1
|
Raspin K, O'Malley DE, Marthick JR, Donovan S, Malley RC, Banks A, Redwig F, Skala M, Dickinson JL, FitzGerald LM. Analysis of a large prostate cancer family identifies novel and recurrent gene fusion events providing evidence for inherited predisposition. Prostate 2022; 82:540-550. [PMID: 34994974 DOI: 10.1002/pros.24300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/27/2021] [Accepted: 12/21/2021] [Indexed: 11/12/2022]
Abstract
There is strong interest in the characterisation of gene fusions and their use to enhance clinical practices in prostate cancer (PrCa). Significantly, ~50% of prostate tumours harbour a gene fusion. Inherited factors are thought to predispose to these events but, to date, only one study has investigated gene fusions in a familial context. Here, we examined the prevalence and diversity of gene fusions in 14 tumours from a single large PrCa family, PcTas9, using the TruSight® RNA Fusion Panel and Sanger sequencing validation. These fusions were then explored in The Cancer Genome Atlas (TCGA) PrCa data set (n = 494). Overall, 64.3% of PcTas9 tumours harboured a gene fusion, including known erythroblast transformation-specific (ETS) fusions involving ERG and ETV1, and two novel gene fusions, C19orf48:ETV4 and RYBP:FOXP1. Although 3' ETS genes were overexpressed in PcTas9 and TCGA tumour samples, 3' fusion of FOXP1 did not appear to alter its expression. In addition, PcTas9 fusion carriers were more likely to have lower-grade disease than noncarriers (p = 0.02). Likewise, TCGA tumours with high-grade disease were less likely to harbour fusions (p = 0.03). Our study further implicates an inherited predisposition to PrCa gene fusion events, which are associated with less aggressive tumours. This knowledge could lead to clinical strategies to predict men at risk for fusion-positive PrCa and, thus, identify patients who are more or less at risk of aggressive disease and/or responsive to particular therapies.
Collapse
Affiliation(s)
- Kelsie Raspin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Dannielle E O'Malley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - James R Marthick
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | | | - Roslyn C Malley
- Hobart Pathology, Hobart, TAS, Australia
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Annette Banks
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Frank Redwig
- Department of Urology, Royal Hobart Hospital, Hobart, TAS, Australia
| | - Marketa Skala
- WP Holman Clinic, Royal Hobart Hospital, Hobart, TAS, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Liesel M FitzGerald
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
2
|
Abdelsalam RA, Khalifeh I, Box A, Kalantarian M, Ghosh S, Abou-Ouf H, Lotfi T, Shahait M, Palanisamy N, Bismar TA. Molecular characterization of prostate cancer in Middle Eastern population highlights differences with Western populations with prognostic implication. J Cancer Res Clin Oncol 2020; 146:1701-1709. [PMID: 32350606 DOI: 10.1007/s00432-020-03221-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/17/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND To investigate the incidence and prognostication of ERG, PTEN and SPINK1 protein expressions in prostate cancer cohort of Middle Eastern descent in comparison to published data from Western population. METHODS Immunohistochemistry for ERG, PTEN and SPINK1 was performed in a cohort of localized PCA (n = 340). The data were correlated to pathological and clinical outcomes and compared to Western populations. RESULTS ERG expression and PTEN loss were noted in 123/288 (42.7%) and 91/297 (30.6%) of patients, respectively. SPINK1 expression was assessed in a subset of cases, noted in 6/150 (4%) of patients. Only ERG expression was associated with grade groups, being more common in the lower grade groups (1-3 vs 4-5; p = 0.04). In contrast to the Western population, PTEN loss foci were more likely to be ERG negative, observed in 81% of tumor foci and patients with PTEN neg/ERG pos were more likely to exhibit biochemical recurrence (OR 2.831; 95% CI 1.10-726, p = 0.03). This association remained significant in multivariate analysis (OR 2.68; 95% CI 0.98-7.33, p = 0.05), after adjusting for GG, path stage and surgical margin. CONCLUSION This study documents significant differences in key molecular events in PCA in Middle Eastern population compared to Western populations that could explain differences in PCA incidence, progression and prognostication. ERG, PTEN and SPINK1 genomic alteration occur less frequently and the enrichment of ERG for PTEN loss is not observed. Additionally, patients with combined PTEN loss/ERG positive are at highest risk for BCR vs North American Caucasian population where PTEN loss alone seems to be associated with the worst clinical outcome. The data presented here further support differences in clonal evolution between Middle Eastern and Western population in relation to PCA and add further insight to understanding PCA molecular pathways.
Collapse
Affiliation(s)
- Ramy A Abdelsalam
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
- Department of Pathology, Mansoura University, Mansoura, Egypt
| | - Ibrahim Khalifeh
- Department of Pathology and Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Alan Box
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Maria Kalantarian
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Sunita Ghosh
- Alberta Health Services-Cancer Control and Department of Medical Oncology, University of Alberta, Edmonton, AB, Canada
| | - Hatem Abou-Ouf
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada
| | - Tamara Lotfi
- Department of Pathology and Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammed Shahait
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Nallasivam Palanisamy
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI, USA
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, Calgary, AB, Canada.
- Departments of Oncology, Biochemistry and Molecular Biology, Calgary, AB, Canada.
- Arnie Charbonneau Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada.
- Rockyview General Hospital, 7007, 14th st, Calgary, SW, ABT2V1P9, Canada.
| |
Collapse
|
3
|
Offermann A, Roth D, Hupe MC, Hohensteiner S, Becker F, Joerg V, Carlsson J, Kuempers C, Ribbat-Idel J, Tharun L, Sailer V, Kirfel J, Svensson M, Andren O, Lubczyk V, Kuefer R, Merseburger AS, Perner S. TRIM24 as an independent prognostic biomarker for prostate cancer. Urol Oncol 2019; 37:576.e1-576.e10. [PMID: 31178279 DOI: 10.1016/j.urolonc.2019.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Simply applicable biomarkers for prostate cancer patients predicting the clinical course are urgently needed. Recently, TRIM24 has been identified to promote androgen receptor signaling and to correlate with an aggressive prostate cancer phenotype. Based on these data, we proofed TRIM24 as a prognostic biomarker for risk stratification. MATERIALS AND METHODS We performed TRIM24 immunohistochemistry on 2 independent cohorts including a total of 806 primary tumors, 26 locally advanced/recurrent tumors, 30 lymph node metastases, 30 distant metastases, and 129 benign prostatic samples from 497 patients as well as on 246 prostate needle biopsies. Expression data were correlated with clinic-pathological data including biochemical recurrence-free survival (bRFS) as endpoint. RESULTS Benign samples show no or low TRIM24 expression in 94%, while tumor tissues demonstrate significant higher levels. Strongest expression is observed in advanced and metastatic tumors. In multivariate analyses, TRIM24 up-regulation on radical prostatectomy specimens correlates with shorter bRFS independent of other prognostic parameters. 5-(10-) year bRFS rates for TRIM24 negative, low, medium and high expressing tumors are 93.1(93.1)%, 75.4(68.5)%, 54.9(47.5)% and 43.1(32.3)%, respectively. Of interest, tumors diagnosed as indolent disease, TRIM24 expression stratifies patients into specific risk groups. Increased TRIM24 expression associates with higher grade group, positive nodal status and extraprostatic tumor growth. TRIM24 assessment on prostate needle biopsies taken prior to treatment decision at time of initial diagnosis significantly correlates with recurrence after surgery. CONCLUSION Using 2 large independent radical prostatectomy specimen cohorts, we found that TRIM24 expression predicts patients' risk to develop disease recurrence with high accuracy and independent from other established biomarkers. Further, this is the first study exploring TRIM24 expression on prostate needle biopsies which represents the clinically relevant tissue type on which biomarkers guide treatment decisions. Thus, we strongly suggest introducing TRIM24 evaluation in prostate needle biopsies in clinical routine as an inexpensive and simple immunohistochemical test.
