1
|
Schoots IG, Ahmed HU, Albers P, Asbach P, van den Bergh RCN, Godtman RA, van Leeuwen PJ, Nordström T, Punwani S, Wallström J, Padhani AR. Magnetic Resonance Imaging-based Biopsy Strategies in Prostate Cancer Screening: A Systematic Review. Eur Urol 2025:S0302-2838(25)00341-0. [PMID: 40514255 DOI: 10.1016/j.eururo.2025.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/15/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND AND OBJECTIVE Prostate cancer (PCa) screening using prostate-specific antigen (PSA) thresholding and systematic biopsies reduces advanced disease presentations and cancer-specific mortality, but also leads to overdiagnosis. Magnetic resonance imaging (MRI) integration may maintain screening benefits, while reducing overdiagnosis and unnecessary biopsies. This review analyses the benefit-harm balance when MRI is integrated as first-line and second-stage (after PSA >3 ng/ml) test in PCa screening. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines, we performed a PROSPERO-registered systematic review (CRD420251006926). Literature searches identified five first-line and four second-stage MRI screening studies. We assessed MRI strategies (first-line/second-stage and risk thresholds), biopsy avoidance, and biopsy methods (targeted/systematic) for histological outcomes (grade group [GG] ≥2/GG 1 cancer detection and benign biopsies). Benefit-to-harm ratios of >1 suggest a positive net benefit. KEY FINDINGS AND LIMITATIONS First-line MRI screening detects twice as many men with GG ≥2 cancer as second-stage MRI screening but has more MRI-negative men (range, 66-89% vs 56-61%). Second-stage MRI significantly reduced biopsy rates (range, 42-79%) compared with systematic biopsy rates in all PSA-positive men. Subsequently, GG ≥2/GG 1 cancer detection ratios increased in MRI-positive men undergoing targeted and systematic biopsies (range, 1.9-6.2) and targeted biopsies alone (range, 1.8-7.0), compared with systematic biopsies alone (range, 0.8-1.4). First-line and second-stage MRI screening allowed biopsy avoidance in three to 125 and two to 15 men, respectively, for each benign diagnosis. All benefit-to-harm ratios showed positive net benefits (>1). Heterogeneity in the study protocols limits generalisability. CONCLUSIONS AND CLINICAL IMPLICATIONS Targeted biopsies in second-stage MRI screening optimise clinically significant PCa detection, while reducing the number of biopsies. First-line MRI screening requires further assessments of its feasibility. PCa screening quality assurance requires standardised MRI interpretations and biopsy protocols.
Collapse
Affiliation(s)
- Ivo G Schoots
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Center of Early Diagnosis, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Hashim U Ahmed
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, UK; Imperial Urology, Division of Cancer, Cardiovascular Medicine and Surgery, Imperial College Healthcare NHS Trust, London, UK
| | - Peter Albers
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany; Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Asbach
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Rebecka A Godtman
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Urology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pim J van Leeuwen
- Center of Early Diagnosis, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tobias Nordström
- Department of Urology and Clinical Sciences, Danderyd Hospital, Danderyd, Sweden; Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Shonit Punwani
- Centre for Medical Imaging, University College London, London, UK
| | - Jonas Wallström
- Department of Radiology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| |
Collapse
|
2
|
Beatrici E, De Carne F, Frego N, Moretto S, Paciotti M, Fasulo V, Uleri A, Garofano G, Avolio PP, Chiarelli G, Contieri R, Arena P, Saitta C, Sordelli F, Saita A, Hurle R, Casale P, Buffi N, Lazzeri M, Lughezzani G. Optimizing Prostate Cancer Diagnostic Work-Up Through Micro-Ultrasound: Minimizing Unnecessary Procedures and Reducing Overdiagnoses. Prostate 2025; 85:603-611. [PMID: 39876544 PMCID: PMC11934833 DOI: 10.1002/pros.24862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/08/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION We aim to critically assess Microultrasound (mUS) clinical performance in an outpatient setting, focusing on its ability to reduce unnecessary diagnostic procedures, potentially reshape prostate cancer (PCa) diagnostic protocols, and increase the ability to rule out clinically significant (Gleason Score ≥ 3 + 4) PCa (csPCa). MATERIALS AND METHODS Between November 2018 and April 2022, we conducted a prospective study involving men who underwent mUS examination due to clinical symptoms, PSA elevation, or opportunistic early detection of PCa. Experienced urologists performed mUS assessments in an outpatient setting using the prostate risk identification using micro-ultrasound (PRI-MUS) protocol to identify lesions suspicious of csPCa (PRI-MUS score ≥ 3). Men with negative mUS results were followed through consistent phone follow-up calls and visits until October 2023 to assess their diagnostic and therapeutic pathways. Using Cox regression models adjusted for PSA levels, DRE results, age, and previous biopsy history, we calculated the hazard ratio (HR) for biopsy-free (BFS), defined as the time from mUS to biopsy or last follow-up, cancer-free survival (CFS), and clinically significant cancer-free survival (csCFS) within the cohort based on mUS results. RESULTS Overall, 425 men were enrolled. The median (IQR) age was 66 (59-72) years, PSA levels were 5.7 (4.0-7.9) ng/mL, prostate volume was 44 (31.5-62.1) mL, and the median follow-up was 39 months (27-53). mUS identified lesions suggesting csPCa in 201/425 (47.3%) men. Overall, mUS resulted negative in 224/425 (52.7%) men, of whom 207/224 (92.4%) did not undergo subsequent mpMRI, while 22/224 (9.8%) proceeded with mpMRI according to the referring physician's decision. The latter detected suspicious lesions in 12/22 cases (54.5%), but only 2/12 (16.7%) were confirmed by biopsy as csPCa. Among those with negative mUS results, 192/224 (85.7%) men avoided additional biopsies during follow-up. Men with negative mUS results exhibited superior BFS (aHR: 0.17; p < 0.001), CFS (aHR:0.12; p < 0.001), and csCFS (aHR:0.09; p < 0.001) survival rates compared to their mUS-positive counterparts. CONCLUSIONS Our findings suggest that mUS can potentially refine patient stratification and transform PCa screening and diagnostic protocols. Pending validation by other studies, a wider implementation of mUS could optimize resource allocation, minimize wastage, and reserve additional costly tests.
Collapse
Affiliation(s)
- Edoardo Beatrici
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Fabio De Carne
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Nicola Frego
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Stefano Moretto
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Marco Paciotti
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Vittorio Fasulo
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Alessandro Uleri
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Giuseppe Garofano
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Pier Paolo Avolio
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Giuseppe Chiarelli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Roberto Contieri
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Paola Arena
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Cesare Saitta
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Federica Sordelli
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Alberto Saita
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Rodolfo Hurle
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Paolo Casale
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - NicolòMaria Buffi
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Massimo Lazzeri
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| | - Giovanni Lughezzani
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleItaly
- Department of UrologyIRCCS Humanitas Research HospitalRozzanoItaly
| |
Collapse
|
3
|
Shore N, Armstrong AJ, Barata P, Byrne L, Hafron J, Young S, Paller C, Wise DR, Ventii K, Samadi A, Arangua P, Werahera PN, Lorentz J. Implementing and Optimizing Universal Germline Genetic Testing for Patients With Prostate Cancer in Clinical Practice. Urology 2025; 199:1-10. [PMID: 39909130 DOI: 10.1016/j.urology.2025.01.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE To advocate for universal germline genetic testing (UGGT) in prostate cancer and provide practical recommendations for its implementation. METHODS Although guidelines for germline genetic testing in prostate cancer have progressed, usage remains limited and inconsistent due to barriers including access, cost, and variable guideline adherence. These issues prevent some patients with germline pathogenic/likely pathogenic variants from benefiting from risk assessment, precision therapies (eg, PARP inhibitors, PD-1 inhibitors), and potential clinical trials. Despite these benefits, studies indicate that germline genetic testing use remains low, especially in prostate cancer care. The PROCLAIM trial (Shore et al, 2023) highlighted that nearly half of patients with pathogenic variants are missed under National Comprehensive Cancer Network guidelines, particularly impacting non-white patients and those with incomplete family history data. Additional racial and socioeconomic disparities further hinder access and variant interpretation accuracy. Given these challenges, UGGT for all prostate cancer patients has been proposed to improve care equity and decision-making. In March 2024, prostate cancer experts convened to discuss strategies for UGGT implementation. RESULTS The outcome of that meeting includes recommendations for integrating UGGT into oncology and urology practices and have been outlined in this paper. CONCLUSION To maximize the benefits while mitigating the potential risks of UGGT, it is essential to address implementation details, including careful gene panel selection, variants of uncertain significance reporting and management, appropriate genetics follow-up, and seamless integration of test reports into electronic medical records for accessibility by patients and providers.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC.
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Department of Medicine, Division of Medical Oncology, Duke University, Durham, NC
| | - Pedro Barata
- Division of Solid Tumor Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
| | - Lindsey Byrne
- Division of Human Genetics, Department of Internal Medicine, Comprehensive Cancer Center, College of Medicine, The Ohio State University, Werner Medical Center, Columbus, OH
| | | | - Sarah Young
- Labcorp Genetics Inc (formerly Invitae Corp), San Francisco, CA
| | - Channing Paller
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - David R Wise
- Department of Medicine, Laura & Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Karen Ventii
- Prostate Conditions Education Council, Centennial, CO
| | - Ali Samadi
- Prostate Conditions Education Council, Centennial, CO
| | - Paul Arangua
- Prostate Conditions Education Council, Centennial, CO
| | | | - Justin Lorentz
- Sunnybrook Health Sciences Center, University of Toronto, Ontario, Canada
| |
Collapse
|
4
|
Vatrano S, Pepe P, Pepe L, Vella N, Alario C, Chiarandà A, Taranto C, Scillieri R, Mauceri C, Fraggetta F. BRCA mutations and prostate cancer: should urologist improve daily clinical practice? Arch Ital Urol Androl 2025:13635. [PMID: 40247739 DOI: 10.4081/aiua.2025.13635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 04/19/2025] Open
Abstract
INTRODUCTION To evaluate BRCA1-2 (breast cancer) detection in men with high risk PCa, including the oncological consequences for the patient and family members. MATERIALS AND METHODS From January 2023 to December 2024, 52 men (median age 73 years;) with confirmed PCa diagnosis underwent somatic and germline BRCA1 and BRCA2 assessment; 11/52 (21%) patients documented a family history of cancer. Patients were at different clinical stages: high-grade (71% had a Gleason score ≥ 8), locally advanced (54% of cases) and/or metastatic PCa (46% of cases) at initial diagnosis, hormone-sensitive and/or castration-resistant PCa (38.2% of cases) at clinical progression. Formalin-fixed paraffin-embedded (FFPE) tissues and next generation sequencing (NGS) analyses of BRCA genes were evaluated on 52 samples (prostate biopsies or definitive samples) collected at Gravina Hospital (Caltagirone, Italy) from different Sicilian pathology departments. The therapeutic and clinical impact of genetic testing for BRCA somatic and germline mutations was evaluated for patients and their families. RESULTS All FFPE cases were successfully genotyped, with a good library and sequencing CQ metrics for all genes of interest; 10/52 (19.2%) patients had somatic or germline BRCA mutations, specifically, 3/52 (5.7%) had somatic and 7/52 (13.5%) had germline mutations. In the seven cases with germline variants, 4/7 (57%) had a family history of PCa or other diseases, while the remaining 3/7 (43%) patients had no hereditary predisposition. All identified genetic variants were related to the BRCA2 gene; after genetic screening of the corresponding relatives, various members of the analysed families carried the mutation identified in the proband, so that cancer prevention and/or active surveillance was possible. CONCLUSIONS NGS analysis for BRCA genetic testing using FFPE tissue in the clinical setting of patients with high-grade and/or metastatic PCa appears to be a valuable tool, not only for therapeutic purposes, but also to identify families with genetic predisposition who may be underdiagnosed according to canonical criteria.
Collapse
|
5
|
McHugh JK, Bancroft EK, Saunders E, Brook MN, McGrowder E, Wakerell S, James D, Rageevakumar R, Benton B, Taylor N, Myhill K, Hogben M, Kinsella N, Sohaib AA, Cahill D, Hazell S, Withey SJ, Mcaddy N, Page EC, Osborne A, Benafif S, Jones AB, Patel D, Huang DY, Kaur K, Russell B, Nicholson R, Croft F, Sobczak J, McNally C, Mutch F, Bennett S, Kingston L, Karlsson Q, Dadaev T, Saya S, Merson S, Wood A, Dennis N, Hussain N, Thwaites A, Hussain S, Rafi I, Ferris M, Kumar P, James ND, Pashayan N, Kote-Jarai Z, Eeles RA. Assessment of a Polygenic Risk Score in Screening for Prostate Cancer. N Engl J Med 2025; 392:1406-1417. [PMID: 40214032 PMCID: PMC7617604 DOI: 10.1056/nejmoa2407934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
BACKGROUND The incidence of prostate cancer is increasing. Screening with an assay of prostate-specific antigen (PSA) has a high rate for false positive results. Genomewide association studies have identified common germline variants in persons with prostate cancer, which can be used to calculate a polygenic risk score associated with risk of prostate cancer. METHODS We recruited persons 55 to 69 years of age from primary care centers in the United Kingdom. Using germline DNA extracted from saliva, we derived polygenic risk scores from 130 variants known to be associated with an increased risk of prostate cancer. Participants with a polygenic risk score in the 90th percentile or higher were invited to undergo prostate cancer screening with multiparametric magnetic resonance imaging (MRI) and transperineal biopsy, irrespective of PSA level. RESULTS Among 40,292 persons invited to participate, 8953 (22.2%) expressed interest in participating and 6393 had their polygenic risk score calculated; 745 (11.7%) had a polygenic risk score in the 90th percentile or higher and were invited to undergo screening. Of these 745 participants, 468 (62.8%) underwent MRI and prostate biopsy; prostate cancer was detected in 187 participants (40.0%). The median age at diagnosis was 64 years (range, 57 to 73). Of the 187 participants with cancer, 103 (55.1%) had prostate cancer classified as intermediate or higher risk according to the 2024 National Comprehensive Cancer Network (NCCN) criteria, so treatment was indicated; cancer would not have been detected in 74 (71.8%) of these participants according to the prostate cancer diagnostic pathway currently used in the United Kingdom (high PSA level and positive MRI results). In addition, 40 of the participants with cancer (21.4%) had disease classified as unfavorable intermediate risk or as high or very high risk according to NCCN criteria. CONCLUSIONS In a prostate cancer screening program involving participants in the top decile of risk as determined by a polygenic risk score, the percentage found to have clinically significant disease was higher than the percentage that would have been identified with the use of PSA or MRI. (Funded by the European Research Council Seventh Framework Program and others; BARCODE1 ClinicalTrials.gov number, NCT03857477.).
