51
|
|
52
|
Freeland J, Crowell PD, Giafaglione JM, Boutros PC, Goldstein AS. Aging of the progenitor cells that initiate prostate cancer. Cancer Lett 2021; 515:28-35. [PMID: 34052326 PMCID: PMC8494000 DOI: 10.1016/j.canlet.2021.05.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
Many organs experience a loss of tissue mass and a decline in regenerative capacity during aging. In contrast, the prostate continues to grow in volume. In fact, age is the most important risk factor for prostate cancer. However, the age-related factors that influence the composition, morphology and molecular features of prostate epithelial progenitor cells, the cells-of-origin for prostate cancer, are poorly understood. Here, we review the evidence that prostate luminal progenitor cells are expanded with age. We explore the age-related changes to the microenvironment that may influence prostate epithelial cells and risk of transformation. Finally, we raise a series of questions about models of aging and regulators of prostate aging which need to be addressed. A fundamental understanding of aging in the prostate will yield critical insights into mechanisms that promote the development of age-related prostatic disease.
Collapse
Affiliation(s)
- Jack Freeland
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, USA
| | - Preston D Crowell
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, USA
| | - Jenna M Giafaglione
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, USA
| | - Paul C Boutros
- Departments of Human Genetics & Urology, Jonsson Comprehensive Cancer Center and Institute for Precision Health, University of California, Los Angeles, USA
| | - Andrew S Goldstein
- Departments of Molecular, Cell and Developmental Biology & Urology, Broad Stem Cell Research Center and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, USA.
| |
Collapse
|
53
|
Mangiola S, McCoy P, Modrak M, Souza-Fonseca-Guimaraes F, Blashki D, Stuchbery R, Keam SP, Kerger M, Chow K, Nasa C, Le Page M, Lister N, Monard S, Peters J, Dundee P, Williams SG, Costello AJ, Neeson PJ, Pal B, Huntington ND, Corcoran NM, Papenfuss AT, Hovens CM. Transcriptome sequencing and multi-plex imaging of prostate cancer microenvironment reveals a dominant role for monocytic cells in progression. BMC Cancer 2021; 21:846. [PMID: 34294073 PMCID: PMC8296706 DOI: 10.1186/s12885-021-08529-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/23/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Prostate cancer is caused by genomic aberrations in normal epithelial cells, however clinical translation of findings from analyses of cancer cells alone has been very limited. A deeper understanding of the tumour microenvironment is needed to identify the key drivers of disease progression and reveal novel therapeutic opportunities. RESULTS In this study, the experimental enrichment of selected cell-types, the development of a Bayesian inference model for continuous differential transcript abundance, and multiplex immunohistochemistry permitted us to define the transcriptional landscape of the prostate cancer microenvironment along the disease progression axis. An important role of monocytes and macrophages in prostate cancer progression and disease recurrence was uncovered, supported by both transcriptional landscape findings and by differential tissue composition analyses. These findings were corroborated and validated by spatial analyses at the single-cell level using multiplex immunohistochemistry. CONCLUSIONS This study advances our knowledge concerning the role of monocyte-derived recruitment in primary prostate cancer, and supports their key role in disease progression, patient survival and prostate microenvironment immune modulation.
Collapse
Affiliation(s)
- Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick McCoy
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Martin Modrak
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Fernando Souza-Fonseca-Guimaraes
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Daniel Blashki
- The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Ryan Stuchbery
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Simon P Keam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Michael Kerger
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ken Chow
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Chayanica Nasa
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Melanie Le Page
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Natalie Lister
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Simon Monard
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Justin Peters
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Phil Dundee
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Scott G Williams
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Anthony J Costello
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Bhupinder Pal
- The Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, Australia
| | - Nicholas D Huntington
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Urology, Frankston Hospital, Frankston, Victoria, Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
54
|
Considine B, Adeniran A, Hurwitz ME. Current Understanding and Management of Intraductal Carcinoma of the Prostate. Curr Oncol Rep 2021; 23:110. [PMID: 34272624 DOI: 10.1007/s11912-021-01090-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW This review will discuss current understanding and management approaches of Intraductal carcinoma of the prostate (IDC-P). IDC-P is a histological finding characterized by neoplastic cells that expand but do not invade prostate ducts. RECENT FINDINGS The presence of IDC-P on a prostate biopsy is almost always associated with an invasive disease component and is independently associated with worse clinical outcomes in both early and late disease. These tumors are enriched for mutations in homologous DNA recombination repair (HRR) leading to high genomic instability. Multiparametric MRI with targeted biopsy may aid in diagnosis. Given the poor clinical outcomes associated with this histologic entity, its presence in biopsies should warrant consideration of aggressive management.
Collapse
Affiliation(s)
- Bryden Considine
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA
| | - Adebowale Adeniran
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA
| | - Michael E Hurwitz
- Yale Comprehensive Cancer Center, 333 Cedar St, New Haven, CT, 06510, USA.
| |
Collapse
|
55
|
Zhao J, Sun G, Zhu S, Dai J, Chen J, Zhang M, Ni Y, Zhang H, Shen P, Zhao X, Zhang B, Pan X, Nie L, Yin X, Liang J, Zhang X, Wang Z, Zhu X, Liao B, Liu Z, Armstrong CM, Gao AC, Huang H, Chen N, Zeng H. Circulating tumour DNA reveals genetic traits of patients with intraductal carcinoma of the prostate. BJU Int 2021; 129:345-355. [PMID: 34185954 DOI: 10.1111/bju.15530] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/05/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To investigate the genetic alterations of patients with prostate cancer (PCa) with and without intraductal carcinoma of the prostate (IDC-P). PATIENTS AND METHODS We performed targeted sequencing of plasma cell-free DNA on 161 patients with prostate adenocarcinoma (PAC) with IDC-P and 84 without IDC-P. Genomic alterations were compared between these two groups. The association between genetic alterations and patients' survival outcomes was also explored. RESULTS We identified that 29.8% (48/161) and 21.4% (18/84) of patients with and without IDC-P harboured genomic alterations in DNA repair pathways, respectively (P = 0.210). Pathogenic germline DNA repair alterations were frequently detected in IDC-P carriers compared to IDC-P non-carriers (11.8% [19/161] vs 2.4% [two of 84], P = 0.024). Germline BReast CAncer type 2 susceptibility protein (BRCA2) and somatic cyclin-dependent kinase 12 (CDK12) defects were specifically identified in IDC-P carriers relative to PAC (BRCA2: 8.7% [14/161] vs 0% and CDK12: 6.8% [11/161] vs 1.2% [one of 84]). Patients with IDC-P had a distinct androgen receptor (AR) pathway alteration, characterised by an enrichment of nuclear receptor corepressor 2 (NCOR2) mutations compared with patients with pure PAC (21.1% [34/161] vs 6.0% [five of 84], P = 0.004). Increased AR alterations were detected in patients harbouring tumours with an IDC-P proportion of ≥10% vs those with an IDC-P proportion of <10% (6.4% [five of 78] vs 18.1% [15/83], P = 0.045). For IDC-P carriers, tumour protein p53 (TP53) mutation was associated with shorter castration-resistant-free survival (median 10.9 vs 28.9 months, P = 0.026), and BRCA2 alteration was related to rapid prostate-specific antigen progression for those receiving abiraterone treatment (median 9.1 vs 11.9 months, P = 0.036). CONCLUSION Our findings provide genomic evidence explaining the aggressive phenotype of tumours with IDC-P, highlighting the potential therapeutic strategies for this patient population.
Collapse
Affiliation(s)
- Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Haoran Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaochen Zhao
- Departments of Biochemistry and Molecular Biology and Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Bei Zhang
- Departments of Biochemistry and Molecular Biology and Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Xiuyi Pan
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhipeng Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xudong Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Banghua Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Allen C Gao
- Department of Urology, University of California Davis, Davis, CA, USA
| | | | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
56
|
Lawrence MG, Porter LH, Clouston D, Murphy DG, Frydenberg M, Taylor RA, Risbridger GP. Knowing what's growing: Why ductal and intraductal prostate cancer matter. Sci Transl Med 2021; 12:12/533/eaaz0152. [PMID: 32132214 DOI: 10.1126/scitranslmed.aaz0152] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Prostate cancer is a common malignancy, but only some tumors are lethal. Accurately identifying these tumors will improve clinical practice and instruct research. Aggressive cancers often have distinctive pathologies, including intraductal carcinoma of the prostate (IDC-P) and ductal adenocarcinoma. Here, we review the importance of these pathologies because they are often overlooked, especially in genomics and preclinical testing. Pathology, genomics, and patient-derived models show that IDC-P and ductal adenocarcinoma accompany multiple markers of poor prognosis. Consequently, "knowing what is growing" will help translate preclinical research to pinpoint and treat high-risk prostate cancer in the clinic.
Collapse
Affiliation(s)
- Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC 3000, Australia.,Epworth HealthCare, Melbourne, VIC 3000, Australia
| | - Mark Frydenberg
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.,Australian Urology Associates, Melbourne, VIC 3000, Australia.,Department of Urology, Cabrini Health, Malvern, VIC 3144, Australia
| | - Renea A Taylor
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC 3800, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
57
|
Wilkinson S, Ye H, Lis RT, Sowalsky AG. Reply to Alessia Cimadamore, Liang Cheng, Antonio Lopez-Beltran, Marina Scarpelli, and Rodolfo Montironi's Letter to the Editor re: Scott Wilkinson, Huihui Ye, Fatima Karzai, et al. Nascent Prostate Cancer Heterogeneity Drives Evolution and Resistance to Intense Hormonal Therapy. Eur Urol. In press. https://doi.org/10.1016/j.eururo.2021.03.009: Focus on Intraductal Carcinoma of the Prostate. Eur Urol 2021; 80:e83-e84. [PMID: 34148711 DOI: 10.1016/j.eururo.2021.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 01/26/2023]
Affiliation(s)
- Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Huihui Ye
- Department of Pathology and Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Rosina T Lis
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
58
|
Kidd SG, Carm KT, Bogaard M, Olsen LG, Bakken AC, Løvf M, Lothe RA, Axcrona K, Axcrona U, Skotheim RI. High expression of SCHLAP1 in primary prostate cancer is an independent predictor of biochemical recurrence, despite substantial heterogeneity. Neoplasia 2021; 23:634-641. [PMID: 34107378 PMCID: PMC8192444 DOI: 10.1016/j.neo.2021.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 01/08/2023]
Abstract
In primary prostate cancer, the common multifocality and heterogeneity are major obstacles in finding robust prognostic tissue biomarkers. The long noncoding RNA SCHLAP1 has been suggested, but its prognostic value has not been investigated in the context of tumor heterogeneity. In the present study, expression of SCHLAP1 was investigated using real-time RT-PCR in a multisampled series of 778 tissue samples from radical prostatectomies of 164 prostate cancer patients (median follow-up time 7.4 y). The prognostic value of SCHLAP1 was evaluated with biochemical recurrence as endpoint. In total, 29% of patients were classified as having high expression of SCHLAP1 in at least one malignant sample. Among these, inter- and intrafocal heterogeneity was detected in 72% and 56%, respectively. High expression of SCHLAP1 was shown to be a predictor of biochemical recurrence in both uni- and multivariable cox regression analyses (P < 0.001 and P = 0.02). High expression of SCHLAP1 was also significantly associated with adverse clinicopathological characteristics, including grade group, high pT stage, invasive cribriform growth/intraductal carcinoma of the prostate, and reactive stroma. In conclusion, high expression of SCHLAP1 in at least one malignant sample is a robust prognostic biomarker in primary prostate cancer. For the first time, high SCHLAP1 expression has been associated with the aggressive histopathologic feature reactive stroma. The expression of SCHLAP1 is highly heterogeneous, and analysis of multiple samples is therefore crucial in determination of the SCHLAP1 status of a patient.
