1
|
Segalés L, Juanpere N, Gallarín N, Lorenzo M, López D, Perera-Bel J, Rodriguez-Vida A, Fumadó L, Cecchini L, Bellmunt J, Lloreta-Trull J, Hernández-Llodrà S. Immunohistochemical markers as predictors of prognosis in multifocal prostate cancer. Virchows Arch 2024; 485:281-290. [PMID: 38017230 PMCID: PMC11329545 DOI: 10.1007/s00428-023-03699-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/09/2023] [Accepted: 11/04/2023] [Indexed: 11/30/2023]
Abstract
The impact of tumor focality on prostate cancer (PCa) prognosis has been addressed in several studies with conflicting results. Tumor foci from multifocal (MF) PCa can show highly heterogeneous molecular features. Our aim was to analyze the protein expression of PTEN, SPOP, SLC45A3, ETV1, ERG and the "triple hit" (ERG overexpression, PTEN plus SLC45A3 loss) in unifocal (UF) and MF PCa, to evaluate their value as prognostic markers according to focality, and the role of tumor heterogeneity in MF disease. PTEN, SPOP, SLC45A3, ETV1 and ERG immunohistochemical expression was evaluated in 185 PCa from 9 TMAs, 51 UF and 134 MF. In a subset of 69 MF cases, the dominant and secondary foci (DF and SF) were compared. Heterogeneity was considered when both tumor foci presented different expression patterns. Relationship with clinicopathological features was also analyzed. MF PCa was diagnosed in significantly younger patients when compared to UF ones (p = 0.007). ETV1 overexpression was associated with UF disease (p = 0.028). A shorter time to PSA recurrence was related to SLC45A3 wt expression in UF PCa (p = 0.052), and to SPOP expression loss (p = 0.043) or "triple hit" phenotype in MF PCa (p = 0.041). In MF cases, PTEN loss, SLC45A3 loss and "triple hit" phenotype were associated with the DF and had significant heterogeneity. In conclusion, our results indicate that UF and MF PCa have relevant and consistent molecular differences. The analysis of an immunohistochemical panel, composed by PTEN, SPOP, SLC45A3, ETV1 and ERG, could be useful to predict outcome in MF cases.
Collapse
Affiliation(s)
- Laura Segalés
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nuria Juanpere
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | | | - Marta Lorenzo
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - David López
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | | | - Alejo Rodriguez-Vida
- Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medical Oncology, Hospital del Mar, CIBERONC, Barcelona, Spain
| | - Lluís Fumadó
- Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Lluís Cecchini
- Department of Urology, Hospital del Mar, Barcelona, Spain
| | - Joaquim Bellmunt
- Hospital del Mar Research Institute, Barcelona, Spain
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Josep Lloreta-Trull
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | | |
Collapse
|
2
|
Muthusamy S, Smith SC. Contemporary Diagnostic Reporting for Prostatic Adenocarcinoma: Morphologic Aspects, Molecular Correlates, and Management Perspectives. Adv Anat Pathol 2024; 31:188-201. [PMID: 38525660 DOI: 10.1097/pap.0000000000000444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
The diagnosis and reporting of prostatic adenocarcinoma have evolved from the classic framework promulgated by Dr Donald Gleason in the 1960s into a complex and nuanced system of grading and reporting that nonetheless retains the essence of his remarkable observations. The criteria for the "Gleason patterns" originally proposed have been continually refined by consensuses in the field, and Gleason scores have been stratified into a patient-friendly set of prognostically validated and widely adopted Grade Groups. One product of this successful grading approach has been the opportunity for pathologists to report diagnoses that signal carefully personalized management, placing the surgical pathologist's interpretation at the center of patient care. At one end of the continuum of disease aggressiveness, personalized diagnostic care means to sub-stratify patients with more indolent disease for active surveillance, while at the other end of the continuum, reporting histologic markers signaling aggression allows sub-stratification of clinically significant disease. Whether contemporary reporting parameters represent deeper nuances of more established ones (eg, new criteria and/or quantitation of Gleason patterns 4 and 5) or represent additional features reported alongside grade (intraductal carcinoma, cribriform patterns of carcinoma), assessment and grading have become more complex and demanding. Herein, we explore these newer reporting parameters, highlighting the state of knowledge regarding morphologic, molecular, and management aspects. Emphasis is made on the increasing value and stakes of histopathologists' interpretations and reporting into current clinical risk stratification and treatment guidelines.
Collapse
Affiliation(s)
| | - Steven Christopher Smith
- Department of Pathology, VCU School of Medicine, Richmond, VA
- Department of Surgery, Division of Urology, VCU School of Medicine, Richmond, VA
- Richmond Veterans Affairs Medical Center, Richmond, VA
- Massey Comprehensive Cancer Center, VCU Health, Richmond, VA
| |
Collapse
|
3
|
Skotheim RI, Bogaard M, Carm KT, Axcrona U, Axcrona K. Prostate cancer: Molecular aspects, consequences, and opportunities of the multifocal nature. Biochim Biophys Acta Rev Cancer 2024; 1879:189080. [PMID: 38272101 DOI: 10.1016/j.bbcan.2024.189080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Prostate cancer is unique compared to other major cancers due to the presence of multiple primary malignant foci in the majority of patients at the time of diagnosis. Each malignant focus has distinct somatic mutations and gene expression patterns, which represents a challenge for the development of prognostic tests for localized prostate cancer. Additionally, the molecular heterogeneity of advanced prostate cancer has important implications for management, particularly for patients with metastatic and locally recurrent cancer. Studies have shown that prostate cancers with mutations in DNA damage response genes are more sensitive to drugs inhibiting the poly ADP-ribose polymerase (PARP) enzyme. However, testing for such mutations should consider both spatial and temporal heterogeneity. Here, we summarize studies where multiregional genomics and transcriptomics analyses have been performed for primary prostate cancer. We further discuss the vast interfocal heterogeneity and how prognostic biomarkers and a molecular definition of the index tumor should be developed. The concept of focal treatments in prostate cancer has been evolving as a demand from patients and clinicians and is one example where there is a need for defining an index tumor. Here, biomarkers must have proven value for individual malignant foci. The potential discovery and implementation of biomarkers that are agnostic to heterogeneity are also explored as an alternative to multisample testing. Thus, deciding upon whole-organ treatment, such as radical prostatectomy, should depend on information from biomarkers which are informative for the whole organ.
Collapse
Affiliation(s)
- Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Kristina T Carm
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
4
|
Voulgari O, Goutas D, Pergaris A, Belogiannis K, Thymara E, Kavantzas N, Lazaris AC. Correlations of PTEN and ERG Immunoexpression in Prostate Carcinoma and Lesions Related to Its Natural History: Clinical Perspectives. Curr Issues Mol Biol 2023; 45:2767-2780. [PMID: 37185705 PMCID: PMC10136580 DOI: 10.3390/cimb45040181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Purpose: The aim of our study was to observe the associations between the ETS-related gene (ERG) and the phosphatase and tensin homolog gene (PTEN) immunoexpression in prostate cancer and related lesions and highlight the clinical significance of these findings. Methods: We evaluated the immunohistochemical expression of ERG and PTEN in a series of 151 invasive prostate adenocarcinomas, including low-grade (Gleason grade pattern 3) and high-grade (Gleason grade patterns 4, 5) morphological patterns which corresponded to 45.5% and 54.4% of the cases, respectively. Additionally, we evaluated the immunoexpression of the two markers both in foci of high-grade prostatic intraepithelial neoplasia (HGPIN), as a precursor lesion of cancer, and in foci of intraductal carcinoma of the prostate (IDCP). Finally, to ensure the malignant nature of the prostate glands examined, we employed p63 and alpha-methylacyl-CoA racemase (AMACR) expression. Results: We found that PTEN loss was observed in 50.7%, and ERG positivity was detected in 41.8% of our cancerous samples. In HGPIN, PTEN loss appeared to be linked with a high-grade adjacent invasive carcinoma component which also displayed PTEN loss. As far as IDCP is concerned, ERG immunonegativity was correlated with adjacent high-grade invasive cancer, which was also ERG immunonegative. Conclusions: Our findings suggest that the clonal expansion of invasive cancer appears to be associated with distinct immunophenotypic cellular alterations of both early and late cancer-related histological lesions. Patients with PTEN loss in HGPIN in prostate biopsies should be closely monitored due to the increased likelihood of having an associated invasive high-grade carcinoma that may have not been sampled. Given the clinical significance that derives from PTEN expression in HGPIN lesions, we suggest the routine use of PTEN immunohistochemistry in prostate cancer biopsies in which HGPIN is the only finding.
Collapse
|
5
|
Ozbek B, Ertunc O, Erickson A, Vidal ID, Gomes-Alexandre C, Guner G, Hicks JL, Jones T, Taube JM, Sfanos KS, Yegnasubramanian S, De Marzo AM. Multiplex immunohistochemical phenotyping of T cells in primary prostate cancer. Prostate 2022; 82:706-722. [PMID: 35188986 DOI: 10.1002/pros.24315] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/03/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Most prostate cancers are "immune cold" and poorly responsive to immune checkpoint inhibitors. However, the mechanisms responsible for the lack of a robust antitumor adaptive immune response in the prostate are poorly understood, which hinders the development of novel immunotherapeutic approaches. AIMS Most inflammatory infiltrates in the prostate are centered around benign glands and stroma, which can confound the molecular characterization of the antitumor immune response. We sought to analytically validate a chromogenic-based multiplex immunohistochemistry (IHC) approach applicable to whole slide digital image analysis to quantify T cell subsets from the tumor microenvironment of primary prostatic adenocarcinomas. As an initial application, we tested the hypothesis that PTEN loss leads to an altered antitumor immune response by comparing matched regions of tumors within the same individual with and without PTEN loss. MATERIALS & METHODS Using the HALO Image Analysis Platform (Indica Labs), we trained a classifier to quantify the densities of eight T cell phenotypes separately in the tumor epithelial and stromal subcompartments. RESULTS The iterative chromogenic approach using 7 different antibodies on the same slide provides highly similar findings to results using individually stained slides with single antibodies. Our main findings in carcinomas (benign removed) include the following: i) CD4+ T cells are present at higher density than CD8+ T cells; ii) all T cell subsets are present at higher densities in the stromal compartment compared to the epithelial tumor compartment; iii) most CD4+ and CD8+ T cells are PD1+; iv) cancer foci with PTEN loss harbored increased numbers of T cells compared to regions without PTEN loss, in both stromal and epithelial compartments; and v) the increases in T cells in PTEN loss regions were associated with ERG gene fusion status. DISCUSSION This modular approach can apply to any IHC-validated antibody combination and sets the groundwork for more detailed spatial analyses. CONCLUSION Iterative chromogenic IHC can be used for whole slide analysis of prostate tissue samples and can complement transcriptomic results including those using single cell and spatial genomic approaches.