Collapse
Affiliation(s)
- Jana K McHugh
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | | | | | | | | | | | - Denzil James
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | | | - Barbara Benton
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Natalie Taylor
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Kathryn Myhill
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Matthew Hogben
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Netty Kinsella
- Royal Marsden NHS Foundation Trust, London
- Translational Oncology and Urology Research Center for Cancer, Society, and Public Health, King's College London, London
| | | | | | | | | | | | - Elizabeth C Page
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Andrea Osborne
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Sarah Benafif
- Institute of Cancer Research, London
- University College London Hospitals NHS Foundation Trust, London
| | - Ann-Britt Jones
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | - Dhruv Patel
- InHealth Limited, InHealth Group HQ, High Wycombe, United Kingdom
| | - Dean Y Huang
- InHealth Limited, InHealth Group HQ, High Wycombe, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | - Sibel Saya
- Institute of Cancer Research, London
- University of Melbourne, Melbourne, VIC, Australia
| | | | | | - Nening Dennis
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | | | | | | | - Imran Rafi
- St. George's University of London, London
| | | | | | - Nicholas D James
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| | | | | | - Rosalind A Eeles
- Institute of Cancer Research, London
- Royal Marsden NHS Foundation Trust, London
| |
Collapse
|
6
|
Al-Monajjed R, Albers P, Hadaschik B. Screening for Prostate Cancer with Prostate-specific Antigen. EUR UROL SUPPL 2025; 74:71-72. [PMID: 40225289 PMCID: PMC11992575 DOI: 10.1016/j.euros.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 04/15/2025] Open
Affiliation(s)
- Rouvier Al-Monajjed
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Albers
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany
| |
Collapse
|
7
|
Rosino Sánchez A, García Torralba E, Girela Baena E, Macías Cerrolaza JA, Tudela Pallares J, Zafra Povés M, Barceló Bayonas I, Muñoz Guillermo V, Fernández Aparicio T. Early diagnosis of prostate cancer in healthy men with germline mutations in DNA Damage Response (DDR) pathways: A literature review and proposal for a screening strategy. Actas Urol Esp 2025; 49:501686. [PMID: 39947293 DOI: 10.1016/j.acuroe.2025.501686] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 02/19/2025]
Abstract
INTRODUCTION Men with mutations in DNA damage response (DDR) pathways have a higher risk of developing prostate neoplasia compared to the general population. The best studied alterations are mutations in BRCA1/2, ATM and MMR-Lynch syndrome. MATERIAL AND METHODS A review of the clinical and prognostic implications of mutations in DDR pathways, as well as an evaluation of the different screening strategies available for affected patients. OBJECTIVE To propose an early diagnostic strategy for men with mutations in DDR pathways. RESULTS Current guidelines do not provide clear, specific recommendations for this subgroup of men. Among mutations in the MMR pathway, the germline MSH2 mutation is most strongly associated with prostate cancer. Men with germline mutations in BRCA1/2, ATM, and MSH2 have a higher incidence of prostate neoplasia, tend to develop the disease at a younger age, and are more likely to have aggressive forms of the disease. Furthermore, men with BRCA1/2 mutations have a lower cancer-specific survival rate compared to the general population. In these patients, PSA levels have important limitations in detecting prostate cancer. Multiparametric MRI of the prostate may be more effective than periodic PSA testing. CONCLUSIONS Patients with mutations in DDR pathways are at increased risk for aggressive prostate neoplasms and require earlier and more intensive screening. PSA-based screening has notable limitations. A screening strategy incorporating multiparametric MRI could offer a more effective strategy for this patient group.
Collapse
Affiliation(s)
| | - E García Torralba
- Unidad de Consejo Genético, Hospital Morales Meseguer, Murcia, Spain
| | - E Girela Baena
- Servicio de Radiología, Hospital Morales Meseguer, Murcia, Spain
| | | | - J Tudela Pallares
- Servicio de Anatomía Patológica, Hospital Morales Meseguer, Murcia, Spain
| | - M Zafra Povés
- Unidad de Consejo Genético, Hospital Morales Meseguer, Murcia, Spain
| | | | | | | |
Collapse
|
8
|
Presutti S, Panio E, Nero C, Iacovelli R, Tagliaferri L, Campetella M, Sacco E, Pallotta G, Rocco B, Sighinolfi MC. BRCA2 and genetic mutations in prostate cancer: an updated practical guide on "when and how" testing across international guidelines. Minerva Urol Nephrol 2025; 77:152-155. [PMID: 40298342 DOI: 10.23736/s2724-6051.25.06395-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Affiliation(s)
- Simona Presutti
- Department of Urological Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy -
- Sacred Heart Catholic University, Rome, Italy -
| | - Enrico Panio
- Department of Urological Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Sacred Heart Catholic University, Rome, Italy
| | - Camilla Nero
- Sacred Heart Catholic University, Rome, Italy
- Department of Woman, Child and Public Health, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Roberto Iacovelli
- Sacred Heart Catholic University, Rome, Italy
- Department of Oncology, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Luca Tagliaferri
- Sacred Heart Catholic University, Rome, Italy
- Radiation Oncology Ward Unit, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
| | - Marco Campetella
- Department of Urology, Isola Tiberina-Gemelli Isola Hospital, Rome, Italy
| | - Emilio Sacco
- Sacred Heart Catholic University, Rome, Italy
- Department of Urology, Isola Tiberina-Gemelli Isola Hospital, Rome, Italy
| | - Giuseppe Pallotta
- Department of Urological Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Sacred Heart Catholic University, Rome, Italy
| | - Bernardo Rocco
- Department of Urological Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Sacred Heart Catholic University, Rome, Italy
| | - Maria C Sighinolfi
- Department of Urological Sciences, IRCCS A. Gemelli University Polyclinic Foundation, Rome, Italy
- Sacred Heart Catholic University, Rome, Italy
| |
Collapse
|
9
|
Fasulo V, Chiarelli G, Garofano G, Ripamonti CB, Barile M, Bianchi P, Morenghi E, Benetti A, Aljoulani M, Finocchiaro A, Paciotti M, Avolio PP, Beatrici E, Arena P, Saita A, Hurle R, Maura F, Da Rin G, Asselta R, Capalbo A, Soldà G, Casale P, Buffi NM, Lughezzani G, Lazzeri M. Impact of prostate cancer screening in European ancestry un-affected men with germline DNA repair pathogenic variants. BJUI COMPASS 2025; 6:BCO2424. [PMID: 40066468 PMCID: PMC11891281 DOI: 10.1002/bco2.424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/13/2024] [Indexed: 03/21/2025] Open
Abstract
Background and Objective Prostate cancer (PCa) is a significant global health concern, ranking as the second most prevalent cancer among men worldwide. Genetic factors, particularly germline pathogenic variants (PVs) in DNA repair genes (DRGs), play a crucial role in PCa predisposition. Our study aimed to assess patients' adherence to a targeted PCa screening program targeting high-risk individuals with DRG PVs and evaluate the potential reduction in biopsy and MRI rates by employing our screening protocol. Methods We conducted a prospective ongoing trial evaluating targeted PCa screening in men with documented PVs in DRGs. Screening involved annual assessment of medical history, physical examination, prostate-specific antigen (PSA) testing, Prostate Health Index (PHI), and multiparametric magnetic resonance imaging (mpMRI) when indicated. Descriptive statistics were used to analyse patient characteristics, and adherence to screening was evaluated at three time points: baseline (T0), one year (T1), and two years (T2) from enrolment. Key Findings and Limitations A total of 101 high-risk individuals were enrolled, with a median age of 52 years. Adherence to screening was high, with 72.3% of patients attending the first annual follow-up (T1) and 100% attending the second follow-up (T2). Despite elevated PSA levels in some patients, no PCa was detected during the study period. However, our screening protocol demonstrated the potential in reducing unnecessary biopsies and MRIs, particularly in patients with elevated PSA but low PHI values. Limitations include the ongoing nature of the study, small sample size, and lack of non-carrier controls. Conclusions and Clinical Implications Our findings described a new PCa screening strategy integrated with genetic risk factors. The incorporation of PHI shows promise in improving the efficiency of diagnostic procedures while minimizing unnecessary interventions. High adherence among high-risk individuals underscores the potential effectiveness of targeted screening programs.
Collapse
|
10
|
Nakamura S, Kojima Y, Takeuchi S. Causative Genes of Homologous Recombination Deficiency (HRD)-Related Breast Cancer and Specific Strategies at Present. Curr Oncol 2025; 32:90. [PMID: 39996890 PMCID: PMC11854191 DOI: 10.3390/curroncol32020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Recently, homologous recombination deficiency (HRD) has become a new target for hereditary cancers. Molecular-based approaches for hereditary cancers in the clinical setting have been reviewed. In particular, the efficacy of the PARP inhibitor has been considered by several clinical trials for various kinds of hereditary cancers. This indicates that the PARP inhibitor can be effective for any kind of BRCA mutated cancers, regardless of the organ-specific cancer. Homologous recombination deficiency (HRD) has become a new target for hereditary cancers, indicating the necessity to confirm the status of HRD-related genes. ARID1A, ATM, ATRX, PALB2, BARD1, RAD51C and CHEK2 are known as HRD-related genes for which simultaneous examination as part of panel testing is more suitable. Both surgical and medical oncologists should learn the basis of genetics including HRD. An understanding of the basic mechanism of homologous repair recombination (HRR) in BRCA-related breast cancer is mandatory for all surgical or medical oncologists because PARP inhibitors may be effective for these cancers and a specific strategy of screening for non-cancers exists. The clinical behavior of each gene should be clarified based on a large-scale database in the future, or, in other words, on real-world data. Firstly, HRD-related genes should be examined when the hereditary nature of a cancer is placed in doubt after an examination of the relevant family history. Alternatively, HRD score examination is a solution by which to identify HRD-related genes at the first step. If lifetime risk is estimated at over 20%, an annual breast MRI is necessary for high-risk screening. However, there are limited data to show its benefit compared with BRCA. Therefore, a large-scale database, including clinical information and a long-term follow-up should be established, after which a periodical assessment is mandatory. The clinical behavior of each gene should be clarified based on a large-scale database, or, in other words, real-world data.
Collapse
Affiliation(s)
- Seigo Nakamura
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
- Division of Breast Surgical Oncology, Department of Surgery, Showa University, Tokyo 142-8666, Japan
| | - Yasuyuki Kojima
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
- Division of Breast Surgical Oncology, Department of Surgery, Showa University, Tokyo 142-8666, Japan
| | - Sayoko Takeuchi
- Institute for Clinical Genetics and Genomics, Showa University, Tokyo 142-8555, Japan; (Y.K.); (S.T.)
| |
Collapse
|
11
|
Bottillo I, Sciarra A, Bevilacqua G, Gentilucci A, Sciarra B, Santarelli V, Salciccia S, Bacigalupo F, Pastacaldi F, Ciccone MP, De Marchis L, Santini D, Magliocca FM, Merenda E, Forte F, Grammatico P. Early Detection of the Pathogenetic Variants of Homologous Recombination Repair Genes in Prostate Cancer: Critical Analysis and Experimental Design. BIOLOGY 2025; 14:117. [PMID: 40001885 PMCID: PMC11851859 DOI: 10.3390/biology14020117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
It has been shown that the pathogenic variants (PVs) of the DNA Damage Response (DDR) genes, whether of a germinal or somatic nature, represent a predictive biomarker of high sensitivity to treatment with inhibitors of the enzyme poly-ADP-ribose polymerase (PARP) in patients with hormone-resistant metastatic prostate cancer (HRPCa). Moreover, the detection of PVs of the Homologous Recombination Repair (HRR) genes in PCa patients can help to define the patient's prognosis and the choice of the therapeutic procedure. Among men with metastatic PCa, the frequency of PVs in HRR genes ranges from 11% to 33%, which is a significantly higher rate compared to non-metastatic PCa, where the incidence is between 5% and 10%. Next-Generation Sequencing (NGS) results were more commonly obtained from newly acquired somatic samples compared to archived samples (prostate biopsy or prostatectomy). We developed an experimental multidisciplinary prospective study in patients with a new diagnosis of high-risk PCa at biopsy. The aim was to evaluate the presence of PVs of different HRR genes in patients with the first diagnosis of PCa in relation to a metastatic or non-metastatic stage, tumor aggressiveness, and early risk of progression. Among 43 initial tumor samples from 22 patients, 25 samples from 12 patients were selected for library preparation based on their DNA concentration and quality. After the NGS, 14 different DNA variants were prioritized. Oncogenetic and likely oncogenetic variants were found in the ATM, BRCA1, PTEN, KMT2D, and CDH1 genes. Moreover, variants of uncertain significance were found in ATM, DDR2, FANCA, FOXA1, PLCB4, PTCH1, and RB1.
Collapse
Affiliation(s)
- Irene Bottillo
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00161 Rome, Italy; (I.B.); (F.B.); (F.P.); (M.P.C.); (P.G.)
| | - Alessandro Sciarra
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, Viale Policlinico 155, 00161 Rome, Italy; (G.B.); (A.G.); (V.S.); (S.S.)
| | - Giulio Bevilacqua
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, Viale Policlinico 155, 00161 Rome, Italy; (G.B.); (A.G.); (V.S.); (S.S.)
| | - Alessandro Gentilucci
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, Viale Policlinico 155, 00161 Rome, Italy; (G.B.); (A.G.); (V.S.); (S.S.)
| | - Beatrice Sciarra
- Department of Pharmaceutic Chemistry, University Sapienza, 00161 Rome, Italy;
| | - Valerio Santarelli
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, Viale Policlinico 155, 00161 Rome, Italy; (G.B.); (A.G.); (V.S.); (S.S.)
| | - Stefano Salciccia
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, Viale Policlinico 155, 00161 Rome, Italy; (G.B.); (A.G.); (V.S.); (S.S.)
| | - Francesca Bacigalupo
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00161 Rome, Italy; (I.B.); (F.B.); (F.P.); (M.P.C.); (P.G.)
| | - Francesco Pastacaldi
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00161 Rome, Italy; (I.B.); (F.B.); (F.P.); (M.P.C.); (P.G.)
| | - Maria Pia Ciccone
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00161 Rome, Italy; (I.B.); (F.B.); (F.P.); (M.P.C.); (P.G.)
| | - Laura De Marchis
- Department Oncology, University Sapienza, 00161 Rome, Italy; (L.D.M.); (D.S.)
| | - Daniele Santini
- Department Oncology, University Sapienza, 00161 Rome, Italy; (L.D.M.); (D.S.)
| | | | - Elisabetta Merenda
- Department of Pathology, University Sapienza, 00161 Rome, Italy; (F.M.M.); (E.M.)
| | - Flavio Forte
- Urology Unit, Vannini Hospital, 00190 Rome, Italy;
| | - Paola Grammatico
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, 00161 Rome, Italy; (I.B.); (F.B.); (F.P.); (M.P.C.); (P.G.)
| |
Collapse
|
12
|
Stroomberg HV, Brasso K, Blak AA, Byrjalsen A, Hansen TVO, Røder A. Prostate-specific antigen screening at low thresholds of men with pathogenic BRCA1/2 variants. Prostate Cancer Prostatic Dis 2025:10.1038/s41391-025-00938-z. [PMID: 39838196 DOI: 10.1038/s41391-025-00938-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/19/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Men with pathogenic BRCA1/2 variants are at higher risk of prostate cancer We included men with likely pathogenic/pathogenic (LP/P) variants in BRCA1/2 in a prostate-specific antigen (PSA) screening program after cascade germline testing since 2014. PSA was tested yearly and an age-specific low PSA threshold for biopsy was used, to determine if a low PSA threshold for biopsy is justified for men with pathogenic BRCA1/2 variants. METHODS From 2014 to 2023 a total of 340 men were included in the program. We report demographics, clinical characteristics, and treatment outcomes at 7 years. RESULTS The cumulative incidence of a primary biopsy was 37% (95CI: 31‒43) after 7 years. Incidence of prostate cancer diagnosis was 11% (95CI: 7.1‒15). Men referred were 7.8 (95CI: 5.3‒11, p < 0.001) times more likely to be diagnosed with prostate cancer than the general Danish male population. The cumulative incidence of biochemical failure (PSA > = 0.2 ng/ml) 4 years after RP was 22% (95CI: 2.3‒41). The main limitation is that not all men underwent a pre-biopsy MRI. CONCLUSION We found a high incidence of prostate cancer in men with LP/P BRCA1/2 variants, but this may be explained by the low PSA threshold for scheduling biopsies. More studies are needed to compare this patient population to men with other germline features. The high risk of recurrence after curative therapy is worrisome and requires further evaluation as to whether this is a biological phenomenon.