Collapse
Affiliation(s)
- Susanne G Kidd
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristina T Carm
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mari Bogaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Linn Guro Olsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Anne Cathrine Bakken
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Marthe Løvf
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karol Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | - Ulrika Axcrona
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Pathology, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Rolf I Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway; Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
59
|
Epstein JI, Amin MB, Fine SW, Algaba F, Aron M, Baydar DE, Beltran AL, Brimo F, Cheville JC, Colecchia M, Comperat E, da Cunha IW, Delprado W, DeMarzo AM, Giannico GA, Gordetsky JB, Guo CC, Hansel DE, Hirsch MS, Huang J, Humphrey PA, Jimenez RE, Khani F, Kong Q, Kryvenko ON, Kunju LP, Lal P, Latour M, Lotan T, Maclean F, Magi-Galluzzi C, Mehra R, Menon S, Miyamoto H, Montironi R, Netto GJ, Nguyen JK, Osunkoya AO, Parwani A, Robinson BD, Rubin MA, Shah RB, So JS, Takahashi H, Tavora F, Tretiakova MS, True L, Wobker SE, Yang XJ, Zhou M, Zynger DL, Trpkov K. The 2019 Genitourinary Pathology Society (GUPS) White Paper on Contemporary Grading of Prostate Cancer. Arch Pathol Lab Med 2021; 145:461-493. [PMID: 32589068 DOI: 10.5858/arpa.2020-0015-ra] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Controversies and uncertainty persist in prostate cancer grading. OBJECTIVE.— To update grading recommendations. DATA SOURCES.— Critical review of the literature along with pathology and clinician surveys. CONCLUSIONS.— Percent Gleason pattern 4 (%GP4) is as follows: (1) report %GP4 in needle biopsy with Grade Groups (GrGp) 2 and 3, and in needle biopsy on other parts (jars) of lower grade in cases with at least 1 part showing Gleason score (GS) 4 + 4 = 8; and (2) report %GP4: less than 5% or less than 10% and 10% increments thereafter. Tertiary grade patterns are as follows: (1) replace "tertiary grade pattern" in radical prostatectomy (RP) with "minor tertiary pattern 5 (TP5)," and only use in RP with GrGp 2 or 3 with less than 5% Gleason pattern 5; and (2) minor TP5 is noted along with the GS, with the GrGp based on the GS. Global score and magnetic resonance imaging (MRI)-targeted biopsies are as follows: (1) when multiple undesignated cores are taken from a single MRI-targeted lesion, an overall grade for that lesion is given as if all the involved cores were one long core; and (2) if providing a global score, when different scores are found in the standard and the MRI-targeted biopsy, give a single global score (factoring both the systematic standard and the MRI-targeted positive cores). Grade Groups are as follows: (1) Grade Groups (GrGp) is the terminology adopted by major world organizations; and (2) retain GS 3 + 5 = 8 in GrGp 4. Cribriform carcinoma is as follows: (1) report the presence or absence of cribriform glands in biopsy and RP with Gleason pattern 4 carcinoma. Intraductal carcinoma (IDC-P) is as follows: (1) report IDC-P in biopsy and RP; (2) use criteria based on dense cribriform glands (>50% of the gland is composed of epithelium relative to luminal spaces) and/or solid nests and/or marked pleomorphism/necrosis; (3) it is not necessary to perform basal cell immunostains on biopsy and RP to identify IDC-P if the results would not change the overall (highest) GS/GrGp part per case; (4) do not include IDC-P in determining the final GS/GrGp on biopsy and/or RP; and (5) "atypical intraductal proliferation (AIP)" is preferred for an intraductal proliferation of prostatic secretory cells which shows a greater degree of architectural complexity and/or cytological atypia than typical high-grade prostatic intraepithelial neoplasia, yet falling short of the strict diagnostic threshold for IDC-P. Molecular testing is as follows: (1) Ki67 is not ready for routine clinical use; (2) additional studies of active surveillance cohorts are needed to establish the utility of PTEN in this setting; and (3) dedicated studies of RNA-based assays in active surveillance populations are needed to substantiate the utility of these expensive tests in this setting. Artificial intelligence and novel grading schema are as follows: (1) incorporating reactive stromal grade, percent GP4, minor tertiary GP5, and cribriform/intraductal carcinoma are not ready for adoption in current practice.
Collapse
Affiliation(s)
- Jonathan I Epstein
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada.,Urology (Epstein), David Geffen School of Medicine at UCLA, Los Angeles, California (Huang).,and Oncology (Epstein), The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science, Memphis (Amin)
| | - Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Fine)
| | - Ferran Algaba
- Department of Pathology, Fundacio Puigvert, Barcelona, Spain (Algaba)
| | - Manju Aron
- Department of Pathology, University of Southern California, Los Angeles (Aron)
| | - Dilek E Baydar
- Department of Pathology, Faculty of Medicine, Koç University, İstanbul, Turkey (Baydar)
| | - Antonio Lopez Beltran
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal (Beltran)
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montréal, Quebec, Canada (Brimo)
| | - John C Cheville
- Department of Pathology, Mayo Clinic, Rochester, Minnesota (Cheville, Jimenez)
| | - Maurizio Colecchia
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (Colecchia)
| | - Eva Comperat
- Department of Pathology, Hôpital Tenon, Sorbonne University, Paris, France (Comperat)
| | | | | | - Angelo M DeMarzo
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada
| | - Giovanna A Giannico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Giannico, Gordetsky)
| | - Jennifer B Gordetsky
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee (Giannico, Gordetsky)
| | - Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston (Guo)
| | - Donna E Hansel
- Department of Pathology, Oregon Health and Science University, Portland (Hansel)
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (Hirsch)
| | - Jiaoti Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California (Huang)
| | - Peter A Humphrey
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut (Humphrey)
| | - Rafael E Jimenez
- Department of Pathology, Mayo Clinic, Rochester, Minnesota (Cheville, Jimenez)
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, New York (Khani, Robinson)
| | - Qingnuan Kong
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, Shandong, China (Kong).,Kong is currently located at Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Oleksandr N Kryvenko
- Departments of Pathology and Laboratory Medicine and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida (Kryvenko)
| | - L Priya Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan (Kunju, Mehra)
| | - Priti Lal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (Lal)
| | - Mathieu Latour
- Department of Pathology, CHUM, Université de Montréal, Montréal, Quebec, Canada (Latour)
| | - Tamara Lotan
- From the Departments of Pathology (Epstein, DeMarzo, Lotan), McGill University Health Center, Montréal, Quebec, Canada
| | - Fiona Maclean
- Douglass Hanly Moir Pathology, Faculty of Medicine and Health Sciences Macquarie University, North Ryde, Australia (Maclean)
| | - Cristina Magi-Galluzzi
- Department of Pathology, The University of Alabama at Birmingham, Birmingham (Magi-Galluzzi, Netto)
| | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan (Kunju, Mehra)
| | - Santosh Menon
- Department of Surgical Pathology, Tata Memorial Hospital, Parel, Mumbai, India (Menon)
| | - Hiroshi Miyamoto
- Departments of Pathology and Laboratory Medicine and Urology, University of Rochester Medical Center, Rochester, New York (Miyamoto)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, Ancona, Italy (Montironi)
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham (Magi-Galluzzi, Netto)
| | - Jane K Nguyen
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio (Nguyen)
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia (Osunkoya)
| | - Anil Parwani
- Department of Pathology, Ohio State University, Columbus (Parwani, Zynger)
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, New York (Khani, Robinson)
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland (Rubin)
| | - Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas (Shah)
| | - Jeffrey S So
- Institute of Pathology, St Luke's Medical Center, Quezon City and Global City, Philippines (So)
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan (Takahashi)
| | - Fabio Tavora
- Argos Laboratory, Federal University of Ceara, Fortaleza, Brazil (Tavora)
| | - Maria S Tretiakova
- Department of Pathology, University of Washington School of Medicine, Seattle (Tretiakova, True)
| | - Lawrence True
- Department of Pathology, University of Washington School of Medicine, Seattle (Tretiakova, True)
| | - Sara E Wobker
- Departments of Pathology and Laboratory Medicine and Urology, University of North Carolina, Chapel Hill (Wobker)
| | - Ximing J Yang
- Department of Pathology, Northwestern University, Chicago, Illinois (Yang)
| | - Ming Zhou
- Department of Pathology, Tufts Medical Center, Boston, Massachusetts (Zhou)
| | - Debra L Zynger
- Department of Pathology, Ohio State University, Columbus (Parwani, Zynger)
| | - Kiril Trpkov
- and Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada (Trpkov)
| |
Collapse
|
60
|
Hesterberg AB, Gordetsky JB, Hurley PJ. Cribriform Prostate Cancer: Clinical Pathologic and Molecular Considerations. Urology 2021; 155:47-54. [PMID: 34058243 DOI: 10.1016/j.urology.2021.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Intraductal cribriform (IDC) and invasive cribriform morphologies are associated with worse prostate cancer outcomes. Limited retrospective studies have associated IDC and cribriform morphology with germline mutations in DNA repair genes, particularly BRCA2. These findings, which prompted the National Comprehensive Cancer Network (NCCN) Guidelines for Prostate Cancer and Genetic/Familial High- Risk Assessment to consider germline testing for individuals with IDC/cribriform histology, have been questioned in a recent prospective study. A deepened understanding of the molecular mechanisms driving disease aggressiveness in cribriform morphology is critical to provide more clarity in clinical decision making. This review summarizes the current understanding of IDC and cribriform prostate cancer, with an emphasis on clinical outcomes and molecular alterations.
Collapse
Affiliation(s)
| | - Jennifer B Gordetsky
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN; Department of Urology, Vanderbilt University Medical Center, Nashville, TN
| | - Paula J Hurley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Urology, Vanderbilt University Medical Center, Nashville, TN; Vanderbilt-Ingram Cancer Center, Nashville, TN.
| |
Collapse
|
61
|
Hesterberg AB, Rios BL, Wolf EM, Tubbs C, Wong HY, Schaffer KR, Lotan TL, Giannico GA, Gordetsky JB, Hurley PJ. A distinct repertoire of cancer-associated fibroblasts is enriched in cribriform prostate cancer. J Pathol Clin Res 2021; 7:271-286. [PMID: 33600062 PMCID: PMC8073007 DOI: 10.1002/cjp2.205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/11/2020] [Accepted: 01/13/2021] [Indexed: 12/27/2022]
Abstract
Outcomes for men with localized prostate cancer vary widely, with some men effectively managed without treatment on active surveillance, while other men rapidly progress to metastatic disease despite curative-intent therapies. One of the strongest prognostic indicators of outcome is grade groups based on the Gleason grading system. Gleason grade 4 prostate cancer with cribriform morphology is associated with adverse outcomes and can be utilized clinically to improve risk stratification. The underpinnings of disease aggressiveness associated with cribriform architecture are not fully understood. Most studies have focused on genetic and molecular alterations in cribriform tumor cells; however, less is known about the tumor microenvironment in cribriform prostate cancer. Cancer-associated fibroblasts (CAFs) are a heterogeneous population of fibroblasts in the tumor microenvironment that impact cancer aggressiveness. The overall goal of this study was to determine if cribriform prostate cancers are associated with a unique repertoire of CAFs. Radical prostatectomy whole-tissue sections were analyzed for the expression of fibroblast markers (ASPN in combination with FAP, THY1, ENG, NT5E, TNC, and PDGFRβ) in stroma adjacent to benign glands and in Gleason grade 3, Gleason grade 4 cribriform, and Gleason grade 4 noncribriform prostate cancer by RNAscope®. Halo® Software was used to quantify percent positive stromal cells and expression per positive cell. The fibroblast subtypes enriched in prostate cancer were highly heterogeneous. Both overlapping and distinct populations of low abundant fibroblast subtypes in benign prostate stroma were enriched in Gleason grade 4 prostate cancer with cribriform morphology compared to Gleason grade 4 prostate cancer with noncribriform morphology and Gleason grade 3 prostate cancer. In addition, gene expression was distinctly altered in CAF subtypes adjacent to cribriform prostate cancer. Overall, these studies suggest that cribriform prostate cancer has a unique tumor microenvironment that may distinguish it from other Gleason grade 4 morphologies and lower Gleason grades.
