1
|
Tao J, Bian X, Zhou J, Zhang M. From microscopes to molecules: The evolution of prostate cancer diagnostics. Cytojournal 2024; 21:29. [PMID: 39391208 PMCID: PMC11464998 DOI: 10.25259/cytojournal_36_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 10/12/2024] Open
Abstract
In the ever-evolving landscape of oncology, the battle against prostate cancer (PCa) stands at a transformative juncture, propelled by the integration of molecular diagnostics into traditional cytopathological frameworks. This synthesis not only heralds a new epoch of precision medicine but also significantly enhances our understanding of the disease's genetic intricacies. Our comprehensive review navigates through the latest advancements in molecular biomarkers and their detection technologies, illuminating the potential these innovations hold for the clinical realm. With PCa persisting as one of the most common malignancies among men globally, the quest for early and precise diagnostic methods has never been more critical. The spotlight in this endeavor shines on the molecular diagnostics that reveal the genetic underpinnings of PCa, offering insights into its onset, progression, and resistance to conventional therapies. Among the genetic aberrations, the TMPRSS2-ERG fusion and mutations in genes such as phosphatase and tensin homolog (PTEN) and myelocytomatosis viral oncogene homolog (MYC) are identified as significant players in the disease's pathology, providing not only diagnostic markers but also potential therapeutic targets. This review underscores a multimodal diagnostic approach, merging molecular diagnostics with cytopathology, as a cornerstone in managing PCa effectively. This strategy promises a future where treatment is not only tailored to the individual's genetic makeup but also anticipates the disease's trajectory, offering hope for improved prognosis and quality of life for patients.
Collapse
Affiliation(s)
- Junyue Tao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaokang Bian
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
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
|
Zhu S, Xu N, Zeng H. Molecular complexity of intraductal carcinoma of the prostate. Cancer Med 2024; 13:e6939. [PMID: 38379333 PMCID: PMC10879723 DOI: 10.1002/cam4.6939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 02/22/2024] Open
Abstract
Intraductal carcinoma of the prostate (IDC-P) is an aggressive subtype of prostate cancer characterized by the growth of tumor cells within the prostate ducts. It is often found alongside invasive carcinoma and is associated with poor prognosis. Understanding the molecular mechanisms driving IDC-P is crucial for improved diagnosis, prognosis, and treatment strategies. This review summarizes the molecular characteristics of IDC-P and their prognostic indications, comparing them to conventional prostate acinar adenocarcinoma, to gain insights into its unique behavior and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| | - Nanwei Xu
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
4
|
Patel P, Harmon S, Iseman R, Ludkowski O, Auman H, Hawley S, Newcomb LF, Lin DW, Nelson PS, Feng Z, Boyer HD, Tretiakova MS, True LD, Vakar-Lopez F, Carroll PR, Cooperberg MR, Chan E, Simko J, Fazli L, Gleave M, Hurtado-Coll A, Thompson IM, Troyer D, McKenney JK, Wei W, Choyke PL, Bratslavsky G, Turkbey B, Siemens DR, Squire J, Peng YP, Brooks JD, Jamaspishvili T. Artificial Intelligence-Based PTEN Loss Assessment as an Early Predictor of Prostate Cancer Metastasis After Surgery: A Multicenter Retrospective Study. Mod Pathol 2023; 36:100241. [PMID: 37343766 PMCID: PMC10592257 DOI: 10.1016/j.modpat.2023.100241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023]
Abstract
Phosphatase and tensin homolog (PTEN) loss is associated with adverse outcomes in prostate cancer and can be measured via immunohistochemistry. The purpose of the study was to establish the clinical application of an in-house developed artificial intelligence (AI) image analysis workflow for automated detection of PTEN loss on digital images for identifying patients at risk of early recurrence and metastasis. Postsurgical tissue microarray sections from the Canary Foundation (n = 1264) stained with anti-PTEN antibody were evaluated independently by pathologist conventional visual scoring (cPTEN) and an automated AI-based image analysis pipeline (AI-PTEN). The relationship of PTEN evaluation methods with cancer recurrence and metastasis was analyzed using multivariable Cox proportional hazard and decision curve models. Both cPTEN scoring by the pathologist and quantification of PTEN loss by AI (high-risk AI-qPTEN) were significantly associated with shorter metastasis-free survival (MFS) in univariable analysis (cPTEN hazard ratio [HR], 1.54; CI, 1.07-2.21; P = .019; AI-qPTEN HR, 2.55; CI, 1.83-3.56; P < .001). In multivariable analyses, AI-qPTEN showed a statistically significant association with shorter MFS (HR, 2.17; CI, 1.49-3.17; P < .001) and recurrence-free survival (HR, 1.36; CI, 1.06-1.75; P = .016) when adjusting for relevant postsurgical clinical nomogram (Cancer of the Prostate Risk Assessment [CAPRA] postsurgical score [CAPRA-S]), whereas cPTEN does not show a statistically significant association (HR, 1.33; CI, 0.89-2; P = .2 and HR, 1.26; CI, 0.99-1.62; P = .063, respectively) when adjusting for CAPRA-S risk stratification. More importantly, AI-qPTEN was associated with shorter MFS in patients with favorable pathological stage and negative surgical margins (HR, 2.72; CI, 1.46-5.06; P = .002). Workflow also demonstrated enhanced clinical utility in decision curve analysis, more accurately identifying men who might benefit from adjuvant therapy postsurgery. This study demonstrates the clinical value of an affordable and fully automated AI-powered PTEN assessment for evaluating the risk of developing metastasis or disease recurrence after radical prostatectomy. Adding the AI-qPTEN assessment workflow to clinical variables may affect postoperative surveillance or management options, particularly in low-risk patients.
Collapse
Affiliation(s)
- Palak Patel
- Department of Cell Biology at The Arthur and Sonia Labatt Brain Tumour Research Centre at the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephanie Harmon
- Molecular Imaging Branch, National Cancer Institute, Bethesda, Maryland; Artificial Intelligence Resource, National Cancer Institute, Bethesda, Maryland
| | - Rachael Iseman
- Division of Cancer Biology and Genetics, Queen's University, Kingston, Ontario, Canada
| | - Olga Ludkowski
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | - Lisa F Newcomb
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Daniel W Lin
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ziding Feng
- Program of Biostatistics and Biomathematics, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Hilary D Boyer
- Program of Biostatistics and Biomathematics, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Maria S Tretiakova
- Department of Pathology, University of Washington Medical Center, Seattle, Washington
| | - Larry D True
- Department of Pathology, University of Washington Medical Center, Seattle, Washington
| | - Funda Vakar-Lopez
- Department of Pathology, University of Washington Medical Center, Seattle, Washington
| | - Peter R Carroll
- Department of Urology, University of California San Francisco and Helen Diller Family, Comprehensive Cancer Center, San Francisco, California
| | - Matthew R Cooperberg
- Department of Urology, University of California San Francisco and Helen Diller Family, Comprehensive Cancer Center, San Francisco, California
| | - Emily Chan
- Department of Urology, University of California San Francisco and Helen Diller Family, Comprehensive Cancer Center, San Francisco, California
| | - Jeff Simko
- Department of Urology, University of California San Francisco and Helen Diller Family, Comprehensive Cancer Center, San Francisco, California; Department of Pathology, University of California San Francisco, San Francisco, California
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Antonio Hurtado-Coll
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Dean Troyer
- Department of Pathology, Eastern Virginia Medical School, Norfolk, Virginia; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia
| | | | - Wei Wei
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Peter L Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, Maryland
| | | | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, Bethesda, Maryland; Artificial Intelligence Resource, National Cancer Institute, Bethesda, Maryland
| | - D Robert Siemens
- Department of Urology, Queen's University, Kingston, Ontario, Canada
| | - Jeremy Squire
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Yingwei P Peng
- Department of Public Health Sciences, Queen's University, Kingston, Ontario, Canada; Department of Mathematics and Statistics, Queen's University, Kingston, Ontario, Canada
| | - James D Brooks
- Department of Urology, Stanford University Medical Center, Stanford, California
| | - Tamara Jamaspishvili
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada; Department of Pathology and Molecular Medicine, SUNY Upstate Medical University, Syracuse, New York.
| |
Collapse
|
5
|
Alfahed A, Ebili HO, Almoammar NE, Alasiri G, AlKhamees OA, Aldali JA, Al Othaim A, Hakami ZH, Abdulwahed AM, Waggiallah HA. Prognostic Values of Gene Copy Number Alterations in Prostate Cancer. Genes (Basel) 2023; 14:genes14050956. [PMID: 37239316 DOI: 10.3390/genes14050956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Whilst risk prediction for individual prostate cancer (PCa) cases is of a high priority, the current risk stratification indices for PCa management have severe limitations. This study aimed to identify gene copy number alterations (CNAs) with prognostic values and to determine if any combination of gene CNAs could have risk stratification potentials. Clinical and genomic data of 500 PCa cases from the Cancer Genome Atlas stable were retrieved from the Genomic Data Commons and cBioPortal databases. The CNA statuses of a total of 52 genetic markers, including 21 novel markers and 31 previously identified potential prognostic markers, were tested for prognostic significance. The CNA statuses of a total of 51/52 genetic markers were significantly associated with advanced disease at an odds ratio threshold of ≥1.5 or ≤0.667. Moreover, a Kaplan-Meier test identified 27/52 marker CNAs which correlated with disease progression. A Cox Regression analysis showed that the amplification of MIR602 and deletions of MIR602, ZNF267, MROH1, PARP8, and HCN1 correlated with a progression-free survival independent of the disease stage and Gleason prognostic group grade. Furthermore, a binary logistic regression analysis identified twenty-two panels of markers with risk stratification potentials. The best model of 7/52 genetic CNAs, which included the SPOP alteration, SPP1 alteration, CCND1 amplification, PTEN deletion, CDKN1B deletion, PARP8 deletion, and NKX3.1 deletion, stratified the PCa cases into a localised and advanced disease with an accuracy of 70.0%, sensitivity of 85.4%, specificity of 44.9%, positive predictive value of 71.67%, and negative predictive value of 65.35%. This study validated prognostic gene level CNAs identified in previous studies, as well as identified new genetic markers with CNAs that could potentially impact risk stratification in PCa.
