101
|
Tan DSP, Lambros MBK, Natrajan R, Reis-Filho JS. Getting it right: designing microarray (and not 'microawry') comparative genomic hybridization studies for cancer research. J Transl Med 2007; 87:737-54. [PMID: 17558419 DOI: 10.1038/labinvest.3700593] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The development of high-resolution microarray-based comparative genomic hybridization (aCGH), using cDNA, bacterial artificial chromosome (BAC) and oligonucleotide probes, is providing tremendous opportunities for translational research by facilitating detailed analysis of entire cancer genomes in a single experiment. However, this technology will only fulfil its promise if studies incorporating aCGH are designed with a full understanding of its current limitations and the strategies available to circumvent them. While there have been several excellent reviews on the current status of this technology, there is currently very little guidance available regarding the appropriate design of experiments incorporating aCGH (including the strengths and weaknesses of each platform), and how best to combine the results obtained from aCGH with other 'omic' technologies, including gene expression. In this review, we present the key design issues that need to be considered in order to optimize aCGH studies, including sample selection, the definition of appropriate experimental objectives, arguments for and against the various microarray platforms that are currently available, and methods for data validation and integration. It is envisaged that future well-designed aCGH studies will enhance our understanding of the genetic basis of cancer, and lead to the identification of novel predictive and prognostic cancer biomarkers, as well as molecular therapeutic targets in cancer.
Collapse
Affiliation(s)
- David S P Tan
- Molecular Pathology Team, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | |
Collapse
|
102
|
Lambros MBK, Natrajan R, Reis-Filho JS. Chromogenic and fluorescent in situ hybridization in breast cancer. Hum Pathol 2007; 38:1105-22. [PMID: 17640550 DOI: 10.1016/j.humpath.2007.04.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 04/21/2007] [Accepted: 04/23/2007] [Indexed: 11/29/2022]
Abstract
Fluorescent (FISH) and chromogenic (CISH) in situ hybridization have recently become part of the diagnostic armamentarium of breast pathologists. HER2 gene testing by FISH and/or CISH has become an integral part of the diagnostic workup for patients with breast cancer. In this era of high throughput technologies, these techniques have proven instrumental for the validation of results from microarray-based comparative genomic hybridization and for the identification of novel oncogenes and tumor suppressor genes. Furthermore, FISH and CISH applied to tissue microarrays have expedited the characterization of genomic changes associated with specific breast cancer molecular subtypes and the identification of novel prognostic and predictive markers. In this review, we provide in this review a critical assessment of CISH and FISH and the impact of the analysis of amplification of specific oncogenes (eg, HER2, EGFR, MYC, CCND1, and FGFR1) and deletion of tumor suppressor genes (eg, BRCA1 and BRCA2) on our understanding of breast cancer.
Collapse
Affiliation(s)
- Maryou B K Lambros
- Molecular Pathology Laboratory, The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, SW3 6JB London, UK
| | | | | |
Collapse
|
103
|
Elsheikh S, Green AR, Aleskandarany MA, Grainge M, Paish CE, Lambros MBK, Reis-Filho JS, Ellis IO. CCND1 amplification and cyclin D1 expression in breast cancer and their relation with proteomic subgroups and patient outcome. Breast Cancer Res Treat 2007; 109:325-35. [PMID: 17653856 DOI: 10.1007/s10549-007-9659-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 06/12/2007] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Despite strong evidence regarding the role of CCND1 amplification and protein overexpression in breast carcinoma, the associations between CCND1 amplification/cyclin D1 overexpression and clinicopathological variables and clinical outcome remain controversial. AIMS OF THE STUDY (1) to correlate cyclin D1 expression with gene amplification; (2) to analyse the correlations between CCND1 amplification and overexpression with clinicopathological features and patients' outcome in invasive breast cancer; (3) to define the prevalence and clinical significance of cyclin D1 overexpression and CCND1 amplification in ER positive breast carcinomas (4) to define the prevalence of cyclin D1 overexpression and CCND1 amplification in breast cancers with basal-like immunophenotype. MATERIALS AND METHODS CCND1 amplification and protein expression were assessed on a tissue microarray containing 880 unselected invasive breast cancer cases, by means of chromogenic in situ hybridisation using the Spotlight CCND1 amplification probe and immunohistochemistry, using the rabbit monoclonal antibody SP4. RESULTS A total of 59/613 tumours (9.6%) showed CCND1 amplification and 224/514 (43.6%) showed strong cyclin D1 expression. A strong positive correlation between CCND1 amplification and higher levels of cyclin D1 expression was found (P < 0.001). Basal-like cancers showed infrequent CCND1 amplification and cyclin D1 overexpression (P < 0.001). Both CCND1 amplification and cyclin D1 expression were associated with positive ER status. CCND1 gene amplification was an independent prognostic factor for patients with ER positive breast cancer. CONCLUSION Our results demonstrate a strong correlation between CCND1 amplification and its protein expression in breast cancer. However, protein expression is more pervasive than gene amplification and associated with ER expression.
Collapse
Affiliation(s)
- Somaia Elsheikh
- Department of Histopathology, School of Molecular Medical Sciences, Nottingham City Hospital NHS Trust, University of Nottingham and University Hospitals NHS Trust, Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
104
|
Lavoué V, Graesslin O, Classe JM, Fondrinier E, Angibeau H, Levêque J. Management of lobular neoplasia diagnosed by core needle biopsy: study of 52 biopsies with follow-up surgical excision. Breast 2007; 16:533-9. [PMID: 17629481 DOI: 10.1016/j.breast.2007.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 04/16/2007] [Accepted: 04/20/2007] [Indexed: 10/23/2022] Open
Abstract
Lobular neoplasia (LN) is a risk factor for bilateral breast cancer without consensus as to its appropriate management. The authors report on a retrospective multi-institutional study concerning 52 patients in whom a diagnosis of LN was made after core needle biopsy (CNB) and who subsequently underwent surgical excision. The excision specimens revealed seven cases of invasive carcinoma and three cases of ductal carcinoma in situ, indicating an underestimation of lesions at CNB in 19% of cases, and in particular in those patients with pleomorphic LN, and when clinical, radiological masses were detected. This lesion is increasingly being diagnosed by CNB due to widespread screening. Follow-up surgical excision should be performed in order to examine the whole lesion in the case of masses or when the histologic specimen reveals a pleomorphic subtype. In other cases, annual mammographic surveillance should be undertaken due to the persistent long-term risk of developing bilateral breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Biopsy, Needle/statistics & numerical data
- Breast Neoplasms/epidemiology
- Breast Neoplasms/etiology
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Carcinoma, Ductal, Breast/epidemiology
- Carcinoma, Ductal, Breast/etiology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Intraductal, Noninfiltrating/epidemiology
- Carcinoma, Intraductal, Noninfiltrating/etiology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/surgery
- Carcinoma, Lobular/epidemiology
- Carcinoma, Lobular/etiology
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/surgery
- Female
- France/epidemiology
- Humans
- Middle Aged
- Retrospective Studies
- Risk Factors
Collapse
Affiliation(s)
- Vincent Lavoué
- Department of Obstetric Gynecology, CHU Tenon, 4 rue de la Chine 75020 Paris, France
| | | | | | | | | | | |
Collapse
|
105
|
Leibl S, Regitnig P, Moinfar F. Flat epithelial atypia (DIN 1a, atypical columnar change): an underdiagnosed entity very frequently coexisting with lobular neoplasia. Histopathology 2007; 50:859-65. [PMID: 17543075 DOI: 10.1111/j.1365-2559.2007.02700.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS Flat epithelial atypia of the breast [FEA; synonyms: ductal intraepithelial neoplasia (DIN) 1a, atypical columnar change] is increasingly recognized by pathologists and shows distinct genetic alterations. The aim of this study was to determine its biological significance as an incidental finding in breast biopsy specimens. METHODS AND RESULTS On the assumption that both FEA and lobular neoplasia (LN) derive from progenitor cells in the terminal ductal-lobular unit, we investigated the association between FEA and LN semiquantitatively in 111 excisional breast biopsy specimens which contained LN, but did not contain ductal carcinoma in situ (DCIS) or invasive carcinoma. Ninety-six cases (86.5%) revealed coexistence of LN and FEA (P < 0001). The distribution of LN was focal in 41 cases (37%), multifocal in 50 (45%) and extensive in 20 (18%) cases. FEA was identified as focal, multifocal and extensive in 29 (26%), 42 (38%) and 25 (23%) cases, respectively. Distribution patterns of LN and FEA showed no statistically significant correlation. CONCLUSIONS Due to the striking association between LN and FEA in our material, one may speculate that these two lesions are biologically related and that FEA is an early but non-obligate precursor lesion similar to LN. Based on this assumption, regular clinical and mammographic follow-up of patients with FEA would be prudent.