Collapse
Affiliation(s)
| | - Brenda L Rios
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Elysa M Wolf
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Colby Tubbs
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Hong Yuen Wong
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Kerry R Schaffer
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Tamara L Lotan
- Department of PathologyJohns Hopkins School of MedicineBaltimoreMDUSA
| | - Giovanna A Giannico
- Department of PathologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of UrologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Jennifer B Gordetsky
- Department of PathologyVanderbilt University Medical CenterNashvilleTNUSA
- Department of UrologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Paula J Hurley
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of UrologyVanderbilt University Medical CenterNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
62
|
Molecular Expression of Some Oncogenes and Predisposing Behaviors Contributing to the Aggressiveness of Prostate Cancer. Rep Biochem Mol Biol 2021; 10:60-68. [PMID: 34277869 DOI: 10.52547/rbmb.10.1.60] [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: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 11/18/2022]
Abstract
Background Prostate cancer is the second most common cancer in men in Iran. It can be treated in the early stages of the disease; therefore, early diagnosis can be lifesaving. The aim of this study was to investigate the molecular expression of some oncogenes and predisposing behaviors contributing to the aggressiveness of prostate cancer. Methods In this case-control study, prostate cancer specimens were collected from both patients and healthy volunteers. Several factors such as age, family history, smoking, and stage of the disease, were investigated based on the criteria of this study. Real-time PCR was used to measure the expression of four oncogenes. Statistical analysis of our data was carried out using SPSS software version 22. Results The X2 test showed that there was a difference in the incidence of prostate cancer in different age groups (X2= 9.30; p= 0.026). Although data analysis by the X2 test showed that family history had a significant effect on prostate cancer (X2= 14.43; p= 0.001), smoking did not show a significant effect on the incidence of this disorder (X2= 4.67; p= 0.097). The T2N1M0 stage is the most common form of prostate cancer in patients with family history of prostate cancer and the habit of smoking. Also, the expression of KRAS1P, GLB1L2, SChLAP1 and PACSIN3 oncogenes reduced in prostate cancer samples compared to the control group. Conclusion Overall, functional interpretation of gene expression in the prostate tissue can affect tumor progression. Yet, further practical studies are required to reveal the accurate underlying mechanisms.
Collapse
|
63
|
Porter LH, Bakshi A, Pook D, Clark A, Clouston D, Kourambas J, Goode DL, Risbridger GP, Taylor RA, Lawrence MG. Androgen receptor enhancer amplification in matched patient-derived xenografts of primary and castrate-resistant prostate cancer. J Pathol 2021; 254:121-134. [PMID: 33620092 DOI: 10.1002/path.5652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/30/2022]
Abstract
Amplifications of the androgen receptor (AR) occur in up to 80% of men with castration-resistant prostate cancer (CRPC). Recent studies highlighted that these amplifications not only span the AR gene but usually encompass a distal enhancer. This represents a newly recognised, non-coding mechanism of resistance to AR-directed therapies, including enzalutamide. To study disease progression before and after AR amplification, we used tumour samples from a castrate-sensitive primary tumour and castrate-resistant metastasis of the same patient. For subsequent functional and genomic studies, we established serially transplantable patient-derived xenografts (PDXs). Whole genome sequencing showed that alterations associated with poor prognosis, such as TP53 and PTEN loss, existed before androgen deprivation therapy, followed by co-amplification of the AR gene and enhancer after the development of metastatic CRPC. The PDX of the primary tumour, without the AR amplification, was sensitive to AR-directed treatments, including castration, enzalutamide, and apalutamide. The PDX of the metastasis, with the AR amplification, had higher AR and AR-V7 expression in castrate conditions, and was resistant to castration, apalutamide, and enzalutamide in vivo. Treatment with a BET inhibitor outperformed the AR-directed therapies for the metastasis, resulting in tumour regression for some, but not all, grafts. Therefore, this study provides novel matched PDXs to test potential treatments that target the overabundance of AR in tumours with AR enhancer amplifications. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Laura H Porter
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Andrew Bakshi
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Pook
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Medical Oncology, Monash Health, Clayton, VIC, Australia
| | - Ashlee Clark
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - John Kourambas
- Department of Medicine, Monash Health, Casey Hospital, Berwick, VIC, Australia
| | -
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Melbourne Urological Research Alliance (MURAL), Biomedicine Discovery Institute Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - David L Goode
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Gail P Risbridger
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Renea A Taylor
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Mitchell G Lawrence
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
64
|
Hollemans E, Verhoef EI, Bangma CH, Rietbergen J, Roobol MJ, Helleman J, van Leenders GJLH. Clinical outcome comparison of Grade Group 1 and Grade Group 2 prostate cancer with and without cribriform architecture at the time of radical prostatectomy. Histopathology 2021; 76:755-762. [PMID: 31944367 PMCID: PMC7216977 DOI: 10.1111/his.14064] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/08/2019] [Accepted: 01/13/2020] [Indexed: 11/30/2022]
Abstract
Aims Invasive cribriform and intraductal carcinoma are associated with aggressive disease in Grade Group 2 (GG2) prostate cancer patients. However, the characteristics and clinical outcome of patients with GG2 prostate cancer without cribriform architecture (GG2−) as compared with those with Grade Group 1 (GG1) prostate cancer are unknown. The aim of this study was to investigate the clinical and pathological characteristics of GG1 and GG2− prostate cancer in radical prostatectomy specimens. Methods and results We reviewed 835 radical prostatectomy specimens for Grade Group, pT stage, surgical margin status, and the presence of cribriform architecture. Biochemical recurrence‐free survival and metastasis were used as clinical outcomes. GG1 prostate cancer was seen in 207 patients, and GG2 prostate cancer was seen in 420 patients, of whom 228 (54%) showed cribriform architecture (GG2+) and 192 (46%) did not. GG2− patients had higher prostate‐specific antigen levels (9.4 ng/ml versus 7.0 ng/ml; P < 0.001), more often had extraprostatic extension (36% versus 11%; P < 0.001) and had more positive surgical margins (27% versus 17%; P = 0.01) than GG1 patients. GG2− patients had shorter biochemical recurrence‐free survival (hazard ratio 2.7, 95% confidence interval 1.4–4.9; P = 0.002) than GG1 patients. Lymph node and distant metastasis were observed neither in GG2− nor in GG1 patients, but occurred in 22 of 228 (10%) GG2+ patients. Conclusion In conclusion, patients with GG2− prostate cancer at radical prostatectomy have more advanced disease and shorter biochemical recurrence‐free survival than those with GG1 prostate cancer, but both groups have a very low risk of developing metastasis.
Collapse
Affiliation(s)
- Eva Hollemans
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Chris H Bangma
- Department of Urology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - John Rietbergen
- Department of Urology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Jozien Helleman
- Department of Urology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | | |
Collapse
|
65
|
Lozano R, Salles DC, Sandhu S, Aragón IM, Thorne H, López-Campos F, Rubio-Briones J, Gutierrez-Pecharroman AM, Maldonado L, di Domenico T, Sanz A, Prieto JD, García I, Pacheco MI, Garcés T, Llacer C, Romero-Laorden N, Zambrana F, López-Casas PP, Lorente D, Mateo J, Pritchard CC, Antonarakis ES, Olmos D, Lotan TL, Castro E. Association between BRCA2 alterations and intraductal and cribriform histologies in prostate cancer. Eur J Cancer 2021; 147:74-83. [PMID: 33626496 DOI: 10.1016/j.ejca.2021.01.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Intraductal (IDC) and cribriform (CRIB) histologies in prostate cancer have been associated with germline BRCA2 (gBRCA2) mutations in small retrospective series, leading to the recommendation of genetic testing for patients with IDC in the primary tumour. PATIENTS AND METHODS To examine the association of gBRCA2 mutations and other tumour molecular features with IDC and/or cribriform (CRIB) histologies, we conducted a case-control study in which primary prostate tumours from 58 gBRCA2 carriers were matched (1:2) by Gleason Grade Group and specimen type to 116 non-carriers. Presence/absence of IDC and CRIB morphologies was established by two expert uropathologists blinded to gBRCA2 status. Fluorescent in-situ hybridization (FISH) and next-generation sequencing (NGS) were used to detect BRCA2 alterations, PTEN deletions and TMPRSS2-ERG fusions. Chi-squared tests were used to compare the frequency of IDC and CRIB in gBRCA2 carriers and controls and to assess associations with other variables. Logistic regression models were constructed to identify independent factors associated with both histology patterns. RESULTS No significant differences between gBRCA2 carriers and non-carriers were observed in the prevalence of IDC (36% gBRCA2 versus 50% non-carriers, p = 0.085) or CRIB (53% gBRCA2 versus 43% non-carriers p = 0.197) patterns. However, IDC histology was independently associated with bi-allelic BRCA2 alterations (OR 4.3, 95%CI 1.1-16.2) and PTEN homozygous loss (OR 5.2, 95%CI 2.1-13.1). CRIB morphology was also independently associated with bi-allelic BRCA2 alterations (OR 5.6, 95%CI 1.7-19.3). CONCLUSIONS While we found no association between gBRCA2 mutations and IDC or CRIB histologies, bi-allelic BRCA2 loss in primary prostate tumours was significantly associated with both variant morphologies, independently of other clinical-pathologic factors.
Collapse
Affiliation(s)
- Rebeca Lozano
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Shahneen Sandhu
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Isabel M Aragón
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Heather Thorne
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Fernando López-Campos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Rubio-Briones
- Urology Department, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Ana M Gutierrez-Pecharroman
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Department of Pathology, Hospital de Getafe, Getafe, Spain
| | - Laneisha Maldonado
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Tomas di Domenico
- Bioinformatics Unit, Spanish National Cancer Research Center, Madrid, Spain
| | - Alejandro Sanz
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Juan D Prieto
- Department of Pathology, Hospital Universitarios Virgen de la Victoria Málaga, Spain
| | - Isabel García
- Department of Pathology, Hospital Universitarios Virgen de la Victoria Málaga, Spain
| | - María I Pacheco
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - Teresa Garcés
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Casilda Llacer
- Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain; Medical Oncology, UGCI, Hospitales Universitarios Virgen de la Victoria y Regional de Málaga, Málaga, Spain
| | | | | | - Pedro P López-Casas
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain
| | - David Lorente
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Medical Oncology, Hospital Provincial de Castellón, Castellón de la Plana, Spain
| | - Joaquin Mateo
- Prostate Cancer Traslational Research Unit, Vall'Hebron Institute of Oncology, Spain
| | | | - Emmanuel S Antonarakis
- Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David Olmos
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Elena Castro
- Prostate Cancer Clinical Research Unit, Spanish National Cancer Research Centre, Madrid, Spain; Genitourinary Cancer Translational Research Group, Instituto de Investigación Biomédica de Málaga, Spain; Medical Oncology, UGCI, Hospitales Universitarios Virgen de la Victoria y Regional de Málaga, Málaga, Spain.
| |
Collapse
|
66
|
Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, Gillessen S, Van der Kwast T, Bristow RG. Prostate cancer. Nat Rev Dis Primers 2021. [PMID: 33542230 DOI: 10.1038/s41572-020-0024.3-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2023]
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
Affiliation(s)
- Richard J Rebello
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
| | - Christoph Oing
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK
- Department of Oncology, Haematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Centre Eppendorf, Hamburg, Germany
| | - Karen E Knudsen
- Sidney Kimmel Cancer Center at Jefferson Health and Thomas Jefferson University, Philadelphia, PA, USA
| | - Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, Manhattan, NY, USA
| | - David C Johnson
- Department of Urology, University of North Carolina, Chapel Hill, NC, USA
| | - Robert E Reiter
- Department of Urology, Jonssen Comprehensive Cancer Center UCLA, Los Angeles, CA, USA
| | | | - Theodorus Van der Kwast
- Laboratory Medicine Program, Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Robert G Bristow
- Cancer Research UK Manchester Institute, University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
67
|
Abstract
Prostate cancer is a complex disease that affects millions of men globally, predominantly in high human development index regions. Patients with localized disease at a low to intermediate risk of recurrence generally have a favourable outcome of 99% overall survival for 10 years if the disease is detected and treated at an early stage. Key genetic alterations include fusions of TMPRSS2 with ETS family genes, amplification of the MYC oncogene, deletion and/or mutation of PTEN and TP53 and, in advanced disease, amplification and/or mutation of the androgen receptor (AR). Prostate cancer is usually diagnosed by prostate biopsy prompted by a blood test to measure prostate-specific antigen levels and/or digital rectal examination. Treatment for localized disease includes active surveillance, radical prostatectomy or ablative radiotherapy as curative approaches. Men whose disease relapses after prostatectomy are treated with salvage radiotherapy and/or androgen deprivation therapy (ADT) for local relapse, or with ADT combined with chemotherapy or novel androgen signalling-targeted agents for systemic relapse. Advanced prostate cancer often progresses despite androgen ablation and is then considered castration-resistant and incurable. Current treatment options include AR-targeted agents, chemotherapy, radionuclides and the poly(ADP-ribose) inhibitor olaparib. Current research aims to improve prostate cancer detection, management and outcomes, including understanding the fundamental biology at all stages of the disease.