Collapse
Affiliation(s)
- Hein V Stroomberg
- Copenhagen Prostate Cancer Center, Department of Urology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Biotech Research & Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.
| | - Klaus Brasso
- Copenhagen Prostate Cancer Center, Department of Urology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anna A Blak
- Copenhagen Prostate Cancer Center, Department of Urology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anna Byrjalsen
- Department of Clinical Genetics, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Thomas van Overeem Hansen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Genetics, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Andreas Røder
- Copenhagen Prostate Cancer Center, Department of Urology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Hübner A, Bußhoff I, Lakes J, Al-Monajjed R, Radtke JP, Albers P. [Early detection of prostate cancer-individualized, risk-adapted and successful]. UROLOGIE (HEIDELBERG, GERMANY) 2025; 64:14-23. [PMID: 39609267 DOI: 10.1007/s00120-024-02478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Population-based screening for prostate cancer (PC) is still controversially discussed. Furthermore, an organized, risk-adapted screening program is already being called for across Europe. Although large randomized controlled trials have shown that prostate-specific antigen (PSA)-based screening can significantly reduce PC-specific mortality, all known screening strategies still frequently lead to overdiagnosis and consecutively to overtreatment of clinically insignificant PC. OBJECTIVE This review article presents the current evidence regarding PC screening. RESULTS The current German early detection program, which is based on the annually recommended digital rectal examination (DRE), lacks specificity and sensitivity. In Germany, an opportunistic PSA screening is currently being carried out, which is no longer recommended due to an unfavorable benefit-risk ratio. Since 2022, the European Commission has been calling for the development of organized, risk-adapted screening strategies that combine the PSA test with magnetic resonance imaging (MRI). The integration of MRI improves the detection of clinically relevant PC and reduces the overdiagnosis of low-risk cancers. In the future, an algorithm for early detection of PC needs to be developed, which, in an organized and personalized screening, combines the PSA test with MRI and possibly genetic markers. CONCLUSION The early detection of prostate cancer should be organized at the population level and integrated into personalized, evidence-based screening strategies.
Collapse
Affiliation(s)
- Anne Hübner
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Isabelle Bußhoff
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Jale Lakes
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Rouvier Al-Monajjed
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland.
- Abteilung für Personalisierte Früherkennung des Prostatakarzinoms, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland.
| | - Jan Philipp Radtke
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland
- Abteilung für Radiologie, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland
| | - Peter Albers
- Klinik für Urologie, Universitätsklinik Düsseldorf, Heinrich-Heine-Universität, Moorenstr. 5, 40225, Düsseldorf, Deutschland
- Abteilung für Personalisierte Früherkennung des Prostatakarzinoms, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Deutschland
| |
Collapse
|
14
|
Oudard S, Timsit MO, Maillet D, Mouillet G, Campedel L, Colomba É, Dourthe LM, Eymard JC, Gobert A, Jamet C, Joly C, Serrate C, Ploussard G. [Metastatic castration-resistant prostate cancer and PARP inhibitors: From tumor genomics to new therapeutic combinations]. Bull Cancer 2025; 112:61-81. [PMID: 39232886 DOI: 10.1016/j.bulcan.2024.05.008] [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: 02/07/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 09/06/2024]
Abstract
Castration-resistant metastatic prostate cancer remains lethal and a therapeutic challenge. Current strategies are geared towards the personalization of treatments based on the identification of relevant molecular targets, including genomic alterations involved in tumoral processes. Among these novel targeted therapies, poly-ADP-ribose polymerase inhibitors (PARPi), by blocking the action of enzymes involved in deoxyribonucleic acid (DNA) repair, induce the destruction of cells carrying defects in homologous recombination repair, often associated with alterations in genes involved in this mechanism. Thus, determining the presence of a molecular anomaly, particularly alterations in the BRCA1/2 genes, is a prerequisite for initiating PARPi monotherapy. In patients with metastatic castration-resistant prostate cancer , around 20-30 % carry this type of mutation. In this population, single-agent studies have demonstrated PARPi ability to prolong overall survival, and to improve symptom control, including pain. Other studies are underway to assess their effectiveness in combination with other therapies, and it already appears that association with new-generation hormone therapy can further prolong radiological progression-free survival, regardless of the mutation status of the genes involved in DNA repair, indicating a synergistic action between PARPi and new-generation hormone therapy.
Collapse
Affiliation(s)
- Stéphane Oudard
- Hôpital Européen Georges-Pompidou, service de cancérologie médicale, Paris, France.
| | - Marc-Olivier Timsit
- Université de Paris, service urologie, cancérologie génito-urinaire et transplantation rénale, Paris, France; Hôpital Necker-Enfants malades, service d'urologie, Paris, France
| | - Denis Maillet
- Hospices civils de Lyon (IC-HCL), Institut de cancérologie, service d'oncologie médicale, Lyon, France; Faculté de médecine Jacques-Lisfranc, Saint-Étienne, France
| | | | - Luca Campedel
- Université Clermont-Auvergne, CHU de Gabriel-Montpied, service d'oncologie, Clermont-Ferrand, France
| | - Émeline Colomba
- Université Paris-Saclay, Institut Gustave Roussy, service de médecine oncologique, Villejuif, France
| | | | | | - Aurélien Gobert
- Centre hospitalier privé Saint-Grégoire, ICRB, Rennes, France
| | - Claire Jamet
- Centre hospitalier Saint-Louis, service d'oncologie médicale, La Rochelle, France
| | - Charlotte Joly
- Hôpital Henri-Mondor, service d'oncologie, Créteil, France
| | - Camille Serrate
- Groupe hospitalier Diaconesses Croix Saint-Simon, service d'oncologie médicale, Paris, France
| | - Guillaume Ploussard
- IUCT Oncopôle Toulouse, service d'urologie, Toulouse, France; Clinique La Croix du Sud, UROSUD, Toulouse, France
| |
Collapse
|
15
|
Guo A, Wu C, Cao J, Zhu K, Ding S. Real-world efficacy and safety of combined first-line treatment with PARP inhibitors and novel hormonal therapy in mCRPC patients with HRR gene mutations. Front Genet 2024; 15:1505163. [PMID: 39712485 PMCID: PMC11659292 DOI: 10.3389/fgene.2024.1505163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
Objective This study evaluated the real-world efficacy and safety of combining PARP inhibitors with novel hormonal therapy (NHT) as a first-line treatment in Chinese patients with metastatic castration-resistant prostate cancer (mCRPC) harboring homologous recombination repair (HRR) gene mutations. Methods We enrolled 41 mCRPC patients who received at least 1 month of combined treatment with PARP inhibitors and NHT. Patients were divided into two groups: Cohort A (mutations in BRCA1, BRCA2, or ATM genes) and Cohort B (mutations in other HRR genes). The primary endpoint was imaging-based progression-free survival (PFS), with secondary endpoints including objective response rate (ORR), disease control rate (DCR), overall survival (OS), PSA50 response, and adverse events (AEs). To ensure accurate research results and control confounding factors, we will employ multivariate Cox proportional hazards models to evaluate key variables affecting mCRPC patient survival outcomes. Results This study enrolled 41 patients, 22 in Cohort A and 19 in Cohort B. The median PFS for all patients was 21.8 months, and the median OS had yet to be reached. The overall ORR was 48.8%, and the DCR was 61.0%. Specifically, the median PFS for Cohort A was 21.8 months compared to 14.5 months for Cohort B. The median OS had yet to be reached for either cohort. Regarding efficacy, 81.8% of patients in Cohort A and 73.7% in Cohort B achieved a PSA50 response. Imaging assessments showed ORRs of 54.6% for Cohort A and 42.1% for Cohort B, with DCRs of 72.7% and 47.4%, respectively. 85.4% of patients experienced grade 1 or 2 adverse events, and 51.2% encountered grade 3 or 4. In the multivariate Cox regression analysis focusing on PFS, the Gleason score was identified as a significant predictor (HR = 5.8, 95% CI: 1.65-20.2, p = 0.006). Conclusion Combined first-line treatment with PARP inhibitors and NHT is effective and well-tolerated in mCRPC patients with HRR gene mutations, particularly those with BRCA1, BRCA2, or ATM mutations. These findings underscore the potential of this therapeutic combination in managing mCRPC in the Chinese population, suggesting a favorable outcome for those with specific genetic backgrounds.
Collapse
Affiliation(s)
- Andong Guo
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chenrui Wu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jishuang Cao
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Kejia Zhu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Sentai Ding
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
An M, Chen C, Xiang J, Li Y, Qiu P, Tang Y, Liu X, Gu Y, Qin N, He Y, Zhu M, Jiang Y, Dai J, Jin G, Ma H, Wang C, Hu Z, Shen H. Systematic identification of pathogenic variants of non-small cell lung cancer in the promoters of DNA-damage repair genes. EBioMedicine 2024; 110:105480. [PMID: 39631147 DOI: 10.1016/j.ebiom.2024.105480] [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: 06/04/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Deficiency in DNA-damage repair (DDR) genes, often due to disruptive coding variants, is linked to higher cancer risk. Our previous study has revealed the association between rare loss-of-function variants in DDR genes and the risk of lung cancer. However, it is still challenging to study the predisposing role of rare regulatory variants of these genes. METHODS Based on whole-genome sequencing data from 2984 patients with non-small cell lung cancer (NSCLC) and 3020 controls, we performed massively parallel reporter assays on 1818 rare variants located in the promoters of DDR genes. Pathway- or gene-level burden analyses were performed using Firth's logistic regression or generalized linear model. FINDINGS We identified 750 rare functional regulatory variants (frVars) that showed allelic differences in transcriptional activity within the promoter regions of DDR genes. Interestingly, the burden of frVars was significantly elevated in cases (odds ratio [OR] = 1.17, p = 0.026), whereas the burden of variants prioritized solely based on bioinformatics annotation was comparable between cases and controls (OR = 1.04, p = 0.549). Among the frVars, 297 were down-regulated transcriptional activity (dr-frVars) and 453 were up-regulated transcriptional activity (ur-frVars); especially, dr-frVars (OR = 1.30, p = 0.008) rather than ur-frVars (OR = 1.06, p = 0.495) were significantly associated with risk of NSCLC. Individuals with NSCLC carried more dr-frVars from Fanconi anemia, homologous recombination, and nucleotide excision repair pathways. In addition, we identified seven genes (i.e., BRCA2, GTF2H1, DDB2, BLM, ALKBH2, APEX1, and RAD51B) with promoter dr-frVars that were associated with lung cancer susceptibility. INTERPRETATION Our findings indicate that functional promoter variants in DDR genes, in addition to protein-truncating variants, can be pathogenic and contribute to lung cancer susceptibility. FUNDING National Natural Science Foundation of China, Youth Foundation of Jiangsu Province, Research Unit of Prospective Cohort of Cardiovascular Diseases and Cancer of Chinese Academy of Medical Sciences, and Natural Science Foundation of Jiangsu Province.
Collapse
Affiliation(s)
- Mingxing An
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Congcong Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China
| | - Jun Xiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yang Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pinyu Qiu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yiru Tang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xinyue Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yayun Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Na Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuanlin He
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China.
| | - Zhibin Hu
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
17
|
Amini AE, Hunter AE, Almashad A, Feng AJ, Patel ND, O'Dea MR, McCormick SR, Rodgers LH, Salari K. Magnetic Resonance Imaging-based Prostate Cancer Screening in Carriers of Pathogenic Germline Mutations: Interim Results from the Initial Screening Round of the Prostate Cancer Genetic Risk Evaluation and Screening Study. Eur Urol Oncol 2024; 7:1358-1366. [PMID: 38453598 DOI: 10.1016/j.euo.2024.01.015] [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: 12/03/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND The risk of early-onset and clinically aggressive prostate cancer is elevated in carriers of certain rare pathogenic germline mutations. The utility of augmenting traditional prostate-specific antigen (PSA)-based screening measures with multiparametric magnetic resonance imaging (MRI) in this population is not yet known. OBJECTIVE To evaluate MRI-based screening in comparison with traditional PSA-based screening among individuals at an elevated genetic risk for prostate cancer. DESIGN, SETTING, AND PARTICIPANTS Male germline carriers of pathogenic/likely pathogenic variants in any of 19 prostate cancer risk genes between the ages of 35 and 74 yr with no prior history of prostate cancer were recruited. Intervention Enrolled participants underwent screening with annual PSA, digital rectal examination (DRE), and triennial multiparametric MRI. Individuals with abnormal DRE, elevated age-adjusted PSA (>1.5 ng/ml for 35-49 yr, >2.0 ng/ml for 50-54 yr, and >3.0 ng/ml for 55-74 yr), or suspicious multiparametric MRI (Prostate Imaging Reporting and Data System [PI-RADS] ≥3 lesion) were offered prostate biopsy. Outcome measurements and statistical analysis Endpoints were diagnosis of any and clinically significant prostate cancer, and alternative screening strategies were compared by a decision curve analysis. RESULTS AND LIMITATIONS To date, 101 males have completed the first round of screening. The greatest proportion of participants are carriers of BRCA2 (n = 44), BRCA1 (n = 35), and ATM (n = 7) variants. Twenty-one have undergone biopsy, resulting in the detection of nine cases of cancer (seven clinically significant). For the detection of clinically significant prostate cancer, abnormal MRI (PI-RADS ≥3) demonstrated 100% sensitivity (7/7) with a negative predictive value (NPV) of 100%, whereas PSA-based screening alone had 57% (4/7) sensitivity with an NPV of 73%. Of six screening strategies evaluated in the decision curve analysis, MRI-based screening alone achieved superior net benefit at all threshold probabilities compared with PSA screening-detecting one additional cancer case per 7.5 patients, while avoiding more unnecessary biopsies at the same threshold probability. CONCLUSIONS Disease prevalence is high among carriers of prostate cancer-associated pathogenic germline mutations. Early results suggest that MRI-based screening enhances early detection of clinically significant disease beyond PSA screening alone. PATIENT SUMMARY In this study, we present the interim results from the PROGRESS prostate cancer screening trial. We found that in certain germline carriers of prostate cancer risk mutations, magnetic resonance imaging-based screening enhances detection of prostate cancer while reducing biopsies triggered, in comparison with traditional prostate-specific antigen screening strategies.
Collapse
Affiliation(s)
- Andrew E Amini
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandra E Hunter
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aya Almashad
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aileen J Feng
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Neel D Patel
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Margaret R O'Dea
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shelley R McCormick
- Center for Cancer Risk Assessment, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Linda H Rodgers
- Center for Cancer Risk Assessment, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Cancer Risk Assessment, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
18
|
Tuffaha H, Edmunds K, Fairbairn D, Roberts MJ, Chambers S, Smith DP, Horvath L, Arora S, Scuffham P. Guidelines for genetic testing in prostate cancer: a scoping review. Prostate Cancer Prostatic Dis 2024; 27:594-603. [PMID: 37202470 PMCID: PMC11543603 DOI: 10.1038/s41391-023-00676-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Genetic testing, to identify pathogenic or likely pathogenic variants in prostate cancer, is valuable in guiding treatment decisions for men with prostate cancer and to inform cancer prevention and early detection options for their immediate blood relatives. There are various guidelines and consensus statements for genetic testing in prostate cancer. Our aim is to review genetic testing recommendations across current guidelines and consensus statements and the level of evidence supporting those recommendations. METHODS A scoping review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-analyses extension for scoping review (PRISMA-ScR) guidelines. Electronic database searches and manual searches of grey literature, including websites of key organisations were conducted. Using the Population, Concept, Context (PCC) framework, this scoping review included: men with prostate cancer or men at high risk of prostate cancer and their biological families; existing guidelines and consensus statements with supporting evidence for genetic testing of men with prostate cancer from any geographical location worldwide. RESULTS Of the 660 citations identified, 23 guidelines and consensus statements met the inclusion criteria for the scoping review. Based on different levels of evidence about who should be tested and how, a diverse range of recommendations were identified. There was general consensus among the guidelines and consensus statements that men with metastatic disease be offered genetic testing; however, there was less consensus in relation to genetic testing in localised prostate cancer. While there was some consensus in relation to which genes to test, recommendations varied regarding who to test, testing methods and implementation. CONCLUSION While genetic testing in prostate cancer is routinely recommended and numerous guidelines exist, there is still considerable lack of consensus regarding who should be tested and how they should be tested. Further evidence is needed to inform value-based genetic testing strategies for implementation in practice.