Collapse
Affiliation(s)
- S Leibl
- Institute of Pathology, Medical University of Graz, Graz, Austria.
| | | | | |
Collapse
|
106
|
Svensson Månsson S, Reis-Filho J, Landberg G. Transcriptional upregulation and unmethylation of the promoter region of p16 in invasive basal cell carcinoma cells and partial co-localization with the gamma 2 chain of laminin-332. J Pathol 2007; 212:102-11. [PMID: 17370299 DOI: 10.1002/path.2152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Accepted: 01/25/2007] [Indexed: 01/06/2023]
Abstract
Basal cell carcinoma cells show low proliferation rates at the invasive front and a concordant upregulation of the cdk-inhibitor p16, limiting proliferative capacity. Little is known about the mechanisms of p16 regulation in normal and malignant cells apart from that many transcription factors such as Ets1, Ets2, SP1, SP3, JunB and the polycomb protein Bmi1 have the potential to induce or repress p16 expression. Therefore, the aim of this study was to determine how p16 is regulated in basal cell carcinoma with special focus on its upregulation in invasive cells. By analysing various microdissected areas of basal cell carcinoma using real-time quantitative PCR we observed upregulation of p16 mRNA in invasive tumour cells compared to centrally localized tumour cells. The methylation status of the p16 promoter, analysed by methylation-specific PCR, also showed diminished methylation in tumour cells at the invasive front, supporting the hypothesis that promoter methylation can affect the transcriptional activation of p16 in vivo. There was only sporadic co-localization of Ets, or ERK1/2 phosphorylation with p16 upregulation at the invasive front, suggesting that these factors were not directly involved in the regulation of p16. Furthermore, the gamma 2 chain of laminin-332 has been reported to be increased at the invasive front compared to the central areas of many tumours. Interestingly, in basal cell carcinoma we observed partial co-localization between p16 and the gamma 2 chain of laminin-332 in tumour cells towards areas of ulceration and in the majority of clearly infiltrative tumour cells but not in p16 positive tumour cells with a more pushing invasive growth pattern. These data suggest that concurrent p16 upregulation and decreased proliferation are more general phenomena in different types of invasive growth patterns in basal cell carcinomas and that these only partially overlap with the gamma 2 chain of laminin-332 associated invasion patterns.
Collapse
Affiliation(s)
- S Svensson Månsson
- Division of Pathology, Department of Laboratory Medicine, Lund University, UMAS, SE-205 02 Malmö, Sweden
| | | | | |
Collapse
|
107
|
Tischkowitz M, Xia B, Sabbaghian N, Reis-Filho JS, Hamel N, Li G, van Beers EH, Li L, Khalil T, Quenneville LA, Omeroglu A, Poll A, Lepage P, Wong N, Nederlof PM, Ashworth A, Tonin PN, Narod SA, Livingston DM, Foulkes WD. Analysis of PALB2/FANCN-associated breast cancer families. Proc Natl Acad Sci U S A 2007; 104:6788-93. [PMID: 17420451 PMCID: PMC1871863 DOI: 10.1073/pnas.0701724104] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
No more than approximately 30% of hereditary breast cancer has been accounted for by mutations in known genes. Most of these genes, such as BRCA1, BRCA2, TP53, CHEK2, ATM, and FANCJ/BRIP1, function in DNA repair, raising the possibility that germ line mutations in other genes that contribute to this process also predispose to breast cancer. Given its close relationship with BRCA2, PALB2 was sequenced in affected probands from 68 BRCA1/BRCA2-negative breast cancer families of Ashkenazi Jewish, French Canadian, or mixed ethnic descent. The average BRCAPRO score was 0.58. A truncating mutation (229delT) was identified in one family with a strong history of breast cancer (seven breast cancers in three female mutation carriers). This mutation and its associated breast cancers were characterized with another recently reported but unstudied mutation (2521delA) that is also associated with a strong family history of breast cancer. There was no loss of heterozygosity in tumors with either mutation. Moreover, comparative genomic hybridization analysis showed major similarities to that of BRCA2 tumors but with some notable differences, especially loss of 18q, a change that was previously unknown in BRCA2 tumors and less common in sporadic breast cancer. This study supports recent observations that PALB2 mutations are present, albeit not frequently, in breast cancer families. The apparently high penetrance noted in this study suggests that at least some PALB2 mutations are associated with a substantially increased risk for the disease.
Collapse
Affiliation(s)
- Marc Tischkowitz
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada H2W 1S6
- Segal Cancer Centre and
| | - Bing Xia
- Dana–Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115
| | - Nelly Sabbaghian
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Segal Cancer Centre and
| | - Jorge S. Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, United Kingdom
| | - Nancy Hamel
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- The Research Institute, McGill University Health Centre, Montréal, QC, Canada H3G 1A4
| | - Guilan Li
- Dana–Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115
| | | | - Lili Li
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Segal Cancer Centre and
| | - Tayma Khalil
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada H2W 1S6
- Segal Cancer Centre and
| | - Louise A. Quenneville
- Department of Pathology, McGill University Sir M. B. Davis-Jewish General Hospital, 3755 Côte St-Catherine, Montréal, QC, Canada H3T 1E2
| | - Atilla Omeroglu
- Department of Pathology, McGill University, Montréal, QC, Canada, H3A 2B4
| | - Aletta Poll
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada M5G 1N9; and
| | - Pierre Lepage
- McGill University and Genome Québec Innovation Centre, Montréal, QC, Canada H3A 1A4
| | - Nora Wong
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada H2W 1S6
- Segal Cancer Centre and
| | - Petra M. Nederlof
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Alan Ashworth
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, United Kingdom
| | - Patricia N. Tonin
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada H2W 1S6
- The Research Institute, McGill University Health Centre, Montréal, QC, Canada H3G 1A4
| | - Steven A. Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada M5G 1N9; and
| | - David M. Livingston
- Dana–Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115
- To whom correspondence may be addressed. E-mail:
| | - William D. Foulkes
- Program in Cancer Genetics, Departments of Oncology and Human Genetics and
- Departments of Medicine and Human Genetics, McGill University, Montréal, QC, Canada H2W 1S6
- Segal Cancer Centre and
- The Research Institute, McGill University Health Centre, Montréal, QC, Canada H3G 1A4
- To whom correspondence may be addressed at:
Cancer Prevention Centre, Segal Cancer Centre, Sir M. B. Davis-Jewish General Hospital, 3755 Côte St-Catherine, Montréal, QC, Canada H3T 1E2. E-mail:
| |
Collapse
|
108
|
Pinder SE, Provenzano E, Reis-Filho JS. Lobular in situ neoplasia and columnar cell lesions: diagnosis in breast core biopsies and implications for management. Pathology 2007; 39:208-16. [PMID: 17454750 DOI: 10.1080/00313020701230849] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Histopathologists are encountering intra-lobular epithelial proliferations more frequently in core biopsies taken from lesions identified in mammographic breast screening programmes. In particular, columnar cell lesions are increasingly being seen in core biopsies taken for the histological assessment of mammographically detected microcalcifications. The morphological features of lobular neoplasia are relatively well known, but columnar cell lesions, particularly forms with atypical features, are less widely recognised. The biological and clinical significance of both of these intra-lobular processes is controversial, (1) as indicators of adjacent malignancy when encountered in core biopsy, (2) the relative risk conferred of development of subsequent malignancy, and (3) their precursor behaviour. For this reason, the optimal clinical management of these lesions, particularly when encountered on core biopsy, is unclear. This review provides an update on the histological diagnosis of lobular neoplasia and columnar cell lesions and outlines recent clinico-pathological and molecular findings with discussion on clinical management.