Collapse
|
68
|
Comedonecrosis Gleason pattern 5 is associated with worse clinical outcome in operated prostate cancer patients. Mod Pathol 2021; 34:2064-2070. [PMID: 34175896 PMCID: PMC8514328 DOI: 10.1038/s41379-021-00860-4] [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: 03/08/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 01/04/2023]
Abstract
Individual growth patterns and cribriform architecture are increasingly considered in risk stratification and clinical decision-making in men with prostate cancer. Our objective was to establish the prognostic value of individual Gleason 5 patterns in a radical prostatectomy (RP) cohort. We reviewed 1064 RPs and recorded Grade Group (GG), pT-stage, surgical margin status, Gleason 4 and 5 growth patterns as well as intraductal carcinoma. The clinical endpoints were biochemical recurrence and post-operative distant metastasis. Gleason pattern 5 was present in 339 (31.9%) RPs, of which 47 (4.4%) presented as primary, 166 (15.6%) as secondary, and 126 (11.8%) as tertiary pattern. Single cells/cords were present in 321 (94.7%) tumors with Gleason pattern 5, solid fields in 90 (26.5%), and comedonecrosis in invasive carcinoma in 32 (9.4%) tumors. Solid fields demonstrated either a small nested morphology (n = 50, 14.7%) or medium to large solid fields (n = 61, 18.0%). Cribriform architecture was present in 568 (53.4%) RPs. Medium to large solid fields and comedonecrosis coincided with cribriform architecture in all specimens, and were not observed in cribriform-negative cases. In multivariable analysis adjusted for Prostate-Specific Antigen, pT-stage, GG, surgical margin status and lymph node metastases, cribriform architecture (Hazard Ratio (HR) 9.9; 95% Confidence Interval (CI) 3.9-25.5, P < 0.001) and comedonecrosis (HR 2.1, 95% CI 1.2-3.7, P = 0.01) were independent predictors for metastasis-free survival, while single cells/cords (HR 1.2; 95% CI 0.7-1.8, P = 0.55) and medium to large solid fields (HR 1.6, 95% CI 0.9-2.7, P = 0.09) were not. In conclusion, comedonecrosis in invasive carcinoma is an independent prognostic Gleason 5 pattern for metastasis-free survival after RP. These data support the current recommendations to routinely include cribriform pattern in pathology reports and indicate that comedonecrosis should also be commented on.
Collapse
|
69
|
Rubin MA, Bristow RG, Thienger PD, Dive C, Imielinski M. Impact of Lineage Plasticity to and from a Neuroendocrine Phenotype on Progression and Response in Prostate and Lung Cancers. Mol Cell 2020; 80:562-577. [PMID: 33217316 PMCID: PMC8399907 DOI: 10.1016/j.molcel.2020.10.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/06/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
Intratumoral heterogeneity can occur via phenotype transitions, often after chronic exposure to targeted anticancer agents. This process, termed lineage plasticity, is associated with acquired independence to an initial oncogenic driver, resulting in treatment failure. In non-small cell lung cancer (NSCLC) and prostate cancers, lineage plasticity manifests when the adenocarcinoma phenotype transforms into neuroendocrine (NE) disease. The exact molecular mechanisms involved in this NE transdifferentiation remain elusive. In small cell lung cancer (SCLC), plasticity from NE to nonNE phenotypes is driven by NOTCH signaling. Herein we review current understanding of NE lineage plasticity dynamics, exemplified by prostate cancer, NSCLC, and SCLC.
Collapse
Affiliation(s)
- Mark A Rubin
- Department for BioMedical Research, University of Bern and Inselspital, 3010 Bern, Switzerland; Bern Center for Precision Medicine, University of Bern and Inselspital, 3010 Bern, Switzerland.
| | - Robert G Bristow
- Manchester Cancer Research Centre and Cancer Research UK Manchester Institute, University of Manchester, Macclesfield SK10 4TG, UK
| | - Phillip D Thienger
- Department for BioMedical Research, University of Bern and Inselspital, 3010 Bern, Switzerland
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Macclesfield SK10 4TG, UK
| | - Marcin Imielinski
- Pathology and Laboratory Medicine and Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
70
|
Abstract
Prostate cancer (PCa) is a clinically heterogeneous disease and has poor patient outcome when tumours progress to castration-resistant and metastatic states. Understanding the mechanistic basis for transition to late stage aggressive disease is vital for both assigning patient risk status in the localised setting and also identifying novel treatment strategies to prevent progression. Subregions of intratumoral hypoxia are found in all solid tumours and are associated with many biologic drivers of tumour progression. Crucially, more recent findings show the co-presence of hypoxia and genomic instability can confer a uniquely adverse prognosis in localised PCa patients. In-depth informatic and functional studies suggests a role for hypoxia in co-operating with oncogenic drivers (e.g. loss of PTEN) and suppressing DNA repair capacity to alter clonal evolution due to an aggressive mutator phenotype. More specifically, hypoxic suppression of homologous recombination represents a “contextual lethal“ vulnerability in hypoxic prostate tumours which could extend the application of existing DNA repair targeting agents such as poly-ADP ribose polymerase inhibitors. Further investigation is now required to assess this relationship on the background of existing genomic alterations relevant to PCa, and also characterise the role of hypoxia in driving early metastatic spread. On this basis, PCa patients with hypoxic tumours can be better stratified into risk categories and treated with appropriate therapies to prevent progression.
Collapse
Affiliation(s)
- Jack Ashton
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert Bristow
- Translational Oncogenomics, CRUK Manchester Institute and CRUK Manchester Centre, Manchester, United Kingdom
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
71
|
Burdak-Rothkamm S, Mansour WY, Rothkamm K. DNA Damage Repair Deficiency in Prostate Cancer. Trends Cancer 2020; 6:974-984. [DOI: 10.1016/j.trecan.2020.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
72
|
Zong Y, Montironi R, Massari F, Jiang Z, Lopez-Beltran A, Wheeler TM, Scarpelli M, Santoni M, Cimadamore A, Cheng L. Intraductal Carcinoma of the Prostate: Pathogenesis and Molecular Perspectives. Eur Urol Focus 2020; 7:955-963. [PMID: 33132109 DOI: 10.1016/j.euf.2020.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/15/2020] [Accepted: 10/15/2020] [Indexed: 11/18/2022]
Abstract
Intraductal carcinoma of the prostate (IDC-P), a clinicopathological entity characterized by malignant prostatic epithelial cells growing within ducts and/or acini, has a distinct architectural pattern, cytological features, and biological behavior. Whereas most IDC-P tumors could be derived from adjacent high-grade invasive cancer via retrograde spreading of cancer cells along benign ducts and acini, a small subset of IDC-P may arise from the transformation and intraductal proliferation of precancerous cells induced by various oncogenic events. These isolated IDC-P tumors possess a distinct mutational profile and may function as a carcinoma in situ lesion with de novo intraductal outgrowth of malignant cells. Further molecular characterization of these two types of IDC-P and better understanding of the mechanisms underlying IDC-P formation and progression could be translated into valuable biomarkers for differential diagnosis and actionable targets for therapeutic interventions. PATIENT SUMMARY: Intraductal carcinoma of the prostate is an aggressive type of prostate cancer associated with high risk for local recurrence and distant metastasis. In this review, we discussed pathogenesis, biomarkers, differential diagnoses, and therapeutic strategies for this tumor.
Collapse
Affiliation(s)
- Yang Zong
- Department of Pathology, University of Massachusetts Memorial Medical Center, Worcester, MA, USA
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Francesco Massari
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Zhong Jiang
- Department of Pathology, University of Massachusetts Memorial Medical Center, Worcester, MA, USA
| | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | - Thomas M Wheeler
- Department of Pathology and Laboratory Medicine, Baylor St. Luke's Medical Center, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
73
|
Fiard G, Norris JM, Nguyen TA, Stavrinides V, Olivier J, Emberton M, Moore CM. What to expect from a non-suspicious prostate MRI? A review. Prog Urol 2020; 30:986-999. [PMID: 33008718 DOI: 10.1016/j.purol.2020.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/06/2020] [Accepted: 09/04/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Many guidelines now recommend multiparametric MRI (mpMRI) prior to an initial or repeat prostate biopsy. However, clinical decision making for men with a non-suspicious mpMRI (Likert or PIRADS score 1-2) varies. OBJECTIVES To review the most recent literature to answer three questions. (1) Should we consider systematic biopsy if mpMRI is not suspicious? (2) Are there additional predictive factors that can help decide which patient should have a biopsy? (3) Can the low visibility of some cancers be explained and what are the implications? SOURCES A narrative review was performed in Medline databases using two searches with the terms "MRI" and "prostate cancer" and ("diagnosis" or "biopsy") and ("non-suspicious" or "negative" or "invisible"); "prostate cancer MRI visible". References of the selected articles were screened for additional articles. STUDY SELECTION Studies published in the last 5 years in English language were assessed for eligibility and selected if data was available to answer one of the three study questions. RESULTS Considering clinically significant cancer as ISUP grade≥2, the negative predictive value (NPV) of mpMRI in various settings and populations ranges from 76% to 99%, depending on cancer prevalence and the type of confirmatory reference test used. NPV is higher among patients with prior negative biopsy (88-96%), and lower for active surveillance patients (85-90%). The PSA density (PSAd) with a threshold of PSAd<0.15ng/ml/ml was the most studied and relevant predictive factor used in combination with mpMRI to rule out clinically significant cancer. Finally, mpMRI-invisible tumours appear to differ from a histopathological and genetic point of view, conferring clinical advantage to invisibility. LIMITATIONS Most published data come from expert centres and results may not be reproducible in all settings. CONCLUSION mpMRI has high diagnostic accuracy and in cases of negative mpMRI, PSA density can be used to determine which patient should have a biopsy. Growing knowledge of the mechanisms and genetics underlying MRI visibility will help develop more accurate risk calculators and biomarkers.