Collapse
Affiliation(s)
- Busra Ozbek
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Onur Ertunc
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Erickson
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Igor D Vidal
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carolina Gomes-Alexandre
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gunes Guner
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jessica L Hicks
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tracy Jones
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Janis M Taube
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen S Sfanos
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Brady Urological Research Institute, Johns Hopkins, Baltimore, Maryland, USA
| | - Srinivasan Yegnasubramanian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Brady Urological Research Institute, Johns Hopkins, Baltimore, Maryland, USA
| | - Angelo M De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Brady Urological Research Institute, Johns Hopkins, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Fontugne J, Cai PY, Alnajar H, Bhinder B, Park K, Ye H, Beg S, Sailer V, Siddiqui J, Blattner-Johnson M, Croyle JA, Noorzad Z, Calagua C, MacDonald TY, Axcrona U, Bogaard M, Axcrona K, Scherr DS, Sanda MG, Johannessen B, Chinnaiyan AM, Elemento O, Skotheim RI, Rubin MA, Barbieri CE, Mosquera JM. Collision tumors revealed by prospectively assessing subtype-defining molecular alterations in 904 individual prostate cancer foci. JCI Insight 2022; 7:155309. [PMID: 35050902 PMCID: PMC8876549 DOI: 10.1172/jci.insight.155309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/19/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Prostate cancer is multifocal with distinct molecular subtypes. The utility of genomic subtyping has been challenged due to inter- and intrafocal heterogeneity. We sought to characterize the subtype-defining molecular alterations of primary prostate cancer across all tumor foci within radical prostatectomy (RP) specimens and determine the prevalence of collision tumors. METHODS From the Early Detection Research Network cohort, we identified 333 prospectively collected RPs from 2010 to 2014 and assessed ETS-related gene (ERG), serine peptidase inhibitor Kazal type 1 (SPINK1), phosphatase and tensin homolog (PTEN), and speckle type BTB/POZ protein (SPOP) molecular status. We utilized dual ERG/SPINK1 immunohistochemistry and fluorescence in situ hybridization to confirm ERG rearrangements and characterize PTEN deletion, as well as high-resolution melting curve analysis and Sanger sequencing to determine SPOP mutation status. RESULTS Based on index focus alone, ERG, SPINK1, PTEN, and SPOP alterations were identified in 47.5%, 10.8%, 14.3%, and 5.1% of RP specimens, respectively. In 233 multifocal RPs with ERG/SPINK1 status in all foci, 139 (59.7%) had discordant molecular alterations between foci. Collision tumors, as defined by discrepant ERG/SPINK1 status within a single focus, were identified in 29 (9.4%) RP specimens. CONCLUSION Interfocal molecular heterogeneity was identified in about 60% of multifocal RP specimens, and collision tumors were present in about 10%. We present this phenomenon as a model for the intrafocal heterogeneity observed in previous studies and propose that future genomic studies screen for collision tumors to better characterize molecular heterogeneity. FUNDING Early Detection Research Network US National Cancer Institute (NCI) 5U01 CA111275-09, Center for Translational Pathology at Weill Cornell Medicine (WCM) Department of Pathology and Laboratory Medicine, US NCI (WCM SPORE in Prostate Cancer, P50CA211024-01), R37CA215040, Damon Runyon Cancer Research Foundation, US MetLife Foundation Family Clinical Investigator Award, Norwegian Cancer Society (grant 208197), and South-Eastern Norway Regional Health Authority (grant 2019016 and 2020063).
Collapse
Affiliation(s)
| | - Peter Y Cai
- Department of Urology, Weill Cornell Medicine, New York, United States of America
| | - Hussein Alnajar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Kyung Park
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Huihui Ye
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, United States of America
| | - Shaham Beg
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Verena Sailer
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Javed Siddiqui
- Michigan Center for Translational Pathology and Department of Pathology, The University of Michigan Medical School, Ann Arbor, United States of America
| | - Mirjam Blattner-Johnson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Jaclyn A Croyle
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Zohal Noorzad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | - Carla Calagua
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, United States of America
| | - Theresa Y MacDonald
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, United States of America
| | - Ulrika Axcrona
- Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Douglas S Scherr
- Department of Urology, New York Presbyterian Hospital-Weill Cornell Medical College, New York, United States of America
| | - Martin G Sanda
- Department of Urology, Beth Israel Deaconess Medical Center, Boston, United States of America
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology and Department of Pathology, The University of Michigan Medical School, Ann Arbor, United States of America
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States of America
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| | | | - Juan M Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States of America
| |
Collapse
|
7
|
Segura-Moreno YY, Sanabria-Salas MC, Varela R, Mesa JA, Serrano ML. Decoding the heterogeneous landscape in the development prostate cancer. Oncol Lett 2021; 21:376. [PMID: 33777200 PMCID: PMC7988715 DOI: 10.3892/ol.2021.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/02/2020] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer (PCa) is characterized as being histologically and molecularly heterogeneous; however, this is not only incorrect among individuals, but also at the multiple foci level, which originates in the prostate gland itself. The reasons for such heterogeneity have not been fully elucidated; however, understanding these may be crucial in determining the course of the disease. PCa is characterized by a complex network of chromosomal rearrangements, which simultaneously deregulate multiple genes; this could explain the appearance of exclusive events associated with molecular subtypes, which have been extensively investigated to establish clinical management and the development of therapies targeted to this type of cancer. From a clinical aspect, the prognosis of the patient has focused on the characteristics of the index lesion (the largest focus in PCa); however, a significant percentage of patients (11%) also exhibit an aggressive secondary foci, which may determine the prognosis of the disease, and could be the determining factor of why, in different studies, the classification of the subtypes does not have an association with prognosis. Due to the aforementioned reasons, the analysis of molecular subtypes in several foci, from the same individual could assist in determining the association between clinical evolution and management of patients with PCa. Castration-resistant PCa (CRPC) has the worst prognosis and develops following androgen ablation therapy. Currently, there are two models to explain the development of CRPC: i) The selection model and ii) the adaptation model; both of which, have been found to include alterations described in the molecular subtypes, such as Enhancer of zeste 2 polycomb repressive complex 2 subunit overexpression, isocitrate dehydrogenase (NAPD+)1 and forkhead box A1 mutations, suggesting that the presence of specific molecular alterations could predict the development of CRPC. This type of analysis could lead to a biological understanding of PCa, to develop personalized medicine strategies, which could improve the response to treatment thus, avoiding the development of resistance. Therefore, the present review discusses the primary molecular factors, to which variable heterogeneity in PCa progress has been attributed.
Collapse
Affiliation(s)
- Yenifer Yamile Segura-Moreno
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| | | | - Rodolfo Varela
- Department of Urology, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Urology, National University of Colombia, University City, Bogota 111321, Colombia
| | - Jorge Andrés Mesa
- Department of Pathology, National Institute of Cancerology, Bogota 110411, Colombia
| | - Martha Lucia Serrano
- Cancer Biology Research Group, National Institute of Cancerology, Bogota 110411, Colombia.,Department of Chemistry, Faculty of Sciences, National University of Colombia, University City, Bogota 111321, Colombia
| |
Collapse
|
8
|
Ma L, He H, Jiang K, Jiang P, He H, Feng S, Chen K, Shao J, Deng G. FAM46C inhibits cell proliferation and cell cycle progression and promotes apoptosis through PTEN/AKT signaling pathway and is associated with chemosensitivity in prostate cancer. Aging (Albany NY) 2020; 12:6352-6369. [PMID: 32283544 PMCID: PMC7185131 DOI: 10.18632/aging.103030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/23/2020] [Indexed: 04/11/2023]
Abstract
Family with sequence similarity 46 member C (FAM46C) is a non-canonical poly(A) polymerase that is associated with tumorigenesis. However, its role in prostate cancer development is not fully understood. Herein, we determined expression pattern of FAM46C in prostate cancer and further identified its effect on the tumorigenesis and chemosensitivity. FAM46C expression was decreased in prostate cancer tissues and cell lines compared with corresponding controls. FAM46C expression was significantly associated with the Gleason score, tumor size and overall survival. FAM46C knockdown in 22RV1 and DU145 cells significantly inhibited apoptosis and promoted cell proliferation and cell cycle progression as well as activation of AKT. FAM46C overexpression had an inverse effect in DU145 cells and inhibited tumor growth in vivo. FAM46C inhibited cell proliferation and cell cycle progression and induced apoptosis via the PTEN/AKT signaling pathway. FAM46C promoted PTEN expression through inhibiting PTEN ubiquitination. The prostate cancer cells and patient-derived xenograft (PDX) mice with high-FAM46C-expressing demonstrated an enhanced chemosensitivity to docetaxel. These findings suggest that FAM46C control cell proliferation, cell cycle and apoptosis through PTEN/AKT signaling pathway and is associated with chemosensitivity of prostate cancer. Modulation of their levels may offer a new approach for improving anti-tumor efficacy for chemotherapeutic agents in prostate cancer.
Collapse
Affiliation(s)
- Libin Ma
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang, China
| | - Huadong He
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Kang Jiang
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Peiwu Jiang
- Surgical Department I, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, China
| | - Han He
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Shengjia Feng
- Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310006, Zhejiang, China
| | - Kean Chen
- Department of Urology, The Second Hospital of Jiaxing, Jiaxing 314001, Zhejiang, China
| | - Jia Shao
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Gang Deng
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| |
Collapse
|
9
|
Immunohistochemical expression of mismatch repair proteins (MSH2, MSH6, MLH1, and PMS2) in prostate cancer: correlation with grade groups (WHO 2016) and ERG and PTEN status. Virchows Arch 2019; 475:223-231. [DOI: 10.1007/s00428-019-02591-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 04/05/2019] [Accepted: 06/03/2019] [Indexed: 10/26/2022]
|
10
|
Vidotto T, Saggioro FP, Jamaspishvili T, Chesca DL, Picanço de Albuquerque CG, Reis RB, Graham CH, Berman DM, Siemens DR, Squire JA, Koti M. PTEN-deficient prostate cancer is associated with an immunosuppressive tumor microenvironment mediated by increased expression of IDO1 and infiltrating FoxP3+ T regulatory cells. Prostate 2019; 79:969-979. [PMID: 30999388 DOI: 10.1002/pros.23808] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Accumulating evidence shows that tumor cell-specific genomic changes can influence the cross talk between cancer cells and the surrounding tumor microenvironment (TME). Loss of the PTEN tumor suppressor gene is observed in 20% to 30% of prostate cancers (PCa) when first detected and the rate increases with PCa progression and advanced disease. Recent findings implicate a role for PTEN in cellular type I interferon response and immunosuppression in PCa. However, the way that PTEN inactivation alters antitumor immune response in PCa is poorly understood. MATERIALS AND METHODS To investigate the changes associated with PTEN loss and an immunosuppressive TME in PCa, we used CIBERSORT to estimate the relative abundance of 22 immune-cell types from 741 primary and 96 metastatic tumors. Our in silico findings were then validated by immunohistochemical analysis of immune cells and IDO1 and PDL1 checkpoint proteins in a cohort of 94 radical prostatectomy specimens. RESULTS FoxP3+ T regulatory cells (Tregs) were significantly increased in PTEN-deficient PCa in all three public domain cohorts. Loss of PTEN in bone metastases was associated with lower CD8+ T-cell abundance, but in liver metastasis, FoxP3+ Tregs were present at higher levels. PTEN-deficient lymph node metastasis had a distinct profile, with high levels of CD8+ T cells. Moreover, we found that metastatic PCa presents higher abundance of FoxP3+ Treg when compared to primary lesions. Since PTEN-deficient tumors are likely to be immunosuppressed as a consequence of increased FoxP3+ Tregs, we then evaluated the localization and expression of IDO1, PDL1 immune checkpoints, and the corresponding density of FoxP3+ Treg and CD8+ T cells using our validation cohort (n = 94). We found that IDO1 protein expression and FoxP3+ Treg density were higher in neoplastic glands compared with benign adjacent tissue. Moreover, higher densities of FoxP3+ Treg cells in both stromal (P = 0.04) and tumor (P = 0.006) compartments were observed in PTEN-deficient tumors compared to tumors that retained PTEN activity. Similarly, IDO1 protein expression was significantly increased in the tumor glands of PTEN-deficient PCa (P < 0.0001). Spearman correlation analysis showed that IDO1 expression was significantly associated with FoxP3+ Treg and CD8+ T-cell density (P < 0.01). CONCLUSIONS Our findings imply that PTEN deficiency is linked to an immunosuppressive state in PCa with distinct changes in the frequency of immune cell types in tumors from different metastatic sites. Our data suggest that determining PTEN status may also help guide the selection of patients for future immunotherapy trials in localized and metastatic PCa.