Collapse
Affiliation(s)
- Anne Offermann
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Doris Roth
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Silke Hohensteiner
- Department of Pathology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Finn Becker
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Vincent Joerg
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Jessica Carlsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Christiane Kuempers
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Julika Ribbat-Idel
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Lars Tharun
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Verena Sailer
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Maria Svensson
- Department of Research and Education, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ove Andren
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Verena Lubczyk
- Department of Pathology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Rainer Kuefer
- Department of Urology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | | | - Sven Perner
- Institute of Pathology, University Hospital Schleswig-Holstein, Luebeck, Germany; Pathology of the Research Center Borstel, Leibniz Lung Center, Borstel, Germany.
| |
Collapse
|
4
|
The knockdown of the mediator complex subunit MED30 suppresses the proliferation and migration of renal cell carcinoma cells. Ann Diagn Pathol 2018; 34:18-26. [DOI: 10.1016/j.anndiagpath.2017.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/17/2017] [Accepted: 12/18/2017] [Indexed: 12/13/2022]
|
5
|
Zhou CK, Young D, Yeboah ED, Coburn SB, Tettey Y, Biritwum RB, Adjei AA, Tay E, Niwa S, Truelove A, Welsh J, Mensah JE, Hoover RN, Sesterhenn IA, Hsing AW, Srivastava S, Cook MB. TMPRSS2:ERG Gene Fusions in Prostate Cancer of West African Men and a Meta-Analysis of Racial Differences. Am J Epidemiol 2017; 186:1352-1361. [PMID: 28633309 PMCID: PMC5860576 DOI: 10.1093/aje/kwx235] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022] Open
Abstract
The prevalence of fusions of the transmembrane protease, serine 2, gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2:ERG, in prostate cancer varies by race. However, such somatic aberration and its association with prognostic factors have neither been studied in a West African population nor been systematically reviewed in the context of racial differences. We used immunohistochemistry to assess oncoprotein encoded by the ERG gene as the established surrogate of ERG fusion genes among 262 prostate cancer biopsies from the Ghana Prostate Study (2004-2006). Poisson regression with robust variance estimation provided prevalence ratios and 95% confidence intervals of ERG expression in relation to patient characteristics. We found that 47 of 262 (18%) prostate cancers were ERG-positive, and being negative for ERG staining was associated with higher Gleason score. We further conducted a systematic review and meta-analysis of TMPRSS2:ERG fusions in relation to race, Gleason score, and tumor stage, combining results from Ghana with 40 additional studies. Meta-analysis showed the prevalence of TMPRSS2:ERG fusions in prostate cancer to be highest in men of European descent (49%), followed by men of Asian (27%) and then African (25%) descent. The lower prevalence of TMPRSS2:ERG fusions in men of African descent implies that alternative genomic mechanisms might explain the disproportionately high prostate cancer burden in such populations.
Collapse
Affiliation(s)
- Cindy Ke Zhou
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Denise Young
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Rockville, Maryland
| | | | - Sally B Coburn
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Yao Tettey
- University of Ghana Medical School, Accra, Ghana
| | | | | | - Evelyn Tay
- University of Ghana Medical School, Accra, Ghana
| | | | | | - Judith Welsh
- NIH Library, National Institutes of Health, Bethesda, Maryland
| | | | - Robert N Hoover
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Isabell A Sesterhenn
- Genitourinary Pathology, Joint Pathology Center, Department of Defense, Silver Spring, Maryland
| | - Ann W Hsing
- Stanford Prevention Research Center and Cancer Institute, Palo Alto, California
- Department of Health Research and Policy, Stanford School of Medicine, Palo Alto, California
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Rockville, Maryland
| | - Michael B Cook
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
6
|
Klümper N, Syring I, Vogel W, Schmidt D, Müller SC, Ellinger J, Shaikhibrahim Z, Brägelmann J, Perner S. Mediator Complex Subunit MED1 Protein Expression Is Decreased during Bladder Cancer Progression. Front Med (Lausanne) 2017; 4:30. [PMID: 28367434 PMCID: PMC5355444 DOI: 10.3389/fmed.2017.00030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 03/03/2017] [Indexed: 01/03/2023] Open
Abstract
Introduction Bladder cancer (BCa) is among the most frequent cancer entities and relevantly contributes to cancer-associated deaths worldwide. The multi-protein Mediator complex is a central regulator of the transcriptional machinery of protein-coding genes and has been described to be altered in several malignancies. MED1, a subunit of the tail module, was described to negatively modulate expression of metastasis-related genes and to be downregulated in melanoma and lung cancer. In contrast, MED1 hyperactivity was described in breast and prostate cancer, likely due its function as a hub for nuclear hormone receptors. So far, only little is known about the function of the Mediator complex in BCa. The aim of this study was therefore to investigate the role of MED1 in BCa as a prognostic biomarker and a biomarker of disease progression. Methods The protein expression of MED1 was assessed by immunohistochemistry (IHC) on tissue microarrays from 224 patients: benign urothelium n = 31, non-muscle invasive BCa (pTis, pT1) n = 72, and muscle invasive BCa (pT2–T4) n = 121. Comprehensive clinicopathological information including follow-up were available. Quantification of MED1 protein expression was evaluated by the semiquantitative image analysis program Definiens. Results MED1 expression significantly decreased during BCa progression from benign urothelium to advanced BCa. Muscle invasion, the crucial step in BCa progression, was associated with low MED1 protein expression. Accordingly, decreased MED1 expression was found in primary BCa samples with positive lymphonodal status and distant metastases. Furthermore, cancer-specific survival was significantly worse in the group of low MED1 expression. Conclusion Our findings show that the downregulation of MED1 is associated with muscle invasion, metastatic spread, and shorter overall survival in BCa.