Collapse
Affiliation(s)
- G Fiard
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College London Hospital NHS Foundation Trust, London, UK; Department of Urology, Grenoble Alpes University Hospital, Grenoble, France; Université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France.
| | - J M Norris
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College London Hospital NHS Foundation Trust, London, UK
| | - T A Nguyen
- Department of urology, université de Brest, CHRU, Brest, France
| | - V Stavrinides
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College London Hospital NHS Foundation Trust, London, UK
| | - J Olivier
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of urology, Lille university, CHU Lille, Lille, France
| | - M Emberton
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College London Hospital NHS Foundation Trust, London, UK
| | - C M Moore
- UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College London Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
74
|
Fine SW, Trpkov K, Amin MB, Algaba F, Aron M, Baydar DE, Beltran AL, Brimo F, Cheville JC, Colecchia M, Comperat E, Costello T, da Cunha IW, Delprado W, DeMarzo AM, Giannico GA, Gordetsky JB, Guo CC, Hansel DE, Hirsch MS, Huang J, Humphrey PA, Jimenez RE, Khani F, Kong MX, Kryvenko ON, Kunju LP, Lal P, Latour M, Lotan T, Maclean F, Magi-Galluzzi C, Mehra R, Menon S, Miyamoto H, Montironi R, Netto GJ, Nguyen JK, Osunkoya AO, Parwani A, Pavlovich CP, Robinson BD, Rubin MA, Shah RB, So JS, Takahashi H, Tavora F, Tretiakova MS, True L, Wobker SE, Yang XJ, Zhou M, Zynger DL, Epstein JI. Practice patterns related to prostate cancer grading: results of a 2019 Genitourinary Pathology Society clinician survey. Urol Oncol 2020; 39:295.e1-295.e8. [PMID: 32948433 DOI: 10.1016/j.urolonc.2020.08.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE To survey urologic clinicians regarding interpretation of and practice patterns in relation to emerging aspects of prostate cancer grading, including quantification of high-grade disease, cribriform/intraductal carcinoma, and impact of magnetic resonance imaging-targeted needle biopsy. MATERIALS AND METHODS The Genitourinary Pathology Society distributed a survey to urology and urologic oncology-focused societies and hospital departments. Eight hundred and thirty four responses were collected and analyzed using descriptive statistics. RESULTS Eighty percent of survey participants use quantity of Gleason pattern 4 on needle biopsy for clinical decisions, less frequently with higher Grade Groups. Fifty percent interpret "tertiary" grade as a minor/<5% component. Seventy percent of respondents would prefer per core grading as well as a global/overall score per set of biopsies, but 70% would consider highest Gleason score in any single core as the grade for management. Seventy five percent utilize Grade Group terminology in patient discussions. For 45%, cribriform pattern would affect management, while for 70% the presence of intraductal carcinoma would preclude active surveillance. CONCLUSION This survey of practice patterns in relationship to prostate cancer grading highlights similarities and differences between contemporary pathology reporting and its clinical application. As utilization of Gleason pattern 4 quantification, minor tertiary pattern, cribriform/intraductal carcinoma, and the incorporation of magnetic resonance imaging-based strategies evolve, these findings may serve as a basis for more nuanced communication and guide research efforts involving pathologists and clinicians.
Collapse
Affiliation(s)
- Samson W Fine
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.
| | - Kiril Trpkov
- Department of Pathology and Lab Medicine, University of Calgary and Alberta Precision Labs, Calgary, AB, Canada
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science, Memphis, TN
| | - Ferran Algaba
- Department of Pathology, Fundacio Puigvert, Barcelona, Spain
| | - Manju Aron
- Department of Pathology, University of Southern California, Los Angeles, CA
| | - Dilek E Baydar
- Department of Pathology, Faculty of Medicine, Koç University, İstanbul, Turkey
| | | | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montréal, QC, Canada
| | | | - Maurizio Colecchia
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eva Comperat
- Department of Pathology, Hôpital Tenon, Sorbonne University, Paris, France
| | - Tony Costello
- Department of Urology, Royal Melbourne Hospital, Melbourne, Australia
| | | | | | - Angelo M DeMarzo
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Giovanna A Giannico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jennifer B Gordetsky
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Charles C Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Donna E Hansel
- Department of Pathology, Oregon Health and Science University Portland OR, USA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, NC
| | | | | | - Francesca Khani
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, NY
| | - Max X Kong
- Department of Pathology, Kaiser Permanente Sacramento Medical Center, CA
| | - Oleksandr N Kryvenko
- Departments of Pathology and Laboratory Medicine and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - L Priya Kunju
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Priti Lal
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mathieu Latour
- Department of Pathology, CHUM, Université de Montréal, Montréal, QC, Canada
| | - Tamara Lotan
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | | | | | - Rohit Mehra
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Santosh Menon
- Department of Surgical Pathology, Tata Memorial Hospital, Parel, Mumbai, India
| | - Hiroshi Miyamoto
- Departments of Pathology and Laboratory Medicine and Urology, University of Rochester Medical Center, Rochester, NY
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, Ancona, Italy
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL
| | - Jane K Nguyen
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Adeboye O Osunkoya
- Department of Pathology, Emory University School of Medicine, Atlanta, GA
| | - Anil Parwani
- Department of Pathology, Ohio State University, Columbus, OH
| | - Christian P Pavlovich
- Departments of Urology and Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine and Urology, Weill Cornell Medicine, New York, NY
| | - Mark A Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey S So
- Institute of Pathology, St Luke's Medical Center, Quezon City and Global City, Philippines
| | - Hiroyuki Takahashi
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Fabio Tavora
- Argos Laboratory, Federal University of Ceara, Fortaleza, Brazil
| | - Maria S Tretiakova
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Lawrence True
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| | - Sara E Wobker
- Departments of Pathology and Laboratory Medicine and Urology, University of North Carolina, Chapel Hill, NC
| | - Ximing J Yang
- Department of Pathology, Northwestern University, Chicago, IL
| | - Ming Zhou
- Department of Pathology, Tufts Medical Center, Boston, MA
| | - Debra L Zynger
- Department of Pathology, Ohio State University, Columbus, OH
| | - Jonathan I Epstein
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD; Departments of Urology and Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
75
|
van Leenders GJLH, Verhoef EI, Hollemans E. Prostate cancer growth patterns beyond the Gleason score: entering a new era of comprehensive tumour grading. Histopathology 2020; 77:850-861. [PMID: 32683729 PMCID: PMC7756302 DOI: 10.1111/his.14214] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
Abstract
The Gleason grading system is one of the most important factors in clinical decision‐making for prostate cancer patients, and is entirely based on the classification of tumour growth patterns. In recent years it has become clear that some individual growth patterns themselves have independent prognostic value, and could be used for better personalised risk stratification. In this review we summarise recent literature on the clinicopathological value and molecular characteristics of individual prostate cancer growth patterns, and show how these, most particularly cribriform architecture, could alter treatment decisions for prostate cancer patients.
Collapse
Affiliation(s)
| | - Esther I Verhoef
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Eva Hollemans
- Department of Pathology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
76
|
Haider S, Tyekucheva S, Prandi D, Fox NS, Ahn J, Xu AW, Pantazi A, Park PJ, Laird PW, Sander C, Wang W, Demichelis F, Loda M, Boutros PC. Systematic Assessment of Tumor Purity and Its Clinical Implications. JCO Precis Oncol 2020; 4:2000016. [PMID: 33015524 PMCID: PMC7529507 DOI: 10.1200/po.20.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 02/03/2023] Open
Abstract
PURPOSE The tumor microenvironment is complex, comprising heterogeneous cellular populations. As molecular profiles are frequently generated using bulk tissue sections, they represent an admixture of multiple cell types (including immune, stromal, and cancer cells) interacting with each other. Therefore, these molecular profiles are confounded by signals emanating from many cell types. Accurate assessment of residual cancer cell fraction is crucial for parameterization and interpretation of genomic analyses, as well as for accurately interpreting the clinical properties of the tumor. MATERIALS AND METHODS To benchmark cancer cell fraction estimation methods, 10 estimators were applied to a clinical cohort of 333 patients with prostate cancer. These methods include gold-standard multiobserver pathology estimates, as well as estimates inferred from genome, epigenome, and transcriptome data. In addition, two methods based on genomic and transcriptomic profiles were used to quantify tumor purity in 4,497 tumors across 12 cancer types. Bulk mRNA and microRNA profiles were subject to in silico deconvolution to estimate cancer cell-specific mRNA and microRNA profiles. RESULTS We present a systematic comparison of 10 tumor purity estimation methods on a cohort of 333 prostate tumors. We quantify variation among purity estimation methods and demonstrate how this influences interpretation of clinico-genomic analyses. Our data show poor concordance between pathologic and molecular purity estimates, necessitating caution when interpreting molecular results. Limited concordance between DNA- and mRNA-derived purity estimates remained a general pan-cancer phenomenon when tested in an additional 4,497 tumors spanning 12 cancer types. CONCLUSION The choice of tumor purity estimation method may have a profound impact on the interpretation of genomic assays. Taken together, these data highlight the need for improved assessment of tumor purity and quantitation of its influences on the molecular hallmarks of cancers.
Collapse
Affiliation(s)
- Syed Haider
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada,The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom,Syed Haider, PhD, The Institute of Cancer Research, 237 Fulham Rd, London, United Kingdom; Twitter: @theboutroslab, @UCLAJCCC; e-mail:
| | - Svitlana Tyekucheva
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Davide Prandi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Natalie S. Fox
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, DC
| | - Andrew Wei Xu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | | | - Peter J. Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | | | - Chris Sander
- cBio Center, Dana-Farber Cancer Institute, Boston, MA,Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Wenyi Wang
- The University of Texas MD Anderson Cancer Center Department of Bioinformatics and Computational Biology, Houston
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy,Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY
| | - Massimo Loda
- Department of Pathology, Weill Medical College of Cornell University, New York, NY,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA,Department of Urology, University of California, Los Angeles, CA,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA,Institute for Precision Health, University of California, Los Angeles, CA
| | | |
Collapse
|
77
|
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
|
78
|
Kato M, Hirakawa A, Kobayashi Y, Yamamoto A, Naito Y, Tochigi K, Sano T, Ishida S, Funahashi Y, Fujita T, Matsukawa Y, Hattori R, Tsuzuki T. Effect of core needle biopsy number on intraductal carcinoma of the prostate (IDC-P) diagnosis in patients with metastatic hormone-sensitive prostate cancer. Int J Clin Oncol 2020; 25:2130-2137. [PMID: 32748295 DOI: 10.1007/s10147-020-01756-0] [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: 05/31/2020] [Accepted: 07/21/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND The number of core needle biopsies in metastatic prostate cancer cases are sometimes reduced to avoid various complications. We analyzed whether core needle biopsy number influence IDC-P detection rate in patients with metastatic castration-sensitive prostate cancer (mHSPC). METHODS We retrospectively evaluated data from 150 patients diagnosed with mHSPC. Subjects were allocated to three groups according to the number of core biopsies performed: ≤ 5, 6-9, and ≥ 10. The study endpoints were the cancer-specific survival (CSS) and overall survival (OS) rates. RESULTS For patients who underwent ≥ 10 core biopsies, a significant difference on CSS was detected between with or without IDC-P (P = 0.016). On the other hand, the difference decreased as the number of core biopsies became smaller (6-9; P = 0.322 and ≤ 5; P = 0.815). A similar trend was identified for the OS outcome. A significant difference on OS was also found between with or without IDC-P in patients who underwent ≥ 10 and 6-9 core needle biopsies (P = 0.0002 and 0.017, respectively), but not in those who underwent ≤ 5 core biopsies (P = 0.341). IDC-P served as a stronger prognostic marker for CSS and OS than did the other factors included in the multivariate analysis for patients had ≥ 10 core biopsies (P = 0.016, and P = 0.0014, respectively). CONCLUSIONS Given the IDC-P detection and its value as a prognostic marker, we propose the performance of ≥ 10 core biopsy procedures in patients diagnosed with mHSPC to minimize the sampling error of the IDC-P.
