1
|
Beauchamp L, Indulkar S, Erak E, Salimian M, Matoso A. Tissue-Based Biomarkers Important for Prognostication and Management of Genitourinary Tumors, Including Surrogate Markers of Genomic Alterations. Surg Pathol Clin 2025; 18:175-189. [PMID: 39890303 DOI: 10.1016/j.path.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
A better understanding of the molecular alterations that underlie urologic malignancies and advances in targeted therapies has impacted classification, prognostication, and treatment. In bladder tumors, these advances include the development of antibody-drug conjugates targeting nectin-4 and Trop-2, as well as human epidermal growth factor receptor 2 and immunotherapy. In prostate cancer, assessment of the percentage of Gleason pattern 4, presence of cribriform glands, and molecular alterations, including PTEN and mismatch repair protein loss, have become standard for clinical care. In renal malignancies, alterations in TSC1/2, mammalian target of rapamycin, anaplastic lymphoma kinase, and other genes impact classification and therapeutic decisions.
Collapse
Affiliation(s)
- Leonie Beauchamp
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Shreeya Indulkar
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Eric Erak
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | - Andres Matoso
- Departments of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| |
Collapse
|
2
|
Potterveld SK, Williamson SR, Al-Obaidy KI, Akgul M, Chan E, Giannico GA, Sangoi AR. GATA3 Expression in Prostatic Adenosquamous Carcinoma: A Potential Diagnostic Pitfall. Int J Surg Pathol 2025; 33:85-91. [PMID: 38562047 DOI: 10.1177/10668969241241640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Urothelial carcinoma and prostatic adenocarcinoma can have overlapping histologic features and in some instances pose challenges to pathologists. GATA binding protein 3 (GATA3) immunohistochemistry (IHC) is a well-established tool to aid in this specific diagnostic dilemma as it has been shown to be a sensitive marker for urothelial carcinoma and a putatively specific marker in excluding prostatic adenocarcinoma. However, in encountering an index tumor of prostatic adenosquamous carcinoma positive for GATA3, herein we sought to investigate this potential diagnostic pitfall in a larger series of tumors. In this study, we retrospectively reviewed prostatic adenosquamous carcinomas diagnosed in 17 patients across the authors' institutions and personal consult collections in the past 10 years. GATA3 IHC was either reviewed or performed on tumors not previously tested. We also recorded other immunostains that were performed at initial diagnosis. Positivity for GATA3 was found in 9 of 17 (53%) tumors, all within squamous regions (2 tumors also showed concomitant moderate GATA3 positivity within glandular elements). The GATA3 positive tumors were all positive for p63 in the 7 tumors where p63 was also performed. Of all tumors tested, NKX3.1 was positive in 100% (13/13) of the glandular elements (3 tumors also showed NKX3.1 concomitant positivity within squamous regions). In summary, when encountering a carcinoma with mixed glandular/squamous features in which prostatic origin is being considered, awareness of GATA3 immunoreactivity in a subset of prostatic adenosquamous carcinoma is critical to avoid diagnostic pitfalls.
Collapse
Affiliation(s)
| | | | | | - Mahmut Akgul
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Emily Chan
- Department of Pathology, Stanford Medical Center, Stanford, CA, USA
| | | | - Ankur R Sangoi
- Department of Pathology, Stanford Medical Center, Stanford, CA, USA
| |
Collapse
|
3
|
Lee E, Oliveira LD, Dairo O, Nourmohammadi Abadchi S, Cha E, Mendes AA, Wang JH, Song DY, Lotan TL. PTEN Loss Is Associated with Adverse Outcomes in the Setting of Salvage Radiation Therapy. Eur Urol Oncol 2024; 7:1513-1519. [PMID: 38964997 DOI: 10.1016/j.euo.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Salvage radiation therapy (SRT) is a mainstay of treatment for biochemical relapse following radical prostatectomy; however, few studies have examined genomic biomarkers in this context. OBJECTIVE We characterized the prognostic impact of previously identified deleterious molecular phenotypes-loss of PTEN, ERG expression, and TP53 mutation-for patients undergoing SRT. DESIGN, SETTING, AND PARTICIPANTS We leveraged an institutional database of 320 SRT patients with available tissue and follow-up. Tissue microarrays were used for genetically validated immunohistochemistry assays. INTERVENTION All men underwent SRT with or without androgen deprivation therapy OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Univariable and multivariable Cox-proportional hazard models assessed the association of molecular phenotypes with biochemical recurrence-free (bRFS) and metastasis-free (MFS) survival after SRT. RESULTS AND LIMITATIONS Loss of PTEN (n = 123, 43%) and ERG expression (n = 118, 39%) were common in this cohort, while p53 overexpression (signifying TP53 missense mutation) was infrequent (n = 21, 7%). In univariable analyses, any loss of PTEN portended worse bRFS (hazard ratio [HR] 1.86; 95% confidence interval 1.36-2.57) and MFS (HR 1.89; 1.21-2.94), with homogeneous PTEN loss being associated with the highest risk of MFS (HR 2.47; 1.54-3.95). Similarly, p53 overexpression predicted worse bRFS (HR 1.95; 1.14-3.32) and MFS (HR 2.79; 1.50-5.19). ERG expression was associated with worse MFS only (HR 1.6; 1.03-2.48). On the multivariable analysis adjusting for known prognostic features, homogeneous PTEN loss remained predictive of adverse bRFS (HR 1.82; 1.12-2.96) and MFS (HR 2.08; 1.06-4.86). The study is limited by its retrospective and single-institution design. CONCLUSIONS PTEN loss by immunohistochemistry is an independent adverse prognostic factor for bRFS and MFS in prostate cancer patients treated with SRT. Future trials will determine the optimal approach to treating SRT patients with adverse molecular prognostic features. PATIENT SUMMARY Loss of the PTEN tumor suppressor protein is associated with worse outcomes after salvage radiotherapy, independent of other clinical or pathologic patient characteristics.
Collapse
Affiliation(s)
- Emerson Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lia DePaula Oliveira
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Oluwademilade Dairo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Eumee Cha
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jarey H Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Y Song
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Urology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Teoman G, Livaoglu A, Kucuk H, Murtezaoglu AR. The importance of histomorphological features and ERG expression in the diagnosis of malignancy in cases with atypical small acinar proliferation. J Pathol Transl Med 2024; 58:134-140. [PMID: 38766739 PMCID: PMC11106607 DOI: 10.4132/jptm.2024.03.18] [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: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Atypical small acinar proliferation (ASAP) cases typically require rebiopsy, which are invasive and associated with increased risk of complications. Our aim in this study was to determine the importance of laboratory and histological findings and E-26 transformation-specific-related gene (ERG) expression in the diagnosis of malignancy. METHODS Between March 2016 and March 2022, 84 patients who were diagnosed with ASAP on biopsy or rebiopsy were included in the study. Clinical-laboratory features of age, serum prostate-specific antigen level, and histopathological features were compared and included multifocality, number of suspicious acini, nuclear enlargement, nucleolar prominence, hyperchromasia, cytoplasmic amphophilia, luminal amorphous acellular secretion, crystalloid presence, infiltrative appearance, inflammation, atrophy, α-methyl acyl-CoA racemase, p63, and/or high molecular weight cytokeratin were analyzed. In addition, ERG expression was evaluated immunohistochemically. RESULTS Statistically significant correlation was found between nucleolar prominence, nuclear hyperchromasia, crystalloid presence, infiltrative pattern, and prostate cancer (p < .001). In 19 of 84 cases (22.6%) ERG was positive in the nucleus. Prostate cancer was diagnosed at rebiopsy in 15 of the 19 ERG-positive cases (78.9%). A statistically significant correlation was found between ERG positivity and prostate cancer (p= .002). CONCLUSIONS Our findings suggest that evaluation of these markers during initial transrectal ultrasound biopsies may decrease and prevent unnecessary prostate rebiopsy.
Collapse
Affiliation(s)
- Gizem Teoman
- Department of Pathology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Türkiye
| | - Ayten Livaoglu
- Department of Pathology, Kanuni Training and Research Hospital, Health Sciences University, Trabzon, Türkiye
| | - Hatice Kucuk
- Department of Pathology, Kanuni Training and Research Hospital, Health Sciences University, Trabzon, Türkiye
| | - Afşın Rahman Murtezaoglu
- Department of Pathology, Kanuni Training and Research Hospital, Health Sciences University, Trabzon, Türkiye
| |
Collapse
|
5
|
Omar M, Xu Z, Rand SB, Alexanderani MK, Salles DC, Valencia I, Schaeffer EM, Robinson BD, Lotan TL, Loda M, Marchionni L. Semi-Supervised, Attention-Based Deep Learning for Predicting TMPRSS2:ERG Fusion Status in Prostate Cancer Using Whole Slide Images. Mol Cancer Res 2024; 22:347-359. [PMID: 38284821 PMCID: PMC10985477 DOI: 10.1158/1541-7786.mcr-23-0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/26/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
IMPLICATIONS Our study illuminates the potential of deep learning in effectively inferring key prostate cancer genetic alterations from the tissue morphology depicted in routinely available histology slides, offering a cost-effective method that could revolutionize diagnostic strategies in oncology.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhuoran Xu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sophie B. Rand
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Daniela C. Salles
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
6
|
Li Y, Su H, Liu K, Zhao Z, Wang Y, Chen B, Xia J, Yuan H, Huang DS, Gu Y. Individualized detection of TMPRSS2-ERG fusion status in prostate cancer: a rank-based qualitative transcriptome signature. World J Surg Oncol 2024; 22:49. [PMID: 38331878 PMCID: PMC10854045 DOI: 10.1186/s12957-024-03314-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/13/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND TMPRSS2-ERG (T2E) fusion is highly related to aggressive clinical features in prostate cancer (PC), which guides individual therapy. However, current fusion prediction tools lacked enough accuracy and biomarkers were unable to be applied to individuals across different platforms due to their quantitative nature. This study aims to identify a transcriptome signature to detect the T2E fusion status of PC at the individual level. METHODS Based on 272 high-throughput mRNA expression profiles from the Sboner dataset, we developed a rank-based algorithm to identify a qualitative signature to detect T2E fusion in PC. The signature was validated in 1223 samples from three external datasets (Setlur, Clarissa, and TCGA). RESULTS A signature, composed of five mRNAs coupled to ERG (five ERG-mRNA pairs, 5-ERG-mRPs), was developed to distinguish T2E fusion status in PC. 5-ERG-mRPs reached 84.56% accuracy in Sboner dataset, which was verified in Setlur dataset (n = 455, accuracy = 82.20%) and Clarissa dataset (n = 118, accuracy = 81.36%). Besides, for 495 samples from TCGA, two subtypes classified by 5-ERG-mRPs showed a higher level of significance in various T2E fusion features than subtypes obtained through current fusion prediction tools, such as STAR-Fusion. CONCLUSIONS Overall, 5-ERG-mRPs can robustly detect T2E fusion in PC at the individual level, which can be used on any gene measurement platform without specific normalization procedures. Hence, 5-ERG-mRPs may serve as an auxiliary tool for PC patient management.
Collapse
Affiliation(s)
- Yawei Li
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Hang Su
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Kaidong Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhangxiang Zhao
- The Sino-Russian Medical Research Center of Jinan University, The Institute of Chronic Disease of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuquan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bo Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jie Xia
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - Huating Yuan
- School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China
| | - De-Shuang Huang
- Bioinformatics and BioMedical Bigdata Mining Laboratory, School of Big Health, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Yunyan Gu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
7
|
Dairo O, DePaula Oliveira L, Schaffer E, Vidotto T, Mendes AA, Lu J, Huynh SV, Hicks J, Sowalsky AG, De Marzo AM, Joshu CE, Hanratty B, Sfanos KS, Isaacs WB, Haffner MC, Lotan TL. FASN Gene Methylation is Associated with Fatty Acid Synthase Expression and Clinical-genomic Features of Prostate Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:152-163. [PMID: 38112617 PMCID: PMC10795515 DOI: 10.1158/2767-9764.crc-23-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/05/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Fatty acid synthase (FASN) catalyzes the synthesis of long-chain saturated fatty acids and is overexpressed during prostatic tumorigenesis, where it is the therapeutic target in several ongoing trials. However, the mechanism of FASN upregulation in prostate cancer remains unclear. Here, we examine FASN gene CpG methylation pattern by InfiniumEPIC profiling and whole-genome bisulfite sequencing across multiple racially diverse primary and metastatic prostate cancer cohorts, comparing with FASN protein expression as measured by digitally quantified IHC assay and reverse phase protein array analysis or FASN gene expression. We demonstrate that the FASN gene body is hypomethylated and overexpressed in primary prostate tumors compared with benign tissue, and FASN gene methylation is significantly inversely correlated with FASN protein or gene expression in both primary and metastatic prostate cancer. Primary prostate tumors with ERG gene rearrangement have increased FASN expression and we find evidence of FASN hypomethylation in this context. FASN expression is also significantly increased in prostate tumors from carriers of the germline HOXB13 G84E mutation compared with matched controls, consistent with a report that HOXB13 may contribute to epigenetic regulation of FASN in vitro. However, in contrast to previous studies, we find no significant association of FASN expression or methylation with self-identified race in models that include ERG status across two independent primary tumor cohorts. Taken together, these data support a potential epigenetic mechanism for FASN regulation in the prostate which may be relevant for selecting patients responsive to FASN inhibitors. SIGNIFICANCE Here, we leverage multiple independent primary and metastatic prostate cancer cohorts to demonstrate that FASN gene body methylation is highly inversely correlated with FASN gene and protein expression. This finding may shed light on epigenetic mechanisms of FASN regulation in prostate cancer and provides a potentially useful biomarker for selecting patients in future trials of FASN inhibitors.