Collapse
Affiliation(s)
- Sarah E Pinder
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | |
Collapse
|
109
|
Katz A, Saad ED, Porter P, Pusztai L. Primary systemic chemotherapy of invasive lobular carcinoma of the breast. Lancet Oncol 2007; 8:55-62. [PMID: 17196511 DOI: 10.1016/s1470-2045(06)71011-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Invasive lobular carcinoma is the second most frequent histological type of breast cancer and its incidence is increasing. It has unique clinical, biological, and molecular features. Invasive lobular carcinoma is almost invariably positive for the oestrogen receptor and, when compared with invasive ductal carcinoma, it is typically of a lower grade. Even though invasive lobular carcinoma represents a distinct clinical entity, the same criteria used for invasive ductal carcinoma are currently applied to establish the need for primary or adjuvant systemic chemotherapy. We reviewed randomised trials of neoadjuvant and adjuvant chemotherapy and noted that insufficient evidence is available to support or withhold use of chemotherapy in patients with invasive lobular carcinoma. Thus, the benefit from systemic chemotherapy for individuals with this form of breast disease is unclear. Invasive lobular carcinoma deserves to be investigated separately in prospective clinical trials to define the best treatment and prevention strategies.
Collapse
Affiliation(s)
- Artur Katz
- Centro Paulista de Oncologia and Hospital Albert Einstein, Sao Paulo, Brazil.
| | | | | | | |
Collapse
|
110
|
Joosse SA, van Beers EH, Nederlof PM. Automated array-CGH optimized for archival formalin-fixed, paraffin-embedded tumor material. BMC Cancer 2007; 7:43. [PMID: 17343727 PMCID: PMC1829401 DOI: 10.1186/1471-2407-7-43] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Accepted: 03/07/2007] [Indexed: 01/10/2023] Open
Abstract
Background Array Comparative Genomic Hybridization (aCGH) is a rapidly evolving technology that still lacks complete standardization. Yet, it is of great importance to obtain robust and reproducible data to enable meaningful multiple hybridization comparisons. Special difficulties arise when aCGH is performed on archival formalin-fixed, paraffin-embedded (FFPE) tissue due to its variable DNA quality. Recently, we have developed an effective DNA quality test that predicts suitability of archival samples for BAC aCGH. Methods In this report, we first used DNA from a cancer cell-line (SKBR3) to optimize the aCGH protocol for automated hybridization, and subsequently optimized and validated the procedure for FFPE breast cancer samples. We aimed for highest throughput, accuracy, and reproducibility applicable to FFPE samples, which can also be important in future diagnostic use. Results Our protocol of automated array-CGH on archival FFPE ULS-labeled DNA showed very similar results compared with published data and our previous manual hybridization method. Conclusion This report combines automated aCGH on unamplified archival FFPE DNA using non-enzymatic ULS labeling, and describes an optimized protocol for this combination resulting in improved quality and reproducibility.
Collapse
Affiliation(s)
- Simon A Joosse
- Division of Experimental Therapy, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Erik H van Beers
- Division of Experimental Therapy, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Petra M Nederlof
- Department of Pathology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
111
|
Yoder BJ, Wilkinson EJ, Massoll NA. Molecular and Morphologic Distinctions between Infiltrating Ductal and Lobular Carcinoma of the Breast. Breast J 2007; 13:172-9. [PMID: 17319859 DOI: 10.1111/j.1524-4741.2007.00393.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Histopathologic distinction between ductal and lobular carcinomas of the breast has been made since 1941. Together, these two subtypes account for >95% of all mammary carcinomas. With the recent advances in molecular techniques, our understanding of the biology behind these carcinomas has greatly expanded. The genomic aberrations in mammary carcinoma are highly complex and appear to be more associated with tumor grade rather than any histopathologic subtype. Protein and RNA expression profiling reveals a classification of mammary carcinoma that has some overlap with traditional histopathology and can at least partially explain clinical behavior. The goal of this review is to present what is currently known about the molecular profiles of infiltrating ductal and lobular carcinoma and how they relate to conventional histopathology and biologic behavior.
Collapse
MESH Headings
- Antigens, CD
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cadherins/genetics
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Chromosome Aberrations
- Female
- Gene Expression Profiling
- Genes, erbB-2/genetics
- Genomic Instability
- Humans
- Mutation
- Receptor, ErbB-2/metabolism
Collapse
Affiliation(s)
- Brian J Yoder
- Department of Pathology, University of Florida, Gainesville, Florida 33805, USA.
| | | | | |
Collapse
|
112
|
Pierga JY, Reis-Filho JS, Cleator SJ, Dexter T, MacKay A, Simpson P, Fenwick K, Iravani M, Salter J, Hills M, Jones C, Ashworth A, Smith IE, Powles T, Dowsett M. Microarray-based comparative genomic hybridisation of breast cancer patients receiving neoadjuvant chemotherapy. Br J Cancer 2007; 96:341-51. [PMID: 17133270 PMCID: PMC2359992 DOI: 10.1038/sj.bjc.6603483] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 10/12/2006] [Accepted: 10/23/2006] [Indexed: 12/29/2022] Open
Abstract
We analysed the molecular genetic profiles of breast cancer samples before and after neoadjuvant chemotherapy with combination doxorubicin and cyclophosphamide (AC). DNA was obtained from microdissected frozen breast core biopsies from 44 patients before chemotherapy. Additional samples were obtained before the second course of chemotherapy (D21) and after the completion of the treatment (surgical specimens) in 17 and 21 patients, respectively. Microarray-based comparative genome hybridisation was performed using a platform containing approximately 5800 bacterial artificial chromosome clones (genome-wide resolution: 0.9 Mb). Analysis of the 44 pretreatment biopsies revealed that losses of 4p, 4q, 5q, 12q13.11-12q13.12, 17p11.2 and 17q11.2; and gains of 1p, 2p, 7q, 9p, 11q, 19p and 19q were significantly associated with oestrogen receptor negativity. 16q21-q22.1 losses were associated with lobular and 8q24 gains with ductal types. Losses of 5q33.3-q4 and 18p11.31 and gains of 6p25.1-p25.2 and Xp11.4 were associated with HER2 amplification. No correlations between DNA copy number changes and clinical response to AC were found. Microarray-based comparative genome hybridisation analysis of matched pretreatment and D21 biopsies failed to identify statistically significant differences, whereas a comparison between matched pretreatment and surgical samples revealed a statistically significant acquired copy number gain on 11p15.2-11p15.5. The modest chemotherapy-driven genomic changes, despite profound loss of cell numbers, suggest that there is little therapeutic selection of resistant non-modal cell lineages.
