN from invasive disease in this model. Additionally, the proliferation marker Ki67 is a superior marker for differentiating between normal and hyperplastic ducts, prior to the development of MIN. Based on this, we developed a scoring matrix for discriminating normal, hyperplasia, MIN and invasive lesions in this spontaneous mammary tumorigenesis model. This study demonstrates heterogeneous expression of myoepithelial proteins throughout tumour development, and highlights the need to characterise the most appropriate markers in other models of early breast cancer to allow accurate classification of disease state.
Collapse
Affiliation(s)
- Hendrika M. Duivenvoorden
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, Melbourne, VIC, Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, Melbourne, VIC, Australia
| | - Sandra A. O’Toole
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Australian Clinical Labs, Bella Vista, NSW, Australia
- * E-mail: (BSP); (SAOT)
| | - Belinda S. Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, Melbourne, VIC, Australia
- * E-mail: (BSP); (SAOT)
| |
Collapse
|
25
|
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
|
26
|
Elastic stains in the evaluation of DCIS with comedo necrosis in breast cancers. Virchows Arch 2017; 472:1007-1014. [PMID: 29101458 DOI: 10.1007/s00428-017-2259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
Abstract
As concerns the microscopic morphology of ductal carcinoma in situ (DCIS), neoplastic cells are surrounded by both a myoepithelial cell layer and a basement membrane as expected from the outer structure of ducts and lobules. However, in some cases, it is impossible to state whether the structures involved by the disease are ducts or lobules. Altogether 1220 anatomic structures involved by DCIS displaying comedo necrosis from 27 slides of 21 patients (seen on both haematoxylin and eosin-stained and orcein-stained slides) were identified as representing ducts, likely ducts, unclassifiable structures, likely acini or acini on the basis of their distribution and resemblance to normal anatomic structures. All structures were then rated as having a circumferential elastic layer (as normal ducts), a partial elastic layer around more or less than half of the periphery or having no peripheral elastic layer at all (as normal acini). Structures classified as ducts or likely ducts were likely to have an elastic coating, whereas acini and likely acini had no such coating. Unclassifiable structures were generally devoid of an elastic layer. Structures (and cases) that were likely to represent neoductgenesis as proposed by Zhou et al. (Int J Breast Cancer 2014;2014:581706) were generally unclassifiable and devoid of outer elastic layer. Many duct-like structures in DCIS with comedo necrosis are devoid of elastic layer typical of normal ducts, suggesting that these structures are abnormal despite conservation of the myoepithelium and the basement membrane.
Collapse
|
27
|
P40 Immunostain Does Not Outperform p63 as a Myoepithelial Cell Marker in the Daily Practice of Breast Pathology. Appl Immunohistochem Mol Morphol 2017; 26:599-604. [PMID: 28549030 DOI: 10.1097/pai.0000000000000507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
P40 antibody has been shown to be a more specific squamous and basal cell marker compared with p63. As detection of myoepithelial cells (MECs) plays a critical role in breast pathology, and the fact that p40 targets an isoform of p63, this study was designed to compare these antibodies in a variety of lesions, especially those with an sclerotic stroma and carcinoma in situ. All studied lesions were selected from the daily cases of the 3 authors and stained with p63, p40, and calponin immunohistochemical stains. Thirty-four cases (and 19 internal controls) were included. Seventy percent constituted sclerotic lesions (12 cases) and ductal carcinoma in situ (12 cases). P40 and p63 stained all lesions and showed a similar patchy staining pattern in 50% of ductal carcinoma in situ and sclerotic lesions. Compared with internal controls, p40 and p63 demonstrated decreased staining intensity in up to 70% and 8% of all cases, respectively, with no cross-reactivity with mesenchymal cells and minor cross-reactivity with epithelial cells. In our study, p40 did not outperform p63 as a MEC marker. p40 showed a decreased intensity in a higher number of cases (P<0.0001). In our opinion, p63 continues to be the best nuclear marker for the detection of MECs in the daily practice of breast pathology.
Collapse
|
28
|
Wang Y, Jindal S, Martel M, Wu Y, Schedin P, Troxell M. Myoepithelial cells in lobular carcinoma in situ: distribution and immunophenotype. Hum Pathol 2016; 55:126-34. [PMID: 27195907 DOI: 10.1016/j.humpath.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/15/2016] [Accepted: 05/05/2016] [Indexed: 12/18/2022]
Abstract
Myoepithelial cells have important physical and paracrine roles in breast tissue development, maintenance, and tumor suppression. Recent molecular and immunohistochemical studies have demonstrated phenotypic alterations in ductal carcinoma in situ-associated myoepithelial cells. Although the relationship of lobular carcinoma in situ (LCIS) and myoepithelial cells was described in 1980, further characterization of LCIS-associated myoepithelial cells is lacking. We stained 27 breast specimens harboring abundant LCIS with antibodies to smooth muscle myosin heavy chain, smooth muscle actin, and calponin. Dual stains for E-cadherin/smooth muscle myosin heavy chain and CK7/p63 were also performed. In each case, the intensity and distribution of staining in LCIS-associated myoepithelial cells were compared with normal breast tissue on the same slide. In 78% of the cases, LCIS-associated myoepithelial cells demonstrated decreased staining intensity for one or more myoepithelial markers. The normal localization of myoepithelial cells (flat against the basement membrane, pattern N) was seen in 96% of LCIS, yet 85% of cases had areas with myoepithelial cell cytoplasm oriented perpendicular to the basement membrane (pattern P), and in 30% of cases, myoepithelial cells appeared focally admixed with LCIS cells (pattern C). This study characterizes detailed architectural and immunophenotypic alterations of LCIS-associated myoepithelial cells. The finding of variably diminished staining favors application of several myoepithelial immunostains in clinical practice. The interaction of LCIS with myoepithelial cells, especially in light of the perpendicular and central architectural arrangements, deserves further mechanistic investigation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239
| | - Maritza Martel
- Department of Pathology, Providence Health and Services, Portland, OR 97213
| | - Yaping Wu
- Department of Pathology, Providence Health and Services, Portland, OR 97213
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239; Oregon Health & Science University, Knight Cancer Institute, Portland, OR 97239
| | - Megan Troxell
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239; Oregon Health & Science University, Knight Cancer Institute, Portland, OR 97239.