Collapse
Affiliation(s)
- Oluwademilade Dairo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Ethan Schaffer
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Thiago Vidotto
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Adrianna A. Mendes
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sophie Vo Huynh
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jessica Hicks
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, NCI, Bethesda, Maryland
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Corrine E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Brian Hanratty
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Karen S. Sfanos
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - William B. Isaacs
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michael C. Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Li J, Wilkerson ML, Deng FM, Liu H. The Application and Pitfalls of Immunohistochemical Markers in Challenging Diagnosis of Genitourinary Pathology. Arch Pathol Lab Med 2024; 148:13-32. [PMID: 37074862 DOI: 10.5858/arpa.2022-0493-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2023] [Indexed: 04/20/2023]
Abstract
CONTEXT.— The morphologic features of different entities in genitourinary pathology overlap, presenting a diagnostic challenge, especially when diagnostic materials are limited. Immunohistochemical markers are valuable when morphologic features alone are insufficient for definitive diagnosis. The World Health Organization classification of urinary and male genital tumors has been updated for 2022. An updated review of immunohistochemical markers for newly classified genitourinary neoplasms and their differential diagnosis is needed. OBJECTIVE.— To review immunohistochemical markers used in the diagnosis of genitourinary lesions in the kidney, bladder, prostate, and testis. We particularly emphasized difficult differential diagnosis and pitfalls in immunohistochemistry application and interpretation. New markers and new entities in the 2022 World Health Organization classifications of genitourinary tumors are reviewed. Recommended staining panels for commonly encountered difficult differential diagnoses and potential pitfalls are discussed. DATA SOURCES.— Review of current literature and our own experience. CONCLUSIONS.— Immunohistochemistry is a valuable tool in the diagnosis of problematic lesions of the genitourinary tract. However, the immunostains must be carefully interpreted in the context of morphologic findings with a thorough knowledge of pitfalls and limitations.
Collapse
Affiliation(s)
- Jianhong Li
- From the Department of Pathology, Geisinger Medical Center, Danville, Pennsylvania (Li, Wilkerson, Liu)
| | - Myra L Wilkerson
- From the Department of Pathology, Geisinger Medical Center, Danville, Pennsylvania (Li, Wilkerson, Liu)
| | - Fang-Ming Deng
- the Department of Pathology, New York University Grossman School of Medicine, New York City (Deng)
| | - Haiyan Liu
- From the Department of Pathology, Geisinger Medical Center, Danville, Pennsylvania (Li, Wilkerson, Liu)
| |
Collapse
|
9
|
Ma C, Wang X, Dai JY, Turman C, Kraft P, Stopsack KH, Loda M, Pettersson A, Mucci LA, Stanford JL, Penney KL. Germline Genetic Variants Associated with Somatic TMPRSS2:ERG Fusion Status in Prostate Cancer: A Genome-Wide Association Study. Cancer Epidemiol Biomarkers Prev 2023; 32:1436-1443. [PMID: 37555839 PMCID: PMC10592169 DOI: 10.1158/1055-9965.epi-23-0275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/12/2023] [Accepted: 08/04/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND The prostate cancer subtype defined by the presence of TMPRSS2:ERG has been shown to be molecularly and epidemiologically distinct. However, few studies have investigated germline genetic variants associating with TMPRSS2:ERG fusion status. METHODS We performed a genome-wide association study with 396 TMPRSS2:ERG(+) cases, 390 TMPRSS2:ERG(-) cases, and 2,386 cancer-free controls from the Physicians' Health Study (PHS), the Health Professionals Follow-up Study (HPFS), and a Seattle-based Fred Hutchinson (FH) Cancer Center Prostate Cancer Study. We applied logistic regression models to test the associations between ∼5 million SNPs with TMPRSS2:ERG fusion status accounting for population stratification. RESULTS We did not identify genome-wide significant variants comparing the TMPRSS2:ERG(+) to the TMPRSS2:ERG(-) prostate cancer cases in the meta-analysis. When comparing TMPRSS2:ERG(+) prostate cancer cases with controls without prostate cancer, 10 genome-wide significant SNPs on chromosome 17q24.3 were observed in the meta-analysis. When comparing TMPRSS2:ERG(-) prostate cancer cases with controls without prostate cancer, two SNPs on chromosome 8q24.21 in the meta-analysis reached genome-wide significance. CONCLUSIONS We observed SNPs at several known prostate cancer risk loci (17q24.3, 1q32.1, and 8q24.21) that were differentially and exclusively associated with the risk of developing prostate tumors either with or without the gene fusion. IMPACT Our findings suggest that tumors with the TMPRSS2:ERG fusion exhibit a different germline genetic etiology compared with fusion negative cases.
Collapse
Affiliation(s)
- Chaoran Ma
- Department of Nutrition, University of Massachusetts Amherst, Amherst, MA
| | - Xiaoyu Wang
- Division of Public Health Sciences, Fred Hutchison Cancer Center, Seattle, WA
| | - James Y. Dai
- Division of Public Health Sciences, Fred Hutchison Cancer Center, Seattle, WA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Konrad H. Stopsack
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Andreas Pettersson
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Janet L. Stanford
- Division of Public Health Sciences, Fred Hutchison Cancer Center, Seattle, WA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA
| | - Kathryn L. Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
10
|
Erak E, Oliveira LD, Mendes AA, Dairo O, Ertunc O, Kulac I, Baena-Del Valle JA, Jones T, Hicks JL, Glavaris S, Guner G, Vidal ID, Markowski M, de la Calle C, Trock BJ, Meena A, Joshi U, Kondragunta C, Bonthu S, Singhal N, De Marzo AM, Lotan TL. Predicting Prostate Cancer Molecular Subtype With Deep Learning on Histopathologic Images. Mod Pathol 2023; 36:100247. [PMID: 37307876 PMCID: PMC11225718 DOI: 10.1016/j.modpat.2023.100247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
Microscopic examination of prostate cancer has failed to reveal a reproducible association between molecular and morphologic features. However, deep-learning algorithms trained on hematoxylin and eosin (H&E)-stained whole slide images (WSI) may outperform the human eye and help to screen for clinically-relevant genomic alterations. We created deep-learning algorithms to identify prostate tumors with underlying ETS-related gene (ERG) fusions or PTEN deletions using the following 4 stages: (1) automated tumor identification, (2) feature representation learning, (3) classification, and (4) explainability map generation. A novel transformer-based hierarchical architecture was trained on a single representative WSI of the dominant tumor nodule from a radical prostatectomy (RP) cohort with known ERG/PTEN status (n = 224 and n = 205, respectively). Two distinct vision transformer-based networks were used for feature extraction, and a distinct transformer-based model was used for classification. The ERG algorithm performance was validated across 3 RP cohorts, including 64 WSI from the pretraining cohort (AUC, 0.91) and 248 and 375 WSI from 2 independent RP cohorts (AUC, 0.86 and 0.89, respectively). In addition, we tested the ERG algorithm performance in 2 needle biopsy cohorts comprised of 179 and 148 WSI (AUC, 0.78 and 0.80, respectively). Focusing on cases with homogeneous (clonal) PTEN status, PTEN algorithm performance was assessed using 50 WSI reserved from the pretraining cohort (AUC, 0.81), 201 and 337 WSI from 2 independent RP cohorts (AUC, 0.72 and 0.80, respectively), and 151 WSI from a needle biopsy cohort (AUC, 0.75). For explainability, the PTEN algorithm was also applied to 19 WSI with heterogeneous (subclonal) PTEN loss, where the percentage tumor area with predicted PTEN loss correlated with that based on immunohistochemistry (r = 0.58, P = .0097). These deep-learning algorithms to predict ERG/PTEN status prove that H&E images can be used to screen for underlying genomic alterations in prostate cancer.
Collapse
Affiliation(s)
- Eric Erak
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | - Onur Ertunc
- Department of Pathology, Suleyman Demirel University, Turkey
| | | | | | - Tracy Jones
- Department of Pathology, Johns Hopkins University School of Medicine
| | - Jessica L Hicks
- Department of Pathology, Johns Hopkins University School of Medicine
| | | | | | | | - Mark Markowski
- Department of Oncology, Johns Hopkins University School of Medicine
| | | | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine
| | | | | | | | | | | | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine; Department of Oncology, Johns Hopkins University School of Medicine; Department of Urology, Johns Hopkins University School of Medicine
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine; Department of Oncology, Johns Hopkins University School of Medicine; Department of Urology, Johns Hopkins University School of Medicine.
| |
Collapse
|
11
|
Salles DC, Mendes AA, Han M, Partin AW, Trock BJ, Jing Y, Lotan TL. ERG Status at the Margin Is Associated With Biochemical Recurrence After Radical Prostatectomy With Positive Surgical Margins. Mod Pathol 2023; 36:100147. [PMID: 36828362 PMCID: PMC10442458 DOI: 10.1016/j.modpat.2023.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/22/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Positive surgical margins at radical prostatectomy are associated with an increased risk of biochemical recurrence (BCR). However, there is considerable variability in outcomes, suggesting that molecular biomarkers-when assessed specifically at the margin tumor tissue-may be useful to stratify prognosis in this group. We used a case-cohort design for the outcome of BCR, selecting 215 patients from a cohort of 813 patients undergoing prostatectomy treated at the Johns Hopkins from 2008 to 2017 with positive margins and available clinical data. Tissue microarrays were created from the tumor adjacent to the positive margin and stained for PTEN, ERG, and Ki-67. Cases were scored dichotomously (PTEN and ERG) or by the Ki-67 staining index using previously validated protocols. The analysis used Cox proportional hazards models weighted for the case-cohort design. Overall, 20% (37/185) of evaluable cases had PTEN loss and 38% (71/185) had ERG expression, and the median Ki-67 expression was 0.42%. In multivariable analysis adjusting for the CAPRA-S score, adjuvant radiation, and grade group at the positive margin, ERG-positive tumors were associated with a higher risk of BCR compared to those that were ERGnegative (hazard ratio [HR], 2.4; 95% CI, 1.2-4.9; P = .012) regardless of PTEN status at the margin, and adding ERG to clinicopathologic variables increased the concordance index from 0.827 to 0.847. PTEN loss was associated with an increased risk of BCR on univariable analysis (HR, 3.19; 95% CI, 1.72-5.92; P = .0002), but this association did not remain after adjusting for clinicopathologic variables (HR, 1.06; 95% CI, 0.49-2.29; P = .890). Thus, in the setting of prostate tumors with positive surgical margins after prostatectomy, ERG-positive tumors with or without PTEN loss at the positive margin are associated with a significantly higher risk of BCR after adjusting for clinicopathologic variables. If validated, ERG status may be helpful in decision-making surrounding adjuvant therapy after prostatectomy.
Collapse
Affiliation(s)
- Daniela C Salles
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Adrianna A Mendes
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Misop Han
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan W Partin
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuezhou Jing
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
12
|
SARSIK KUMBARACI B, KANAT E, AYKUTLU U, KIZILAY F, ŞEN S. Prostatın benign, prekürsör ve malign epitelyal proliferasyonlarında ERG ile PTEN ekspresyonlarının araştırılması ve bulguların klinikopatolojik korelasyonu. EGE TIP DERGISI 2022. [DOI: 10.19161/etd.1209075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Amaç: Prostat kanseri farklı klinik gidişata ve geniş bir tedavi yelpazesine sahip, klinik ve moleküler olarak oldukça heterojen bir kanser türüdür. Özellikle “prostatik intraepitelyal neoplazi” (PİN), “atipik intraduktal proliferasyon” (AİP) ve “intraduktal karsinom” (İDK) benzer morfolojik özelliklere sahip olması açısından ayırıcı tanı zorluğu yaratan tanılar olup, hasta tedavi ve takibi de farklı olan antitelerdir. Çalışmamızda bu lezyonlarda ERG ve PTEN ekspresyon düzeylerini belirlemeyi ve bu biyobelirteçlerin prognostik ve diagnostik değerini araştırmayı amaçladık. Gereç ve Yöntem: EÜTF Tıbbi Patoloji Anabilim Dalında 2011-2012 yılında radikal prostatektomi veya iğne biyopsi materyallerinde “Adenokarsinom” tanısı almış 87 olgu çalışmaya alındı. Histopatolojik olarak AİP, İDK ve PİN içeren alanlar belirlendi. immunohistokimyasal olarak bu alanlarda ERG ve PTEN ekspresyonları değerlendirildi.Bulgular: Olguların 6’sında İDK, 29’unda AİP ve 52’sinde PİN belirlendi. İDK AİP, DG 3 ve üstünde olan tümörlerde daha fazla görüldü. İDK ve AİP in eşlik ettiği prostat karsinomlarının sağ kalım süresi daha kısaydı (p=0.043). İDK ve AİP içeren tümörlerde ERG ve PTEN durumu invaziv komponentle uyum içindeydi. Ayrıca tüm İDK alanlarında ERG pozitifti. PTEN ile heterojen boyanma görülmüş olup, PTEN’in invaziv karsinom ve İDK alanlarında negatifliği daha fazlaydı (p=0,63). ERG pozitifliği ve PTEN negatifliği istatistiksel olarak anlamlı olmamakla birlikte AİP tanısını desteklediği dikkati çekti.Sonuç: Özellikle ayırıcı tanı sorunu yaratan intraduktal lezyonlarda ERG pozitifliği ve PTEN negatifliği klinik öneme sahip prostat karsinomuna eşlik edebileceği için özellikle biyopsilerde gözardı edilmemeli ve hasta tedavi ile takibi buna göre yapılmalıdır.