Collapse
Affiliation(s)
- Haitham Tuffaha
- Centre for the Business and Economics of Health, University of Queensland, Brisbane, QLD, Australia.
| | - Kim Edmunds
- Centre for the Business and Economics of Health, University of Queensland, Brisbane, QLD, Australia
| | - David Fairbairn
- Pathology Queensland, The Royal Brisbane Women's Hospital, Brisbane, QLD, Australia
| | - Matthew J Roberts
- UQ Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Suzanne Chambers
- The Faculty of Health Sciences, Australian Catholic University, Brisbane, NSW, Australia
| | - David P Smith
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Lisa Horvath
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
- Clinical Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Shiksha Arora
- Centre for the Business and Economics of Health, University of Queensland, Brisbane, QLD, Australia
| | - Paul Scuffham
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
19
|
Chou WH, Chalker C, Sokolova AO, Isharwal S. Prostate cancer and genetic contributions. Andrology 2024:10.1111/andr.13812. [PMID: 39611376 PMCID: PMC12119970 DOI: 10.1111/andr.13812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/17/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Prostate cancer remains a lethal disease for many men. Knowledge of genetic contributions to this condition has increasingly been used in its management. In this narrative review, we summarize various genetic alterations and syndromes associated with prostate cancer, including hereditary breast and ovarian cancer syndrome, Lynch syndrome, and hereditary prostate cancer, among others. Indications for germline testing are reviewed, as well as incorporation of genetic data at different phases of management for prostate cancer, such as screening and monitoring, and treatment of localized and metastatic disease.
Collapse
Affiliation(s)
- Wesley H. Chou
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
| | - Cameron Chalker
- Division of Hematology/Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Alexandra O. Sokolova
- Division of Hematology/Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Sudhir Isharwal
- Department of Urology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
20
|
Hemminki K, Kiemeney LA, Morgans AK, Ranniko A, Pichler R, Hemminki O, Culig Z, Mulders P, Bangma CH. Hereditary and Familial Traits in Urological Cancers and Their Underlying Genes. EUR UROL SUPPL 2024; 69:13-20. [PMID: 39314911 PMCID: PMC11416669 DOI: 10.1016/j.euros.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Early recognition of hereditary urological cancers may influence diagnostic and therapeutic decision-making, and potentially alter the fate of patients and family members. Here, we introduce readers to the current knowledge on germline genetic testing and clinical practice in prostate, bladder, renal, and testicular carcinoma. Considering all urological cancer patients, routine inquiries about familial cancer history should become a standard practice in clinical settings. If suspicion arises, patients can opt for two avenues: referral to genetic counseling or undergoing genetic tests after consultation with the treating urologist. Patient summary Tumors of the urogenital tract (prostate, kidney, bladder, and testes) can sometimes be related to genetic mutations that are present in all the cells of the body. Such mutations can be inherited and run in families. Therefore, it is relevant to obtain information on the incidence of all cancers in the family history. The information obtained may initiate genetic testing, leading to the identification of mutations that are related to cancer in the current or next generation. In addition, these mutations may offer alternative treatment options for patients.
Collapse
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of Medicine, Charles University, Pilsen, Czech Republic
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Alicia K. Morgans
- Survivorship Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Antti Ranniko
- Research Program in Systems Oncology and ICAN-Digital Precision Cancer Medicine Flagship, Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Renate Pichler
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Otto Hemminki
- Research Program in Systems Oncology and ICAN-Digital Precision Cancer Medicine Flagship, Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Zoran Culig
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Mulders
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Chris H. Bangma
- Department of Urology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
21
|
Teppala S, Scuffham P, Edmunds K, Roberts MJ, Fairbairn D, Smith DP, Horvath L, Tuffaha H. The Cost-Effectiveness of Germline BReast CAncer Gene Testing in Metastatic Prostate Cancer Followed by Cascade Testing of First-Degree Relatives of Mutation Carriers. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:1515-1527. [PMID: 38977196 DOI: 10.1016/j.jval.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVES Patients with metastatic prostate cancer (mPCa) with BReast CAncer gene (BRCA) mutations benefit from targeted treatments (eg, olaparib). In addition, family members of affected patients have increased risk of hereditary cancers and benefit from early detection and prevention. International guidelines recommend genetic testing in mPCa; however, the value for money of testing patients with mPCa and cascade testing of blood-related family members has not been assessed. In this context, we evaluated the cost-effectiveness of germline BRCA testing in patients with mPCa followed by cascade testing of first-degree relatives (FDRs) of mutation carriers. METHODS We conducted a cost-utility analysis of germline BRCA testing using 2 scenarios: (1) testing patients with mPCa only and (2) testing patients with mPCa and FDRs of those who test positive. A semi-Markov multi-health-state transition model was constructed using a lifetime time horizon. The analyses were performed from an Australian payer perspective. Decision uncertainty was characterized using probabilistic analyses. RESULTS Compared with no testing, BRCA testing in mPCa was associated with an incremental cost of AU$3731 and a gain of 0.014 quality-adjusted life-years (QALYs), resulting in an incremental cost-effectiveness ratio of AU$265 942/QALY. Extending testing to FDRs of variant-positive patients resulted in an incremental cost-effectiveness ratio of AU$16 392/QALY. Probability of cost-effectiveness at a willingness-to-pay of AU$75 000/QALY was 0% in the standalone mPCa analysis and 100% in the cascade testing analysis. CONCLUSION BRCA testing when performed as a standalone strategy in patients with mPCa may not be cost-effective but demonstrates significant value for money after the inclusion of cascade testing of FDRs of mutation carriers.
Collapse
Affiliation(s)
- Srinivas Teppala
- Center for Applied Health Economics, Griffith University, Brisbane, QLD, Australia.
| | - Paul Scuffham
- Center for Applied Health Economics, Griffith University, Brisbane, QLD, Australia
| | - Kim Edmunds
- Center for the Business and Economics of Health, The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Roberts
- UQ Center for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - David Fairbairn
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - David P Smith
- The Daffodil Center, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Lisa Horvath
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia; Clinical Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Haitham Tuffaha
- Center for the Business and Economics of Health, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Ploussard G, Baboudjian M, Barret E, Brureau L, Fiard G, Fromont G, Olivier J, Dariane C, Mathieu R, Rozet F, Peyrottes A, Roubaud G, Renard-Penna R, Sargos P, Supiot S, Turpin L, Rouprêt M. French AFU Cancer Committee Guidelines - Update 2024-2026: Prostate cancer - Diagnosis and management of localised disease. THE FRENCH JOURNAL OF UROLOGY 2024; 34:102717. [PMID: 39581668 DOI: 10.1016/j.fjurol.2024.102717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/22/2024] [Accepted: 08/02/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE The aim of the Oncology Committee of the French Urology Association is to propose updated recommendations for the diagnosis and management of localized prostate cancer (PCa). METHODS A systematic review of the literature from 2022 to 2024 was conducted by the CCAFU on the elements of diagnosis and therapeutic management of localized PCa, evaluating references with their level of evidence. RESULTS The recommendations set out the genetics, epidemiology and diagnostic methods of PCa, as well as the concepts of screening and early detection. MRI, the reference imaging test for localized cancer, is recommended before prostate biopsies are performed. Molecular imaging is an option for disease staging. Performing biopsies via the transperineal route reduces the risk of infection. Active surveillance is the standard treatment for tumours with a low risk of progression. Therapeutic methods are described in detail, and recommended according to the clinical situation. CONCLUSION This update of French recommendations should help to improve the management of localized PCa.
Collapse
Affiliation(s)
- Guillaume Ploussard
- Department of Urology, La Croix du Sud Hospital, Quint-Fonsegrives, France; Department of Radiotherapy, Institut Curie, Paris, France.
| | | | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | - Laurent Brureau
- Department of Urology, CHU de Pointe-à-Pitre, University of Antilles, University of Rennes, Inserm, EHESP, Institut de Recherche en Santé, Environnement et Travail (Irset), UMR_S 1085, 97110 Pointe-à-Pitre, Guadeloupe
| | - Gaëlle Fiard
- Department of Urology, Grenoble Alpes University Hospital, Université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France
| | | | | | - Charles Dariane
- Department of Urology, Hôpital européen Georges-Pompidou, AP-HP, Paris, France; Paris University, U1151 Inserm, INEM, Necker, Paris, France
| | | | - François Rozet
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | | | - Guilhem Roubaud
- Department of Medical Oncology, Institut Bergonié, 33000 Bordeaux, France
| | - Raphaële Renard-Penna
- Sorbonne University, AP-HP, Radiology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Paul Sargos
- Department of Radiotherapy, Institut Bergonié, 33000 Bordeaux, France
| | - Stéphane Supiot
- Radiotherapy Department, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Léa Turpin
- Nuclear Medicine Department, Hôpital Foch, Suresnes, France
| | - Morgan Rouprêt
- Sorbonne University, GRC 5 Predictive Onco-Uro, AP-HP, Urology, Pitié-Salpêtrière Hospital, 75013 Paris, France
| |
Collapse
|
23
|
Lorentz J, Woollcombe J, Loblaw A, Liu S, Vesprini D. Screening guidelines for individuals at increased risk for prostate cancer. Can Urol Assoc J 2024; 18:E301-E307. [PMID: 38896481 PMCID: PMC11477514 DOI: 10.5489/cuaj.8710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Individuals at increased risk for prostate cancer (PCa) are inconsistently defined in national and international guidelines. The National Comprehensive Cancer Network (NCCN) defines people at increased risk for PCa to include those with a concerning family history, West African/Caribbean/African-American individuals, and those who have germline mutations in known PCa-related genes. Recommendations for screening are also inconsistently defined in national and international guidelines. The NCCN and American Urological Association recommend that individuals at increased risk for PCa be screened with prostate-specific antigen and digital rectal exam starting at age 40. Defining increased risk groups and defining lifetime risk is an ongoing academic process that can be facilitated through patient registries of these cohorts at academic centers.
Collapse
Affiliation(s)
- Justin Lorentz
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Julia Woollcombe
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Andrew Loblaw
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Stanley Liu
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Danny Vesprini
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
24
|
Ficarra V, Bartoletti R, Borghesi M, Caffo O, DE Nunzio C, Falagario UG, Gandaglia G, Giannarini G, Minervini A, Mirone V, Porpiglia F, Rocco B, Salonia A, Verze P, Carrieri G. Organized prostate cancer screening program: a proposal from the Italian Society of Urology (SIU). Minerva Urol Nephrol 2024; 76:519-529. [PMID: 39320244 DOI: 10.23736/s2724-6051.24.06117-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
To contrast opportunistic PCa screening, the European Union Council suggested extending screening programs to PCa by recommending the implementation of a stepwise approach in the EU Countries to evaluate the feasibility and effectiveness of an organized program based on PSA testing in combination with additional MRI as a follow-up test. The objective of this expert-based document is to propose an organized PCa screening program according to the EU Council recommendations. The Italian Society of Urology (SIU) developed a team of experts with the aim to report 1) the most recent epidemiologic data about incidence, prevalence, and mortality of PCa; 2) the most important risk factors to identify categories of men with an increased risk to eventually develop the disease; 3) the most relevant studies presenting data on population-based screening; and 4) the current recommendations of the leading International Guidelines. According to previous evidence, the Panel proposed some indications to develop a new organized PCa screening program for asymptomatic men with a life-expectancy of at least fifteen years. The SIU Panel strongly supports the implementation of a pilot, organized PCa screening program inviting asymptomatic men in the age range of 50-55 years. Invited men who are already performing opportunistic screening will be randomized to continue opportunistic screening or to cross into the organized protocol. Men with PSA level ≤3 ng/mL and familiarity for PCa received a DRE as well as all those with PSA levels >3 ng/mL. All other men with PSA levels greater than 3 ng/mL proceed to secondary testing represented by mpMRI. Men with Prostate Imaging-Reporting and Data System (PI-RADS) lesions 3 and PSAD 0.15 ng/mL/cc or higher as well as those with PI-RADS 4-5 lesions proceed to targeted plus systematic prostate biopsy. The primary outcome of the proposed pilot PCa screening program will be the detection rate of clinically significant PCa defined as a tumor with a ISUP Grade Group ≥2. Main secondary outcomes will be the detection rate of aggressive PCa (ISUP Grade Group ≥4); the detection rate of insignificant PCa (ISUP Grade Group 1); the number of unnecessary prostate biopsy avoided, the metastasis-free survival, and the overall survival. Men will be invited over a one-year period. Preliminary analyses will be planned 2 and 5 years after the baseline enrollment. According to the recent EU Council recommendations on cancer screening, pilot studies evaluating the feasibility and effectiveness of PCa screening programs using PSA as the primary and mpMRI as the secondary screening test in selected cohorts of patients must be strongly promoted by scientific societies and supported by national governments.
Collapse
Affiliation(s)
- Vincenzo Ficarra
- Urologic Section, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy -
- Urologic Section, Department of Oncology, AOU G. Martino, Messina, Italy -
| | - Riccardo Bartoletti
- Urology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marco Borghesi
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genoa, Genoa, Italy
| | - Orazio Caffo
- Division of Oncology, Santa Chiara Hospital, Trento, Italy
| | - Cosimo DE Nunzio
- Department of Urology, Sant'Andrea Hospital, La Sapienza University, Rome, Italy
| | - Ugo G Falagario
- Department of Urology and Kidney Transplantation, University of Foggia, Foggia, Italy
| | - Giorgio Gandaglia
- Department of Urology, Urological Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Gianluca Giannarini
- Urology Unit, "Santa Maria della Misericordia" University Hospital, Udine, Italy
| | - Andrea Minervini
- Unit of Oncologic Minimally-Invasive Urology and Andrology, Careggi Hospital, University of Florence, Florence, Italy
| | | | - Francesco Porpiglia
- Division of Urology, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Bernardo Rocco
- Europa Uomo Italia Nonprofit Organization, Milan, Italy
- Unit of Urology, ASST Santi Paolo e Carlo, Milan, Italy
| | - Andrea Salonia
- Department of Urology, Urological Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Paolo Verze
- Fondazione Prevenzione Ricerca Oncologia, Salerno, Italy
| | - Giuseppe Carrieri
- Department of Urology and Kidney Transplantation, University of Foggia, Foggia, Italy
| |
Collapse
|
25
|
Hall R, Bancroft E, Pashayan N, Kote-Jarai Z, Eeles RA. Genetics of prostate cancer: a review of latest evidence. J Med Genet 2024; 61:915-926. [PMID: 39137963 DOI: 10.1136/jmg-2024-109845] [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: 01/02/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024]
Abstract
Prostate cancer (PrCa) is a largely heritable and polygenic disease. It is the most common cancer in people with prostates (PwPs) in Europe and the USA, including in PwPs of African descent. In the UK in 2020, 52% of all cancers were diagnosed at stage I or II. The National Health Service (NHS) long-term plan is to increase this to 75% by 2028, to reduce absolute incidence of late-stage disease. In the absence of a UK PrCa screening programme, we should explore how to identify those at increased risk of clinically significant PrCa.Incorporating genomics into the PrCa screening, diagnostic and treatment pathway has huge potential for transforming patient care. Genomics can increase efficiency of PrCa screening by focusing on those with genetic predisposition to cancer-which when combined with risk factors such as age and ethnicity, can be used for risk stratification in risk-based screening (RBS) programmes. The goal of RBS is to facilitate early diagnosis of clinically significant PrCa and reduce overdiagnosis/overtreatment in those unlikely to experience PrCa-related symptoms in their lifetime. Genetic testing can guide PrCa management, by identifying those at risk of lethal PrCa and enabling access to novel targeted therapies.PrCa is curable if diagnosed below stage III when most people do not experience symptoms. RBS using genetic profiling could be key here if we could show better survival outcomes (or reduction in cancer-specific mortality accounting for lead-time bias), in addition to more cost efficiency than age-based screening alone. Furthermore, PrCa outcomes in underserved communities could be optimised if genetic testing was accessible, minimising health disparities.