| |
Collapse
|
29
|
Wood A, Houghton SL, Biswas A. A Comparative Study of Immunohistochemical Myoepithelial Cell Markers in Cutaneous Benign Cystic Apocrine Lesions. Am J Dermatopathol 2016; 38:475-83. [PMID: 26630681 DOI: 10.1097/dad.0000000000000431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The use of immunohistochemical markers for myoepithelial cells (MEC) is a useful tool in the distinction of benign from malignant epithelial neoplasms. Although their use in breast tumors is well recognized, little is known concerning its application in comparable cutaneous lesions. Using benign cutaneous cystic apocrine lesions as a study model, the aim of this study was to compare 5 immunohistochemical markers [calponin, p63, smooth muscle actin (SMA), cytokeratin 14, and CD10] in their effectiveness to highlight MEC. Cases of apocrine hidrocystoma and cystadenoma (n = 44) were reviewed with a particular emphasis on proliferative features and apocrine change. The MEC staining pattern and the intensity and distribution scores in proliferative (n = 29) and nonproliferative (n = 15) lesions were assessed, and the differences between the 2 groups were statistically analyzed using Fisher exact test. Calponin and SMA stained MEC in the most consistent manner. Being a nuclear stain, p63 was easy to interpret but typically showed discontinuous staining. Cytokeratin 14 not only effectively highlighted MEC but also stained some luminal epithelial cells in an unpredictable manner. Because of prominent background dermal fibroblast staining, CD10 was often difficult to interpret. Only SMA and p63 showed a statistically significant difference in MEC staining intensity scores between the proliferative and nonproliferative groups. Our results show that immunohistological staining for MEC in benign cystic apocrine lesions of the skin is variable. The authors recommend that a panel of markers that includes calponin and p63 be used and highlight the need for awareness of specific caveats associated with individual markers.
Collapse
Affiliation(s)
- Andrew Wood
- *Resident, Pathology, Western General Hospital, Edinburgh, United Kingdom; †Biomedical Scientist, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom; and ‡Consultant Dermatopathologist, Western General Hospital, Edinburgh, United Kingdom
| | | | | |
Collapse
|
30
|
Khazai L, Rosa M. Use of Immunohistochemical Stains in Epithelial Lesions of the Breast. Cancer Control 2016; 22:220-5. [PMID: 26068768 DOI: 10.1177/107327481502200214] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND During the last few decades, immunohistochemistry (IHC) has become an integral part of pathology. Although hematoxylin and eosin (H & E) stain remains the fundamental basis for diagnostic pathology of the breast, IHC stains provide useful and sometimes vital information. Moreover, considering the role of hormonal therapy in hormone receptor-positive breast tumors, as well as the availability of targeted chemotherapeutic agents for HER2-positive cases, IHC studies represent a major part of workups. METHODS A literature search was performed to explore the uses of IHC stains related to the diagnoses of breast lesions and prognostic/predictive information. RESULTS Selective use of IHC stains in conjunction with H & E examination helps resolve most diagnostic issues encountered by surgical pathologists during their day-to-day practice. Pathologists should be familiar with the use of each immunostain and its limitations to avoid interpretative errors. CONCLUSIONS IHC stains help guide the differential diagnosis of challenging epithelial lesions of the breast. They should be selectively and judiciously used and their findings must be interpreted with the differential diagnoses in mind and with an understanding of possible pitfalls.