Collapse
Affiliation(s)
- Banu SARSIK KUMBARACI
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, TIBBİ PATOLOJİ ANABİLİM DALI
| | - Emre KANAT
- UŞAK ÜNİVERSİTESİ, TIP FAKÜLTESİ, DAHİLİ TIP BİLİMLERİ BÖLÜMÜ, ACİL TIP ANABİLİM DALI
| | - Umut AYKUTLU
- Acıbadem Sağlık Grubu, Altunizade Hastanesi, Patoloji Laboratuvarı
| | - Fuat KIZILAY
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, ÜROLOJİ ANABİLİM DALI
| | - Sait ŞEN
- EGE ÜNİVERSİTESİ, TIP FAKÜLTESİ, CERRAHİ TIP BİLİMLERİ BÖLÜMÜ, TIBBİ PATOLOJİ ANABİLİM DALI
| |
Collapse
|
13
|
Bakbak H, Sayar E, Kaur HB, Salles DC, Patel RA, Hicks J, Lotan TL, De Marzo AM, Gulati R, Epstein JI, Haffner MC. Clonal relationships of adjacent Gleason pattern 3 and Gleason pattern 5 lesions in Gleason Scores 3+5=8 and 5+3=8. Hum Pathol 2022; 130:18-24. [PMID: 36309296 PMCID: PMC10542864 DOI: 10.1016/j.humpath.2022.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2022]
Abstract
Genomic studies have demonstrated a high level of intra-tumoral heterogeneity in prostate cancer. There is strong evidence suggesting that individual tumor foci can arise as genetically distinct, clonally independent lesions. However, recent studies have also demonstrated that adjacent Gleason pattern (GP) 3 and GP4 lesions can originate from the same clone but follow divergent genetic and morphologic evolution. The clonal relationship of adjacent GP3 and GP5 lesions has thus far not been investigated. Here we analyzed a cohort of 14 cases-11 biopsy and 3 radical prostatectomy specimens-with a Gleason score of 3 + 5 = 8 or 5 + 3 = 8 present in the same biopsy or in a single dominant tumor nodule at radical prostatectomy. Clonal and subclonal relationships between GP3 and GP5 lesions were assessed using genetically validated immunohistochemical assays for ERG, PTEN, and P53. 9/14 (64%) cases showed ERG reactivity in both GP3 and GP5 lesions. Only 1/14 (7%) cases showed a discordant pattern with ERG staining present only in GP3. PTEN expression was lost in 2/14 (14%) cases with perfect concordance between GP5 and GP3. P53 nuclear reactivity was present in 1/14 (7%) case in both GP5 and GP3. This study provides first evidence that the majority of adjacent GP3 and GP5 lesions share driver alterations and are clonally related. In addition, we observed a lower-than-expected rate of PTEN loss in GP5 in the context of Gleason score 3 + 5 = 8 or 5 + 3 = 8 tumors.
Collapse
Affiliation(s)
- Hasim Bakbak
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Erolcan Sayar
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Harsimar B Kaur
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Daniela C Salles
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Radhika A Patel
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jessica Hicks
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Tamara L Lotan
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Angelo M De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jonathan I Epstein
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA.
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA; Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, 21287, MD, USA; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, 98195, WA, USA.
| |
Collapse
|
14
|
Mullen D, Nowak K, Chetty R. Gene of the month: ERG. J Clin Pathol 2022; 75:577-580. [PMID: 35738886 DOI: 10.1136/jcp-2022-208350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 11/04/2022]
Abstract
The ERG gene belongs to the erythroblastosis transformation specific family of transcription factors and encodes for the transcription regulator protein ERG. It is located on chromosome 22q22 and is a nuclear transcription factor. In normal physiology, ERG protein is expressed in endothelial cells and is involved in processes including, but not limited to, angiogenesis and haematopoiesis. Of diagnostic value in clinical practice, ERG immunohistochemistry is a useful marker of endothelial differentiation for both benign and malignant vascular lesions. It is also reliable for identifying ERG gene translocated malignancies such as EWS/FUS::ERG Ewing's sarcoma and TMPSSR2::ERG prostatic carcinoma.
Collapse
Affiliation(s)
- Dorinda Mullen
- Department of Pathology, University Health Network Laboratory Medicine Program, University of Toronto, Toronto, Ontario, Canada
| | - Klaudia Nowak
- Department of Pathology, University Health Network Laboratory Medicine Program, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
15
|
OUP accepted manuscript. Carcinogenesis 2022; 43:779-786. [DOI: 10.1093/carcin/bgac036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/04/2022] [Accepted: 04/21/2022] [Indexed: 11/14/2022] Open
|
16
|
Vidal I, Zheng Q, Hicks JL, Chen J, Platz EA, Trock BJ, Kulac I, Baena-Del Valle JA, Sfanos KS, Ernst S, Jones T, Maynard JP, Glavaris SA, Nelson WG, Yegnasubramanian S, De Marzo AM. GSTP1 positive prostatic adenocarcinomas are more common in Black than White men in the United States. PLoS One 2021; 16:e0241934. [PMID: 34191807 PMCID: PMC8244883 DOI: 10.1371/journal.pone.0241934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
GSTP1 is a member of the Glutathione-S-transferase (GST) family silenced by CpG island DNA hypermethylation in 90-95% of prostate cancers. However, prostate cancers expressing GSTP1 have not been well characterized. We used immunohistochemistry against GSTP1 to examine 1673 primary prostatic adenocarcinomas on tissue microarrays (TMAs) with redundant sampling from the index tumor from prostatectomies. GSTP1 protein was positive in at least one TMA core in 7.7% of cases and in all TMA cores in 4.4% of cases. The percentage of adenocarcinomas from Black patients who had any GSTP1 positive TMA cores was 14.9%, which was 2.5 times higher than the percentage from White patients (5.9%; P < 0.001). Further, the percentages of tumors from Black patients who had all TMA spots positive for GSTP1 (9.5%) was 3-fold higher than the percentage from White patients (3.2%; P<0.001). In terms of association with other molecular alterations, GSTP1 positivity was enriched in ERG positive cancers among Black men. By in situ hybridization, GSTP1 mRNA expression was concordant with protein staining, supporting the lack of silencing of at least some GSTP1 alleles in GSTP1-positive tumor cells. This is the first report revealing that GSTP1-positive prostate cancers are substantially over-represented among prostate cancers from Black compared to White men. This observation should prompt additional studies to determine whether GSTP1 positive cases represent a distinct molecular subtype of prostate cancer and whether GSTP1 expression could provide a biological underpinning for the observed disparate outcomes for Black men.
Collapse
Affiliation(s)
- Igor Vidal
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Qizhi Zheng
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jessica L. Hicks
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jiayu Chen
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Elizabeth A. Platz
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Bruce J. Trock
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | | | - Karen S. Sfanos
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sarah Ernst
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Tracy Jones
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Janielle P. Maynard
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Stephanie A. Glavaris
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William G. Nelson
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Srinivasan Yegnasubramanian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Angelo M. De Marzo
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
- The Brady Urological Research Institute at Johns Hopkins, Baltimore, Maryland, United States of America
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Homologous recombination deficiency (HRD) score in germline BRCA2- versus ATM-altered prostate cancer. Mod Pathol 2021; 34:1185-1193. [PMID: 33462368 PMCID: PMC8154637 DOI: 10.1038/s41379-020-00731-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 01/28/2023]
Abstract
The homologous recombination deficiency (HRD) score integrates three DNA-based measures of genomic instability, and has been understudied in prostate cancer. Given the recent FDA approval of two PARP inhibitors for prostate cancer, HRD score analysis could help to refine treatment selection. We assessed HRD score (defined as the sum of loss-of-heterozygosity, telomeric allelic imbalance, and large-scale state transitions) in three cohorts of primary prostate cancer, including a Johns Hopkins University (JHU) cohort with germline mutations in BRCA2, ATM, or CHEK2 (n = 64), the TCGA cohort (n = 391), and the PROGENE cohort (n = 102). In the JHU cohort, tumors with germline BRCA2 mutations had higher HRD scores (median = 27) than those with germline ATM or CHEK2 mutations (median = 16.5 [p = 0.029] and 9 [p < 0.001], respectively). For TCGA tumors without underlying HR pathway mutations, the median HRD score was 11, significantly lower than ovarian carcinoma lacking BRCA1/2 mutations (median = 28). In the absence of HR gene mutations, the median HRD score was unexpectedly higher among prostate cancers with TP53 mutations versus those without (17 vs. 11; p = 0.015); this finding was confirmed in the PROGENE cohort (24 vs. 16; p = 0.001). Finally, among eight BRCA2-altered patients who received olaparib, progression-free survival trended longer in those with HRD scores above versus below the median (14.9 vs. 9.9 months). We conclude that HRD scores are low in primary prostate cancer and higher in cases with germline BRCA2 or somatic TP53 mutations. Germline BRCA2-altered cases have significantly higher HRD scores than germline ATM-altered or CHEK2-altered cases, consistent with the lower efficacy of PARP inhibitors among the latter.
Collapse
|
18
|
Jamaspishvili T, Patel PG, Niu Y, Vidotto T, Caven I, Livergant R, Fu W, Kawashima A, How N, Okello JB, Guedes LB, Ouellet V, Picanço C, Koti M, Reis RB, Saad F, Mes-Masson AM, Lotan TL, Squire JA, Peng YP, Siemens DR, Berman DM. Risk Stratification of Prostate Cancer Through Quantitative Assessment of PTEN Loss (qPTEN). J Natl Cancer Inst 2021; 112:1098-1104. [PMID: 32129857 DOI: 10.1093/jnci/djaa032] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 12/25/2019] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Phosphatase and tensin homolog (PTEN) loss has long been associated with adverse findings in early prostate cancer. Studies to date have yet to employ quantitative methods (qPTEN) for measuring of prognostically relevant amounts of PTEN loss in postsurgical settings and demonstrate its clinical application. METHODS PTEN protein levels were measured by immunohistochemistry in radical prostatectomy samples from training (n = 410) and validation (n = 272) cohorts. PTEN loss was quantified per cancer cell and per tissue microarray core. Thresholds for identifying clinically relevant PTEN loss were determined using log-rank statistics in the training cohort. Univariate (Kaplan-Meier) and multivariate (Cox proportional hazards) analyses on various subpopulations were performed to assess biochemical recurrence-free survival (BRFS) and were independently validated. All statistical tests were two-sided. RESULTS PTEN loss in more than 65% cancer cells was most clinically relevant and had statistically significant association with reduced BRFS in training (hazard ratio [HR] = 2.48, 95% confidence interval [CI] = 1.59 to 3.87; P < .001) and validation cohorts (HR = 4.22, 95% CI = 2.01 to 8.83; P < .001). The qPTEN scoring method identified patients who recurred within 5.4 years after surgery (P < .001). In men with favorable risk of biochemical recurrence (Cancer of the Prostate Risk Assessment - Postsurgical scores <5 and no adverse pathological features), qPTEN identified a subset of patients with shorter BRFS (HR = 5.52, 95% CI = 2.36 to 12.90; P < .001) who may be considered for intensified monitoring and/or adjuvant therapy. CONCLUSIONS Compared with previous qualitative approaches, qPTEN improves risk stratification of postradical prostatectomy patients and may be considered as a complementary tool to guide disease management after surgery.
Collapse
Affiliation(s)
- Tamara Jamaspishvili
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Palak G Patel
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Yi Niu
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Thiago Vidotto
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada.,Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Isabelle Caven
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Rachel Livergant
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Winnie Fu
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Atsunari Kawashima
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.,Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Nathan How
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - John B Okello
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Liana B Guedes
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Veronique Ouellet
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Clarissa Picanço
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Madhuri Koti
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada.,Urology, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Rodolfo B Reis
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14048-900, Brazil
| | - Fred Saad
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Department of Surgery, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Anne-Marie Mes-Masson
- Institut du Cancer de Montréal and Centre de Recherche du Centre hospitalier de l, 'Université de Montréal, Montréal, Québec H2X 0A9, Canada.,Department of Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Jeremy A Squire
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Yingwei P Peng
- Division of Cancer Care and Epidemiology, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Public Health Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.,Mathematics and Statistics, Queen's University, Kingston, ON K7L 3N6, Canada
| | | | - David M Berman
- Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON K7L 3N6, Canada.,Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.,Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 2V7, Canada
| |
Collapse
|
19
|
Hewitt SM. The Challenges of Optimizing Biomarkers to Guide Clinical Decision Making. J Natl Cancer Inst 2021; 112:1079-1080. [PMID: 32129860 DOI: 10.1093/jnci/djaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Kaur H, Salles DC, Murali S, Hicks JL, Nguyen M, Pritchard CC, De Marzo AM, Lanchbury JS, Trock BJ, Isaacs WB, Timms KM, Antonarakis ES, Lotan TL. Genomic and Clinicopathologic Characterization of ATM-deficient Prostate Cancer. Clin Cancer Res 2020; 26:4869-4881. [PMID: 32694154 PMCID: PMC7501149 DOI: 10.1158/1078-0432.ccr-20-0764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/28/2020] [Accepted: 07/15/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE The ATM (ataxia telangiectasia mutated) gene is mutated in a subset of prostate cancers, and ATM mutation may confer specific therapeutic vulnerabilities, although ATM-deficient prostate cancers have not been well-characterized. EXPERIMENTAL DESIGN We genetically validated a clinical grade IHC assay to detect ATM protein loss and examined the frequency of ATM loss among tumors with pathogenic germline ATM mutations and genetically unselected primary prostate carcinomas using tissue microarrays (TMAs). Immunostaining results were correlated with targeted somatic genomic sequencing and clinical outcomes. RESULTS ATM protein loss was found in 13% (7/52) of primary Gleason pattern 5 cancers with available sequencing data and was 100% sensitive for biallelic ATM inactivation. In a separate cohort with pathogenic germline ATM mutations, 74% (14/19) had ATM protein loss of which 70% (7/10) of evaluable cases had genomic evidence of biallelic inactivation, compared with zero of four of cases with intact ATM expression. By TMA screening, ATM loss was identified in 3% (25/831) of evaluable primary tumors, more commonly in grade group 5 (17/181; 9%) compared with all other grades (8/650; 1%; P < 0.0001). Of those with available sequencing, 80% (4/5) with homogeneous ATM protein loss and 50% (6/12) with heterogeneous ATM protein loss had detectable pathogenic ATM alterations. In surgically treated patients, ATM loss was not significantly associated with clinical outcomes in random-effects Cox models after adjusting for clinicopathologic variables. CONCLUSIONS ATM loss is enriched among high-grade prostate cancers. Optimal evaluation of ATM status requires both genomic and IHC studies and will guide development of molecularly targeted therapies.