Collapse
Affiliation(s)
- Thiago Vidotto
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiano P Saggioro
- Department of Pathology and Legal Medicine, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Tamara Jamaspishvili
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Canada
| | - Deise L Chesca
- Department of Pathology and Legal Medicine, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Rodolfo B Reis
- Medical Genetics Division, Clinics Hospital of Ribeirão Preto, Ribeirão Preto, Brazil
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - David M Berman
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Canada
| | - D Robert Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| | - Jeremy A Squire
- Department of Genetics, Medicine School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Madhuri Koti
- Cancer Biology and Genetics, Queen's Cancer Research Institute, Queen's University, Kingston, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
- Department of Urology, Queen's University, Kingston, Canada
| |
Collapse
|
11
|
Segalés L, Juanpere N, Lorenzo M, Albero-González R, Fumadó L, Cecchini L, Bellmunt J, Lloreta-Trull J, Hernández-Llodrà S. Strong cytoplasmic ETV1 expression has a negative impact on prostate cancer outcome. Virchows Arch 2019; 475:457-466. [DOI: 10.1007/s00428-019-02573-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 01/28/2023]
|
12
|
Iczkowski KA. Large-Gland Proliferations of the Prostate. Surg Pathol Clin 2018; 11:687-712. [PMID: 30447836 DOI: 10.1016/j.path.2018.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Large-gland proliferations of the prostate have gained considerable attention in the past decade. The differential diagnosis is quite broad but can be refined using histologic criteria and, sometimes, immunostains. Pathologists have come to realize that cribriform and intraductal as well as ductal carcinomas are particularly aggressive patterns, and should name them in diagnostic reporting when present.
Collapse
Affiliation(s)
- Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| |
Collapse
|
13
|
Association of ERG/PTEN status with biochemical recurrence after radical prostatectomy for clinically localized prostate cancer. Med Oncol 2018; 35:152. [PMID: 30291535 DOI: 10.1007/s12032-018-1212-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022]
Abstract
We have previously demonstrated a significant correlative relationship between PTEN deletion and ERG rearrangement, both in the development of clinically localized prostate cancers and metastases. Herein, we evaluate the cooperative role of ERG and PTEN in oncological outcomes after radical prostatectomy for clinically localized prostate cancer. We evaluated ERG and PTEN status using three previously described cohorts. The first cohort included 235 clinically localized prostate cancer cases represented on tissue microarrays (TMA), evaluated using previously validated FISH assays for ERG and PTEN. The second cohort included 167 cases of clinically localized prostate cancer on TMAs evaluated for PTEN by FISH, and for PTEN and ERG by dual IHC. The third cohort comprised 59 clinically localized prostate cancer cases assessed by array comparative genomic hybridization (aCGH). Kaplan-Meir plots and long rank tests were used to assess the association of ERG and PTEN status with biochemical recurrence after radical prostatectomy for clinically localized prostate cancer. Of the 317 cases eligible for analyses with evaluable ERG and PTEN status, 88 (27.8%) patients developed biochemical recurrence over a median follow-up of 5.7 years. Overall, 45% (142/317) of cases demonstrated ERG rearrangement and 20% (62/317) of cases demonstrated PTEN loss. Hemizygous and homozygous deletion of PTEN was seen in 10% (18/175) and 3% (5/175) of ERG-negative cases, respectively. In contrast, hemizygous and homozygous deletion of PTEN was seen in 11% (15/142) and 17% (24/123) of ERG-positive cases, respectively. PTEN loss (heterozygous or homozygous) was significantly associated with shorter time to biochemical recurrence compared to no PTEN loss (p < 0.001). However, ERG rearrangement versus no rearrangement was not associated with time to PSA recurrence (p = 0.15). Patients who exhibited ERG rearrangement and loss of PTEN had no significant difference in time to recurrence compared to patients with wild-type ERG and loss of PTEN (p = 0.30). Our findings confirm a mutual cooperative role of ERG and PTEN in the pathogenesis of prostate cancer, particularly for homozygous PTEN deletion. ERG did not stratify outcome either alone or in combination with PTEN in this cohort.
Collapse
|
14
|
Köseoğlu H. Genetics in the Prostate Cancer. Prostate Cancer 2018. [DOI: 10.5772/intechopen.77259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
15
|
Abstract
PURPOSE OF REVIEW This review will examine the taxonomy of PCa subclasses across disease states, explore the relationship among specific alterations, and highlight current clinical relevance. RECENT FINDINGS Prostate cancer (PCa) is driven by multiple genomic alterations, with distinct patterns and clinical implications. Alterations occurring early in the timeline of the disease define core subtypes of localized, treatment-naive PCa. With time, an increase in number and severity of genomic alterations adds molecular complexity and is associated with progression to metastasis. These later events are not random and are influenced by the underlying subclasses. All the subclasses of localized disease initially respond to androgen deprivation therapy (ADT), but with progression to castrate-resistant PCa (CRPC), mechanisms of resistance against ADT shift the molecular landscape. In CRPC, resistance mechanisms largely define the biology and sub-classification of these cancers, while clinical relevance and opportunities for precision therapy are still being defined.
Collapse
Affiliation(s)
- Kaveri Arora
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, BRB 1452, 413 East 69th Street, New York, NY, 10021, USA.,Department of Urology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher E Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Belfer Research Building, BRB 1452, 413 East 69th Street, New York, NY, 10021, USA. .,Department of Urology, Weill Cornell Medicine, New York, NY, USA. .,Englander Institute for Precision Medicine of Weill Cornell Medicine and NewYork-Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
16
|
Use of multicolor fluorescence in situ hybridization to detect deletions in clinical tissue sections. J Transl Med 2018; 98:403-413. [PMID: 29339834 DOI: 10.1038/s41374-017-0007-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/01/2017] [Accepted: 12/01/2017] [Indexed: 12/19/2022] Open
Abstract
A variety of laboratory methods are available for the detection of deletions of tumor suppressor genes and losses of their proteins. The clinical utility of fluorescence in situ hybridization (FISH) for the identification of deletions of tumor suppressor genes has previously been limited by difficulties in the interpretation of FISH signal patterns. The first deletion FISH assays using formalin-fixed paraffin-embedded tissue sections had to deal with a significant background level of signal losses affecting nuclei that are truncated by the cutting process of slide preparation. Recently, more efficient probe designs, incorporating probes adjacent to the tumor suppressor gene of interest, have increased the accuracy of FISH deletion assays so that true chromosomal deletions can be readily distinguished from the false signal losses caused by sectioning artifacts. This mini-review discusses the importance of recurrent tumor suppressor gene deletions in human cancer and reviews the common FISH methods being used to detect the genomic losses encountered in clinical specimens. The use of new probe designs to recognize truncation artifacts is illustrated with a four-color PTEN FISH set optimized for prostate cancer tissue sections. Data are presented to show that when section thickness is reduced, the frequency of signal truncation losses is increased. We also provide some general guidelines that will help pathologists and cytogeneticists run routine deletion FISH assays and recognize sectioning artifacts. Finally, we summarize how recently developed sequence-based approaches are being used to identify recurrent deletions using small DNA samples from tumors.
Collapse
|
17
|
Yang S, Wang Y, Gao H, Wang B. MicroRNA-30a-3p overexpression improves sepsis-induced cell apoptosis in vitro and in vivo via the PTEN/PI3K/AKT signaling pathway. Exp Ther Med 2018; 15:2081-2087. [PMID: 29434809 PMCID: PMC5776646 DOI: 10.3892/etm.2017.5644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/10/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to explain the mechanism of miR-30a-3p overexpression in sepsis-induced cell apoptosis in vitro and in vivo. For the in vitro cell experiments, H9c2 cells were divided into three groups, including the untreated normal control (NC), lipopolysaccharide (LPS)-treated and miRNA (treated with LPS and transfection with miRNA-30a-3p) groups. The cell proliferation and apoptosis were evaluated by MTT assay and flow cytometry, respectively. The relative protein expression levels were measured by western blot assay. In the in vivo experiment, a sepsis rat model was established by intraperitoneal injection of LPS. Sprague Dawley rats were divided into three groups, including the NC, LPS-injected and miRNA (in which model rats were injected with miR-30a-3p vector at the caudal vein) groups. The myocardial morphology in different groups was observed by hematoxylin and eosin staining. In addition, tissue apoptosis and protein expression levels were evaluated by TUNEL and western blot assay, respectively. The results of cell experiments indicated that the cell proliferation rate was significantly increased and the cell apoptosis rate was significantly downregulated in the miR-30a-3p group compared with the LPS group (both P<0.05). The relative protein expression of phosphatase and tensin homolog (PTEN) was markedly decreased in the miRNA group compared with the LPS group, while the levels of phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) were significantly increased in the miRNA group (all P<0.05). In the in vivo experiments, the myocardial morphology of the miRNA group was improved compared with that of the LPS group. Compared with the LPS group, cell apoptosis in the miRNA group was significantly downregulated (P<0.05), while the relative protein levels (PTEN, PI3K and AKT) in the tissues were also significantly altered (P<0.05). In conclusion, miR-30a-3p overexpression may improve the sepsis-induced cell apoptosis in vitro and in vivo via the PTEN/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Shuying Yang
- Intensive Care Unit, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Yongqiang Wang
- Intensive Care Unit, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Hongmei Gao
- Intensive Care Unit, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Bing Wang
- Intensive Care Unit, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| |
Collapse
|
18
|
Ullman D, Dorn D, Rais-Bahrami S, Gordetsky J. Clinical Utility and Biologic Implications of Phosphatase and Tensin Homolog (PTEN) and ETS-related Gene (ERG) in Prostate Cancer. Urology 2017; 113:59-70. [PMID: 29225123 DOI: 10.1016/j.urology.2017.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) and ETS-related gene (ERG) mutations are commonly found in prostate cancer. Although mouse studies have demonstrated that PTEN and ERG cooperatively interact during tumorigenesis, human studies examining these genes have been inconclusive. A systematic PubMed search including original articles assessing the pathogenesis of PTEN and ERG in prostate cancer was performed. Studies examining ERG's prognostic significance have conflicting results. Studies examining PTEN and ERG simultaneously found these genes are likely to occur together, but cooperative tumorigenesis functions have not been conclusively established. PTEN mutations are associated with a range of prognostic features. However, the practical clinical utility of this information remains to be determined.
Collapse
Affiliation(s)
- David Ullman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - David Dorn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer Gordetsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL; Department of Urology, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
19
|
Prostate cancer, PI3K, PTEN and prognosis. Clin Sci (Lond) 2017; 131:197-210. [PMID: 28057891 DOI: 10.1042/cs20160026] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/12/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
Loss of function of the PTEN tumour suppressor, resulting in dysregulated activation of the phosphoinositide 3-kinase (PI3K) signalling network, is recognized as one of the most common driving events in prostate cancer development. The observed mechanisms of PTEN loss are diverse, but both homozygous and heterozygous genomic deletions including PTEN are frequent, and often accompanied by loss of detectable protein as assessed by immunohistochemistry (IHC). The occurrence of PTEN loss is highest in aggressive metastatic disease and this has driven the development of PTEN as a prognostic biomarker, either alone or in combination with other factors, to distinguish indolent tumours from those likely to progress. Here, we discuss these factors and the consequences of PTEN loss, in the context of its role as a lipid phosphatase, as well as current efforts to use available inhibitors of specific components of the PI3K/PTEN/TOR signalling network in prostate cancer treatment.
