Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter T. Simpson is active.

Publication


Featured researches published by Peter T. Simpson.


The Journal of Pathology | 2005

Molecular evolution of breast cancer.

Peter T. Simpson; Jorge S. Reis-Filho; Theodora Gale; Sunil R. Lakhani

Molecular analysis of invasive breast cancer and its precursors has furthered our understanding of breast cancer progression. In the past few years, new multi‐step pathways of breast cancer progression have been delineated through genotypic–phenotypic correlations. Nuclear grade, more than any other pathological feature, is strongly associated with the number and pattern of molecular genetic abnormalities in breast cancer cells. Thus, there are two distinct major pathways to the evolution of low‐ and high‐grade invasive carcinomas: whilst the former consistently show oestrogen receptor (ER) and progesterone receptor (PgR) positivity and 16q loss, the latter are usually ER/PgR‐negative and show Her‐2 overexpression/amplification and complex karyotypes. The boundaries between the evolutionary pathways of well‐differentiated/low‐grade ductal and lobular carcinomas have been blurred, with changes in E‐cadherin expression being one of the few distinguishing features between the two. In addition, lesions long thought to be precursors of breast carcinomas, such as hyperplasia of usual type, are currently considered mere risk indicators, whilst columnar cell lesions are now implicated as non‐obligate precursors of atypical ductal hyperplasia (ADH) and well‐differentiated ductal carcinoma in situ (DCIS). However, only through the combination of comprehensive morphological analysis and cutting‐edge molecular tools can this knowledge be translated into clinical practice and patient management. Copyright


Histopathology | 2006

Metaplastic breast carcinomas are basal‐like tumours

Jorge S. Reis-Filho; Fernanda Milanezi; Dawn Steele; Kay Savage; Peter T. Simpson; J. M. Nesland; Emílio Marcelo Pereira; Sunil R. Lakhani; Fernando Schmitt

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.


The American Journal of Surgical Pathology | 2005

Columnar cell lesions of the breast: The missing link in breast cancer progression? A morphological and molecular analysis.

Peter T. Simpson; Theo Gale; Jorge S. Reis-Filho; Chris Jones; Suzanne Parry; John P. Sloane; Andrew M. Hanby; Sarah Pinder; Andrew H S Lee; S Humphreys; Ian O. Ellis; Sunil R. Lakhani

Columnar cell lesions (CCLs) of the breast are a spectrum of lesions that have posed difficulties to pathologists for many years, prompting discussion concerning their biologic and clinical significance. We present a study of CCL in context with hyperplasia of usual type (HUT) and the more advanced lesions ductal carcinoma in situ (DCIS) and invasive ductal carcinoma. A total of 81 lesions from 18 patients were subjected to a comprehensive morphologic review based upon a modified version of Schnitts classification system for CCL, immunophenotypic analysis (estrogen receptor [ER], progesterone receptor [PgR], Her2/neu, cytokeratin 5/6 [CK5/6], cytokeratin 14 [CK14], E-cadherin, p53) and for the first time, a whole genome molecular analysis by comparative genomic hybridization. Multiple CCLs from 3 patients were studied in particular detail, with topographic information and/or showing a morphologic spectrum of CCL within individual terminal duct lobular units. CCLs were ER and PgR positive, CK5/6 and CK14 negative, exhibit low numbers of genetic alterations and recurrent 16q loss, features that are similar to those of low grade in situ and invasive carcinoma. The molecular genetic profiles closely reflect the degree of proliferation and atypia in CCL, indicating some of these lesions represent both a morphologic and molecular continuum. In addition, overlapping chromosomal alterations between CCL and more advanced lesions within individual terminal duct lobular units suggest a commonality in molecular evolution. These data further support the hypothesis that CCLs are a nonobligate, intermediary step in the development of some forms of low grade in situ and invasive carcinoma.


Clinical Cancer Research | 2006

FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas

Jorge S. Reis-Filho; Peter T. Simpson; Nicholas C. Turner; Maryou Ballo Lambros; Chris Jones; Alan Mackay; Anita Grigoriadis; David Sarrió; Kay Savage; Tim Dexter; Marjan Iravani; Kerry Fenwick; Barbara L. Weber; David Hardisson; Fernando Schmitt; José Palacios; Sunil R. Lakhani; Alan Ashworth

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.


Virchows Archiv | 2003

Distribution of p63, cytokeratins 5/6 and cytokeratin 14 in 51 normal and 400 neoplastic human tissue samples using TARP-4 multi-tumor tissue microarray.