Collapse
Affiliation(s)
- Abdulaziz Alfahed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Henry Okuchukwu Ebili
- Department of Morbid Anatomy and Histopathology, Olabisi Onabanjo University, Ago-Iwoye P.M.B. 2002, Nigeria
| | - Nasser Eissa Almoammar
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Glowi Alasiri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud University, Riyadh 13317, Saudi Arabia
| | - Osama A AlKhamees
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Jehad A Aldali
- Department of Pathology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratories, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan 82817, Saudi Arabia
| | - Abdulhadi M Abdulwahed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| |
Collapse
|
6
|
Harmon SA, Gesztes W, Young D, Mehralivand S, McKinney Y, Sanford T, Sackett J, Cullen J, Rosner IL, Srivastava S, Merino MJ, Wood BJ, Pinto PA, Choyke PL, Dobi A, Sesterhenn IA, Turkbey B. Prognostic Features of Biochemical Recurrence of Prostate Cancer Following Radical Prostatectomy Based on Multiparametric MRI and Immunohistochemistry Analysis of MRI-guided Biopsy Specimens. Radiology 2021; 299:613-623. [PMID: 33847515 PMCID: PMC8165944 DOI: 10.1148/radiol.2021202425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/07/2020] [Accepted: 02/16/2021] [Indexed: 12/18/2022]
Abstract
Background Although prostate MRI is routinely used for the detection and staging of localized prostate cancer, imaging-based assessment and targeted molecular sampling for risk stratification are an active area of research. Purpose To evaluate features of preoperative MRI and MRI-guided biopsy immunohistochemistry (IHC) findings associated with biochemical recurrence (BCR) of prostate cancer after surgery. Materials and Methods In this retrospective case-control study, patients underwent multiparametric MRI before MRI-guided biopsy followed by radical prostatectomy between 2008 and 2016. Lesions were retrospectively scored with the Prostate Imaging Reporting and Data System (PI-RADS) (version 2) by radiologists who were blinded to the clinical-pathologic results. The IHC staining, including stains for the ETS-related gene, phosphatase and tensin homolog, androgen receptor, prostate specific antigen, and p53, was performed with targeted biopsy specimens of the index lesion (highest suspicion at MRI and pathologic grade) and scored by pathologists who were blinded to clinical-pathologic outcomes. Cox proportional hazards regression analysis was used to evaluate associations with recurrence-free survival (RFS). Results The median RFS was 31.7 months (range, 1-101 months) for 39 patients (median age, 62 years; age range, 47-76 years) without BCR and 14.6 months (range, 1-61 months) for 40 patients (median age, 59 years; age range, 47-73 years) with BCR. MRI features that showed a significant relationship with the RFS interval included an index lesion with a PI-RADS score of 5 (hazard ratio [HR], 2.10; 95% CI: 1.05, 4.21; P = .04); index lesion burden, defined as ratio of index lesion volume to prostate volume (HR, 1.55; 95% CI: 1.2, 2.1; P = .003); and suspicion of extraprostatic extension (EPE) (HR, 2.18; 95% CI: 1.1, 4.2; P = .02). Presurgical multivariable analysis indicated that suspicion of EPE at MRI (adjusted HR, 2.19; 95% CI: 1.1, 4.3; P = .02) and p53 stain intensity (adjusted HR, 2.22; 95% CI: 1.0, 4.7; P = .04) were significantly associated with RFS. Conclusion MRI features, including Prostate Imaging Reporting and Data System score, index lesion burden, extraprostatic extension, and preoperative guided biopsy p53 immunohistochemistry stain intensity are associated with biochemical relapse of prostate cancer after surgery. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Costa in this issue.
Collapse
Affiliation(s)
| | | | - Denise Young
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Sherif Mehralivand
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Yolanda McKinney
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Thomas Sanford
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Jonathan Sackett
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Jennifer Cullen
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Inger L. Rosner
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Shiv Srivastava
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Maria J. Merino
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Bradford J. Wood
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Peter A. Pinto
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Peter L. Choyke
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Albert Dobi
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Isabell A. Sesterhenn
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| | - Baris Turkbey
- From the Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute (S.A.H.); Molecular Imaging Branch (S.A.H., S.M., Y.M., T.S., J.S., P.L.C., B.T.), Laboratory of Pathology (M.J.M.), Center for Interventional Oncology (B.J.W.), and Urologic Oncology Branch (S.M., P.A.P.), National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B85, Bethesda, Md 20892; Center for Prostate Disease Research, John P. Murtha Cancer Center, Department of Surgery, Uniformed Services University of the Health Sciences (W.G., D.Y., J.C., I.L.R., S.S., A.D., I.A.S.) and Urology Service (I.L.R.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Genitourinary Pathology, Joint Pathology Center, Silver Spring, Md (I.A.S.)
| |
Collapse
|
7
|
Jamaspishvili T, Patel PG, Niu Y, Vidotto T, Caven I, Livergant R, Fu W, Kawashima A, How N, Okello JB, Guedes LB, Ouellet V, Picanço C, Koti M, Reis RB, Saad F, Mes-Masson AM, Lotan TL, Squire JA, Peng YP, Siemens DR, Berman DM. Risk Stratification of Prostate Cancer Through Quantitative Assessment of PTEN Loss (qPTEN). J Natl Cancer Inst 2021; 112:1098-1104. [PMID: 32129857 DOI: 10.1093/jnci/djaa032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 12/25/2019] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Phosphatase and tensin homolog (PTEN) loss has long been associated with adverse findings in early prostate cancer. Studies to date have yet to employ quantitative methods (qPTEN) for measuring of prognostically relevant amounts of PTEN loss in postsurgical settings and demonstrate its clinical application. METHODS PTEN protein levels were measured by immunohistochemistry in radical prostatectomy samples from training (n = 410) and validation (n = 272) cohorts. PTEN loss was quantified per cancer cell and per tissue microarray core. Thresholds for identifying clinically relevant PTEN loss were determined using log-rank statistics in the training cohort. Univariate (Kaplan-Meier) and multivariate (Cox proportional hazards) analyses on various subpopulations were performed to assess biochemical recurrence-free survival (BRFS) and were independently validated. All statistical tests were two-sided. RESULTS PTEN loss in more than 65% cancer cells was most clinically relevant and had statistically significant association with reduced BRFS in training (hazard ratio [HR] = 2.48, 95% confidence interval [CI] = 1.59 to 3.87; P < .001) and validation cohorts (HR = 4.22, 95% CI = 2.01 to 8.83; P < .001). The qPTEN scoring method identified patients who recurred within 5.4 years after surgery (P < .001). In men with favorable risk of biochemical recurrence (Cancer of the Prostate Risk Assessment - Postsurgical scores <5 and no adverse pathological features), qPTEN identified a subset of patients with shorter BRFS (HR = 5.52, 95% CI = 2.36 to 12.90; P < .001) who may be considered for intensified monitoring and/or adjuvant therapy. CONCLUSIONS Compared with previous qualitative approaches, qPTEN improves risk stratification of postradical prostatectomy patients and may be considered as a complementary tool to guide disease management after surgery.
Collapse
Affiliation(s)
- Tamara Jamaspishvili
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Palak G Patel
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Yi Niu
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Thiago Vidotto
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada.,Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Isabelle Caven
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Rachel Livergant
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Winnie Fu
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Atsunari Kawashima
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.,Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Nathan How
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - John B Okello
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Liana B Guedes
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Veronique Ouellet
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Clarissa Picanço
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Madhuri Koti
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada.,Urology, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Rodolfo B Reis
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil
| | - Fred Saad
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Department of Surgery, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Anne-Marie Mes-Masson
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Department of Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jeremy A Squire
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Yingwei P Peng
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Public Health Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.,Mathematics and Statistics, Queen's University, Kingston, ON K7L 3N6, Canada
| | | | - David M Berman
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada
| |
Collapse
|
8
|
Sopyllo K, Erickson AM, Mirtti T. Grading Evolution and Contemporary Prognostic Biomarkers of Clinically Significant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13040628. [PMID: 33562508 PMCID: PMC7914622 DOI: 10.3390/cancers13040628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Prostate cancer treatment decisions are based on clinical stage and histological diagnosis, including Gleason grading assessed by a pathologist, in biopsies. Prior to staging and grading, serum or blood prostate-specific antigen (PSA) levels are measured and often trigger diagnostic examinations. However, PSA is best suited as a marker of cancer relapse after initial treatment. In this review, we first narratively describe the evolution of histological grading, the current status of Gleason pattern-based diagnostics and glance into future methodology of risk assessment by histological examination. In the second part, we systematically review the biomarkers that have been shown, independent from clinical characteristics, to correlate with clinically relevant end-points, i.e., occurrence of metastases, disease-specific mortality and overall survival after initial treatment of localized prostate cancer. Abstract Gleason grading remains the strongest prognostic parameter in localized prostate adenocarcinoma. We have here outlined the evolution and contemporary practices in pathological evaluation of prostate tissue samples for Gleason score and Grade group. The state of more observer-independent grading methods with the aid of artificial intelligence is also reviewed. Additionally, we conducted a systematic review of biomarkers that hold promise in adding independent prognostic or predictive value on top of clinical parameters, Grade group and PSA. We especially focused on hard end points during the follow-up, i.e., occurrence of metastasis, disease-specific mortality and overall mortality. In peripheral blood, biopsy-detected prostate cancer or in surgical specimens, we can conclude that there are more than sixty biomarkers that have been shown to have independent prognostic significance when adjusted to conventional risk assessment or grouping. Our search brought up some known putative markers and panels, as expected. Also, the synthesis in the systematic review indicated markers that ought to be further studied as part of prospective trials and in well characterized patient cohorts in order to increase the resolution of the current clinico-pathological prognostic factors.
Collapse
Affiliation(s)
- Konrad Sopyllo
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Andrew M. Erickson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK;
| | - Tuomas Mirtti
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Department of Pathology, HUS Diagnostic Centre, Helsinki University Hospital, 00029 Helsinki, Finland
- Correspondence:
| |
Collapse
|
9
|
Harmon SA, Patel PG, Sanford TH, Caven I, Iseman R, Vidotto T, Picanço C, Squire JA, Masoudi S, Mehralivand S, Choyke PL, Berman DM, Turkbey B, Jamaspishvili T. High throughput assessment of biomarkers in tissue microarrays using artificial intelligence: PTEN loss as a proof-of-principle in multi-center prostate cancer cohorts. Mod Pathol 2021; 34:478-489. [PMID: 32884130 PMCID: PMC9152638 DOI: 10.1038/s41379-020-00674-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Phosphatase and tensin homolog (PTEN) loss is associated with adverse outcomes in prostate cancer and has clinical potential as a prognostic biomarker. The objective of this work was to develop an artificial intelligence (AI) system for automated detection and localization of PTEN loss on immunohistochemically (IHC) stained sections. PTEN loss was assessed using IHC in two prostate tissue microarrays (TMA) (internal cohort, n = 272 and external cohort, n = 129 patients). TMA cores were visually scored for PTEN loss by pathologists and, if present, spatially annotated. Cores from each patient within the internal TMA cohort were split into 90% cross-validation (N = 2048) and 10% hold-out testing (N = 224) sets. ResNet-101 architecture was used to train core-based classification using a multi-resolution ensemble approach (×5, ×10, and ×20). For spatial annotations, single resolution pixel-based classification was trained from patches extracted at ×20 resolution, interpolated to ×40 resolution, and applied in a sliding-window fashion. A final AI-based prediction model was created from combining multi-resolution and pixel-based models. Performance was evaluated in 428 cores of external cohort. From both cohorts, a total of 2700 cores were studied, with a frequency of PTEN loss of 14.5% in internal (180/1239) and external 13.5% (43/319) cancer cores. The final AI-based prediction of PTEN status demonstrated 98.1% accuracy (95.0% sensitivity, 98.4% specificity; median dice score = 0.811) in internal cohort cross-validation set and 99.1% accuracy (100% sensitivity, 99.0% specificity; median dice score = 0.804) in internal cohort test set. Overall core-based classification in the external cohort was significantly improved in the external cohort (area under the curve = 0.964, 90.6% sensitivity, 95.7% specificity) when further trained (fine-tuned) using 15% of cohort data (19/124 patients). These results demonstrate a robust and fully automated method for detection and localization of PTEN loss in prostate cancer tissue samples. AI-based algorithms have potential to streamline sample assessment in research and clinical laboratories.