Collapse
Affiliation(s)
- Laila Khazai
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | |
Collapse
|
31
|
Cserni G, Wells CA, Kaya H, Regitnig P, Sapino A, Floris G, Decker T, Foschini MP, van Diest PJ, Grabau D, Reiner A, DeGaetano J, Chmielik E, Cordoba A, Andreu X, Zolota V, Charafe-Jauffret E, Ryska A, Varga Z, Weingertner N, Bellocq JP, Liepniece-Karele I, Callagy G, Kulka J, Bürger H, Figueiredo P, Wesseling J, Amendoeira I, Faverly D, Quinn CM, Bianchi S. Consistency in recognizing microinvasion in breast carcinomas is improved by immunohistochemistry for myoepithelial markers. Virchows Arch 2016; 468:473-81. [DOI: 10.1007/s00428-016-1909-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/24/2015] [Accepted: 01/14/2016] [Indexed: 11/29/2022]
|
32
|
Mardekian SK, Bombonati A, Palazzo JP. Ductal carcinoma in situ of the breast: the importance of morphologic and molecular interactions. Hum Pathol 2015; 49:114-23. [PMID: 26826418 DOI: 10.1016/j.humpath.2015.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/02/2015] [Accepted: 11/05/2015] [Indexed: 12/21/2022]
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a lesion characterized by significant heterogeneity, in terms of morphology, immunohistochemical staining, molecular signatures, and clinical expression. For some patients, surgical excision provides adequate treatment, but a subset of patients will experience recurrence of DCIS or progression to invasive ductal carcinoma (IDC). Recent years have seen extensive research aimed at identifying the molecular events that characterize the transition from normal epithelium to DCIS and IDC. Tumor epithelial cells, myoepithelial cells, and stromal cells undergo alterations in gene expression, which are most important in the early stages of breast carcinogenesis. Epigenetic modifications, such as DNA methylation, together with microRNA alterations, play a major role in these genetic events. In addition, tumor proliferation and invasion is facilitated by the lesional microenvironment, which includes stromal fibroblasts and macrophages that secrete growth factors and angiogenesis-promoting substances. Characterization of DCIS on a molecular level may better account for the heterogeneity of these lesions and how this manifests as differences in patient outcome and response to therapy. Molecular assays originally developed for assessing likelihood of recurrence in IDC are recently being applied to DCIS, with promising results. In the future, the classification of DCIS will likely incorporate molecular findings along with histologic and immunohistochemical features, allowing for personalized prognostic information and therapeutic options for patients with DCIS. This review summarizes current data regarding the molecular characterization of DCIS and discusses the potential clinical relevance.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biopsy
- Breast Neoplasms/chemistry
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma/chemistry
- Carcinoma/genetics
- Carcinoma/pathology
- Carcinoma, Intraductal, Noninfiltrating/chemistry
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Disease Progression
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Predisposition to Disease
- Humans
- Immunohistochemistry
- Mastectomy
- Molecular Diagnostic Techniques
- Neoplasm Recurrence, Local
- Phenotype
- Predictive Value of Tests
- Reproducibility of Results
- Treatment Outcome
Collapse
Affiliation(s)
- Stacey K Mardekian
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107.
| | - Alessandro Bombonati
- Department of Pathology, Albert Einstein Medical Center, Philadelphia, PA 19141.
| | - Juan P Palazzo
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107.
| |
Collapse
|
33
|
Russell TD, Jindal S, Agunbiade S, Gao D, Troxell M, Borges VF, Schedin P. Myoepithelial cell differentiation markers in ductal carcinoma in situ progression. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3076-89. [PMID: 26343330 PMCID: PMC4630168 DOI: 10.1016/j.ajpath.2015.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 07/14/2015] [Accepted: 07/21/2015] [Indexed: 11/20/2022]
Abstract
We describe a preclinical model that investigates progression of early-stage ductal carcinoma in situ (DCIS) and report that compromised myoepithelial cell differentiation occurs before transition to invasive disease. Human breast cancer MCF10DCIS.com cells were delivered into the mouse mammary teat by intraductal injection in the absence of surgical manipulations and accompanying wound-healing confounders. DCIS-like lesions developed throughout the mammary ducts with full representation of human DCIS histologic patterns. Tumor cells were incorporated into the normal mammary epithelium, developed ductal intraepithelial neoplasia and DCIS, and progressed to invasive carcinoma, suggesting the model provides a rigorous approach to study early stages of breast cancer progression. Mammary glands were evaluated for myoepithelium integrity with immunohistochemical assays. Progressive loss of the myoepithelial cell differentiation markers p63, calponin, and α-smooth muscle actin was observed in the mouse myoepithelium surrounding DCIS-involved ducts. p63 loss was an early indicator, calponin loss intermediate, and α-smooth muscle actin a later indicator of compromised myoepithelium. Loss of myoepithelial calponin was specifically associated with gain of the basal marker p63 in adjacent tumor cells. In single time point biopsies obtained from 16 women diagnosed with pure DCIS, a similar loss in myoepithelial cell markers was observed. These results suggest that further research is warranted into the role of myoepithelial cell p63 and calponin expression on DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Tanya D Russell
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Samiat Agunbiade
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dexiang Gao
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Megan Troxell
- Department of Pathology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Virginia F Borges
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; University of Colorado Cancer Center, Aurora, Colorado
| | - Pepper Schedin
- Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
34
|
Kővári B, Báthori Á, Cserni G. CD10 Immunohistochemical Expression in Apocrine Lesions of the Breast. Pathobiology 2015; 82:259-63. [PMID: 26562027 DOI: 10.1159/000440664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/24/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In the breast, CD10 is expressed by myoepithelial cells (MECs), and apocrine metaplasia has also been mentioned as being positive with this marker. Apocrine lesions have been explored for the expression of CD10. METHODS The apocrine lesions studied included 11 cysts, 6 cases of apocrine adenosis, 2 of apocrine metaplasia or hyperplasia in papilloma, 13 ductal carcinomas in situ (DCIS) and invasive carcinomas (14 ductal and 4 lobular). RESULTS Benign apocrine lesions showed complete or partial luminal CD10 staining, although most cases included parts without staining, and 2 lesions were completely negative. The MECs were often but not always positive. Nine of the 13 cases of apocrine DCIS displayed no luminal staining, but 4 demonstrated very focal luminal positivity. The MECs around the DCIS showed a spectrum of staining from nil to strong and complete. Only 4 invasive carcinomas demonstrated luminal/membranous staining. Cytoplasmic CD10 positivity was seen focally in 4 invasive cancers and in 3 DCIS. CONCLUSION CD10 positivity is luminal/membranous in most benign apocrine lesions, the staining being nonuniversal and sometimes focal. Analogous staining in apocrine malignancies seems rarer in DCIS and even rarer in invasive apocrine carcinomas, but atypical cytoplasmic positivity may also occur. CD10 is not an ideal myoepithelial marker in apocrine lesions.