Collapse
Affiliation(s)
- Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Akihiro Hirakawa
- Division of Biostatistics and Data Science, Clinical Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yumiko Kobayashi
- Statistical Analysis Section, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Akiyuki Yamamoto
- Department of Urology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kosuke Tochigi
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shohei Ishida
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yasuhito Funahashi
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Takashi Fujita
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, 65 Tsumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ryohei Hattori
- Department of Urology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| |
Collapse
|
79
|
Reporting Practices and Resource Utilization in the Era of Intraductal Carcinoma of the Prostate: A Survey of Genitourinary Subspecialists. Am J Surg Pathol 2020; 44:673-680. [PMID: 31876580 DOI: 10.1097/pas.0000000000001417] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Intraductal carcinoma of the prostate (IDC-P) has been recently recognized by the World Health Organization classification of prostatic tumors as a distinct entity, most often occurring concurrently with invasive prostatic adenocarcinoma (PCa). Whether documented admixed with PCa or in its rare pure form, numerous studies associate this entity with clinical aggressiveness. Despite increasing clinical experience and requirement of IDC-P documentation in protocols for synoptic reporting, the specifics of its potential contribution to assessment of grade group (GG) and cancer quantitation of PCa in both needle biopsies (NBx) and radical prostatectomy (RP) specimens remain unclear. Moreover, there are no standard guidelines for incorporating basal cell marker immunohistochemistry (IHC) in the diagnosis of IDC-P, either alone or as part of a cocktail with AMACR/racemase. An online survey containing 26 questions regarding diagnosis, reporting practices, and IHC resource utilization, focusing on IDC-P, was undertaken by 42 genitourinary subspecialists from 9 countries. The degree of agreement or disagreement regarding approaches to individual questions was classified as significant majority (>75%), majority (51% to 75%), minority (26% to 50%) and significant minority (≤25%). IDC-P with or without invasive cancer is considered a contraindication for active surveillance by the significant majority (95%) of respondents, although a majority (66%) also agreed that the clinical significance/behavior of IDC-P on NBx or RP with PCa required further study. The majority do not upgrade PCa based on comedonecrosis seen only in the intraductal component in NBx (62%) or RP (69%) specimens. Similarly, recognizable IDC-P with GG1 PCa was not a factor in upgrading in NBx (78%) or RP (71%) specimens. The majority (60%) of respondents include readily recognizable IDC-P in assessment of linear extent of PCa at NBx. A significant majority (78%) would use IHC to confirm or exclude intraductal carcinoma if other biopsies showed no PCa, while 60% would use it to confirm IDC-P with invasive PCa in NBx if it would change the overall GG assignment. Nearly half (48%, a minority) would use IHC to confirm IDC-P for accurate Gleason pattern 4 quantitation. A majority (57%) report the percentage of IDC-P when present, in RP specimens. When obvious Gleason pattern 4 or 5 PCa is present in RP or NBx, IHC is rarely to almost never used to confirm the presence of IDC-P by the significant majority (88% and 90%, respectively). Most genitourinary pathologists consider IDC-P to be an adverse prognostic feature independent of the PCa grade, although recommendations for standardization are needed to guide reporting of IDC-P vis a vis tumor quantitation and final GG assessment. The use of IHC varies widely and is performed for a multitude of indications, although it is used most frequently in scenarios where confirmation of IDC-P would impact the GG assigned. Further study and best practices recommendations are needed to provide guidance with regards to the most appropriate indications for IHC use in scenarios regarding IDC-P.
Collapse
|
80
|
Bicak M, Lückerath K, Kalidindi T, Phelps ME, Strand SE, Morris MJ, Radu CG, Damoiseaux R, Peltola MT, Peekhaus N, Ho A, Veach D, Malmborg Hager AC, Larson SM, Lilja H, McDevitt MR, Klein RJ, Ulmert D. Genetic signature of prostate cancer mouse models resistant to optimized hK2 targeted α-particle therapy. Proc Natl Acad Sci U S A 2020; 117:15172-15181. [PMID: 32532924 PMCID: PMC7334567 DOI: 10.1073/pnas.1918744117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hu11B6 is a monoclonal antibody that internalizes in cells expressing androgen receptor (AR)-regulated prostate-specific enzyme human kallikrein-related peptidase 2 (hK2; KLK2). In multiple rodent models, Actinium-225-labeled hu11B6-IgG1 ([225Ac]hu11B6-IgG1) has shown promising treatment efficacy. In the present study, we investigated options to enhance and optimize [225Ac]hu11B6 treatment. First, we evaluated the possibility of exploiting IgG3, the IgG subclass with superior activation of complement and ability to mediate FC-γ-receptor binding, for immunotherapeutically enhanced hK2 targeted α-radioimmunotherapy. Second, we compared the therapeutic efficacy of a single high activity vs. fractionated activity. Finally, we used RNA sequencing to analyze the genomic signatures of prostate cancer that progressed after targeted α-therapy. [225Ac]hu11B6-IgG3 was a functionally enhanced alternative to [225Ac]hu11B6-IgG1 but offered no improvement of therapeutic efficacy. Progression-free survival was slightly increased with a single high activity compared to fractionated activity. Tumor-free animals succumbing after treatment revealed no evidence of treatment-associated toxicity. In addition to up-regulation of canonical aggressive prostate cancer genes, such as MMP7, ETV1, NTS, and SCHLAP1, we also noted a significant decrease in both KLK3 (prostate-specific antigen ) and FOLH1 (prostate-specific membrane antigen) but not in AR and KLK2, demonstrating efficacy of sequential [225Ac]hu11B6 in a mouse model.
Collapse
Affiliation(s)
- Mesude Bicak
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genome Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Katharina Lückerath
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Teja Kalidindi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Michael E Phelps
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095;
| | - Sven-Erik Strand
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, 223 81 Lund, Sweden
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Caius G Radu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
| | - Mari T Peltola
- Department of Biochemistry-Biotechnology, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Norbert Peekhaus
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
| | - Austin Ho
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
| | - Darren Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Diaprost AB, 223 63 Lund, Sweden
| | | | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065
| | - Hans Lilja
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Translational Medicine, Lund University, 221 00 Lund, Sweden
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, OX3 7DQ Oxford, United Kingdom
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Michael R McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn Institute for Data Science and Genome Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029;
| | - David Ulmert
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095;
- Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095
| |
Collapse
|
81
|
Harland N, Stenzl A, Todenhöfer T. Role of Multiparametric Magnetic Resonance Imaging in Predicting Pathologic Outcomes in Prostate Cancer. World J Mens Health 2020; 39:38-47. [PMID: 32648376 PMCID: PMC7752518 DOI: 10.5534/wjmh.200030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/10/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
Multiparametric magnetic resonance imaging (mpMRI) and the introduction of standardized protocols for its interpretation have had a significant impact on the field of prostate cancer (PC). Multiple randomized controlled trials have shown that the sensitivity for detection of clinically significant PC is increased when mpMRI results are the basis for indication of a prostate biopsy. The added value with regards to sensitivity has been strongest for patients with persistent suspicion for PC after a prior negative biopsy. Although enhanced sensitivity of mpMRI is convincing, studies that have compared mpMRI with prostatectomy specimens prepared by whole-mount section analysis have shown a significant number of lesions that were not detected by mpMRI. In this context, the importance of an additional systematic biopsy (SB) is still being debated. While SB in combination with targeted biopsies leads to an increased detection rate, most of the tumors detected by SB only are considered clinically insignificant. Currently, multiple risk calculation tools are being developed that include not only clinical parameters but mpMRI results in addition to clinical parameters in order to improve risk stratification for PC, such as the Partin tables. In summary, mpMRI of the prostate has become a standard procedure recommended by multiple important guidelines for the diagnostic work-up of patients with suspicion of PC.
Collapse
Affiliation(s)
- Niklas Harland
- Department of Urology, University Hospital Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tübingen, Germany.,Medical School, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Tilman Todenhöfer
- Medical School, Eberhard-Karls-University Tübingen, Tübingen, Germany.,Clinical Trial Unit, Studienpraxis Urologie, Nürtingen, Germany.
| |
Collapse
|
82
|
Kishan AU, Romero T, Alshalalfa M, Liu Y, Tran PT, Nickols NG, Ye H, Sajed D, Rettig MB, Reiter RE, Garraway IP, Spratt DE, Freedland SJ, Zhao X, Li Z, Deek M, Livingstone J, Mahal BA, Nguyen PL, Feng FY, Den RB, Schaeffer EM, Lotan TL, Karnes RJ, Klein EA, Ross AE, Grogan T, Davicioni E, Elashoff D, Boutros PC, Weidhaas JB. Transcriptomic Heterogeneity of Gleason Grade Group 5 Prostate Cancer. Eur Urol 2020; 78:327-332. [PMID: 32461072 DOI: 10.1016/j.eururo.2020.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/10/2020] [Indexed: 01/01/2023]
Abstract
Gleason grade group (GG) 5 prostate cancer has been associated with an aggressive natural history, and retrospective data support a role for treatment intensification. However, clinical outcomes remain heterogeneous in this cohort, and intensified treatments carry an increased risk of adverse events. We sought to explore the transcriptomic heterogeneity of GG 5 tumors by querying transcriptomic data from the tumors of 2138 patients with GG 5 disease who underwent prostatectomy. Four distinct consensus clusters were identified with respect to differential transcriptional activation of hallmark pathways, with distinct molecular subtyping profiles and different average genomic risks (AGRs). One cluster, accounting for 325 tumors (15.2% of the population), was enriched for genes related to the cell cycle/proliferation, metabolic pathways, androgen response pathways, and DNA repair, and had a higher AGR than the other clusters (p < 0.001). This clustering, with an identification of a high genomic risk cluster, was subsequently validated in a separate cohort of 1921 patients as well as a third cohort of 201 patients. The latter cohort had outcomes available, and it was found that patients in the high genomic risk cluster had significantly worse distant metastasis-free survival than the other clusters. Tumors in this high genomic risk cluster of GG 5 disease may be particularly likely to benefit from treatment intensification. PATIENT SUMMARY: In this report, we examined differences in gene expression in tumors from men with Gleason grade group 5 prostate cancer. We identified significant diversity, with one specific subgroup of tumors associated with expression profiles that suggest a worse prognosis.
Collapse
Affiliation(s)
- Amar U Kishan
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA; Department of Urology, University of California, Los Angeles, CA, USA.
| | - Tahmineh Romero
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Mohammed Alshalalfa
- Department of Radiation Oncology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Yang Liu
- Decipher Biosciences, San Diego, CA, USA
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas G Nickols
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA; Department of Radiation Oncology, Veteran Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Huihui Ye
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Dipti Sajed
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Matthew B Rettig
- Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Division of Hematology and Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Robert E Reiter
- Department of Urology, University of California, Los Angeles, CA, USA
| | - Isla P Garraway
- Department of Urology, University of California, Los Angeles, CA, USA; Division of Urology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Steven J Freedland
- Department of Surgery, Division of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Surgery, Division of Urology, Veteran Affairs Healthcare System, Durham, NC, USA
| | - Xin Zhao
- Decipher Biosciences, San Diego, CA, USA
| | - Ziwen Li
- Decipher Biosciences, San Diego, CA, USA
| | - Matthew Deek
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie Livingstone
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| | - Brandon A Mahal
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul L Nguyen
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Felix Y Feng
- Department of Radiation Oncology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Robert B Den
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edward M Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tamara L Lotan
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Eric A Klein
- Glickman Urological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Tristan Grogan
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | - David Elashoff
- Statistics Core, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Paul C Boutros
- Department of Urology, University of California, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Joanne B Weidhaas
- Department of Radiation Oncology, University of California, Los Angeles, CA, USA
| |
Collapse
|
83
|
Evolution of prostate cancer histopathology. Curr Opin Urol 2020; 29:587-592. [PMID: 31453865 DOI: 10.1097/mou.0000000000000669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW During the last 15 years several updates in the Gleason grading have been made. With the help of pertinent research results pathologists have gained a better insight into the meanings of several prostate cancer (PCa) patterns and know better how to classify them in the Gleason grade system. RECENT FINDINGS During the last years PCa with cribriform architecture has be given much attention. Many data have also been published about the meaning of comedonecrosis and its relationship with Gleason pattern 4 and 5. The correlationship between comedonecrosis and intraductal PCa has also been highlighted in the recent literature. Intraductal PCa is one of the most described topics at the moment with implications to treatment such as radiation therapy. We also highlight several practical issues such as the differences of grading in prostate biopsies and prostatectomies and describe the problematic of reporting a minor high-grade pattern. SUMMARY Many new and recent data have allowed to refine diagnosis in PCa and improve the patients's treatment. We show that comedonecrosis can be overgraded and insist on the implication with cribriform and intraducatal carcinomas. Furthermore, we describe the importance of these PCa types especially in the consideration of further treatment.