Collapse
|
20
|
Hernández-Llodrà S, Juanpere N, de Muga S, Lorenzo M, Gil J, Font-Tello A, Agell L, Albero-González R, Segalés L, Merino J, Serrano L, Fumadó L, Cecchini L, Lloreta-Trull J. ERG overexpression plus SLC45A3 (prostein) and PTEN expression loss: Strong association of the triple hit phenotype with an aggressive pathway of prostate cancer progression. Oncotarget 2017; 8:74106-74118. [PMID: 29088771 PMCID: PMC5650326 DOI: 10.18632/oncotarget.18266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 05/15/2017] [Indexed: 12/18/2022] Open
Abstract
TMPRSS2 and SLC45A3 rearrangements may coexist in the same tumor. ERG rearrangements and PTEN loss are concomitant events in prostate cancer (PrCa), and can cooperate in progression. We have reported that mRNA expression of TMPRSS2-ERG and SLC45A3-ERG rearrangements plus PTEN loss define an aggressive tumor subset. The aim of this study has been to validate these results by immunohistochemistry in a large cohort of tumors. ERG, SLC45A3 and PTEN immunostaining and their association with pathological features and PSA progression-free survival were analyzed in 220 PrCa (PSMAR-Biobank, Barcelona, Spain). ERG protein expression was found in 46.8% and SLC45A3 and PTEN loss in 30% and 34% tumors, respectively. Single ERG positive immunostaining was associated with GS = 6 tumors (p = 0.016), double ERG+/PTEN loss with GS = 7 (p = 0.008) and Grade Group 2 (GG) or GG3 cases (p = 0.042), ERG+/SLC45A3 loss/PTEN loss ("triple hit") with GS ≥ 8 (p < 0.0001) and GG4 or GG5 tumors (p = 0.0003). None of GS = 6 nor = GG1 cases showed this combination. In the GS ≥ 8 group, ERG+ (p = 0.002), PTEN loss (p = 0.009) and "triple hit" (p = 0.003) were associated with Gleason pattern 3 component, and single SLC45A3 loss (p = 0.036) with GS ≥ 8 without pattern 3. The number of aberrant events and the triple hit were strongly associated with shorter PSA progression-free survival. In GS = 6 PrCa, single ERG+ was also associated with progression. ERG+ identifies a distinct pathway of PrCa. Additional assessment of PTEN and SLC45A3 adds relevant prognostic information. The triple hit phenotype (ERG+/SLC45A3 loss/PTEN loss) is associated with progression and could be used for patient stratification, treatment and follow-up.
Collapse
Affiliation(s)
| | - Nuria Juanpere
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Silvia de Muga
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Lorenzo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Joan Gil
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laia Agell
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laura Segalés
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laia Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Fumadó
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Cecchini
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Josep Lloreta-Trull
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| |
Collapse
|
21
|
Inhibition of the PI3K/AKT/mTOR pathway activates autophagy and compensatory Ras/Raf/MEK/ERK signalling in prostate cancer. Oncotarget 2017; 8:56698-56713. [PMID: 28915623 PMCID: PMC5593594 DOI: 10.18632/oncotarget.18082] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/24/2017] [Indexed: 12/19/2022] Open
Abstract
The PI3K/AKT/mTOR pathway is frequently activated in advanced prostate cancer, due to loss of the tumour suppressor PTEN, and is an important axis for drug development. We have assessed the molecular and functional consequences of pathway blockade by inhibiting AKT and mTOR kinases either in combination or as individual drug treatments. In established prostate cancer cell lines, a decrease in cell viability and in phospho-biomarker expression was observed. Although apoptosis was not induced, a G1 growth arrest was observed in PTEN null LNCaP cells, but not in BPH1 or PC3 cells. In contrast, when the AKT inhibitor AZD7328 was applied to patient-derived prostate cultures that retained expression of PTEN, activation of a compensatory Ras/MEK/ERK pathway was observed. Moreover, whilst autophagy was induced following treatment with AZD7328, cell viability was less affected in the patient-derived cultures than in cell lines. Surprisingly, treatment with a combination of both AZD7328 and two separate MEK1/2 inhibitors further enhanced phosphorylation of ERK1/2 in primary prostate cultures. However, it also induced irreversible growth arrest and senescence. Ex vivo treatment of a patient-derived xenograft (PDX) of prostate cancer with a combination of AZD7328 and the mTOR inhibitor KU-0063794, significantly reduced tumour frequency upon re-engraftment of tumour cells. The results demonstrate that single agent targeting of the PI3K/AKT/mTOR pathway triggers activation of the Ras/MEK/ERK compensatory pathway in near-patient samples. Therefore, blockade of one pathway is insufficient to treat prostate cancer in man.
Collapse
|
22
|
Ronen S, Abbott DW, Kravtsov O, Abdelkader A, Xu Y, Banerjee A, Iczkowski KA. PTEN loss and p27 loss differ among morphologic patterns of prostate cancer, including cribriform. Hum Pathol 2017; 65:85-91. [PMID: 28504208 DOI: 10.1016/j.humpath.2017.04.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/18/2017] [Accepted: 04/29/2017] [Indexed: 12/31/2022]
Abstract
The presence and extent of cribriform pattern of prostate cancer portend recurrence and cancer death. The relative expressions within this morphology of the prognostically adverse loss of PTEN, and the downstream inactivation of cell cycle inhibitor p27/Kip1 had been uncertain. In this study, we examined 52 cases of cribriform cancer by immunohistochemistry for PTEN, p27, and CD44 variant (v)7/8, and a subset of 17 cases by chromogenic in situ hybridization (ISH) using probes for PTEN or CDKN1B (gene for p27). The fractions of epithelial pixels positive by immunohistochemistry and ISH were digitally assessed for benign acini, high-grade prostatic intraepithelial neoplasia, and 8 morphologic patterns of cancer. Immunostaining results demonstrated that (1) PTEN loss was significant for fused small acini, cribriform-central cells, small cribriform acini, and Gleason grade 5 cells in comparison with other acini; (2) p27 loss was significant only for cribriform-peripheral cells and borderline significant for fused small acini in comparison with benign acini; and (3) CD44v7/8 showed expression loss in cribriform-peripheral cells; other comparisons were not significant. ISH showed that cribriform cancer had significant PTEN loss normalized to benign acini (P<.02), whereas Gleason 3 cancer or fused small acini did not. With CDKN1B, the degree of signal loss among various cancer morphologies was insignificant. In conclusion, molecular disparities emerged between the fused small acini and cribriform patterns of Gleason 4 cancer. PTEN or p27 loss as prognostic factors demands distinct assessment in the varieties of Gleason 4 cancer, and in the biphenotypic peripheral versus central populations in cribriform structures.
Collapse
Affiliation(s)
- Shira Ronen
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel W Abbott
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Oleksandr Kravtsov
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amrou Abdelkader
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yayun Xu
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anjishnu Banerjee
- Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kenneth A Iczkowski
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
23
|
Fisher KW, Zhang S, Wang M, Montironi R, Wang L, Baldrige LA, Wang JY, MacLennan GT, Williamson SR, Lopez-Beltran A, Cheng L. TMPRSS2-ERGgene fusion is rare compared to PTENdeletions in stage T1a prostate cancer. Mol Carcinog 2017; 56:814-820. [DOI: 10.1002/mc.22535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Kurt W. Fisher
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| | - Shaobo Zhang
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| | - Mingsheng Wang
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| | - Rodolfo Montironi
- Department of Urology; Institute of Pathological Anatomy and Histopathology; Polytechnic University of the Marche Region (Ancona); United Hospitals; Ancona Italy
| | - Lisha Wang
- Michigan Center for Translational Pathology; University of Michigan; Ann Arbor Michigan
| | - Lee A. Baldrige
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| | - Jonas Y. Wang
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| | - Gregory T. MacLennan
- Departments of Pathology and Laboratory Medicine; Case Western Reserve University; Cleveland Ohio
| | - Sean R. Williamson
- Department of Pathology and Laboratory Medicine; Henry Ford Health System; Detroit Michigan
- Josephine Ford Cancer Institute; Henry Ford Health System; Detroit Michigan
- Department of Pathology; Wayne State University School of Medicine; Detroit Michigan
| | - Antonio Lopez-Beltran
- Faculty of Medicine, Department of Pathology and Surgery, Cordoba University Spain and Champalimaud Clinical Center; Cordoba University; Lisbon Portugal
| | - Liang Cheng
- Department of Pathology; Indiana University School of Medicine; Indianapolis Indiana
| |
Collapse
|
24
|
Downes MR, Satturwar S, Trudel D, van der Kwast TH. Evaluation of ERG and PTEN protein expression in cribriform architecture prostate carcinomas. Pathol Res Pract 2017; 213:34-38. [DOI: 10.1016/j.prp.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/19/2016] [Indexed: 10/20/2022]
|
25
|
Giannico GA, Arnold SA, Gellert LL, Hameed O. New and Emerging Diagnostic and Prognostic Immunohistochemical Biomarkers in Prostate Pathology. Adv Anat Pathol 2017; 24:35-44. [PMID: 27941540 PMCID: PMC10182893 DOI: 10.1097/pap.0000000000000136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The diagnosis of minimal prostatic adenocarcinoma can be challenging on prostate needle biopsy, and immunohistochemistry may be used to support the diagnosis of cancer. The International Society of Urologic Pathology currently recommends the use of the basal cell markers high-molecular-weight cytokeraratin and p63, and α-methylacyl-coenzyme-A racemase. However, there are caveats associated with the interpretation of these markers, particularly with benign mimickers. Another issue is that of early detection of presence and progression of disease and prediction of recurrence after clinical intervention. There remains a lack of reliable biomarkers to accurately predict low-risk cancer and avoid over treatment. As such, aggressive forms of prostate cancer may be missed and indolent disease may be subjected to unnecessary radical therapy. New biomarker discovery promises to improve early detection and prognosis and to provide targets for therapeutic interventions. In this review, we present the emerging immunohistochemical biomarkers of prostate cancer PTEN, ERG, FASN, MAGI-2, and SPINK1, and address their diagnostic and prognostic advantages and limitations.
Collapse
Affiliation(s)
- Giovanna A. Giannico
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Shanna A. Arnold
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
- Department of Veterans Affairs, Nashville, TN
| | - Lan L. Gellert
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Omar Hameed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| |
Collapse
|
26
|
Huang KC, Evans A, Donnelly B, Bismar TA. SPINK1 Overexpression in Localized Prostate Cancer: a Rare Event Inversely Associated with ERG Expression and Exclusive of Homozygous PTEN Deletion. Pathol Oncol Res 2016; 23:399-407. [PMID: 27738792 DOI: 10.1007/s12253-016-0119-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022]
Abstract
SPINK1 is proposed as potential prognostic marker in prostate cancer (PCA). However, its relation to PTEN and ERG in localized PCA remains unclear. The study population consisted of two independent cohorts of men treated by radical prostatectomy for localized PCA (discovery n = 218 and validation n = 129). Patterns of association between SPINK1 and each of ERG and PTEN were evaluated by immunohistochemistry and fluorescence in situ hybridization. Associations between SPINK1 expression and various pathologic parameters and clinical outcome were also investigated. SPINK1 was expressed in 15.3 % and 10.9 % of cases in the discovery and validation cohort, respectively. SPINK expression was observed in 5.56 % of high-grade prostatic intraepithelial neoplasia and 1.1 % of adjacent morphologically benign prostatic glands. SPINK1 and ERG expression were almost exclusive, with only 1.0 % of the cases co-expressing both in the same core sample. SPINK1 interfocal and within-core heterogeneity was noted in 29.2 % and 64.6 % of cases, respectively. SPINK1 expression was not significantly associated with PTEN deletion in the two cohorts (p = 0.871 for discovery cohort and p = 0.293 for validation cohort). While SPINK1 expression did occur with hemizygous PTEN deletion, there was a complete absence of SPINK1 expression in PCA showing homozygous PTEN deletion, which was confirmed in the validation cohort (p = 0.02). Despite SPINK1's association with higher Gleason score (>7) (p = 0.02), it was not associated with other pathological parameters or biochemical recurrence post-radical prostatectomy. We documented absolute exclusivity between SPINK1 overexpression and homozygous PTEN deletion in localized PCA. SPINK1 and ERG expressions are exclusive events in PCA. SPINK1 is not of added prognostic value in localized PCA.