Collapse
Affiliation(s)
- Stephanie A Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Palak G Patel
- Division of Cancer Biology & Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
- Department of Cell Biology at The Arthur and Sonia Labatt Brain Tumour Research Centre at the Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas H Sanford
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Urology, Upstate Medical University, Syracuse, NY, USA
| | - Isabelle Caven
- Division of Cancer Biology & Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Rachael Iseman
- Division of Cancer Biology & Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Thiago Vidotto
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Clarissa Picanço
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Samira Masoudi
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sherif Mehralivand
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter L Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David M Berman
- Division of Cancer Biology & Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tamara Jamaspishvili
- Division of Cancer Biology & Genetics, Cancer Research Institute, Queen's University, Kingston, ON, Canada.
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
10
|
Vlajnic T, Bubendorf L. Molecular pathology of prostate cancer: a practical approach. Pathology 2020; 53:36-43. [PMID: 33234230 DOI: 10.1016/j.pathol.2020.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
While localised prostate cancer can be cured by local treatment, 'high-risk' prostate cancer often progresses to castration resistant disease and remains incurable with a dismal prognosis. In recent years, technical advances and development of novel methodologies have largely contributed to a better understanding of underlying molecular mechanisms that promote tumour growth and progression. Consecutively, novel therapeutic strategies for treatment of prostate cancer have emerged during the last decade, calling for the identification of predictive biomarkers. The concept of personalised medicine is to tailor treatment according to the specific tumour profile of an individual patient. Moreover, acquired molecular changes during tumour evolution and in response to therapy selection pressure require adapted predictive marker testing at different time points during the disease. In this setting, the pathologist plays a critical role in patient management and treatment selection. In this review, we provide a comprehensive overview of the current knowledge of molecular aspects of prostate cancer and their potential utility in the context of different therapeutic approaches. Furthermore, we discuss methods for molecular marker testing in routine clinical practice, with a focus on castration resistant prostate cancer.
Collapse
Affiliation(s)
- Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
11
|
Liu W, Hou J, Petkewicz J, Na R, Wang CH, Sun J, Gallagher J, Bogachkov YY, Swenson L, Regner M, Resurreccion WK, Isaacs WB, Brendler CB, Crawford S, Zheng SL, Helfand BT, Xu J. Feasibility and performance of a novel probe panel to detect somatic DNA copy number alterations in clinical specimens for predicting prostate cancer progression. Prostate 2020; 80:1253-1262. [PMID: 32803894 DOI: 10.1002/pros.24057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/03/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND To assess the feasibility of a novel DNA-based probe panel to detect copy number alterations (CNAs) in prostate tumor DNA and its performance for predicting clinical progression. METHODS A probe panel was developed and optimized to measure CNAs in trace amounts of tumor DNA (2 ng) isolated from formalin-fixed paraffin-embedded tissues. Ten genes previously associated with aggressive disease were targeted. The panel's feasibility and performance were assessed in 175 prostate cancer (PCa) patients who underwent radical prostatectomy with a median 10-year follow-up, including 42 men who developed disease progression (either metastasis and/or PCa-specific death). Association with disease progression was tested using univariable and multivariable analyses. RESULTS The probe panel detected CNAs in all 10 genes in tumor DNA isolated from either diagnostic biopsies or surgical specimens. A four-gene model (PTEN/MYC/BRCA2/CDKN1B) had the strongest association with disease progression; 64.3% of progressors and 22.5% of non-progressors had at least one CNA in these four genes, odds ratio (OR) (95% confidence interval) = 6.21 (2.77-13.87), P = 8.48E-06. The association with disease progression remained significant after adjusting for known clinicopathological variables. Among the seven progressors of the 65 patients with clinically low-risk disease, three (42.9%) had at least one CNA in these four genes. CONCLUSIONS The probe panel can detect CNAs in trace amounts of tumor DNA from biopsies or surgical tissues at the time of diagnosis or surgery. CNAs independently predict metastatic/lethal cancer, particularly among men with clinically low-risk disease at diagnosis. If validated, this may improve current abilities to assess tumor aggressiveness.
Collapse
Affiliation(s)
- Wennuan Liu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Jun Hou
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Jacqueline Petkewicz
- John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Rong Na
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Chi-Hsiung Wang
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Jishan Sun
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Johnie Gallagher
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Yedida Y Bogachkov
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Laura Swenson
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - MaryAnn Regner
- Department of Pathology, NorthShore University HealthSystem, Evanston, Illinois
| | - W Kyle Resurreccion
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - William B Isaacs
- Department of Urology and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Charles B Brendler
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Susan Crawford
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - S Lilly Zheng
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Brian T Helfand
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
- John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Jianfeng Xu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois
| |
Collapse
|
12
|
Erickson AM, Lokman U, Lahdensuo K, Tornberg S, Visapaa H, Bergroth R, Santti H, Petas A, Rannikko AS, Mirtti T. PTEN and ERG expression in MRI-ultrasound guided fusion biopsy correlated with radical prostatectomy findings in men with prostate cancer. Prostate 2020; 80:1118-1127. [PMID: 32634262 DOI: 10.1002/pros.24040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Conventional systematic prostate biopsies (SBx) have multiple limitations, and magnetic resonance imaging (MRI)-ultrasound fusion targeting is increasingly applied (fusion biopsies [FBx]). In our previous studies, we have shown that loss of the tumor suppressor gene phosphatase and tensin homolog (PTEN) in radical prostatectomy (RP) specimens predicts poor disease-specific survival, and in active surveillance (AS), PTEN loss in SBx predicts an adverse AS outcome, although SBx PTEN status does not correlate well with the corresponding RP status. Here, we have hypothesized that PTEN and erythroblast transformation-specific related gene (ERG) status in FBx correlate better with RP than they would in SBx. METHODS A total of 106 men, who had undergone FBx and subsequent RP in a single center between June 2015 and May 2017 were included. Fifty-three of the men had concomitant or previous SBx's. All biopsy and RP specimens were collected, and tissue microarrays (TMA) were constructed from RP specimens. Immunohistochemical stainings for PTEN and ERG expression were conducted on biopsies and RP TMAs and results were compared by using Fisher's exact test. RESULTS The immunohistochemical predictive power of FBx, determined by the concordance of biopsy PTEN and ERG status with RP, is superior to SBx (77.6% vs 66.7% in PTEN, 92.4% vs 66.6% in ERG). FBx was superior to SBx in correlation with RP Gleason Grade Groups and MRI prostate imaging reporting and data system scores. CONCLUSION FBx grading correlates with RP histology and MRI findings and predicts the biomarker status in the RP specimens more accurately than SBx. A longer follow-up is needed to evaluate if this translates to better prediction of disease outcomes, especially in AS and radiation therapy where prostatectomy specimens are not available for prognostication.
Collapse
Affiliation(s)
- Andrew M Erickson
- Department of Pathology (HUS Diagnostic Center) and Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Utku Lokman
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Urology, Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | - Kanerva Lahdensuo
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sara Tornberg
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Visapaa
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Robin Bergroth
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Henrikki Santti
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anssi Petas
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti S Rannikko
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology (HUS Diagnostic Center) and Medicum, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Mohler JL, Antonarakis ES, Armstrong AJ, D'Amico AV, Davis BJ, Dorff T, Eastham JA, Enke CA, Farrington TA, Higano CS, Horwitz EM, Hurwitz M, Ippolito JE, Kane CJ, Kuettel MR, Lang JM, McKenney J, Netto G, Penson DF, Plimack ER, Pow-Sang JM, Pugh TJ, Richey S, Roach M, Rosenfeld S, Schaeffer E, Shabsigh A, Small EJ, Spratt DE, Srinivas S, Tward J, Shead DA, Freedman-Cass DA. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 17:479-505. [PMID: 31085757 DOI: 10.6004/jnccn.2019.0023] [Citation(s) in RCA: 869] [Impact Index Per Article: 217.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The NCCN Guidelines for Prostate Cancer include recommendations regarding diagnosis, risk stratification and workup, treatment options for localized disease, and management of recurrent and advanced disease for clinicians who treat patients with prostate cancer. The portions of the guidelines included herein focus on the roles of germline and somatic genetic testing, risk stratification with nomograms and tumor multigene molecular testing, androgen deprivation therapy, secondary hormonal therapy, chemotherapy, and immunotherapy in patients with prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Joseph E Ippolito
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Jesse McKenney
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - George Netto
- University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | | | | | - Sylvia Richey
- St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | - Mack Roach
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | - Edward Schaeffer
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Ahmad Shabsigh
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Eric J Small
- UCSF Helen Diller Family Comprehensive Cancer Center
| | | | | | - Jonathan Tward
- Huntsman Cancer Institute at the University of Utah; and
| | | | | |
Collapse
|
14
|
Norris JM, Simpson BS, Parry MA, Allen C, Ball R, Freeman A, Kelly D, Kim HL, Kirkham A, You S, Kasivisvanathan V, Whitaker HC, Emberton M. Genetic Landscape of Prostate Cancer Conspicuity on Multiparametric Magnetic Resonance Imaging: A Systematic Review and Bioinformatic Analysis. EUR UROL SUPPL 2020; 20:37-47. [PMID: 33000006 PMCID: PMC7497895 DOI: 10.1016/j.euros.2020.06.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Context Multiparametric magnetic resonance imaging (mpMRI) detects most, but not all, clinically significant prostate cancer. The genetic basis of prostate cancer visibility and invisibility on mpMRI remains uncertain. Objective To systematically review the literature on differential gene expression between mpMRI-visible and mpMRI-invisible prostate cancer, and to use bioinformatic analysis to identify enriched processes or cellular components in genes validated in more than one study. Evidence acquisition We performed a systematic literature search of the Medline, EMBASE, PubMed, and Cochrane databases up to January 2020 in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. The primary endpoint was differential genetic features between mpMRI-visible and mpMRI-invisible tumours. Secondary endpoints were explanatory links between gene function and mpMRI conspicuity, and the prognostic value of differential gene enrichment. Evidence synthesis We retrieved 445 articles, of which 32 met the criteria for inclusion. Thematic synthesis from the included studies showed that mpMRI-visible cancer tended towards enrichment of molecular features associated with increased disease aggressivity, including phosphatase and tensin homologue (PTEN) loss and higher genomic classifier scores, such as Oncotype and Decipher. Three of the included studies had accompanying publicly available data suitable for further bioinformatic analysis. An over-representation analysis of these datasets revealed increased expression of genes associated with extracellular matrix components in mpMRI-visible tumours. Conclusions Prostate cancer that is visible on mpMRI is generally enriched with molecular features of tumour development and aggressivity, including activation of proliferative signalling, DNA damage, and inflammatory processes. Additionally, there appears to be concordant cellular components and biological processes associated with mpMRI conspicuity, as highlighted by bioinformatic analysis of large genetic datasets. Patient summary Prostate cancer that is detected by magnetic resonance imaging (MRI) tends to have genetic features that are associated with more aggressive disease. This suggests that MRI can be used to assess the likelihood of aggressive prostate cancer, based on tumour visibility.