Collapse
Affiliation(s)
- Harsimar Kaur
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Daniela C Salles
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sanjana Murali
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jessica L Hicks
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Bruce J Trock
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William B Isaacs
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Emmanuel S Antonarakis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland.
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
Kaur H, Samarska I, Lu J, Faisal F, Maughan BL, Murali S, Asrani K, Alshalalfa M, Antonarakis ES, Epstein JI, Joshu CE, Schaeffer EM, Mosquera JM, Lotan TL. Neuroendocrine differentiation in usual-type prostatic adenocarcinoma: Molecular characterization and clinical significance. Prostate 2020; 80:1012-1023. [PMID: 32649013 PMCID: PMC9524879 DOI: 10.1002/pros.24035] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/14/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Small cell neuroendocrine (NE) carcinomas of the prostate classically lose androgen receptor (AR) expression, may harbor loss of the RB1, TP53, and PTEN tumor suppressor genes, and are associated with a poor prognosis. However usual-type adenocarcinomas may also contain areas of NE differentiation, and in this context the molecular features and biological significance are less certain. METHODS We examined the molecular phenotype and oncologic outcomes of primary prostate adenocarcinomas with ≥5% NE differentiation (≥5% chromogranin A-positive NE cells in any given tumor spot on tissue microarray) using three independent study sets: a set of tumors with paneth cell-like NE differentiation (n = 26), a retrospective case-cohort of intermediate- and high-risk patients enriched for adverse outcomes (n = 267), and primary tumors from a retrospective series of men with eventual castration-resistant metastatic prostate cancer (CRPC) treated with abiraterone or enzalutamide (n = 55). RESULTS Benign NE cells expressed significantly lower quantified AR levels compared with paired benign luminal cells (P < .001). Similarly, paneth-like NE carcinoma cells or carcinoma cells expressing chromogranin A expressed significantly lower quantified AR levels than paired non-NE carcinoma cells (P < .001). Quantified ERG protein expression, was also lower in chromogranin A-labeled adenocarcinoma cells compared with unlabeled cells (P < .001) and tumors with NE differentiation showed lower gene expression scores for AR activity compared with those without. Despite evidence of lower AR signaling, adenocarcinomas with NE differentiation did not differ by prevalence of TP53 missense mutations, or PTEN or RB1 loss, compared with those without NE differentiation. Finally, NE differentiation was not associated with time to metastasis in intermediate- and high-risk patients (P = .6 on multivariate analysis), nor with progression-free survival in patients with CRPC treated with abiraterone or enzalutamide (P = .9). CONCLUSION NE differentiation in usual-type primary prostate adenocarcinoma is a molecularly and clinically distinct form of lineage plasticity from that occurring in small cell NE carcinoma.
Collapse
Affiliation(s)
- Harsimar Kaur
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Iryna Samarska
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Farzana Faisal
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Benjamin L. Maughan
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sanjana Murali
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - Jonathan I. Epstein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Edward M. Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
22
|
Lau HD, Clark M. Metastatic squamous cell carcinoma transformed from prostatic adenocarcinoma following androgen deprivation therapy: A case report with clinicopathologic and molecular findings. Diagn Cytopathol 2020; 48:E14-E17. [PMID: 32628337 DOI: 10.1002/dc.24539] [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] [Received: 05/22/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022]
Abstract
Squamous cell carcinoma (SCC) of the prostate is a rare and clinically aggressive entity that may arise de novo or through transformation of prostatic adenocarcinoma, typically following hormonal or radiation therapy. Confirmation of prostatic origin, especially when evaluating a metastatic focus, often requires correlation with clinical and imaging findings, as the morphologic and immunohistochemical features of SCC are not organ-specific. Comprehensive genomic profiling (CGP) may provide additional information useful for confirming the primary site and for identifying potential targeted therapy options. CGP data may also contribute to our understanding of the molecular basis of squamous differentiation in prostatic malignancies. However, these data are limited, and to our knowledge, there are only three previously published cases of prostatic SCC with reported CGP findings. Herein, we report a case of metastatic keratinizing SCC diagnosed by core needle biopsy in a 68-year-old man with a history of prostatic adenocarcinoma status post radical prostatectomy and androgen deprivation therapy (ADT). NKX3.1 immunohistochemistry was negative. CGP was performed, and a TMPRSS2-ERG fusion, among other genetic alterations, was detected, supporting a diagnosis of metastatic SCC transformed from prostatic adenocarcinoma following ADT. This case supports the use of CGP or other molecular techniques not only to query potential targeted therapy options but also to refine the diagnosis and confirm the primary site of disease in cases with non-specific morphologic and immunophenotypic features, such as SCC.
Collapse
Affiliation(s)
- Hubert D Lau
- VA Palo Alto Health Care System, Pathology and Laboratory Service, Palo Alto, California, USA
| | - Melissa Clark
- VA Palo Alto Health Care System, Pathology and Laboratory Service, Palo Alto, California, USA
| |
Collapse
|
23
|
Eineluoto JT, Sandeman K, Pohjonen J, Sopyllo K, Nordling S, Stürenberg C, Malén A, Kilpeläinen TP, Santti H, Petas A, Matikainen M, Pellinen T, Järvinen P, Kenttämies A, Rannikko A, Mirtti T. Associations of PTEN and ERG with Magnetic Resonance Imaging Visibility and Assessment of Non-organ-confined Pathology and Biochemical Recurrence After Radical Prostatectomy. Eur Urol Focus 2020; 7:1316-1323. [PMID: 32620540 DOI: 10.1016/j.euf.2020.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/18/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diagnosing clinically significant prostate cancer (PCa) is challenging, but may be facilitated by biomarkers and multiparametric magnetic resonance imaging (MRI). OBJECTIVE To determine the association between biomarkers phosphatase and tensin homolog (PTEN) and ETS-related gene (ERG) with visible and invisible PCa lesions in MRI, and to predict biochemical recurrence (BCR) and non-organ-confined (non-OC) PCa by integrating clinical, MRI, and biomarker-related data. DESIGN, SETTING, AND PARTICIPANTS A retrospective analysis of a population-based cohort of men with PCa, who underwent preoperative MRI followed by radical prostatectomy (RP) during 2014-2015 in Helsinki University Hospital (n = 346), was conducted. A tissue microarray corresponding to the MRI-visible and MRI-invisible lesions in RP specimens was constructed and stained for PTEN and ERG. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Associations of PTEN and ERG with MRI-visible and MRI-invisible lesions were examined (Pearson's χ2 test), and predictions of non-OC disease together with clinical and MRI parameters were determined (area under the receiver operating characteristic curve and logistic regression analyses). BCR prediction was analyzed by Kaplan-Meier and Cox proportional hazard analyses. RESULTS AND LIMITATIONS Patients with MRI-invisible lesions (n = 35) had less PTEN loss and ERG-positive expression compared with patients (n = 90) with MRI-visible lesions (17.2% vs 43.3% [p = 0.006]; 8.6% vs 20.0% [p = 0.125]). Patients with invisible lesions had better, but not statistically significantly improved, BCR-free survival probability in Kaplan-Meier analyses (p = 0.055). Rates of BCR (5.7% vs 21.1%; p = 0.039), extraprostatic extension (11.4% vs 44.6%; p < 0.001), seminal vesicle invasion (0% vs 21.1%; p = 0.003), and lymph node metastasis (0% vs 12.2%; p = 0.033) differed between the groups in favor of patients with MRI-invisible lesions. Biomarkers had no independent role in predicting non-OC disease or BCR. The short follow-up period was a limitation. CONCLUSIONS PTEN loss, BCR, and non-OC RP findings were more often encountered with MRI-visible lesions. PATIENT SUMMARY Magnetic resonance imaging (MRI) of the prostate misses some cancer lesions. MRI-invisible lesions seem to be less aggressive than MRI-visible lesions.
Collapse
Affiliation(s)
- Juho T Eineluoto
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - Kevin Sandeman
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Joona Pohjonen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Konrad Sopyllo
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Stig Nordling
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Carolin Stürenberg
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Adrian Malén
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas P Kilpeläinen
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Henrikki Santti
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anssi Petas
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Matikainen
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Petrus Järvinen
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anu Kenttämies
- HUS Medical Imaging Center Department of Diagnostic Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Rannikko
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Mirtti
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
24
|
Telomere lengths differ significantly between small-cell neuroendocrine prostate carcinoma and adenocarcinoma of the prostate. Hum Pathol 2020; 101:70-79. [PMID: 32389660 DOI: 10.1016/j.humpath.2020.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/28/2020] [Indexed: 01/05/2023]
Abstract
Small-cell neuroendocrine carcinoma (SCNC) of the prostate is an aggressive subtype with frequent TP53 mutation and RB1 inactivation; however, the molecular phenotype remains an area of investigation. Here, we compared telomere lengths in prostatic SCNC and usual-type prostatic adenocarcinoma (AdCa). We studied 32 cases of prostatic SCNC (including 11 cases with concurrent AdCa) and 347 cases of usual-type AdCa on tissue microarrays. Telomere lengths in tumor cells were qualitatively compared with those in normal cells using a telomere-specific fluorescence in situ hybridization assay. ERG, PTEN, and TP53 status were assessed in a proportion of cases using genetically validated immunohistochemistry protocols. Clinicopathological and molecular characteristics of cases were compared between the telomere groups using the chi-square test.A significantly higher proportion of prostatic SCNC cases (50%, 16/32) showed normal/long telomeres compared with AdCa cases (11%, 39/347; P < 0.0001). In 82% (9/11) of cases with concurrent SCNC and AdCa, the paired components were concordant for telomere length status. Among AdCa cases, the proportion of cases with normal/long telomeres significantly increased with increasing tumor grade group (P = 0.01) and pathologic stage (P = 0.02). Cases with normal/long telomeres were more likely to be ERG positive (P = 0.04) and to have TP53 missense mutation (P = 0.01) than cases with short telomeres.Normal or long telomere lengths are significantly more common in prostatic SCNC than in AdCa and are similar between concurrent SCNC and AdCa tumors, supporting a common origin. Among AdCa cases, longer telomere lengths are significantly associated with high-risk pathologic and molecular features.
Collapse
|
25
|
An Algorithmic Immunohistochemical Approach to Define Tumor Type and Assign Site of Origin. Adv Anat Pathol 2020; 27:114-163. [PMID: 32205473 DOI: 10.1097/pap.0000000000000256] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunohistochemistry represents an indispensable complement to an epidemiology and morphology-driven approach to tumor diagnosis and site of origin assignment. This review reflects the state of my current practice, based on 15-years' experience in Pathology and a deep-dive into the literature, always striving to be better equipped to answer the age old questions, "What is it, and where is it from?" The tables and figures in this manuscript are the ones I "pull up on the computer" when I am teaching at the microscope and turn to myself when I am (frequently) stuck. This field is so exciting because I firmly believe that, through the application of next-generation immunohistochemistry, we can provide better answers than ever before. Specific topics covered in this review include (1) broad tumor classification and associated screening markers; (2) the role of cancer epidemiology in determining pretest probability; (3) broad-spectrum epithelial markers; (4) noncanonical expression of broad tumor class screening markers; (5) a morphologic pattern-based approach to poorly to undifferentiated malignant neoplasms; (6) a morphologic and immunohistochemical approach to define 4 main carcinoma types; (7) CK7/CK20 coordinate expression; (8) added value of semiquantitative immunohistochemical stain assessment; algorithmic immunohistochemical approaches to (9) "garden variety" adenocarcinomas presenting in the liver, (10) large polygonal cell adenocarcinomas, (11) the distinction of primary surface ovarian epithelial tumors with mucinous features from metastasis, (12) tumors presenting at alternative anatomic sites, (13) squamous cell carcinoma versus urothelial carcinoma, and neuroendocrine neoplasms, including (14) the distinction of pheochromocytoma/paraganglioma from well-differentiated neuroendocrine tumor, site of origin assignment in (15) well-differentiated neuroendocrine tumor and (16) poorly differentiated neuroendocrine carcinoma, and (17) the distinction of well-differentiated neuroendocrine tumor G3 from poorly differentiated neuroendocrine carcinoma; it concludes with (18) a discussion of diagnostic considerations in the broad-spectrum keratin/CD45/S-100-"triple-negative" neoplasm.