Collapse
Affiliation(s)
- J-Y Pierga
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
- Medical Oncology Department, Institut Curie, Paris, cedex 5, France
| | - J S Reis-Filho
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - S J Cleator
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - T Dexter
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - A MacKay
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - P Simpson
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - K Fenwick
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - M Iravani
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - J Salter
- Academic Department of Biochemistry, Royal Marsden Hospital NHS Trust, Fulham Road, London, SW3 6JJ, UK
| | - M Hills
- Academic Department of Biochemistry, Royal Marsden Hospital NHS Trust, Fulham Road, London, SW3 6JJ, UK
| | - C Jones
- Section of Paediatric Oncology, Institute of Cancer Research, London, SM2 5NG, UK
| | - A Ashworth
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
| | - I E Smith
- Breast Cancer Unit, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - T Powles
- Breast Cancer Unit, Royal Marsden Hospital, London, SW3 6JJ, UK
| | - M Dowsett
- Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, SW3 6JB, UK
- Academic Department of Biochemistry, Royal Marsden Hospital NHS Trust, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
113
|
Arriola E, Lambros MBK, Jones C, Dexter T, Mackay A, Tan DSP, Tamber N, Fenwick K, Ashworth A, Dowsett M, Reis-Filho JS. Evaluation of Phi29-based whole-genome amplification for microarray-based comparative genomic hybridisation. J Transl Med 2007; 87:75-83. [PMID: 17170740 DOI: 10.1038/labinvest.3700495] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
For the optimal performance of high throughput genomic technologies sufficient yields of high-quality DNA are crucial. Following microdissection, most samples fail to produce sufficient quantities of DNA for genome-wide experiments. Various PCR-based amplification methods have been used, but these usually produce nonuniform representations of the genome. Bacteriophage Phi29 DNA polymerase random-primed DNA amplification is based on isothermal multiple displacement amplification. We sought to define the genome representation of this method in a bacterial artificial chromosome microarray comparative genomic hybridisation (aCGH) platform. Test genomic female DNA was amplified using Phi29 amplification at four different starting concentrations (0.5, 5, 10 and 50 ng). These products were combined with unamplified and amplified genomic female DNA as reference. In addition, 50 ng of DNA from five microdissected breast cancer frozen samples, were amplified using the same method. Three combinations were performed: unamplified test with unamplified reference, amplified test with unamplified reference and both amplified tumour and reference DNA. aCGH was performed with an in-house 16 K BAC platform (a resolution of approximately 100 Kb). Pearson's correlation tests and hierarchical clustering were performed to compare the profiles obtained. aCGH profiles obtained with amplified test and unamplified reference female genomic DNA showed copy number biases throughout the genome. These biases were more conspicuous with smaller amounts of starting material and mapped to regions of known copy number polymorphisms. When similar concentrations of test and reference DNA were amplified, the biases were significantly reduced, rendering accurate profiles. For the tumours, representative profiles were obtained when both test and reference DNA were amplified. Phi29 amplification induces copy number biases and unamplified material remains the gold standard for copy number analysis. For accurate results using Phi29 amplification, samples subjected to aCGH analysis should be combined with reference DNA amplified with the same method, using similar amounts of starting template.
Collapse
Affiliation(s)
- Edurne Arriola
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Abstract
Lobular neoplasia is a relatively uncommon lesion, which is frequently diagnosed in biopsy specimens taken for other reasons. Although the histological features of this lesion are well known, its biological significance as a "risk indicator" or "breast cancer precursor" has been a matter of debate. This review provides an update on recent clinicopathological and molecular data on lobular neoplasia and how these have changed the way these lesions are perceived and, most importantly, managed. Furthermore, the current recommendations for the management of lobular neoplasia diagnosed on core needle biopsies proposed in the National Health Service Breast Cancer Screening guidelines are discussed.
Collapse
Affiliation(s)
- Jorge S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK.
| | | |
Collapse
|
115
|
Rodriguez-Pinilla SM, Jones RL, Lambros MBK, Arriola E, Savage K, James M, Pinder SE, Reis-Filho JS. MYC amplification in breast cancer: a chromogenic in situ hybridisation study. J Clin Pathol 2006; 60:1017-23. [PMID: 17158641 PMCID: PMC1972423 DOI: 10.1136/jcp.2006.043869] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
AIMS To analyse the correlation between MYC amplification and various clinicopathological features and outcome in a cohort of 245 patients with invasive breast carcinoma treated with surgery followed by anthracycline-based chemotherapy. Given the high prevalence of MYC amplification in tumours of BRCA1 mutation carriers and the similarities between these and sporadic "basal-like" carcinomas, the prevalence of MYC amplification in "basal-like" breast carcinomas was investigated. METHODS MYC gene copy number was assessed on tissue microarrays containing duplicate cores of 245 invasive breast carcinomas by means of chromogenic in situ hybridisation using SpotLight C-MYC amplification probe and chromosome 8 centromeric probe (CEP8). Signals were evaluated at 400x magnification; 30 morphologically unequivocal neoplastic cells in each core were counted for the presence of the gene and CEP8 probes. RESULTS Amplification was defined as a MYC:CEP8 ratio >2. Signals for both MYC and CEP8 were assessable in 196/245 (80%) tumours. MYC amplification was found in 19/196 cases (9.7%) and was not associated with tumour size, histological grade, positivity for oestrogen receptor, progesterone receptor, HER2, epidermal growth factor, cytokeratins 14, 5/6 and 17, MIB1 or p53. Only 4% of basal-like carcinomas showed MYC amplification, compared to 8.75% and 10.7% of luminal and HER2 tumours respectively. On univariate analysis, MYC amplification displayed a significant association with shorter metastasis-free and overall survival and proved to be an independent prognostic factor on multivariate survival analysis. CONCLUSION MYC amplification is not associated with "basal-like" phenotype and proved to be an independent prognostic factor for breast cancer patients treated with anthracycline-based chemotherapy.
Collapse
|
116
|
Natrajan R, Reis-Filho JS, Little SE, Messahel B, Brundler MA, Dome JS, Grundy PE, Vujanic GM, Pritchard-Jones K, Jones C. Blastemal Expression of Type I Insulin-Like Growth Factor Receptor in Wilms' Tumors Is Driven by Increased Copy Number and Correlates with Relapse. Cancer Res 2006; 66:11148-55. [PMID: 17145858 DOI: 10.1158/0008-5472.can-06-1931] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most Wilms' tumors are of low stage, favorable histology, and have a high likelihood of cure with current multimodal therapy. Despite this, there remains a group of patients whose tumors recur for whom intensive salvage regimens result in survival of only 50%. Fitting a Cox proportional hazards model to microarray-based comparative genomic hybridization (aCGH) data on 68 Wilms' tumor samples, we identified a significant correlation between increased copy number at chromosome 15q26.3 insulin-like growth factor I receptor (IGFIR) and tumor relapse (adjusted P = 0.014). Wilms' tumors (13%) exhibited a low-level gain corresponding to three to four copies of the gene by aCGH analysis, 9 of 10 of which exhibited high IGFIR mRNA levels. Although IGFIR protein expression was restricted to the epithelial cells of fetal kidney and Wilms' tumors in most cases, 12% of tumors were also found to express IGFIR in the blastemal compartment. Blastemal IGFIR protein expression was associated with an increased copy number and a shorter relapse-free survival time (P = 0.027, log-rank test). In addition to the membrane localization, IGFIR was localized to the perinuclear region of the blastemal cells in 6% of Wilms' tumors. These data provide evidence that an increase in IGFIR gene copy number results in aberrant expression in the blastemal compartment of some Wilms' tumors and is associated with an adverse outcome in these patients. These findings suggest the possibility of use of targeted agents in the therapy of these children.
Collapse
Affiliation(s)
- Rachael Natrajan
- Paediatric Oncology, Institute of Cancer Research/Royal Marsden National Health Service Trust, Sutton, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Reis-Filho JS, Simpson PT, Turner NC, Lambros MB, Jones C, Mackay A, Grigoriadis A, Sarrio D, Savage K, Dexter T, Iravani M, Fenwick K, Weber B, Hardisson D, Schmitt FC, Palacios J, Lakhani SR, Ashworth A. FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas. Clin Cancer Res 2006; 12:6652-62. [PMID: 17121884 DOI: 10.1158/1078-0432.ccr-06-1164] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Classic lobular carcinomas (CLC) account for 10% to 15% of all breast cancers. At the genetic level, CLCs show recurrent physical loss of chromosome16q coupled with the lack of E-cadherin (CDH1 gene) expression. However, little is known about the putative therapeutic targets for these tumors. The aim of this study was to characterize CLCs at the molecular genetic level and identify putative therapeutic targets. EXPERIMENTAL DESIGN We subjected 13 cases of CLC to a comprehensive molecular analysis including immunohistochemistry for E-cadherin, estrogen and progesterone receptors, HER2/neu and p53; high-resolution comparative genomic hybridization (HR-CGH); microarray-based CGH (aCGH); and fluorescent and chromogenic in situ hybridization for CCND1 and FGFR1. RESULTS All cases lacked the expression of E-cadherin, p53, and HER2, and all but one case was positive for estrogen receptors. HR-CGH revealed recurrent gains on 1q and losses on 16q (both, 85%). aCGH showed a good agreement with but higher resolution and sensitivity than HR-CGH. Recurrent, high level gains at 11q13 (CCND1) and 8p12-p11.2 were identified in seven and six cases, respectively, and were validated with in situ hybridization. Examination of aCGH and the gene expression profile data of the cell lines, MDA-MB-134 and ZR-75-1, which harbor distinct gains of 8p12-p11.2, identified FGFR1 as a putative amplicon driver of 8p12-p11.2 amplification in MDA-MB-134. Inhibition of FGFR1 expression using small interfering RNA or a small-molecule chemical inhibitor showed that FGFR1 signaling contributes to the survival of MDA-MB-134 cells. CONCLUSIONS Our findings suggest that receptor FGFR1 inhibitors may be useful as therapeutics in a subset of CLCs.