Collapse
Affiliation(s)
- Joseph M Norris
- UCL Division of Surgery & Interventional Science, University College London, London, UK.,London Deanery of Urology, London, UK.,Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Benjamin S Simpson
- UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - Marina A Parry
- UCL Cancer Institute, University College London, London, UK
| | - Clare Allen
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rhys Ball
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Alex Freeman
- Department of Pathology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Daniel Kelly
- School of Healthcare Sciences, Cardiff University, Wales, UK
| | - Hyung L Kim
- Department of Urology, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Alex Kirkham
- Department of Radiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, West Hollywood, CA, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA, USA
| | - Veeru Kasivisvanathan
- UCL Division of Surgery & Interventional Science, University College London, London, UK.,Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hayley C Whitaker
- UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - Mark Emberton
- UCL Division of Surgery & Interventional Science, University College London, London, UK.,Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
15
|
Nóbrega MD, Cilião HL, Souza MFD, Souza MRD, Serpeloni JM, Fuganti PE, Cólus IMDS. Association of polymorphisms of PTEN, AKT1, PI3K, AR, and AMACR genes in patients with prostate cancer. Genet Mol Biol 2020; 43:e20180329. [PMID: 32484847 PMCID: PMC7271063 DOI: 10.1590/1678-4685-gmb-2018-0329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Polymorphic variants in the PTEN (rs2735343), PI3K (rs2699887), AKT1 (rs2494750), AR (rs17302090), and AMACR (rs3195676) genes were evaluated as possible molecular markers of susceptibility, prognosis, and progression of prostate cancer (PCa), in a case-control study. Samples consisted of 277 patients with PCa and 277 controls from Londrina, PR, Brazil. SNPs were analyzed by real-time PCR. A family history of cancer, including PCa, as well as level of schooling were risk factors for PCa. The data were obtained via logistic regression, using odds ratios with a CI 95%. The genotypes of AKT1 and AKT1+AR demonstrated an association with protection for the disease. The combination of SNPs with the histopathological tumor data between allele variants of AMACR, AKT1+AR, and AKT1+AMACR indicated an association with protection against seminal vesicle invasion. The polymorphisms AKT1+AR and PI3K+AR were associated with protection against tumor bilaterality. The genotype combinations PTEN+AMACR and PTEN+AR were associated with the risk of extracapsular extension. Of the five genes studied, two were associated with protection for PCa, four were associated with protection for some prognostic variables, and only one was associated with risk. Thus, these SNPs are candidates for markers to discriminate men with better or worse prognosis for PCa.
Collapse
Affiliation(s)
- Monyse de Nóbrega
- Universidade Estadual de Londrina (UEL), Departamento de Biologia Geral, Londrina, PR, Brazil
| | - Heloisa Lizotti Cilião
- Universidade Estadual de Londrina (UEL), Departamento de Biologia Geral, Londrina, PR, Brazil
| | | | - Milene Roldão de Souza
- Universidade Estadual de Londrina (UEL), Departamento de Biologia Geral, Londrina, PR, Brazil
| | - Juliana Mara Serpeloni
- Universidade Estadual de Londrina (UEL), Departamento de Biologia Geral, Londrina, PR, Brazil
| | | | | |
Collapse
|
16
|
Eggener SE, Rumble RB, Armstrong AJ, Morgan TM, Crispino T, Cornford P, van der Kwast T, Grignon DJ, Rai AJ, Agarwal N, Klein EA, Den RB, Beltran H. Molecular Biomarkers in Localized Prostate Cancer: ASCO Guideline. J Clin Oncol 2019; 38:1474-1494. [PMID: 31829902 DOI: 10.1200/jco.19.02768] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE This guideline provides recommendations for available tissue-based prostate cancer biomarkers geared toward patient selection for active surveillance, identification of clinically significant disease, choice of postprostatectomy adjuvant versus salvage radiotherapy, and to address emerging questions such as the relative value of tissue biomarkers compared with magnetic resonance imaging. METHODS An ASCO multidisciplinary Expert Panel, with representatives from the European Association of Urology, American Urological Association, and the College of American Pathologists, conducted a systematic literature review of localized prostate cancer biomarker studies between January 2013 and January 2019. Numerous tissue-based molecular biomarkers were evaluated for their prognostic capabilities and potential for improving management decisions. Here, the Panel makes recommendations regarding the clinical use and indications of these biomarkers. RESULTS Of 555 studies identified, 77 were selected for inclusion plus 32 additional references selected by the Expert Panel. Few biomarkers had rigorous testing involving multiple cohorts and only 5 of these tests are commercially available currently: Oncotype Dx Prostate, Prolaris, Decipher, Decipher PORTOS, and ProMark. With various degrees of value and validation, multiple biomarkers have been shown to refine risk stratification and can be considered for select men to improve management decisions. There is a paucity of prospective studies assessing short- and long-term outcomes of patients when these markers are integrated into clinical decision making. RECOMMENDATIONS Tissue-based molecular biomarkers (evaluating the sample with the highest volume of the highest Gleason pattern) may improve risk stratification when added to standard clinical parameters, but the Expert Panel endorses their use only in situations in which the assay results, when considered as a whole with routine clinical factors, are likely to affect a clinical decision. These assays are not recommended for routine use as they have not been prospectively tested or shown to improve long-term outcomes-for example, quality of life, need for treatment, or survival. Additional information is available at www.asco.org/genitourinary-cancer-guidelines.
Collapse
Affiliation(s)
| | | | | | - Todd M Morgan
- University of Michigan School of Medicine, Ann Arbor, MI
| | | | - Philip Cornford
- Royal Liverpool University Hospital, Liverpool, United Kingdom
| | | | | | - Alex J Rai
- Columbia University Irving Medical Center, New York, NY
| | | | | | | | | |
Collapse
|
17
|
Liu R, Zhou J, Xia S, Li T. The impact of PTEN deletion and ERG rearrangement on recurrence after treatment for prostate cancer: a systematic review and meta-analysis. Clin Transl Oncol 2019; 22:694-702. [DOI: 10.1007/s12094-019-02170-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/28/2019] [Indexed: 11/28/2022]
|
18
|
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: 40] [Impact Index Per Article: 8.0] [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
|
19
|
Bramhecha YM, Rouzbeh S, Guérard KP, Scarlata E, Brimo F, Chevalier S, Hamel L, Aprikian AG, Lapointe J. The combination of PTEN deletion and 16p13.3 gain in prostate cancer provides additional prognostic information in patients treated with radical prostatectomy. Mod Pathol 2019; 32:128-138. [PMID: 30140035 DOI: 10.1038/s41379-018-0107-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/16/2018] [Accepted: 06/16/2018] [Indexed: 12/17/2022]
Abstract
Prostate cancer is a clinically heterogeneous disease and accurately risk-stratifying patients is a key clinical challenge. We hypothesized that the concurrent identification of the DNA copy number alterations 10q23.3 (PTEN) deletion and 16p13.3 (PDPK1) gain, related to the PI3K/AKT survival pathway, would improve prognostication. We assessed PTEN deletion status using fluorescence in situ hybridization (FISH) and evaluated its clinical significance in combination with the 16p13.3 gain in a set of 332 primary radical prostatectomy cases on a tissue microarray with clinical follow-up. The PTEN deletion was detected in 34% (97/287) of the evaluable tumors and was significantly associated with high Gleason grade group (P < 0.0001) and advanced pathological tumor stage (pT-stage, P < 0.001). The PTEN deletion emerged as a significant predictor of biochemical recurrence independent of the standard clinicopathologic parameters (hazard ratio: 3.00, 95% confidence interval: 1.81-4.98; P < 0.0001) and further stratified patients with low and intermediate risk of biochemical recurrence [Gleason grade group 1-2 (≤3 + 4), Gleason grade group 2 (3 + 4), pT2, prostate-specific antigen ≤ 10, low and intermediate CAPRA-S score; log-rank P ≤ 0.007]. A PTEN deletion also increased the risk of distant metastasis (log-rank, P = 0.001), further supporting its role in prostate cancer progression. Combining both 16p13.3 gain and PTEN deletion improved biochemical recurrence risk stratification and provided prognostic information beyond the established CAPRA-S score (co-alteration: hazard ratio: 4.70, 95% confidence interval: 2.12-10.42; P < 0.0001). Our study demonstrates the potential clinical utility of PTEN genomic deletion in low-intermediate risk patients and highlights the enhanced prognostication achieved when assessed in combination with another genomic biomarker related to the PI3K/AKT pathway, thereby supporting their promising usefulness in clinical management of prostate cancer.
Collapse
Affiliation(s)
- Yogesh M Bramhecha
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Shaghayegh Rouzbeh
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Karl-Philippe Guérard
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Eleonora Scarlata
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Simone Chevalier
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lucie Hamel
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Armen G Aprikian
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jacques Lapointe
- Division of Urology, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada. .,Division of Experimental Medicine, McGill University and the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
20
|
Comparing Prognostic Utility of a Single-marker Immunohistochemistry Approach with Commercial Gene Expression Profiling Following Radical Prostatectomy. Eur Urol 2018; 74:668-675. [DOI: 10.1016/j.eururo.2018.08.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/13/2018] [Indexed: 11/18/2022]
|
21
|
Guedes LB, Morais CL, Fedor H, Hicks J, Gurel B, Melamed J, Lee P, Gopalan A, Knudsen BS, True LD, Scher HI, Fine SW, Trock BJ, De Marzo AM, Lotan TL. Effect of Preanalytic Variables on an Automated PTEN Immunohistochemistry Assay for Prostate Cancer. Arch Pathol Lab Med 2018; 143:338-348. [DOI: 10.5858/arpa.2018-0068-oa] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Phosphatase and tensin homolog (PTEN) is a promising prognostic and potentially predictive biomarker in prostate cancer.
Objective.—
To assess the effects of preanalytic variables on an analytically validated and fully automated PTEN immunohistochemistry assay.
Design.—
PTEN immunohistochemistry was performed on Ventana immunostaining systems. In benign prostate tissues, immunostaining intensity across variable conditions was assessed by digital image analysis. In prostate tumor tissues, immunostaining was scored visually.
Results.—
Delay of fixation for 4 hours or longer at room temperature or 48 hours or longer at 4°C and duration of formalin fixation did not significantly alter immunostaining intensity. Intensity of staining was highest in 10% formalin compared with other fixatives. Tumor tissues with PTEN loss processed using protocols from 11 academic institutions were all evaluable and scored identically. PTEN immunostaining of needle biopsies where tissue blocks had been stored for less than 10 years was more frequently scored as nonevaluable compared with blocks that had been stored for 10 years or longer. This effect was less evident for radical prostatectomy specimens, where low rates of nonevaluable staining were seen for 23 years or more of storage. Storage of unstained slides for 5 years at room temperature prior to immunostaining resulted in equivalent scoring compared with freshly cut slides. Machine-to-machine variability assessed across 3 Ventana platforms and 2 institutions was negligible in 12 tumors, and platform-to-platform variability was also minor comparing Ventana and Leica instruments across 77 tumors (κ = 0.926).