Collapse
Affiliation(s)
- Bence Kővári
- Department of Pathology, University of Szeged, Szeged, Hungary
| | | | | |
Collapse
|
35
|
Chen L, Yin X, Lu S, Chen G, Dong L. Basal cytokeratin phenotypes of myoepithelial cells indicates the origin of ductal carcinomas in situ of the breast. Appl Immunohistochem Mol Morphol 2015; 23:558-64. [PMID: 26336082 DOI: 10.1097/pai.0000000000000123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Terminal duct lobular unit (TDLU) is widely accepted as the origin of ductal carcinoma in situ of breast. The differentiation states of myoepithelial cells of breast ductal system hint the development of breast hyperplastic lesions. Basal cytokeratin (CK) phenotypes indicate the differentiation of myoepithelial cells. Using antibodies of CK5/6, CK14, and CK17, this study reports the basal CK phenotypes of myoepithelial cells in 20 foci of normal breast, 20 usual ductal hyperplasias, 36 ductal carcinomas in situ (DCIS), and 28 sclerosing adenosis (SA). The results showed that the positive staining of basal CKs of myoepithelial cells in normal ducts were significantly higher than those in normal lobules. The basal CK expression of myoepithelial cells of DCIS and usual ductal hyperplasia was similar to that of normal duct, whereas that of SA was similar to that of normal lobule. We propose a modified model of TDLU origin of intraductal carcinoma that most of DCIS originate from terminal ducts of TDLU, whereas most SA originate from lobules.
Collapse
Affiliation(s)
- Ling Chen
- *Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu †Department of Pathology, Maternity and Child Health Care Hospital, Jiaxing ‡Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou §Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
36
|
Kov�ri B, Sz�sz AM, Kulka J, Maru�ic Z, �arcevic B, Tiszlavicz L, Cserni G. Evaluation of p40 as a Myoepithelial Marker in Different Breast Lesions. Pathobiology 2015; 82:166-71. [DOI: 10.1159/000375127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
37
|
Phenotypic and Functional Characterization of Ductal Carcinoma In Situ-Associated Myoepithelial Cells. Clin Breast Cancer 2015; 15:335-42. [PMID: 25700939 DOI: 10.1016/j.clbc.2015.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/14/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is contained by myoepithelial cells that are morphologically similar to normal breast tissue myoepithelial cells. However, phenotypic and functional characteristics of DCIS-associated myoepithelial cells are not known. In this study, we aimed to assess the characteristics of DCIS-associated myoepithelial cells. MATERIALS AND METHODS Immunophenotypic and functional characteristics of myoepithelial cells of pure DCIS, the DCIS component of infiltrating duct carcinoma (IDC), and the adjacent normal breast tissue of both groups (30 cases in each group) was assessed using phenotypic (CK5/6, CK14, p63, and calponin) and functional markers (maspin and CXCL14). RESULTS There was a decrease in expression of CK14, p63, and calponin in pure DCIS-associated myoepithelial cells compared with normal breast tissue myoepithelial cells (43.3% vs. 80.3%, 3.3% vs. 70%, 46.6 vs. 93.3%, respectively) and in the DCIS component of IDC compared with normal breast tissue myoepithelial cells (56.6% vs. 100%, 3.3% vs. 73.3%, 56.6% vs. 96.6%, respectively). CK5/6 expression was low to absent in myoepithelial cells of pure DCIS and the DCIS component of IDC as well as normal breast tissue myoepithelial cells. Maspin was expressed in all samples of normal breast tissue; however, 20% of pure DCIS and 26.6% of the DCIS component of IDC showed decreased expression. CXCL14 expression was greater in pure DCIS compared with adjacent normal breast tissue and the DCIS component of IDC. CONCLUSION Decreased expression of myoepithelial cell markers in DCIS suggests that DCIS-associated myoepithelial cells are phenotypically different from their normal counterparts. Two or more markers, preferably p63 and calponin, should be used to distinguish in situ from invasive breast carcinomas.