Collapse
MESH Headings
- Biomarkers, Tumor/analysis
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cadherins/metabolism
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/metabolism
- Cell Line, Tumor
- Cell Survival
- Chromosome Aberrations
- Chromosomes, Human, Pair 11
- Chromosomes, Human, Pair 8
- Down-Regulation
- Epigenesis, Genetic
- Gene Amplification
- Gene Expression Regulation, Neoplastic
- Genetic Heterogeneity
- Humans
- Immunohistochemistry/methods
- Nucleic Acid Hybridization/methods
- Oligonucleotide Array Sequence Analysis/methods
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/physiology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Jorge Sergio Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Ng G, Huang J, Roberts I, Coleman N. Defining ploidy-specific thresholds in array comparative genomic hybridization to improve the sensitivity of detection of single copy alterations in cell lines. J Mol Diagn 2006; 8:449-58. [PMID: 16931585 PMCID: PMC1867620 DOI: 10.2353/jmoldx.2006.060033] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Array comparative genomic hybridization (CGH) is being widely used to screen for recurrent genomic copy number alterations in neoplasms, with imbalances typically detected through the application of gain and loss thresholds. Review of array CGH publications for the year 2005 showed that a wide range of thresholds are used. However, the effect of sample ploidy on the sensitivity of these thresholds for single copy alterations (SCAs) has not been evaluated. Here, we describe a method to evaluate the detection accuracy of thresholds for detecting SCAs in cell line array CGH data. By applying a hidden Markov model-based method, we segmented array CGH data from well-karyotyped cell lines and generated ploidy-specific sensitivity-specificity plots, from which we identified optimum thresholds relevant to sample ploidy. We demonstrate that commonly used nonploidy-specific thresholds are suboptimal in their ability to call SCAs, particularly when applied to hypertriploid or tetraploid cell lines. We conclude that the use of ploidy-specific thresholds improves the sensitivity of thres-hold-based array CGH for detecting SCAs in cell lines. Because polyploidy is a common feature of cancer cells, the application of ploidy-specific thresholds to cell lines (and potentially to clinical samples) may improve the detection sensitivity of SCAs of biological significance.
Collapse
Affiliation(s)
- Grace Ng
- Medical Research Council Cancer Cell Unit, Hutchison/MRC Research Centre, Box 197, Hills Rd., Cambridge CB2 2XZ, UK
| | | | | | | |
Collapse
|
119
|
Natrajan R, Williams RD, Hing SN, Mackay A, Reis-Filho JS, Fenwick K, Iravani M, Valgeirsson H, Grigoriadis A, Langford CF, Dovey O, Gregory SG, Weber BL, Ashworth A, Grundy PE, Pritchard-Jones K, Jones C. Array CGH profiling of favourable histology Wilms tumours reveals novel gains and losses associated with relapse. J Pathol 2006; 210:49-58. [PMID: 16823893 DOI: 10.1002/path.2021] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite the excellent survival of Wilms tumour patients treated with multimodality therapy, approximately 15% will suffer from tumour relapse, where response rates are markedly reduced. We have carried out microarray-based comparative genomic hybridisation on a series of 76 Wilms tumour samples, enriched for cases which recurred, to identify changes in DNA copy number associated with clinical outcome. Using 1Mb-spaced genome-wide BAC arrays, the most significantly different genomic changes between favourable histology tumours that did (n = 37), and did not (n = 39), subsequently relapse were gains on 1q, and novel deletions at 12q24 and 18q21. Further relapse-associated loci included losses at 1q32.1, 2q36.3-2q37.1, and gain at 13q31. 1q gains correlated strongly with loss of 1p and/or 16q. In 3 of 11 cases with concurrent 1p(-)/1q(+), a breakpoint was identified at 1p13. Multiple low-level sub-megabase gains along the length of 1q were identified using chromosome 1 tiling-path arrays. One such recurrent region at 1q22-q23.1 included candidate genes RAB25, NES, CRABP2, HDGF and NTRK1, which were screened for mRNA expression using quantitative RT-PCR. These data provide a high-resolution catalogue of genomic copy number changes in relapsing favourable histology Wilms tumours.
Collapse
MESH Headings
- Chromosome Aberrations
- Chromosome Deletion
- Chromosomes, Human, Pair 1/genetics
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 8/genetics
- DNA, Neoplasm/genetics
- Genes, Wilms Tumor/physiology
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Neoplasm Recurrence, Local/genetics
- Oligonucleotide Array Sequence Analysis/methods
- RNA, Messenger/analysis
- RNA, Neoplasm/analysis
- Treatment Outcome
- Wilms Tumor/genetics
- Wilms Tumor/pathology
Collapse
Affiliation(s)
- R Natrajan
- Paediatric Oncology, Institute of Cancer Research/Royal Marsden NHS Trust, Sutton, Surrey SM2 5NG, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Reis-Filho JS, Milanezi F, Steele D, Savage K, Simpson PT, Nesland JM, Pereira EM, Lakhani SR, Schmitt FC. Metaplastic breast carcinomas are basal-like tumours. Histopathology 2006; 49:10-21. [PMID: 16842242 DOI: 10.1111/j.1365-2559.2006.02467.x] [Citation(s) in RCA: 258] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS Recently, an immunohistochemical panel comprising antibodies against HER2, oestrogen receptor (ER), epidermal growth factor receptor (EGFR) and cytokeratin (CK) 5/6 was reported to identify basal-like breast carcinomas, as defined by cDNA microarrays. Our aim was to analyse a series of metaplastic breast carcinomas (MBCs) using this panel plus two other basal markers (CK14 and p63) and progesterone receptor (PR), to define how frequently MBCs show a basal-like immunophenotype. METHODS AND RESULTS Sixty-five cases were retrieved from the pathology archives of the authors' institutions and reviewed by three of the authors. Immunohistochemistry with antibodies for HER2, ER, EGFR, CK5/6, CK14 and p63 was performed according to standard methods. All but six cases (91%) showed the typical immunoprofile of basal-like tumours (ER- and HER2-, EGFR+ and/or CK5/6+). When CK14 and p63 were added to the panel, two additional cases could be classified as basal-like. The majority of MBCs lacked PR, except 4/19 (21%) carcinomas with squamous metaplasia. CONCLUSIONS Our results demonstrate that MBCs show a basal-like phenotype, regardless of the type of metaplastic elements. Moreover, as these neoplasms frequently overexpress EGFR (57%), patients with MBC may benefit from treatment with anti-EGFR drugs.
Collapse
Affiliation(s)
- J S Reis-Filho
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Bürger H, Kersting C, Hungermann D, Decker T, Böcker W. [The significance of "normal tissue" in the development of breast cancer: new concepts of early carcinogenesis]. DER PATHOLOGE 2006; 27:319-25. [PMID: 16896674 DOI: 10.1007/s00292-006-0857-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Only little information on the primary molecularbiological events involved in early breast is available. In particular, the definition of postulated precursor lesions of invasive breast cancer, such as ductal hyperplasia or ductal carcinoma in situ, is under an intense, controversial discussion in terms of pathogenesis and tumor biology. The most recent research on biological regulation mechanisms and genetic alterations in morphologically normally appearing breast tissue give rise for a reinterpretation for the most common progression models of breast cancer. The detection of genetic alterations within normal breast tissue in particular challenges the commonly postulated relationship between invasive and in situ breast carcinomas on the one hand, and benign, proliferative breast lesions on the other. The concerns about these relationship are further supported by the description of different cellular compartments within the normal female breast, including a "progenitor cell compartment" with different cytokeratin expression patterns, which can be transferred towards well known or suspected precursor lesions of invasive and in situ breast cancer. The aim of this manuscript is to provide an overview of the most recent results and developments in breast pathology, and to describe the consequences of our changing understanding of breast carcinogenesis.