Collapse
|
26
|
Borkowetz A, Froehner M, Rauner M, Conrad S, Erdmann K, Mayr T, Datta K, Hofbauer LC, Baretton GB, Wirth M, Fuessel S, Toma M, Muders MH. Neuropilin‐2 is an independent prognostic factor for shorter cancer‐specific survival in patients with acinar adenocarcinoma of the prostate. Int J Cancer 2019; 146:2619-2627. [DOI: 10.1002/ijc.32679] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/24/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022]
Affiliation(s)
| | | | - Martina Rauner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III Technische Universität Dresden Germany
| | - Stefanie Conrad
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III Technische Universität Dresden Germany
| | - Kati Erdmann
- Department of Urology Technische Universität Dresden Germany
| | - Thomas Mayr
- Institute of Pathology, Technische Universität Dresden Germany
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology University of Nebraska Medical Center Omaha NE
| | - Lorenz C. Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III Technische Universität Dresden Germany
| | - Gustavo B. Baretton
- Institute of Pathology, Technische Universität Dresden Germany
- Tumor and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine, Technische Universität Dresden Dresden Germany
| | - Manfred Wirth
- Department of Urology Technische Universität Dresden Germany
| | - Susanne Fuessel
- Department of Urology Technische Universität Dresden Germany
| | - Marietta Toma
- Institute of Pathology, Technische Universität Dresden Germany
| | | |
Collapse
|
27
|
Kei S, Adeyi OA. Practical Application of Lineage-Specific Immunohistochemistry Markers: Transcription Factors (Sometimes) Behaving Badly. Arch Pathol Lab Med 2019; 144:626-643. [PMID: 31385722 DOI: 10.5858/arpa.2019-0226-ra] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Transcription factors (TFs) are proteins that regulate gene expression and control RNA transcription from DNA. Lineage-specific TFs have increasingly been used by pathologists to determine tumor lineage, especially in the setting of metastatic tumors of unknown primary, among other uses. With experience gathered from its daily application and increasing pitfalls reported from immunohistochemical studies, these often-touted highly specific TFs are not as reliable as once thought. OBJECTIVES.— To summarize the established roles of many of the commonly used TFs in clinical practice and to discuss known and potential sources for error (eg, false-positivity from cross-reactivity, aberrant, and overlap "lineage-specific" expression) in their application and interpretation. DATA SOURCES.— Literature review and the authors' personal practice experience were used. Several examples selected from the University Health Network (Toronto, Ontario, Canada) are illustrated. CONCLUSIONS.— The application of TF diagnostic immunohistochemistry has enabled pathologists to better assess the lineage/origin of primary and metastatic tumors. However, the awareness of potential pitfalls is essential to avoid misdiagnosis.
Collapse
Affiliation(s)
- Si Kei
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (Dr Lou); and the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Adeyi)
| | - Oyedele A Adeyi
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (Dr Lou); and the Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis (Dr Adeyi)
| |
Collapse
|
28
|
Isaacsson Velho P, Lim D, Wang H, Park JC, Kaur HB, Almutairi F, Carducci MA, Denmeade SR, Markowski MC, Isaacs WB, Antonarakis ES, Pritchard CC, Eisenberger MA, Lotan TL. Molecular Characterization and Clinical Outcomes of Primary Gleason Pattern 5 Prostate Cancer After Radical Prostatectomy. JCO Precis Oncol 2019; 3:PO.19.00081. [PMID: 31650100 PMCID: PMC6812513 DOI: 10.1200/po.19.00081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2019] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Very high-risk prostate cancer (PC) is associated with poor response to local and systemic treatments; however, few cases have been molecularly profiled. We studied clinical outcomes and molecular profiles of patients with clinically localized primary Gleason pattern 5 PC. PATIENTS AND METHODS Clinicopathologic features, targeted somatic and germline sequencing, and PTEN, TP53, and ERG status by immunohistochemistry were assessed in patients undergoing surgery from 2005 to 2015; 60 consecutive patients were identified with Gleason score 5 + 4 = 9 or 5 + 5 = 10 PC after radical prostatectomy with available tissue and clinical follow-up. Clinicopathologic and genomic parameters were correlated with biochemical relapse, metastasis-free survival, time to castration resistance, and overall survival using Cox proportional hazards models. RESULTS Of patients with somatic sequencing data and clinical follow-up, 34% had DNA repair gene mutations, including 22% (11 of 49) with homologous recombination and 12% (six of 49) with mismatch repair gene alterations. Homologous recombination mutations were germline in 82% (nine of 11) of patients. In addition, 33% (16 of 49) had TP53 mutation, and 51% (29 of 57) had PTEN loss. Overall, 43% developed metastasis, with a time to castration resistance of 12 months. On multivariable analysis of clinicopathologic variables, only ductal/intraductal histology (hazard ratio, 4.43; 95% CI, 1.76 to 11.15; P = .002) and seminal vesicle invasion (hazard ratio, 5.14; 95% CI, 1.83 to 14.47; P = .002) were associated with metastasis. Among genomic alterations, only TP53 mutation and PTEN loss were associated with metastasis on univariable analysis, and neither remained significant in multivariable analyses. These data are retrospective and hypothesis generating. CONCLUSION Potentially actionable homologous recombination and mismatch repair alterations are observed in a significant proportion of patients with very high-risk PC at the time of radical prostatectomy. These findings could inform the design of prospective trials in this patient population.
Collapse
Affiliation(s)
| | - David Lim
- Johns Hopkins University, Baltimore, MD
| | - Hao Wang
- Johns Hopkins University, Baltimore, MD
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Becker F, Joerg V, Hupe MC, Roth D, Krupar R, Lubczyk V, Kuefer R, Sailer V, Duensing S, Kirfel J, Merseburger AS, Brägelmann J, Perner S, Offermann A. Increased mediator complex subunit CDK19 expression associates with aggressive prostate cancer. Int J Cancer 2019; 146:577-588. [PMID: 31271443 DOI: 10.1002/ijc.32551] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/23/2022]
Abstract
The Mediator complex is a transcriptional regulator interacting with transcription factors and RNA-polymerase-II. Recently, we identified its subunit CDK19 to be specifically expressed in prostate cancer (PCa) and to be functionally implicated in PCa aggressiveness. Aim of our study was to comprehensively characterize the protein expression of CDK19 and its paralog CDK8 in PCa. We performed immunohistochemistry (IHC) for CDK19/CDK8 on a large cohort including needle biopsies from 202 patients, 799 primary tumor foci of radical prostatectomy specimens from 415 patients, 120 locally advanced tumor foci obtained by palliative transurethral resection, 140 lymph node metastases, 67 distant metastases and 82 benigns. Primary tumors were stained for the proliferation marker Ki67, androgen receptor (AR) and ERG. For 376 patients, clinic-pathologic data were available. Primary endpoint was disease-recurrence-free survival (DFS). Nuclear CDK19 and CDK8 expression increases during progression showing the highest intensity in metastatic and castration-resistant tumors. High CDK19 expression on primary tumors correlates with DFS independently from Gleason grade and PSA. Five-year-DFS rates of patients with primary tumors expressing no, moderate and high CDK19 are 73.7, 56.9 and 30.4%, respectively. CDK19 correlates with Gleason grade, T-stage, Ki67 proliferation-index, nuclear AR expression and ERG-status. Therapeutic options for metastatic and castration-resistant PCa remain limited. In the current study, we confirmed an important role of the Mediator subunit CDK19 in advanced PCa supporting current developments to target CDK19 and its paralog CDK8. Furthermore, CDK19 protein expression has the potential to predict disease recurrence independently from established biomarkers thus contributing to individual management for PCa patients.
Collapse
Affiliation(s)
- Finn Becker
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Vincent Joerg
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Marie C Hupe
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Doris Roth
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | | | - Verena Lubczyk
- Department of Pathology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Rainer Kuefer
- Department of Urology, Klinik am Eichert Alb Fils Kliniken, Goeppingen, Germany
| | - Verena Sailer
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Jutta Kirfel
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - Johannes Brägelmann
- Molecular Pathology, Institute of Pathology, University Hospital of Cologne, Cologne, Germany.,Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne, Germany
| | - Sven Perner
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Anne Offermann
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| |
Collapse
|
30
|
Khani F, Wobker SE, Hicks JL, Robinson BD, Barbieri CE, De Marzo AM, Epstein JI, Pritchard CC, Lotan TL. Intraductal carcinoma of the prostate in the absence of high‐grade invasive carcinoma represents a molecularly distinct type of
in situ
carcinoma enriched with oncogenic driver mutations. J Pathol 2019; 249:79-89. [DOI: 10.1002/path.5283] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/28/2019] [Accepted: 04/15/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Francesca Khani
- Department of Pathology and Laboratory Medicine Weill Cornell Medicine New York NY USA
- Department of Urology Weill Cornell Medicine New York NY USA
| | - Sara E Wobker
- Department of Pathology and Laboratory Medicine UNC Chapel Hill Chapel Hill NC USA
| | - Jessica L Hicks
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine Weill Cornell Medicine New York NY USA
- Department of Urology Weill Cornell Medicine New York NY USA
| | | | - Angelo M De Marzo
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Urology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Oncology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Jonathan I Epstein
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Urology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Oncology Johns Hopkins University School of Medicine Baltimore MD USA
| | - Colin C Pritchard
- Department of Laboratory Medicine University of Washington Seattle WA USA
| | - Tamara L Lotan
- Department of Pathology Johns Hopkins University School of Medicine Baltimore MD USA
- Department of Oncology Johns Hopkins University School of Medicine Baltimore MD USA
| |
Collapse
|
31
|
Schweizer MT, Antonarakis ES, Bismar TA, Guedes LB, Cheng HH, Tretiakova MS, Vakar-Lopez F, Klemfuss N, Konnick EQ, Mostaghel EA, Hsieh AC, Nelson PS, Yu EY, Montgomery RB, True LD, Epstein JI, Lotan TL, Pritchard CC. Genomic Characterization of Prostatic Ductal Adenocarcinoma Identifies a High Prevalence of DNA Repair Gene Mutations. JCO Precis Oncol 2019; 3. [PMID: 31123724 DOI: 10.1200/po.18.00327] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Ductal prostate cancer (dPC) is a rare variant of prostatic adenocarcinoma associated with poor outcomes. Although its histopathologic features are well characterized, the underlying molecular hallmarks of this aggressive subtype are not well described. We sought to provide a comprehensive overview of the spectrum of mutations associated with dPC. METHODS Three case series across multiple institutions were assembled. All patients had a diagnosis of dPC, and histopathologic classification was confirmed by an expert genitourinary pathologist. Case series 1 included men who were prospectively enrolled in a tumor sequencing study at the University of Washington (n = 22). Case series 2 and 3 included archival samples from men treated at Johns Hopkins Hospital (n = 21) and University of Calgary (n = 8), respectively. Tumor tissue was sequenced on a targeted next-generation sequencing assay, UW-OncoPlex, according to previously published methods. The frequency of pathogenic/likely pathogenic mutations are reported. RESULTS Overall, 25 patients (49%) had at least one DNA damage repair gene alteration, including seven (14%) with a mismatch repair gene mutation and 16 (31%) with a homologous repair mutation. Germline autosomal dominant mutations were confirmed or suspected in 10 patients (20%). Activating mutations in the PI3K pathway (n = 19; 37%), WNT pathway (n = 16; 31%), and MAPK pathway (n = 8; 16%) were common. CONCLUSION This study strongly suggests that dPCs are enriched for actionable mutations, with approximately 50% of patients demonstrating DNA damage repair pathway alteration(s). Patients with dPC should be offered next-generation sequencing to guide standard-of-care treatment (eg, immune checkpoint inhibitors) or triaged toward an appropriate clinical trial (eg, poly [ADP-ribose] polymerase inhibitors).
Collapse
Affiliation(s)
- Michael T Schweizer
- University of Washington, Seattle, WA.,Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | - Heather H Cheng
- University of Washington, Seattle, WA.,Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | - Elahe A Mostaghel
- University of Washington, Seattle, WA.,Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | - Evan Y Yu
- University of Washington, Seattle, WA.,Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | | | | | | |
Collapse
|
32
|
Rezk M, Chandra A, Addis D, Møller H, Youssef M, Dasgupta P, Yamamoto H. ETS-related gene ( ERG) expression as a predictor of oncological outcomes in patients with high-grade prostate cancer treated with primary androgen deprivation therapy: a cohort study. BMJ Open 2019; 9:e025161. [PMID: 30852544 PMCID: PMC6429920 DOI: 10.1136/bmjopen-2018-025161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES To determine whether ETS-related gene (ERG) expression can be used as a biomarker to predict biochemical recurrence and prostate cancer-specific death in patients with high Gleason grade prostate cancer treated with androgen deprivation therapy (ADT) as monotherapy. METHODS A multicentre retrospective cohort study identifying 149 patients treated with primary ADT for metastatic or non-metastatic prostate cancer with Gleason score 8-10 between 1999 and 2006. Patients planned for adjuvant radiotherapy at diagnosis were excluded. Age at diagnosis, ethnicity, prostate-specific antigen and Charlson-comorbidity score were recorded. Prostatic tissue acquired at biopsy or transurethral resection surgery was assessed for immunohistochemical expression of ERG. Failure of ADT defined as prostate specific antigen nadir +2. Vital status and death certification data determined using the UK National Cancer Registry. Primary outcome measures were overall survival (OS) and prostate cancer specific survival (CSS). Secondary outcome was biochemical recurrence-free survival (BRFS). RESULTS The median OS of our cohort was 60.2 months (CI 52.0 to 68.3). ERG expression observed in 51/149 cases (34%). Multivariate Cox proportional hazards analysis showed no significant association between ERG expression and OS (p=0.41), CSS (p=0.92) and BRFS (p=0.31). Cox regression analysis showed Gleason score (p=0.003) and metastatic status (p<1×10-5) to be the only significant predictors of prostate CSS. CONCLUSIONS No significant association was found between ERG status and any of our outcome measures. Despite a limited sample size, our results suggest that ERG does not appear to be a useful biomarker in predicting response to ADT in patients with high risk prostate cancer.