Collapse
|
38
|
Moritani S, Ichihara S, Yatabe Y, Hasegawa M, Iwakoshi A, Hosoda W, Narita M, Nagai Y, Asai M, Ujihira N, Yuba Y, Jijiwa M. Immunohistochemical expression of myoepithelial markers in adenomyoepithelioma of the breast: a unique paradoxical staining pattern of high-molecular weight cytokeratins. Virchows Arch 2014; 466:191-8. [PMID: 25479938 DOI: 10.1007/s00428-014-1687-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 09/10/2014] [Accepted: 11/11/2014] [Indexed: 12/16/2022]
Abstract
To determine which immunohistochemical markers are useful for the identification of neoplastic myoepithelial cells in adenomyoepithelioma of the breast, the expression of seven myoepithelial markers (α-smooth muscle actin (α-SMA), calponin, p63, CD10, cytokeratin 5/6, cytokeratin 14, and S-100) was examined in 19 lesions from 16 patients. The lesion consisted of seven spindle and 12 clear cell lesions. For normal myoepithelial cells, α-SMA, calponin, and p63 were significantly more sensitive than cytokeratin 5/6, cytokeratin 14, and S-100. There was no significant difference in the expression of α-SMA, calponin, p63, and CD10 in neoplastic myoepithelial cells of adenomyoepithelioma regardless of spindle or clear cell types. In spindle cell lesions, high-molecular weight cytokeratins (HMWCK; cytokeratin 5/6 and cytokeratin 14) tended to show higher staining scores and S-100 showed lower staining scores than other markers. In clear cell lesions, HMWCK showed significantly lower staining scores than the other five markers. There was no significant difference in staining scores among the other five markers. HMWCK showed a unique paradoxical staining pattern in clear cell lesions, with diffusely positive inner epithelial cells and completely negative outer myoepithelial cells. Although the sensitivity of HMWCK in clear cell lesions is low, with this unique paradoxical staining pattern and relatively high sensitivity in spindle cell lesions, HMWCK could be useful in diagnosing adenomyoepithelioma. In choosing immunohistochemical markers, any of the seven markers are useful, but combining HMWCK and any one of α-SMA, calponin, and p63 would be a good panel for the diagnosis of adenomyoepithelioma.
Collapse
Affiliation(s)
- Suzuko Moritani
- Department of Advanced Diagnosis, Division of Pathology, Nagoya Medical Center, 4-1-1, Sannomaru, Naka-ku, Nagoya, Aichi, 460-0001, Japan,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Utility of immunohistochemical markers in irradiated breast tissue: an analysis of the role of myoepithelial markers, p53, and Ki-67. Am J Surg Pathol 2014; 38:1128-37. [PMID: 25029119 DOI: 10.1097/pas.0000000000000243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Radiation therapy is an important adjunct to breast-conserving surgery, but the diagnosis of recurrent/de novo carcinoma in a background of radiation atypia can be difficult, especially on small biopsies. Immunostaining for myoepithelial cell proteins is often used to assess invasion in nonirradiated breast tissue, yet these stains have not been investigated specifically in irradiated breast. We studied 29 irradiated breast resection specimens, some with carcinoma in situ (CIS, n=13) and/or invasive carcinoma (n=13). Representative blocks were stained for the myoepithelial proteins p63, smooth muscle myosin heavy chain (SMM), calponin, CK5/6, the proliferative marker Ki-67, and the tumor-suppressor p53. Nonirradiated control tissue was also stained with Ki-67 and p53 (CIS, normal, contralateral). Areas of radiation atypia/atrophy and nearly all CIS in irradiated breast tissue had abundant myoepithelial cells as evidenced by SMM, calponin, and p63 stains, with focal staining attenuation or gaps with SMM and calponin and frequently absent CK5/6 staining. As predicted, myoepithelial cell staining was absent in invasive carcinoma. p63 staining revealed postradiation myoepithelial nuclear morphologic changes. p53 staining was increased, although weak, in irradiated non-neoplastic breast (12% irradiated; 4% nonirradiated); however, irradiated CIS had less p53 staining when compared with control CIS (3% irradiated; 38% nonirradiated). As expected, Ki-67 was increased in carcinoma as compared with non-neoplastic irradiated tissue. Thus, myoepithelial immunostaining is a useful diagnostic adjunct in irradiated breast, with caveats similar to nonirradiated breast. Ki-67 may be helpful in some postradiation specimens; however, p53 staining is not reliable in this setting.