Collapse
Affiliation(s)
- Niklas Klümper
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences , Luebeck , Germany
| | - Isabella Syring
- Clinic for Urology and Pediatric Urology, University Hospital of Bonn , Bonn , Germany
| | - Wenzel Vogel
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences , Luebeck , Germany
| | - Doris Schmidt
- Clinic for Urology and Pediatric Urology, University Hospital of Bonn , Bonn , Germany
| | - Stefan C Müller
- Clinic for Urology and Pediatric Urology, University Hospital of Bonn , Bonn , Germany
| | - Jörg Ellinger
- Clinic for Urology and Pediatric Urology, University Hospital of Bonn , Bonn , Germany
| | - Zaki Shaikhibrahim
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences , Luebeck , Germany
| | - Johannes Brägelmann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Luebeck, Germany; Department of Hematology, Oncology and Rheumatology, University Hospital of Bonn, Bonn, Germany
| | - Sven Perner
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences , Luebeck , Germany
| |
Collapse
|
7
|
Brägelmann J, Klümper N, Offermann A, von Mässenhausen A, Böhm D, Deng M, Queisser A, Sanders C, Syring I, Merseburger AS, Vogel W, Sievers E, Vlasic I, Carlsson J, Andrén O, Brossart P, Duensing S, Svensson MA, Shaikhibrahim Z, Kirfel J, Perner S. Pan-Cancer Analysis of the Mediator Complex Transcriptome Identifies CDK19 and CDK8 as Therapeutic Targets in Advanced Prostate Cancer. Clin Cancer Res 2016; 23:1829-1840. [PMID: 27678455 DOI: 10.1158/1078-0432.ccr-16-0094] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/28/2016] [Accepted: 09/15/2016] [Indexed: 11/16/2022]
Abstract
Purpose: The Mediator complex is a multiprotein assembly, which serves as a hub for diverse signaling pathways to regulate gene expression. Because gene expression is frequently altered in cancer, a systematic understanding of the Mediator complex in malignancies could foster the development of novel targeted therapeutic approaches.Experimental Design: We performed a systematic deconvolution of the Mediator subunit expression profiles across 23 cancer entities (n = 8,568) using data from The Cancer Genome Atlas (TCGA). Prostate cancer-specific findings were validated in two publicly available gene expression cohorts and a large cohort of primary and advanced prostate cancer (n = 622) stained by immunohistochemistry. The role of CDK19 and CDK8 was evaluated by siRNA-mediated gene knockdown and inhibitor treatment in prostate cancer cell lines with functional assays and gene expression analysis by RNAseq.Results: Cluster analysis of TCGA expression data segregated tumor entities, indicating tumor-type-specific Mediator complex compositions. Only prostate cancer was marked by high expression of CDK19 In primary prostate cancer, CDK19 was associated with increased aggressiveness and shorter disease-free survival. During cancer progression, highest levels of CDK19 and of its paralog CDK8 were present in metastases. In vitro, inhibition of CDK19 and CDK8 by knockdown or treatment with a selective CDK8/CDK19 inhibitor significantly decreased migration and invasion.Conclusions: Our analysis revealed distinct transcriptional expression profiles of the Mediator complex across cancer entities indicating differential modes of transcriptional regulation. Moreover, it identified CDK19 and CDK8 to be specifically overexpressed during prostate cancer progression, highlighting their potential as novel therapeutic targets in advanced prostate cancer. Clin Cancer Res; 23(7); 1829-40. ©2016 AACR.
Collapse
Affiliation(s)
- Johannes Brägelmann
- Section for Prostate Cancer Research, University Hospital of Bonn, Bonn, Germany.,Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.,Department of Hematology, Oncology and Rheumatology, University Hospital of Bonn, Bonn, Germany
| | - Niklas Klümper
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Anne Offermann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Anne von Mässenhausen
- Section for Prostate Cancer Research, University Hospital of Bonn, Bonn, Germany.,Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany
| | - Diana Böhm
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Mario Deng
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Angela Queisser
- Section for Prostate Cancer Research, University Hospital of Bonn, Bonn, Germany.,Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany
| | - Christine Sanders
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Isabella Syring
- Section for Prostate Cancer Research, University Hospital of Bonn, Bonn, Germany.,Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.,Clinic for Urology and Pediatric Urology, University Hospital of Bonn, Bonn, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Wenzel Vogel
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Elisabeth Sievers
- Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany
| | - Ignacija Vlasic
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Jessica Carlsson
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ove Andrén
- Department of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Peter Brossart
- Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.,Department of Hematology, Oncology and Rheumatology, University Hospital of Bonn, Bonn, Germany
| | - Stefan Duensing
- Molecular Uro-oncology, Department of Urology, University of Heidelberg, Heidelberg, Germany
| | - Maria A Svensson
- Department of Research and Education, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Zaki Shaikhibrahim
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Hospital of Bonn, Bonn, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany
| | - Sven Perner
- Pathology of the University Medical Center Schleswig-Holstein, Campus Lübeck and the Research Center Borstel, Leibniz Center for Medicine and Biosciences, Lübeck and Borstel, Germany.
| |
Collapse
|
8
|
Abstract
Prostate cancer is a clinically heterogeneous disease, with some men having indolent disease that can safely be observed, while others have aggressive, lethal disease. Over the past decade, researchers have begun to unravel some of the genomic heterogeneity that contributes to these varying clinical phenotypes. Distinct molecular sub-classes of prostate cancer have been identified, and the uniqueness of these sub-classes has been leveraged to predict clinical outcomes, design novel biomarkers for prostate cancer diagnosis, and develop novel therapeutics. Recent work has also elucidated the temporal and spatial heterogeneity of prostate cancer, helping us understand disease pathogenesis, response to therapy, and progression. New genomic techniques have provided us with a window into the remarkable clinical and genomic heterogeneity of prostate cancer, and this new perspective will increasingly impact patient care.
Collapse
Affiliation(s)
- Jonathan Shoag
- Department of Urology, NewYork–Presbyterian Hospital, Weill Cornell Medical College, New York, USA
| | - Christopher E Barbieri
- Department of Urology, NewYork–Presbyterian Hospital, Weill Cornell Medical College, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, USA
| |
Collapse
|
9
|
Klümper N, Syring I, Offermann A, Shaikhibrahim Z, Vogel W, Müller SC, Ellinger J, Strauß A, Radzun HJ, Ströbel P, Brägelmann J, Perner S, Bremmer F. Differential expression of Mediator complex subunit MED15 in testicular germ cell tumors. Diagn Pathol 2015; 10:165. [PMID: 26377566 PMCID: PMC4573996 DOI: 10.1186/s13000-015-0398-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/28/2015] [Indexed: 01/07/2023] Open
Abstract
Background Testicular germ cell tumors (TGCT) are the most common cancer entities in young men with increasing incidence observed in the last decades. For therapeutic management it is important, that TGCT are divided into several histological subtypes. MED15 is part of the multiprotein Mediator complex which presents an integrative hub for transcriptional regulation and is known to be deregulated in several malignancies, such as prostate cancer and bladder cancer role, whereas the role of the Mediator complex in TGCT has not been investigated so far. Aim of the study was to investigate the implication of MED15 in TGCT development and its stratification into histological subtypes. Methods Immunohistochemical staining (IHC) against Mediator complex subunit MED15 was conducted on a TGCT cohort containing tumor-free testis (n = 35), intratubular germ cell neoplasia unclassified (IGCNU, n = 14), seminomas (SEM, n = 107) and non-seminomatous germ cell tumors (NSGCT, n = 42), further subdivided into embryonic carcinomas (EC, n = 30), yolk sac tumors (YST, n = 5), chorionic carcinomas (CC, n = 5) and teratomas (TER, n = 2). Quantification of MED15 protein expression was performed through IHC followed by semi-quantitative image analysis using the Definiens software. Results In tumor-free seminiferous tubules, MED15 protein expression was absent or only low expressed in spermatogonia. Interestingly, the precursor lesions IGCNU exhibited heterogeneous but partly very strong MED15 expression. SEM weakly express the Mediator complex subunit MED15, whereas NSGCT and especially EC show significantly enhanced expression compared to tumor-free testis. Conclusions In conclusion, MED15 is differentially expressed in tumor-free testis and TGCT. While MED15 is absent or low in tumor-free testis and SEM, NSGCT highly express MED15, hinting at the diagnostic potential of this marker to distinguish between SEM and NSGCT. Further, the precursor lesion IGCNU showed increased nuclear MED15 expression in the preinvasive precursor cells, which may provide diagnostic value to distinguish between benign and pre-malignant testicular specimen, and may indicate a role for MED15 in carcinogenesis in TGCT.
Collapse
Affiliation(s)
- Niklas Klümper
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.
| | - Isabella Syring
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany. .,Department of Urology and Pediatric Urology, University Hospital of Bonn, Bonn, Germany.
| | - Anne Offermann
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.
| | - Zaki Shaikhibrahim
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.
| | - Wenzel Vogel
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.
| | - Stefan C Müller
- Department of Urology and Pediatric Urology, University Hospital of Bonn, Bonn, Germany.
| | - Jörg Ellinger
- Department of Urology and Pediatric Urology, University Hospital of Bonn, Bonn, Germany.
| | - Arne Strauß
- Department of Urology, University Medical Center, University of Göttingen, Göttingen, Germany.
| | - Heinz Joachim Radzun
- Institute of Pathology, University Hospital of Göttingen, Robert Koch Str. 40, 37075, Göttingen, Germany.
| | - Philipp Ströbel
- Institute of Pathology, University Hospital of Göttingen, Robert Koch Str. 40, 37075, Göttingen, Germany.
| | - Johannes Brägelmann
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany. .,Department of Hematology/Oncology, University Hospital of Bonn, Bonn, Germany.
| | - Sven Perner
- Section for Prostate Cancer Research, Institute of Pathology, Center for Integrated Oncology Cologne/Bonn, University Hospital of Bonn, Bonn, Germany.
| | - Felix Bremmer
- Institute of Pathology, University Hospital of Göttingen, Robert Koch Str. 40, 37075, Göttingen, Germany.
| |
Collapse
|
10
|
Berg KD, Brasso K, Thomsen FB, Røder MA, Holten-Rossing H, Toft BG, Iversen P, Vainer B. ERG protein expression over time: from diagnostic biopsies to radical prostatectomy specimens in clinically localised prostate cancer. J Clin Pathol 2015; 68:788-94. [DOI: 10.1136/jclinpath-2015-202894] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/17/2015] [Indexed: 11/04/2022]
Abstract
AimsWe evaluated the consistency in ERG protein expression from diagnostic specimens through rebiopsies to radical prostatectomies in patients with clinically localised prostate cancer to investigate the validity of ERG status in biopsies.MethodsERG expression was assessed by immunohistochemistry (IHC) in 625 biopsy sets and 86 radical prostatectomy specimens from 265 patients with prostate cancer managed on active surveillance. For IHC, a rabbit monoclonal primary antibody was used (clone: EPR3864). TMPRSS2-ERG fluorescence in situ hybridisation (FISH) analyses were performed in 74 biopsies using the FISH ZytoLight TriCheck Probe (SPEC ERG/TMPRSS2). FISH results were correlated with IHC findings.ResultsThe concordance between FISH and IHC was 97.3% and IHC demonstrated a sensitivity and specificity for ERG rearrangement of 100% and 95.5%, respectively. Applying IHC, 38.1% of patients were ERG-positive, 53.6% were ERG-negative and 8.3% showed both ERG-positive and negative tumour foci (ERG heterogeneous) at diagnosis. When ERG status was dichotomised (ERG-positive or heterogeneous vs ERG-negative), 95.6%–97.1% of patients did not experience ERG reclassification during the first two rounds of rebiopsies. The concordance in ERG status between biopsies and surgical specimen was 89.5%–94.2% depending on the number of rebiopsies included. Sampling bias was assumed to explain most (81.3%) of the mismatches in ERG status.ConclusionsConsistency in ERG status ranged from 90% to 95% for patients undergoing serial biopsies and radical prostatectomy. This indicates that biopsies can be used reliably to investigate ERG's prognostic and predictive value.
Collapse
|
11
|
Fleischmann A, Saramäki OR, Zlobec I, Rotzer D, Genitsch V, Seiler R, Visakorpi T, Thalmann GN. Prevalence and prognostic significance of TMPRSS2-ERG gene fusion in lymph node positive prostate cancers. Prostate 2014; 74:1647-54. [PMID: 25252136 DOI: 10.1002/pros.22882] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/28/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND TMPRSS2-ERG gene fusion is the most frequent genetic alteration in prostate cancer. However, information about its distribution in lymph node positive prostate cancers and the prognostic significance in these advanced tumors is unknown. METHODS Gene fusion status was determined by fluorescence in situ hybridization on a tissue-microarray constructed from 119 hormone-naïve nodal positive, surgically treated prostate cancers containing samples from the primary tumors and corresponding lymph node metastases. Data were correlated with various tumor features (Gleason score, stage, cancer volume, nodal tumor burden) and biochemical recurrence-free, disease-specific, and overall survival. RESULTS TMPRSS2-ERG fusion was detected in 43.5% of the primary tumors. Conversely, only 29.9% of the metastasizing components showed the fusion. Concordance in TMPRSS2-ERG status between primary tumors and metastases was 70.9% (Kappa 0.39); 20.9% and 8.1% of the patients showed the mutation solely in their primary tumors and metastases, respectively. TMPRSS2-ERG fusion was not correlated with specific histopathological tumor features but predicted favorable biochemical recurrence-free, disease-specific and overall survival independently when present in the primary tumor (P < 0.05 each). CONCLUSION TMPRSS2-ERG fusion is more frequent in primary prostate cancer than in corresponding metastases suggesting no selection of fusion-positive cells in the metastatic process. The gene fusion in primary tumors independently predicts favorable outcome.
Collapse
|
12
|
Comprehensive validation of published immunohistochemical prognostic biomarkers of prostate cancer -what has gone wrong? A blueprint for the way forward in biomarker studies. Br J Cancer 2014; 112:140-8. [PMID: 25422912 PMCID: PMC4453620 DOI: 10.1038/bjc.2014.588] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/17/2014] [Accepted: 10/25/2014] [Indexed: 02/06/2023] Open
Abstract
Background: Treatment planning of localised prostate cancer remains challenging. Besides conventional parameters, a wealth of prognostic biomarkers has been proposed so far. None of which, however, have successfully been implemented in a routine setting so far. The aim of our study was to systematically verify a set of published prognostic markers for prostate cancer. Methods: Following an in-depth PubMed search, 28 markers were selected that have been proposed as multivariate prognostic markers for primary prostate cancer. Their prognostic validity was examined in a radical prostatectomy cohort of 238 patients with a median follow-up of 60 months and biochemical progression as endpoint of the analysis. Immunohistochemical evaluation was performed using previously published cut-off values, but allowing for optimisation if necessary. Univariate and multivariate Cox regression were used to determine the prognostic value of biomarkers included in this study. Results: Despite the application of various cut-offs in the analysis, only four (14%) markers were verified as independently prognostic (AKT1, stromal AR, EZH2, and PSMA) for PSA relapse following radical prostatectomy. Conclusions: Apparently, many immunohistochemistry-based studies on prognostic markers seem to be over-optimistic. Codes of best practice, such as the REMARK guidelines, may facilitate the performance of conclusive and transparent future studies.
Collapse
|
13
|
Adler D, Offermann A, Braun M, Menon R, Syring I, Nowak M, Halbach R, Vogel W, Ruiz C, Zellweger T, Rentsch CA, Svensson M, Andren O, Bubendorf L, Biskup S, Duensing S, Kirfel J, Perner S. MED12 overexpression is a frequent event in castration-resistant prostate cancer. Endocr Relat Cancer 2014; 21:663-675. [PMID: 24938407 DOI: 10.1530/erc-14-0171] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In a recent effort to unravel the molecular basis of prostate cancer (PCa), Barbieri and colleagues using whole-exome sequencing identified a novel recurrently mutated gene, MED12, in 5.4% of primary PCa. MED12, encoding a subunit of the Mediator complex, is a transducer of Wnt/β-catenin signaling, linked to modulation of hedgehog signaling and to the regulation of transforming growth factor beta (TGFβ)-receptor signaling. Therefore, these studies prompted us to investigate the relevance of MED12 in PCa. Expression of MED12, SMAD3 phosphorylation, and proliferation markers was assessed by immunohistochemistry on tissue microarrays from 633 patients. siRNA-mediated knockdown of MED12 was carried out on PCa cell lines followed by cellular proliferation assays, cell cycle analysis, apoptosis assays, and treatments with recombinant TGFβ3. We found nuclear overexpression of MED12 in 40% (28/70) of distant metastatic castration-resistant prostate cancer (CRPC(MET)) and 21% (19/90) of local-recurrent CRPC (CRPC(LOC)) in comparison with frequencies of less than 11% in androgen-sensitive PCa, and no overexpression in benign prostatic tissues. MED12 expression was significantly correlated with high proliferative activity in PCa tissues, whereas knockdown of MED12 decreased proliferation, reduced G1- to S-phase transition, and increased the expression of the cell cycle inhibitor p27. TGFβ signaling activation associates with MED12 nuclear overexpression in tissues and results in a strong increase in MED12 nuclear expression in cell lines. Furthermore, MED12 knockdown reduced the expression of the TGFβ target gene vimentin. Our findings show that MED12 nuclear overexpression is a frequent event in CRPC in comparison with androgen-sensitive PCa and is directly implicated in TGFβ signaling.
Collapse
|
14
|
Falzarano SM, Magi-Galluzzi C. ERG protein expression as a biomarker of prostate cancer. Biomark Med 2014; 7:851-65. [PMID: 24266818 DOI: 10.2217/bmm.13.105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
TMPRSS2-ERG is a recurrent rearrangement specific for prostate cancer, leading to the overexpression of a truncated ERG protein product that is amenable to immunohistochemical detection. Two monoclonal anti-ERG antibodies have currently been validated, with comparable sensitivity and specificity for detecting ERG rearrangement. ERG immunostaining has been applied in different settings to elucidate the role of ERG rearrangement and overexpression in prostate cancer tumorigenesis and progression, as well as to investigate potential diagnostic and prognostic applications. In this article we review the literature on the topic and suggest potential future applications.
Collapse
Affiliation(s)
- Sara Moscovita Falzarano
- R.T. Pathology & Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, L25, Cleveland, OH 44195, USA
| | | |
Collapse
|
15
|
Smith SC, Palanisamy N, Zuhlke KA, Johnson AM, Siddiqui J, Chinnaiyan AM, Kunju LP, Cooney KA, Tomlins SA. HOXB13 G84E-related familial prostate cancers: a clinical, histologic, and molecular survey. Am J Surg Pathol 2014; 38:615-26. [PMID: 24722062 PMCID: PMC3988475 DOI: 10.1097/pas.0000000000000090] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent genetic epidemiologic studies identified a germline mutation in the homeobox transcription factor, HOXB13 G84E, which is associated with markedly increased risk for prostate cancer, particularly early-onset hereditary prostate cancer. The histomorphologic and molecular features of cancers arising in such carriers have not been studied. Here, we reviewed prostatectomy specimens from 23 HOXB13 G84E mutation carriers, mapping the total cancer burden by anatomically distinct cancer focus and evaluating morphologic features. We also assessed basic molecular subtypes for all cancer foci (ERG/SPINK1 status) by dual immunohistochemistry staining on full sections. The cohort showed a median age of 58 years, a median serum PSA level of 5.7 ng/mL, and a median of 6 cancer foci (range, 1 to 14) per case. Of evaluable cases, dominant foci were Gleason score 6 in 23%, 3+4=7 in 41%, 4+3=7 in 23%, and ≥8 in 14%; biochemical recurrence was observed in 1 case over a median of 36 months follow-up. Histologic review found a high prevalence of cases showing cancers with a spectrum of features previously described with pseudohyperplastic carcinomas, with 45% of cases showing a dominant focus with such features. Molecular subtyping revealed a strikingly low prevalence of ERG cancer with increased prevalence of SPINK1 cancer (dominant focus ERG 17%, SPINK1 26%, ERG/SPINK1 52%, single ERG/SPINK1 focus 4%). One ERG/SPINK1 dominant focus showed aberrant p63 immunophenotype. In summary, HOXB13 G84E variant-related prostate cancers show frequent pseudohyperplastic-type features and markedly low prevalence of ERG cancers relative to unselected cases and, especially, to early-onset cohorts. These findings suggest that novel molecular pathways may drive disease in HOXB13 G84E carriers.
Collapse
Affiliation(s)
- Steven C. Smith
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nallasivam Palanisamy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI USA
| | - Kimberly A. Zuhlke
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anna M. Johnson
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI USA
- Howard Hughes Medical Institute
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Lakshmi P Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI USA
| | - Kathleen A. Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI USA
| | - Scott A. Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI USA
| |
Collapse
|
16
|
Xu B, Chevarie-Davis M, Chevalier S, Scarlata E, Zeizafoun N, Dragomir A, Tanguay S, Kassouf W, Aprikian A, Brimo F. The prognostic role of ERG immunopositivity in prostatic acinar adenocarcinoma: a study including 454 cases and review of the literature. Hum Pathol 2014; 45:488-97. [DOI: 10.1016/j.humpath.2013.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/04/2013] [Accepted: 10/10/2013] [Indexed: 10/26/2022]
|
17
|
ERG rearrangement is associated with prostate cancer-related death in Chinese prostate cancer patients. PLoS One 2014; 9:e84959. [PMID: 24516518 PMCID: PMC3917829 DOI: 10.1371/journal.pone.0084959] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/20/2013] [Indexed: 11/19/2022] Open
Abstract
Recently, ETS-related gene (ERG) gene rearrangements, phosphatase tensin homologue (PTEN) deletions and EGFR family aberrations were characterized as potential biomarkers for prostate cancer (PCa) patient management. Although ERG gene rearrangement has been identified in approximately 50% of localized prostate cancers in western countries, the prognostic significance of this critical molecular event remains unknown in Chinese patients. Using fluorescence in situ hybridization (FISH) and immunohistochemistry, we evaluated ERG, PTEN and EGFR family aberrations in a cohort of 224 Chinese prostate cancer patients diagnosed in transurethral resection of the prostate (TUR-P). Overall, ERG rearrangement was detected in 23.2% (44/190) cases, of which 54.5% (24/44) showed deletion of the 5'end of ERG. PTEN deletion was identified in 10.8% (19/176) cases. Amplification of EGFR and HER2 genes was present in 1.1% (2/178) and 5.8% (10/173) of cases, respectively. Significant correlation between ERG rearrangement and PTEN deletion was identified in this cohort. EGFR and HER2 aberrations occurred more frequently in PCas without ERG rearrangement than in those with ERG rearrangement, although this did not reach statistical significance. Overall, ERG rearrangement was associated with pre-operative PSA values (P = 0.038) and cancer-related death (P = 0.02), but not with the age, clinical T stage, Gleason score, or Ki-67 labeling index (LI). Notably, multivariate analysis including known prognostic markers revealed ERG rearrangement was an independent prognostic factor (P = 0.022). Additionally, ERG rearrangement status was helpful to identify patients with poor prognosis from PCa group with low Ki-67 LI. In summary, we reported that ERG rearrangement was associated with cancer-related death in Chinese PCa patients. Determination of ERG rearrangement status allows stratification of PCa patients into different survival categories.
Collapse
|
18
|
Shaikhibrahim Z, Menon R, Braun M, Offermann A, Queisser A, Boehm D, Vogel W, Rüenauver K, Ruiz C, Zellweger T, Svensson M, Andren O, Kristiansen G, Wernert N, Bubendorf L, Kirfel J, Biskup S, Perner S. MED15, encoding a subunit of the mediator complex, is overexpressed at high frequency in castration-resistant prostate cancer. Int J Cancer 2013; 135:19-26. [DOI: 10.1002/ijc.28647] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/13/2013] [Accepted: 11/18/2013] [Indexed: 02/04/2023]
Affiliation(s)
- Zaki Shaikhibrahim
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Roopika Menon
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Martin Braun
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Anne Offermann
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Angela Queisser
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Diana Boehm
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Wenzel Vogel
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Kerstin Rüenauver
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Christian Ruiz
- Institute for Pathology; University Hospital Basel; Basel Switzerland
| | | | - Maria Svensson
- Department of Urology; University Hospital of Örebro; Örebro Sweden
| | - Ove Andren
- Department of Urology; University Hospital of Örebro; Örebro Sweden
- School of Health and Medical Sciences; Örebro University; Örebro Sweden
| | - Glen Kristiansen
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Nicolas Wernert
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Lukas Bubendorf
- Institute for Pathology; University Hospital Basel; Basel Switzerland
| | - Jutta Kirfel
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| | - Saskia Biskup
- Center for Genomics and Transcriptomics, CeGaT GmbH; Tuebingen Germany
| | - Sven Perner
- Department of Prostate Cancer Research; University Hospital of Bonn; Bonn Germany
- Institute of Pathology; University Hospital of Bonn; Bonn Germany
| |
Collapse
|
19
|
Gopalan A, Leversha MA, Dudas ME, Maschino AC, Chang J, Al-Ahmadie HA, Chen YB, Tickoo SK, Reuter VE, Fine SW. TMPRSS2-ERG rearrangement in dominant anterior prostatic tumours: incidence and correlation with ERG immunohistochemistry. Histopathology 2013; 63:279-86. [PMID: 23701505 DOI: 10.1111/his.12153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/26/2013] [Indexed: 11/28/2022]
Abstract
AIM To study prostate cancer zonal differences in TMPRSS2-ERG gene rearrangement. METHODS AND RESULTS We examined 136 well-characterized dominant anterior prostatic tumours, including 61 transition zone (TZ) and 75 anterior peripheral zone (PZ) lesions, defined using strict anatomical considerations. TMPRSS2-ERG FISH and ERG protein immunohistochemistry were performed on tissue microarrays. FISH results, available for 56 TZ and 71 anterior PZ samples, were correlated with ERG staining and TZ-associated 'clear cell' histology. Fewer TZ cancers (four of 56; 7%) were rearranged than anterior PZ cancers (18 of 71; 25%) (P = 0.009); deletion was the sole mechanism of TZ cancer rearrangement. ERG protein overexpression was present in 4% (two of 56; both FISH+) and 30% (21 of 71; 17 FISH+) of TZ and anterior PZ tumours, respectively. 'Clear cell' histology was present in 21 of 56 (38%) TZ and eight of 71 (11%) anterior PZ tumours. Seven per cent of cancers with and 21% without this histology had rearrangement, regardless of zonal origin. CONCLUSIONS TMPRSS2-ERG rearrangement occurs in dominant TZ and anterior PZ prostate cancers, with all rearranged TZ cancers in this cohort showing deletion. ERG immunohistochemistry demonstrated excellent sensitivity (86%) and specificity (96%) for TMPRSS2-ERG rearrangement. TMPRSS2-ERG fusion is rare in TZ tumours and present at a low frequency in tumours displaying 'clear cell' histology.
Collapse
Affiliation(s)
- Anuradha Gopalan
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
PTEN losses exhibit heterogeneity in multifocal prostatic adenocarcinoma and are associated with higher Gleason grade. Mod Pathol 2013; 26:435-47. [PMID: 23018874 DOI: 10.1038/modpathol.2012.162] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prostatic adenocarcinoma is an epithelial malignancy characterized by marked histological heterogeneity. It most often has a multifocal distribution within the gland, and different Gleason grades may be present within different foci. Data from our group and others have shown that the genomic deletion of the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor gene and the disruption of the ETS gene family have a central role in prostate cancer and are likely to be associated with Gleason grade. In this study, prostate cancer samples were systematically analyzed to determine whether there was concordance between PTEN losses and TMPRSS2-ERG fusion rearrangements, within or between foci in multifocal disease, using well-annotated tissue microarrays (TMAs) consisting of 724 cores derived from 142 radical prostatectomy specimens. Three-color fluorescence in situ hybridization analysis of both the PTEN deletion and the TMPRSS2-ERG fusion was used to precisely map genetic heterogeneity, both within and between tumor foci represented on the TMA. PTEN deletion was observed in 56 of 134 (42%) patients (hemizygous=42 and homozygous=14). TMPRSS2-ERG fusion was observed in 63 of 139 (45%) patients. When analyzed by Gleason pattern for a given TMA core, PTEN deletions were significantly associated with Gleason grades 4 or 5 over grade 3 (P<0.001). Although TMPRSS2-ERG fusions showed a strong relationship with PTEN deletions (P=0.007), TMPRSS2-ERG fusions did not show correlation with Gleason grade. The pattern of genetic heterogeneity of PTEN deletion was more diverse than that observed for TMPRSS2-ERG fusions in multifocal disease. However, the marked interfocal discordance for both TMPRSS2-ERG fusions and PTEN deletions was consistent with the concept that multiple foci of prostate cancer arise independently within the same prostate, and that individual tumor foci can have distinct patterns of genetic rearrangements.
Collapse
|
21
|
Brenner JC, Chinnaiyan AM, Tomlins SA. ETS Fusion Genes in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
22
|
Kimura T, Furusato B, Miki J, Yamamoto T, Hayashi N, Takahashi H, Kamata Y, van Leenders GJHL, Visakorpi T, Egawa S. Expression of ERG oncoprotein is associated with a less aggressive tumor phenotype in Japanese prostate cancer patients. Pathol Int 2012; 62:742-8. [DOI: 10.1111/pin.12006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
23
|
Shaikhibrahim Z, Braun M, Nikolov P, Boehm D, Scheble V, Menon R, Fend F, Kristiansen G, Perner S, Wernert N. Rearrangement of the ETS genes ETV-1, ETV-4, ETV-5, and ELK-4 is a clonal event during prostate cancer progression. Hum Pathol 2012; 43:1910-6. [DOI: 10.1016/j.humpath.2012.01.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 01/20/2012] [Accepted: 01/25/2012] [Indexed: 10/28/2022]
|
24
|
Shaikhibrahim Z, Ochsenfahrt J, Fuchs K, Kristiansen G, Perner S, Wernert N. ERG is specifically associated with ETS-2 and ETV-4, but not with ETS-1, in prostate cancer. Int J Mol Med 2012; 30:1029-33. [PMID: 22922762 PMCID: PMC3572757 DOI: 10.3892/ijmm.2012.1097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 06/22/2012] [Indexed: 11/06/2022] Open
Abstract
The erythroblast transformation-specific (ETS) family of transcription factors plays important roles in both physiological and pathological conditions. Even though many studies have focused on single ETS factors within a single tissue and within the context of specific promoters, the functional impact of multiple ETS members present within a specific cell type has not yet been investigated, especially in prostate cancer (PCa). As the most prominent gene rearrangement in PCa leads to the overexpression of the ETS-related gene (ERG), the aim of this study was to investigate whether ERG is part of a complex integrated transcriptional network that involves other ETS factors. More specifically, as the ETS family consists of 27 members, we focused our efforts initially on investigating whether ERG is associated with the three family members, ETS-1, ETS-2 and ETS variant gene‑4 (ETV‑4), in PCa as a proof of principle. Using western blot analysis, we show that ERG, ETS-1, ETS-2 and ETV-4 are expressed in PC3 cell nuclear extracts and in protein lysates prepared from human PCa prostatectomy specimens. Immunoprecipitations using an anti-ERG antibody were used with PC3 cell nuclear extracts as well as with a pooled protein lysate sample prepared from the PCa tissue samples of five patients. Importantly, our results revealed that ERG is specifically associated with ETS-2 and ETV-4, but not with ETS-1, in PC3 cell nuclear extracts and PCa tissue protein lysates. Our findings strongly support the notion that ERG is part of a complex integrated transcriptional network that involves other ETS factors, which are likely to cooperate or influence the activity of ERG in PCa. The functional impact of multiple ETS factors being associated with ERG in PCa requires further study, as it may provide insights into the mechanism by which ERG exerts its influence in PCa and may subsequently contribute to our understanding of the molecular basis of PCa.
Collapse
Affiliation(s)
- Zaki Shaikhibrahim
- Institute of Pathology, University Hospital of Bonn, D-53127 Bonn, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Scheble VJ, Scharf G, Braun M, Ruiz C, Stürm S, Petersen K, Beschorner R, Bachmann A, Zellweger T, Fend F, Kristiansen G, Bubendorf L, Wernert N, Shaikhibrahim Z, Perner S. ERG rearrangement in local recurrences compared to distant metastases of castration-resistant prostate cancer. Virchows Arch 2012; 461:157-62. [DOI: 10.1007/s00428-012-1270-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 04/17/2012] [Accepted: 06/13/2012] [Indexed: 11/30/2022]
|
26
|
Pettersson A, Graff RE, Bauer SR, Pitt MJ, Lis RT, Stack EC, Martin NE, Kunz L, Penney KL, Ligon AH, Suppan C, Flavin R, Sesso HD, Rider JR, Sweeney C, Stampfer MJ, Fiorentino M, Kantoff PW, Sanda MG, Giovannucci EL, Ding EL, Loda M, Mucci LA. The TMPRSS2:ERG rearrangement, ERG expression, and prostate cancer outcomes: a cohort study and meta-analysis. Cancer Epidemiol Biomarkers Prev 2012; 21:1497-509. [PMID: 22736790 DOI: 10.1158/1055-9965.epi-12-0042] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Whether the genomic rearrangement transmembrane protease, serine 2 (TMPRSS2):v-ets erythroblastosis virus E26 oncogene homolog (ERG) has prognostic value in prostate cancer is unclear. METHODS Among men with prostate cancer in the prospective Physicians' Health and Health Professionals Follow-Up Studies, we identified rearrangement status by immunohistochemical assessment of ERG protein expression. We used Cox models to examine associations of ERG overexpression with biochemical recurrence and lethal disease (distant metastases or cancer-specific mortality). In a meta-analysis including 47 additional studies, we used random-effects models to estimate associations between rearrangement status and outcomes. RESULTS The cohort consisted of 1,180 men treated with radical prostatectomy between 1983 and 2005. During a median follow-up of 12.6 years, 266 men experienced recurrence and 85 men developed lethal disease. We found no significant association between ERG overexpression and biochemical recurrence [hazard ratio (HR), 0.99; 95% confidence interval (CI), 0.78-1.26] or lethal disease (HR, 0.93; 95% CI, 0.61-1.43). The meta-analysis of prostatectomy series included 5,074 men followed for biochemical recurrence (1,623 events), and 2,049 men followed for lethal disease (131 events). TMPRSS2:ERG was associated with stage at diagnosis [risk ratio (RR)(≥T3 vs. T2), 1.23; 95% CI, 1.16-1.30) but not with biochemical recurrence (RR, 1.00; 95% CI, 0.86-1.17) or lethal disease (RR, 0.99; 95% CI, 0.47-2.09). CONCLUSIONS These results suggest that TMPRSS2:ERG, or ERG overexpression, is associated with tumor stage but does not strongly predict recurrence or mortality among men treated with radical prostatectomy. IMPACT This is the largest prospective cohort study to examine associations of ERG overexpression and lethal prostate cancer among men treated with radical prostatectomy.