Conclusions.—
Automated PTEN immunostaining is robust to most preanalytic variables in the prostate and may be performed on prostate tumor tissues subjected to a wide range of preanalytic conditions. These data may help guide assay development if PTEN becomes a key predictive biomarker.
Collapse
Affiliation(s)
- Liana B. Guedes
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Carlos L. Morais
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Helen Fedor
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Jessica Hicks
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Bora Gurel
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Jonathan Melamed
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Peng Lee
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Anuradha Gopalan
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Beatrice S. Knudsen
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Lawrence D. True
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Howard I. Scher
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Samson W. Fine
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Bruce J. Trock
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Angelo M. De Marzo
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| | - Tamara L. Lotan
- From the Departments of Pathology (Drs Guedes, Morais, Fedor, Hicks, Gurel, De Marzo, and Lotan), Oncology (Drs Trock, De Marzo, and Lotan), and Urology (Drs Trock and De Marzo), Johns Hopkins University School of Medicine, Baltimore, Maryland; the Department of Pathology, New York University School of Medicine, New York, New York (Drs Melamed and Lee); the Department of Pathology, Memorial Sloan
| |
Collapse
|
22
|
Ouellet V, Aprikian A, Bergeron A, Brimo F, Bristow RG, Chevalier S, Drachenberg D, Fazli L, Fleshner NE, Gleave M, Karakiewicz P, Klotz L, Lacombe L, Lattouf JB, van der Kwast T, Squire JA, Latour M, Trudel D, Mes-Masson AM, Saad F. The Terry Fox Research Institute Canadian Prostate Cancer Biomarker Network: an analysis of a pan-Canadian multi-center cohort for biomarker validation. BMC Urol 2018; 18:78. [PMID: 30200929 PMCID: PMC6131811 DOI: 10.1186/s12894-018-0392-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Refinement of parameters defining prostate cancer (PC) prognosis are urgently needed to identify patients with indolent versus aggressive disease. The Canadian Prostate Cancer Biomaker Network (CPCBN) consists of researchers from four Canadian provinces to create a validation cohort to address issues dealing with PC diagnosis and management. METHODS A total of 1512 radical prostatectomy (RP) specimens from five different biorepositories affiliated with teaching hospitals were selected to constitute the cohort. Tumoral and adjacent benign tissues were arrayed on tissue microarrays (TMAs). A patient clinical database was developed and includes data on diagnosis, treatment and clinical outcome. RESULTS Mean age at diagnosis of patients in the cohort was 61 years. Of these patients, 31% had a low grade (≤6) Gleason score (GS), 55% had GS 7 (40% of 3 + 4 and 15% of 4 + 3) and 14% had high GS (≥8) PC. The median follow-up of the cohort was 113 months. A total of 34% had a biochemical relapse, 4% developed bone metastasis and 3% of patients died from PC while 9% died of other causes. Pathological review of the TMAs confirmed the presence of tumor and benign tissue cores for > 94% of patients. Immunohistochemistry and FISH analyses, performed on a small set of specimens, showed high quality results and no biorepository-specific bias. CONCLUSIONS The CPCBN RP cohort is representative of real world PC disease observed in the Canadian population. The frequency of biochemical relapse and bone metastasis as events allows for a precise assessment of the prognostic value of biomarkers. This resource is available, in a step-wise manner, for researchers who intend to validate prognostic biomarkers in PC. Combining multiple biomarkers with clinical and pathologic parameters that are predictive of outcome will aid in clinical decision-making for patients treated for PC.
Collapse
Affiliation(s)
- Véronique Ouellet
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada
| | - Armen Aprikian
- Research Institute of McGill University Health Center and Department of Surgery (Urology), McGill University, Montréal, Québec, Canada
| | - Alain Bergeron
- CHU de Québec-Université Laval and Department of Surgery, Université Laval, Québec City, Québec, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University Health Centre, Montréal, Québec, Canada
| | - Robert G Bristow
- Department of Medical Biophysics and Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.,University Health Network, Toronto, ON, Canada
| | - Simone Chevalier
- Research Institute of McGill University Health Center and Department of Surgery (Urology), McGill University, Montréal, Québec, Canada
| | - Darrel Drachenberg
- University of Manitoba and Manitoba Prostate Centre, Winnipeg, MB, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Neil E Fleshner
- University Health Network, Toronto, ON, Canada.,Division of Urology, Department of Surgery of University Health Network, University of Toronto, Toronto, ON, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, BC, Canada.,Department of Urologic Sciences, Vancouver, BC, Canada
| | - Pierre Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada.,Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | | | - Louis Lacombe
- CHU de Québec-Université Laval and Department of Surgery, Université Laval, Québec City, Québec, Canada
| | - Jean-Baptiste Lattouf
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada.,Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | | | - Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada.,Department of Genetics and Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mathieu Latour
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada.,Department of Pathology and Cellular Biology, Université de Montréal, Montréal, Québec, Canada
| | - Dominique Trudel
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada.,Department of Pathology and Cellular Biology, Université de Montréal, Montréal, Québec, Canada
| | - Anne-Marie Mes-Masson
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada.,Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Fred Saad
- Institut du cancer de Montréal and Centre de recherche du Centre hospitalier de l'Université de Montréal, 900, St-Denis St, room R10-464, Montréal, Québec, H2X 0A9, Canada. .,Department of Surgery, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
23
|
Magi-Galluzzi C, Isharwal S, Falzarano SM, Tsiatis A, Dee A, Maddala T, Knezevic D, Febbo PG, Lawrence J, Klein EA. The 17-Gene Genomic Prostate Score Assay Predicts Outcome After Radical Prostatectomy Independent of PTEN Status. Urology 2018; 121:132-138. [PMID: 30142405 DOI: 10.1016/j.urology.2018.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/17/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To compare the ability of loss of phosphatase and tensin homolog (PTEN) and Genomic prostate score assay (GPS) in predicting the biochemical-recurrence (BCR) and clinical-recurrence (CR) after radical prostatectomy (RP) for clinically localized prostate cancer (PCa). METHODS Three hundred seventy seven patients with and without CR were retrospectively selected by stratified cohort sampling design from RP database. PTEN status (by immunohistochemistry [IHC] and fluorescence in situ hybridization [FISH]) and GPS results were determined for RP specimens. BCR was defined as Prostate Specific Antigen (PSA) ≥ 0.2 ng/mL or initiation of salvage therapy for a rising PSA. CR was defined as local recurrence and/or distant metastases. RESULTS Baseline mean age, PSA, and GPS score for the cohort were 61.1 years, 8 ng/dL, and 32.8. PTEN loss was noted in 38% patients by FISH and 25% by IHC. The concordance between FISH and IHC for PTEN loss was 66% (Kappa coefficient 0.278; P < .001). On univariable analysis, loss of PTEN by FISH or IHC was associated with BCR and CR (P < .05). However, after adjusting for GPS results, PTEN loss was not a significant predictor for CR or BCR (P > .1). The GPS result remained strongly associated with CR and BCR after adjusting for PTEN status (P < .001). PTEN status and GPS results only weakly correlated. GPS was widely distributed regardless of PTEN status indicating the biological heterogeneity of PCa even in PTEN-deficient cases. CONCLUSION GPS is a significant predictor of aggressive PCa, independent of PTEN status. After adjustment for GPS results, PTEN was not independently associated with recurrence for PCa.
Collapse
Affiliation(s)
- Cristina Magi-Galluzzi
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH; Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Sudhir Isharwal
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Sara M Falzarano
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | - Anne Dee
- Genomic Health, Inc., Redwood City, CA
| | | | | | | | | | - Eric A Klein
- Department of Urology, Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
24
|
Comparison of cell cycle progression score with two immunohistochemical markers (PTEN and Ki-67) for predicting outcome in prostate cancer after radical prostatectomy. World J Urol 2018; 36:1495-1500. [PMID: 29679140 DOI: 10.1007/s00345-018-2290-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 04/05/2018] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Previous studies of the cell cycle progression (CCP) score in surgical specimens of prostate cancer (PCa) in patients treated by radical prostatectomy (RP) demonstrated significant association with time to biochemical recurrence (BCR). In this study, we compared the ability of the CCP score and the expression of PTEN or Ki-67 to predict BCR in a cohort of patients treated by RP. Finally, we constructed the best predictive model for BCR, incorporating biomarkers and relevant clinical variables. MATERIALS AND METHODS The study population consisted of 652 PCa patients enrolled in a retrospective cohort and who had RP surgery in French urological centers from 2000 to 2007. RESULTS Among the 652 patients with CCP scores and complete clinical data, BCR events occurred in 41%, and the median time from surgery to the last follow-up among BCR-free patients was 72 months. In univariate Cox analysis, the continuous CCP score and positive Ki-67 predicted recurrence with a HR of 1.44 (95% CI 1.17-1.75; p = 5.3 × 10-4) and 1.89 (95% CI 1.38-2.57; p = 1.6 × 10-4), respectively. In contrast, PTEN expression was not associated with BCR risk. Of the three biomarkers, only the CCP score remained significantly associated in a multivariable Cox model (p = 0.026). The best model incorporated CAPRA-S and CCP scores as predictors, with HRs of 1.32 and 1.24, respectively. CONCLUSION The CCP score was superior to the two IHC markers (PTEN and Ki-67) for predicting outcome in PCa after RP.
Collapse
|
25
|
Decreased TSPAN1 promotes prostate cancer progression and is a marker for early biochemical recurrence after radical prostatectomy. Oncotarget 2018; 7:63294-63305. [PMID: 27556508 PMCID: PMC5325364 DOI: 10.18632/oncotarget.11448] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/10/2016] [Indexed: 01/22/2023] Open
Abstract
Patients with prostate cancer (PCa) have a variable prognosis. It is challenging to recognize the progressive disease. In this study, we focused on TSPAN1, a new member of the tetraspanin family. Its expression was decreased in progressive PCa and was an independent prognosis factor of biochemical recurrence after radical prostatectomy. In vitro, knockdown and overexpression of TSPAN1 in PCa cell lines showed that TSPAN1 could inhibit cell proliferation and migration. TSPAN1 was positive related to PTEN in both clinical specimen and mouse models. The combination of these two markers could increase their prognosis value especially in low risk patients. In vitro TSPAN1 knockdown resulted in increased Akt phosphorylation and caused evident cell cycle transition from G1 to S phase. Our data suggests that TSPAN1 is a valuable marker to recognize more progressive PCa.
Collapse
|
26
|
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: 1.3] [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
|
27
|
Ong CW, Maxwell P, Alvi MA, McQuaid S, Waugh D, Mills I, Salto‐Tellez M. A gene signature associated with PTEN activation defines good prognosis intermediate risk prostate cancer cases. J Pathol Clin Res 2018; 4:103-113. [PMID: 29665325 PMCID: PMC5903700 DOI: 10.1002/cjp2.94] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/05/2017] [Accepted: 01/13/2018] [Indexed: 12/17/2022]
Abstract
Accurate identification of intermediate risk (Gleason 3 + 4 = 7) prostate cancer patients with low risk of disease progression is an unmet challenge in treatment decision making. Here we describe a gene signature that could guide clinicians in the selection of patients with intermediate stage clinically localized prostate cancer for active surveillance. We examined six major drivers of aggressive disease - PTEN, MYC, RB1, TP53, AURKA, AR - by immunohistochemistry in a focused (N = 69) cohort predominantly consisting of intermediate risk prostate cancer. Fuzzy clustering and unsupervised hierarchical clustering were utilized to determine the correlation of gene expression and methylation values with immunohistochemical expression. From the immunohistochemistry observation, we found that intermediate risk prostate cancer cases could be classified as 'complex' (differential expression of more than one driver) or 'simple' (differential expression of only one). Focussing on the 'simple' cases, expression and methylation profiling generated signatures which correlated tightly only with differential PTEN expression and not with any of the other drivers assessed by immunohistochemistry. From this, we derived a geneset of 35 genes linked to high PTEN expression. Subsequently we determined its prognostic significance in intermediate-risk cases extracted from three publicly available clinical datasets (Total N = 215). Hence, this study shows that, by using immunohistochemistry as an upfront stratifier of intermediate risk prostate cancers, it is possible to identify through differential gene expression profiling a geneset with prognostic power across multiple cohorts. This strategy has not been used previously and the signature has the potential to impact on treatment decisions in patients for whom decision making is currently empirical at best.
Collapse
Affiliation(s)
- Chee W Ong
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - Pamela Maxwell
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - Muhammad A Alvi
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - Stephen McQuaid
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - David Waugh
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - Ian Mills
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| | - Manuel Salto‐Tellez
- Movember FASTMAN Centre of Excellence, Centre for Cancer Research and Cell Biology, Queen's University BelfastNorthern Ireland, UK
| |
Collapse
|
28
|
Jamaspishvili T, Berman DM, Ross AE, Scher HI, De Marzo AM, Squire JA, Lotan TL. Clinical implications of PTEN loss in prostate cancer. Nat Rev Urol 2018; 15:222-234. [PMID: 29460925 DOI: 10.1038/nrurol.2018.9] [Citation(s) in RCA: 375] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Genomic aberrations of the PTEN tumour suppressor gene are among the most common in prostate cancer. Inactivation of PTEN by deletion or mutation is identified in ∼20% of primary prostate tumour samples at radical prostatectomy and in as many as 50% of castration-resistant tumours. Loss of phosphatase and tensin homologue (PTEN) function leads to activation of the PI3K-AKT (phosphoinositide 3-kinase-RAC-alpha serine/threonine-protein kinase) pathway and is strongly associated with adverse oncological outcomes, making PTEN a potentially useful genomic marker to distinguish indolent from aggressive disease in patients with clinically localized tumours. At the other end of the disease spectrum, therapeutic compounds targeting nodes in the PI3K-AKT-mTOR (mechanistic target of rapamycin) signalling pathway are being tested in clinical trials for patients with metastatic castration-resistant prostate cancer. Knowledge of PTEN status might be helpful to identify patients who are more likely to benefit from these therapies. To enable the use of PTEN status as a prognostic and predictive biomarker, analytically validated assays have been developed for reliable and reproducible detection of PTEN loss in tumour tissue and in blood liquid biopsies. The use of clinical-grade assays in tumour tissue has shown a robust correlation between loss of PTEN and its protein as well as a strong association between PTEN loss and adverse pathological features and oncological outcomes. In advanced disease, assessing PTEN status in liquid biopsies shows promise in predicting response to targeted therapy. Finally, studies have shown that PTEN might have additional functions that are independent of the PI3K-AKT pathway, including those affecting tumour growth through modulation of the immune response and tumour microenvironment.
Collapse
Affiliation(s)
- Tamara Jamaspishvili
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, Ontario, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - David M Berman
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, Ontario, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ashley E Ross
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy A Squire
- Department of Pathology and Legal Medicine, University of Sao Paulo, Campus Universitario Monte Alegre, Ribeirão Preto, Brazil
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
29
|
Vidotto T, Tiezzi DG, Squire JA. Distinct subtypes of genomic PTEN deletion size influence the landscape of aneuploidy and outcome in prostate cancer. Mol Cytogenet 2018; 11:1. [PMID: 29308088 PMCID: PMC5753467 DOI: 10.1186/s13039-017-0348-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/01/2017] [Indexed: 12/16/2022] Open
Abstract
Background Inactivation of the PTEN tumor suppressor gene by deletion occurs in 20-30% of prostate cancer tumors and loss strongly correlates with a worse outcome. PTEN loss of function not only leads to activation of the PI3K/AKT pathway, but is also thought to affect genome stability and increase levels of tumor aneuploidy. We performed an in silico integrative genomic and transcriptomic analysis of 491 TCGA prostate cancer tumors. These data were used to map the genomic sizes of PTEN gene deletions and to characterize levels of instability and patterns of aneuploidy acquisition. Results PTEN homozygous deletions had a significant increase in aneuploidy compared to PTEN tumors without an apparent deletion, and hemizygous deletions showed an intermediate aneuploidy profile. A supervised clustering of somatic copy number alterations (SCNA) demonstrated that the size of PTEN deletions was not random, but comprised five distinct subtypes: (1) "Small Interstitial" (70 bp-789Kb); (2) "Large Interstitial" (1-7 MB); (3) "Large Proximal" (3-65 MB); (4) "Large Terminal" (8-64 MB), and (5) "Extensive" (71-132 MB). Many of the deleted fragments in each subtype were flanked by low copy repetitive (LCR) sequences. SCNAs such as gain at 3q21.1-3q29 and deletions at 8p, RB1, TP53 and TMPRSS2-ERG were variably present in all subtypes. Other SCNAs appeared to be recurrent in some deletion subtypes, but absent from others. To determine how the aneuploidy influenced global levels of gene expression, we performed a comparative transcriptome analysis. One deletion subtype (Large Interstitial) was characterized by gene expression changes associated with angiogenesis and cell adhesion, structure, and metabolism. Logistic regression demonstrated that this deletion subtype was associated with a high Gleason score (HR = 2.386; 95% C.I. 1.245-4.572), extraprostatic extension (HR = 2.423, 95% C.I. 1.157-5.075), and metastasis (HR = 7.135; 95% C.I. 1.540-33.044). Univariate and multivariate Cox Regression showed that presence of this deletion subtype was also strongly predictive of disease recurrence. Conclusions Our findings indicate that genomic deletions of PTEN fall into five different size distributions, with breakpoints that often occur close LCR regions, and that each subtype is associated with a characteristic aneuploidy signature. The Large Interstitial deletion had a distinct gene expression signature that was related to cancer progression and was also predictive of a worse prognosis.
Collapse
Affiliation(s)
- Thiago Vidotto
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniel Guimarães Tiezzi
- Deparment of Gynecology and Obstetrics, Clinical Hospital of Ribeirão Preto, Ribeirão Preto, Brazil
| | - Jeremy A Squire
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, 3900 Bandeirantes Avenue, Monte Alegre, Ribeirão Preto, São Paulo 14040-900 Brazil.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| |
Collapse
|
30
|
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: 2.1] [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
|
31
|
Glaser ZA, Gordetsky JB, Porter KK, Varambally S, Rais-Bahrami S. Prostate Cancer Imaging and Biomarkers Guiding Safe Selection of Active Surveillance. Front Oncol 2017; 7:256. [PMID: 29164056 PMCID: PMC5670116 DOI: 10.3389/fonc.2017.00256] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/12/2017] [Indexed: 01/04/2023] Open
Abstract
Background Active surveillance (AS) is a widely adopted strategy to monitor men with low-risk, localized prostate cancer (PCa). Current AS inclusion criteria may misclassify as many as one in four patients. The advent of multiparametric magnetic resonance imaging (mpMRI) and novel PCa biomarkers may offer improved risk stratification. We performed a review of recently published literature to characterize emerging evidence in support of these novel modalities. Methods An English literature search was conducted on PubMed for available original investigations on localized PCa, AS, imaging, and biomarkers published within the past 3 years. Our Boolean criteria included the following terms: PCa, AS, imaging, biomarker, genetic, genomic, prospective, retrospective, and comparative. The bibliographies and diagnostic modalities of the identified studies were used to expand our search. Results Our review identified 222 original studies. Our expanded search yielded 244 studies. Among these, 70 met our inclusion criteria. Evidence suggests mpMRI offers improved detection of clinically significant PCa, and MRI-fusion technology enhances the sensitivity of surveillance biopsies. Multiple studies demonstrate the promise of commercially available screening assays for prediction of AS failure, and several novel biomarkers show promise in this setting. Conclusion In the era of AS for men with low-risk PCa, improved strategies for proper stratification are needed. mpMRI has dramatically enhanced the detection of clinically significant PCa. The advent of novel biomarkers for prediction of aggressive disease and AS failure has shown some initial promise, but further validation is warranted.
Collapse
Affiliation(s)
- Zachary A Glaser
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jennifer B Gordetsky
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kristin K Porter
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
32
|
Geybels MS, Fang M, Wright JL, Qu X, Bibikova M, Klotzle B, Fan JB, Feng Z, Ostrander EA, Nelson PS, Stanford JL. PTEN loss is associated with prostate cancer recurrence and alterations in tumor DNA methylation profiles. Oncotarget 2017; 8:84338-84348. [PMID: 29137428 PMCID: PMC5663600 DOI: 10.18632/oncotarget.20940] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 07/08/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) with loss of the tumor suppressor gene PTEN has an unfavorable prognosis. DNA methylation profiles associated with PTEN loss may provide further insights into the mechanisms underlying these more aggressive, clinically relevant tumors. METHODS The cohort included patients with clinically localized PCa. Samples taken from the primary tumor were used to determine PTEN genomic deletions using FISH, and to analyze epigenome-wide DNA methylation profiles. Patients were followed for PCa recurrence on average for 8 years after diagnosis. RESULTS The study included 471 patients with data on PTEN loss, and the frequency of hemi- and homozygous PTEN loss was 10.0% and 4.5%, respectively. Loss of PTEN was associated with a significantly higher risk of recurrence (any vs. no PTEN loss; HR = 1.74; 95% CI: 1.03-2.93). Hazard ratios for hemi- and homozygous loss were 1.39 (95% CI: 0.73-2.64) and 2.84 (95% CI: 1.30-6.19), respectively. Epigenome-wide methylation profiling identified 4,208 differentially methylated CpGs (FDR Q-value < 0.01) in tumors with any versus no PTEN loss. There were no genome-wide significant differentially methylated CpGs in homo- versus hemizygous deleted tumors. Tumor methylation data were used to build a methylation signature of PTEN loss in our cohort, which was confirmed in TCGA, and included CpGs in ATP11A, GDNF, JAK1, JAM3, and VAPA. CONCLUSION Loss of PTEN was positively associated with PCa recurrence. Prostate tumors with PTEN loss harbor a distinct methylation signature, and these aberrantly methylated CpG sites may mediate tumor progression when PTEN is deleted.
Collapse
Affiliation(s)
- Milan S. Geybels
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
| | - Min Fang
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jonathan L. Wright
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
- Department of Urology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Xiaoyu Qu
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Cytogenetics, Seattle Cancer Care Alliance, Seattle, Washington, USA
| | - Marina Bibikova
- Department of Oncology, Illumina, Inc., San Diego, California, USA
| | - Brandy Klotzle
- Department of Oncology, Illumina, Inc., San Diego, California, USA
| | - Jian-Bing Fan
- Department of Oncology, Illumina, Inc., San Diego, California, USA
- Current address: AnchorDx Corp., Guangzhou 510300, People's Republic of China
| | - Ziding Feng
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Elaine A. Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Peter S. Nelson
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchison Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA
| |
Collapse
|
33
|
Lotan TL, Heumann A, Rico SD, Hicks J, Lecksell K, Koop C, Sauter G, Schlomm T, Simon R. PTEN loss detection in prostate cancer: comparison of PTEN immunohistochemistry and PTEN FISH in a large retrospective prostatectomy cohort. Oncotarget 2017; 8:65566-65576. [PMID: 29029453 PMCID: PMC5630353 DOI: 10.18632/oncotarget.19217] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/27/2017] [Indexed: 11/25/2022] Open
Abstract
PTEN deletion is an established prognostic biomarker in prostate cancer. We compared PTEN immunohistochemistry (IHC) and PTEN fluorescence in situ hybridization (FISH) in the largest existing radical prostatectomy cohort with clinical follow-up data. There was high concordance between IHC and FISH: 93% (3098/3330) of tumors with intact PTEN IHC showed absence of PTEN gene deletion and 66% (720/1087) of cases with PTEN protein loss by IHC showed PTEN gene deletion by FISH. 84% (447/533) of cases with PTEN homozygous gene deletion had PTEN protein loss by IHC. PTEN loss by IHC was associated with reduced PSA recurrence-free survival (RFS) in multivariable models (HR=1.3; 95% CI: 1.16-1.47). Among cases with either PTEN deletion or absence of PTEN deletion by FISH, PTEN loss by IHC was strongly associated with reduced RFS on univariable analysis (p=0.0005 and p<0.0001 respectively). Among cases with intact PTEN by IHC, homozygous (p=0.04) but not heterozygous (p=0.10) PTEN gene deletion was weakly associated with reduced RFS. Among cases with PTEN loss by IHC, both homozygous (p=0.0044) and heterozygous (p=0.0017) PTEN gene deletion were associated with reduced RFS. These data support the utility of PTEN IHC and PTEN FISH as complementary screening tools for PTEN loss in prostate cancer.
Collapse
Affiliation(s)
- Tamara L. Lotan
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Asmus Heumann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jessica Hicks
- Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Christina Koop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Klinik Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Prostate cancer, PI3K, PTEN and prognosis. Clin Sci (Lond) 2017; 131:197-210. [PMID: 28057891 DOI: 10.1042/cs20160026] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [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
|
35
|
Leapman MS, Carroll PR. Risk Stratification of Newly Diagnosed Prostate Cancer with Genomic Platforms. UROLOGY PRACTICE 2017; 4:322-328. [PMID: 37592678 DOI: 10.1016/j.urpr.2016.06.008] [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: 10/20/2022]
Abstract
INTRODUCTION Interest in novel risk stratification tools for men with newly diagnosed prostate cancer has flourished, aiming to offer increasingly accurate predictions of future disease behavior to ultimately better guide clinical management. We highlight the use of genomic platforms attempting to refine clinical decisions at the point of initial diagnosis. METHODS In the context of a benchmark standard of clinical risk stratification tools we reviewed the role of genomic tests, including individual gene expression assays, as well as a growing number of tissue based expression tests assessing multiple gene panels, to improve predictions at initial diagnosis. RESULTS The role of single gene status including TMPRSS2:ERG fusion and PTEN expression has been investigated among men with newly diagnosed prostate cancer. Gene expression profiles incorporating panels of genes associated with prostate cancer outcome have received external validation and have commercial application in assays that incorporate baseline clinical risk to offer predictions of immediate pathological and downstream disease end points. Comparisons of gene signatures have offered insights into relative predictive performance in archival tissue. However, to date no studies appear to directly support a single genomic assay offering superior clinical usefulness for decision making at the time of diagnosis. CONCLUSIONS Risk stratification tools incorporating genomic analysis of prostate cancer have been developed which seek to improve the accuracy of initial predictions. Further study is warranted to define the additive clinical benefit associated with their use if implemented broadly.
Collapse
Affiliation(s)
- Michael S Leapman
- Department of Urology, UCSF - Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Peter R Carroll
- Department of Urology, UCSF - Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
36
|
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: 14] [Impact Index Per Article: 2.0] [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
|
37
|
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: 24] [Impact Index Per Article: 3.4] [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
|
38
|
Peterson LE, Kovyrshina T. Progression inference for somatic mutations in cancer. Heliyon 2017; 3:e00277. [PMID: 28492066 PMCID: PMC5415494 DOI: 10.1016/j.heliyon.2017.e00277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/08/2017] [Accepted: 03/23/2017] [Indexed: 01/05/2023] Open
Abstract
Computational methods were employed to determine progression inference of genomic alterations in commonly occurring cancers. Using cross-sectional TCGA data, we computed evolutionary trajectories involving selectivity relationships among pairs of gene-specific genomic alterations such as somatic mutations, deletions, amplifications, downregulation, and upregulation among the top 20 driver genes associated with each cancer. Results indicate that the majority of hierarchies involved TP53, PIK3CA, ERBB2, APC, KRAS, EGFR, IDH1, VHL, etc. Research into the order and accumulation of genomic alterations among cancer driver genes will ever-increase as the costs of nextgen sequencing subside, and personalized/precision medicine incorporates whole-genome scans into the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Healthcare Policy and Research, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
- Dept. of Biostatistics, School of Public Health, University of Texas – Health Science Center, Houston, TX 77030, USA
- Dept. of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dept. of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Tatiana Kovyrshina
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Mathematics and Statistics, University of Houston – Downtown, Houston, TX 77002, USA
| |
Collapse
|
39
|
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: 0.6] [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
|
40
|
MUC1 Expression by Immunohistochemistry Is Associated with Adverse Pathologic Features in Prostate Cancer: A Multi-Institutional Study. PLoS One 2016; 11:e0165236. [PMID: 27846218 PMCID: PMC5112958 DOI: 10.1371/journal.pone.0165236] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022] Open
Abstract
Background The uncertainties inherent in clinical measures of prostate cancer (CaP) aggressiveness endorse the investigation of clinically validated tissue biomarkers. MUC1 expression has been previously reported to independently predict aggressive localized prostate cancer. We used a large cohort to validate whether MUC1 protein levels measured by immunohistochemistry (IHC) predict aggressive cancer, recurrence and survival outcomes after radical prostatectomy independent of clinical and pathological parameters. Material and Methods MUC1 IHC was performed on a multi-institutional tissue microarray (TMA) resource including 1,326 men with a median follow-up of 5 years. Associations with clinical and pathological parameters were tested by the Chi-square test and the Wilcoxon rank sum test. Relationships with outcome were assessed with univariable and multivariable Cox proportional hazard models and the Log-rank test. Results The presence of MUC1 expression was significantly associated with extracapsular extension and higher Gleason score, but not with seminal vesicle invasion, age, positive surgical margins or pre-operative serum PSA levels. In univariable analyses, positive MUC1 staining was significantly associated with a worse recurrence free survival (RFS) (HR: 1.24, CI 1.03–1.49, P = 0.02), although not with disease specific survival (DSS, P>0.5). On multivariable analyses, the presence of positive surgical margins, extracapsular extension, seminal vesicle invasion, as well as higher pre-operative PSA and increasing Gleason score were independently associated with RFS, while MUC1 expression was not. Positive MUC1 expression was not independently associated with disease specific survival (DSS), but was weakly associated with overall survival (OS). Conclusion In our large, rigorously designed validation cohort, MUC1 protein expression was associated with adverse pathological features, although it was not an independent predictor of outcome after radical prostatectomy.
Collapse
|
41
|
|
42
|
Brooks JD, Wei W, Pollack JR, West RB, Shin JH, Sunwoo JB, Hawley SJ, Auman H, Newcomb LF, Simko J, Hurtado-Coll A, Troyer DA, Carroll PR, Gleave ME, Lin DW, Nelson PS, Thompson IM, True LD, McKenney JK, Feng Z, Fazli L. Loss of Expression of AZGP1 Is Associated With Worse Clinical Outcomes in a Multi-Institutional Radical Prostatectomy Cohort. Prostate 2016; 76:1409-19. [PMID: 27325561 PMCID: PMC5557496 DOI: 10.1002/pros.23225] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Given the uncertainties inherent in clinical measures of prostate cancer aggressiveness, clinically validated tissue biomarkers are needed. We tested whether Alpha-2-Glycoprotein 1, Zinc-Binding (AZGP1) protein levels, measured by immunohistochemistry, and RNA expression, by RNA in situ hybridization (RISH), predict recurrence after radical prostatectomy independent of clinical and pathological parameters. METHODS AZGP1 IHC and RISH were performed on a large multi-institutional tissue microarray resource including 1,275 men with 5 year median follow-up. The relationship between IHC and RISH expression levels was assessed using the Kappa analysis. Associations with clinical and pathological parameters were tested by the Chi-square test and the Wilcoxon rank sum test. Relationships with outcome were assessed with univariable and multivariable Cox proportional hazards models and the Log-rank test. RESULTS Absent or weak expression of AZGP1 protein was associated with worse recurrence free survival (RFS), disease specific survival, and overall survival after radical prostatectomy in univariable analysis. AZGP1 protein expression, along with pre-operative serum PSA levels, surgical margin status, seminal vesicle invasion, extracapsular extension, and Gleason score predicted RFS on multivariable analysis. Similarly, absent or low AZGP1 RNA expression by RISH predicted worse RFS after prostatectomy in univariable and multivariable analysis. CONCLUSIONS In our large, rigorously designed validation cohort, loss of AZGP1 expression predicts RFS after radical prostatectomy independent of clinical and pathological variables. Prostate 76:1409-1419, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- James D Brooks
- Department of Urology, Stanford University, Stanford, California.