Collapse
Affiliation(s)
- Kuo-Cheng Huang
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, 7007, 14sth st sw, Calgary, AB, T2V 1P9, Canada
| | - Andrew Evans
- Department of Pathology, Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Bryan Donnelly
- Department of Urology, University of Calgary, Calgary, AB, Canada
- The Prostate Cancer Center, Calgary, AB, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary and Calgary Laboratory Services, 7007, 14sth st sw, Calgary, AB, T2V 1P9, Canada.
- Departments of Oncology, Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.
- Southern Alberta Cancer Institute and Tom Baker Cancer Center, Calgary, AB, Canada.
- The Prostate Cancer Center, Calgary, AB, Canada.
| |
Collapse
|
27
|
PTEN loss and chromosome 8 alterations in Gleason grade 3 prostate cancer cores predicts the presence of un-sampled grade 4 tumor: implications for active surveillance. Mod Pathol 2016; 29:764-71. [PMID: 27080984 PMCID: PMC4925272 DOI: 10.1038/modpathol.2016.63] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 12/11/2022]
Abstract
Men who enter active surveillance because their biopsy exhibits only Gleason grade 3 (G3) frequently have higher grade tumor missed by biopsy. Thus, biomarkers are needed that, when measured on G3 tissue, can predict the presence of higher grade tumor in the whole prostate. We evaluated whether PTEN loss, chromosome 8q gain (MYC) and/or 8p loss (LPL) measured only on G3 cores is associated with un-sampled G4 tumor. A tissue microarray was constructed of prostatectomy tissue from patients whose prostates exhibited only Gleason score 3+3, only 3+4 or only 4+3 tumor (n=50 per group). Cores sampled only from areas of G3 were evaluated for PTEN loss by immunohistochemistry, and PTEN deletion, LPL/8p loss and MYC/8q gain by fluorescence in situ hybridization. Biomarker results were compared between Gleason score 6 vs 7 tumors using conditional logistic regression. PTEN protein loss, odds ratio=4.99, P=0.033; MYC/8q gain, odds ratio=5.36, P=0.010; and LPL/8p loss, odds ratio=3.96, P=0.003 were significantly more common in G3 cores derived from Gleason 7 vs Gleason 6 tumors. PTEN gene deletion was not statistically significant. Associations were stronger comparing Gleason 4+3 vs 6 than for Gleason 3+4 vs 6. MYC/8q gain, LPL/8p loss and PTEN protein loss measured in G3 tissue microarray cores strongly differentiate whether the core comes from a Gleason 6 or Gleason 7 tumor. If validated to predict upgrading from G3 biopsy to prostatectomy these biomarkers could reduce the likelihood of enrolling high-risk men and facilitate safe patient selection for active surveillance.
Collapse
|
28
|
Hernández S, Font-Tello A, Juanpere N, de Muga S, Lorenzo M, Salido M, Fumadó L, Serrano L, Cecchini L, Serrano S, Lloreta J. Concurrent TMPRSS2-ERG and SLC45A3-ERG rearrangements plus PTEN loss are not found in low grade prostate cancer and define an aggressive tumor subset. Prostate 2016; 76:854-65. [PMID: 26959281 DOI: 10.1002/pros.23176] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/16/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND SLC45A3 is the second most common ERG partner in prostate cancer (PrCa). Coexisting TMPRSS2 and SLC45A3 rearrangements are found in a subset of cases, but the meaning is still unknown. METHODS SLC45A3-ERG and TMPRSS2-ERG rearrangements and their association with ERG and PTEN expression and with clinical and pathological features have been analyzed in 80 PrCa (PSMAR-Biobank, Barcelona, Spain). ERG and PTEN mRNA were assessed by qRT-PCR; TMPRSS2-ERG and SLC45A3-ERG by RT-PCR, FISH, and direct sequencing; and ERG expression by IHC. The endpoints were Gleason score (GS), stage, and PSA progression-free survival. RESULTS Single TMPRSS2-ERG was found in 51.6% GS ≤ 7 and 22.2% GS ≥ 8 tumors (P = 0.027). SLC45A3-ERG was found in 25 cases, 20 of them with concurrent TMPRSS2-ERG rearrangement: 11.5% GS = 6, 22.2% GS = 7, and 50% GS ≥ 8 tumors (P = 0.013). Double rearrangements were associated with higher levels of ERG mRNA (P = 0.04). Double rearrangement plus PTEN loss was detected in 0% GS = 6; 14.7% GS = 7, and 29.4% GS ≥ 8 tumors (P = 0.032). Furthermore, this triple change was present in 19.2% stage T3-4 but not in any of stage T2 tumors (P = 0.05). No relationship was found with PSA progression-free survival. CONCLUSIONS Single TMPRSS2-ERG translocation is associated with low grade PrCa. Subsequent development of SLC45A3-ERG results in higher ERG expression. The combination of double rearrangement plus PTEN loss, according to our series, is never found in low grade, low stage tumors. These findings could be potentially useful in therapeutic decision making in PrCa. Tumors with combined TMPRSS2-ERG/SLC45A3-ERG fusions plus PTEN loss should be excluded from watchful waiting and are candidates for intensive therapy. Prostate 76:854-865, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Silvia Hernández
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alba Font-Tello
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Núria Juanpere
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Silvia de Muga
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Lorenzo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Marta Salido
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Fumadó
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Laia Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Cecchini
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Sergio Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Josep Lloreta
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| |
Collapse
|
29
|
In prostate cancer needle biopsies, detections of PTEN loss by fluorescence in situ hybridization (FISH) and by immunohistochemistry (IHC) are concordant and show consistent association with upgrading. Virchows Arch 2016; 468:607-17. [DOI: 10.1007/s00428-016-1904-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/15/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022]
|
30
|
Integrated analysis of the genomic instability of PTEN in clinically insignificant and significant prostate cancer. Mod Pathol 2016; 29:143-56. [PMID: 26612463 DOI: 10.1038/modpathol.2015.136] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 11/08/2022]
Abstract
Patients with clinically insignificant prostate cancer remain a major over-treated population. PTEN loss is one of the most recurrent alterations in prostate cancer associated with an aggressive phenotype, however, the occurrence of PTEN loss in insignificant prostate cancer has not been reported and its role in the separation of insignificant from significant prostate cancer is unclear. An integrated analysis of PTEN loss was, therefore, performed for structural variations, point mutations and protein expression in clinically insignificant (48 cases) and significant (76 cases) prostate cancers treated by radical prostatectomy. Whole-genome mate pair sequencing was performed on tumor cells isolated by laser capture microdissection to characterize PTEN structural alterations. Fluorescence in situ hybridization probes were constructed from the sequencing data to detect the spectrum of these PTEN alterations. PTEN loss by mate pair sequencing and fluorescence in situ hybridization occurred in 2% of insignificant, 13% of large volume Gleason score 6, and 46% of Gleason score 7 and higher cancers. In Gleason score 7 cancers with PTEN loss, PTEN alterations were detected in both Gleason pattern 3 and 4 in 57% of cases by mate pair sequencing, 75% by in situ hybridization and 86% by immunohistochemistry. PTEN loss by sequencing was strongly associated with TMPRSS2-ERG fusion, biochemical recurrence, PTEN loss by in situ hybridization and protein loss by immunohistochemistry. The complex nature of PTEN rearrangements was unveiled by sequencing, detailing the heterogeneous events leading to homozygous loss of PTEN. PTEN point mutation was present in 5% of clinically significant tumors and not in insignificant cancer or high-grade prostatic intraepithelial neoplasia. PTEN loss is infrequent in clinically insignificant prostate cancer, and is associated with higher grade tumors. Detection of PTEN loss in Gleason score 6 cancer in a needle biopsy specimen indicates a higher likelihood of clinically significant prostate cancer.
Collapse
|
31
|
Qu X, Jeldres C, Glaskova L, Friedman C, Schroeder S, Nelson PS, Porter C, Fang M. Identification of Combinatorial Genomic Abnormalities Associated with Prostate Cancer Early Recurrence. J Mol Diagn 2016; 18:215-24. [PMID: 26752304 DOI: 10.1016/j.jmoldx.2015.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 10/06/2015] [Accepted: 10/14/2015] [Indexed: 11/29/2022] Open
Abstract
Multiple biomarkers are needed to distinguish aggressive from indolent prostate cancer. We tested the prognostic utility of a three-marker fluorescent in situ hybridization (FISH) panel (TMPRSS2/ERG rearrangements, AR gain, and PTEN deletion) in a retrospective cohort (n = 210; median follow-up, 5.7 years). PTEN deletion was associated with an increased risk of biochemical recurrence (BcR; hazard ratio, 3.58; 95% CI, 1.39-9.22; P < 0.01) by multivariable Cox regression analyses and earlier BcR (P < 0.02) by Kaplan-Meier analysis. AR gain coexisted with X-chromosome gain and was associated with advanced tumor stage. When this panel was applied, two categories of combinatorial abnormalities proved clinically important. First, PTEN deletion without TMPRSS2/ERG rearrangement was enriched in pT3/4 tumors (70% versus 48%) and tumors with Gleason grades of 8 to 9 (60% versus 17%) compared with the entire cohort. These patients had earlier BcR than patients with normal FISH panel results (P < 0.01). In contrast, patients with PTEN deletion and ERG rearrangement had a BcR rate similar to patients who tested normal for all three markers (P > 0.1). Second, AR gain and concurrent trisomy 10 without TMPRSS2/ERG rearrangement were enriched in pT3/4 tumors and tumors with Gleason grades of 8 to 9. The three-marker FISH panel demonstrated prognostic utility and identified genomic aberrations associated with advanced disease state and early BcR in prostate cancer.
Collapse
Affiliation(s)
- Xiaoyu Qu
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington
| | - Claudio Jeldres
- Department of Urology, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | - Peter S Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Christopher Porter
- Department of Urology, Virginia Mason Medical Center, Seattle, Washington.
| | - Min Fang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
32
|
Gibbs SL, Genega E, Salemi J, Kianzad V, Goodwill HL, Xie Y, Oketokoun R, Khurd P, Kamen A, Frangioni JV. Near-infrared fluorescent digital pathology for the automation of disease diagnosis and biomarker assessment. Mol Imaging 2016; 14. [PMID: 25812603 DOI: 10.2310/7290.2015.00005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hematoxylin-eosin (H&E) staining of tissue has been the mainstay of pathology for more than a century. However, the learning curve for H&E tissue interpretation is long, whereas intra- and interobserver variability remain high. Computer-assisted image analysis of H&E sections holds promise for increased throughput and decreased variability but has yet to demonstrate significant improvement in diagnostic accuracy. Addition of biomarkers to H&E staining can improve diagnostic accuracy; however, coregistration of immunohistochemical staining with H&E is problematic as immunostaining is completed on slides that are at best 4 μm apart. Simultaneous H&E and immunostaining would alleviate coregistration problems; however, current opaque pigments used for immunostaining obscure H&E. In this study, we demonstrate that diagnostic information provided by two or more independent wavelengths of near-infrared (NIR) fluorescence leave the H&E stain unchanged while enabling computer-assisted diagnosis and assessment of human disease. Using prostate cancer as a model system, we introduce NIR digital pathology and demonstrate its utility along the spectrum from prostate biopsy to whole mount analysis of H&E-stained tissue.