Collapse
Affiliation(s)
- Andreas Pettersson
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Braun M, Stomper J, Boehm D, Vogel W, Scheble VJ, Wernert N, Shaikhibrahim Z, Fend F, Kristiansen G, Perner S. Improved method of detecting the ERG gene rearrangement in prostate cancer using combined dual-color chromogenic and silver in situ hybridization. J Mol Diagn 2012; 14:322-7. [PMID: 22642898 DOI: 10.1016/j.jmoldx.2012.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Revised: 01/26/2012] [Accepted: 01/31/2012] [Indexed: 12/29/2022] Open
Abstract
The recently detected TMPRSS2-ERG fusion gene was revealed as a recurrent and prevalent prostate cancer (PCa)-specific event, potentially qualifying it for clinical use. To detect this alteration, fluorescence in situ hybridization (FISH) is the method of choice. However, FISH has some disadvantages for widespread adoption in clinical practice. Subsequently, chromogenic in situ hybridization, which uses organic chromogens, and enzymatic metallography silver in situ hybridization have emerged as promising bright-field alternatives. Compared with chromogenic in situ hybridization, silver in situ hybridization signals are very distinct and superior with regard to signal clarity and resolution, but the method excludes multicolor protocols. Based on the ERG break-apart FISH assay, we established a dual-color ERG break-apart assay using combined chromogenic in situ hybridization and silver in situ hybridization (CS-ISH) and compared these results with those obtained by FISH. We assessed 178 PCa and 10 benign specimens for their ERG rearrangement status by applying dual-color FISH and CS-ISH ERG break-apart assays to consecutive sections. We observed a highly significant concordance (97.7%) between FISH- and CS-ISH-based results (Pearson's correlation coefficient = 0.955, P < 0.001). Our findings demonstrate that the ERG rearrangement status can reliably be assessed by CS-ISH. Further, the CS-ISH technique combines the accuracy and precision of FISH with the ease of bright-field microscopy. This tool allows a much broader spectrum of applications in which to study the biological role and clinical use of ERG rearrangements in PCa.
Collapse
Affiliation(s)
- Martin Braun
- Institute of Pathology, University Hospital of Bonn, Sigmund-Freud-Straße 25, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kron K, Liu L, Trudel D, Pethe V, Trachtenberg J, Fleshner N, Bapat B, van der Kwast T. Correlation of ERG expression and DNA methylation biomarkers with adverse clinicopathologic features of prostate cancer. Clin Cancer Res 2012; 18:2896-904. [PMID: 22452941 DOI: 10.1158/1078-0432.ccr-11-2901] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Fusion of the TMPRSS2 gene with the ERG oncogene and aberrant DNA methylation patterns are commonly found in prostate cancer. The aim of this study was to analyze the relationship between ERG expression, DNA methylation of three biomarkers, and clinicopathologic features of prostate cancer. EXPERIMENTAL DESIGN Immunohistochemistry for ERG protein was conducted as a surrogate for TMPRSS2-ERG fusions. We analyzed methylation of CYP26A1, TBX15, and HOXD3 in 219 prostatectomy specimens by the quantitative MethyLight assay. DNA methylation was compared between ERG-positive and -negative cases and correlations of ERG and DNA methylation with clinicopathologic features were analyzed using χ(2), Spearman correlation, logistic regression, and Cox regression. RESULTS ERG expression varied according to Gleason pattern (almost absent in pattern II, highest in pattern III, and lower in pattern IV/V) and showed a strong positive correlation with methylation levels of CYP26A1, TBX15, and HOXD3 (Spearman P < 0.005). TBX15 and HOXD3 methylation were significantly associated with pathologic stage, Gleason score, and Gleason pattern (P ≤ 0.015). In multivariate regression analysis, PSA, TBX15 high methylation, and HOXD3 high methylation were significantly associated with stage (P < 0.05), whereas ERG expression was negatively correlated with Gleason score (P = 0.003). In univariate time-to-recurrence analysis, a combination of HOXD3/TBX15 high methylation predicted recurrence in ERG-positive and -negative cases (P < 0.05). CONCLUSIONS CYP26A1, TBX15, and HOXD3 are methylation markers of prostate cancer associated with ERG expression and clinicopathologic variables, suggesting that incorporation of these markers may be useful in a pre- and posttreatment clinical setting.
Collapse
Affiliation(s)
- Ken Kron
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Bismar TA, Dolph M, Teng LH, Liu S, Donnelly B. ERG protein expression reflects hormonal treatment response and is associated with Gleason score and prostate cancer specific mortality. Eur J Cancer 2012; 48:538-46. [DOI: 10.1016/j.ejca.2012.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/22/2011] [Accepted: 01/03/2012] [Indexed: 10/14/2022]
|
30
|
ERG protein expression and genomic rearrangement status in primary and metastatic prostate cancer--a comparative study of two monoclonal antibodies. Prostate Cancer Prostatic Dis 2012; 15:165-9. [PMID: 22231490 DOI: 10.1038/pcan.2011.67] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Overexpression of the ERG protein is highly prevalent in prostate cancer (PCa) and commonly results from gene fusions involving the ERG gene. Recently, N-terminal epitope-targeted mouse and a C-terminal epitope-targeted rabbit monoclonal anti-ERG antibody (ERG-MAbs) have been introduced for the detection of the ERG protein. Independent studies reported that immunohistochemistry (IHC) with both ERG-MAbs highly correlates with the underlying ERG gene rearrangement status. However, comparative studies of both antibodies are lacking. Here, we are among the first to compare the mouse ERG-MAb with the rabbit ERG-MAb for their concordance on the same PCa cohort. Furthermore, we assessed whether the ERG protein expression is conserved in lymph node and distant PCa metastases. METHODS We evaluated tissue microarrays of 278 specimens containing 265 localized PCa, 29 lymph node, 30 distant metastases and 13 normal prostatic tissues. We correlated ERG protein expression with ERG rearrangement status using an ERG break-apart fluorescence in-situ hybridization assay and IHC of both ERG-MAbs. RESULTS ERG expression and ERG rearrangement status were highly concordant regardless of whether the mouse or rabbit ERG-MAb was used (97.8% versus 98.6%, respectively). Of interest, both ERG antibodies reliably detected the ERG expression in lymph node and distant PCa metastases, of which a subset underwent decalcification. Lymphocytes only revealed immunoreactivity using the rabbit ERG-MAb. If ERG protein expression was present in localized PCa, we observed the same pattern in the corresponding lymph node metastases. CONCLUSIONS By demonstrating a broad applicability of IHC to study ERG protein expression using either antibody, this study adds an important step toward a facilitated routine clinical application. Further, we demonstrate that the clonal nature of the ERG rearrangement is not restricted to the genomic level, but proceeds in the proteome. Together, our results simplify future efforts to further eliucidate the biological role of ERG in PCa.
Collapse
|
31
|
|
32
|
Perner S. [Dangerous liaisons in prostate cancer. Clinical and biological implications of recurrent gene fusions]. DER PATHOLOGE 2011; 31 Suppl 2:121-5. [PMID: 20798944 DOI: 10.1007/s00292-010-1345-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Prostate cancer is a common and clinically heterogeneous disease. Understanding the biology of prostate cancer is necessary to best determinate the risk of disease progression and develop novel therapeutic approaches to prevent or slow down disease progression. The recent discovery and subsequent characterization of recurrent gene rearrangements of ETS genes - most frequently ERG - in the majority of prostate cancers is a milestone in translational prostate cancer research. Although multiple molecular alterations have been detected in prostate cancer, a detailed understanding of gene fusion in prostate cancer should help explain the clinical and biologic diversity in addition to providing a rationale for a molecular sub-classification of the disease. This review describes the path from the identification of common ETS gene rearrangements in prostate cancer to possible applications in the treatment of patients, on to the potential scientific implications arising from their discovery.
Collapse
Affiliation(s)
- S Perner
- Institut für Pathologie, Comprehensive Cancer Center, Universitätsklinikum Tübingen, Liebermeisterstr. 8, 72076 Tübingen.
| |
Collapse
|