Collapse
Affiliation(s)
- H Bürger
- Gerhard-Domagk-Institut für Pathologie, Institut für Klinische Chemie und Laboratoriumsmedizin, Westfälische Wilhelms-Universität, Domagkstrasse 17, 48149, Münster, Germany.
| | | | | | | | | |
Collapse
|
122
|
Horsley SW, Mackay A, Iravani M, Fenwick K, Valgeirsson H, Dexter T, Ashworth A, Kearney L. Array CGH of fusion gene-positive leukemia-derived cell lines reveals cryptic regions of genomic gain and loss. Genes Chromosomes Cancer 2006; 45:554-64. [PMID: 16523483 DOI: 10.1002/gcc.20317] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Human leukemia-derived cell lines containing characteristic chromosomal translocations and inversions have been instrumental in identifying fusion genes implicated in the pathogenesis of the corresponding leukemia. Although chimeric fusion genes usually provide early and essential steps in the development of leukemia, they are not in themselves sufficient, requiring additional genetic events. The nature of these secondary, cooperating genetic events is not known. The advent of genome wide microarray-based methods for assessing copy number changes made it possible to search for cytogenetically invisible regions of chromosome imbalance. We used BAC microarrays with a resolution of 1 Mb to determine whether cryptic regions of deletion or gain were associated with specific leukemia-associated fusion genes in a series of cell lines. To complement the array analysis, we also applied 24-color karyotyping by M-FISH. This revealed cryptic chromosomal translocations and regions of loss or gain in all the cell lines studied. The chromosomal origin of previously unidentified marker chromosomes was revealed. In all cases, chromosomes described as monosomic were shown to be involved in unbalanced translocations with concurrent loss and/or gain of chromosomal material. The extent of these amplified and deleted regions was more accurately defined. Finally, small regions of deletion and amplification, often including genes known to be involved in leukemia progression (for example MYC, TP53, CDKN2A, and KIT), were identified.
Collapse
Affiliation(s)
- Sharon W Horsley
- Section of Haemato-Oncology, Institute of Cancer Research, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Affiliation(s)
- Melinda F Lerwill
- James Homer Wright Pathology Laboratories of the Massachusetts General Hospital, Department of Pathology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
124
|
Reis-Filho JS, Savage K, Lambros MBK, James M, Steele D, Jones RL, Dowsett M. Cyclin D1 protein overexpression and CCND1 amplification in breast carcinomas: an immunohistochemical and chromogenic in situ hybridisation analysis. Mod Pathol 2006; 19:999-1009. [PMID: 16648863 DOI: 10.1038/modpathol.3800621] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Conflicting results on the prevalence of cyclin D1 ovexpression and its correlation with CCND1 amplification and outcome of breast cancer patients have been reported. Owing to limited sensitivity and specificity of most antibodies against cyclin D1, evaluation of cyclin D1 immunoexpression is reported to be problematic. The aims of this study were to assess the prevalence of cyclin D1 expression in breast carcinomas using the SP4 rabbit monoclonal antibody; to correlate cyclin D1 expression with amplification, assessed using chromogenic in situ hybridisation (CISH); and to analyse the relationship between CCND1 amplification and overexpression with clinicopathological parameters and outcome in a tissue microarray containing replicate tumour samples from 245 breast cancer patients. Immunohistochemistry for cyclin D1 was performed using the SP4 and the results were scored according to the Allred scoring system. CISH was carried out using the Zymed CCND1 SpotLight probe. CISH signals were counted in 60 morphologically unequivocal neoplastic cells. Amplification was defined as >5 signals per nucleus in more than 50% of cancer cells, or when large gene copy clusters were seen. Strong cyclin D1 expression and CCND1 amplification were found in 67.4 and 14.5% of the cases, respectively. A strong correlation between cyclin D1 overexpression and CCND1 amplification was demonstrated (P<0.0001). Cyclin D1 expression showed a positive correlation with hormone receptor expression (both ER and PgR, P<0.0001). An inverse correlation was observed between an immunohistochemical panel of 'basal-like' markers and both cyclin D1 overexpression (P<0.0001) and CCND1 amplification (P<0.0001). On univariate analysis cyclin D1 expression showed a correlation with longer overall survival (OS). Neither cyclin D1 nor CCND1 were independent prognostic factors for disease-free survival or OS. The results of this study confirm the association between cyclin D1 overexpression and positivity for hormone receptors and the lack of CCND1 amplification in basal-like breast carcinomas.
Collapse
Affiliation(s)
- Jorge S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK.
| | | | | | | | | | | | | |
Collapse
|
125
|
Banerjee S, Reis-Filho JS, Ashley S, Steele D, Ashworth A, Lakhani SR, Smith IE. Basal-like breast carcinomas: clinical outcome and response to chemotherapy. J Clin Pathol 2006; 59:729-35. [PMID: 16556664 PMCID: PMC1860434 DOI: 10.1136/jcp.2005.033043] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2005] [Indexed: 02/07/2023]
Abstract
BACKGROUND Grade-III invasive ductal carcinomas of no special type (IDCs-NST) constitute a heterogeneous group of tumours with different clinical behaviour and response to chemotherapy. As many as 25% of all grade-III IDCs-NST are known to harbour a basal-like phenotype, as defined by gene expression profiling or immunohistochemistry for basal cytokeratins. Patients with basal-like breast carcinomas (BLBC) are reported to have a shorter disease-free and overall survival. MATERIAL AND METHODS A retrospective analysis of 49 patients with BLBC (as defined by basal cytokeratin expression) and 49 controls matched for age, nodal status and grade was carried out. Histological features, immunohistochemical findings for oestrogen receptor (ER), progesterone receptor (PgR) and HER2, and clinical outcome and survival after adjuvant chemotherapy were compared between the two groups. RESULTS It was more likely for patients with BLBCs to be found negative for ER (p<0.0001), PgR (p<0.0001) and HER2 (p<0.01) than controls. Patients with BLBCs were found to have a significantly higher recurrence rate (p<0.05) and were associated with significantly shorter disease-free and overall survival (both p<0.05). In the group of patients who received anthracycline-based adjuvant chemotherapy (BLBC group, n = 47; controls, n = 49), both disease-free and overall survival were found to be significantly shorter in the BLBC group (p<0.05). CONCLUSIONS BLBCs are a distinct clinical and pathological entity, characterised by high nuclear grade, lack of hormone receptors and HER2 expression and a more aggressive clinical course. Standard adjuvant chemotherapy seems to be less effective in these tumours and new therapeutic approaches are indicated.
Collapse
MESH Headings
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/secondary
- Chemotherapy, Adjuvant
- Female
- Humans
- Keratins/metabolism
- Middle Aged
- Neoplasm Proteins/metabolism
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Survival Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- S Banerjee
- Breast Unit, Royal Marsden Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
126
|
Abstract
The introduction of comparative genomic hybridization (CGH) in 1992 opened new avenues in genomic investigation; in particular, it advanced analysis of solid tumours, including breast cancer, because it obviated the need to culture cells before their chromosomes could be analyzed. The current generation of CGH analysis uses ordered arrays of genomic DNA sequences and is therefore referred to as array-CGH or matrix-CGH. It was introduced in 1998, and further increased the potential of CGH to provide insight into the fundamental processes of chromosomal instability and cancer. This review provides a critical evaluation of the data published on array-CGH and breast cancer, and discusses some of its expected future value and developments.