Jorge S. Reis-Filho; Peter T. Simpson; Albino Martins; Ana Preto; Fátima Gärtner; Fernando Schmitt

Abstractp63, cytokeratin (CK) 5/6 and CK 14 have been employed in diagnostic pathology as markers of basal, squamous and myoepithelial differentiation in several types of human neoplasms; however, there is scant data on the concurrent expression of these markers in large series of human neoplasms. We analyzed the distribution of these three immunohistochemical markers in 51 normal human tissue samples, 350 carcinomas, 25 malignant melanomas (MMs), and 25 glioblastomas using three serial sections of tissue array research program (TARP)-4 multi-tumor tissue microarray. Also, we performed double immunostainings to characterize the differential distribution of p63/CK 5/6 and p63/CK 14 in normal breast, salivary gland and skin. p63, CK 5/6 and CK 14 were expressed in basal cells of the prostate and respiratory epithelia and in breast and bronchial myoepithelial cells. p63 was also expressed in cytotrophoblast cells of human placenta and in scattered cells of lymph node germinal center. CK 5/6 and CK 14 also stained the cytoplasm of basal cells of esophageal stratified squamous epithelium and transitional epithelial cells of the bladder. No mesenchymal, neural, endothelial, smooth muscle or adipose cells were stained by any of the markers. p63, CK 5/6, and CK 14 were respectively expressed in 92.6%, 75.0%, and 52.9% of the squamous cell carcinomas of the lung, 10.2%, 20.0%, and 7.4% of the ductal carcinomas of the breast, 12.9%, 34.4%, and 11.8% of the serous and 25.0%, 0%, and 0% of the endometrioid carcinomas of the ovary. Lung, prostate and colonic adenocarcinomas, as well as MMs and glioblastomas were only rarely decorated by one of the markers. Only matched samples of 16 squamous cell carcinomas and two ductal carcinomas of the breast co-expressed these three markers. In double immunostainings, p63-CK 5/6, as well as p63-CK 14 were co-expressed by basal/myoepithelial cells of the salivary glands and basal cells of the epidermis. Our results demonstrate that p63, CK 5/6 and CK 14 may be used together in immunohistochemical panels to characterize squamous differentiation in poorly differentiated carcinomas or carcinomas of unknown origin.


The Journal of Pathology | 2006

EGFR amplification and lack of activating mutations in metaplastic breast carcinomas

Jorge S. Reis-Filho; Céline Pinheiro; Mb Lambros; Fernanda Milanezi; Sílvia Carvalho; Kay Savage; Peter T. Simpson; Chris Jones; Sally Swift; Alan Mackay; Rui M. Reis; Jason L. Hornick; Emílio Marcelo Pereira; Fátima Baltazar; Christopher D. M. Fletcher; Alan Ashworth; Sunil R. Lakhani; Fernando Schmitt

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. Copyright


The Journal of Pathology | 2005

Pleomorphic lobular carcinoma of the breast: role of comprehensive molecular pathology in characterization of an entity

Jorge S. Reis-Filho; Peter T. Simpson; Chris Jones; Dawn Steele; Alan Mackay; Marjan Iravani; Kerry Fenwick; Haukur Valgeirsson; Maryou B. Lambros; Alan Ashworth; José Palacios; Fernando Schmitt; Sunil R. Lakhani

Immunohistochemical analysis of E‐cadherin has changed the way lobular neoplasia is perceived. It has helped to classify difficult cases of carcinoma in situ with indeterminate features and led to the identification of new variants of lobular carcinoma. Pleomorphic lobular carcinoma (PLC) and pleomorphic lobular carcinoma in situ (PLCIS), recently described variants of invasive and in situ classic lobular carcinoma, are reported to be associated with more aggressive clinical behaviour. Although PLC/PLCIS show morphological features of classic lobular neoplasia and lack E‐cadherin expression, it is still unclear whether these lesions evolve through the same genetic pathway as lobular carcinomas or are high‐grade ductal neoplasms that have lost E‐cadherin. Here we have analysed a case of extensive PLCIS and invasive PLC associated with areas of E‐cadherin‐negative carcinoma in situ with indeterminate features, using immunohistochemistry, chromogenic in situ hybridization, high‐resolution comparative genomic hybridization (CGH) and array‐based CGH. We observed that all lesions lacked E‐cadherin and β‐catenin and showed gain of 1q and loss of 16q, features that are typical of lobular carcinomas but are not seen in high‐grade ductal lesions. In addition, amplifications of c‐myc and HER2 were detected in the pleomorphic components, which may account for the high‐grade features in this case and the reported aggressive clinical behaviour of these lesions. Taken together, these data suggest that at least some PLCs may evolve from the same precursor or through the same genetic pathway as classic lobular carcinomas. Copyright


Oncogene | 2003

cDNA microarray analysis of genes associated with ERBB2 (HER2/ neu ) overexpression in human mammary luminal epithelial cells

Alan Mackay; Chris Jones; Tim Dexter; Ricardo L.A. Silva; Karen Bulmer; Allison Jones; Peter T. Simpson; Robert A. Harris; Parmjit S. Jat; A. Munro Neville; Luiz F. L. Reis; Sunil R. Lakhani; Michael J. O'Hare