Collapse
Affiliation(s)
- Rose Hall
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| | | | | | | | - Rosalind A Eeles
- The Royal Marsden NHS Foundation Trust, London, UK
- Institute for Cancer Research, London, UK
| |
Collapse
|
26
|
Cheng HH, Shevach JW, Castro E, Couch FJ, Domchek SM, Eeles RA, Giri VN, Hall MJ, King MC, Lin DW, Loeb S, Morgan TM, Offit K, Pritchard CC, Schaeffer EM, Szymaniak BM, Vassy JL, Katona BW, Maxwell KN. BRCA1, BRCA2, and Associated Cancer Risks and Management for Male Patients: A Review. JAMA Oncol 2024; 10:1272-1281. [PMID: 39052257 PMCID: PMC12080741 DOI: 10.1001/jamaoncol.2024.2185] [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] [Indexed: 07/27/2024]
Abstract
Importance Half of all carriers of inherited cancer-predisposing variants in BRCA1 and BRCA2 are male, but the implications for their health are underrecognized compared to female individuals. Germline variants in BRCA1 and BRCA2 (also known as pathogenic or likely pathogenic variants, referred to here as BRCA1/2 PVs) are well known to significantly increase the risk of breast and ovarian cancers in female carriers, and knowledge of BRCA1/2 PVs informs established cancer screening and options for risk reduction. While risks to male carriers of BRCA1/2 PVs are less characterized, there is convincing evidence of increased risk for prostate cancer, pancreatic cancer, and breast cancer in males. There has also been a rapid expansion of US Food and Drug Administration-approved targeted cancer therapies, including poly ADP ribose polymerase (PARP) inhibitors, for breast, pancreatic, and prostate cancers associated with BRCA1/2 PVs. Observations This narrative review summarized the data that inform cancer risks, targeted cancer therapy options, and guidelines for early cancer detection. It also highlighted areas of emerging research and clinical trial opportunities for male BRCA1/2 PV carriers. These developments, along with the continued relevance to family cancer risk and reproductive options, have informed changes to guideline recommendations for genetic testing and strengthened the case for increased genetic testing for males. Conclusions and Relevance Despite increasing clinical actionability for male carriers of BRCA1/2 PVs, far fewer males than female individuals undergo cancer genetic testing. Oncologists, internists, and primary care clinicians should be vigilant about offering appropriate genetic testing to males. Identifying more male carriers of BRCA1/2 PVs will maximize opportunities for cancer early detection, targeted risk management, and cancer treatment for males, along with facilitating opportunities for risk reduction and prevention in their family members, thereby decreasing the burden of hereditary cancer.
Collapse
Affiliation(s)
- Heather H. Cheng
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
- University of Washington, Department of Medicine (Hematology and Oncology), Seattle, WA
| | - Jeffrey W. Shevach
- Duke University School of Medicine, Division of Medical Oncology, Durham, NC
| | - Elena Castro
- Hospital Universitario 12 de Octubre, Department of Medical Oncology, Madrid, Spain
| | - Fergus J. Couch
- Mayo Clinic, Division of Experimental Pathology and Laboratory Medicine, Rochester, NY
| | - Susan M. Domchek
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
| | - Rosalind A. Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London UK
| | - Veda N. Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | - Michael J. Hall
- Fox Chase Cancer Center, Department of Clinical Genetics, Philadelphia, PA
| | - Mary-Claire King
- University of Washington, Department of Medicine (Medical Genetics) and Department of Genome Sciences, Seattle, WA
| | - Daniel W. Lin
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
- University of Washington, Department of Urology, Seattle, WA
| | - Stacy Loeb
- New York University School of Medicine, Department of Urology and Population Health, New York, NY
- Manhattan Veterans Affairs, Department of Surgery/Urology, New York, NY
| | - Todd M. Morgan
- University of Michigan, Department of Urology, Ann Arbor, MI
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, Clinical Genetics Service, New York, NY
| | - Colin C. Pritchard
- University of Washington, Department of Laboratory Medicine and Pathology, Seattle, WA
- Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Edward M. Schaeffer
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL
| | - Brittany M. Szymaniak
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL
| | - Jason L. Vassy
- Harvard Medical School at VA Boston Healthcare System, Boston, MA
| | - Bryson W. Katona
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
| | - Kara N. Maxwell
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA
| |
Collapse
|
27
|
Bancroft EK, Page EC, Brook MN, Pope J, Thomas S, Myhill K, Helfand BT, Talaty P, Ong KR, Douglas E, Cook J, Rosario DJ, Salinas M, Buys SS, Anson J, Davidson R, Longmuir M, Side L, Eccles DM, Tischkowitz M, Taylor A, Cruellas M, Ballestero EP, Cleaver R, Varughese M, Barwell J, LeButt M, Greenhalgh L, Hart R, Azzabi A, Jobson I, Cogley L, Evans DG, Rothwell J, Taylor N, Hogben M, Saya S, Eeles RA, Aaronson NK. The psychosocial impact of prostate cancer screening for BRCA1 and BRCA2 carriers. BJU Int 2024; 134:484-500. [PMID: 38839570 DOI: 10.1111/bju.16432] [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] [Indexed: 06/07/2024]
Abstract
OBJECTIVES To report the long-term outcomes from a longitudinal psychosocial study that forms part of the 'Identification of Men with a genetic predisposition to ProstAte Cancer: Targeted Screening in men at higher genetic risk and controls' (IMPACT) study. The IMPACT study is a multi-national study of targeted prostate cancer (PrCa) screening in individuals with a known germline pathogenic variant (GPV) in either the BReast CAncer gene 1 (BRCA1) or the BReast CAncer gene 2 (BRCA2). SUBJECTS AND METHODS Participants enrolled in the IMPACT study were invited to complete a psychosocial questionnaire prior to each annual screening visit for a minimum of 5 years. The questionnaire included questions on sociodemographics and the following measures: Hospital Anxiety and Depression Scale, Impact of Event Scale, 36-item Short-Form Health Survey, Memorial Anxiety Scale for PrCa, Cancer Worry Scale, risk perception and knowledge. RESULTS A total of 760 participants completed questionnaires: 207 participants with GPV in BRCA1, 265 with GPV in BRCA2 and 288 controls (non-carriers from families with a known GPV). We found no evidence of clinically concerning levels of general or cancer-specific distress or poor health-related quality of life in the cohort as a whole. Individuals in the control group had significantly less worry about PrCa compared with the carriers; however, all mean scores were low and within reported general population norms, where available. BRCA2 carriers with previously high prostate-specific antigen (PSA) levels experience a small but significant increase in PrCa anxiety (P = 0.01) and PSA-specific anxiety (P < 0.001). Cancer risk perceptions reflected information provided during genetic counselling and participants had good levels of knowledge, although this declined over time. CONCLUSION This is the first study to report the longitudinal psychosocial impact of a targeted PrCa screening programme for BRCA1 and BRCA2 carriers. The results reassure that an annual PSA-based screening programme does not have an adverse impact on psychosocial health or health-related quality of life in these higher-risk individuals. These results are important as more PrCa screening is targeted to higher-risk groups.
Collapse
Affiliation(s)
- Elizabeth K Bancroft
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | | | - Mark N Brook
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Jennifer Pope
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | - Sarah Thomas
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
| | - Kathryn Myhill
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
| | - Brian T Helfand
- Division of Urology, John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, IL, USA
| | - Pooja Talaty
- Division of Urology, John and Carol Walter Center for Urological Health, NorthShore University HealthSystem, Evanston, IL, USA
| | - Kai-Ren Ong
- West Midlands Regional Clinical Genetics Service, Birmingham Women's Hospital, Birmingham, UK
| | - Emma Douglas
- West Midlands Regional Clinical Genetics Service, Birmingham Women's Hospital, Birmingham, UK
| | - Jackie Cook
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | | | - Monica Salinas
- Hereditary Cancer Program, ICO (Catalan Institute of Oncology), Barcelona, Spain
| | - Saundra S Buys
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jo Anson
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Rosemarie Davidson
- West of Scotland Genetic Service, Queen Elizabeth University Hospital, Glasgow, UK
| | - Mark Longmuir
- West of Scotland Genetic Service, Queen Elizabeth University Hospital, Glasgow, UK
| | - Lucy Side
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Diana M Eccles
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
- The University of Southampton Medical School, Southampton, UK
| | - Marc Tischkowitz
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK
- Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Amy Taylor
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Mara Cruellas
- Hereditary Cancer Genetics Group, Medical Oncology Department, Hospital Vall d'Hebron, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Eduard Perez Ballestero
- Hereditary Cancer Genetics Group, Medical Oncology Department, Hospital Vall d'Hebron, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Ruth Cleaver
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Mohini Varughese
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Julian Barwell
- University of Leicester, Leicester, UK
- University Hospitals Leicester, Leicester, UK
| | | | - Lynn Greenhalgh
- Liverpool Centre for Genomic Medicine, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Rachel Hart
- Liverpool Centre for Genomic Medicine, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Ashraf Azzabi
- Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Irene Jobson
- Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Lynn Cogley
- Peninsular Genetics, Derriford Hospital, Plymouth, UK
| | - D Gareth Evans
- Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jeanette Rothwell
- Genomic Medicine, Manchester Academic Health Sciences Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Natalie Taylor
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
| | - Matthew Hogben
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
| | - Sibel Saya
- Oncogenetics Team, Institute of Cancer Research, London, UK
- The University of Melbourne, Melbourne, Australia
| | - Rosalind A Eeles
- Oncogenetics Team, Royal Marsden NHS Foundation Trust, London, UK
- Oncogenetics Team, Institute of Cancer Research, London, UK
| | | |
Collapse
|
28
|
Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Brunckhorst O, Darraugh J, Eberli D, De Meerleer G, De Santis M, Farolfi A, Gandaglia G, Gillessen S, Grivas N, Henry AM, Lardas M, van Leenders GJLH, Liew M, Linares Espinos E, Oldenburg J, van Oort IM, Oprea-Lager DE, Ploussard G, Roberts MJ, Rouvière O, Schoots IG, Schouten N, Smith EJ, Stranne J, Wiegel T, Willemse PPM, Tilki D. EAU-EANM-ESTRO-ESUR-ISUP-SIOG Guidelines on Prostate Cancer-2024 Update. Part I: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur Urol 2024; 86:148-163. [PMID: 38614820 DOI: 10.1016/j.eururo.2024.03.027] [Citation(s) in RCA: 284] [Impact Index Per Article: 284.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/14/2024] [Accepted: 03/27/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND AND OBJECTIVE The European Association of Urology (EAU)-European Association of Nuclear Medicine (EANM)-European Society for Radiotherapy and Oncology (ESTRO)-European Society of Urogenital Radiology (ESUR)-International Society of Urological Pathology (ISUP)-International Society of Geriatric Oncology (SIOG) guidelines provide recommendations for the management of clinically localised prostate cancer (PCa). This paper aims to present a summary of the 2024 version of the EAU-EANM-ESTRO-ESUR-ISUP-SIOG guidelines on the screening, diagnosis, and treatment of clinically localised PCa. METHODS The panel performed a literature review of all new data published in English, covering the time frame between May 2020 and 2023. The guidelines were updated, and a strength rating for each recommendation was added based on a systematic review of the evidence. KEY FINDINGS AND LIMITATIONS A risk-adapted strategy for identifying men who may develop PCa is advised, generally commencing at 50 yr of age and based on individualised life expectancy. The use of multiparametric magnetic resonance imaging in order to avoid unnecessary biopsies is recommended. When a biopsy is considered, a combination of targeted and regional biopsies should be performed. Prostate-specific membrane antigen positron emission tomography imaging is the most sensitive technique for identifying metastatic spread. Active surveillance is the appropriate management for men with low-risk PCa, as well as for selected favourable intermediate-risk patients with International Society of Urological Pathology grade group 2 lesions. Local therapies are addressed, as well as the management of persistent prostate-specific antigen after surgery. A recommendation to consider hypofractionation in intermediate-risk patients is provided. Patients with cN1 PCa should be offered a local treatment combined with long-term intensified hormonal treatment. CONCLUSIONS AND CLINICAL IMPLICATIONS The evidence in the field of diagnosis, staging, and treatment of localised PCa is evolving rapidly. These PCa guidelines reflect the multidisciplinary nature of PCa management. PATIENT SUMMARY This article is the summary of the guidelines for "curable" prostate cancer. Prostate cancer is "found" through a multistep risk-based screening process. The objective is to find as many men as possible with a curable cancer. Prostate cancer is curable if it resides in the prostate; it is then classified into low-, intermediary-, and high-risk localised and locally advanced prostate cancer. These risk classes are the basis of the treatments. Low-risk prostate cancer is treated with "active surveillance", a treatment with excellent prognosis. For low-intermediary-risk active surveillance should also be discussed as an option. In other cases, active treatments, surgery, or radiation treatment should be discussed along with the potential side effects to allow shared decision-making.
Collapse
Affiliation(s)
- Philip Cornford
- Department of Urology, Liverpool University Hospitals NHS Trust, Liverpool, UK.
| | | | | | | | | | - Julie Darraugh
- European Association of Urology, Arnhem, The Netherlands
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Gert De Meerleer
- Department of Radiation Oncology, University Hospital Leuven, Leuven, Belgium
| | - Maria De Santis
- Department of Urology, Universitätsmedizin Berlin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giorgio Gandaglia
- Division of Oncology/Unit of Urology, Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland (IOSI), EOC, Bellinzona, Switzerland; Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Nikolaos Grivas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ann M Henry
- Leeds Cancer Centre, St. James's University Hospital and University of Leeds, Leeds, UK
| | - Michael Lardas
- Department of Urology, Metropolitan General Hospital, Athens, Greece
| | | | - Matthew Liew
- Department of Urology, Liverpool University Hospitals NHS Trust, Liverpool, UK
| | | | - Jan Oldenburg
- Akershus University Hospital (Ahus), Lørenskog, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Inge M van Oort
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU Medical Center, Amsterdam, The Netherlands
| | | | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland Centre for Clinical Research, Herston, QLD, Australia
| | - Olivier Rouvière
- Department of Imaging, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France; Université de Lyon, Université Lyon 1, UFR Lyon-Est, Lyon, France
| | - Ivo G Schoots
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Emma J Smith
- European Association of Urology, Arnhem, The Netherlands
| | - Johan Stranne
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Urology, Sahlgrenska University Hospital-Västra Götaland, Gothenburg, Sweden
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Peter-Paul M Willemse
- Department of Urology, Cancer Center University Medical Center Utrecht, Utrecht, The Netherlands
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| |
Collapse
|
29
|
Miyahira AK, Kamran SC, Jamaspishvili T, Marshall CH, Maxwell KN, Parolia A, Zorko NA, Pienta KJ, Soule HR. Disrupting prostate cancer research: Challenge accepted; report from the 2023 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2024; 84:993-1015. [PMID: 38682886 DOI: 10.1002/pros.24721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION The 2023 Coffey-Holden Prostate Cancer Academy (CHPCA) Meeting, themed "Disrupting Prostate Cancer Research: Challenge Accepted," was convened at the University of California, Los Angeles, Luskin Conference Center, in Los Angeles, CA, from June 22 to 25, 2023. METHODS The 2023 marked the 10th Annual CHPCA Meeting, a discussion-oriented scientific think-tank conference convened annually by the Prostate Cancer Foundation, which centers on innovative and emerging research topics deemed pivotal for advancing critical unmet needs in prostate cancer research and clinical care. The 2023 CHPCA Meeting was attended by 81 academic investigators and included 40 talks across 8 sessions. RESULTS The central topic areas covered at the meeting included: targeting transcription factor neo-enhancesomes in cancer, AR as a pro-differentiation and oncogenic transcription factor, why few are cured with androgen deprivation therapy and how to change dogma to cure metastatic prostate cancer without castration, reducing prostate cancer morbidity and mortality with genetics, opportunities for radiation to enhance therapeutic benefit in oligometastatic prostate cancer, novel immunotherapeutic approaches, and the new era of artificial intelligence-driven precision medicine. DISCUSSION This article provides an overview of the scientific presentations delivered at the 2023 CHPCA Meeting, such that this knowledge can help in facilitating the advancement of prostate cancer research worldwide.