Collapse
Affiliation(s)
- Erik H van Beers
- Division of Experimental Therapy, Netherlands Cancer Institute NKI-AVL, Amsterdam
| | - Petra M Nederlof
- Department of Pathology, Netherlands Cancer Institute NKI-AVL, Amsterdam, The Netherlands
| |
Collapse
|
127
|
Moriya T, Kasajima A, Ishida K, Kariya Y, Akahira JI, Endoh M, Watanabe M, Sasano H. New trends of immunohistochemistry for making differential diagnosis of breast lesions. Med Mol Morphol 2006; 39:8-13. [PMID: 16575508 DOI: 10.1007/s00795-006-0309-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Accepted: 01/13/2006] [Indexed: 11/24/2022]
Abstract
Immunohistochemistry is widely used for pathological diagnosis of breast lesions. Other than hormone receptors and HER2/neu analysis for primary breast carcinomas, several markers may be useful for differential diagnoses, although in limited situations. To decide the malignant potential of intraductal proliferative lesions, analysis for the staining pattern of cytokeratins may be a good reference. Most ductal carcinoma in situ cases are diffusely positive for luminal cell markers (CK8, CK18, CK19), but negative for basal cell markers (CK5/6 and CK14). However, usual ductal hyperplasia may show the mosaic staining patterns for any of these markers, which may indicate a heterogeneous cell population in benign lesions. Myoepithelial markers (alpha-SMA, myosin, calponin, p63, CD10) are almost consistently positive for benign papillomas but they do not completely distinguish intraductal papillary carcinomas. Preservation of myoepithelial layer is the diagnostic key when looking at benign sclerosing lesions, including carcinoma with pseudoinvasive structures. E-cadherin is mostly positive for ductal carcinomas but negative for lobular carcinomas. Some of the lobular carcinomas are positive for 34betaE12, but they are consistently negative for CK5/6. Comparison with histopathological findings of hematoxylin and eosin is essential to make proper diagnosis in the individual case.
Collapse
Affiliation(s)
- Takuya Moriya
- Department of Pathology, Tohoku University Hospital, Aoba-ku, Sendai, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Morandi L, Marucci G, Foschini MP, Cattani MG, Pession A, Riva C, Eusebi V. Genetic similarities and differences between lobular in situ neoplasia (LN) and invasive lobular carcinoma of the breast. Virchows Arch 2006; 449:14-23. [PMID: 16612623 DOI: 10.1007/s00428-006-0192-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 02/11/2006] [Indexed: 11/25/2022]
Abstract
One of the most controversial issues in breast pathology is whether lobular neoplasia (LN) is a risk factor or a precursor lesion of invasive lobular carcinoma (ILC). This is consequent to the fact that no conclusive data on the biology of LN exist. Molecular studies of LN and ILC are scanty, variable, and not consistent. Clonality of 12 cases of LN and ILC present simultaneously in the same block has been studied. Cells from both lesions were obtained by microdissection and were studied for mitochondrial DNA (mtDNA), D-loop sequencing, and neighbor-joining trees. Eight of the same cases were studied with comparative genomic hybridization (CGH) array to have additional data consistent with mtDNA. In all cases, loss of heterozygosity was studied for D16S496,locus 16q22.1 related to e-cadherin. It appears that no fewer than eight cases were genetically very similar (clonal) with mtDNA. Seven of these cases appeared also clonal with CGH array. It is concluded that in the present series, LN and ILC are genetically related lesions in the majority of cases and that LN might be the precursor of ILC.
Collapse
Affiliation(s)
- Luca Morandi
- Section of Pathology, Department of Oncology, University of Bologna, Ospedale Bellaria, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
129
|
Lambros MBK, Simpson PT, Jones C, Natrajan R, Westbury C, Steele D, Savage K, Mackay A, Schmitt FC, Ashworth A, Reis-Filho JS. Unlocking pathology archives for molecular genetic studies: a reliable method to generate probes for chromogenic and fluorescent in situ hybridization. J Transl Med 2006; 86:398-408. [PMID: 16446704 DOI: 10.1038/labinvest.3700390] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Chromogenic (CISH) and fluorescent (FISH) in situ hybridization have emerged as reliable techniques to identify amplifications and chromosomal translocations. CISH provides a spatial distribution of gene copy number changes in tumour tissue and allows a direct correlation between copy number changes and the morphological features of neoplastic cells. However, the limited number of commercially available gene probes has hindered the use of this technique. We have devised a protocol to generate probes for CISH that can be applied to formalin-fixed, paraffin-embedded tissue sections (FFPETS). Bacterial artificial chromosomes (BACs) containing fragments of human DNA which map to specific genomic regions of interest are amplified with phi29 polymerase and random primer labelled with biotin. The genomic location of these can be readily confirmed by BAC end pair sequencing and FISH mapping on normal lymphocyte metaphase spreads. To demonstrate the reliability of the probes generated with this protocol, four strategies were employed: (i) probes mapping to cyclin D1 (CCND1) were generated and their performance was compared with that of a commercially available probe for the same gene in a series of 10 FFPETS of breast cancer samples of which five harboured CCND1 amplification; (ii) probes targeting cyclin-dependent kinase 4 were used to validate an amplification identified by microarray-based comparative genomic hybridization (aCGH) in a pleomorphic adenoma; (iii) probes targeting fibroblast growth factor receptor 1 and CCND1 were used to validate amplifications mapping to these regions, as defined by aCGH, in an invasive lobular breast carcinoma with FISH and CISH; and (iv) gene-specific probes for ETV6 and NTRK3 were used to demonstrate the presence of t(12;15)(p12;q25) translocation in a case of breast secretory carcinoma with dual colour FISH. In summary, this protocol enables the generation of probes mapping to any gene of interest that can be applied to FFPETS, allowing correlation of morphological features with gene copy number.
Collapse
MESH Headings
- Adenoma, Pleomorphic/genetics
- Adenoma, Pleomorphic/pathology
- Biological Specimen Banks
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/pathology
- Chromogenic Compounds
- Chromosomes, Artificial, Bacterial
- DNA Probes/biosynthesis
- Female
- Humans
- In Situ Hybridization/methods
- Oligonucleotide Array Sequence Analysis
- Paraffin Embedding
- Pathology/methods
- Salivary Gland Neoplasms/genetics
- Salivary Gland Neoplasms/pathology
- Sensitivity and Specificity
- Tissue Fixation
- Translocation, Genetic
Collapse
Affiliation(s)
- Maryou B K Lambros
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Di Palma S, Lambros MBK, Savage K, Jones C, Mackay A, Dexter T, Iravani M, Fenwick K, Ashworth A, Reis-Filho JS. Oncocytic change in pleomorphic adenoma: molecular evidence in support of an origin in neoplastic cells. J Clin Pathol 2006; 60:492-9. [PMID: 16467165 PMCID: PMC1994546 DOI: 10.1136/jcp.2005.031369] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Cells with oncocytic change (OC) are a common finding in salivary glands (SGs) and in SG tumours. When found within pleomorphic adenomas (PAs), cells with OC may be perceived as evidence of malignancy, and lead to a misdiagnosis of carcinoma ex pleomorphic adenoma (CaExPa). AIM To describe a case of PA with atypical OC, resembling a CaExPa. A genomewide molecular analysis was carried out to compare the molecular genetic features of the two components and to determine whether the oncocytic cells originated from PA cells, entrapped normal cells, or whether these cells constitute an independent tumour. MATERIALS AND METHODS Representative blocks were immunohistochemically analysed with antibodies raised against cytokeratin (Ck) 5/6, Ck8/18, Ck14, vimentin, p63, alpha-smooth muscle actin (ASMA), S100 protein, anti-mitochondria antibody, beta-catenin, HER2, Ki67, p53 and epidermal growth factor receptor. Typical areas of PA and OC were microdissected and subjected to microarray-based comparative genomic hybridisation (aCGH). Chromogenic in situ hybridisation (CISH) was performed with in-house generated probes to validate the aCGH findings. RESULTS PA cells showed the typical immunohistochemical profile, including positivity for Ck5/6, Ck8/18, Ck14, vimentin, ASMA, S100 protein, p63, epidermal growth factor receptor and beta-catenin, whereas oncocytic cells showed a luminal phenotype, expression of anti-mitochondria antibody and reduced beta-catenin staining. Both components showed low proliferation rates and lacked p53 reactivity. aCGH revealed a similar amplification in both components, mapping to 12q13.3-q21.1, which was further validated by CISH. No HER2 gene amplification or overexpression was observed. The foci of oncocytic metaplasia showed an additional low-level gain of 6p25.2-p21.31. CONCLUSION The present data demonstrate that the bizarre atypical cells of the present case show evidence of clonality but no features of malignancy. In addition, owing to the presence of a similar genome amplification pattern in both components, it is proposed that at least in some cases, OC may originate from PA cells.