To investigate changes in gene expression associated with ERBB2, expression profiling of immortalized human mammary luminal epithelial cells and variants expressing a moderate and high level of ERBB2 has been carried out using cDNA microarrays corresponding to approximately 6000 unique genes/ESTs. A total of 61 significantly up- or downregulated (2.0-fold) genes were identified and further validated by RT–PCR analysis as well as microarray comparisons with a spontaneously ERBB2- overexpressing breast cancer cell line and ERBB2-positive primary breast tumors. The expression and clinical relevance of proteins predicted to be associated with ERBB2 overexpression in breast cancers were analysed together with their clinical relevance by antibody screening using a tissue array. Differentially regulated genes include those involved in cell–matrix interactions including proline 4-hydroxylase (P4HA2), galectin 1 (LGALS1) and galectin 3 (LGALS3), fibronectin 1 (FN1) and p-cadherin (CDH3), and cell proliferation (CRIP1, IGFBP3) and transformation (S100P, S100A4). A number of genes associated with MYC signalling were also differentially expressed, including NDRG1, USF2 and the epithelial membrane proteins 1 and 3 (EMP1, EMP3). These data represent profiles of the transcriptional changes associated with ERBB2-related pathways in the breast, and identify novel and potentially useful targets for prognosis and therapy.


Breast Cancer Research | 2005

Metaplastic breast carcinomas exhibit EGFR, but not HER2, gene amplification and overexpression: immunohistochemical and chromogenic in situ hybridization analysis

Jorge S. Reis-Filho; Fernanda Milanezi; Sílvia Carvalho; Peter T. Simpson; Dawn Steele; Kay Savage; Maryou B. Lambros; Emílio Marcelo Pereira; J. M. Nesland; Sunil R. Lakhani; Fernando Schmitt

IntroductionMetaplastic 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.MethodTwenty-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 40× and 63× objective lenses.ResultsNineteen (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.ConclusionMetaplastic 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.


The Journal of Pathology | 2008

Molecular profiling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas

Peter T. Simpson; Jorge S. Reis-Filho; Mb Lambros; Chris Jones; Dawn Steele; Alan Mackay; Marjan Iravani; Kerry Fenwick; Tim Dexter; A. Jones; Lynne Reid; L. Da Silva; Sandra J. Shin; David Hardisson; Alan Ashworth; Fernando Schmitt; José Palacios; Sunil R. Lakhani

Pleomorphic lobular carcinomas (PLC) of the breast display histological features associated with classic invasive lobular carcinoma (ILC), yet they also exhibit more conspicuous nuclear atypia and pleomorphism, and an aggressive clinical behaviour. From a breast cancer progression perspective, it is unclear whether PLC is a variant of ILC or is a high‐grade invasive ductal carcinoma (IDC) that has lost E‐cadherin. The molecular features of 26 PLC were studied using immunohistochemistry [oestrogen receptor (ER), progesterone receptor (PR), HER2, p53 and E‐cadherin], 0.9 Mb resolution, microarray‐based comparative genomic hybridization (aCGH), fluorescent (FISH) and chromogenic (CISH) in situ hybridization and loss of heterozygosity. Comparative analysis was performed with aCGH data from PLC with classic ILC (16 cases) and high grade IDC (35 cases). PLCs were frequently ER‐ and PR‐positive, E‐cadherin‐negative and occasionally HER2‐ and p53‐positive. Recurrent copy number changes identified by aCGH included gains on 1q, 8q, 11q, 12q, 16p and 17q and losses on 8p, 11q, 13q, 16q and Xq. Highly recurrent 1q+ (100% of cases), 16p+ (93%), 11q− (53%) and 16q− (93%) and evidence of the der(1;16)/der(16)t(1;16) rearrangement, as detected by FISH, suggested that PLC had a ‘lobular genotype’. Focal amplifications were evident at 8p12‐p11, 8q24, 11q13.1‐q14.1, 12q14, 17q12 and 20q13. Loss of BRCA2 was detected in 40% of PLC by LOH. Comparative analysis of aCGH data suggested the molecular features of PLC (ER/PR‐positive, E‐cadherin‐negative, 1q+, 11q−, 16p+ and 16q−) were more closely related to those of ILC than IDC, implicating an overlapping developmental pathway for these lobular tumour types. Molecular alterations found in PLC that are more typical of high‐grade IDC than ILC (p53 and HER2 positivity, 8q+, 17q24‐q25+, 13q− and amplification of 8q24, 12q14, 17q12 and 20q13) are likely to drive the high‐grade and more aggressive biology of PLC. Copyright

Collaboration


Dive into the Peter T. Simpson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lynne Reid

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kum Kum Khanna

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Georgia Chenevix-Trench

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jodi M. Saunus

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Fares Al-Ejeh

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge