151
|
Abstract
PURPOSE OF REVIEW The recent publication of The Cancer Genome Atlas molecular taxonomy of primary prostate cancer highlights the increased understanding of the genomic basis of human prostate cancer, but also emphasizes the complexity and heterogeneity of prostate cancer. RECENT FINDINGS Seven molecular subclasses have been defined on the basis of early genomic alterations, which are largely mutually exclusive. SUMMARY We review the recent advances in the genomic understanding of human prostate cancer, with focus on molecular subclassification. Broadly, prostate cancer can be classified based upon whether specific genomic rearrangements, such as the Transmembrane Protease, Serine 2-ETS-related gene fusion occur or whether specific alterations such as Speckle-type POZ protein and forkhead box A1 mutations occur. The molecular drivers remain to be identified in a further quarter of human prostate cancers. Depending upon the molecular subclassification and the coincident genomic alterations, specific clinical insights can be gained from this information, including associations with pathologic factors, race, and prognosis, as well as the possibility for future precision therapies.
Collapse
|
152
|
Hendriks RJ, van Oort IM, Schalken JA. Blood-based and urinary prostate cancer biomarkers: a review and comparison of novel biomarkers for detection and treatment decisions. Prostate Cancer Prostatic Dis 2016; 20:12-19. [DOI: 10.1038/pcan.2016.59] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/26/2016] [Accepted: 10/24/2016] [Indexed: 11/09/2022]
|
153
|
Rodrigues DN, Boysen G, Sumanasuriya S, Seed G, Marzo AMD, de Bono J. The molecular underpinnings of prostate cancer: impacts on management and pathology practice. J Pathol 2016; 241:173-182. [PMID: 27753448 DOI: 10.1002/path.4826] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 12/20/2022]
Abstract
Prostate cancer (PCa) is a clinically heterogeneous disease and current treatment strategies are based largely on anatomical and pathological parameters. In the recent past, several DNA sequencing studies of primary and advanced PCa have revealed recurrent patterns of genomic aberrations that expose mechanisms of resistance to available therapies and potential new drug targets. Suppression of androgen receptor (AR) signalling is the cornerstone of advanced prostate cancer treatment. Genomic aberrations of the androgen receptor or alternative splicing of its mRNA are increasingly recognised as biomarkers of resistance to AR-targeted therapies such as abiraterone or enzalutamide. Genomic aberrations of the PI3K-AKT axis, in particular affecting PTEN, are common in PCa, and compounds targeting different kinases in this pathway are showing promise in clinical trials. Both germline and somatic defects in DNA repair genes have been shown to sensitise some patients to therapy with PARP inhibition. In addition, abnormalities in mismatch-repair genes are associated with response to immune checkpoint inhibition in other solid tumours and present a tantalising therapeutic avenue to be pursued. Aberrations in CDK4/6-RB1 pathway genes occur in a subset of PCas, may associate with differential sensitivity to treatment, and are likely to have clinical implications beyond prognostication. Inhibitors of CDK4/6 are already being tested in prostate cancer clinical trials. Furthermore, deletions of RB1 are strongly associated with a neuroendocrine phenotype, a rare condition characterized by a non-AR-driven transcriptomic profile. Finally, aberrations in genes involved in regulating the chromatin structure are an emerging area of interest. Deletions of CHD1 are not infrequent in PCa and may associate with increased AR activity and genomic instability, and these tumours could benefit from DNA-damaging therapies. This review summarises how genomic discoveries in PCa are changing the treatment landscape of advanced CRPC, both by identifying biomarkers of resistance and by identifying vulnerabilities to be targeted. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Daniel Nava Rodrigues
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.,Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Gunther Boysen
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.,Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - Semini Sumanasuriya
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.,Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| | - George Seed
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Johann de Bono
- The Institute of Cancer Research, 15 Cotswold Road, Sutton, Surrey, SM2 5NG, UK.,Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK
| |
Collapse
|
154
|
Lahdensuo K, Erickson A, Saarinen I, Seikkula H, Lundin J, Lundin M, Nordling S, Bützow A, Vasarainen H, Boström PJ, Taimen P, Rannikko A, Mirtti T. Loss of PTEN expression in ERG-negative prostate cancer predicts secondary therapies and leads to shorter disease-specific survival time after radical prostatectomy. Mod Pathol 2016; 29:1565-1574. [PMID: 27562498 DOI: 10.1038/modpathol.2016.154] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 11/09/2022]
Abstract
The clinical course of prostate cancer is highly variable. Current prognostic variables, stage, and Gleason score have limitations in assessing treatment regimens for individual patients, especially in the intermediate-risk group of Gleason score 7. ERG:TMPRSS2 fusion and loss of PTEN are some of the most common genetic alterations in prostate cancer. Immunohistochemistry of PTEN and ERG has generated interest as a promising method for more precise outcome prediction but requires further validation in population-based cohorts. We studied the predictive value of ERG and PTEN expression by immunohistochemistry in two large radical prostatectomy cohorts comprising 815 patients with extensive follow-up information. Clinical end points were initiation of secondary therapy, overall survival, and disease-specific survival. Predictions of clinical outcomes were also assessed according to androgen receptor (AR) activity. PTEN loss, especially in ERG-negative cancers, predicted initiation of secondary treatments and shortened disease-specific survival time, as well as stratifying Gleason score 7 patients into different prognostic groups with regard to secondary treatments and disease-specific survival. High AR immunoreactivity in ERG-negative cancers with PTEN loss predicted worse disease-specific survival. We also observed that in Gleason score 7 ERG-negative cases with PTEN loss and high AR expression have significantly shorter disease-specific survival time compared with ERG-positive cases. Our conclusion is that loss of PTEN is a strong determining factor for shorter disease-specific survival time and initiation of secondary therapies after radical prostatectomy. The predictive value of PTEN immunoreactivity is further accentuated in ERG-negative cancers with high AR expression. Negative PTEN expression, accompanied by ERG status, can be used to stratify patients with Gleason score 7 into different survival groups. Assessment of PTEN and ERG status could provide an additional tool for initial diagnostics when determining the prognosis and subsequent follow-up regimen for patients treated by radical prostatectomy.
Collapse
Affiliation(s)
- Kanerva Lahdensuo
- Department of Urology, Meilahti Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Andrew Erickson
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Irena Saarinen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Heikki Seikkula
- Department of Urology, University of Turku and Turku University Hospital, Turku, Finland
| | - Johan Lundin
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Mikael Lundin
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Stig Nordling
- Department of Pathology, HUSLAB and Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anna Bützow
- United Medix Laboratories, Pathology, Helsinki, Finland
| | - Hanna Vasarainen
- Department of Urology, Meilahti Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Peter J Boström
- Department of Urology, University of Turku and Turku University Hospital, Turku, Finland
| | - Pekka Taimen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Antti Rannikko
- Department of Urology, Meilahti Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland.,Department of Pathology, HUSLAB and Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
155
|
Silva MP, Barros-Silva JD, Ersvær E, Kildal W, Hveem TS, Pradhan M, Vieira J, Teixeira MR, Danielsen HE. Cancer Prognosis Defined by the Combined Analysis of 8q, PTEN and ERG. Transl Oncol 2016; 9:575-582. [PMID: 27916292 PMCID: PMC5143339 DOI: 10.1016/j.tranon.2016.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022] Open
Abstract
Overtreatment is a major concern in men diagnosed with prostate cancer. The aim of this study was to evaluate the combined prognostic role of three frequent molecular alterations in prostate cancer, namely relative 8q gain, ERG overexpression, and loss of PTEN expression, in a series of 136 patients with prostate cancer treated with prostatectomy and with a long follow-up. Fluorescent in situ hybridization was used to detect the relative copy number of 8q and immunohistochemistry was used for quantitative assessment of ERG and PTEN expression. During a median follow-up period of 117.8 months, 66 (49%) patients had disease recurrence. Relative 8q gain, ERG overexpression, and loss of PTEN expression were observed in 18%, 56%, and 33% of the cases, respectively. No association with patient recurrence-free survival was found for relative 8q gain or ERG overexpression on their own, whereas loss of PTEN expression was associated with worse recurrence-free survival (P=.006). Interestingly, in the subgroup of patients with normal PTEN expression, we found that the combined relative 8q gain/ERG overexpression is associated with high risk of recurrence (P=.008), suggesting that alternative mechanisms exist for progression into clinically aggressive disease. Additionally, in intermediate-risk patients with normal PTEN expression in their tumors, the combination of 8q gain/ERG overexpression was associated with a poor recurrence-free survival (P<.001), thus indicating independent prognostic value. This study shows that the combined analysis of 8q, ERG and PTEN contributes to an improved clinical outcome stratification of prostate cancer patients treated with radical prostatectomy.
Collapse
Affiliation(s)
- Maria P Silva
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal; Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - João D Barros-Silva
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal; Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Elin Ersvær
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Tarjei Sveinsgjerd Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Oslo, Norway; Department of Informatics, University of Oslo, 0310 Oslo, Norway
| | - Manohar Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Joana Vieira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Manuel R Teixeira
- Department of Genetics, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal; Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Oslo, Norway; Department of Informatics, University of Oslo, 0310 Oslo, Norway; Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
156
|
Wilczak W, Rashed S, Hube-Magg C, Kluth M, Simon R, Büscheck F, Clauditz TS, Grupp K, Minner S, Tsourlakis MC, Möller-Koop C, Graefen M, Adam M, Haese A, Wittmer C, Sauter G, Izbicki JR, Huland H, Schlomm T, Steurer S, Krech T, Lebok P. Up-regulation of mismatch repair genes MSH6, PMS2 and MLH1 parallels development of genetic instability and is linked to tumor aggressiveness and early PSA recurrence in prostate cancer. Carcinogenesis 2016; 38:19-27. [DOI: 10.1093/carcin/bgw116] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/06/2016] [Accepted: 10/31/2016] [Indexed: 11/13/2022] Open
|
157
|
Abstract
PURPOSE OF REVIEW The initial management of localized prostate cancer is increasingly complex with the identification of a growing number of prognostic subgroups. Molecular and genetic biomarkers have been proposed to help clinicians and patients navigate treatment decisions. RECENT FINDINGS Three commercially available tests, the Genomic Prostate score, Cell Cycle Progression score, and Genomic Classifier appear to currently have the most supporting data for their use in localized prostate cancer. All three have been shown to identify men at higher risk for poor outcome following radical prostatectomy in retrospective studies whereas the first two have also shown promise in addressing which men might be appropriate for active surveillance. Only the Genomic Classifier has data supporting its use as a predictive marker in addition to a prognostic marker. SUMMARY Over the past several years, the management of localized prostate cancer has seen the development of several novel biomarkers aimed at improving decision making. Although a lack of prospective validation makes it challenging to know how best to change management based on the results from any of the tests, the growing body of retrospective data suggests significant promise in this arena.
Collapse
|
158
|
Kim H, Skowronski J, Den RB. Prognostic outlier genes for enhanced prostate cancer treatment. Future Oncol 2016; 13:249-261. [PMID: 27728977 DOI: 10.2217/fon-2016-0223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
AIM To review the current landscape of outlier genes in the field of prostate cancer. METHODS A comprehensive review was performed. RESULTS Prostate cancer continues to be a significant worldwide health issue. In the era of personalized medicine, more emphasis is being placed on the ability to determine the timing, intensity and type of treatment, according to each patient's unique disease. Several commercial tests are available to determine the risk of aggressive prostate cancer based on genomic biomarkers and gene expression. Outlier genes represent a form of cancer classification that focuses on bimodal expression of a gene in a specific subset of patients. Outlier genes identified in prostate cancer include TMPRSS2-ERG, SPINK1, ScHLAP1, NVL, SMC4 and SQLE. CONCLUSION Classifying patient prostate cancers by outlier genes may allow for individualized cancer therapies and improved cancer therapy outcomes.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jenna Skowronski
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
159
|
Recent progress and perspectives on prostate cancer biomarkers. Int J Clin Oncol 2016; 22:214-221. [PMID: 27730440 PMCID: PMC5378754 DOI: 10.1007/s10147-016-1049-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/29/2016] [Indexed: 01/28/2023]
Abstract
The application of prostate-specific antigen (PSA) in prostate cancer (PC) screening, diagnosis, and prognosis has improved the clinical management of PC patients. However, the PSA assay has been faced with criticism due to its potential association with over-diagnosis and subsequent overtreatment of patients with indolent disease. The United States Preventive Services Task Force incited much debate over PSA-based screening in 2012 by recommending against this approach. However, the PSA assay remains the first-line tool for the early detection of PC. This debate highlights the unmet need for non-invasive PC biomarkers with greater sensitivity and specificity that are capable of distinguishing aggressive disease from indolent disease, predicting treatment response, and guiding treatment decisions. Recent investigations into putative PC biomarkers have focused on PSA isoform assays (prostate health index, 4-kallikurein panel), PC-associated genes in the urine (PCA3 and TMPRSS2-ERG), glycan-associated biomarkers (S2, 3PSA, GCNT1, and tri- and tetra-antennary serum N-glycans), and circulating tumor cells. Although substantial efforts to identify novel PC biomarkers that might replace PSA have been put forth, the majority of the putative PC biomarkers reported in the last few years are still under investigation or validation. This review provides an overview of the current state of PC biomarker research and focuses on a few promising PC biomarkers in development.
Collapse
|
160
|
Spratt DE, Zumsteg ZS, Feng FY, Tomlins SA. Translational and clinical implications of the genetic landscape of prostate cancer. Nat Rev Clin Oncol 2016; 13:597-610. [PMID: 27245282 PMCID: PMC5030163 DOI: 10.1038/nrclinonc.2016.76] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the past several years, analyses of data from high-throughput studies have elucidated many fundamental insights into prostate cancer biology. These insights include the identification of molecular alterations and subtypes that drive tumour progression, recurrent aberrations in signalling pathways, the existence of substantial intertumoural and intratumoural heterogeneity, Darwinian evolution in response to therapeutic pressures and the complicated multidirectional patterns of spread between primary tumours and metastatic sites. However, these concepts have not yet been fully translated into clinical tools to improve prognostication, prediction and personalization of treatment of patients with prostate cancer. The current and future clinical implications of 'omics' level knowledge is not only revolutionizing our understanding of prostate cancer biology, but is also shaping ongoing, and future clinical investigations and practice. In this Review, we summarize these advances, and the remaining challenges surrounding tumour heterogeneity and the ability to overcome treatment resistance are also described.
Collapse
Affiliation(s)
- Daniel E Spratt
- Department of Radiation Oncology, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | - Zachary S Zumsteg
- Department of Radiation Oncology, Cedars Sinai Medical Center, 8700 Beverly Blvd, West Hollywood, CA 90048, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, 1524 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109-2200, Ann Arbor, Michigan, USA
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
- Department of Urology, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
- Michigan Center for Translational Pathology, University of Michigan Medical School, 1524 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109-2200, Ann Arbor, Michigan, USA
| |
Collapse
|
161
|
Tosoian JJ, Almutairi F, Morais CL, Glavaris S, Hicks J, Sundi D, Humphreys E, Han M, De Marzo AM, Ross AE, Tomlins SA, Schaeffer EM, Trock BJ, Lotan TL. Prevalence and Prognostic Significance of PTEN Loss in African-American and European-American Men Undergoing Radical Prostatectomy. Eur Urol 2016; 71:697-700. [PMID: 27477529 DOI: 10.1016/j.eururo.2016.07.026] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/20/2016] [Indexed: 10/21/2022]
Abstract
African-American (AA) men have a higher risk of lethal prostate cancer (PCa) compared to European-American (EA) men. However, the molecular basis of this difference, if any, remains unclear. In EA PCa, PTEN loss, but not ERG rearrangement, has been associated with poor outcomes in most studies. Although ERG rearrangement is less common in AA compared to EA PCa, the relative frequency of PTEN loss and the association of PTEN/ERG molecular subtypes with outcomes is unknown for AA PCa. We examined PTEN/ERG status by immunohistochemistry in self-identified AA patients undergoing radical prostatectomy at Johns Hopkins with tumor tissue available on tissue microarray (TMA; n=169) and matched these cases by pathologic parameters to 169 EA patients from the same TMAs. The rate of PTEN loss was significantly lower in AA compared to EA PCa (18% vs 34%; p=0.001), similar to the lower rate of ERG expression (25% vs 51%; p<0.001). To examine the association of PTEN/ERG status with oncologic outcomes, we created an additional TMA of 87 AA tumors with Gleason score > 4 + 3 = 7. Among the total population of AA men with outcome data from all TMAs (n=222), PTEN loss was associated with higher risk of biochemical recurrence (hazard ratio [HR] 2.25, 95% confidence interval [CI] 1.33-3.82) and metastasis (HR 3.90, 95% CI 1.46-10.4) in multivariable models. PATIENT SUMMARY PTEN and ERG alterations in prostate cancer are less likely in African-American than in European-American men. However, PTEN loss remains associated with poor prostate cancer outcomes among African-American men.
Collapse
Affiliation(s)
- Jeffrey J Tosoian
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Fawaz Almutairi
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Carlos L Morais
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie Glavaris
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jessica Hicks
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Debasish Sundi
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Urology, UT MD Anderson Cancer Center, Houston, TX, USA
| | | | - Misop Han
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashley E Ross
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Scott A Tomlins
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Edward M Schaeffer
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Urology, Northwestern University, Chicago, IL, USA
| | - Bruce J Trock
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
162
|
Vascular morphology differentiates prostate cancer mortality risk among men with higher Gleason grade. Cancer Causes Control 2016; 27:1043-7. [PMID: 27379990 DOI: 10.1007/s10552-016-0782-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Higher Gleason grade is associated with prostate cancer mortality; however, there is significant heterogeneity in this association. We evaluated whether vessel morphology, a biomarker of angiogenesis, aided in distinguishing mortality risks among men with high Gleason grading. METHODS We characterized vessel morphology (area and irregularity) among 511 patients diagnosed with prostate cancer during 1986 to 2000, re-reviewed Gleason grade, and followed men through 2012. Men were grouped according to integrated vessel lumen irregularity and vessel area across Gleason grade. The more angiogenic group was identified as those with more irregular vessel lumen and smaller vessel area. Crude rates (95 % confidence intervals) and survival probability were estimated across Gleason grade and vessel morphology. RESULTS During a median 14-year follow-up, 62 men developed bone metastases or died of prostate cancer. Lethality rates were uniformly low within Gleason grade categories 6 and 7(3 + 4), regardless of vessel morphology. However, among men with Gleason grades of 7(4 + 3) or 8-10, the more angiogenic group was associated with fourfold higher risk of lethal outcomes compared to those with less angiogenic potential. Ten-year survival probability ranged from 95 to 74 % according to the extent of vessel morphology (p < 0.0001, log-rank test). CONCLUSIONS Vessel morphology may aid Gleason grading in predicting prostate cancer mortality risks among men diagnosed with high-grade Gleason cancers.
Collapse
|
163
|
Krstanoski Z, Vokac NK, Zagorac A, Pospihalj B, Munda M, Dzeroski S, Golouh R. TMPRSS2:ERG gene aberrations may provide insight into pT stage in prostate cancer. BMC Urol 2016; 16:35. [PMID: 27377958 PMCID: PMC4932765 DOI: 10.1186/s12894-016-0160-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 06/15/2016] [Indexed: 11/10/2022] Open
Abstract
Background TMPRSS2:ERG gene aberration may be a novel marker that improves risk stratification of prostate cancer before definitive cancer therapy, but studies have been inconclusive. Methods The study cohort consisted of 202 operable prostate cancer Slovenian patients who underwent laparoscopic radical prostatectomy. We retrospectively constructed tissue microarrays of their prostatic specimens for fluorescence in situ hybridization, with appropriate signals obtained in 148 patients for subsequent statistical analyses. Results The following genetic aberrations were found: TMPRSS2:ERG fusion, TMPRSS2 split (a non-ERG translocation) and ERG split (an ERG translocation without involvement of TMPRSS2). TMPRSS2:ERG gene fusion happened in 63 patients (42 %), TMPRSS2 split in 12 patients and ERG split in 8 patients. Association was tested between TMPRSS2:ERG gene fusion and several clinicopathological variables, i.e., pT stage, extended lymph node dissection status, and Gleason score, correcting for multiple comparisons. Only the association with pT stage was significant at p = 0.05: Of 62 patients with pT3 stage, 34 (55 %) had TMPRSS2:ERG gene fusion. In pT3 stage patients, stronger (but not significant) association between eLND status and TMPRSS2:ERG gene fusion was detected. We detected TMPRSS2:ERG gene fusion in 64 % of the pT3 stage patients where we did not perform an extended lymph node dissection. Conclusions Our results indicate that it is possible to predict pT3 stage at final histology from TMPRSS2:ERG gene fusion at initial core needle biopsy. FISH determination of TMPRSS2:ERG gene fusion may be particularly useful for patients scheduled to undergo a radical prostatectomy in order to improve oncological and functional results.
Collapse
Affiliation(s)
- Zoran Krstanoski
- Department of Urology, General Hospital Slovenj Gradec, Gosposvetska 1, 2380, Slovenj Gradec, Slovenia.
| | - Nadja Kokalj Vokac
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
| | - Andreja Zagorac
- Laboratory of Medical Genetics, University Medical Centre Maribor, Maribor, Slovenia
| | - Boris Pospihalj
- Division of Gynecology, Department of Gynecological Pathology and Cytology, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Miha Munda
- Institute for Anatomy, Histology and Embryology, Medical Faculty, University of Maribor, Maribor, Slovenia
| | | | - Rastko Golouh
- Institute of Pathology, Medical Faculty University of Maribor, Maribor, Slovenia
| |
Collapse
|
164
|
Abstract
Although most prostate cancer (PCa) cases are not life-threatening, approximately 293 000 men worldwide die annually due to PCa. These lethal cases are thought to be caused by coordinated genomic alterations that accumulate over time. Recent genome-wide analyses of DNA from subjects with PCa have revealed most, if not all, genetic changes in both germline and PCa tumor genomes. In this article, I first review the major, somatically acquired genomic characteristics of various subtypes of PCa. I then recap key findings on the relationships between genomic alterations and clinical parameters, such as biochemical recurrence or clinical relapse, metastasis and cancer-specific mortality. Finally, I outline the need for, and challenges with, validation of recent findings in prospective studies for clinical utility. It is clearer now than ever before that the landscape of somatically acquired aberrations in PCa is highlighted by DNA copy number alterations (CNAs) and TMPRSS2-ERG fusion derived from complex rearrangements, numerous single nucleotide variations or mutations, tremendous heterogeneity, and continuously punctuated evolution. Genome-wide CNAs, PTEN loss, MYC gain in primary tumors, and TP53 loss/mutation and AR amplification/mutation in advanced metastatic PCa have consistently been associated with worse cancer prognosis. With this recently gained knowledge, it is now an opportune time to develop DNA-based tests that provide more accurate patient stratification for prediction of clinical outcome, which will ultimately lead to more personalized cancer care than is possible at present.
Collapse
Affiliation(s)
- Wennuan Liu
- Program for Personalized Cancer Care, Research Institute, NorthShore University HealthSystem, Evanston, IL, USA
| |
Collapse
|
165
|
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
|
166
|
Roudier MP, Winters BR, Coleman I, Lam HM, Zhang X, Coleman R, Chéry L, True LD, Higano CS, Montgomery B, Lange PH, Snyder LA, Srivistava S, Corey E, Vessella RL, Nelson PS, Üren A, Morrissey C. Characterizing the molecular features of ERG-positive tumors in primary and castration resistant prostate cancer. Prostate 2016; 76:810-22. [PMID: 26990456 PMCID: PMC5589183 DOI: 10.1002/pros.23171] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/10/2016] [Indexed: 11/11/2022]
Abstract
BACKGROUND The TMPRSS2-ERG gene fusion is detected in approximately half of primary prostate cancers (PCa) yet the prognostic significance remains unclear. We hypothesized that ERG promotes the expression of common genes in primary PCa and metastatic castration-resistant PCa (CRPC), with the objective of identifying ERG-associated pathways, which may promote the transition from primary PCa to CRPC. METHODS We constructed tissue microarrays (TMA) from 127 radical prostatectomy specimens, 20 LuCaP patient-derived xenografts (PDX), and 152 CRPC metastases obtained immediately at time of death. Nuclear ERG was assessed by immunohistochemistry (IHC). To characterize the molecular features of ERG-expressing PCa, a subset of IHC confirmed ERG+ or ERG- specimens including 11 radical prostatectomies, 20 LuCaP PDXs, and 45 CRPC metastases underwent gene expression analysis. Genes were ranked based on expression in primary PCa and CRPC. Common genes of interest were targeted for IHC analysis and expression compared with biochemical recurrence (BCR) status. RESULTS IHC revealed that 43% of primary PCa, 35% of the LuCaP PDXs, and 18% of the CRPC metastases were ERG+ (12 of 48 patients [25%] had at least one ERG+ metastasis). Based on gene expression data and previous literature, two proteins involved in calcium signaling (NCALD, CACNA1D), a protein involved in inflammation (HLA-DMB), CD3 positive immune cells, and a novel ERG-associated protein, DCLK1 were evaluated in primary PCa and CRPC metastases. In ERG+ primary PCa, a weak association was seen with NCALD and CACNA1D protein expression. HLA-DMB association with ERG was decreased and CD3 cell number association with ERG was changed from positive to negative in CRPC metastases compared to primary PCa. DCLK1 was upregulated at the protein level in unpaired ERG+ primary PCa and CRPC metastases (P = 0.0013 and P < 0.0001, respectively). In primary PCa, ERG status or expression of targeted proteins was not associated with BCR-free survival. However, for primary PCa, ERG+DCLK1+ patients exhibited shorter time to BCR (P = 0.06) compared with ERG+DCLK1- patients. CONCLUSIONS This study examined ERG expression in primary PCa and CRPC. We have identified altered levels of inflammatory mediators associated with ERG expression. We determined expression of DCLK1 correlates with ERG expression and may play a role in primary PCa progression to metastatic CPRC. Prostate 76:810-822, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Martine P Roudier
- Department of Urology, University of Washington, Seattle, WA
- To whom all correspondence should be addressed: Colm Morrissey Ph.D. Genitourinary Cancer Research Laboratory, Department of Urology, Box 356510, University of Washington, Seattle, WA 98195, Telephone: 206-543-1461, Fax: 206-543-1146,
| | - Brian R Winters
- Department of Urology, University of Washington, Seattle, WA
- To whom all correspondence should be addressed: Colm Morrissey Ph.D. Genitourinary Cancer Research Laboratory, Department of Urology, Box 356510, University of Washington, Seattle, WA 98195, Telephone: 206-543-1461, Fax: 206-543-1146,
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Hung-Ming Lam
- Department of Urology, University of Washington, Seattle, WA
| | - Xiaotun Zhang
- Department of Urology, University of Washington, Seattle, WA
| | | | - Lisly Chéry
- Department of Urology, University of Washington, Seattle, WA
| | | | | | | | - Paul H. Lange
- Department of Urology, University of Washington, Seattle, WA
- Department of Veterans Affairs Medical Center, Seattle, WA
| | | | - Shiv Srivistava
- Uniformed Services University of the Health Sciences, Rockville, MD
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA
| | - Robert L. Vessella
- Department of Urology, University of Washington, Seattle, WA
- Department of Veterans Affairs Medical Center, Seattle, WA
| | - Peter S. Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Aykut Üren
- Georgetown University Medical Center, Washington, D. C
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA
| |
Collapse
|
167
|
Lotan TL, Wei W, Morais CL, Hawley ST, Fazli L, Hurtado-Coll A, Troyer D, McKenney JK, Simko J, Carroll PR, Gleave M, Lance R, Lin DW, Nelson PS, Thompson IM, True LD, Feng Z, Brooks JD. PTEN Loss as Determined by Clinical-grade Immunohistochemistry Assay Is Associated with Worse Recurrence-free Survival in Prostate Cancer. Eur Urol Focus 2016; 2:180-188. [PMID: 27617307 DOI: 10.1016/j.euf.2015.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND PTEN is the most commonly deleted tumor suppressor gene in primary prostate cancer (PCa) and its loss is associated with poor clinical outcomes and ERG gene rearrangement. OBJECTIVE We tested whether PTEN loss is associated with shorter recurrence-free survival (RFS) in surgically treated PCa patients with known ERG status. DESIGN SETTING AND PARTICIPANTS A genetically validated, automated PTEN immunohistochemistry (IHC) protocol was used for 1275 primary prostate tumors from the Canary Foundation retrospective PCa tissue microarray cohort to assess homogeneous (in all tumor tissue sampled) or heterogeneous (in a subset of tumor tissue sampled) PTEN loss. ERG status as determined by a genetically validated IHC assay was available for a subset of 938 tumors. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Associations between PTEN and ERG status were assessed using Fisher's exact test. Kaplan-Meier and multivariate weighted Cox proportional models for RFS were constructed. RESULTS AND LIMITATIONS When compared to intact PTEN, homogeneous (hazard ratio [HR] 1.66, p = 0.001) but not heterogeneous (HR 1.24, p = 0.14) PTEN loss was significantly associated with shorter RFS in multivariate models. Among ERG-positive tumors, homogeneous (HR 3.07, p < 0.0001) but not heterogeneous (HR 1.46, p = 0.10) PTEN loss was significantly associated with shorter RFS. Among ERG-negative tumors, PTEN did not reach significance for inclusion in the final multivariate models. The interaction term for PTEN and ERG status with respect to RFS did not reach statistical significance (p = 0.11) for the current sample size. CONCLUSIONS These data suggest that PTEN is a useful prognostic biomarker and that there is no statistically significant interaction between PTEN and ERG status for RFS. PATIENT SUMMARY We found that loss of the PTEN tumor suppressor gene in prostate tumors as assessed by tissue staining is correlated with shorter time to prostate cancer recurrence after radical prostatectomy.
Collapse
Affiliation(s)
- Tamara L Lotan
- Pathology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA; Oncology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Wei Wei
- MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos L Morais
- Pathology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Dean Troyer
- Pathology Department, Eastern Virginia Medical School, Norfolk, VA, USA; Urology Department, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | - Jeffrey Simko
- Pathology Department, University of California-San Francisco, San Francisco, CA, USA; Urology Department, University of California-San Francisco, San Francisco, CA, USA
| | - Peter R Carroll
- Urology Department, University of California-San Francisco, San Francisco, CA, USA
| | | | - Raymond Lance
- Urology Department, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Daniel W Lin
- Urology Department, University of Washington, Seattle, WA, USA
| | - Peter S Nelson
- Oncology Department, University of Washington, Seattle, WA, USA; Pathology Department, University of Washington, Seattle, WA, USA
| | - Ian M Thompson
- Urology Department, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lawrence D True
- Pathology Department, University of California-San Francisco, San Francisco, CA, USA
| | | | - James D Brooks
- Urology Department, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
168
|
Morais CL, Guedes LB, Hicks J, Baras AS, De Marzo AM, Lotan TL. ERG and PTEN status of isolated high-grade PIN occurring in cystoprostatectomy specimens without invasive prostatic adenocarcinoma. Hum Pathol 2016; 55:117-25. [PMID: 27189342 DOI: 10.1016/j.humpath.2016.04.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/14/2016] [Accepted: 04/22/2016] [Indexed: 11/17/2022]
Abstract
High-grade prostatic intraepithelial neoplasia (HGPIN) is widely believed to represent a precursor to invasive prostatic adenocarcinoma. However, recent molecular studies have suggested that retrograde spread of invasive adenocarcinoma into pre-existing prostatic ducts can morphologically mimic HGPIN. Thus, previous molecular studies characterizing morphologically identified HGPIN occurring in radical prostatectomies or needle biopsies with concurrent invasive carcinoma may be partially confounded by intraductal spread of invasive tumor. To assess ERG and PTEN status in HGPIN foci likely to represent true precursor lesions in the prostate, we studied isolated HGPIN occurring without associated invasive adenocarcinoma in cystoprostatectomies performed at Johns Hopkins between 2009 and 2014. Of 344 cystoprostatectomies, 33% (115/344) contained invasive prostatic adenocarcinoma in the partially submitted prostate (10 blocks/case on average) and were excluded from the study. Of the remaining cases without sampled cancer, 32% (73/229) showed 133 separate foci of HGPIN and were immunostained for ERG and PTEN using genetically validated protocols. Of foci of HGPIN with evaluable staining, 7% (8/107) were positive for ERG. PTEN loss was not seen in any HGPIN lesion (0/88). Because these isolated HGPIN foci at cystoprostatectomy are unlikely to represent retrograde spread of invasive tumor, our study suggests that ERG rearrangement, but not PTEN loss, is present in a minority of potential neoplastic precursor lesions in the prostate.
Collapse
Affiliation(s)
- Carlos L Morais
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231
| | - Liana B Guedes
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231
| | - Jessica Hicks
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231
| | | | - Angelo M De Marzo
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231; Urology, Johns Hopkins School of Medicine, Baltimore, MD 21231; Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21231
| | - Tamara L Lotan
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21231; Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21231.
| |
Collapse
|
169
|
Gaudreau PO, Stagg J, Soulières D, Saad F. The Present and Future of Biomarkers in Prostate Cancer: Proteomics, Genomics, and Immunology Advancements. BIOMARKERS IN CANCER 2016; 8:15-33. [PMID: 27168728 PMCID: PMC4859450 DOI: 10.4137/bic.s31802] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/29/2022]
Abstract
Prostate cancer (PC) is the second most common form of cancer in men worldwide. Biomarkers have emerged as essential tools for treatment and assessment since the variability of disease behavior, the cost and diversity of treatments, and the related impairment of quality of life have given rise to a need for a personalized approach. High-throughput technology platforms in proteomics and genomics have accelerated the development of biomarkers. Furthermore, recent successes of several new agents in PC, including immunotherapy, have stimulated the search for predictors of response and resistance and have improved the understanding of the biological mechanisms at work. This review provides an overview of currently established biomarkers in PC, as well as a selection of the most promising biomarkers within these particular fields of development.
Collapse
Affiliation(s)
- Pierre-Olivier Gaudreau
- Hematologist and Medical Oncologist, Notre-Dame Hospital, CHUM Research Center, Montreal, QC, Canada
| | - John Stagg
- Associate Professor, Department of Pharmacy, Cancer Axis—Montreal Cancer Institute, Montreal, QC, Canada
| | - Denis Soulières
- Hematologist and Medical Oncologist, Notre-Dame Hospital, CHUM Research Center, Montreal, QC, Canada
- Associate Professor, Department of Medicine, University of Montreal, QC, Canada
| | - Fred Saad
- Professor and Chief of Urology, CHUM—Pavillon R, Montreal, QC, Canada
| |
Collapse
|
170
|
Yang Z, Yu L, Wang Z. PCA3 and TMPRSS2-ERG gene fusions as diagnostic biomarkers for prostate cancer. Chin J Cancer Res 2016; 28:65-71. [PMID: 27041928 DOI: 10.3978/j.issn.1000-9604.2016.01.05] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The incidence of prostate cancer (PCa) is rising steadily among males in many countries. Serum prostate-specific antigen (PSA) is widely applied to clinical diagnosis and screening of PCa. However, the so-called grey area of PSA levels 4.0-10.0 ng/mL has a low specificity of 25-40% resulting in a high rate of negative biopsy and overtreatment. So in order to treat PCa patients in early stage, there is an urgent need for new biomarkers in PCa diagnosis. The PCA3 gene, a non-coding RNA (ncRNA) that is highly expressed in prostate cancer (PCa) cells, has been identified as a molecular biomarkers to detect PCa, of which PCA3 has already under clinical application. PCA3 is strongly overexpressed in malignant prostate tissue compared to benign or normal adjacent one. Newly, PCA3 is considered to be a promising biomarker in clinical diagnosis and targeted therapy. The diagnostic significance of PCA3, however, is awaiting further researches. Moreover, it has been demonstrated recently that TMPRSS2-ERG gene fusion is identified as the predominant genetic change in patients diagnosed with PCa. Recent study revealed that combination of the PCA3 and TMPRSS2-ERG gene fusion test optimizes PCa detection compared with that of single biomarker, which would lead to a considerable reduction of the number of prostate biopsies. In this review, we focused on the potential use of PCA3 and TMPRSS2-ERG gene fusion detection in the diagnosis of PCa.
Collapse
Affiliation(s)
- Zheng Yang
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Lu Yu
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Zhe Wang
- 1 State Key Laboratory of Cancer Biology, Department of Pathology, Xi Jing Hospital, Xi'an 710032, China ; 2 The First Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
171
|
Pre-diagnostic circulating sex hormone levels and risk of prostate cancer by ERG tumour protein expression. Br J Cancer 2016; 114:939-44. [PMID: 26986253 PMCID: PMC4984801 DOI: 10.1038/bjc.2016.61] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/31/2016] [Accepted: 02/17/2016] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Experimental studies have shown androgen receptor stimulation to facilitate formation of the TMPRSS2:ERG gene fusion in prostate cell lines. No study has tested whether higher pre-diagnostic circulating sex hormone levels in men increase risk of developing TMPRSS2:ERG-positive prostate cancer specifically. METHODS We conducted a nested case-control study of 200 prostate cancer cases and 1057 controls from the Physicians' Health Study and Health Professionals Follow-up Study. We examined associations between pre-diagnostic circulating levels of total testosterone, free testosterone, DHT, androstanediol glucuronide, estradiol, and SHBG and risk of prostate cancer by TMPRSS2:ERG status. TMPRSS2:ERG was estimated by ERG immunohistochemistry. We used multivariable unconditional polytomous logistic regression to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for risk of ERG-positive (n=94) and, separately, ERG-negative (n=106) disease. RESULTS Free testosterone was significantly associated with the risk of ERG-positive prostate cancer (OR: 1.37, 95% CI: 1.05-1.77), but not ERG-negative prostate cancer (OR: 1.09, 95% CI: 0.86-1.38) (Pdiff=0.17). None of the remaining hormones evaluated showed clear differential associations with ERG-positive vs ERG-negative disease. CONCLUSIONS These findings provide some suggestive evidence that higher pre-diagnostic free testosterone levels are associated with an increased risk of developing TMPRSS2:ERG-positive prostate cancer.
Collapse
|
172
|
Penney KL, Pettersson A, Shui IM, Graff RE, Kraft P, Lis RT, Sesso HD, Loda M, Mucci LA. Association of Prostate Cancer Risk Variants with TMPRSS2:ERG Status: Evidence for Distinct Molecular Subtypes. Cancer Epidemiol Biomarkers Prev 2016; 25:745-9. [PMID: 26941365 DOI: 10.1158/1055-9965.epi-15-1078] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Numerous genetic variants have been confirmed as prostate cancer risk factors. These variants may confer susceptibility to the development of specific molecular alterations during tumor initiation and progression. The TMPRSS2:ERG gene fusion occurs in roughly 50% of prostate cancers. Genetic risk variants may influence the development of this fusion. We sought to determine whether prostate cancer risk variants are differentially associated with TMPRSS2:ERG fusion-positive and negative cancer. METHODS In the Health Professionals Follow-up Study and Physicians' Health Study Tumor Cohort, we evaluated the associations of 39 prostate cancer risk SNPs with TMPRSS2:ERG fusion status, measured by ERG protein expression. Logistic regression was performed to generate OR and 95% confidence intervals. The primary outcome was ERG(+) (n = 227) versus ERG(-) (n = 260) prostate cancer. A secondary outcome was ERG(+) or ERG(-) cancer versus controls without cancer. RESULTS Six of 39 SNPs were significantly associated (P < 0.05) with ERG(+) versus ERG(-) disease. Three SNPs were exclusively associated with the risk of ERG(+), one with risk of ERG(-), and two with associations trending in opposite directions for ERG(+) and ERG(-) Only two significant SNPs would be expected by chance. CONCLUSIONS Prostate cancer genetic risk variants are differentially associated with the development of ERG(+) and ERG(-) prostate cancer. IMPACT Our findings suggest the molecular process of prostate carcinogenesis may be distinct for men with different underlying genetic predisposition. When examining risk factors for prostate cancer, the integration of molecular subtypes may enhance understanding of the etiology of this disease. Cancer Epidemiol Biomarkers Prev; 25(5); 745-9. ©2016 AACR.
Collapse
Affiliation(s)
- Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Irene M Shui
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rebecca E Graff
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rosina T Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Howard D Sesso
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. The Broad Institute, Cambridge, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
173
|
Graff RE, Pettersson A, Lis RT, Ahearn TU, Markt SC, Wilson KM, Rider JR, Fiorentino M, Finn S, Kenfield SA, Loda M, Giovannucci EL, Rosner B, Mucci LA. Dietary lycopene intake and risk of prostate cancer defined by ERG protein expression. Am J Clin Nutr 2016; 103:851-60. [PMID: 26817504 PMCID: PMC4763492 DOI: 10.3945/ajcn.115.118703] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 12/14/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND There is limited evidence that supports etiologically distinct molecular subtypes of prostate cancer, the identification of which may improve prevention. Given their antioxidant properties, we hypothesized that lycopene and tomato sauce may be especially protective against diseases harboring the common gene fusion transmembrane protease, serine 2 (TMPRSS2):v-ets avian erythroblastosis virus E26 oncogene homolog (ERG). OBJECTIVE We aimed to examine associations between estimated lycopene and tomato sauce intake and the risk of prostate cancer defined by ERG protein expression subtype. DESIGN Our study population consisted of a prospective cohort of 46,719 men from the Health Professionals Follow-Up Study. TMPRSS2:ERG was assessed by ERG immunohistochemistry on tumor tissue microarrays constructed from radical prostatectomy specimens. We used multivariable competing risk models to calculate HRs and 95% CIs for the risk of ERG-positive and, separately, ERG-negative disease. We implemented inverse probability weighting to account for evaluating ERG status only in surgically treated cases. RESULTS During 23 y of follow-up, 5543 men were diagnosed with prostate cancer, among whom 884 were assayed for ERG (426 ERG-positive). With inclusion of only the latter cases, increasing cumulative average tomato sauce intake was associated with a decreased risk of prostate cancer overall (≥2 servings/wk compared with <1 serving/mo; multivariable HR: 0.70; 95% CI: 0.52, 0.95; P-trend = 0.002). With respect to molecular subtypes, cumulative average tomato sauce intake was associated with a decreased risk of ERG-positive disease (HR: 0.54; 95% CI: 0.37, 0.81; P-trend = 0.004) but not with ERG-negative disease (HR: 0.96; 95% CI: 0.62, 1.50; P-trend = 0.10) (P-heterogeneity = 0.04). Increasing quintiles of lycopene intake were associated with a decreased risk of both subtypes (P-heterogeneity = 0.79). Inverse probability weighting did not materially change the results. CONCLUSIONS Our results lend some support to the hypothesis that prostate cancers that harbor TMPRSS2:ERG may be etiologically distinct from fusion-negative cancers. In particular, tomato sauce consumption may play a role in reducing TMPRSS2:ERG-positive disease.
Collapse
Affiliation(s)
- Rebecca E Graff
- Departments of Epidemiology, Departments ofEpidemiology and Biostatistics and
| | - Andreas Pettersson
- Departments of Epidemiology, Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Rosina T Lis
- Department of Pathology and Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Kathryn M Wilson
- Departments of Epidemiology, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jennifer R Rider
- Departments of Epidemiology, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michelangelo Fiorentino
- Departments of Epidemiology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA; Pathology Unit, Addarii Institute, S Orsola-Malpighi Hospital, Bologna, Italy; and
| | - Stephen Finn
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA; Department of Histopathology, Trinity College, Dublin, Ireland
| | - Stacey A Kenfield
- Departments of Epidemiology, Urology, University of California, San Francisco, San Francisco, CA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Massimo Loda
- Department of Pathology and Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Edward L Giovannucci
- Departments of Epidemiology, Nutrition, and Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Bernard Rosner
- Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lorelei A Mucci
- Departments of Epidemiology, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
174
|
Lu LI, Zhang H, Pang J, Hou GL, Lu MH, Gao X. ERG rearrangement as a novel marker for predicting the extra-prostatic extension of clinically localised prostate cancer. Oncol Lett 2016; 11:2532-2538. [PMID: 27073512 DOI: 10.3892/ol.2016.4282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/13/2016] [Indexed: 11/06/2022] Open
Abstract
Currently, there are no well-established preoperative clinicopathological parameters for predicting extra-prostatic extension (EPE) in patients with clinically localised prostate cancer (PCa). The transmembrane protease serine 2 (TMPRSS2)-ETS-related gene (ERG) fusion gene is a specific biomarker of PCa and is considered a prognostic predictor. The aim of the present study was to assess the value of this marker for predicting EPE in patients with clinically localised PCa. In total, 306 PCa patients with clinically localised disease, including 220 patients (71.9%) with organ-confined disease and 86 EPE cases (28.1%), were included in the study. Receiver operating characteristic curves and logistic regression were employed to establish the optimal cut-off value and to investigate whether ERG rearrangement was an independent predictor for the EPE of clinically localised PCa. A leave-one-out cross-validation (LOOCV) model was implemented to validate the predictive power of ERG rearrangement. An increase in ERG rearrangements was identified to be associate'd with EPE, and the optimal cut-off for predicting EPE was determined to be 2.25%, with a sensitivity of 70.24% [95% confidence interval (CI), 62.6-78.9%], a specificity of 80.43% (95% CI, 75.4-85.1%), and an area under the curve (AUC) of 0.781 (95% CI, 0.730-0.826). In the LOOCV model, ERG rearrangement also demonstrated good performance for predicting EPE (sensitivity, 76.923%; specificity, 71.429%; 95% CI for AUC, 0.724-0.958). In addition, a high Gleason score (≥7) and a cT2c classification upon biopsy were independent factors for EPE.
Collapse
Affiliation(s)
- L I Lu
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Hao Zhang
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jun Pang
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Guo-Liang Hou
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Min-Hua Lu
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xin Gao
- Department of Urology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
175
|
Bakkar A, Alshalalfa M, Petersen LF, Abou-Ouf H, Al-Mami A, Hegazy SA, Feng F, Alhajj R, Bijian K, Alaoui-Jamali MA, Bismar TA. microRNA 338-3p exhibits tumor suppressor role and its down-regulation is associated with adverse clinical outcome in prostate cancer patients. Mol Biol Rep 2016; 43:229-40. [DOI: 10.1007/s11033-016-3948-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 02/08/2016] [Indexed: 02/07/2023]
|
176
|
Linn DE, Penney KL, Bronson RT, Mucci LA, Li Z. Deletion of Interstitial Genes between TMPRSS2 and ERG Promotes Prostate Cancer Progression. Cancer Res 2016; 76:1869-81. [PMID: 26880803 DOI: 10.1158/0008-5472.can-15-1911] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/23/2016] [Indexed: 11/16/2022]
Abstract
TMPRSS2-ERG gene fusions that occur frequently in human prostate cancers can be generated either through insertional chromosomal rearrangement or by intrachromosomal deletion. Genetically, a key difference between these two mechanisms is that the latter results in deletion of a ∼3-Mb interstitial region containing genes with unexplored roles in prostate cancer. In this study, we characterized two mouse models recapitulating TMPRSS2-ERG insertion or deletion events in the background of prostate-specific PTEN deficiency. We found that only the mice that lacked the interstitial region developed prostate adenocarcinomas marked by poor differentiation and epithelial-to-mesenchymal transition. Mechanistic investigations identified several interstitial genes, including Ets2 and Bace2, whose reduced expression correlated in the gene homologs in human prostate cancer with biochemical relapse and lethal disease. Accordingly, PTEN-deficient mice with prostate-specific knockout of Ets2 exhibited marked progression of prostate adenocarcinomas that was partly attributed to activation of MAPK signaling. Collectively, our findings established that Ets2 is a tumor suppressor gene in prostate cancer, and its loss along with other genes within the TMPRSS2-ERG interstitial region contributes to disease progression. Cancer Res; 76(7); 1869-81. ©2016 AACR.
Collapse
Affiliation(s)
- Douglas E Linn
- Division of Genetics, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kathryn L Penney
- Channing Division of Network Medicine, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | | | - Lorelei A Mucci
- Channing Division of Network Medicine, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
177
|
|
178
|
Novel Research on Fusion Genes and Next-Generation Sequencing. Prostate Cancer 2016. [DOI: 10.1016/b978-0-12-800077-9.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
179
|
Carozzi F, Tamburrino L, Bisanzi S, Marchiani S, Paglierani M, Di Lollo S, Crocetti E, Buzzoni C, Burroni E, Greco L, Baldi E, Sani C. Are biomarkers evaluated in biopsy specimens predictive of prostate cancer aggressiveness? J Cancer Res Clin Oncol 2016; 142:201-12. [PMID: 26210155 DOI: 10.1007/s00432-015-2015-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 07/07/2015] [Indexed: 01/29/2023]
Abstract
PURPOSE To evaluate biomarkers involved in biological pathways for prostate cancer (PCa) progression, measured in biopsy specimens, in order to distinguish patients at higher risk for fatal PCa and thus improve the initial management of disease. METHODS Retrospective case-control study. In 129 PCa patients who underwent ultrasound-guided needle prostate biopsy and subsequent radical prostatectomy from 1987 to 1999 at the University Hospital of Careggi, we evaluated: (1) mRNA expression of the serine 2 (TMPRSS2): erythroblastosis virus E26 oncogene homolog (ERG); (2) expression of matrix metalloproteinases (MMP)-2 and 9 (epithelial and stromal); (3) expression of androgen receptor; (4) expression of prognostic marker Ki67 (MIB1); (5) presence and typing of human papilloma virus; (6) DNA methylation of CpG islands of several genes involved in PCa progression. RESULTS The cohort consists of 38 cases (patients with PCa and died of PCa within 10 years from diagnosis) and 91 controls (patients with PCa but alive 10 years after diagnosis). Gleason bioptic score, epithelial MMP expression and SERPINB5 methylation correlated with statistically significant increase in death risk OR. Compared with patients with high level of MMP, patients with low level of MMP had OR for specific death 4.78 times higher (p = 0.0066). After adjustment for age and Gleason score, none of the investigated biomarkers showed increased OR for PCa death. CONCLUSIONS Our preliminary results suggest that evaluation, in prostate biopsy specimens, of a panel of biomarkers known to be involved in PCa progression is poorly indicative of tumor outcome.
Collapse
Affiliation(s)
- Francesca Carozzi
- Laboratory Cancer Prevention, Cancer Prevention and Research Institute (ISPO), Via Cosimo il Vecchio 2, 50139, Florence, Italy
| | - Lara Tamburrino
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Simonetta Bisanzi
- Laboratory Cancer Prevention, Cancer Prevention and Research Institute (ISPO), Via Cosimo il Vecchio 2, 50139, Florence, Italy
| | - Sara Marchiani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Milena Paglierani
- DAI Biomedicina SOD Istologia Patologica e Diagnostica Molecolare, AOU Careggi, Florence, Italy
| | - Simonetta Di Lollo
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Emanuele Crocetti
- Clinical and Descriptive Epidemiology Unit, Cancer Prevention and Research Institute (ISPO), Florence, Italy
| | - Carlotta Buzzoni
- Clinical and Descriptive Epidemiology Unit, Cancer Prevention and Research Institute (ISPO), Florence, Italy
| | - Elena Burroni
- Laboratory Cancer Prevention, Cancer Prevention and Research Institute (ISPO), Via Cosimo il Vecchio 2, 50139, Florence, Italy
| | - Luana Greco
- Laboratory Cancer Prevention, Cancer Prevention and Research Institute (ISPO), Via Cosimo il Vecchio 2, 50139, Florence, Italy
| | - Elisabetta Baldi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Cristina Sani
- Laboratory Cancer Prevention, Cancer Prevention and Research Institute (ISPO), Via Cosimo il Vecchio 2, 50139, Florence, Italy.
| |
Collapse
|
180
|
Sanguedolce F, Cormio A, Brunelli M, D'Amuri A, Carrieri G, Bufo P, Cormio L. Urine TMPRSS2: ERG Fusion Transcript as a Biomarker for Prostate Cancer: Literature Review. Clin Genitourin Cancer 2015; 14:117-21. [PMID: 26774207 DOI: 10.1016/j.clgc.2015.12.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/25/2015] [Accepted: 12/09/2015] [Indexed: 11/25/2022]
Abstract
Prostate cancer (PCa) is one of the most common male malignancies. Serum prostate-specific antigen (PSA) is one of the most valuable biomarkers in tumor biology and remains the standard marker in detecting and monitoring PCa. However, the high number of serum PSA false positive and false negative results make the identification of novel biomarkers extremely welcome to improve our diagnostic accuracy in detecting PCa and distinguishing the aggressive from the indolent ones. In this study, we analyzed the current role of urinary gene fusion transcripts involving v-ets erythroblastosis virus E26 oncogene homolog, commonly known as ERG, and the androgen-regulated gene transmembrane protease, serine 2 (TMPRSS2), as a biomarker for PCa. Used as a single marker, urinary TMPRSS2:ERG has low sensitivity but high specificity. However, its combination with the other urinary marker PCa antigen 3 (PCA3) has been reported to provide high specificity and sensitivity. Finally, a commercially available assay combining serum PSA with urinary PCA3 and TMPRSS2:ERG provides a 90% specificity and 80% sensitivity in diagnosing PCa. Urinary TMPRSS2:ERG also seems to be indicative of PCa aggressiveness upon biopsy. Should these findings be confirmed in larger studies, urinary TMPRSS2:ERG might become a valuable test not only for diagnosing PCa but also for distinguishing the aggressive tumors from the indolent ones.
Collapse
Affiliation(s)
| | - Antonella Cormio
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, University of Verona, Verona, Italy
| | | | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| | - Pantaleo Bufo
- Department of Pathology, University of Foggia, Foggia, Italy
| | - Luigi Cormio
- Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| |
Collapse
|
181
|
Biomarkers for prostate cancer: present challenges and future opportunities. Future Sci OA 2015; 2:FSO72. [PMID: 28031932 PMCID: PMC5137959 DOI: 10.4155/fso.15.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 01/30/2023] Open
Abstract
Prostate cancer (PCa) has variable biological potential with multiple treatment options. A more personalized approach, therefore, is needed to better define men at higher risk of developing PCa, discriminate indolent from aggressive disease and improve risk stratification after treatment by predicting the likelihood of progression. This may improve clinical decision-making regarding management, improve selection for active surveillance protocols and minimize morbidity from treatment. Discovery of new biomarkers associated with prostate carcinogenesis present an opportunity to provide patients with novel genetic signatures to better understand their risk of developing PCa and help forecast their clinical course. In this review, we examine the current literature evaluating biomarkers in PCa. We also address current limitations and present several ideas for future studies.
Collapse
|
182
|
Geybels MS, Alumkal JJ, Luedeke M, Rinckleb A, Zhao S, Shui IM, Bibikova M, Klotzle B, van den Brandt PA, Ostrander EA, Fan JB, Feng Z, Maier C, Stanford JL. Epigenomic profiling of prostate cancer identifies differentially methylated genes in TMPRSS2:ERG fusion-positive versus fusion-negative tumors. Clin Epigenetics 2015; 7:128. [PMID: 26692910 PMCID: PMC4676897 DOI: 10.1186/s13148-015-0161-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022] Open
Abstract
Background About half of all prostate cancers harbor the TMPRSS2:ERG (T2E) gene fusion. While T2E-positive and T2E-negative tumors represent specific molecular subtypes of prostate cancer (PCa), previous studies have not yet comprehensively investigated how these tumor subtypes differ at the epigenetic level. We therefore investigated epigenome-wide DNA methylation profiles of PCa stratified by T2E status. Results The study included 496 patients with clinically localized PCa who had a radical prostatectomy as primary treatment for PCa. Fluorescence in situ hybridization (FISH) “break-apart” assays were used to determine tumor T2E-fusion status, which showed that 266 patients (53.6 %) had T2E-positive PCa. The study showed global DNA methylation differences between tumor subtypes. A large number of differentially methylated CpG sites were identified (false-discovery rate [FDR] Q-value <0.00001; n = 27,876) and DNA methylation profiles accurately distinguished between tumor T2E subgroups. A number of top-ranked differentially methylated CpGs in genes (FDR Q-values ≤1.53E−29) were identified: C3orf14, CACNA1D, GREM1, KLK10, NT5C, PDE4D, RAB40C, SEPT9, and TRIB2, several of which had a corresponding alteration in mRNA expression. These genes may have various roles in the pathogenesis of PCa, and the calcium-channel gene CACNA1D is a known ERG-target. Analysis of The Cancer Genome Atlas (TCGA) data provided confirmatory evidence for our findings. Conclusions This study identified substantial differences in DNA methylation profiles of T2E-positive and T2E-negative tumors, thereby providing further evidence that different underlying oncogenic pathways characterize these molecular subtypes. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0161-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milan S Geybels
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Joshi J Alumkal
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR USA
| | - Manuel Luedeke
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Antje Rinckleb
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Shanshan Zhao
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NC Research Triangle Park, USA
| | - Irene M Shui
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | | | | | - Piet A van den Brandt
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Elaine A Ostrander
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD USA
| | - Jian-Bing Fan
- Illumina, Inc., San Diego, CA USA ; Present Address: AnchorDx Corp., Guangzhou, 510300 People's Republic of China
| | | | - Christiane Maier
- Institute of Human Genetics and Department of Urology, Faculty of Medicine, University of Ulm, Ulm, Germany
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA USA ; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA USA
| |
Collapse
|
183
|
Prognostic relevance of proliferation markers (Ki-67, PHH3) within the cross-relation of ERG translocation and androgen receptor expression in prostate cancer. Pathology 2015; 47:629-36. [DOI: 10.1097/pat.0000000000000320] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
184
|
Gerke TA, Martin NE, Ding Z, Nuttall EJ, Stack EC, Giovannucci E, Lis RT, Stampfer MJ, Kantoff PW, Parmigiani G, Loda M, Mucci LA. Evaluating a 4-marker signature of aggressive prostate cancer using time-dependent AUC. Prostate 2015; 75:1926-33. [PMID: 26469352 PMCID: PMC4831584 DOI: 10.1002/pros.23090] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 08/25/2015] [Indexed: 11/05/2022]
Abstract
BACKGROUND We previously identified a protein tumor signature of PTEN, SMAD4, SPP1, and CCND1 that, together with clinical features, was associated with lethal outcomes among prostate cancer patients. In the current study, we sought to validate the molecular model using time-dependent measures of AUC and predictive values for discriminating lethal from non-lethal prostate cancer. METHODS Using data from the initial study, we fit survival models for men with prostate cancer who were participants in the Physicians' Health Study (PHS; n = 276). Based on these models, we generated prognostic risk scores in an independent population, the Health Professionals Follow-up Study (HPFS; n = 347) to evaluate external validity. In each cohort, men were followed prospectively from cancer diagnosis through 2011 for development of distant metastasis or cancer mortality. We measured protein tumor expression of PTEN, SMAD4, SPP1, and CCND1 on tissue microarrays. RESULTS During a median of 11.9 and 14.3 years follow-up in the PHS and HPFS cohorts, 24 and 32 men (9%) developed lethal disease. When used as a prognostic factor in a new population, addition of the four markers to clinical variables did not improve discriminatory accuracy through 15 years of follow-up. CONCLUSIONS Although the four markers have been identified as key biological mediators in metastatic progression, they do not provide independent, long-term prognostic information beyond clinical factors when measured at diagnosis. This finding may underscore the broad heterogeneity in aggressive prostate tumors and highlight the challenges that may result from overfitting in discovery-based research.
Collapse
Affiliation(s)
- Travis A. Gerke
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Neil E. Martin
- Department of Radiation Oncology, Brigham and Women’s Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zhihu Ding
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth J. Nuttall
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Edward C. Stack
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Edward Giovannucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Rosina T. Lis
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Meir J. Stampfer
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Phillip W. Kantoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Giovanni Parmigiani
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Massimo Loda
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
185
|
Ahearn TU, Pettersson A, Ebot EM, Gerke T, Graff RE, Morais CL, Hicks JL, Wilson KM, Rider JR, Sesso HD, Fiorentino M, Flavin R, Finn S, Giovannucci EL, Loda M, Stampfer MJ, De Marzo AM, Mucci LA, Lotan TL. A Prospective Investigation of PTEN Loss and ERG Expression in Lethal Prostate Cancer. J Natl Cancer Inst 2015; 108:djv346. [PMID: 26615022 DOI: 10.1093/jnci/djv346] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 10/19/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND PTEN is a tumor suppressor frequently deleted in prostate cancer that may be a useful prognostic biomarker. However, the association of PTEN loss with lethal disease has not been tested in a large, predominantly surgically treated cohort. METHODS In the Health Professionals Follow-up Study and Physicians' Health Study, we followed 1044 incident prostate cancer cases diagnosed between 1986 and 2009 for cancer-specific and all-cause mortality. A genetically validated PTEN immunohistochemistry (IHC) assay was performed on tissue microarrays (TMAs). TMPRSS2:ERG status was previously assessed in a subset of cases by a genetically validated IHC assay for ERG. Cox proportional hazards models adjusting for age and body mass index at diagnosis, Gleason grade, and clinical or pathologic TNM stage were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the association with lethal disease. All statistical tests were two-sided. RESULTS On average, men were followed 11.7 years, during which there were 81 lethal events. Sixteen percent of cases had complete PTEN loss in all TMA cores and 9% had heterogeneous PTEN loss across cores. After adjustment for clinical-pathologic variables, complete PTEN loss was associated with lethal progression (HR = 1.8, 95% CI = 1.2 to 2.9). The association of PTEN loss (complete or heterogeneous) with lethal progression was only among men with ERG-negative (HR = 3.1, 95% CI = 1.7 to 5.7) but not ERG-positive (HR = 1.2, 95% CI = 0.7 to 2.2) tumors. CONCLUSIONS PTEN loss is independently associated with increased risk of lethal progression, particularly in the ERG fusion-negative subgroup. These validated and inexpensive IHC assays may be useful for risk stratification in prostate cancer.
Collapse
Affiliation(s)
- Thomas U Ahearn
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Andreas Pettersson
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ericka M Ebot
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Travis Gerke
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rebecca E Graff
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carlos L Morais
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jessica L Hicks
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kathryn M Wilson
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer R Rider
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Howard D Sesso
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michelangelo Fiorentino
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard Flavin
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Stephen Finn
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Edward L Giovannucci
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Massimo Loda
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Meir J Stampfer
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Angelo M De Marzo
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lorelei A Mucci
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tamara L Lotan
- Department of Epidemiology (TUA, AP, EME, TG, REG, KMW, JRR, HDS, ELG, MJS, LAM) and Department of Nutrition (ELG, MJS), Harvard T. H. Chan School of Public Health, Boston, MA; Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (AP); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (REG); Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (CLM, JLH, AMDM, TLL); Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (JRR, ELG, MJS, LAM, KMW); Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA (HDS); Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy (MF); Department of Histopathology Research, Trinity College, Dublin, Ireland (RF, SF); Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA (ML); Department of Oncology (AMDM, TLL) and Department of Urology (ADMD), Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
186
|
The Molecular Taxonomy of Primary Prostate Cancer. Cell 2015; 163:1011-25. [PMID: 26544944 PMCID: PMC4695400 DOI: 10.1016/j.cell.2015.10.025] [Citation(s) in RCA: 2300] [Impact Index Per Article: 230.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/14/2015] [Accepted: 10/06/2015] [Indexed: 12/12/2022]
Abstract
There is substantial heterogeneity among primary prostate cancers, evident in the spectrum of molecular abnormalities and its variable clinical course. As part of The Cancer Genome Atlas (TCGA), we present a comprehensive molecular analysis of 333 primary prostate carcinomas. Our results revealed a molecular taxonomy in which 74% of these tumors fell into one of seven subtypes defined by specific gene fusions (ERG, ETV1/4, and FLI1) or mutations (SPOP, FOXA1, and IDH1). Epigenetic profiles showed substantial heterogeneity, including an IDH1 mutant subset with a methylator phenotype. Androgen receptor (AR) activity varied widely and in a subtype-specific manner, with SPOP and FOXA1 mutant tumors having the highest levels of AR-induced transcripts. 25% of the prostate cancers had a presumed actionable lesion in the PI3K or MAPK signaling pathways, and DNA repair genes were inactivated in 19%. Our analysis reveals molecular heterogeneity among primary prostate cancers, as well as potentially actionable molecular defects.
Collapse
|
187
|
Soulié M, Portier G, Salomon L. [Oncological principles for local control of primary tumor]. Prog Urol 2015; 25:918-32. [PMID: 26519960 DOI: 10.1016/j.purol.2015.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 07/30/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Review of the databases of carcinogenesis and the principles of local control of the primary tumor in order to decrease the risk of tumor progression and predict metastatic behavior. MATERIALS AND METHODS Review of the literature using Medline databases based on scientific relevance. Research was centered on the characteristics of solid tumor development, the basics of local control of the primary tumor, latest advance in genomics and the oncological principles applied on prostate cancer surgery. RESULTS The cornerstone in order to cure a local or locally advanced cancer is to eradicate the primary tumor. This should be done using effective methods that can assure local control, decrease the risk of progression and metastasis. The oncological surgery is the most important step in order to have this tumor control, beside radiotherapy and systemic therapy associated. In localized prostate cancer, surgery remains the gold standard between the multiple therapeutic modalities proposed. CONCLUSION The local control of solid malignant tumor is primordial in order to change the natural history of the disease and decrease its risk of progression. This is the goal of oncological surgery, and starting from these principles radical prostatectomy was favored.
Collapse
Affiliation(s)
- M Soulié
- Département d'urologie-andrologie-transplantation rénale, CHU Rangueil, 1, avenue Jean-Poulhès, 31059 Toulouse cedex 9, France.
| | - G Portier
- Service de chirurgie digestive, CHU Purpan, place Baylac, 31059 Toulouse cedex 9, France
| | - L Salomon
- Service d'urologie et de transplantation rénale et pancréatique, CHU Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil cedex, France
| |
Collapse
|
188
|
Pellegrini KL, Sanda MG, Moreno CS. RNA biomarkers to facilitate the identification of aggressive prostate cancer. Mol Aspects Med 2015; 45:37-46. [PMID: 26022941 PMCID: PMC4637232 DOI: 10.1016/j.mam.2015.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/20/2015] [Indexed: 02/07/2023]
Abstract
A large number of men are diagnosed with prostate cancer each year, but many will not experience morbidity or mortality as a result of their cancers. Therefore, biomarkers for prostate cancer are necessary to carefully select patients for initial diagnostic biopsy or to facilitate care decisions for men who have already been diagnosed with prostate cancer. RNA-based approaches to biomarker discovery allow the investigation of non-coding RNAs, gene fusion transcripts, splice variants, and multi-gene expression panels in tissue, urine, or blood as opportunities to improve care decisions. This review focuses on RNA biomarkers that are available as commercial assays, and therefore already available for potential clinical use, as well as providing an overview of newer RNA biomarkers that are in earlier stages of clinical development.
Collapse
Affiliation(s)
- Kathryn L Pellegrini
- Department of Urology, Emory University School of Medicine, Winship Cancer Institute at Emory University, Atlanta, GA 30322, USA
| | - Martin G Sanda
- Department of Urology, Emory University School of Medicine, Winship Cancer Institute at Emory University, Atlanta, GA 30322, USA
| | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Winship Cancer Institute at Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
189
|
Berg KD, Røder MA, Thomsen FB, Vainer B, Gerds TA, Brasso K, Iversen P. The predictive value of ERG protein expression for development of castration-resistant prostate cancer in hormone-naïve advanced prostate cancer treated with primary androgen deprivation therapy. Prostate 2015; 75:1499-509. [PMID: 26053696 DOI: 10.1002/pros.23026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/30/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Biomarkers predicting response to primary androgen deprivation therapy (ADT) and risk of castration-resistant prostate cancer (CRPC) is lacking. We aimed to analyse the predictive value of ERG expression for development of CRPC. METHODS In total, 194 patients with advanced and/or metastatic prostate cancer (PCa) treated with first-line castration-based ADT were included. ERG protein expression was analysed in diagnostic specimens using immunohistochemistry (anti-ERG, EPR3864). Time to CRPC was compared between ERG subgroups using multiple cause-specific Cox regression stratified on ERG-status. Risk reclassification and time-dependent area under the ROC curves were used to assess the discriminative ability of ERG-status. Time to PSA-nadir, proportion achieving PSA-nadir ≤0.2 ng/ml, and risk of PCa-specific death were secondary endpoints. RESULTS Median follow-up was 6.8 years (IQR: 4.9-7.3). In total, 105 patients (54.1%) were ERG-positive and 89 (45.9%) were ERG-negative. No difference in risk of CRPC was observed between ERG subgroups (P = 0.51). Median time to CRPC was 3.9 years (95%CI: 3.2-5.1) and 4.5 years (95%CI: 2.3-not reached) in the ERG-positive and ERG-negative group, respectively. Compared to a model omitting ERG-status, the ERG-stratified model showed comparable AUC values 1 year (77.6% vs. 78.0%, P = 0.82), 2 years (71.7% vs. 71.8%, P = 0.85), 5 years (68.5% vs. 69.9%, P = 0.32), and 8 years (67.9% vs. 71.4%, P = 0.21) from ADT initiation. No differences in secondary endpoints were observed. CONCLUSIONS ERG expression was not associated with risk of CRPC suggesting that ERG is not a candidate biomarker for predicting response to primary ADT in patients diagnosed with advanced and/or metastatic PCa.
Collapse
Affiliation(s)
- Kasper D Berg
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin A Røder
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Frederik B Thomsen
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ben Vainer
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Gerds
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Brasso
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peter Iversen
- Copenhagen Prostate Cancer Center, Department of Urology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
190
|
Tomlins SA, Alshalalfa M, Davicioni E, Erho N, Yousefi K, Zhao S, Haddad Z, Den RB, Dicker AP, Trock BJ, DeMarzo AM, Ross AE, Schaeffer EM, Klein EA, Magi-Galluzzi C, Karnes RJ, Jenkins RB, Feng FY. Characterization of 1577 primary prostate cancers reveals novel biological and clinicopathologic insights into molecular subtypes. Eur Urol 2015; 68:555-67. [PMID: 25964175 PMCID: PMC4562381 DOI: 10.1016/j.eururo.2015.04.033] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/21/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Prostate cancer (PCa) molecular subtypes have been defined by essentially mutually exclusive events, including ETS gene fusions (most commonly involving ERG) and SPINK1 overexpression. Clinical assessment may aid in disease stratification, complementing available prognostic tests. OBJECTIVE To determine the analytical validity and clinicopatholgic associations of microarray-based molecular subtyping. DESIGN, SETTING, AND PARTICIPANTS We analyzed Affymetrix GeneChip expression profiles for 1577 patients from eight radical prostatectomy cohorts, including 1351 cases assessed using the Decipher prognostic assay (GenomeDx Biosciences, San Diego, CA, USA) performed in a laboratory with Clinical Laboratory Improvements Amendment certification. A microarray-based (m-) random forest ERG classification model was trained and validated. Outlier expression analysis was used to predict other mutually exclusive non-ERG ETS gene rearrangements (ETS(+)) or SPINK1 overexpression (SPINK1(+)). OUTCOME MEASUREMENTS Associations with clinical features and outcomes by multivariate logistic regression analysis and receiver operating curves. RESULTS AND LIMITATIONS The m-ERG classifier showed 95% accuracy in an independent validation subset (155 samples). Across cohorts, 45% of PCas were classified as m-ERG(+), 9% as m-ETS(+), 8% as m-SPINK1(+), and 38% as triple negative (m-ERG(-)/m-ETS(-)/m-SPINK1(-)). Gene expression profiling supports three underlying molecularly defined groups: m-ERG(+), m-ETS(+), and m-SPINK1(+)/triple negative. On multivariate analysis, m-ERG(+) tumors were associated with lower preoperative serum prostate-specific antigen and Gleason scores, but greater extraprostatic extension (p<0.001). m-ETS(+) tumors were associated with seminal vesicle invasion (p=0.01), while m-SPINK1(+)/triple negative tumors had higher Gleason scores and were more frequent in Black/African American patients (p<0.001). Clinical outcomes were not significantly different among subtypes. CONCLUSIONS A clinically available prognostic test (Decipher) can also assess PCa molecular subtypes, obviating the need for additional testing. Clinicopathologic differences were found among subtypes based on global expression patterns. PATIENT SUMMARY Molecular subtyping of prostate cancer can be achieved using extra data generated from a clinical-grade, genome-wide expression-profiling prognostic assay (Decipher). Transcriptomic and clinical analysis support three distinct molecular subtypes: (1) m-ERG(+), (2) m-ETS(+), and (3) m-SPINK1(+)/triple negative (m-ERG(-)/m-ETS(-)/m-SPINK1(-)). Incorporation of subtyping into a clinically available assay may facilitate additional applications beyond routine prognosis.
Collapse
Affiliation(s)
- Scott A Tomlins
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | | | - Elai Davicioni
- GenomeDx Bioscience Inc., Vancouver, British Columbia, Canada
| | - Nicholas Erho
- GenomeDx Bioscience Inc., Vancouver, British Columbia, Canada
| | - Kasra Yousefi
- GenomeDx Bioscience Inc., Vancouver, British Columbia, Canada
| | - Shuang Zhao
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zaid Haddad
- GenomeDx Bioscience Inc., Vancouver, British Columbia, Canada
| | - Robert B Den
- Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam P Dicker
- Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | - Bruce J Trock
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Angelo M DeMarzo
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashley E Ross
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Edward M Schaeffer
- James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Eric A Klein
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Robert B Jenkins
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Felix Y Feng
- 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 Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
191
|
Morais CL, Herawi M, Toubaji A, Albadine R, Hicks J, Netto GJ, De Marzo AM, Epstein JI, Lotan TL. PTEN loss and ERG protein expression are infrequent in prostatic ductal adenocarcinomas and concurrent acinar carcinomas. Prostate 2015; 75:1610-9. [PMID: 26178158 PMCID: PMC4537350 DOI: 10.1002/pros.23042] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/27/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prostatic ductal adenocarcinoma is an unusual and aggressive morphologic subtype of prostate cancer. PTEN gene deletion and ERG gene rearrangement are among the most common genomic changes in acinar prostate cancers. Though ductal adenocarcinoma most commonly occurs with synchronous usual-type acinar adenocarcinoma, little is known about the molecular phenotype of these mixed tumors. METHODS We used genetically validated immunohistochemistry (IHC) assays to assess PTEN and ERG status in a group of 37 surgically treated ductal adenocarcinomas and 18 synchronous acinar adenocarcinomas where we have previously reported ERG gene rearrangement status by fluorescence in situ hybridization (FISH). A group of 34 stage and grade-matched pure acinar adenocarcinoma cases was studied as a control. RESULTS ERG IHC was highly concordant with ERG FISH results, with 100% (36/36) concordance among ductal adenocarcinomas and 91% (31/34) concordance among 34 pure acinar carcinomas. Similar to previous FISH results, ERG expression by IHC was significantly less common among ductal adenocarcinomas (11% or 4/37) and their synchronous acinar tumors (6% or 1/18) compared to matched pure acinar adenocarcinoma cases (50% or 17/34; P = 0.0005 and 0.002, respectively). PTEN loss by IHC was also less common among ductal adenocarcinomas (18% or 6/34) and their synchronous acinar tumors (22% or 4/18) compared to matched pure acinar carcinomas (50% or 17/34; P = 0.01 and 0.08, respectively). As expected, PTEN loss was enriched among ERG positive compared to ERG-negative tumors in the pure acinar tumor control group (2.5-fold enrichment; P = 0.04) however this was not observed among the ductal adenocarcinomas (1.5 fold enrichment; P = NS). Of ductal adenocarcinomas with an evaluable synchronous acinar component, ERG status was concordant in 94% (17/18) and PTEN status was concordant in 94% (16/17). CONCLUSIONS Based on PTEN and ERG, ductal adenocarcinomas and their concurrent acinar carcinomas may be clonally related in some cases and show important molecular differences from pure acinar carcinoma.
Collapse
Affiliation(s)
- Carlos L. Morais
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mehsati Herawi
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Antoun Toubaji
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Roula Albadine
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jessica Hicks
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - George J. Netto
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jonathan I. Epstein
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Urology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
192
|
Abstract
Prostate cancer (PCa) has become to have the highest incidence and the second mortality rate in western countries, affecting men's health to a large extent. Although prostate-specific antigen (PSA) was discovered to help diagnose the cancer in an early stage for decades, its specificity is relative low, resulting in unnecessary biopsy for healthy people and over-treatment for patients. Thus, it is imperative to identify more and more effective biomarkers for early diagnosis of PCa in order to distinguish patients from healthy populations, which helps guide an early treatment to lower disease-related mortality by noninvasive or minimal invasive approaches. This review generally describes the current early diagnostic biomarkers of PCa in addition to PSA and summarizes the advantages and disadvantages of these biomarkers.
Collapse
Affiliation(s)
| | | | - Ying-Hao Sun
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
193
|
Pal RP, Kockelbergh RC, Pringle JH, Cresswell L, Hew R, Dormer JP, Cooper C, Mellon JK, Barwell JG, Hollox EJ. Immunocytochemical detection of ERG expression in exfoliated urinary cells identifies with high specificity patients with prostate cancer. BJU Int 2015; 117:686-96. [DOI: 10.1111/bju.13184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Raj P. Pal
- Department of Cancer Studies and Molecular Medicine; University of Leicester; Leicester UK
- Department of Urology; University Hospitals of Leicester NHS Trust; Leicester UK
| | - Roger C. Kockelbergh
- Department of Urology; University Hospitals of Leicester NHS Trust; Leicester UK
| | - John Howard Pringle
- Department of Cancer Studies and Molecular Medicine; University of Leicester; Leicester UK
| | - Lara Cresswell
- Department of Cytogenetics; University Hospitals of Leicester NHS Trust; Leicester UK
| | - Roger Hew
- Department of Cellular Pathology; University Hospitals of Leicester NHS Trust; Leicester UK
| | - John P. Dormer
- Department of Cellular Pathology; University Hospitals of Leicester NHS Trust; Leicester UK
| | - Colin Cooper
- Department of Cancer Genetics; University of East Anglia; Norwich UK
| | - John Kilian Mellon
- Department of Urology; University Hospitals of Leicester NHS Trust; Leicester UK
| | | | | |
Collapse
|
194
|
Abstract
PURPOSE OF REVIEW Genomic instability is a fundamental feature of human cancer, leading to the activation of oncogenes and inactivation of tumor suppressors. In prostate cancer (PCA), structural genomic rearrangements, resulting in gene fusions, amplifications, and deletions, are a critical mechanism effecting these alterations. Here, we review recent literature regarding the importance of genomic rearrangements in the pathogenesis of PCA and the potential impact on patient care. RECENT FINDINGS Next-generation sequencing has revealed a striking abundance, complexity, and heterogeneity of genomic rearrangements in PCA. These recent studies have nominated a number of processes in predisposing PCA to genomic rearrangements, including androgen-induced transcription. SUMMARY Structural rearrangements are the critical mechanism resulting in the characteristic genomic changes associated with PCA pathogenesis and progression. Future studies will determine whether the impact of these events on tumor phenotypes can be translated to clinical utility for patient prognosis and choices of management strategies.