Collapse
Affiliation(s)
- Mark Rezk
- NIHR Biomedical Research Centre, Kings College London, London, UK
- Intensive Care Unit, Torbay and South Devon NHS Foundation Trust, Torquay, UK
| | - Ashish Chandra
- Department of Histopathology and Cytology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Daniel Addis
- Department of Histopathology and Cytology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Henrik Møller
- Department of Cancer Epidemiology & Populational Health, Kings College London, London, UK
| | - Mina Youssef
- General Surgery Department, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Surgical Oncology Department, National Cancer Institute, Cairo, Egypt
| | - Prokar Dasgupta
- NIHR Biomedical Research Centre, Kings College London, London, UK
| | - Hide Yamamoto
- Department of Urology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Department of Urology, Maidstone Hospital, Maidstone, UK
| |
Collapse
|
33
|
Koide H, Kimura T, Inaba H, Sato S, Iwatani K, Yorozu T, Furusato B, Kamata Y, Miki J, Kiyota H, Takahashi H, Egawa S. Comparison of ERG and SPINK1 expression among incidental and metastatic prostate cancer in Japanese men. Prostate 2019; 79:3-8. [PMID: 30051483 DOI: 10.1002/pros.23705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND TMPRSS2:ERG fusion is the most common genetic event in prostate cancer (PCa). However, its association with prognosis is controversial. Overexpression of serine protease inhibitor Kazal-type 1 (SPINK1) was almost exclusively defined in ERG-negative PCa in most studies. This study aimed to determine the association between ERG and SPINK1 expression and the biological aggressiveness of PCa by analyzing their expression in incidental and metastatic cohorts. METHODS A total of 143 cystoprostatectomy specimens of invasive bladder cancer and 98 biopsy specimens from de novo metastatic PCa were analyzed. The prostate gland of cystoprostatectomy specimens was fixed and sliced in step sections. Immunohistochemistry of ERG and SPINK1 was conducted, and the results were correlated with the clinicopathological characteristics of the patients. RESULTS The overall prevalence of incidental cancer was 32.2% (46/143). The frequencies of both ERG and SPINK1 expression were not significantly different between incidental and metastatic cohorts (15.2% and 14.3%; P = 1.00, and 6.5% and 12.2%; P = 0.38, respectively). In the metastatic cohort, any pre-treatment factors were not significantly associated with the frequencies of ERG and SPINK1 expression. However, SPINK1 expression was significantly associated with a shorter time to castration-resistant PCa (CRPC) (P = 0.048). Meanwhile, overall survival was not significantly associated with the expression status of ERG and SPINK1 (P = 0.71). CONCLUSIONS ERG and SPINK1 expression may not have significant influence on the metastatic behavior of PCa. SPINK1 expression was significantly associated with a shorter time to CRPC in metastatic PCa. The expression profile of ERG and SPINK1 may be a useful predictor for effect of androgen deprivation therapy in patients with metastatic castration-sensitive PCa.
Collapse
Affiliation(s)
- Haruhisa Koide
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Inaba
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Shun Sato
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Kosuke Iwatani
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yorozu
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Bungo Furusato
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuko Kamata
- Division of Oncology, Jikei University School of Medicine, Tokyo, Japan
| | - Jun Miki
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kiyota
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Pathology, Jikei University School of Medicine, Tokyo, Japan
| | - Shin Egawa
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Lange T, Oh-Hohenhorst SJ, Joosse SA, Pantel K, Hahn O, Gosau T, Dyshlovoy SA, Wellbrock J, Feldhaus S, Maar H, Gehrcke R, Kluth M, Simon R, Schlomm T, Huland H, Schumacher U. Development and Characterization of a Spontaneously Metastatic Patient-Derived Xenograft Model of Human Prostate Cancer. Sci Rep 2018; 8:17535. [PMID: 30510249 PMCID: PMC6277427 DOI: 10.1038/s41598-018-35695-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022] Open
Abstract
Here we describe the establishment and characterization of an AR+, PSMA+, ERG+, PTEN-/-, CHD1+/- patient-derived xenograft (PDX) model termed 'C5', which has been developed from a 60 years old patient suffering from castration-resistant prostate cancer (CRPC). The patient underwent radical prostatectomy, showed early tumor marker PSA recurrence and, one year after surgery, abiraterone resistance. Subcutaneous C5 tumors can be serially transplanted between mice and grow within ~90 days to 1.5-2 cm³ tumors in SCID Balb/c mice (take rate 100%), NOD-scid IL2Rgnull (NSG) mice (100%) and C57BL/6 pfp-/-/rag2-/- mice (66%). In contrast, no tumor growth is observed in female mice. C5 tumors can be cryopreserved and show the same growth characteristics in vivo afterwards. C5 tumor cells do not grow stably in vitro, neither under two- nor three-dimensional cell culture conditions. Upon serial transplantation, some C5 tumors spontaneously disseminated to distant sites with an observable trend towards higher metastatic cell loads in scid compared to NSG mice. Lung metastases could be verified by histology by means of anti-PSMA immunohistochemistry, exclusively demonstrating single disseminated tumor cells (DTCs) and micro-metastases. Upon surgical resection of the primary tumors, such pulmonary foci rarely grew out to multi-cellular metastatic colonies despite doubled overall survival span. In the brain and bone marrow, the metastatic cell load present at surgery even disappeared during the post-surgical period. We provide shallow whole genome sequencing and whole exome sequencing data of C5 tumors demonstrating the copy number aberration/ mutation status of this PCa model and proving genomic stability over several passages. Moreover, we analyzed genomic and transcriptomic alterations during metastatic progression achieved by serial transplantation. This study describes a novel PCa PDX model that enables future research on several aspects of metastatic PCa, particularly for the AR+ , ERG+ , PTEN-/- PCa subtype.
Collapse
Affiliation(s)
- Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Su Jung Oh-Hohenhorst
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Hahn
- Department of Urology, University Medical Center Goettingen, Robert-Koch-Strasse 40, 37075, Goettingen, Germany
| | - Tobias Gosau
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Sergey A Dyshlovoy
- Laboratory of Experimental Oncology, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald-Tumorzentrum, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russian Federation
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Feldhaus
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Hanna Maar
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Renate Gehrcke
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Urology, Charité University Hospital, Berlin, Germany
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
35
|
García-Perdomo HA, Chaves MJ, Osorio JC, Sanchez A. Association between TMPRSS2:ERG fusion gene and the prostate cancer: systematic review and meta-analysis. Cent European J Urol 2018; 71:410-419. [PMID: 30680235 PMCID: PMC6338815 DOI: 10.5173/ceju.2018.1752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/28/2018] [Accepted: 11/01/2018] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION To identify the association between the TMPRSS2:ERG fusion gene, their variants and the onset of localized prostate cancer. MATERIAL AND METHODS A systematic search strategy was carried out through MEDLINE, EMBASE, LILACS, CENTRAL and unpublished literature. We included randomized control trials, cohort, case-control and cross-sectional studies that involved patients >18 years-old assessing the association between TMPRSS2 fusion gene, its single nucleotide polymorphisms and prostate cancer. The primary outcome was prostate cancer defined by histology of the tumor coming from transrectal ultrasound guided biopsy, transurethral resection of the prostate or radical prostatectomy. We assessed the risk of bias with QUADAS2 and performed a meta-analysis with Stata 14. RESULTS We found 241 records with the search strategies. After duplicates were removed, 18 studies were included in qualitative analysis and 15 studies in meta-analysis. All included studies that had no applicability concerns and low risk of bias for flow and timing. Nine studies had an unclear risk of bias for index and reference tests, since they did not describe the blinding assessment appropriately. Regarding the association between TMPRSS2:ERG and prostate cancer, we found an odds ratio (OR) 2.24 and a 95% confidence interval (CI) (1.29 to 3.91). Regarding the kind of sample, urine showed an OR 2.79 and a 95% CI (1.12 to 6.98) and when using a DNA molecular template, the OR was 3.55 with a 95% CI (1.08 to 11.65). CONCLUSIONS There was an association between TMPRSS2:ERG fusion gene with the diagnosis of prostate cancer, mainly in urine samples and DNA-based molecular templates. TMPRSS2:ERG might be used as the gold standard biomarker for diagnosis and stratification of PCa.
Collapse
|
36
|
Vicentini C, Cantù C, Antonello D, Simbolo M, Mafficini A, Luchini C, Rusev B, Porcaro AB, Iacovelli R, Fassan M, Corbo V, Brunelli M, Artibani W, Scarpa A, Lawlor RT. ERG alterations and mTOR pathway activation in primary prostate carcinomas developing castration-resistance. Pathol Res Pract 2018; 214:1675-1680. [PMID: 30190183 DOI: 10.1016/j.prp.2018.08.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/03/2023]
Abstract
INTRODUCTION One of the most common sites of distant metastasization of prostate cancer is bone, but to date reliable biomarkers able to predict the risk and timing of bone metastasization are still lacking. PATIENTS AND METHODS Surgically resected paraffin embedded samples from 12 primary prostate cancers that developed metachronous bone metastasis at different time points were studied (six cases within 2 years, six cases after 5 years from surgery). A targeted next-generation DNA and RNA sequencing able to assess simultaneously mutations, copy number alterations and fusion events of multiple genes was used. Immunohistochemistry was used to assess mTOR pathway activation. RESULTS Rearrangements of ETS family genes, molecular alterations in PTEN and TP53 genes were detected in 10, 6 and 5 cancers, respectively. Nine samples showed TMPRSS2-ERG fusions, which were associated with increased ERG expression at immunohistochemistry. mTOR pathway activation was documented in 6 patients, with a clear trend of prevalence in late-metastatic patients (p = 0.08). CONCLUSIONS A simultaneous next-generation targeted DNA and RNA sequencing is applicable on routine formalin-fixed paraffin-embedded tissues to assess the multigene molecular asset of individual prostate cancers. This approach, coupled with immunohistochemistry for ERG and mTOR pathway proteins, may help to better characterize prostate cancer molecular features with a potential impact on clinical decisions.
Collapse
Affiliation(s)
- Caterina Vicentini
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Cinzia Cantù
- ARC-NET Research Centre, University of Verona, Verona, Italy
| | - Davide Antonello
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Surgery, General Surgery B, University of Verona, Verona, Italy
| | - Michele Simbolo
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Borislav Rusev
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | | | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua, PD, Italy
| | - Vincenzo Corbo
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | - Aldo Scarpa
- ARC-NET Research Centre, University of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, Verona, Italy.
| | - Rita T Lawlor
- ARC-NET Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
37
|
Lee SR, Choi YD, Cho NH. Association between pathologic factors and ERG expression in prostate cancer: finding pivotal networking. J Cancer Res Clin Oncol 2018; 144:1665-1683. [PMID: 29948147 DOI: 10.1007/s00432-018-2685-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/06/2018] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate associations between pathologic factors and erythroblast transformation-specific (ETS)-related gene (ERG) expression in prostate cancer patients. Using next-generation sequencing, we identified target genes and regulatory networks. METHODS ERG expression in 60 radical prostatectomies was compared with pathological findings by association rule mining with the Apriori algorithm. Whole-exome and RNA sequencing were performed on three formalin-fixed, paraffin-embedded ERG-positive and negative prostate cancer samples. A network diagram identifying dominant altered genes was constructed using Cytoscape open-source bioinformatics platform and GeneMania plugin. RESULTS Pathologic conditions positive for perineural invasion, apical margins, and Gleason score 3 + 4 = 7 were significantly more likely to be ERG-positive than other pathologic conditions (p = 0.0008), suggesting an association between ERG positivity, perineural invasion, apical margins, and Gleason score 3 + 4 = 7 (Firth's logistic regression: OR 42.565, 95% CI 1.670-1084.847, p = 0.0232). Results of whole-exome and RNA sequencing identified 97 somatic mutations containing common mutated genes. Regulatory network analysis identified NOTCH1, MEF2C, STAT3, LCK, CACNA2D3, PCSK7, MEF2A, PDZD2, TAB1, and ASGR1 as pivotal genes. NOTCH1 appears to function as a hub, because it had the highest node degree and betweenness. NOTCH1 staining was found 8 of 60 specimens (13%), with a significant association between ERG and NOTCH1 positivity (p = 0.001). CONCLUSIONS Evaluating the association between ERG expression and pathologic factors, and identifying the regulatory network and pivotal hub may help to understand the clinical significance of ERG-positive prostate cancer.