Collapse
|
40
|
Abe E, Suzuki K, Hayashi N, Yang Y, Li CP, Uno M, Akiyama F, Yamauchi H, Nakamura S, Tsugawa K, Tsunoda H, Ohde S, Sasano H. Clinicopathological significance of 'atypical ductal proliferation' in core needle biopsy of the breast. Pathol Int 2014; 64:58-66. [PMID: 24629173 DOI: 10.1111/pin.12135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/13/2014] [Indexed: 11/30/2022]
Abstract
Atypical ductal proliferation or ADP has been used in histopathological diagnosis of core needle biopsy (CNB) but its details have not been well studied. Therefore, we examined the clinicopathological characteristics of the initial CNB cases diagnosed as 'ADP ' who subsequently turned out to be malignant, and compared the findings to those that did not. Among 101 cases initially diagnosed as ADP in CNB, the second biopsy revealed no carcinoma (38), ductal carcinoma in situ (DCIS) (45) and invasive carcinoma (18). Significant differences were detected between those which turned out to be carcinoma and those that did not, in the status of myoepithelial cells identified by p63 immunohistochemistry (P = 0.026) and ultrasound (US) categories (P < 0.001). We further compared the histopathological characteristics of those initially diagnosed as ADP and subsequently as DCIS or invasive ductal carcinoma (IDC) with those initially diagnosed as such. DCIS or IDC cases initially diagnosed as ADP had significantly lower Ki67 labeling index (P < 0.01, P < 0.01) and histological grade using Van nuys prognostic index (P < 0.01) or Nottingham histological grades (P < 0.01) respectively than those initially as DCIS or IDC. An assessment of myoepithelial components with US findings might contribute to determine the subsequent clinical algorithm of the patients diagnosed as ADP at initial CNB.
Collapse
Affiliation(s)
- Eriko Abe
- Department of Pathology, St. Luke's International Hospital, Tokyo; Department of Pathology, Tohoku University Hospital, Sendai
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sclerosing adenosis and risk of breast cancer. Breast Cancer Res Treat 2014; 144:205-12. [PMID: 24510013 PMCID: PMC3924024 DOI: 10.1007/s10549-014-2862-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 01/24/2014] [Indexed: 01/16/2023]
Abstract
Over one million American women have a benign breast biopsy annually. Sclerosing adenosis (SA) is a common, but poorly understood benign breast lesion demonstrating increased numbers of distorted lobules accompanied by stromal fibrosis. Few studies of its association with breast cancer have been conducted, with contradictory results. We studied SA in the Mayo Benign Breast Disease (BBD) Cohort, which includes women who had benign biopsies at Mayo-Rochester 1967–2001. Breast cancer risk in defined subsets was assessed using standardized incidence ratios (SIRs), relative to the Iowa Surveillance, Epidemiology, and End Results registry. This BBD cohort of 13,434 women was followed for a median of 15.7 years. SA was present in 3,733 women (27.8 %) who demonstrated an SIR for breast cancer of 2.10 (95 % CI 1.91–2.30) versus an SIR of 1.52 (95 % CI 1.42–1.63) for the 9,701 women without SA. SA was present in 62.4 % of biopsies with proliferative disease without atypia and 55.1 % of biopsies with atypical hyperplasia. The presence of SA stratified risk in subsets of women defined by age, involution status, and family history. However, SA does not further stratify risk in women diagnosed with other forms of proliferative breast disease, either with or without atypia. SA is a common proliferative lesion of the breast which, as a single feature, conveys an approximate doubling of breast cancer risk. Its role in breast carcinogenesis remains undefined; its presence may aid in risk prediction for women after a breast biopsy.
Collapse
|
42
|
Nie X, He J, Li Y, Pan DZ, Pan HX, Weng MX, Yang XP, Liu CP, Huang T. Accurate assessment of HER2 gene status for invasive component of breast cancer by combination of immunohistochemistry and chromogenic In Situ hybridization. ACTA ACUST UNITED AC 2013; 33:379-384. [PMID: 23771664 DOI: 10.1007/s11596-013-1128-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Indexed: 11/25/2022]
Abstract
The specimens of ductal carcinoma in situ (DCIS) with early invasion, and specimens collected by core needle biopsy (CNB) tend to contain limited amount of invasive component, so it is imperative to explore a new technique which can assess HER2 gene status accurately for the limited invasive cancer component in these specimens. Dual staining technique of combining immunohistochemistry (IHC) for myoepithelial cells and single or dual probe chromogenic in situ hybridization (CISH) for HER2 gene was performed on routinely processed paraffin sections from 20 cases diagnosed as having DCIS with invasive cancer. Among them, 10 had fluorescence in situ hybridization (FISH)-confirmed amplification of HER2 and 10 had FISH-confirmed non-amplification of HER2. We successfully detected HER2 genetic signals and myoepithelial IHC markers (SMM-HC or CK5/6) simultaneously on a single section in all 20 specimens. Myoepithelial markers and HER2 signals detected by dual staining assay were consistent with those by individual technique performed alone. HER2 gene amplification results determined by dual staining assay were 100% consistent with those of FISH. Dual staining technique which allows simultaneous detection of myoepithelial marker protein and cancerous HER2 gene is feasible, and it has potential to be used in clinical practice for effective determination of HER2 amplification in limited invasive component.