Collapse
Affiliation(s)
- Andrea K Miyahira
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tamara Jamaspishvili
- Department of Pathology and Laboratory Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Catherine H Marshall
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kara N Maxwell
- Department of Medicine-Hematology/Oncology and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Abhijit Parolia
- Department of Pathology, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas A Zorko
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- University of Minnesota Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Howard R Soule
- Science Department, Prostate Cancer Foundation, Santa Monica, California, USA
| |
Collapse
|
30
|
Grindedal EM, Zucknick M, Stormorken A, Rønne E, Tandstad NM, Isaacs WB, Axcrona K, Mæhle L. Outcomes of 10 years of PSA screening for prostate cancer in Norwegian men with Lynch syndrome. Prostate 2024; 84:945-953. [PMID: 38629217 DOI: 10.1002/pros.24711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Pathogenic germline variants in the mismatch repair (MMR) genes are associated with an increased risk of prostate cancer (PCa). Since 2010 we have recommended MMR carriers annual PSA testing from the age of 40. Prospective studies of the outcome of long-term PSA screening are lacking. This study aimed to investigate the incidence and characteristics of PCa in Norwegian MMR carriers attending annual PSA screening (PSA threshold >3.0 ng/mL) to evaluate whether our recommendations should be continued. METHODS This is a prospective observational study of 225 male MMR carriers who were recommended annual PSA screening by the Section of Inherited Cancer, Oslo University Hospital from 2010 and onwards. Incidence and tumor characteristics (age, PSA at diagnosis, Gleason score, TNM score) were described. IHC and MSI-analyses were done on available tumors. Standardized incidence ratio (SIR) was calculated based on data from the Cancer Registry of Norway. RESULTS Twenty-two of 225 (9.8%) had been diagnosed with PCa, including 10/69 (14.5%) MSH2 carriers and 8/61 (13.1%) MSH6 carriers. Ten of 20 (50%) tumors had Gleason score ≥4 + 3 on biopsy and 6/11 (54.5%) had a pathological T3a/b stage. Eight of 17 (47.1%) tumors showed abnormal staining on IHC and 3/13 (23.1%) were MSI-high. SIR was 9.54 (95% CI 5.98-14.45) for all MMR genes, 13.0 (95% CI 6.23-23.9) for MSH2 and 13.74 for MSH6 (95% CI 5.93-27.08). CONCLUSIONS Our results indicate that the MMR genes, and especially MSH2 and MSH6, are associated with a significant risk of PCa, and a high number of tumors show aggressive characteristics. While the impact of screening on patient outcomes remains to be more firmly established, the high SIR values we observe provide support for continued PSA screening of MSH2 and MSH6 carriers. Studies are needed to provide optimal recommendations for PSA-threshold and to evaluate whether MLH1 and PMS2 carriers should not be recommended screening.
Collapse
Affiliation(s)
| | - Manuela Zucknick
- Department of Biostatistics, Oslo Centre for Biostatistics and Epidemiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Astrid Stormorken
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Elin Rønne
- Department of Pathology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Nora M Tandstad
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - William B Isaacs
- Brady Urological Institute, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Karol Axcrona
- Department of Urology, Akershus University Hospital, Lørenskog, Norway
- Department of Molecular Oncology, Institute of Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Lovise Mæhle
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
31
|
van Harten MJ, Roobol MJ, van Leeuwen PJ, Willemse PPM, van den Bergh RCN. Evolution of European prostate cancer screening protocols and summary of ongoing trials. BJU Int 2024; 134:31-42. [PMID: 38469728 DOI: 10.1111/bju.16311] [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: 03/13/2024]
Abstract
Population-based organised repeated screening for prostate cancer has been found to reduce disease-specific mortality, but with substantial overdiagnosis leading to overtreatment. Although only very few countries have implemented a screening programme on a national level, individual prostate-specific antigen (PSA) testing is common. This opportunistic testing may have little favourable impact, while stressing the side-effects. The classic early detection protocols as were state-of-the-art in the 1990s applied a PSA and digital rectal examination threshold for sextant systematic prostate biopsy, with a fixed interval for re-testing, and limited indication for expectant management. In the three decades since these trials were started, different important improvements have become available in the cascade of screening, indication for biopsy, and treatment. The main developed aspects include: better identification of individuals at risk (using early/baseline PSA, family history, and/or genetic profile), individualised re-testing interval, optimised and individualised starting and stopping age, with gradual invitation at a fixed age rather than invitation of a wider range of age groups, risk stratification for biopsy (using PSA density, risk calculator, magnetic resonance imaging, serum and urine biomarkers, or combinations/sequences), targeted biopsy, transperineal biopsy approach, active surveillance for low-risk prostate cancer, and improved staging of disease. All these developments are suggested to decrease the side-effects of screening, while at least maintaining the advantages, but Level 1 evidence is lacking. The knowledge gained and new developments on early detection are being tested in different prospective screening trials throughout Europe. In addition, the European Union-funded PRostate cancer Awareness and Initiative for Screening in the European Union (PRAISE-U) project will compare and evaluate different screening pilots throughout Europe. Implementation and sustainability will also be addressed. Modern screening approaches may reduce the burden of the second most frequent cause of cancer-related death in European males, while minimising side-effects. Also, less efficacious opportunistic early detection may be indirectly reduced.
Collapse
Affiliation(s)
- Meike J van Harten
- Cancer Center, Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Monique J Roobol
- Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Peter-Paul M Willemse
- Cancer Center, Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roderick C N van den Bergh
- Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
- St Antonius Hospital, Utrecht, The Netherlands
| |
Collapse
|
32
|
Vlaming M, Ausems MGEM, Schijven G, van Oort IM, Kets CM, Komdeur FL, van der Kolk LE, Oldenburg RA, Sijmons RH, Kiemeney LALM, Bleiker EMA. Men with metastatic prostate cancer carrying a pathogenic germline variant in breast cancer genes: disclosure of genetic test results to relatives. Fam Cancer 2024; 23:165-175. [PMID: 38722431 PMCID: PMC11153271 DOI: 10.1007/s10689-024-00377-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/14/2024] [Indexed: 06/06/2024]
Abstract
Some patients with metastatic prostate cancer carry a pathogenic germline variant (PV) in a gene, that is mainly associated with an increased risk of breast cancer in women. If they test positive for such a PV, prostate cancer patients are encouraged to disclose the genetic test result to relatives who are at risk in case the carrier status changes the relatives' medical care. Our study aimed to investigate how men who learned they carry a PV in BRCA1, BRCA2, PALB2, CHEK2 or ATM disclosed their carrier status to at-risk relatives and to assess the possible psychological burden for the carrier and their perception of the burden for relatives. In total, 23 men with metastatic prostate cancer carrying a PV completed the IRI questionnaire about family communication; 14 also participated in a semi-structured interview. Patients felt highly confident in discussing the genetic test result with relatives. The diagnosis of prostate cancer was experienced as a burden, whereas being informed about genetic testing results did in most cases not add to this burden. Two patients encountered negative experiences with family communication, as they considered the genetic test result to be more urgent than their relatives. This mixed-methods study shows that metastatic prostate cancer patients with a PV in genes mainly associated with increased risk of breast cancer feel well-equipped to communicate about this predisposition in their families. Carriers felt motivated to disclose their genetic test result to relatives. Most of them indicated that the disclosure was not experienced as a psychological burden.
Collapse
Affiliation(s)
- Michiel Vlaming
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Heidelberglaan 100, CX Utrecht, 3584, The Netherlands
| | - Margreet G E M Ausems
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Heidelberglaan 100, CX Utrecht, 3584, The Netherlands
| | - Gina Schijven
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Heidelberglaan 100, CX Utrecht, 3584, The Netherlands
| | - Inge M van Oort
- Department of Urology, Radboud University Medical Center, Geert Grooteplein Zuid 10, GA Nijmegen, 6525, The Netherlands
| | - C Marleen Kets
- Department of Human Genetics, Radboud University Medical Center, Geert Grooteplein Zuid 10, GA Nijmegen, 6525, The Netherlands
| | - Fenne L Komdeur
- Department of Human Genetics, Amsterdam University Medical Centers, Meibergdreef 9, AZ Amsterdam, 1105, The Netherlands
| | - Lizet E van der Kolk
- Department of Clinical Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, CX Amsterdam, 1066, The Netherlands
| | - Rogier A Oldenburg
- Department of Clinical Genetics, Erasmus Medical Center, Dr. Molewaterplein 40, GD Rotterdam, 3015, The Netherlands
| | - Rolf H Sijmons
- Department of Genetics, University Medical Center Groningen, Hanzeplein 1, GZ Groningen, 9713, The Netherlands
| | - Lambertus A L M Kiemeney
- Department of Urology, Radboud University Medical Center, Geert Grooteplein Zuid 10, GA Nijmegen, 6525, The Netherlands
- Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein Zuid 10, GA Nijmegen, 6525, The Netherlands
| | - Eveline M A Bleiker
- Department of Clinical Genetics, the Netherlands Cancer Institute, Plesmanlaan 121, CX Amsterdam, 1066, The Netherlands.
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Plesmanlaan 121, CX Amsterdam, 1066, The Netherlands.
- Department of Clinical Genetics, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, 2333, The Netherlands.
| |
Collapse
|
33
|
Chalker C, Chun B, Sokolova AO. Germline and somatic mutations in prostate cancer: Implications for treatment. Curr Probl Cancer 2024; 50:101101. [PMID: 38718711 DOI: 10.1016/j.currproblcancer.2024.101101] [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: 12/21/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 06/16/2024]
Abstract
Genetic testing is an integral part of the workup of metastatic prostate cancer, in part, because the results can have a profound impact on the subsequent management of this disease. There are now several Food & Drug Administration (FDA) approved therapeutics available for patients with prostate cancer and certain genetic abnormalities - most notably, mutations in DNA damage repair (DDR) pathways such mismatch repair (MMR) and homologous recombination repair (HRR). In this review of the current literature, we discuss the indications for somatic and germline testing, the genetic changes of particular clinical relevance, the associated therapeutic options, and the clinical data supporting their use. We also highlight select trials-in-progress and future directions for the field.
Collapse
Affiliation(s)
- Cameron Chalker
- Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239.
| | - Brie Chun
- Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239
| | - Alexandra O Sokolova
- Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239
| |
Collapse
|
34
|
Trevisan L, Godino L, Battistuzzi L, Innella G, Luppi E, Buzzatti G, Gismondi V, Blondeaux E, Bonelli LA, Turchetti D, Varesco L. Cascade testing in Italian Hereditary Breast Ovarian Cancer families: a missed opportunity for cancer prevention? Fam Cancer 2024; 23:197-207. [PMID: 37968543 DOI: 10.1007/s10689-023-00349-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Healthy carriers of BRCA1/2 pathogenic variants (PVs) may benefit from risk-reducing measures of proven efficacy. The main approach to identify these individuals is cascade testing, and strategies to support this complex process are under investigation. In Italy, cascade testing has received little attention; therefore, we analyzed the uptake and characteristics of BRCA1/2 cascade testing in families diagnosed with HBOC between 2017 and 2019 at two Italian genetics centers. All blood relatives aged 18 years or older at September 2022 and who could be involved in the first step of cascade testing (i.e., all the living relatives closest to the proband) were included. In addition to first-degree relatives, individuals who were second-, third- or fourth-degree relatives were included if the closest relative(s) was/were deceased. Overall, 213 families were included (103, Genoa; 110, Bologna). Most probands were women affected by breast and/or ovarian cancer (86.4%, Genoa; 84.5%, Bologna), and the branch segregating the PV was known/suspected in 62% of families (62.1%, Genoa; 60.9%, Bologna). Overall, the uptake of cascade testing was 22.8% (25.8%, Genoa; 19.9%, Bologna; OR = 0.59: 95%CI 0.43-0.82). It was strongly associated with female gender (OR = 3.31, 95%CI 2.38-4.59), age ≤ 70 years (< 30 years OR = 3.48, 95%CI 1.85-6.56; 30-70 years OR = 3.08, 95%CI 2.01-4.71), first-degree relationship with the proband (OR = 16.61, 95%CI 10.50-26.28) and segregation of the PV in both the maternal (OR = 2.54, 95%CI 1.72-3.75) and the paternal branch (OR = 4.62, 95%CI 3.09-6.91). These real-world data may be important to inform the design and implementation of strategies aimed at improving the uptake of HBOC cascade testing in Italy.
Collapse
Affiliation(s)
- Lucia Trevisan
- Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lea Godino
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Linda Battistuzzi
- Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanni Innella
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Elena Luppi
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Giulia Buzzatti
- Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Viviana Gismondi
- Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Eva Blondeaux
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genova, 16132, Italy.
| | - Luigina Ada Bonelli
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, Genova, 16132, Italy
| | - Daniela Turchetti
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Liliana Varesco
- Unit of Hereditary Cancer, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
35
|
De Vrieze M, Hübner A, Al-Monajjed R, Albers P, Radtke JP, Schimmöller L, Boschheidgen M. [Prostate cancer screening-current overview]. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:479-487. [PMID: 38743100 DOI: 10.1007/s00117-024-01312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND The harm-to-benefit ratio of prostate cancer (PCa) screening remains controversial mainly due to the unfavorable test characteristics of prostate-specific antigen (PSA) as a screening test. METHODS In this nonsystematic review, we present a current overview of the body of evidence on prostate cancer screening with a focus on the role of magnetic resonance imaging (MRI) of the prostate. RESULTS Evidence generated in large randomized controlled trials showed that PSA-based screening significantly decreases cancer-specific mortality. The main obstacle in developing and implementing PCa screening strategies is the resulting overdiagnosis and as a consequence overtreatment of indolent cancers. Opportunistic screening is characterized by an adverse benefit-to-harm ratio and should, therefore, not be recommended. The German Statutory Early Detection Program for prostate cancer, which consists of a digital rectal examination (DRE) as a stand-alone screening test, is not evidence-based, neither specific nor sensitive enough and results in unnecessary diagnostics. The European Commission recently urged member states to develop population-based and organized risk-adapted PSA-based screening programs, which are currently tested in the ongoing German PROBASE trial. Finetuning of the diagnostic pathway following PSA-testing seems key to improve its positive and negative predictive value and thereby making PCa screening more accurate. Incorporation of prostatic MRI into screening strategies leads to more accurate diagnosis of clinically significant prostate cancer, while diagnosis of indolent cancers is reduced. In the future, molecular liquid-based biomarkers have the potential to complement or even replace PSA in PCa screening and further personalize screening strategies. Active surveillance as an alternative to immediate radical therapy of demographically increasing PCa diagnoses can potentially further improve the benefit-to-harm ratio of organized screening. CONCLUSION Early detection of PCa should be organized on a population level into personalized and evidence-based screening strategies. Multiparametric MRI of the prostate may play a key role in this setting.
Collapse
Affiliation(s)
- Maxime De Vrieze
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Deutschland
| | - Anne Hübner
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Rouvier Al-Monajjed
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Deutschland.