Collapse
|
33
|
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
|
34
|
Haffner MC, Weier C, Xu MM, Vaghasia A, Gürel B, Gümüşkaya B, Esopi DM, Fedor H, Tan HL, Kulac I, Hicks J, Isaacs WB, Lotan TL, Nelson WG, Yegnasubramanian S, De Marzo AM. Molecular evidence that invasive adenocarcinoma can mimic prostatic intraepithelial neoplasia (PIN) and intraductal carcinoma through retrograde glandular colonization. J Pathol 2015; 238:31-41. [PMID: 26331372 DOI: 10.1002/path.4628] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/28/2015] [Accepted: 08/10/2015] [Indexed: 12/23/2022]
Abstract
Prostate cancer often manifests as morphologically distinct tumour foci and is frequently found adjacent to presumed precursor lesions such as high-grade prostatic intraepithelial neoplasia (HGPIN). While there is some evidence to suggest that these lesions can be related and exist on a pathological and morphological continuum, the precise clonal and temporal relationships between precursor lesions and invasive cancers within individual tumours remain undefined. Here, we used molecular genetic, cytogenetic, and histological analyses to delineate clonal, temporal, and spatial relationships between HGPIN and cancer lesions with distinct morphological and molecular features. First, while confirming the previous finding that a substantial fraction of HGPIN lesions associated with ERG-positive cancers share rearrangements and overexpression of ERG, we found that a significant subset of such HGPIN glands exhibit only partial positivity for ERG. This suggests that such ERG-positive HGPIN cells either rapidly invade to form adenocarcinoma or represent cancer cells that have partially invaded the ductal and acinar space in a retrograde manner. To clarify these possibilities, we used ERG expression status and TMPRSS2-ERG genomic breakpoints as markers of clonality, and PTEN deletion status to track temporal evolution of clonally related lesions. We confirmed that morphologically distinct HGPIN and nearby invasive cancer lesions are clonally related. Further, we found that a significant fraction of ERG-positive, PTEN-negative HGPIN and intraductal carcinoma (IDC-P) lesions are most likely clonally derived from adjacent PTEN-negative adenocarcinomas, indicating that such PTEN-negative HGPIN and IDC-P lesions arise from, rather than give rise to, the nearby invasive adenocarcinoma. These data suggest that invasive adenocarcinoma can morphologically mimic HGPIN through retrograde colonization of benign glands with cancer cells. Similar clonal relationships were also seen for intraductal carcinoma adjacent to invasive adenocarcinoma. These findings represent a potentially undervalued indicator of pre-existing invasive prostate cancer and have significant implications for prostate cancer diagnosis and risk stratification.
Collapse
Affiliation(s)
- Michael C Haffner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Christopher Weier
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Meng Meng Xu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ajay Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bora Gürel
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Berrak Gümüşkaya
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - David M Esopi
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Helen Fedor
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsueh-Li Tan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ibrahim Kulac
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jessica Hicks
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - William B Isaacs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tamara L Lotan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA.,Brady Urological Institute, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Martin AM, Nirschl TR, Nirschl CJ, Francica BJ, Kochel CM, van Bokhoven A, Meeker AK, Lucia MS, Anders RA, DeMarzo AM, Drake CG. Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis 2015. [PMID: 26260996 DOI: 10.1038/pcan.2015.39.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Primary prostate cancers are infiltrated with programmed death-1 (PD-1) expressing CD8+ T-cells. However, in early clinical trials, men with metastatic castrate-resistant prostate cancer did not respond to PD-1 blockade as a monotherapy. One explanation for this unresponsiveness could be that prostate tumors generally do not express programmed death ligand-1 (PD-L1), the primary ligand for PD-1. However, lack of PD-L1 expression in prostate cancer would be surprising, given that phosphatase and tensin homolog (PTEN) loss is relatively common in prostate cancer and several studies have shown that PTEN loss correlates with PD-L1 upregulation--constituting a mechanism of innate immune resistance. This study tested whether prostate cancer cells were capable of expressing PD-L1, and whether the rare PD-L1 expression that occurs in human specimens correlates with PTEN loss. METHODS Human prostate cancer cell lines were evaluated for PD-L1 expression and loss of PTEN by flow cytometry and western blotting, respectively. Immunohistochemical (IHC) staining for PTEN was correlated with PD-L1 IHC using a series of resected human prostate cancer samples. RESULTS In vitro, many prostate cancer cell lines upregulated PD-L1 expression in response to inflammatory cytokines, consistent with adaptive immune resistance. In these cell lines, no association between PTEN loss and PD-L1 expression was apparent. In primary prostate tumors, PD-L1 expression was rare, and was not associated with PTEN loss. CONCLUSIONS These studies show that some prostate cancer cell lines are capable of expressing PD-L1. However, in human prostate cancer, PTEN loss is not associated with PD-L1 expression, arguing against innate immune resistance as a mechanism that mitigates antitumor immune responses in this disease.
Collapse
Affiliation(s)
- A M Martin
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - T R Nirschl
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - C J Nirschl
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - B J Francica
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - C M Kochel
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - A van Bokhoven
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - A K Meeker
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - M S Lucia
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - R A Anders
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - A M DeMarzo
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - C G Drake
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
36
|
Paucity of PD-L1 expression in prostate cancer: innate and adaptive immune resistance. Prostate Cancer Prostatic Dis 2015; 18:325-32. [PMID: 26260996 PMCID: PMC4641011 DOI: 10.1038/pcan.2015.39] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/29/2015] [Accepted: 07/08/2015] [Indexed: 12/31/2022]
Abstract
Background Primary prostate cancers are infiltrated with PD-1 expressing CD8+ T cells. However, in early clinical trials, men with mCRPC did not respond to PD-1 blockade as a monotherapy. One explanation for this unresponsiveness could be that prostate tumors generally do not express PD-L1, the primary ligand for PD-1. However, lack of PD-L1 expression in prostate cancer would be surprising, given that PTEN loss is relatively common in prostate cancer and several studies have shown that PTEN loss correlates with PD-L1 up-regulation - constituting a mechanism of innate immune resistance. This study tested whether prostate cancer cells were capable of expressing PD-L1, and whether the rare PD-L1 expression that occurs in human specimens correlates with PTEN loss. Methods Human prostate cancer cell lines were evaluated for PD-L1 expression and loss of PTEN by flow cytometry and western blotting, respectively. Immunohistochemical (IHC) staining for PTEN was correlated with PD-L1 IHC using a series of resected human prostate cancer samples. Results In vitro, many prostate cancer cell lines up-regulated PD-L1 expression in response to inflammatory cytokines, consistent with adaptive immune resistance. In these cell lines, no association between PTEN loss and PD-L1 expression was apparent. In primary prostate tumors, PD-L1 expression was rare, and was not associated with PTEN loss. Conclusions These studies show that some prostate cancer cell lines are capable of expressing PD-L1. However, in human prostate cancer, PTEN loss is not associated with PD-L1 expression, arguing against innate immune resistance as a mechanism that mitigates anti-tumor immune responses in this disease.
Collapse
|
37
|
Font-Tello A, Juanpere N, de Muga S, Lorenzo M, Lorente JA, Fumado L, Serrano L, Serrano S, Lloreta J, Hernández S. Association of ERG and TMPRSS2-ERG with grade, stage, and prognosis of prostate cancer is dependent on their expression levels. Prostate 2015; 75:1216-26. [PMID: 25939480 DOI: 10.1002/pros.23004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/26/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND There is controversy in the literature on the role of the fusion TMPRSS2-ERG in the pathogenesis and progression of prostate cancer. The quantitative differences in TMPRSS2-ERG fusion expression have received very limited attention in the literature. METHODS We have quantitatively analyzed the mRNA levels of TMPRSS2-ERG, ERG, PTEN, and AR (n = 83), as well as ERG immunostaining (n = 78) in a series of prostate tumors. RESULTS Among the TMPRSS2-ERG cases (n = 57), high fusion levels were associated with GS ≥8 (P = 0.025). ERG mRNA overexpression was associated with GS ≥8 (P = 0.047), and with stage T3-T4 tumors (P = 0.032). Among the ERG overexpressing cases (n = 54), higher expression levels were found in 92.3% of GS ≥8 tumors (P = 0.02). ERG immunostaining, regardless of staining intensity, was also associated with high stage (P = 0.05). There was a statistical association between ERG immunostaining and PSA progression-free survival (Log Rank test, P = 0.048). Decreased PTEN expression was associated with TMPRSS2-ERG (P = 0.01), ERG mRNA overexpression (P = 0.003) and ERG immunostaining (P = 0.007). Furthermore, decreased PTEN expression, alone (P = 0.041) and also combined with TMPRSS2-ERG (P = 0.04) or with ERG overexpression (P = 0.04) was associated with GS ≥7 tumors. CONCLUSIONS Although more studies are needed to further clarify their role, our findings emphasize that the expression levels of the TMPRSS2-ERG fusion and ERG mRNA, rather than their mere presence, are related to a more aggressive phenotype, have an effect on prognosis and could be molecular markers of progression for prostate cancer. Furthermore, ERG immunohistochemistry could be also a potentially useful prognostic factor.
Collapse
Affiliation(s)
- Alba Font-Tello
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Núria Juanpere
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Silvia de Muga
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Lorenzo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - José A Lorente
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Lluis Fumado
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Laia Serrano
- Department of Pathology, Hospital Universitari Germans Tries i Pujol, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Sergio Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Josep Lloreta
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Silvia Hernández
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
38
|
Troyer DA, Jamaspishvili T, Wei W, Feng Z, Good J, Hawley S, Fazli L, McKenney JK, Simko J, Hurtado-Coll A, Carroll PR, Gleave M, Lance R, Lin DW, Nelson PS, Thompson IM, True LD, Brooks JD, Squire JA. A multicenter study shows PTEN deletion is strongly associated with seminal vesicle involvement and extracapsular extension in localized prostate cancer. Prostate 2015; 75:1206-15. [PMID: 25939393 PMCID: PMC4475421 DOI: 10.1002/pros.23003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 03/20/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Loss of the phosphatase and tensin homolog (PTEN) tumor suppressor gene is a promising marker of aggressive prostate cancer. Active surveillance and watchful waiting are increasingly recommended to patients with small tumors felt to be low risk, highlighting the difficulties of Gleason scoring in this setting. There is an urgent need for predictive biomarkers that can be rapidly deployed to aid in clinical decision-making. Our objectives were to assess the incidence and ability of PTEN alterations to predict aggressive disease in a multicenter study. METHODS We used recently developed probes optimized for sensitivity and specificity in a four-color FISH deletion assay to study the Canary Retrospective multicenter Prostate Cancer Tissue Microarray (TMA). This TMA was constructed specifically for biomarker validation from radical prostatectomy specimens, and is accompanied by detailed clinical information with long-term follow-up. RESULTS In 612 prostate cancers, the overall rate of PTEN deletion was 112 (18.3%). Hemizygous PTEN losses were present in 55/612 (9.0%) of cancers, whereas homozygous PTEN deletion was observed in 57/612 (9.3%) of tumors. Significant associations were found between PTEN status and pathologic stage (P < 0.0001), seminal vesicle invasion (P = 0.0008), extracapsular extension (P < 0.0001), and Gleason score (P = 0.0002). In logistic regression analysis of clinical and pathological variables, PTEN deletion was significantly associated with extracapsular extension, seminal vesicle involvement, and higher Gleason score. In the 406 patients in which clinical information was available, PTEN homozygous (P = 0.009) deletion was associated with worse post-operative recurrence-free survival (number of events = 189), pre-operative prostate specific antigen (PSA) (P < 0.001), and pathologic stage (P = 0.03). CONCLUSION PTEN status assessed by FISH is an independent predictor for recurrence-free survival in multivariate models, as were seminal vesicle invasion, extracapsular extension, and Gleason score, and preoperative PSA. Furthermore, these data demonstrate that the assay can be readily introduced at first diagnosis in a cost effective manner analogous to the use of FISH for analysis of HER2/neu status in breast cancer. Combined with published research beginning 17 years ago, both the data and tools now exist to implement a PTEN assay in the clinic.