Collapse
Affiliation(s)
- Silvana Di Palma
- Royal Surrey County Hospital, University of Surrey, Guildford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Natrajan R, Little SE, Sodha N, Reis-Filho JS, Mackay A, Fenwick K, Ashworth A, Perlman EJ, Dome JS, Grundy PE, Pritchard-Jones K, Jones C. Analysis by array CGH of genomic changes associated with the progression or relapse of Wilms' tumour. J Pathol 2006; 211:52-9. [PMID: 17103382 DOI: 10.1002/path.2087] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Despite aggressive salvage regimens, approximately half of all children who suffer a Wilms' tumour recurrence will die of their disease. Although there are increasing data on molecular genetic prognostic factors present in the tumour at diagnosis, there is little information regarding the molecular events that occur with Wilms' tumour progression and relapse. In the present study, microarray-based comparative genomic hybridization (aCGH) analysis has been carried out on 58 Wilms' tumour samples, which included 38 untreated primary and 20 recurrent tumours. A higher degree of copy number changes was observed in the recurrent tumours (33.0% genomic clones) than in the primary tumour (21.2%). Paired analysis highlighted the acquisition of 15q gain with high levels of IGF1R expression in the tumour recurrence in two cases. The most statistically significant abnormality acquired between diagnosis and relapse was loss of 17p. One case that experienced 17p loss was classified as favourable histology at diagnosis, but exhibited diffuse anaplasia at recurrence and had a homozygous TP53 deletion. Another instructive case with a constitutional 11p13 deletion presented with bilateral tumours and suffered two subsequent recurrences in the left kidney. A somatic WT1 mutation was found only in the right kidney tumour, while the constitutional 11p13 deletion was the only abnormality detected in the initial left kidney tumour by aCGH. The two subsequent relapses in the left kidney contained an accumulation of additional genetic alterations, including an independent WT1 mutation.
Collapse
Affiliation(s)
- R Natrajan
- Paediatric Oncology, Institute of Cancer Research/Royal Marsden NHS Trust, Sutton, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Reis-Filho JS, Pinheiro C, Lambros MBK, Milanezi F, Carvalho S, Savage K, Simpson PT, Jones C, Swift S, Mackay A, Reis RM, Hornick JL, Pereira EM, Baltazar F, Fletcher CDM, Ashworth A, Lakhani SR, Schmitt FC. EGFR amplification and lack of activating mutations in metaplastic breast carcinomas. J Pathol 2006; 209:445-53. [PMID: 16739104 DOI: 10.1002/path.2004] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Metaplastic breast carcinomas are reported to harbour epidermal growth factor receptor (EGFR) overexpression in up to 80% of the cases, but EGFR gene amplification is the underlying genetic mechanism in around one-third of these. In this study, EGFR gene amplification as defined by chromogenic in situ hybridization and protein overexpression was examined in a cohort of 47 metaplastic breast carcinomas. Furthermore, the presence of activating EGFR mutations in exons 18, 19, 20, and 21 was investigated. Thirty-two cases showed EGFR overexpression and of these, 11 (34%) harboured EGFR gene amplification. In addition, EGFR amplification showed a statistically significant association with EGFR overexpression (p < 0.0094) and was restricted to carcinomas with homologous metaplasia. Ten cases, five with and five without EGFR amplification, were subjected to microarray-based CGH, which demonstrated that EGFR copy number gain may occur by amplification of a discrete genomic region or by gains of the short arm of chromosome 7 with a breakpoint near the EGFR gene locus, the minimal region of amplification mapping to EGFR, LANCL2, and SEC61G. No activating EGFR mutations were identified, suggesting that this is unlikely to be a common alternative underlying genetic mechanism for EGFR expression in metaplastic breast carcinomas. Given that metaplastic breast carcinomas are resistant to conventional chemotherapy or hormone therapy regimens and that tumours with EGFR amplification are reported to be sensitive to EGFR tyrosine kinase inhibitors, these findings indicate that further studies are warranted to explore EGFR tyrosine kinase inhibitors as potential therapeutic agents for metaplastic breast carcinomas harbouring amplification of 7p11.2.
Collapse
Affiliation(s)
- J S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Espié M, Hocini H, Cuvier C, Giacchetti S, Bourstyn E, de Roquancourt A. [Breast lobular carcinoma in situ: diagnosis and evolution]. ACTA ACUST UNITED AC 2005; 33:964-9. [PMID: 16324870 DOI: 10.1016/j.gyobfe.2005.10.022] [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: 08/11/2005] [Accepted: 10/19/2005] [Indexed: 10/25/2022]
Abstract
The incidence of lobular cancers in situ is increasing, especially in post-menopausal women. Whereas this form of disease was regarded for a long time as nothing but a risk factor of the occurrence of later infiltrating carcinoma, it now tends to represent a precancerous state whose progression to subsequent infiltrating carcinoma does not inevitably occur. The clinical and radiological diagnosis remains difficult and the choice of therapies varies according to teams, ranging from mere surveillance to mastectomy.
Collapse
Affiliation(s)
- M Espié
- Centre des maladies du sein, hôpital Saint-Louis, Paris, France.
| | | | | | | | | | | |
Collapse
|
134
|
Reis-Filho JS, Milanezi F, Carvalho S, Simpson PT, Steele D, Savage K, Lambros MBK, Pereira EM, Nesland JM, Lakhani SR, Schmitt FC. Metaplastic breast carcinomas exhibit EGFR, but not HER2, gene amplification and overexpression: immunohistochemical and chromogenic in situ hybridization analysis. Breast Cancer Res 2005; 7:R1028-35. [PMID: 16280056 PMCID: PMC1410747 DOI: 10.1186/bcr1341] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2005] [Revised: 09/12/2005] [Accepted: 09/29/2005] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTION Metaplastic breast carcinomas constitute a heterogeneous group of neoplasms, accounting for less than 1% of all invasive mammary carcinomas. Approximately 70-80% of metaplastic breast carcinomas overexpress the epidermal growth factor receptor (EGFR). Human epidermal growth factor receptor (HER)2 and EGFR have attracted much attention in the medical literature over the past few years owing to the fact that humanized monoclonal antibodies against HER2 and therapies directed against the extracellular ligand-binding domain or the intracellular tyrosine kinase domain of EGFR have proven successful in treating certain types of human cancer. We investigated whether HER2 and EGFR overexpression was present and evaluated gene amplification in a series of metaplastic breast carcinomas. METHOD Twenty-five metaplastic breast carcinomas were immunohistochemically analyzed using a monoclonal antibody (31G7) for EGFR and two antibodies for HER2 (Herceptest and CB11) and scored using the Herceptest scoring system. Gene amplification was evaluated by chromogenic in situ hybridization using Zymed Spot-Light EGFR and HER2 amplification probe. The results were evaluated by bright field microscopy under 40x and 63x objective lenses. RESULTS Nineteen (76%) metaplastic breast carcinomas exhibited EGFR ovexpression, and among these EGFR amplification (defined either by large gene clusters or >5 signals/nucleus in >50% of neoplastic cells) was detected in seven cases (37%): three carcinomas with squamous differentiation and four spindle cell carcinomas. One case exhibited HER2 overexpression of grade 2+ (>10% of cells with weak to moderate complete membrane staining), but HER2 gene amplification was not detected. CONCLUSION Metaplastic breast carcinomas frequently overexpressed EGFR, which was associated with EGFR gene amplification in one-third of cases. Our findings suggest that some patients with metaplastic breast carcinomas might benefit from novel therapies targeting EGFR. Because most metaplastic breast carcinomas overexpress EGFR without gene amplification, further studies to evaluate EGFR activating mutations are warranted.
Collapse
Affiliation(s)
- Jorge S Reis-Filho
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
- IPATIMUP – Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - Fernanda Milanezi
- IPATIMUP – Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - Silvia Carvalho
- IPATIMUP – Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | - Pete T Simpson
- Molecular & Cellular Pathology, Mayne Medical School, University of Queensland, Queensland Institute of Medical Research and Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Dawn Steele
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | - Kay Savage
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | - Maryou BK Lambros
- The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK
| | | | - Jahn M Nesland
- The Norwegian Radium Hospital, University of Oslo, Montebello, Norway
| | - Sunil R Lakhani
- Molecular & Cellular Pathology, Mayne Medical School, University of Queensland, Queensland Institute of Medical Research and Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Fernando C Schmitt
- IPATIMUP – Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| |
Collapse
|