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Deutschland.
| | - Peter Albers
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Deutschland
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Jan Philipp Radtke
- Department of Urology, University Hospital Düsseldorf, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Deutschland
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Deutschland
| | - Lars Schimmöller
- University Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, 40225, Düsseldorf, Deutschland
- Department of Urology, University Hospital Düsseldorf, Department of Diagnostic, Interventional Radiology and Nuclear Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Deutschland
| | - Matthias Boschheidgen
- University Dusseldorf, Medical Faculty, Department of Diagnostic and Interventional Radiology, 40225, Düsseldorf, Deutschland
| |
Collapse
|
36
|
Kumsa FA, Fowke JH, Hashtarkhani S, White BM, Shrubsole MJ, Shaban-Nejad A. The association between neighborhood obesogenic factors and prostate cancer risk and mortality: the Southern Community Cohort Study. ARXIV 2024:arXiv:2405.18456v1. [PMID: 38855542 PMCID: PMC11160857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Prostate cancer is one of the leading causes of cancer-related mortality among men in the United States. We examined the role of neighborhood obesogenic attributes on prostate cancer risk and mortality in the Southern Community Cohort Study (SCCS). Methods From the total of 34,166 SCCS male participants, 28,356 were included in the analysis. We assessed the relationship between neighborhood obesogenic factors [neighborhood socioeconomic status (nSES) and neighborhood obesogenic environment indices including the restaurant environment index, the retail food environment index, parks, recreational facilities, and businesses] and prostate cancer risk and mortality by controlling for individual-level factors using a multivariable Cox proportional hazards model. We further stratified prostate cancer risk analysis by race and body mass index (BMI). Results Median follow-up time was 133 months [interquartile range (IQR): 103, 152], and the mean age was 51.62 (SD: ± 8.42) years. There were 1,524 (5.37%) prostate cancer diagnoses and 98 (6.43%) prostate cancer deaths during follow-up. Compared to participants residing in the wealthiest quintile, those residing in the poorest quintile had a higher risk of prostate cancer (aHR = 1.32, 95% CI 1.12-1.57, p = 0.001), particularly among non-obese men with a BMI < 30 (aHR = 1.46, 95% CI 1.07-1.98, p = 0.016). The restaurant environment index was associated with a higher prostate cancer risk in overweight (BMI ≥ 25) White men (aHR = 3.37, 95% CI 1.04-10.94, p = 0.043, quintile 1 vs. None). Obese Black individuals without any neighborhood recreational facilities had a 42% higher risk (aHR = 1.42, 95% CI 1.04-1.94, p = 0.026) compared to those with any access. Compared to residents in the wealthiest quintile and most walkable area, those residing within the poorest quintile (aHR = 3.43, 95% CI 1.54-7.64, p = 0.003) or the least walkable area (aHR = 3.45, 95% CI 1.22-9.78, p = 0.020) had a higher risk of prostate cancer death. Conclusion Living in a lower-nSES area was associated with a higher prostate cancer risk, particularly among Black men. Restaurant and retail food environment indices were also associated with a higher prostate cancer risk, with stronger associations within overweight White individuals. Finally, residing in a low-SES neighborhood or the least walkable areas were associated with a higher risk of prostate cancer mortality.
Collapse
Affiliation(s)
- Fekede Asefa Kumsa
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Jay H. Fowke
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Soheil Hashtarkhani
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Brianna M. White
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Martha J. Shrubsole
- Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Arash Shaban-Nejad
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| |
Collapse
|
37
|
Piombino C, Pipitone S, Tonni E, Mastrodomenico L, Oltrecolli M, Tchawa C, Matranga R, Roccabruna S, D’Agostino E, Pirola M, Bacchelli F, Baldessari C, Baschieri MC, Dominici M, Sabbatini R, Vitale MG. Homologous Recombination Repair Deficiency in Metastatic Prostate Cancer: New Therapeutic Opportunities. Int J Mol Sci 2024; 25:4624. [PMID: 38731844 PMCID: PMC11083429 DOI: 10.3390/ijms25094624] [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: 03/25/2024] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
More than 20% of metastatic prostate cancer carries genomic defects involving DNA damage repair pathways, mainly in homologous recombination repair-related genes. The recent approval of olaparib has paved the way to precision medicine for the treatment of metastatic prostate cancer with PARP inhibitors in this subset of patients, especially in the case of BRCA1 or BRCA2 pathogenic/likely pathogenic variants. In face of this new therapeutic opportunity, many issues remain unsolved. This narrative review aims to describe the relationship between homologous recombination repair deficiency and prostate cancer, the techniques used to determine homologous recombination repair status in prostate cancer, the crosstalk between homologous recombination repair and the androgen receptor pathway, the current evidence on PARP inhibitors activity in metastatic prostate cancer also in homologous recombination repair-proficient tumors, as well as emerging mechanisms of resistance to PARP inhibitors. The possibility of combination therapies including a PARP inhibitor is an attractive option, and more robust data are awaited from ongoing phase II and phase III trials outlined in this manuscript.
Collapse
Affiliation(s)
- Claudia Piombino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Stefania Pipitone
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elena Tonni
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Luciana Mastrodomenico
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marco Oltrecolli
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Cyrielle Tchawa
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Rossana Matranga
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Sara Roccabruna
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Elisa D’Agostino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Marta Pirola
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Francesca Bacchelli
- Clinical Trials Office, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Cristina Baschieri
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Roberto Sabbatini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (C.P.); (S.P.); (E.T.); (L.M.); (M.O.); (C.T.); (R.M.); (S.R.); (E.D.); (M.P.); (C.B.); (M.D.); (R.S.)
| |
Collapse
|
38
|
Shevach JW, Candelieri-Surette D, Lynch JA, Hubbard RA, Alba PR, Glanz K, Parikh RB, Maxwell KN. Racial Differences in Germline Genetic Testing Completion Among Males With Pancreatic, Breast, or Metastatic Prostate Cancers. J Natl Compr Canc Netw 2024; 22:237-243. [PMID: 38631387 PMCID: PMC11361447 DOI: 10.6004/jnccn.2023.7105] [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: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 04/19/2024]
Abstract
BACKGROUND Germline genetic testing is a vital component of guideline-recommended cancer care for males with pancreatic, breast, or metastatic prostate cancers. We sought to determine whether there were racial disparities in germline genetic testing completion in this population. PATIENTS AND METHODS This retrospective cohort study included non-Hispanic White and Black males with incident pancreatic, breast, or metastatic prostate cancers between January 1, 2019, and September 30, 2021. Two nationwide cohorts were examined: (1) commercially insured individuals in an administrative claims database, and (2) Veterans receiving care in the Veterans Health Administration. One-year germline genetic testing rates were estimated by using Kaplan-Meier methods. Cox proportional hazards regression was used to test the association between race and genetic testing completion. Causal mediation analyses were performed to investigate whether socioeconomic variables contributed to associations between race and germline testing. RESULTS Our cohort consisted of 7,894 males (5,142 commercially insured; 2,752 Veterans). One-year testing rates were 18.0% (95% CI, 16.8%-19.2%) in commercially insured individuals and 14.2% (95% CI, 11.5%-15.0%) in Veterans. Black race was associated with a lower hazard of testing among commercially insured individuals (adjusted hazard ratio [aHR], 0.73; 95% CI, 0.58-0.91; P=.005) but not among Veterans (aHR, 0.99; 95% CI, 0.75-1.32; P=.960). In commercially insured individuals, income (aHR, 0.90; 95% CI, 0.86-0.96) and net worth (aHR, 0.92; 95% CI, 0.86-0.98) mediated racial disparities, whereas education (aHR, 0.98; 95% CI, 0.94-1.01) did not. CONCLUSIONS Overall rates of guideline-recommended genetic testing are low in males with pancreatic, breast, or metastatic prostate cancers. Racial disparities in genetic testing among males exist in a commercially insured population, mediated by net worth and household income; these disparities are not seen in the equal-access Veterans Health Administration. Alleviating financial and access barriers may mitigate racial disparities in genetic testing.
Collapse
Affiliation(s)
- Jeffrey W. Shevach
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC
| | | | - Julie A. Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Rebecca A. Hubbard
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA
| | - Patrick R. Alba
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Karen Glanz
- Perelman School of Medicine and School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Ravi B. Parikh
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kara N. Maxwell
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
39
|
Kumsa FA, Fowke JH, Hashtarkhani S, White BM, Shrubsole MJ, Shaban-Nejad A. The association between neighborhood obesogenic factors and prostate cancer risk and mortality: the Southern Community Cohort Study. Front Oncol 2024; 14:1343070. [PMID: 38720808 PMCID: PMC11078097 DOI: 10.3389/fonc.2024.1343070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/18/2024] [Indexed: 05/12/2024] Open
Abstract
Background Prostate cancer is one of the leading causes of cancer-related mortality among men in the United States. We examined the role of neighborhood obesogenic attributes on prostate cancer risk and mortality in the Southern Community Cohort Study (SCCS). Methods From the total of 34,166 SCCS male participants, 28,356 were included in the analysis. We assessed the relationship between neighborhood obesogenic factors [neighborhood socioeconomic status (nSES) and neighborhood obesogenic environment indices including the restaurant environment index, the retail food environment index, parks, recreational facilities, and businesses] and prostate cancer risk and mortality by controlling for individual-level factors using a multivariable Cox proportional hazards model. We further stratified prostate cancer risk analysis by race and body mass index (BMI). Results Median follow-up time was 133 months [interquartile range (IQR): 103, 152], and the mean age was 51.62 (SD: ± 8.42) years. There were 1,524 (5.37%) prostate cancer diagnoses and 98 (6.43%) prostate cancer deaths during follow-up. Compared to participants residing in the wealthiest quintile, those residing in the poorest quintile had a higher risk of prostate cancer (aHR = 1.32, 95% CI 1.12-1.57, p = 0.001), particularly among non-obese men with a BMI < 30 (aHR = 1.46, 95% CI 1.07-1.98, p = 0.016). The restaurant environment index was associated with a higher prostate cancer risk in overweight (BMI ≥ 25) White men (aHR = 3.37, 95% CI 1.04-10.94, p = 0.043, quintile 1 vs. None). Obese Black individuals without any neighborhood recreational facilities had a 42% higher risk (aHR = 1.42, 95% CI 1.04-1.94, p = 0.026) compared to those with any access. Compared to residents in the wealthiest quintile and most walkable area, those residing within the poorest quintile (aHR = 3.43, 95% CI 1.54-7.64, p = 0.003) or the least walkable area (aHR = 3.45, 95% CI 1.22-9.78, p = 0.020) had a higher risk of prostate cancer death. Conclusion Living in a lower-nSES area was associated with a higher prostate cancer risk, particularly among Black men. Restaurant and retail food environment indices were also associated with a higher prostate cancer risk, with stronger associations within overweight White individuals. Finally, residing in a low-SES neighborhood or the least walkable areas were associated with a higher risk of prostate cancer mortality.
Collapse
Affiliation(s)
- Fekede Asefa Kumsa
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Jay H. Fowke
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Soheil Hashtarkhani
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Brianna M. White
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| | - Martha J. Shrubsole
- Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Arash Shaban-Nejad
- Department of Pediatrics, College of Medicine, The University of Tennessee Health Science Center (UTHSC) - Oak Ridge National Laboratory (ORNL) Center for Biomedical Informatics, Memphis, TN, United States
| |
Collapse
|
40
|
Kalampokis N, Zabaftis C, Spinos T, Karavitakis M, Leotsakos I, Katafigiotis I, van der Poel H, Grivas N, Mitropoulos D. Review on the Role of BRCA Mutations in Genomic Screening and Risk Stratification of Prostate Cancer. Curr Oncol 2024; 31:1162-1169. [PMID: 38534919 PMCID: PMC10969585 DOI: 10.3390/curroncol31030086] [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: 01/27/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Somatic and germline alterations can be commonly found in prostate cancer (PCa) patients. The aim of our present study was to perform a comprehensive review of the current literature in order to examine the impact of BRCA mutations in the context of PCa as well as their significance as genetic biomarkers. (2) Methods: A narrative review of all the available literature was performed. Only "landmark" publications were included. (3) Results: Overall, the number of PCa patients who harbor a BRCA2 mutation range between 1.2% and 3.2%. However, BRCA2 and BRCA1 mutations are responsible for most cases of hereditary PCa, increasing the risk by 3-8.6 times and up to 4 times, respectively. These mutations are correlated with aggressive disease and poor prognosis. Gene testing should be offered to patients with metastatic PCa, those with 2-3 first-degree relatives with PCa, or those aged < 55 and with one close relative with breast (age ≤ 50 years) or invasive ovarian cancer. (4) Conclusions: The individualized assessment of BRCA mutations is an important tool for the risk stratification of PCa patients. It is also a population screening tool which can guide our risk assessment strategies and achieve better results for our patients and their families.
Collapse
Affiliation(s)
- Nikolaos Kalampokis
- Department of Urology, G. Hatzikosta General Hospital, 45001 Ioannina, Greece;
| | - Christos Zabaftis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Theodoros Spinos
- Department of Urology, University of Patras Hospital, 26504 Patras, Greece;
| | - Markos Karavitakis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Leotsakos
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Katafigiotis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Henk van der Poel
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Nikolaos Grivas
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Dionysios Mitropoulos
- Department of Urology, Medical School, National & Kapodistrian University of Athens, 14122 Athens, Greece;
| |
Collapse
|
41
|
Amini AE, Salari K. Incorporating Genetic Risk Into Prostate Cancer Care: Implications for Early Detection and Precision Oncology. JCO Precis Oncol 2024; 8:e2300560. [PMID: 38412389 DOI: 10.1200/po.23.00560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/29/2024] Open
Abstract
The availability and cost of germline and somatic genetic testing have dramatically improved over the past two decades, enabling precision medicine approaches in oncology, with significant implications for prostate cancer (PCa) care. Roughly 12% of individuals with advanced disease are carriers of rare pathogenic germline variants that predispose to particularly aggressive and earlier-onset disease. Several of these variants are already established as clinically actionable by modern precision oncology therapeutics, while others may come to aid the selection of active surveillance, definitive local therapies, and systemic therapies. Concurrently, the number of common variants (ie, incorporated into polygenic risk scores) associated with PCa risk has continued to grow, but with several important considerations both at the intersection of race and ancestry and for early detection of aggressive disease. Family history has historically been used as a proxy for this inherited genetic risk of PCa, but recently emerging evidence examining this relation has shifted our understanding of how best to leverage this tool in PCa care. This review seeks to clarify and contextualize the existing and emerging precision oncology paradigms that use inherited genetic risk in PCa care, for both early detection and localized disease management.
Collapse
Affiliation(s)
- Andrew E Amini
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
42
|
Incorvaia L, Perez A, Marchetti C, Brando C, Gristina V, Cancelliere D, Pivetti A, Contino S, Di Giovanni E, Barraco N, Bono M, Giurintano A, Bazan Russo TD, Gottardo A, Cutaia S, Pedone E, Peri M, Corsini LR, Fanale D, Galvano A, Scambia G, Badalamenti G, Russo A, Bazan V. Theranostic biomarkers and PARP-inhibitors effectiveness in patients with non-BRCA associated homologous recombination deficient tumors: Still looking through a dirty glass window? Cancer Treat Rev 2023; 121:102650. [PMID: 37939446 DOI: 10.1016/j.ctrv.2023.102650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Breast cancer susceptibility gene 1 (BRCA1) and breast cancer susceptibility gene 2 (BRCA2) deleterious variants were the first and, still today, the main biomarkers of poly(ADP)ribose polymerase (PARP)-inhibitors (PARPis) benefit. The recent, increased, numbers of individuals referred for counseling and multigene panel testing, and the remarkable expansion of approved PARPis, not restricted to BRCA1/BRCA2-Pathogenic Variants (PVs), produced a strong clinical need for non-BRCA biomarkers. Significant limitations of the current testing and assays exist. The different approaches that identify the causes of Homologous Recombination Deficiency (HRD), such as the germline and somatic Homologous Recombination Repair (HRR) gene PVs, the testing showing its consequences, such as the genomic scars, or the novel functional assays such as the RAD51 foci testing, are not interchangeable, and should not be considered as substitutes for each other in clinical practice for guiding use of PARPi in non-BRCA, HRD-associated tumors. Today, the deeper knowledge on the significant relationship among all proteins involved in the HRR, not limited to BRCA, expands the possibility of a successful non-BRCA, HRD-PARPi synthetic lethality and, at the same time, reinforces the need for enhanced definition of HRD biomarkers predicting the magnitude of PARPi benefit.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Claudia Marchetti
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniela Cancelliere
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Alessia Pivetti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Silvia Contino
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Emilia Di Giovanni
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marco Bono
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Ambra Giurintano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Andrea Gottardo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Sofia Cutaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Marta Peri
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy.