Collapse
|
195
|
Brooks JD, Wei W, Hawley S, Auman H, Newcomb L, Boyer H, Fazli L, Simko J, Hurtado-Coll A, Troyer DA, Carroll PR, Gleave M, Lance R, Lin DW, Nelson PS, Thompson IM, True LD, Feng Z, McKenney JK. Evaluation of ERG and SPINK1 by Immunohistochemical Staining and Clinicopathological Outcomes in a Multi-Institutional Radical Prostatectomy Cohort of 1067 Patients. PLoS One 2015; 10:e0132343. [PMID: 26172920 PMCID: PMC4501723 DOI: 10.1371/journal.pone.0132343] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/14/2015] [Indexed: 11/27/2022] Open
Abstract
Distinguishing between patients with early stage, screen detected prostate cancer who must be treated from those that can be safely watched has become a major issue in prostate cancer care. Identification of molecular subtypes of prostate cancer has opened the opportunity for testing whether biomarkers that characterize these subtypes can be used as biomarkers of prognosis. Two established molecular subtypes are identified by high expression of the ERG oncoprotein, due to structural DNA alterations that encode for fusion transcripts in approximately ½ of prostate cancers, and over-expression of SPINK1, which is purportedly found only in ERG-negative tumors. We used a multi-institutional prostate cancer tissue microarray constructed from radical prostatectomy samples with associated detailed clinical data and with rigorous selection of recurrent and non-recurrent cases to test the prognostic value of immunohistochemistry staining results for the ERG and SPINK1 proteins. In univariate analysis, ERG positive cases (419/1067; 39%) were associated with lower patient age, pre-operative serum PSA levels, lower Gleason scores (≤3+4=7) and improved recurrence free survival (RFS). On multivariate analysis, ERG status was not correlated with RFS, disease specific survival (DSS) or overall survival (OS). High-level SPINK1 protein expression (33/1067 cases; 3%) was associated with improved RFS on univariate and multivariate Cox regression analysis. Over-expression of either protein was not associated with clinical outcome. While expression of ERG and SPINK1 proteins was inversely correlated, it was not mutually exclusive since 3 (0.28%) cases showed high expression of both. While ERG and SPINK1 appear to identify discrete molecular subtypes of prostate cancer, only high expression of SPINK1 was associated with improved clinical outcome. However, by themselves, neither ERG nor SPINK1 appear to be useful biomarkers for prognostication of early stage prostate cancer.
Collapse
Affiliation(s)
- James D. Brooks
- Department of Urology, Stanford University, Stanford, California, United States of America
- * E-mail:
| | - Wei Wei
- The Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sarah Hawley
- Canary Foundation, Canary Center at Stanford, Palo Alto, California, United States of America
| | - Heidi Auman
- Canary Foundation, Canary Center at Stanford, Palo Alto, California, United States of America
| | - Lisa Newcomb
- Department of Urology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Hilary Boyer
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Ladan Fazli
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Jeff Simko
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Antonio Hurtado-Coll
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Dean A. Troyer
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Eastern Virginia Medical School, Pathology, Microbiology and Molecular Biology, Norfolk, Virginia, United States of America
| | - Peter R. Carroll
- Department of Urology, University of California San Francisco, San Francisco, California, United States of America
| | - Martin Gleave
- The Prostate Center at Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Raymond Lance
- Department of Urology, Eastern Virginia Medical School, Norfolk, Virginia, United States of America
| | - Daniel W. Lin
- Department of Urology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ian M. Thompson
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lawrence D. True
- Department of Pathology, University of Washington Medical Center, Seattle, Washington, United States of America
| | - Ziding Feng
- The Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jesse K. McKenney
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
196
|
Graff RE, Pettersson A, Lis RT, DuPre N, Jordahl KM, Nuttall E, Rider JR, Fiorentino M, Sesso HD, Kenfield SA, Loda M, Giovannucci EL, Rosner B, Nguyen PL, Sweeney CJ, Mucci LA. The TMPRSS2:ERG fusion and response to androgen deprivation therapy for prostate cancer. Prostate 2015; 75:897-906. [PMID: 25728532 PMCID: PMC4424159 DOI: 10.1002/pros.22973] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/06/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND In the United States, half of men with prostate cancer harbor the androgen-regulated gene fusion TMPRSS2:ERG. We hypothesized that men with TMPRSS2:ERG positive tumors are more responsive to androgen deprivation therapy (ADT). METHODS We studied a cohort of 239 men with prostate cancer from the Physicians' Health Study and Health Professionals Follow-up Study who received ADT during their disease course. Fusion status was assessed on available tumor tissue by immunohistochemistry for ERG protein expression. We used Cox models to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for assessment of prostate cancer-specific mortality after ADT initiation. RESULTS Roughly half of the men had stage T3 or higher tumors at diagnosis and 39% had Gleason 8-10 tumors. During an average follow up of 10.2 years, 42 men died from prostate cancer. There was a non-significant inverse association between positive fusion status and time to death from prostate cancer after ADT (multivariable HR: 0.76; 95% CI: 0.40-1.45). Harboring the TMPRSS2:ERG fusion was associated with a statistically significant lower risk of prostate cancer mortality among men who were treated with orchiectomy (multivariable HR: 0.13; 95% CI: 0.03-0.62), based on 15 events. CONCLUSIONS Our results, combined with those from earlier studies, provide suggestive evidence that men with TMPRSS2:ERG positive tumors may have longer prostate cancer survival after ADT. Larger cohorts are needed for more robust results and to assess whether men with tumors harboring the fusion benefit from treatment with ADT in the (neo) adjuvant or metastatic setting specifically.
Collapse
Affiliation(s)
- Rebecca E. Graff
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | | | - Rosina T. Lis
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Natalie DuPre
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | | | - Elizabeth Nuttall
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Jennifer R. Rider
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Michelangelo Fiorentino
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
- Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Howard D. Sesso
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Department of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Stacey A. Kenfield
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, University of California, San Francisco, San Francisco, CA
| | - Massimo Loda
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Bernard Rosner
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Biostatistics, Harvard School of Public Health, Boston, MA
| | - Paul L. Nguyen
- Department of Radiation Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Boston, MA
| | - Christopher J. Sweeney
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard School of Public Health, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
197
|
Kluth M, Runte F, Barow P, Omari J, Abdelaziz ZM, Paustian L, Steurer S, Christina Tsourlakis M, Fisch M, Graefen M, Tennstedt P, Huland H, Michl U, Minner S, Sauter G, Simon R, Adam M, Schlomm T. Concurrent deletion of 16q23 and PTEN is an independent prognostic feature in prostate cancer. Int J Cancer 2015; 137:2354-63. [PMID: 26009879 DOI: 10.1002/ijc.29613] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/13/2015] [Accepted: 04/22/2015] [Indexed: 01/14/2023]
Abstract
The deletion of 16q23-q24 belongs to the most frequent chromosomal changes in prostate cancer, but the clinical consequences of this alteration have not been studied in detail. We performed fluorescence in situ hybridization analysis using a 16q23 probe in more than 7,400 prostate cancers with clinical follow-up data assembled in a tissue microarray format. Chromosome 16q deletion was found in 21% of cancers, and was linked to advanced tumor stage, high Gleason grade, accelerated cell proliferation, the presence of lymph node metastases (p < 0.0001 each) and positive surgical margin (p = 0.0004). 16q Deletion was more frequent in ERG fusion-positive (27%) as compared to ERG fusion-negative cancers (16%, p < 0.0001), and was linked to other ERG-associated deletions including phosphatase and tensin homolog (PTEN) (p < 0.0001) and 3p13 (p = 0.0303). In univariate analysis, the deletion of 16q was linked to early biochemical recurrence independently from the ERG status (p < 0.0001). Tumors with codeletions of 16q and PTEN had a worse prognosis (p = 0.0199) than those with PTEN or the deletion of 16q alone. Multivariate modeling revealed that the prognostic value of 16q/PTEN deletion patterns was independent from the established prognostic factors. In summary, the results of our study demonstrate that the deletion of 16q and PTEN cooperatively drives prostate cancer progression, and suggests that deletion analysis of 16q and PTEN could be of important clinical value particularly for preoperative risk assessment of the clinically most challenging group of low- and intermediated grade prostate cancers.
Collapse
Affiliation(s)
- Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederic Runte
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philipp Barow
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jazan Omari
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zaid M Abdelaziz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Paustian
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Margit Fisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Graefen
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pierre Tennstedt
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Michl
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Adam
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, Section for Translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
198
|
Tomlins SA, Day JR, Lonigro RJ, Hovelson DH, Siddiqui J, Kunju LP, Dunn RL, Meyer S, Hodge P, Groskopf J, Wei JT, Chinnaiyan AM. Urine TMPRSS2:ERG Plus PCA3 for Individualized Prostate Cancer Risk Assessment. Eur Urol 2015; 70:45-53. [PMID: 25985884 DOI: 10.1016/j.eururo.2015.04.039] [Citation(s) in RCA: 274] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/29/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND TMPRSS2:ERG (T2:ERG) and prostate cancer antigen 3 (PCA3) are the most advanced urine-based prostate cancer (PCa) early detection biomarkers. OBJECTIVE Validate logistic regression models, termed Mi-Prostate Score (MiPS), that incorporate serum prostate-specific antigen (PSA; or the multivariate Prostate Cancer Prevention Trial risk calculator version 1.0 [PCPTrc]) and urine T2:ERG and PCA3 scores for predicting PCa and high-grade PCa on biopsy. DESIGN, SETTING, AND PARTICIPANTS T2:ERG and PCA3 scores were generated using clinical-grade transcription-mediated amplification assays. Pretrained MiPS models were applied to a validation cohort of whole urine samples prospectively collected after digital rectal examination from 1244 men presenting for biopsy. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Area under the curve (AUC) was used to compare the performance of serum PSA (or the PCPTrc) alone and MiPS models. Decision curve analysis (DCA) was used to assess clinical benefit. RESULTS AND LIMITATIONS Among informative validation cohort samples (n=1225 [98%], 80% from patients presenting for initial biopsy), models incorporating T2:ERG had significantly greater AUC than PSA (or PCPTrc) for predicting PCa (PSA: 0.693 vs 0.585; PCPTrc: 0.718 vs 0.639; both p<0.001) or high-grade (Gleason score >6) PCa on biopsy (PSA: 0.729 vs 0.651, p<0.001; PCPTrc: 0.754 vs 0.707, p=0.006). MiPS models incorporating T2:ERG score had significantly greater AUC (all p<0.001) than models incorporating only PCA3 plus PSA (or PCPTrc or high-grade cancer PCPTrc [PCPThg]). DCA demonstrated net benefit of the MiPS_PCPTrc (or MiPS_PCPThg) model compared with the PCPTrc (or PCPThg) across relevant threshold probabilities. CONCLUSIONS Incorporating urine T2:ERG and PCA3 scores improves the performance of serum PSA (or PCPTrc) for predicting PCa and high-grade PCa on biopsy. PATIENT SUMMARY Incorporation of two prostate cancer (PCa)-specific biomarkers (TMPRSS2:ERG and PCA3) measured in the urine improved on serum prostate-specific antigen (or a multivariate risk calculator) for predicting the presence of PCa and high-grade PCa on biopsy. A combined test, Mi-Prostate Score, uses models validated in this study and is clinically available to provide individualized risk estimates.
Collapse
Affiliation(s)
- Scott A Tomlins
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - John R Day
- Hologic/Gen-Probe Inc., San Diego, CA, USA
| | - Robert J Lonigro
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel H Hovelson
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - L Priya Kunju
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rodney L Dunn
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | - John T Wei
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
199
|
Defining ‘progression’ and triggers for curative intervention during active surveillance. Curr Opin Urol 2015; 25:258-66. [DOI: 10.1097/mou.0000000000000158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
200
|
Heterogeneity of PTEN and ERG expression in prostate cancer on core needle biopsies: implications for cancer risk stratification and biomarker sampling. Hum Pathol 2015; 46:698-706. [DOI: 10.1016/j.humpath.2015.01.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 11/22/2022]
|