Collapse
Affiliation(s)
- Dean A Troyer
- Eastern Virginia Medical School, Pathology and Microbiology and Molecular BiologyNorfolk, Virginia
- Department of Pathology, University of Texas Health Science Center at San AntonioSan Antonio, Texas
| | - Tamara Jamaspishvili
- Department of Pathology and Molecular Medicine, Queen’s UniversityKingston, Ontario, Canada
| | - Wei Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Ziding Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer CenterHouston, Texas
| | - Jennifer Good
- Department of Pathology and Molecular Medicine, Queen’s UniversityKingston, Ontario, Canada
| | - Sarah Hawley
- Canary FoundationCanary Center at Stanford, 3155 Porter DrivePalo Alto
| | - Ladan Fazli
- The Prostate Center at Vancouver General Hospital, University of British ColumbiaVancouver, British Columbia, Canada
| | | | - Jeff Simko
- Department of Pathology, University of California San FranciscoSan Francisco, California
- Department of Urology, University of California San FranciscoSan Francisco, California
| | - Antonio Hurtado-Coll
- The Prostate Center at Vancouver General Hospital, University of British ColumbiaVancouver, British Columbia, Canada
| | - Peter R Carroll
- Department of Urology, University of California San FranciscoSan Francisco, California
| | - Martin Gleave
- The Prostate Center at Vancouver General Hospital, University of British ColumbiaVancouver, British Columbia, Canada
| | - Raymond Lance
- Department of Urology, Eastern Virginia Medical SchoolNorfolk, Virginia
| | - Daniel W Lin
- Department of Urology, University of WashingtonSeattle, Washington
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research CenterSeattle, Washington
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center at San AntonioSan Antonio, Texas
| | - Lawrence D True
- Department of Pathology, University of Washington Medical CenterSeattle, Washington
| | - James D Brooks
- Department of Urology, Stanford UniversityStanford, California
| | - Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen’s UniversityKingston, Ontario, Canada
- Department of Pathology and Forensic Medicine, University of São Paulo at Ribeirão PretoBrazil
- * Correspondence to: Dr. Jeremy A. Squire, PhD, Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada; Department of Pathology and Forensic Medicine, University of São Paulo at Ribeirão Preto, Brazil. E-mail:
| |
Collapse
|
39
|
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]
|
40
|
Abstract
PUPOSE OF REVIEW The review covers arguments for and against removing the label of 'cancer' in Gleason score 6 prostate tumors. RECENT FINDINGS While there are a number of factors that determine whether men elect active surveillance, the most powerful predictor remains the Gleason score. Gleason grading remains a robust and powerful predictor of outcome in patients with prostate cancer. A pure Gleason score 6 (GS6) tumor is exceedingly unlikely to cause harm in the near term, and there have been discussions regarding whether the term cancer should still be applied. In this review, we update the largely clinico-pathological arguments that have led to the suggestion to remove the cancer label from GS6 tumors, and we provide counter arguments on the basis of practical matters of needle biopsy sampling, classical histopathology, and molecular biology findings. SUMMARY The implications are that by retaining the label of cancer and implementing the recently proposed concept of prognostic groups, with patients harboring GS6 tumors placed into the lowest category, there is still a strong rationale in support of the choice of active surveillance or watchful waiting for most patients with GS6 lesions.
Collapse
Affiliation(s)
- Ibrahim Kulac
- aDepartment of Pathology bDepartment of Urology cDepartment of Oncology dThe Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins eThe Brady Urological Research Institute
| | | | | | | | | |
Collapse
|
41
|
Hong MK, Macintyre G, Wedge DC, Van Loo P, Patel K, Lunke S, Alexandrov LB, Sloggett C, Cmero M, Marass F, Tsui D, Mangiola S, Lonie A, Naeem H, Sapre N, Phal PM, Kurganovs N, Chin X, Kerger M, Warren AY, Neal D, Gnanapragasam V, Rosenfeld N, Pedersen JS, Ryan A, Haviv I, Costello AJ, Corcoran NM, Hovens CM. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat Commun 2015; 6:6605. [PMID: 25827447 PMCID: PMC4396364 DOI: 10.1038/ncomms7605] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/11/2015] [Indexed: 12/29/2022] Open
Abstract
Tumour heterogeneity in primary prostate cancer is a well-established phenomenon. However, how the subclonal diversity of tumours changes during metastasis and progression to lethality is poorly understood. Here we reveal the precise direction of metastatic spread across four lethal prostate cancer patients using whole-genome and ultra-deep targeted sequencing of longitudinally collected primary and metastatic tumours. We find one case of metastatic spread to the surgical bed causing local recurrence, and another case of cross-metastatic site seeding combining with dynamic remoulding of subclonal mixtures in response to therapy. By ultra-deep sequencing end-stage blood, we detect both metastatic and primary tumour clones, even years after removal of the prostate. Analysis of mutations associated with metastasis reveals an enrichment of TP53 mutations, and additional sequencing of metastases from 19 patients demonstrates that acquisition of TP53 mutations is linked with the expansion of subclones with metastatic potential which we can detect in the blood.
Collapse
Affiliation(s)
- Matthew K.H. Hong
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Geoff Macintyre
- Centre for Neural Engineering, Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
- Diagnostic Genomics, NICTA, Victoria Research Laboratory, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David C. Wedge
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Peter Van Loo
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Department of Human Genetics, KU Leuven, Herestraat 49 Box 602, B-3000 Leuven, Belgium
- Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Keval Patel
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
- Academic Urology Group, Addenbrookes Hospital, Cambridge University, Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Sebastian Lunke
- Centre for Translational Pathology, University of Melbourne, Parkville 3050, Victoria, Australia
| | - Ludmil B. Alexandrov
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Clare Sloggett
- Victorian Life Sciences Computation Initiative, The University of Melbourne, Parkville 3050, Victoria, Australia
| | - Marek Cmero
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
- Centre for Neural Engineering, Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
- Diagnostic Genomics, NICTA, Victoria Research Laboratory, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Francesco Marass
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Dana Tsui
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Stefano Mangiola
- Centre for Neural Engineering, Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andrew Lonie
- Victorian Life Sciences Computation Initiative, The University of Melbourne, Parkville 3050, Victoria, Australia
| | - Haroon Naeem
- Centre for Neural Engineering, Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
- Diagnostic Genomics, NICTA, Victoria Research Laboratory, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nikhil Sapre
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Pramit M. Phal
- Department of Radiology, Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| | - Natalie Kurganovs
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Xiaowen Chin
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Michael Kerger
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Anne Y. Warren
- Department of Histopathology, University Cambridge Hospitals, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - David Neal
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
- Academic Urology Group, Addenbrookes Hospital, Cambridge University, Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Vincent Gnanapragasam
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
- Academic Urology Group, Addenbrookes Hospital, Cambridge University, Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - John S. Pedersen
- TissuPath Specialist Pathology, Mount Waverley 3149, Victoria, Australia
- Monash University Faculty of Medicine, Clayton 3168, Victoria, Australia
| | - Andrew Ryan
- TissuPath Specialist Pathology, Mount Waverley 3149, Victoria, Australia
| | - Izhak Haviv
- Bar-Ilan University Medical School, Safad 1311502, Israel
| | - Anthony J. Costello
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Niall M. Corcoran
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| | - Christopher M. Hovens
- Department of Surgery, Division of Urology, Royal Melbourne Hospital and University of Melbourne, Parkville 3050, Victoria, Australia
- The Epworth Prostate Centre, Epworth Hospital, Richmond 3121, Victoria, Australia
| |
Collapse
|
42
|
Gerlinger M, Catto JW, Orntoft TF, Real FX, Zwarthoff EC, Swanton C. Intratumour heterogeneity in urologic cancers: from molecular evidence to clinical implications. Eur Urol 2015; 67:729-37. [PMID: 24836153 DOI: 10.1016/j.eururo.2014.04.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/21/2014] [Indexed: 02/05/2023]
Abstract
CONTEXT Intratumour heterogeneity (ITH) can impair the precise molecular analysis of tumours and may contribute to difficulties encountered in cancer biomarker qualification and treatment personalisation. OBJECTIVE This review summarises the evidence for genetic ITH in renal, bladder, and prostate carcinomas and potential strategies to address the clinical and translational research challenges arising from ITH. EVIDENCE ACQUISITION Publications that assessed ITH in the relevant urologic cancers were identified in a literature review. EVIDENCE SYNTHESIS ITH with functionally distinct tumour subclones has been identified in all three tumour types. Heterogeneity of actionable genetic changes and of prognostic biomarkers between different tumour regions in the same patient suggests limitations of single biopsy-based molecular analyses for precision medicine approaches. Evolutionary constraints may differ between patients and may allow the prediction of specific evolutionary trajectories. CONCLUSIONS Assessment of multiple tumour regions for precision medicine purposes, monitoring of subclonal dynamics over time, and the preferential targeting of genetic alterations located on the trunk of the phylogenetic tree of individual cancers may accelerate the development of personalised medicine strategies and improve our understanding of treatment failure. PATIENT SUMMARY Genetic alterations can be heterogeneous within urologic tumours, complicating their use as biomarkers for treatment personalisation. We present novel strategies to address these challenges.
Collapse
Affiliation(s)
- Marco Gerlinger
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK.
| | - James W Catto
- Academic Urology Unit, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Torben F Orntoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Molecular Pathology Program, CNIO (Spanish National Cancer Research Centre), Madrid, Spain; Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Charles Swanton
- CR-UK London Research Institute, London, UK; University College London Cancer Institute, London, UK.
| |
Collapse
|
43
|
Tolkach Y, Imkamp F, Godin K, Van Poppel H. Clinically relevant genetic characterization of prostate tumors: how close are we to the goal? Korean J Urol 2015; 56:90-8. [PMID: 25685295 PMCID: PMC4325124 DOI: 10.4111/kju.2015.56.2.90] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 12/23/2014] [Indexed: 11/18/2022] Open
Abstract
Substantial efforts are being made in research on the molecular genetic characterization of prostate cancer. The number of fundamental research programs in prostate cancer molecular biology and genetics is overwhelming. However, a significant gap appears to exist between the huge number of studies on the genetic characterization of prostate cancer, which often have limited translation into clinical practice or simply were not conceived to be so translated, and clinical practice. From a clinical point of view, this balance should be urgently shifted towards rapid translation into urological practice. However, prostate cancer is characterized by prominent genetic heterogeneity, which could be a very difficult barrier to overcome. In this review, we discuss the possible clinical applications of scientific data from fundamental studies of prostate cancer genetics, the main problems with the translation of these data to clinics, and future perspectives.