| | - Wei Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Robert B West
- Department of Pathology, Stanford University, Stanford, California
| | - Jun Ho Shin
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California
| | - John B Sunwoo
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Stanford, California
| | - Sarah J Hawley
- Canary Foundation, Canary Center at Stanford, Palo Alto, California
| | - Heidi Auman
- Canary Foundation, Canary Center at Stanford, Palo Alto, California
| | - Lisa F Newcomb
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Jeff Simko
- Department of Pathology, University of California San Francisco, San Francisco, California
| | - Antonio Hurtado-Coll
- Department of Urologic Sciences and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Dean A Troyer
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Eastern Virginia Medical School, Pathology and Microbiology and Molecular Biology, Norfolk, Virginia
| | - Peter R Carroll
- Department of Urology, University of California San Francisco, San Francisco, California
| | - Martin E Gleave
- Department of Urologic Sciences and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Daniel W Lin
- Department of Urology, University of Washington Medical Center, Seattle, Washington
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Lawrence D True
- Department of Pathology, University of Washington Medical Center, Seattle, Washington
| | | | - Ziding Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ladan Fazli
- Department of Urologic Sciences and Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Yuruk E, Pastuszak AW, Suggs JM, Colakerol A, Serefoglu EC. The association between seminal vesicle size and duration of abstinence from ejaculation. Andrologia 2016; 49. [PMID: 27660049 DOI: 10.1111/and.12707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2016] [Indexed: 01/14/2023] Open
Abstract
There are few data describing the relationship between seminal vesicle (SV) size and duration of abstinence between ejaculations. This study evaluates the association between SV size and duration of abstinence from ejaculation using pelvic magnetic resonance imaging (MRI). Sexually active men 18-68 years old who underwent pelvic MRI for various medical indications were included. The date of last ejaculation was recorded, and the cross-sectional areas of the right and left seminal vesicles were calculated separately using mediolateral and anteroposterior measurements on T2-weighted MRI images. The association between SV area and duration of abstinence between ejaculations was determined via linear regression analysis. The study cohort consisted of 104 men with a mean age of 46.45 ± 11.4 (range 18-68) years old. Mean right and left SV cross-sectional areas were 744.1 ± 351.1 (range: 149.9-1794.7) mm2 and 727.6 ± 359.2 (range 171.4-2248.4) mm2 respectively. The mean duration of abstinence between ejaculations in the cohort was 3.6 ± 2.6 (range 1-15) days. Although no correlation between age and SV area was observed (r = .007, p = .947), linear regression analysis demonstrated a positive correlation between SV area and the duration of abstinence from ejaculation (r = .372, p = .0001). SV cross-sectional area increases with duration of abstinence from ejaculation and can be assessed using MRI. The use of SV size estimation may be applicable in diagnosis, risk stratification and treatment of urological diseases.
Collapse
Affiliation(s)
- E Yuruk
- Bagcilar Research and Training Hospital, Istanbul, Turkey
| | - A W Pastuszak
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, USA.,Division of Male Reproductive Medicine, Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - J M Suggs
- Baylor College of Medicine, Houston, TX, USA
| | - A Colakerol
- Bagcilar Research and Training Hospital, Istanbul, Turkey
| | - E C Serefoglu
- Bagcilar Research and Training Hospital, Istanbul, Turkey
| |
Collapse
|
44
|
Analytic validation of a clinical-grade PTEN immunohistochemistry assay in prostate cancer by comparison with PTEN FISH. Mod Pathol 2016; 29:904-14. [PMID: 27174589 PMCID: PMC4967011 DOI: 10.1038/modpathol.2016.88] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/05/2016] [Accepted: 04/14/2016] [Indexed: 01/09/2023]
Abstract
PTEN loss is a promising prognostic and predictive biomarker in prostate cancer. Because it occurs most commonly via PTEN gene deletion, we developed a clinical-grade, automated, and inexpensive immunohistochemical assay to detect PTEN loss. We studied the sensitivity and specificity of PTEN immunohistochemistry relative to four-color fluorescence in situ hybridization (FISH) for detection of PTEN gene deletion in a multi-institutional cohort of 731 primary prostate tumors. Intact PTEN immunostaining was 91% specific for the absence of PTEN gene deletion (549/602 tumors with two copies of the PTEN gene by FISH showed intact expression of PTEN by immunohistochemistry) and 97% sensitive for the presence of homozygous PTEN gene deletion (absent PTEN protein expression by immunohistochemistry in 65/67 tumors with homozygous deletion). PTEN immunohistochemistry was 65% sensitive for the presence of hemizygous PTEN gene deletion, with protein loss in 40/62 hemizygous tumors. We reviewed the 53 cases where immunohistochemistry showed PTEN protein loss and FISH showed two intact copies of the PTEN gene. On re-review, there was ambiguous immunohistochemistry loss in 6% (3/53) and failure to analyze the same tumor area by both methods in 34% (18/53). Of the remaining discordant cases, 41% (13/32) revealed hemizygous (n=8) or homozygous (n=5) PTEN gene deletion that was focal in most cases (11/13). The remaining 19 cases had two copies of the PTEN gene detected by FISH, representing truly discordant cases. Our automated PTEN immunohistochemistry assay is a sensitive method for detection of homozygous PTEN gene deletions. Immunohistochemistry screening is particularly useful to identify cases with heterogeneous PTEN gene deletion in a subset of tumor glands. Mutations, small insertions, or deletions and/or epigenetic or microRNA-mediated mechanisms may lead to PTEN protein loss in tumors with normal or hemizygous PTEN gene copy number.
Collapse
|
45
|
Lotan TL, Wei W, Morais CL, Hawley ST, Fazli L, Hurtado-Coll A, Troyer D, McKenney JK, Simko J, Carroll PR, Gleave M, Lance R, Lin DW, Nelson PS, Thompson IM, True LD, Feng Z, Brooks JD. PTEN Loss as Determined by Clinical-grade Immunohistochemistry Assay Is Associated with Worse Recurrence-free Survival in Prostate Cancer. Eur Urol Focus 2016; 2:180-188. [PMID: 27617307 DOI: 10.1016/j.euf.2015.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND PTEN is the most commonly deleted tumor suppressor gene in primary prostate cancer (PCa) and its loss is associated with poor clinical outcomes and ERG gene rearrangement. OBJECTIVE We tested whether PTEN loss is associated with shorter recurrence-free survival (RFS) in surgically treated PCa patients with known ERG status. DESIGN SETTING AND PARTICIPANTS A genetically validated, automated PTEN immunohistochemistry (IHC) protocol was used for 1275 primary prostate tumors from the Canary Foundation retrospective PCa tissue microarray cohort to assess homogeneous (in all tumor tissue sampled) or heterogeneous (in a subset of tumor tissue sampled) PTEN loss. ERG status as determined by a genetically validated IHC assay was available for a subset of 938 tumors. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Associations between PTEN and ERG status were assessed using Fisher's exact test. Kaplan-Meier and multivariate weighted Cox proportional models for RFS were constructed. RESULTS AND LIMITATIONS When compared to intact PTEN, homogeneous (hazard ratio [HR] 1.66, p = 0.001) but not heterogeneous (HR 1.24, p = 0.14) PTEN loss was significantly associated with shorter RFS in multivariate models. Among ERG-positive tumors, homogeneous (HR 3.07, p < 0.0001) but not heterogeneous (HR 1.46, p = 0.10) PTEN loss was significantly associated with shorter RFS. Among ERG-negative tumors, PTEN did not reach significance for inclusion in the final multivariate models. The interaction term for PTEN and ERG status with respect to RFS did not reach statistical significance (p = 0.11) for the current sample size. CONCLUSIONS These data suggest that PTEN is a useful prognostic biomarker and that there is no statistically significant interaction between PTEN and ERG status for RFS. PATIENT SUMMARY We found that loss of the PTEN tumor suppressor gene in prostate tumors as assessed by tissue staining is correlated with shorter time to prostate cancer recurrence after radical prostatectomy.
Collapse
Affiliation(s)
- Tamara L Lotan
- Pathology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA; Oncology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Wei Wei
- MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos L Morais
- Pathology Department, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Dean Troyer
- Pathology Department, Eastern Virginia Medical School, Norfolk, VA, USA; Urology Department, Eastern Virginia Medical School, Norfolk, VA, USA
| | | | - Jeffrey Simko
- Pathology Department, University of California-San Francisco, San Francisco, CA, USA; Urology Department, University of California-San Francisco, San Francisco, CA, USA
| | - Peter R Carroll
- Urology Department, University of California-San Francisco, San Francisco, CA, USA
| | | | - Raymond Lance
- Urology Department, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Daniel W Lin
- Urology Department, University of Washington, Seattle, WA, USA
| | - Peter S Nelson
- Oncology Department, University of Washington, Seattle, WA, USA; Pathology Department, University of Washington, Seattle, WA, USA
| | - Ian M Thompson
- Urology Department, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lawrence D True
- Pathology Department, University of California-San Francisco, San Francisco, CA, USA
| | | | - James D Brooks
- Urology Department, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
46
|
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: 3.4] [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]
|
47
|
Garzotto M, Kopp RP. Gene-expression profiling of localized prostate cancer: still miles to go before we sleep. Future Oncol 2016; 12:273-6. [DOI: 10.2217/fon.15.316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mark Garzotto
- Veterans Administration Portland Health Care System, Oregon Health & Science University, Portland, OR, USA
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
- Radiation Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ryan P Kopp
- Veterans Administration Portland Health Care System, Oregon Health & Science University, Portland, OR, USA
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
48
|
Biomarkers for prostate cancer: present challenges and future opportunities. Future Sci OA 2015; 2:FSO72. [PMID: 28031932 PMCID: PMC5137959 DOI: 10.4155/fso.15.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 01/30/2023] Open
Abstract
Prostate cancer (PCa) has variable biological potential with multiple treatment options. A more personalized approach, therefore, is needed to better define men at higher risk of developing PCa, discriminate indolent from aggressive disease and improve risk stratification after treatment by predicting the likelihood of progression. This may improve clinical decision-making regarding management, improve selection for active surveillance protocols and minimize morbidity from treatment. Discovery of new biomarkers associated with prostate carcinogenesis present an opportunity to provide patients with novel genetic signatures to better understand their risk of developing PCa and help forecast their clinical course. In this review, we examine the current literature evaluating biomarkers in PCa. We also address current limitations and present several ideas for future studies.
Collapse
|
49
|
Schweizer MT, Yu EY. Persistent androgen receptor addiction in castration-resistant prostate cancer. J Hematol Oncol 2015; 8:128. [PMID: 26566796 PMCID: PMC4644296 DOI: 10.1186/s13045-015-0225-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/10/2015] [Indexed: 02/07/2023] Open
Abstract
It is now understood that persistent activation of the androgen receptor (AR) signaling pathway often underlies the development of castration-resistant prostate cancer (CRPC). This realization led to renewed interest in targeting the AR and ultimately to the development of the potent next-generation AR-directed agents abiraterone and enzalutamide. While these drugs prolong survival in men with CRPC, they are unfortunately not curative. Perhaps not surprisingly, evidence points to persistent AR signaling as one of the key drivers by which resistances to these agents develops. In this context, activation of the AR signaling program can occur through a number of molecular adaptations, including alterations leading to persistent canonical AR signaling (e.g., AR amplification/overexpression, elucidations/concentration of intratumoral androgens), activation of the AR program via feedback pathways (e.g., AKT/mTOR/Pi3K, HER2/Neu), and activation of the AR program via mutation or substitution (e.g., AR ligand binding domain mutation; AR splice variants; glucocorticoid receptor signaling). This review will provide an overview of the more clinical relevant (i.e., druggable) pathways that have been implicated in the emergence of drug resistance in men with CRPC and highlight some of the ongoing efforts towards developing therapeutics to impair these mechanisms.
Collapse
Affiliation(s)
- Michael T Schweizer
- Division of Oncology, Department of Medicine, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Evan Y Yu
- Division of Oncology, Department of Medicine, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|