Collapse
Affiliation(s)
- Seung-Ryeol Lee
- Department of Urology, CHA Bundang Medical Center, CHA University College of Medicine, Seongnam, South Korea
- Department of Urology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Young-Deuk Choi
- Department of Urology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Nam-Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
38
|
Kamoun A, Cancel-Tassin G, Fromont G, Elarouci N, Armenoult L, Ayadi M, Irani J, Leroy X, Villers A, Fournier G, Doucet L, Boyault S, Brureau L, Multigner L, Diedhiou A, Roupret M, Compérat E, Blanchet P, de Reyniès A, Cussenot O. Comprehensive molecular classification of localized prostate adenocarcinoma reveals a tumour subtype predictive of non-aggressive disease. Ann Oncol 2018; 29:1814-1821. [DOI: 10.1093/annonc/mdy224] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
39
|
Perdomo HAG, Zapata-Copete JA, Sanchez A. Molecular alterations associated with prostate cancer. Cent European J Urol 2018; 71:168-176. [PMID: 30038806 PMCID: PMC6051361 DOI: 10.5173/ceju.2018.1583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/28/2017] [Accepted: 03/16/2018] [Indexed: 01/07/2023] Open
Abstract
Introduction The amount of information and knowledge about pathways and genetic alterations regarding prostate cancer, including the tools available for its study has been recently increasing. Additionally, a variety of molecular signaling pathways control cell proliferation, however, this incompletely understood process is disturbed in cancer cells. Materials and methods A literature review was made using the MEDLINE, Embase and LILACS databases searching for the following keywords: prostate neoplasms, prostate cancer, molecular medicine, genomics, pathways, and cell cycle. Results Different biological mechanisms have been associated with the development of prostate cancer, such as alterations in tumor suppressor genes, oncogenes (TP53, RB1, among others) and CDKIs; DNA methylation; chromosomal alterations and rearrangements; changes in PTEN and PI3K / mTOR; global defects in apoptosis; alterations in the androgen receptor (AR); and epigenetic mechanisms. Conclusions Good clinical practice and a practical approach have to be based on basic knowledge, thus, in this article, the main genetic alterations, mutations and pathways involved in prostate cancer development were reviewed.
Collapse
Affiliation(s)
| | | | - Adalberto Sanchez
- School of Basic Sciences, Department of Physiological Sciences at Universidad del Valle, Cali, Colombia
| |
Collapse
|
40
|
Zhou CK, Young D, Yeboah ED, Coburn SB, Tettey Y, Biritwum RB, Adjei AA, Tay E, Niwa S, Truelove A, Welsh J, Mensah JE, Hoover RN, Sesterhenn IA, Hsing AW, Srivastava S, Cook MB. TMPRSS2:ERG Gene Fusions in Prostate Cancer of West African Men and a Meta-Analysis of Racial Differences. Am J Epidemiol 2017; 186:1352-1361. [PMID: 28633309 PMCID: PMC5860576 DOI: 10.1093/aje/kwx235] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022] Open
Abstract
The prevalence of fusions of the transmembrane protease, serine 2, gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2:ERG, in prostate cancer varies by race. However, such somatic aberration and its association with prognostic factors have neither been studied in a West African population nor been systematically reviewed in the context of racial differences. We used immunohistochemistry to assess oncoprotein encoded by the ERG gene as the established surrogate of ERG fusion genes among 262 prostate cancer biopsies from the Ghana Prostate Study (2004-2006). Poisson regression with robust variance estimation provided prevalence ratios and 95% confidence intervals of ERG expression in relation to patient characteristics. We found that 47 of 262 (18%) prostate cancers were ERG-positive, and being negative for ERG staining was associated with higher Gleason score. We further conducted a systematic review and meta-analysis of TMPRSS2:ERG fusions in relation to race, Gleason score, and tumor stage, combining results from Ghana with 40 additional studies. Meta-analysis showed the prevalence of TMPRSS2:ERG fusions in prostate cancer to be highest in men of European descent (49%), followed by men of Asian (27%) and then African (25%) descent. The lower prevalence of TMPRSS2:ERG fusions in men of African descent implies that alternative genomic mechanisms might explain the disproportionately high prostate cancer burden in such populations.
Collapse
Affiliation(s)
- Cindy Ke Zhou
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Denise Young
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Rockville, Maryland
| | | | - Sally B Coburn
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Yao Tettey
- University of Ghana Medical School, Accra, Ghana
| | | | | | - Evelyn Tay
- University of Ghana Medical School, Accra, Ghana
| | | | | | - Judith Welsh
- NIH Library, National Institutes of Health, Bethesda, Maryland
| | | | - Robert N Hoover
- Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Isabell A Sesterhenn
- Genitourinary Pathology, Joint Pathology Center, Department of Defense, Silver Spring, Maryland
| | - Ann W Hsing
- Stanford Prevention Research Center and Cancer Institute, Palo Alto, California
- Department of Health Research and Policy, Stanford School of Medicine, Palo Alto, California
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Rockville, Maryland
| | - Michael B Cook
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
41
|
Shoag J, Liu D, Blattner M, Sboner A, Park K, Deonarine L, Robinson BD, Mosquera JM, Chen Y, Rubin MA, Barbieri CE. SPOP mutation drives prostate neoplasia without stabilizing oncogenic transcription factor ERG. J Clin Invest 2017; 128:381-386. [PMID: 29202479 PMCID: PMC5749531 DOI: 10.1172/jci96551] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022] Open
Abstract
Nearly 50% of prostate cancers harbor gene fusions that lead to overexpression of the transcription factor ERG, while a mutually exclusive 10% of prostate cancers harbor recurrent mutations in the gene encoding the E3 ubiquitin ligase SPOP. Recent reports suggest that SPOP acts as a ubiquitin ligase for ERG and propose that ERG stabilization is the oncogenic effector of SPOP mutation. Here, we used human prostate cancer samples and showed that the vast majority of human SPOP-mutant cancers do not express ERG. Comparison of SPOP-mutant and ERG-fusion organoid models showed evidence of divergent, rather than common, transcriptional programs. Furthermore, expression of prostate cancer–associated SPOP mutations in genetically engineered mouse models of SPOP-mutant prostate cancer did not result in the expression of ERG protein in histologically normal prostate glands, high-grade prostatic intraepithelial neoplasia, invasive adenocarcinoma, or prostate organoids. In summary, we found no evidence that ERG is an effector of SPOP mutation in human prostate cancer or mouse models.
Collapse
Affiliation(s)
| | - Deli Liu
- Department of Urology, New York Presbyterian Hospital.,HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine.,Sandra and Edward Meyer Cancer Center, and
| | - Mirjam Blattner
- Sandra and Edward Meyer Cancer Center, and.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Andrea Sboner
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA.,The Caryl and Israel Englander Institute for Precision Medicine of Weill Cornell Medicine, and New York-Presbyterian Hospital, New York, New York, USA
| | - Kyung Park
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Brian D Robinson
- Department of Urology, New York Presbyterian Hospital.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA.,The Caryl and Israel Englander Institute for Precision Medicine of Weill Cornell Medicine, and New York-Presbyterian Hospital, New York, New York, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, and.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Mark A Rubin
- Department of Urology, New York Presbyterian Hospital.,Sandra and Edward Meyer Cancer Center, and.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA.,The Caryl and Israel Englander Institute for Precision Medicine of Weill Cornell Medicine, and New York-Presbyterian Hospital, New York, New York, USA.,Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Christopher E Barbieri
- Department of Urology, New York Presbyterian Hospital.,Sandra and Edward Meyer Cancer Center, and.,The Caryl and Israel Englander Institute for Precision Medicine of Weill Cornell Medicine, and New York-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
42
|
Netto GJ, Eich ML, Varambally S. Prostate Cancer: An Update on Molecular Pathology with Clinical Implications. EUR UROL SUPPL 2017. [DOI: 10.1016/j.eursup.2017.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
43
|
Graff RE, Ahearn TU, Pettersson A, Ebot EM, Gerke T, Penney KL, Wilson KM, Markt SC, Pernar CH, Gonzalez-Feliciano AG, Song M, Lis RT, Schmidt DR, Vander Heiden MG, Fiorentino M, Giovannucci EL, Loda M, Mucci LA. Height, Obesity, and the Risk of TMPRSS2:ERG-Defined Prostate Cancer. Cancer Epidemiol Biomarkers Prev 2017; 27:193-200. [PMID: 29167279 DOI: 10.1158/1055-9965.epi-17-0547] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/08/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022] Open
Abstract
Background: The largest molecular subtype of primary prostate cancer is defined by the TMPRSS2:ERG gene fusion. Few studies, however, have investigated etiologic differences by TMPRSS2:ERG status. Because the fusion is hormone-regulated and a man's hormonal milieu varies by height and obesity status, we hypothesized that both may be differentially associated with risk of TMPRSS2:ERG-defined disease.Methods: Our study included 49,372 men from the prospective Health Professionals Follow-up Study. Participants reported height and weight at baseline in 1986 and updated weight biennially thereafter through 2009. Tumor ERG protein expression (a TMPRSS2:ERG marker) was immunohistochemically assessed. We used multivariable competing risks models to calculate HRs and 95% confidence intervals (CIs) for the risk of ERG-positive and ERG-negative prostate cancer.Results: During 23 years of follow-up, we identified 5,847 incident prostate cancers, among which 913 were ERG-assayed. Taller height was associated with an increased risk of ERG-positive disease only [per 5 inches HR 1.24; 95% confidence interval (CI), 1.03-1.50; Pheterogeneity = 0.07]. Higher body mass index (BMI) at baseline (per 5 kg/m2 HR 0.75; 95% CI, 0.61-0.91; Pheterogeneity = 0.02) and updated BMI over time (per 5 kg/m2 HR 0.86; 95% CI, 0.74-1.00; Pheterogeneity = 0.07) were associated with a reduced risk of ERG-positive disease only.Conclusions: Our results indicate that anthropometrics may be uniquely associated with TMPRSS2:ERG-positive prostate cancer; taller height may be associated with greater risk, whereas obesity may be associated with lower risk.Impact: Our study provides strong rationale for further investigations of other prostate cancer risk factors that may be distinctly associated with subtypes. Cancer Epidemiol Biomarkers Prev; 27(2); 193-200. ©2017 AACR.
Collapse
Affiliation(s)
- Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California. .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Thomas U Ahearn
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ericka M Ebot
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Travis Gerke
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kathryn M Wilson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sarah C Markt
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Claire H Pernar
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Mingyang Song
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rosina T Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daniel R Schmidt
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Harvard Radiation Oncology Program, Harvard Medical School, Boston, Massachusetts
| | - Matthew G Vander Heiden
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Lotan TL, Torres A, Zhang M, Tosoian JJ, Guedes LB, Fedor H, Hicks J, Ewing CM, Isaacs SD, Johng D, De Marzo AM, Isaacs WB. Somatic molecular subtyping of prostate tumors from HOXB13 G84E carriers. Oncotarget 2017; 8:22772-22782. [PMID: 28186998 PMCID: PMC5410261 DOI: 10.18632/oncotarget.15196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/21/2017] [Indexed: 11/25/2022] Open
Abstract
A recurrent germline mutation (G84E) in the HOXB13 gene is associated with early onset and family history-positive prostate cancer in patients of European descent, occurring in up to 5% of prostate cancer families. To date, the molecular features of prostate tumors occurring in HOXB13 G84E carriers have not been studied in a large cohort of patients. We identified 101 heterozygous carriers of G84E who underwent radical prostatectomy for prostate cancer between 1985 and 2011 and matched these men by race, age and tumor grade to 99 HOXB13 wild-type controls. Immunostaining for HOXB13, PTEN, ERG, p53 and SPINK1 as well as RNA in situ hybridization for ETV1/4/5 were performed using genetically validated assays. Tumors from G84E carriers generally expressed HOXB13 protein at a level comparable to benign and wild-type glands. ETS gene expression (either ERG or ETV1/4/5) was seen in 36% (36/101) of tumors from G84E carriers compared to 68% (65/96) of the controls (p < 0.0001). PTEN was lost in 11% (11/101) of G84E carriers compared to 25% (25/99) of the controls (p = 0.014). PTEN loss was enriched among ERG-positive compared to ERG-negative tumors in both groups of patients. Nuclear accumulation of the p53 protein, indicative of underlying TP53 missense mutations, was uncommon in both groups, occurring in 1% (1/101) of the G84E carriers versus 2% (2/92) of the controls (p = NS). Taken together, these data suggest that genes other than ERG and PTEN may drive carcinogenesis/progression in the majority of men with germline HOXB13 mutations.