Collapse
Affiliation(s)
- Yuri Tolkach
- Urology and Urologic Oncology Clinic, Hannover Medical School, Hannover, Germany
| | - Florian Imkamp
- Urology and Urologic Oncology Clinic, Hannover Medical School, Hannover, Germany
| | | | - Hendrik Van Poppel
- Department of Urology, University Hospitals of Catholic University of Leuven, Leuven, Belgium
| |
Collapse
|
44
|
PTEN loss is associated with upgrading of prostate cancer from biopsy to radical prostatectomy. Mod Pathol 2015; 28:128-137. [PMID: 24993522 PMCID: PMC4282985 DOI: 10.1038/modpathol.2014.85] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/20/2014] [Accepted: 04/21/2014] [Indexed: 12/16/2022]
Abstract
When distinguishing between indolent and potentially harmful prostate cancers, the Gleason score is the most important variable, but may be inaccurate in biopsies due to tumor under-sampling. This study investigated whether a molecular feature, PTEN protein loss, could help identify which Gleason score 6 tumors on biopsy are likely to be upgraded at radical prostatectomy. Seventy one patients with Gleason score 6 tumors on biopsy upgraded to Gleason score 7 or higher at prostatectomy (cases) were compared with 103 patients with Gleason score 6 on both biopsy and prostatectomy (controls). A validated immunohistochemical assay for PTEN was performed, followed by fluorescence in situ hybridization (FISH) to detect PTEN gene deletion in a subset. PTEN protein loss and clinical-pathologic variables were assessed by logistic regression. Upgraded patients were older than controls (61.8 vs 59.3 years), had higher pre-operative PSA levels (6.5 vs 5.3 ng/ml) and a higher fraction of involved cores (0.42 vs 0.36). PTEN loss by immunohistochemistry was found in 18% (13/71) of upgraded cases compared with 7% (7/103) of controls (P=0.02). Comparison between PTEN immunohistochemistry and PTEN FISH showed the assays were highly concordant, with 97% (65/67) of evaluated biopsies with intact PTEN protein lacking PTEN gene deletion, and 81% (13/16) of the biopsies with PTEN protein loss showing homozygous PTEN gene deletion. Tumors with PTEN protein loss were more likely to be upgraded at radical prostatectomy than those without loss, even after adjusting for age, preoperative PSA, clinical stage and race (odds ratio=3.04 (1.08-8.55; P=0.035)). PTEN loss in Gleason score 6 biopsies identifies a subset of prostate tumors at increased risk of upgrading at radical prostatectomy. These data provide evidence that a genetic event can improve Gleason score accuracy and highlight a path toward the clinical use of molecular markers to augment pathologic grading.
Collapse
|
45
|
Iczkowski KA, Egevad L, Ma J, Harding-Jackson N, Algaba F, Billis A, Camparo P, Cheng L, Clouston D, Comperat EM, Datta MW, Evans AG, Griffiths DF, Guo CC, Hailemariam S, Huang W, Humphrey PA, Jiang Z, Kahane H, Kristiansen G, La Rosa FG, Lopez-Beltran A, MacLennan GT, Magi-Galluzzi C, Merrimen J, Montironi R, Osunkoya AO, Picken MM, Rao N, Shah RB, Shanks JH, Shen SS, Tawfik OW, True LD, Van der Kwast T, Varma M, Wheeler TM, Zynger DL, Sahr N, Bostwick DG. Intraductal carcinoma of the prostate: interobserver reproducibility survey of 39 urologic pathologists. Ann Diagn Pathol 2014; 18:333-42. [DOI: 10.1016/j.anndiagpath.2014.08.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 10/24/2022]
|
46
|
Heterogeneity of DNA methylation in multifocal prostate cancer. Virchows Arch 2014; 466:53-9. [DOI: 10.1007/s00428-014-1678-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/17/2014] [Accepted: 10/23/2014] [Indexed: 10/24/2022]
|
47
|
Tereshchenko IV, Zhong H, Chekmareva MA, Kane-Goldsmith N, Santanam U, Petrosky W, Stein MN, Ganesan S, Singer EA, Moore D, Tischfield JA, DiPaola RS. ERG and CHD1 heterogeneity in prostate cancer: use of confocal microscopy in assessment of microscopic foci. Prostate 2014; 74:1551-9. [PMID: 25175909 PMCID: PMC4181586 DOI: 10.1002/pros.22873] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/10/2014] [Indexed: 11/06/2022]
Abstract
BACKGROUND Biomarkers predicting tumor response are important to emerging targeted therapeutics. Complimentary methods to assess and understand genetic changes and heterogeneity within only few cancer cells in tissue will be a valuable addition for assessment of tumors such as prostate cancer that often have insufficient tumor for next generation sequencing in a single biopsy core. METHODS Using confocal microscopy to identify cell-to-cell relationships in situ, we studied the most common gene rearrangement in prostate cancer (TMPRSS2 and ERG) and the tumor suppressor CHD1 in 56 patients who underwent radical prostatectomy. RESULTS Wild type ERG was found in 22 of 56 patients; ERG copy number was increased in 10/56, and ERG rearrangements confirmed in 24/56 patients. In 24 patients with ERG rearrangements, the mechanisms of rearrangement were heterogeneous, with deletion in 14/24, a split event in 7/24, and both deletions and split events in the same tumor focus in 3/24 patients. Overall, 14/45 (31.1%) of patients had CHD1 deletion, with the majority of patients with CHD1 deletions (13/14) correlating with ERG-rearrangement negative status (P < 0.001). CONCLUSIONS These results demonstrate the ability of confocal microscopy and FISH to identify the cell-to-cell differences in common gene fusions such as TMPRSS2-ERG that may arise independently within the same tumor focus. These data support the need to study complimentary approaches to assess genetic changes that may stratify therapy based on predicted sensitivities.
Collapse
Affiliation(s)
- Irina V Tereshchenko
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, New Jersey
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Single-cell genetic analysis reveals insights into clonal development of prostate cancers and indicates loss of PTEN as a marker of poor prognosis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2671-86. [PMID: 25131421 DOI: 10.1016/j.ajpath.2014.06.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 05/16/2014] [Accepted: 06/16/2014] [Indexed: 12/11/2022]
Abstract
Gauging the risk of developing progressive disease is a major challenge in prostate cancer patient management. We used genetic markers to understand genomic alteration dynamics during disease progression. By using a novel, advanced, multicolor fluorescence in situ hybridization approach, we enumerated copy numbers of six genes previously identified by array comparative genomic hybridization to be involved in aggressive prostate cancer [TBL1XR1, CTTNBP2, MYC (alias c-myc), PTEN, MEN1, and PDGFB] in six nonrecurrent and seven recurrent radical prostatectomy cases. An ERG break-apart probe to detect TMPRSS2-ERG fusions was included. Subsequent hybridization of probe panels and cell relocation resulted in signal counts for all probes in each individual cell analyzed. Differences in the degree of chromosomal and genomic instability (ie, tumor heterogeneity) or the percentage of cells with TMPRSS2-ERG fusion between samples with or without progression were not observed. Tumors from patients that progressed had more chromosomal gains and losses, and showed a higher degree of selection for a predominant clonal pattern. PTEN loss was the most frequent aberration in progressers (57%), followed by TBL1XR1 gain (29%). MYC gain was observed in one progresser, which was the only lesion with an ERG gain, but no TMPRSS2-ERG fusion. According to our results, a probe set consisting of PTEN, MYC, and TBL1XR1 would detect progressers with 86% sensitivity and 100% specificity. This will be evaluated further in larger studies.
Collapse
|
49
|
Fraser M, Berlin A, Bristow RG, van der Kwast T. Genomic, pathological, and clinical heterogeneity as drivers of personalized medicine in prostate cancer. Urol Oncol 2014; 33:85-94. [PMID: 24768356 DOI: 10.1016/j.urolonc.2013.10.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 12/23/2022]
Abstract
Prostate cancer (CaP) is the most commonly diagnosed malignancy in men in the Western world. In North America, more than 275,000 men are diagnosed annually, whereby approximately 1 in 6 men will be diagnosed with CaP in their lifetime, and 1 in 34 men will die from castration-resistant metastatic disease. Unfortunately, current clinical prognostic factors explain only a proportion of the observed variation in clinical outcome from patient to patient. Furthermore, overtreatment of indolent and low-risk cancers leads to inappropriate morbidity following radiotherapy or surgery. As such, better predictors of individualized prognosis and treatment response are urgently needed to triage patients to customized and intensified CaP treatment. Recent developments in next-generation sequencing have made it possible to identify prognostic and predictive signatures based on genomic profiles. We discuss the genetic basis of CaP progression from localized to systemic disease (e.g., point mutations, copy-number alterations, and structural variants) in relation with unique features of CaP biology, including intraprostatic and interprostatic heterogeneity, multifocality and multiclonality, TMPRSS2:ERG, and other ETS-family gene fusions. Finally, we focus on the use of genomic markers as prognostic factors for local failure and for systemic disease, as novel risk-stratification tools, in triaging patients to existing treatment options, and ultimately the potential of genomics for the identification of molecular targets for therapy of CaP.
Collapse
Affiliation(s)
- Michael Fraser
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada
| | - Alejandro Berlin
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Robert G Bristow
- Ontario Cancer Institute and Princess Margaret Cancer Center (University Health Network), Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Theodorus van der Kwast
- Department of Pathology and Laboratory Medicine, Toronto General Hospital (University Health Network), Toronto, Ontario, Canada.
| |
Collapse
|
50
|
Fontugne J, Lee D, Cantaloni C, Barbieri CE, Caffo O, Hanspeter E, Mazzoleni G, Dalla Palma P, Rubin MA, Fellin G, Mosquera JM, Barbareschi M, Demichelis F. Recurrent prostate cancer genomic alterations predict response to brachytherapy treatment. Cancer Epidemiol Biomarkers Prev 2014; 23:594-600. [PMID: 24515272 DOI: 10.1158/1055-9965.epi-13-1180] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the association of recurrent molecular alterations in prostate cancer, such as ERG rearrangements and phosphatase and tensin homolog gene (PTEN) deletions, with oncologic outcomes in patients with prostate cancer treated with brachytherapy. METHODS Ninety-two men underwent I-125 brachytherapy with a 145 Gy delivered dose between 2000 and 2008. Pretreatment prostate biopsies were analyzed by immunohistochemistry (IHC) and FISH for ERG rearrangement and overexpression, PTEN deletion, and expression loss. Univariable and multivariable Cox-regression analyses evaluated association of ERG and PTEN status with biochemical recurrence (BCR). RESULTS Within a median follow-up of 73 months, 11% of patients experienced BCR. Of 80 samples with both IHC and FISH performed for ERG, 46 (57.8%) demonstrated rearrangement by FISH and 45 (56.3%) by IHC. Of 77 samples with both IHC and FISH for PTEN, 14 (18.2%) had PTEN deletion by FISH and 22 (28.6%) by IHC. No significant associations were found between ERG, PTEN status, and clinicopathologic features. Patients with concurrent ERG rearrangement and PTEN deletion demonstrated significantly worse relapse-free survival rates compared with those with ERG or PTEN wild type (P < 0.01). In multivariable Cox regression analysis adjusted for the effects of standard clinicopathologic features, combined ERG rearranged and PTEN deletion was independently associated with BCR (HR = 2.6; P = 0.02). CONCLUSIONS Concurrent ERG rearrangement and PTEN loss was independently associated with time to BCR in patients undergoing brachytherapy. Future studies are needed to validate prostate cancer molecular subtyping for risk stratification. IMPACT Identifying patients in the ERG-rearranged/PTEN-deleted molecular subclass may improve treatment personalization.
Collapse
Affiliation(s)
- Jacqueline Fontugne
- Authors' Affiliations: Departments of Pathology and Laboratory Medicine and Urology; Institute for Precision Medicine; Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, New York; Centre for Integrative Biology, University of Trento; Departments of Medical Oncology, Pathology, and Radiotherapy and Medical Physics, Ospedale Santa Chiara, Trento; and Department of Surgical Pathology, Central Hospital, Bolzano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|