Collapse
Affiliation(s)
- Xiu Nie
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun He
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Li
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dan-Zhen Pan
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua-Xiong Pan
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mi-Xia Weng
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiu-Ping Yang
- Department of Pathology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun-Ping Liu
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Huang
- Department of Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
43
|
Tan PH, Ellis IO. Myoepithelial and epithelial-myoepithelial, mesenchymal and fibroepithelial breast lesions: updates from the WHO Classification of Tumours of the Breast 2012. J Clin Pathol 2013; 66:465-70. [PMID: 23533258 DOI: 10.1136/jclinpath-2012-201078] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the 4th edition of the WHO Classification of Tumours of the Breast, myoepithelial lesions are retitled myoepithelial and epithelial-myoepithelial lesions in order to better reflect the dual participation of luminal and myoepithelial compartments in some key entities. Malignant myoepithelioma, described as a section within the chapter on myoepithelial lesions in the 3rd edition, is recognised in the 4th edition as part of metaplastic carcinoma. Adenomyoepithelioma with malignancy is categorised in terms of the cellular component undergoing malignant transformation. The list of antibodies that can be used for identifying myoepithelial cells is updated. Among mesenchymal lesions, new additions are nodular fasciitis and atypical vascular lesions, while the haemangiopericytoma is removed. The 3rd edition stated that pathological prediction of behaviour of phyllodes tumours is difficult in the individual case. In the 4th edition, some progress has been made in prioritisation and weighting of histological parameters that can potentially estimate probability of recurrence. The WHO Working Group advocates leaning towards a diagnosis of fibroadenoma in cases where there is histological uncertainty in distinction from a benign phyllodes tumour, or adopting the neutral term 'benign fibroepithelial neoplasm', as the clinical behaviour of fibroadenoma overlaps with that of benign phyllodes tumour. The 3rd edition terminology of 'periductal stromal sarcoma' is revised to 'periductal stromal tumour', akin to the widespread consensus to avoid the use of the term 'cystosarcoma' in the context of phyllodes tumours.
Collapse
Affiliation(s)
- Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore
| | | |
Collapse
|
44
|
Abstract
Most diagnoses in breast pathology can be made with H&E sections. Nevertheless immunohistochemistry plays a useful supplementary role. This article reviews the common uses of immunohistochemistry in diagnostic breast pathology. It is important to be aware of the limitations of individual antibodies. Such problems can often be overcome by using panels of antibodies. Quality control is also essential: internal and external controls should show appropriate staining. Immunohistochemistry must be interpreted in combination with the morphology seen on H&E sections. Myoepithelial markers, such as smooth muscle actin, smooth muscle myosin heavy chain and p63, are useful for distinguishing invasive carcinoma from sclerosing lesions and ductal carcinoma in situ (DCIS), and in the classification of papillary lesions. Basal cytokeratins can help distinguish epithelial hyperplasia of usual type (UEH) and clonal proliferations such as DCIS and lobular carcinoma in situ (LCIS). UEH usually shows patchy expression whereas DCIS and other clonal proliferations are typically negative. E-cadherin can usually separate DCIS and LCIS: DCIS typically shows membrane staining and most LCIS is negative. Cytokeratins can be used to detect small nodal metastases or subtle invasive carcinomas such as invasive lobular carcinomas. Immunohistochemistry plays a useful role in diagnosing spindle cell lesions such as a panel of cytokeratins to identify spindle cell carcinomas. Immunohistochemistry is helpful in recognising metastases to the breast. Different antibodies are useful for different tumours: WT1 for ovarian carcinoma; TTF1 for pulmonary adenocarcinoma; S100, melan-A and HMB45 for melanoma; and lymphoid markers for lymphoma.
Collapse
Affiliation(s)
- Andrew H S Lee
- Department of Histopathology, Nottingham University Hospitals, City Hospital Campus, Nottingham NG5 1PB, UK.
| |
Collapse
|
45
|
Resetkova E. A Pictorial Journey Through Variants of Ductal Carcinoma in Situ (DCIS) and DCIS Mimickers. Clin Breast Cancer 2012; 12:347-63. [DOI: 10.1016/j.clbc.2012.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 06/06/2012] [Accepted: 07/09/2012] [Indexed: 11/16/2022]
|
46
|
Dewar R, Fadare O, Gilmore H, Gown AM. Best practices in diagnostic immunohistochemistry: myoepithelial markers in breast pathology. Arch Pathol Lab Med 2011; 135:422-9. [PMID: 21466356 DOI: 10.5858/2010-0336-cp.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Numerous immunohistochemical stains have been shown to exhibit exclusive or preferential positivity in breast myoepithelial cells relative to their luminal/epithelial counterparts. These myoepithelial markers provide invaluable assistance in accurately classifying breast proliferations, especially in core biopsies. Although numerous myoepithelial markers are available, they differ in their sensitivity, specificity, and ease of interpretation, which may be attributed, to a large extent, to the variable immunoreactivity of these markers in stromal cells including myofibroblasts, vessels, luminal/epithelial cells, and tumor cells. OBJECTIVE To review commonly used myoepithelial markers in breast pathology and a selection of diagnostic scenarios where they may be useful. DATA SOURCES The information outlined in this review article is based on our experiences with routine cases and a review of English-language articles published between 1987 and 2008. CONCLUSIONS To demonstrate the presence or absence of myoepithelial cells, a panel-based approach of 2 or more markers is recommended. Markers that most effectively combine sensitivity, specificity, and ease of interpretation include smooth muscle myosin heavy chains, calponin, p75, p63, P-cadherin, basal cytokeratins, maspin, and CD10. These markers, however, display varying cross-reactivity patterns and variably reduced expression in the myoepithelial cells bordering in situ carcinomas. The choice of a myoepithelial marker should be dependent on a combination of factors, including published evidence on its diagnostic utility, its availability, performance characteristics that have been achieved in a given laboratory, and the specific diagnostic scenario. When its use is deemed necessary, immunohistochemistry for myoepithelial cells in breast pathology is most effective when conceptualized as supplemental, rather than central to routine morphologic interpretation.