Collapse
Affiliation(s)
- Tamara L Lotan
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alba Torres
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Miao Zhang
- Departments of Pathology, MD Anderson Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeffrey J Tosoian
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liana B Guedes
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helen Fedor
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jessica Hicks
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles M Ewing
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah D Isaacs
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dorhyun Johng
- Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - William B Isaacs
- Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Suh JH, Park JH, Lee C, Moon KC. Intrafocal heterogeneity of ERG protein expression and gene fusion pattern in prostate cancer. Prostate 2017; 77:1438-1445. [PMID: 28845585 DOI: 10.1002/pros.23405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/08/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Prostate cancer is considered to be highly heterogeneous, with various morphologic features and biologic behaviors. The TMPRSS2-ERG gene fusion is the most frequently observed genetic aberration in prostate cancer. The aim of this study was to elucidate the intrafocal heterogeneity of ERG gene fusion status. METHODS ERG immunohistochemistry (IHC) was performed in samples from 168 prostate cancer patients who had undergone radical prostatectomy, and 40 cases showing ERG-positive IHC staining were selected for tissue microarray (TMA) construction. Two to six representative cores were selected from each tumor focus. In the cases with heterogeneous ERG IHC staining intensity, the areas showing different intensities were separately selected. Using the TMA blocks, IHC and fluorescence in situ hybridization (FISH) were conducted to evaluate the heterogeneity of ERG protein expression and ERG fusion gene patterns, respectively, in a single tumor focus. Heterogeneity of ERG IHC staining was defined as the simultaneous presence of negative and positive cores in the same tumor focus. Heterogeneity of ERG FISH was defined by the presence of cores with positive and negative FISH signals or cores with break-apart and interstitial deletion FISH signals in the same tumor focus. RESULTS A total of 202 TMA cores were isolated from 40 ERG-positive cases. Of the 202 total cores, 19 were negative for ERG IHC staining, and 46 showed 1+, 52 showed 2+, and 85 showed 3+ ERG staining intensity. Eleven cores were negative for ERG FISH signal, 119 cores showed ERG break-apart FISH signals, and the remaining 72 cores revealed interstitial deletion. Intrafocal heterogeneity of ERG IHC staining was found in 20% (8/40) of cases, and intrafocal heterogeneity of ERG gene fusion pattern was found in 32.5% (13/40) of cases. CONCLUSIONS In summary, this study showed significantly frequent intrafocal heterogeneity of ERG protein expression, gene fusion status and fusion pattern. This heterogeneity can be caused by the development of subclones during cancer progression or the intermingling of different tumors.
Collapse
Affiliation(s)
- Ja Hee Suh
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Hwan Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Woo JS, Reddy OL, Koo M, Xiong Y, Li F, Xu H. Application of Immunohistochemistry in the Diagnosis of Pulmonary and Pleural Neoplasms. Arch Pathol Lab Med 2017. [PMID: 28644685 DOI: 10.5858/arpa.2016-0550-ra] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - A vast majority of neoplasms arising from lung or pleura are initially diagnosed based on the histologic evaluation of small transbronchial, endobronchial, or needle core biopsies. Although most diagnoses can be determined by morphology alone, immunohistochemistry can be a valuable diagnostic tool in the workup of problematic cases. OBJECTIVE - To provide a practical approach in the interpretation and immunohistochemical selection of lung/pleura-based neoplasms obtained from small biopsy samples. DATA SOURCES - A literature review of previously published articles and the personal experience of the authors were used in this review article. CONCLUSION - Immunohistochemistry is a useful diagnostic tool in the workup of small biopsies from the lung and pleura sampled by small biopsy techniques.
Collapse
|
47
|
Hernández-Llodrà S, Juanpere N, de Muga S, Lorenzo M, Gil J, Font-Tello A, Agell L, Albero-González R, Segalés L, Merino J, Serrano L, Fumadó L, Cecchini L, Lloreta-Trull J. ERG overexpression plus SLC45A3 (prostein) and PTEN expression loss: Strong association of the triple hit phenotype with an aggressive pathway of prostate cancer progression. Oncotarget 2017; 8:74106-74118. [PMID: 29088771 PMCID: PMC5650326 DOI: 10.18632/oncotarget.18266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 05/15/2017] [Indexed: 12/18/2022] Open
Abstract
TMPRSS2 and SLC45A3 rearrangements may coexist in the same tumor. ERG rearrangements and PTEN loss are concomitant events in prostate cancer (PrCa), and can cooperate in progression. We have reported that mRNA expression of TMPRSS2-ERG and SLC45A3-ERG rearrangements plus PTEN loss define an aggressive tumor subset. The aim of this study has been to validate these results by immunohistochemistry in a large cohort of tumors. ERG, SLC45A3 and PTEN immunostaining and their association with pathological features and PSA progression-free survival were analyzed in 220 PrCa (PSMAR-Biobank, Barcelona, Spain). ERG protein expression was found in 46.8% and SLC45A3 and PTEN loss in 30% and 34% tumors, respectively. Single ERG positive immunostaining was associated with GS = 6 tumors (p = 0.016), double ERG+/PTEN loss with GS = 7 (p = 0.008) and Grade Group 2 (GG) or GG3 cases (p = 0.042), ERG+/SLC45A3 loss/PTEN loss ("triple hit") with GS ≥ 8 (p < 0.0001) and GG4 or GG5 tumors (p = 0.0003). None of GS = 6 nor = GG1 cases showed this combination. In the GS ≥ 8 group, ERG+ (p = 0.002), PTEN loss (p = 0.009) and "triple hit" (p = 0.003) were associated with Gleason pattern 3 component, and single SLC45A3 loss (p = 0.036) with GS ≥ 8 without pattern 3. The number of aberrant events and the triple hit were strongly associated with shorter PSA progression-free survival. In GS = 6 PrCa, single ERG+ was also associated with progression. ERG+ identifies a distinct pathway of PrCa. Additional assessment of PTEN and SLC45A3 adds relevant prognostic information. The triple hit phenotype (ERG+/SLC45A3 loss/PTEN loss) is associated with progression and could be used for patient stratification, treatment and follow-up.
Collapse
Affiliation(s)
| | - Nuria Juanpere
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Silvia de Muga
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Lorenzo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Joan Gil
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laia Agell
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laura Segalés
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Laia Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Fumadó
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Lluís Cecchini
- Department of Urology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| | - Josep Lloreta-Trull
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,Department of Pathology, Hospital del Mar-Parc de Salut Mar-IMIM, Barcelona, Spain
| |
Collapse
|
48
|
Mancarella C, Casanova-Salas I, Calatrava A, García-Flores M, Garofalo C, Grilli A, Rubio-Briones J, Scotlandi K, López-Guerrero JA. Insulin-like growth factor 1 receptor affects the survival of primary prostate cancer patients depending on TMPRSS2-ERG status. BMC Cancer 2017; 17:367. [PMID: 28545426 PMCID: PMC5445474 DOI: 10.1186/s12885-017-3356-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 05/15/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is characterized by clinical and biological heterogeneity and has differential outcomes and mortality rates. Therefore, it is necessary to identify molecular alterations to define new therapeutic strategies based on the risk of progression. In this study, the prognostic relevance of the insulin-like growth factor (IGF) system was examined in molecular subtypes defined by TMPRSS2-ERG (T2E) gene fusion within a series of patients with primary localized PCa. METHODS A cohort of 270 formalin-fixed and paraffin-embedded (FFPE) primary PCa samples from patients with more than 5 years' follow-up was collected. IGF-1R, IGF-1, IGFBP-3 and INSR expression was analyzed using quantitative RT-PCR. The T2E status and immunohistochemical ERG findings were considered in the analyses. The association with both biochemical and clinical progression-free survival (BPFS and PFS, respectively) was evaluated for the different molecular subtypes using the Kaplan-Meier proportional risk log-rank test and the Cox proportional hazards model. RESULTS An association between IGF-1R overexpression and better BPFS was found in T2E-negative patients (35.3% BPFS, p-value = 0.016). Multivariate analysis demonstrated that IGF-1R expression constitutes an independent variable in T2E-negative patients [HR: 0.41. CI 95% (0.2-0.82), p = 0.013]. These data were confirmed using immunohistochemistry of ERG as subrogate of T2E. High IGF-1 expression correlated with prolonged BPFS and PFS independent of the T2E status. CONCLUSIONS IGF-1R, a reported target of T2E, constitutes an independent factor for good prognosis in T2E-negative PCa. Quantitative evaluation of IGF-1/IGF-1R expression combined with molecular assessment of T2E status or ERG protein expression represents a useful marker for tumor progression in localized PCa.
Collapse
Affiliation(s)
- Caterina Mancarella
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Irene Casanova-Salas
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, C/ Prof. Beltrán Báguena, 8, 46009 Valencia, Spain
| | - Ana Calatrava
- Department of Pathology, Fundación Instituto Valenciano de Oncología, C/ Prof. Beltrán Báguena, 8, 46009 Valencia, Spain
| | - Maria García-Flores
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, C/ Prof. Beltrán Báguena, 8, 46009 Valencia, Spain
| | - Cecilia Garofalo
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Andrea Grilli
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, via di Barbiano, 1/10, 40136 Bologna, Italy
| | - José Rubio-Briones
- Department of Urology, Fundación Instituto Valenciano de Oncología, C/ Prof. Beltrán Báguena, 8, 46009 Valencia, Spain
| | - Katia Scotlandi
- CRS Development of Biomolecular Therapies, Experimental Oncology Laboratory, Rizzoli Orthopedic Institute, via di Barbiano, 1/10, 40136 Bologna, Italy
| | - José Antonio López-Guerrero
- Laboratory of Molecular Biology, Fundación Instituto Valenciano de Oncología, C/ Prof. Beltrán Báguena, 8, 46009 Valencia, Spain
| |
Collapse
|
49
|
Torres A, Alshalalfa M, Tomlins SA, Erho N, Gibb EA, Chelliserry J, Lim L, Lam LLC, Faraj SF, Bezerra SM, Davicioni E, Yousefi K, Ross AE, Netto GJ, Schaeffer EM, Lotan TL. Comprehensive Determination of Prostate Tumor ETS Gene Status in Clinical Samples Using the CLIA Decipher Assay. J Mol Diagn 2017; 19:475-484. [PMID: 28341589 PMCID: PMC5417038 DOI: 10.1016/j.jmoldx.2017.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 01/30/2017] [Indexed: 12/19/2022] Open
Abstract
ETS family gene fusions are common in prostate cancer and molecularly define a tumor subset. ERG is the most commonly rearranged, leading to its overexpression, followed by ETV1, ETV4, and ETV5, and these alterations are generally mutually exclusive. We validated the Decipher prostate cancer assay to detect ETS alterations in a Clinical Laboratory Improvement Amendments-accredited laboratory. Benchmarking against ERG immunohistochemistry and ETV1/4/5 RNA in situ hybridization, we examined the accuracy, precision, and reproducibility of gene expression ETS models using formalin-fixed, paraffin-embedded samples. The m-ERG model achieved an area under curve of 95%, with 93% sensitivity and 98% specificity to predict ERG immunohistochemistry status. The m-ETV1, -ETV4, and -ETV5 models achieved areas under curve of 98%, 88%, and 99%, respectively. The models had 100% robustness for ETS status, and scores were highly correlated across sample replicates. Models predicted 41.5% of a prospective radical prostatectomy cohort (n = 4036) to be ERG+, 6.3% ETV1+, 1% ETV4+, and 0.4% ETV5+. Of prostate tumor biopsy samples (n = 509), 41.2% were ERG+, 8.6% ETV1+, 0.4% ETV4+, and none ETV5+. Higher Decipher risk status tumors were more likely to be ETS+ (ERG or ETV1/4/5) in the radical prostatectomy and the biopsy cohorts (P < 0.05). These results support the utility of microarray-based ETS status prediction models for molecular classification of prostate tumors.
Collapse
Affiliation(s)
- Alba Torres
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Scott A Tomlins
- Department of Pathology, University of Michigan, Ann Arbor, Michigan; Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Nicholas Erho
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Ewan A Gibb
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | | | - Lony Lim
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Lucia L C Lam
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Sheila F Faraj
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stephania M Bezerra
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Elai Davicioni
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Kasra Yousefi
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Ashley E Ross
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - George J Netto
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Edward M Schaeffer
- Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Urology, Northwestern University, Chicago, Illinois
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
50
|
Graff RE, Judson G, Ahearn TU, Fiorentino M, Loda M, Giovannucci EL, Mucci LA, Pettersson A. Circulating Antioxidant Levels and Risk of Prostate Cancer by TMPRSS2:ERG. Prostate 2017; 77:647-653. [PMID: 28102015 PMCID: PMC5354965 DOI: 10.1002/pros.23312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/04/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Few studies have considered etiological differences across molecular subtypes of prostate cancer, despite potential to improve opportunities for precision prevention of a disease for which modifiable risk factors have remained elusive. Factors that lead to DNA double-strand breaks, such as oxidative stress, may promote the formation of the TMPRSS2:ERG gene fusion in prostate cancer. We tested the hypothesis that increasing levels of pre-diagnostic circulating antioxidants, which may reduce oxidative stress, are associated with lower risk of developing TMPRSS2:ERG positive prostate cancer. METHODS We conducted a nested case-control study, including 370 cases and 2,470 controls, to evaluate associations between pre-diagnostic α- and β-carotene, α- and γ-tocopherol, β-cryptoxanthin, lutein, lycopene, retinol, and selenium with the risk of prostate cancer by ERG protein expression status (a marker of TMPRSS2:ERG). Multivariable unconditional polytomous logistic regression was used to calculate odds ratios and 95% confidence intervals. RESULTS We did not find any of the antioxidants to be significantly associated with the risk of prostate cancer according to ERG status. CONCLUSIONS The results do not support the hypothesis that circulating pre-diagnostic antioxidant levels protect against developing TMPRSS2:ERG positive prostate cancer. Additional studies are needed to explore mechanisms for the development of TMPRSS2:ERG positive disease. Prostate 77: 647-653, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rebecca E. Graff
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Gregory Judson
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Thomas U. Ahearn
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michelangelo Fiorentino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Massimo Loda
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Andreas Pettersson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|