Collapse
|
84
|
Rebello RJ, Bristow RG. Intermediate Risk Prostate Cancer: Disease Heterogeneity Linked to Measurable Biological Features. Clin Oncol (R Coll Radiol) 2020; 32:298-302. [PMID: 32147317 DOI: 10.1016/j.clon.2019.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/20/2019] [Indexed: 11/26/2022]
Affiliation(s)
- R J Rebello
- Translational Oncogenomics Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| | - R G Bristow
- Translational Oncogenomics Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester Cancer Research Centre, Manchester, UK.
| |
Collapse
|
85
|
Abstract
Intraductal carcinoma of the prostate (IDC-P) is a diagnostic entity characterized by architecturally or cytologically malignant-appearing prostatic glandular epithelium confined to prostatic ducts. Despite its apparent in situ nature, this lesion is associated with aggressive prostatic adenocarcinoma and is a predictor for poor prognosis when identified on biopsy or radical prostatectomy. This review discusses diagnosis, clinical features, histogenesis, and management of IDC-P, as well as current research and controversies surrounding this entity.
Collapse
|
86
|
Haffner MC, Salles DC, Gao G, Epstein JI. Gleason pattern 4 with cribriform morphology on biopsy is associated with adverse clinicopathological findings in a prospective radical prostatectomy cohort. Hum Pathol 2020; 98:74-80. [PMID: 32119879 DOI: 10.1016/j.humpath.2020.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/24/2020] [Indexed: 11/28/2022]
Abstract
The prognostic significance of the Gleason grading system has been well established. However, individual Gleason patterns comprise heterogeneous morphologies which might add additional prognostic information. Recent evidence suggests that Gleason pattern 4 with cribriform growth pattern is associated with an adverse prognosis. To determine the association between cribriform pattern on biopsies and pathological findings on subsequent prostatectomies, we evaluated the presence of cribriform architecture in a prospective cohort of 367 men from 2014 to 2018 treated at a single institution. Cribriform architecture was present in 63.5% of all biopsies and was correlated with the overall extent of Gleason pattern 4. In addition, cribriform morphology on biopsy showed a statistically significant association with higher Gleason grade and increased pathological stage and nodal metastasis. In a subset analysis of cases with Grade Group 2 (Gleason score 3 + 4, n = 208), these associations did not reach statistical significance, but the presence of cribriform growth in this subgroup showed a trend toward increased upgrading to Grade Group 5 (Gleason score 9/10) (1 [0.5%] vs. 5 [2.4%], P = 0.06). This large prospective study comparing biopsy and prostatectomy finding of cribriform architecture demonstrates that cribriform pattern 4 is associated with adverse prognostic features and highlights the relevance for recognizing specific morphologies with distinct biological and clinical features.
Collapse
Affiliation(s)
- Michael C Haffner
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Daniela C Salles
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Guofeng Gao
- Department of Pathology, University of California Davis Health System, Sacramento, CA 92093, USA
| | - Jonathan I Epstein
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
87
|
Kato M, Hirakawa A, Kobayashi Y, Yamamoto A, Ishida R, Kamihira O, Sano T, Majima T, Ishida S, Funahashi Y, Sassa N, Fujita T, Matsukawa Y, Hattori R, Gotoh M, Tsuzuki T. Response of intraductal carcinoma of the prostate to androgen deprivation therapy predicts prostate cancer prognosis in radical prostatectomy patients. Prostate 2020; 80:284-290. [PMID: 31860754 DOI: 10.1002/pros.23942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Intraductal carcinoma of the prostate (IDC-P) has a poor prognosis and is thought to be completely resistant to current therapies, including androgen deprivation therapy (ADT). However, to date, there are no data showing direct evidence of such resistance. METHODS We retrospectively evaluated 145 patients with high-risk prostate cancer who underwent radical prostatectomy (RP) with neoadjuvant ADT between 1991 and 2005. All patient data were collected from slides prepared from needle biopsy (NB) samples of prostate tissue and RP specimens. Data were analyzed in terms of serum level of prostate specific antigen (PSA), Gleason score of NB samples, clinical T stage, the positive cancer core rate, maximum cancer extension rate, presence of Gleason pattern 5, and presence of IDC-P in both NB samples and RP specimens. RESULTS The median initial PSA was 33.2 ng/mL (range, 2.4-296 ng/mL), and the median follow-up period was 109 months (range, 11-257 months). The preoperative median ADT period was 4 months (range, 1-20 months). IDC-P was present in 53 patients (37%) in NB samples and 65 (45%) in RP. The patients were divided into three groups based on the presence or absence of IDC-P in NB/RP samples (IDC-P-negative at biopsy: 92 cases, IDC-P-positive at biopsy with IDC-P disappearance: 15 cases, and IDC-P-positive at biopsy with IDC-P persistence: 38 cases). Overall, 28% of IDC-P-positive cases in NB samples showed the disappearance of IDC-P at RP. IDC-P persistence cases showed the poorest prognosis, while IDC-P disappearance cases had a similar prognosis to that of IDC-P-negative at biopsy cases in terms of disease-free survival, cancer-specific survival, and overall survival (P = .0018, P = .0087, and P = .0034, respectively). CONCLUSIONS Some cases with IDC-P responded to ADT and demonstrated favorable clinical outcomes similar to those of cases without IDC-P. These findings indicate that cases with IDC-P are heterogeneous.
Collapse
Affiliation(s)
- Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Hirakawa
- Department of Biostatistics and Bioinformatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumiko Kobayashi
- Statistical Analysis Section, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Akiyuki Yamamoto
- Department of Urology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Ryo Ishida
- Department of Urology, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Osamu Kamihira
- Department of Urology, Komaki City Hospital, Komaki, Japan
| | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyoshi Majima
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shohei Ishida
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhito Funahashi
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoto Sassa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Fujita
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryohei Hattori
- Department of Urology, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
88
|
Merging new-age biomarkers and nanodiagnostics for precision prostate cancer management. Nat Rev Urol 2020; 16:302-317. [PMID: 30962568 DOI: 10.1038/s41585-019-0178-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The accurate identification and stratified treatment of clinically significant early-stage prostate cancer have been ongoing concerns since the outcomes of large international prostate cancer screening trials were reported. The controversy surrounding clinical and cost benefits of prostate cancer screening has highlighted the lack of strategies for discriminating high-risk disease (that requires early treatment) from low-risk disease (that could be managed using watchful waiting or active surveillance). Advances in molecular subtyping and multiomics nanotechnology-based prostate cancer risk delineation can enable refinement of prostate cancer molecular taxonomy into clinically meaningful and treatable subtypes. Furthermore, the presence of intertumoural and intratumoural heterogeneity in prostate cancer warrants the development of novel nanodiagnostic technologies to identify clinically significant prostate cancer in a rapid, cost-effective and accurate manner. Circulating and urinary next-generation prostate cancer biomarkers for disease molecular subtyping and the newest complementary nanodiagnostic platforms for enhanced biomarker detection are promising tools for precision prostate cancer management. However, challenges in merging both aspects and clinical translation still need to be overcome.
Collapse
|
89
|
Abstract
Long non-coding RNAs (lncRNAs) are regulators of cellular machinery that are commonly dysregulated in genitourinary malignancies. Accordingly, the investigation of lncRNAs is improving our understanding of genitourinary cancers, from development to progression and dissemination. lncRNAs are involved in major oncogenic events in genitourinary malignancies, including androgen receptor (AR) signalling in prostate cancer, hypoxia-inducible factor (HIF) pathway activation in renal cell carcinoma and invasiveness in bladder cancer, as well as multiple other proliferation and survival mechanisms. In line with their putative oncogenic roles, new lncRNA-based classifications are emerging as potent predictors of prognosis. In clinical practice, detection of oncogenic lncRNAs in serum or urine might enable early cancer detection, and lncRNAs might also be promising therapeutic targets for patients with genitourinary cancer. Furthermore, as predictors of sensitivity to anticancer treatments, lncRNAs could be integrated into future precision medicine strategies. Overall, lncRNAs are promising new candidates for molecular studies and for discovery of innovative biomarkers and are putative therapeutic targets in genitourinary oncology.
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW This review summarizes recent advances in prostate cancer (PCa) genetics. RECENT FINDINGS Upwards of 20% of metastatic castration-resistant prostate tumors (mCRPC) carry homologous recombination (HR) repair gene mutations, of which ~ 10% are germline (inherited). Another ~ 5% exhibit microsatellite instability (MSI-H) and/or mismatch repair deficiency (MMRd). Pembrolizumab is approved for tumors with MMRd, thus patients with mCRPC and MMRd are candidates for pembrolizumab. Emerging data indicate that platinum chemotherapy and poly ADP-ribose polymerase inhibitors (PARPi) are effective in PCa exhibiting HR deficiency. NCCN guidelines now recommend germline and somatic tumor testing in specific clinical scenarios due to treatment and family implications. Genetic testing in PCa patients may inform prognosis, treatment options, and have implications for family counseling. PARPi, platinum chemotherapy, and immune checkpoint inhibitors are promising targeted therapies for PCa with specific molecular features. Therapeutic advances, along with importance to relatives, are driving genetic testing in prostate cancer.
Collapse
Affiliation(s)
- Alexandra O Sokolova
- Department of Medicine, University of Washington, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 825 Eastlake Ave E, Seattle, WA, 98109, USA
- VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA, 98108, USA
| | - Heather H Cheng
- Department of Medicine, University of Washington, Washington, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 825 Eastlake Ave E, Seattle, WA, 98109, USA.
| |
Collapse
|
91
|
The Genomic and Molecular Pathology of Prostate Cancer: Clinical Implications for Diagnosis, Prognosis, and Therapy. Adv Anat Pathol 2020; 27:11-19. [PMID: 31503032 DOI: 10.1097/pap.0000000000000245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Prostate cancer (PCa) is the most common noncutaneous malignancy affecting American men and the second most common cause of cancer death. The traditional risk classification schemes for PCa are limited due to the vast clinical and molecular heterogeneity of the disease. Fortunately, recent advancements in sequencing technologies have provided us with valuable insight into the genomics of PCa. To date, a wide array of recurrent genomic alterations in PCa have been identified. Incorporating these distinct molecular subtypes of PCa into prediction models provides opportunities for improved risk stratification and ultimately better patient outcomes. In this review, we summarize the key molecular subtypes of PCa and focus on those genomic alterations that have clinical implications for diagnosis, prognosis, and therapeutic response.