Collapse
Affiliation(s)
- Rajan Dewar
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | |
Collapse
|
47
|
Sänger N, Effenberger KE, Riethdorf S, Van Haasteren V, Gauwerky J, Wiegratz I, Strebhardt K, Kaufmann M, Pantel K. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. Int J Cancer 2011; 129:2522-6. [PMID: 21207426 DOI: 10.1002/ijc.25895] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 11/03/2010] [Accepted: 11/30/2010] [Indexed: 01/16/2023]
Abstract
Detection of disseminated tumor cells (DTCs) in bone marrow is an independent prognostic factor in primary breast cancer. Here, we conducted a proof-of-principle study to evaluate whether this tumor cell spread occurs already in patients with ductal carcinoma in situ (DCIS). After preoperative screening by stereotactic core biopsy, 30 consecutive women with DCIS were included. Bone marrow aspirates, taken at the time of primary surgery, were subjected to DTC detection by a standardized immunoassay using the established monoclonal anti-cytokeratin antibodies A45-B/B3 and AE1/AE3. DTCs were detected in 4 of 19 cases of pure DCIS (21.1%) and in four of seven cases of DCIS with microinvasion (57.1%). After a median follow-up time of 22 months, two initially DTC-positive patients suffered from contralateral carcinoma and contralateral DCIS at months 12 and 30, respectively, whereas the remaining patients were relapse free. Thus, hematogenous tumor cell dissemination into bone marrow is an early event in breast cancer development.
Collapse
Affiliation(s)
- Nicole Sänger
- Department of Gynaecology and Obstetrics, University Hospital Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Polyak K. Molecular markers for the diagnosis and management of ductal carcinoma in situ. J Natl Cancer Inst Monogr 2011; 2010:210-3. [PMID: 20956832 DOI: 10.1093/jncimonographs/lgq019] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is a heterogeneous group of lesions reflecting the proliferation of malignant cells within the ducts of the breast without invasion through the basement membrane. Numerous studies analyzing the molecular profiles of DCIS using genome-wide unbiased and candidate gene approaches have been conducted with the aim of identifying clinically useful markers that would predict the risk of progression to invasion. Results of these investigations defined the heterogeneity of DCIS at the molecular level, but a gene signature predictive of invasive progression has not been identified. Major diagnostic criteria that differentiate DCIS from invasive cancer are the presence of intact basement membrane and myoepithelial cell layer. Based on this, perturbation of normal myoepithelial cell differentiation has been proposed to explain progression to invasion. Comprehensive molecular studies analyzing large cohorts of DCIS with long-term clinical follow-up are necessary to resolve the many remaining questions.
Collapse
Affiliation(s)
- Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St D740C, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Cichon MA, Degnim AC, Visscher DW, Radisky DC. Microenvironmental influences that drive progression from benign breast disease to invasive breast cancer. J Mammary Gland Biol Neoplasia 2010; 15:389-97. [PMID: 21161341 PMCID: PMC3011086 DOI: 10.1007/s10911-010-9195-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 12/03/2010] [Indexed: 12/21/2022] Open
Abstract
Invasive breast cancer represents the endpoint of a developmental process that originates in the terminal duct lobular units and is believed to progress through stages of increasing proliferation, atypical hyperplasia, and carcinoma in situ before the cancer acquires invasive and metastatic capabilities. By comparison with invasive breast cancer, which has been studied extensively, the preceding stages of benign breast disease are more poorly understood. Much less is known about the molecular changes underlying benign breast disease development and progression, as well as the transition from in situ into invasive disease. Even less focus has been given to the specific role of stroma in this progression. The reasons for lack of knowledge about these lesions often come from their small size and limited sample availability. More challenges are posed by limitations of the models used to investigate the lesions preceding invasive breast cancer. However, recent studies have identified alterations in stromal cell function that may be critical for disease progression from benign disease to invasive cancer: key functions of myoepithelial cells that maintain tissue structure are lost, while tissue fibroblasts become activated to produce proteases that degrade the extracellular matrix and trigger the invasive cellular phenotype. Gene expression profiling of stromal alterations associated with disease progression has also identified key transcriptional changes that occur early in disease development. In this review, we will summarize recent studies showing how stromal factors can facilitate progression of ductal carcinoma in situ to invasive disease. We also suggest approaches to identify processes that control earlier stages of disease progression.
Collapse
Affiliation(s)
| | - Amy C. Degnim
- Department of Surgery, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Derek C. Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224 USA
| |
Collapse
|
50
|
Schnitt SJ. Molecular biology of breast tumor progression: a view from the other side. Int J Surg Pathol 2010; 18:170S-173S. [PMID: 20484285 DOI: 10.1177/1066896910370773] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Stuart J Schnitt
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|