Collapse
|
92
|
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
|
93
|
Hua JT, Chen S, He HH. Landscape of Noncoding RNA in Prostate Cancer. Trends Genet 2019; 35:840-851. [DOI: 10.1016/j.tig.2019.08.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/09/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022]
|
94
|
Cimadamore A, Scarpelli M, Raspollini MR, Doria A, Galosi AB, Massari F, Di Nunno V, Cheng L, Lopez-Beltran A, Montironi R. Prostate cancer pathology: What has changed in the last 5 years. Urologia 2019; 87:3-10. [PMID: 31545701 DOI: 10.1177/0391560319876821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent non-cutaneous malignancy in men in the United States. In the last few years, many recommendations have been made available from the 2014 International Society of Urologic Pathology consensus conference, 2016 World Health Organization blue book and 2018 8th edition of American Joint Committee on Cancer Staging System. Here, we focus on four topics which are considered relevant on the basis of their common appearance in routine practice, clinical importance and 'need to improve communication between pathology reports and clinicians': prostate cancer classification, prostate cancer grading, prostate cancer staging, and current definition of clinically significant prostate cancer. Tissue biomarkers that can predict significant disease and/or upgrading and tissue-based genomics for the purpose of diagnosis and prognosis are mentioned briefly.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, Marche Polytechnic University, School of Medicine, United Hospitals, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Marche Polytechnic University, School of Medicine, United Hospitals, Ancona, Italy
| | | | - Andrea Doria
- Section of Pathological Anatomy, Marche Polytechnic University, School of Medicine, United Hospitals, Ancona, Italy
| | - Andrea Benedetto Galosi
- Institute of Urology, Marche Polytechnic University, School of Medicine, United Hospitals, Ancona, Italy
| | - Francesco Massari
- Division of Oncology, Policlinico Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Vincenzo Di Nunno
- Division of Oncology, Policlinico Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, University of Cordoba, Cordoba, Spain
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Marche Polytechnic University, School of Medicine, United Hospitals, Ancona, Italy
| |
Collapse
|
95
|
Zeuschner P, Linxweiler J, Junker K. Non-coding RNAs as biomarkers in liquid biopsies with a special emphasis on extracellular vesicles in urological malignancies. Expert Rev Mol Diagn 2019; 20:151-167. [DOI: 10.1080/14737159.2019.1665998] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Philip Zeuschner
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| | - Johannes Linxweiler
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| |
Collapse
|
96
|
Truong M, Frye T, Messing E, Miyamoto H. Historical and contemporary perspectives on cribriform morphology in prostate cancer. Nat Rev Urol 2019; 15:475-482. [PMID: 29713007 DOI: 10.1038/s41585-018-0013-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Gleason scoring system is widely used for the grading and prognostication of prostate cancer. A Gleason pattern 4 subtype known as cribriform morphology has now been recognized as an aggressive and often lethal pattern of prostate cancer. The vast majority of published and ongoing prostate cancer studies still do not acknowledge the prognostic differences between various Gleason pattern 4 morphologies. As a result, current treatment recommendations are likely to be imprecise and not tailored towards patients who are most likely to die from the disease. Use of active surveillance for patients with Gleason score 3 + 4 prostate cancer has been suggested. However, the success of such paradigms would require cribriform morphology to be reported at the time of prostate biopsy, as patients harbouring such a pattern are poor candidates for surveillance. To date, only a limited number of studies have described the molecular alterations that occur in the cribriform morphological pattern. Further refinement of prostate cancer grading paradigms to distinguish cribriform from noncribriform Gleason pattern 4 is essential.
Collapse
Affiliation(s)
- Matthew Truong
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas Frye
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward Messing
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hiroshi Miyamoto
- Department of Urology, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
97
|
van Leenders GJLH, Kweldam CF, Hollemans E, Kümmerlin IP, Nieboer D, Verhoef EI, Remmers S, Incrocci L, Bangma CH, van der Kwast TH, Roobol MJ. Improved Prostate Cancer Biopsy Grading by Incorporation of Invasive Cribriform and Intraductal Carcinoma in the 2014 Grade Groups. Eur Urol 2019; 77:191-198. [PMID: 31439369 DOI: 10.1016/j.eururo.2019.07.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/31/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Grade groups (GGs) are an important parameter for therapeutic decision making in prostate cancer (Pca) patients. Invasive cribriform and/or intraductal carcinoma (CR/IDC) has an independent prognostic value for disease outcome, but are not included in the GG limiting their clinical use. OBJECTIVE To perform a proof-of-principle study incorporating CR/IDC in the current GG. DESIGN, SETTING, AND PARTICIPANTS All prostate biopsies of 1031 men with screen-detected Pca between 1993 and 2000 were reviewed for the current GG (ranging from 1 to 5) and CR/IDC. The cribriform grade (cGrade) was equal to the GG if CR/IDC was present and GG minus 1 if not. GG1 was cGrade1 if intraductal carcinoma was absent. INTERVENTION Biopsy review for GG and CR/IDC. A total of 406 patients had received radical prostatectomy (RP), 508 radiotherapy (RT), 108 surveillance, and eight hormonal therapy, and the treatment was unknown for one patient. Outcome measurements and statistical analysis disease-specific survival (DSS), metastasis-free survival (MFS), and biochemical recurrence-free survival (BCRFS) after 15.1 yr (interquartile range 10.9-19.7 yr) follow-up were compared using Harrell's C-statistic. RESULTS AND LIMITATIONS The biopsy GGs were 486 GG1, 310 GG2, 104 GG3, 64 GG4, and 67 GG5; cGrade distributions were 738 cGrade1, 102 cGrade2, 91 cGrade3, 58 cGrade4, and 42 cGrade5. The cGrade had a better discriminative value than the GG for DSS (C-index 0.79; 95% confidence interval 0.74-0.83 vs 0.76; 0.71-0.82) and MFS (0.79; 0.74-0.84 vs 0.77; 0.72-0.82). The discriminative value for BCRFS after RP and RT was similar for both models. Different diagnostic, such as use of sextant biopsies, and therapeutic strategies in the 1990s are the limitations of this study. CONCLUSIONS The cGrade is a simple Pca grade modification with better discriminative values for DSS and MFS than the GG, particularly impacting decision making in men with current GG2 Pca. PATIENT SUMMARY Microscopic grading is an important factor for decision making in prostate cancer (Pca) patients. We show that a simple grade modification better predicts Pca outcome and might improve treatment choices.
Collapse
Affiliation(s)
| | - Charlotte F Kweldam
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Eva Hollemans
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Intan P Kümmerlin
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Daan Nieboer
- Department of Urology, Erasmus University Medical Centre, Rotterdam, The Netherlands; Department of Public Health, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Sebastiaan Remmers
- Department of Urology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Luca Incrocci
- Department of Radiotherapy, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Chris H Bangma
- Department of Urology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Monique J Roobol
- Department of Urology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
98
|
Li X, Meng Y. Survival analysis of immune-related lncRNA in low-grade glioma. BMC Cancer 2019; 19:813. [PMID: 31419958 PMCID: PMC6697914 DOI: 10.1186/s12885-019-6032-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Low-grade glioma is grade I-II glioma. Immunotherapy is a promising way of tumor killing. Research on immune molecular mechanisms in low-grade gliomas and discovery of new immune checkpoints for low-grade gliomas are of great importance. METHODS Gene sequencing data and clinical data of low-grade glioma were downloaded from TCGA database. Prognosis related lncRNAs were identified by Cox regression and their possible functions were found by gene enrichment set analysis. RESULTS A total of 529 low-grade glioma samples and 5 non-tumor brain tissue samples are obtained from the TCGA database. Two hundred forty-seven immune-associated lncRNAs are screened. Cox regression showed that 16 immune-related lncRNAs are associated with low-grade glioma prognosis, and 7 lncRNAs are independent risk factors. Gene set enrichment analysis suggests that these molecules are enriched in extracellular region, sequence-specific DNA binding, neuropeptide signaling pathway, transcriptional misregulation in cancer, cytokine-cytokine receptor interaction, protein digestion and absorption, chemokine signaling pathway, etc. CONCLUSION: The identification of immune-related lncRNA may provide new targets for the research of the molecular mechanisms and treatment of low-grade glioma.
Collapse
Affiliation(s)
- Xiaozhi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutong Meng
- Department of Stomatology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning Province, 110004, People's Republic of China.
| |
Collapse
|
99
|
Shah RB, Shore KT, Yoon J, Mendrinos S, McKenney JK, Tian W. PTEN loss in prostatic adenocarcinoma correlates with specific adverse histologic features (intraductal carcinoma, cribriform Gleason pattern 4 and stromogenic carcinoma). Prostate 2019; 79:1267-1273. [PMID: 31111513 DOI: 10.1002/pros.23831] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/29/2019] [Accepted: 04/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The loss of PTEN tumor suppressor gene is one of the most common somatic genetic aberrations in prostate cancer (PCa) and is frequently associated with high-risk disease. Deletion or mutation of at least one PTEN allele has been reported to occur in 20% to 40% of localized PCa and up to 60% of metastases. The goal of this study was to determine if somatic alteration detected by PTEN immunohistochemical loss of expression is associated with specific histologic features. METHODS Two hundred sixty prostate core needle biopsies with PCa were assessed for PTEN loss using an analytically validated immunohistochemical assay. Blinded to PTEN status, each tumor was assessed for the Grade Group (GG) and the presence or absence of nine epithelial features. Presence of stromogenic PCa was also assessed and defined as grade 3 reactive tumor stroma as previously described: the presence of carcinoma associated stromal response with epithelial to stroma ratio of greater than 50% reactive stroma. RESULTS Eight-eight (34%) cases exhibited PTEN loss while 172 (66%) had intact PTEN. PTEN loss was significantly (P < 0.05) associated with increasing GG, poorly formed glands (74% of total cases with loss vs 49% of intact), and three well-validated unfavorable pathological features: intraductal carcinoma of the prostate (IDC-P) (69% of total cases with loss vs 12% of intact), cribriform Gleason pattern 4 (38% of total cases with loss vs 10% of intact) and stromogenic PCa (23% of total cases with loss vs 6% of intact). IDC-P had the highest relative risk (4.993, 95% confidence interval, 3.451-7.223, P < 0.001) for PTEN loss. At least one of these three unfavorable pathological features were present in 67% of PCa exhibiting PTEN loss, while only 11% of PCa exhibited PTEN loss when none of these three unfavorable pathological features were present. CONCLUSIONS PCa with PTEN loss demonstrates a strong correlation with known unfavorable histologic features, particularly IDC-P. This is the first study showing the association of PTEN loss with stromogenic PCa.
Collapse
Affiliation(s)
- Rajal B Shah
- Division of Urologic Pathology, Inform Diagnostics, Irving, Texas
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Karen T Shore
- Weiss School of Natural Sciences, Rice University, Houston, Texas
| | - Jiyoon Yoon
- Division of Urologic Pathology, Inform Diagnostics, Irving, Texas
| | - Savvas Mendrinos
- Division of Urologic Pathology, Inform Diagnostics, Irving, Texas
| | - Jesse K McKenney
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, Ohio
| | - Wei Tian
- Division of Urologic Pathology, Inform Diagnostics, Irving, Texas
| |
Collapse
|
100
|
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications. MEDICINES (BASEL, SWITZERLAND) 2019; 6:E82. [PMID: 31366128 PMCID: PMC6789661 DOI: 10.3390/medicines6030082] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most frequent nonskin cancer and second most common cause of cancer-related deaths in man. Prostate cancer is a clinically heterogeneous disease with many patients exhibiting an aggressive disease with progression, metastasis, and other patients showing an indolent disease with low tendency to progression. Three stages of development of human prostate tumors have been identified: intraepithelial neoplasia, adenocarcinoma androgen-dependent, and adenocarcinoma androgen-independent or castration-resistant. Advances in molecular technologies have provided a very rapid progress in our understanding of the genomic events responsible for the initial development and progression of prostate cancer. These studies have shown that prostate cancer genome displays a relatively low mutation rate compared with other cancers and few chromosomal loss or gains. The ensemble of these molecular studies has led to suggest the existence of two main molecular groups of prostate cancers: one characterized by the presence of ERG rearrangements (~50% of prostate cancers harbor recurrent gene fusions involving ETS transcription factors, fusing the 5' untranslated region of the androgen-regulated gene TMPRSS2 to nearly the coding sequence of the ETS family transcription factor ERG) and features of chemoplexy (complex gene rearrangements developing from a coordinated and simultaneous molecular event), and a second one characterized by the absence of ERG rearrangements and by the frequent mutations in the E3 ubiquitin ligase adapter SPOP and/or deletion of CDH1, a chromatin remodeling factor, and interchromosomal rearrangements and SPOP mutations are early events during prostate cancer development. During disease progression, genomic and epigenomic abnormalities accrued and converged on prostate cancer pathways, leading to a highly heterogeneous transcriptomic landscape, characterized by a hyperactive androgen receptor signaling axis.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|