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
43
|
Paulo P, Cardoso M, Brandão A, Pinto P, Falconi A, Pinheiro M, Cerveira N, Silva R, Santos C, Pinto C, Peixoto A, Maia S, Teixeira MR. Genetic landscape of homologous recombination repair genes in early-onset/familial prostate cancer patients. Genes Chromosomes Cancer 2023; 62:710-720. [PMID: 37436117 DOI: 10.1002/gcc.23190] [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: 03/28/2023] [Revised: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Prostate cancer (PrCa) is one of the three most frequent and deadliest cancers worldwide. The discovery of PARP inhibitors for the treatment of tumors with deleterious variants in homologous recombination repair (HRR) genes has placed PrCa on the roadmap of precision medicine. However, the overall contribution of HRR genes to the 10%-20% of carcinomas arising in men with early-onset/familial PrCa has not been fully clarified. We used targeted next-generation sequencing (T-NGS) covering eight HRR genes (ATM, BRCA1, BRCA2, BRIP1, CHEK2, NBN, PALB2, and RAD51C) and an analysis pipeline querying both small and large genomic variations to clarify their global and relative contribution to hereditary PrCa predisposition in a series of 462 early-onset/familial PrCa cases. Deleterious variants were found in 3.9% of the patients, with CHEK2 and ATM being the most frequently mutated genes (38.9% and 22.2% of the carriers, respectively), followed by PALB2 and NBN (11.1% of the carriers, each), and finally by BRCA2, RAD51C, and BRIP1 (5.6% of the carriers, each). Using the same NGS data, exonic rearrangements were found in two patients, one pathogenic in BRCA2 and one of unknown significance in BRCA1. These results contribute to clarify the genetic heterogeneity that underlies PrCa predisposition in the early-onset and familial disease, respectively.
Collapse
Affiliation(s)
- Paula Paulo
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Marta Cardoso
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Andreia Brandão
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Pedro Pinto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Ariane Falconi
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Manuela Pinheiro
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Nuno Cerveira
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Rui Silva
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
| | - Catarina Santos
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Carla Pinto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Ana Peixoto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
| | - Sofia Maia
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Medical Genetics Unit, Hospital Pediátrico de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Manuel R Teixeira
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP) /RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) /Porto Comprehensive Cancer Center, Porto, Portugal
- Department of Laboratory Genetics, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| |
Collapse
|
44
|
Peyrottes A, Rouprêt M, Fiard G, Fromont G, Barret E, Brureau L, Créhange G, Gauthé M, Baboudjian M, Renard-Penna R, Roubaud G, Rozet F, Sargos P, Ruffion A, Mathieu R, Beauval JB, De La Taille A, Ploussard G, Dariane C. [Early detection of prostate cancer: Towards a new paradigm?]. Prog Urol 2023; 33:956-965. [PMID: 37805291 DOI: 10.1016/j.purol.2023.09.016] [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: 05/03/2023] [Accepted: 09/12/2023] [Indexed: 10/09/2023]
Abstract
Prostate cancer (PCa) is a public health issue. The diagnostic strategy for PCa is well codified and assessed by digital rectal examination, PSA testing and multiparametric MRI, which may or may not lead to prostate biopsies. The formal benefit of organized PCa screening, studied more than 10 years ago at an international scale and for all incomers, is not demonstrated. However, diagnostic and therapeutic modalities have evolved since the pivotal studies. The contribution of MRI and targeted biopsies, the widespread use of active surveillance for unsignificant PCa, the improvement of surgical techniques and radiotherapy… have allowed a better selection of patients and strengthened the interest for an individualized approach, reducing the risk of overtreatment. Aiming to enhance coverage and access to screening for the population, the European Commission recently promoted the evaluation of an organized PCa screening strategy, including MRI. The lack of screening programs has become detrimental to the population and must shift towards an early detection policy adapted to the risk of each individual.
Collapse
Affiliation(s)
- A Peyrottes
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, membre junior, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, Hôpital Européen Georges-Pompidou, AP-HP Centre, Université de Paris, 20 rue Leblanc, 75015 Paris, France.
| | - M Rouprêt
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Sorbonne university, GRC 5 Predictive Onco-Uro, AP-HP, urology, Pitié-Salpétrière hospital, 75013 Paris, France
| | - G Fiard
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, Grenoble Alpes university hospital, université Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, Grenoble, France
| | - G Fromont
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of pathology, CHRU, 37000 Tours, France
| | - E Barret
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, institut mutualiste Montsouris, Paris, France
| | - L Brureau
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, CHU de Pointe-à-Pitre, university of Antilles, university of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), UMR S 1085, 97110 Pointe-à-Pitre, Guadeloupe
| | - G Créhange
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of radiotherapy, institut Curie, Paris, France
| | - M Gauthé
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Sintep nuclear medicine, 38100 Grenoble, France
| | - M Baboudjian
- Department of urology, La Conception Hospital, Aix-Marseille University, AP-HM, Marseille, France
| | - R Renard-Penna
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Sorbonne university, AP-HP, radiology, Pitie-Salpétrière hospital, 75013 Paris, France
| | - G Roubaud
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of medical oncology, institut Bergonié, 33000 Bordeaux, France
| | - F Rozet
- Sorbonne university, GRC 5 Predictive Onco-Uro, AP-HP, urology, Pitié-Salpétrière hospital, 75013 Paris, France; Department of urology, institut mutualiste Montsouris, Paris, France
| | - P Sargos
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of radiotherapy, institut Bergonié, 33000 Bordeaux, France
| | - A Ruffion
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, centre hospitalier Lyon Sud, hospices civils de Lyon, Lyon, France
| | - R Mathieu
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, CHU de Rennes, Rennes, France
| | - J-B Beauval
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, La Croix du Sud Hôpital, Quint-Fonsegrives, France
| | - A De La Taille
- Department of urology, university hospital Henri-Mondor, AP-HP, Créteil, France
| | - G Ploussard
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Department of urology, La Croix du Sud Hôpital, Quint-Fonsegrives, France
| | - C Dariane
- Comité de Cancérologie de l'Association Française d'Urologie, groupe prostate, maison de l'urologie, 11, rue Viète, 75017 Paris, France; Service d'urologie, Hôpital Européen Georges-Pompidou, AP-HP Centre, Université de Paris, 20 rue Leblanc, 75015 Paris, France
| |
Collapse
|
45
|
Chen JY, Wang PY, Liu MZ, Lyu F, Ma MW, Ren XY, Gao XS. Biomarkers for Prostate Cancer: From Diagnosis to Treatment. Diagnostics (Basel) 2023; 13:3350. [PMID: 37958246 PMCID: PMC10649216 DOI: 10.3390/diagnostics13213350] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Prostate cancer (PCa) is a widespread malignancy with global significance, which substantially affects cancer-related mortality. Its spectrum varies widely, from slow-progressing cases to aggressive or even lethal forms. Effective patient stratification into risk groups is crucial to therapeutic decisions and clinical trials. This review examines a wide range of diagnostic and prognostic biomarkers, several of which are integrated into clinical guidelines, such as the PHI, the 4K score, PCA3, Decipher, and Prolaris. It also explores the emergence of novel biomarkers supported by robust preclinical evidence, including urinary miRNAs and isoprostanes. Genetic alterations frequently identified in PCa, including BRCA1/BRCA2, ETS gene fusions, and AR changes, are also discussed, offering insights into risk assessment and precision treatment strategies. By evaluating the latest developments and applications of PCa biomarkers, this review contributes to an enhanced understanding of their role in disease management.
Collapse
Affiliation(s)
- Jia-Yan Chen
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (J.-Y.C.); (F.L.); (M.-W.M.); (X.-Y.R.)
| | - Pei-Yan Wang
- School of Information, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Ming-Zhu Liu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China;
| | - Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (J.-Y.C.); (F.L.); (M.-W.M.); (X.-Y.R.)
| | - Ming-Wei Ma
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (J.-Y.C.); (F.L.); (M.-W.M.); (X.-Y.R.)
| | - Xue-Ying Ren
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (J.-Y.C.); (F.L.); (M.-W.M.); (X.-Y.R.)
| | - Xian-Shu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China; (J.-Y.C.); (F.L.); (M.-W.M.); (X.-Y.R.)
| |
Collapse
|
46
|
Sciarra A, Santarelli V, Santodirocco L, Frisenda M, Salciccia S, Casale P, Forte F, Mariotti G, Moriconi M, Cattarino S, Sciarra B, Bevilacqua G, Gentilucci A. Is It Time to Anticipate the Use of PARP Inhibition in Prostate Cancer Patients? Curr Oncol 2023; 30:8054-8067. [PMID: 37754499 PMCID: PMC10528501 DOI: 10.3390/curroncol30090584] [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: 06/25/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
The increasing diffusion of genetic analysis regarding the pathogenetic variants (PVs) of genes involved in DNA Damage Repair (DDR) mechanisms and the development of Poly ADP ribose polymerase (PARP) inhibitors (PARPis) led to the first valid precision medicine option tailored toward metastatic prostate cancer (mPC). The concept of anticipation in the systemic treatment of mPC was initially adopted for androgen receptor signaling inhibitors (ARSIs) to describe the expansion of their indications, from the setting of the late-stage second-line treatment of metastatic castration-resistant prostate cancer (mCRPC) to first-line therapy in selected cases. There is already mounting evidence in favor of the anticipation of PARPis in the first line of mCRPC therapy, and further evidence in favor of mHSPC is emerging. Many studies have demonstrated the synergism between ARSIs and PARP inhibitors. Recent discoveries regarding the crosstalk between the androgen receptor (AR) and DNA repair mechanisms are disconnecting the use of PARPis from genetic analysis. The new message emerging is that the combination of PARPis with ARSIs may work independently of DDR mutational status. As a matter of fact, most of the recent trials analyzing the combination of PARPis with abiraterone or enzalutamide as a first-line therapy enrolled mCRPC patients irrespective of their mutational status. The PROPEL trial concluded that the advantage of the combination was independent of PV status, despite a higher advantage being reported in the BRCA1/2 mutated subgroup. The MAGNITUDE trial, however, showed a significant advantage only in the DDR mutated subgroup, and the DDR non-mutated cohort was closed for further enrollment. The combination of PARPis with ARSIs represents a significant strategy with a view to the anticipation and intensification of care in mPC. However, it should not nullify the advantages of precision medicine linked to the genetic analysis of DDR genes.
Collapse
Affiliation(s)
- Alessandro Sciarra
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Valerio Santarelli
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Lorenzo Santodirocco
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Marco Frisenda
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Stefano Salciccia
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Paolo Casale
- Urologic Division, Humanitas Hospital, Rozzano, 00100 Milan, Italy;
| | - Flavio Forte
- Urologic Division, Figliesancamillo Hospital, 00198 Rome, Italy;
| | - Gianna Mariotti
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Martina Moriconi
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Susanna Cattarino
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Beatrice Sciarra
- Department of Chemistry, University Sapienza, 00166 Rome, Italy;
| | - Giulio Bevilacqua
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| | - Alessandro Gentilucci
- Department Materno Infantile e Scienze Urologiche, University Sapienza, 00166 Rome, Italy; (V.S.); (L.S.); (M.F.); (S.S.); (G.M.); (M.M.); (S.C.); (G.B.); (A.G.)
| |
Collapse
|
47
|
Ueki A, Yoshida R, Kosaka T, Matsubayashi H. Clinical risk management of breast, ovarian, pancreatic, and prostatic cancers for BRCA1/2 variant carriers in Japan. J Hum Genet 2023; 68:517-526. [PMID: 37088789 DOI: 10.1038/s10038-023-01153-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/21/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Opportunities for genetic counseling and germline BRCA1/2 (BRCA) testing are increasing in Japan owing to cancer genomic profiling testing and companion diagnostics being covered by national health insurance for patients with BRCA-related cancers. These tests are useful not only to judge whether platinum agents and PARP inhibitors are indicated but also to reveal an autosomal-dominant inherited cancer syndrome: hereditary breast and ovarian cancer. In individuals with germline BRCA variants, risk of cancers of the breast, ovary, pancreas, and prostate is significantly increased at various ages of onset, but the stomach, uterus, biliary tract, and skin might also be at risk. For women with pathogenic BRCA variants, breast awareness and image analyses should be initiated in their 20s, and risk-reducing procedures such as mastectomy are recommended starting in their 30s, with salpingo-oophorectomy in their late 30s. For male BRCA pathogenic variant carriers, prostatic surveillance should be applied using serum prostate-specific antigen starting in their 40s. For both sexes, image examinations ideally using endoscopic ultrasound and magnetic resonance cholangiopancreatography and blood testing should begin in their 50s for pancreatic surveillance. Homologous recombination pathway-associated genes are also causative candidates. Variant pathogenicity needs to be evaluated every 6-12 months when results are uncertain for clinical significance. Genetic counseling needs to be offered to the blood relatives of the pathogenic variant carriers with suitable timing. We review the recommended cross-organ BRCA risk management in Japan.
Collapse
Affiliation(s)
- Arisa Ueki
- Department of Clinical Genetics, The Cancer Institute Hospital of JFCR, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Reiko Yoshida
- Institute for Clinical Genetics and Genomics, Showa University, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan.
| |
Collapse
|
48
|
Grypari IM, Tzelepi V, Gyftopoulos K. DNA Damage Repair Pathways in Prostate Cancer: A Narrative Review of Molecular Mechanisms, Emerging Biomarkers and Therapeutic Targets in Precision Oncology. Int J Mol Sci 2023; 24:11418. [PMID: 37511177 PMCID: PMC10380086 DOI: 10.3390/ijms241411418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer (PCa) has a distinct molecular signature, including characteristic chromosomal translocations, gene deletions and defective DNA damage repair mechanisms. One crucial pathway involved is homologous recombination deficiency (HRD) and it is found in almost 20% of metastatic castrate-resistant PCa (mCRPC). Inherited/germline mutations are associated with a hereditary predisposition to early PCa development and aggressive behavior. BRCA2, ATM and CHECK2 are the most frequently HRD-mutated genes. BRCA2-mutated tumors have unfavorable clinical and pathological characteristics, such as intraductal carcinoma. PARP inhibitors, due to the induction of synthetic lethality, have been therapeutically approved for mCRPC with HRD alterations. Mutations are detected in metastatic tissue, while a liquid biopsy is utilized during follow-up, recognizing acquired resistance mechanisms. The mismatch repair (MMR) pathway is another DNA repair mechanism implicated in carcinogenesis, although only 5% of metastatic PCa is affected. It is associated with aggressive disease. PD-1 inhibitors have been used in MMR-deficient tumors; thus, the MMR status should be tested in all metastatic PCa cases. A surrogate marker of defective DNA repair mechanisms is the tumor mutational burden. PDL-1 expression and intratumoral lymphocytes have ambivalent predictive value. Few experimental molecules have been so far proposed as potential biomarkers. Future research may further elucidate the role of DNA damage pathways in PCa, revealing new therapeutic targets and predictive biomarkers.
Collapse
Affiliation(s)
- Ioanna-Maria Grypari
- Cytology Department, Aretaieion University Hospital, National Kapodistrian University of Athens, 11528 Athens, Greece
| | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Kostis Gyftopoulos
- Department of Anatomy, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
49
|
Wei JT, Barocas D, Carlsson S, Coakley F, Eggener S, Etzioni R, Fine SW, Han M, Kim SK, Kirkby E, Konety BR, Miner M, Moses K, Nissenberg MG, Pinto PA, Salami SS, Souter L, Thompson IM, Lin DW. Early Detection of Prostate Cancer: AUA/SUO Guideline Part I: Prostate Cancer Screening. J Urol 2023; 210:46-53. [PMID: 37096582 PMCID: PMC11060750 DOI: 10.1097/ju.0000000000003491] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE The summary presented herein covers recommendations on the early detection of prostate cancer and provides a framework to facilitate clinical decision-making in the implementation of prostate cancer screening, biopsy, and follow-up. This is Part I of a two-part series that focuses on prostate cancer screening. Please refer to Part II for discussion of initial and repeat biopsies as well as biopsy technique. MATERIALS AND METHODS The systematic review utilized to inform this guideline was conducted by an independent methodological consultant. The systematic review was based on searches in Ovid MEDLINE and Embase and Cochrane Database of Systematic Reviews (January 1, 2000-November 21, 2022). Searches were supplemented by reviewing reference lists of relevant articles. RESULTS The Early Detection of Prostate Cancer Panel developed evidence- and consensus-based guideline statements to provide guidance in prostate cancer screening, initial and repeat biopsy, and biopsy technique. CONCLUSIONS Prostate-specific antigen (PSA)-based prostate cancer screening in combination with shared decision-making (SDM) is recommended. Current data regarding risk from population-based cohorts provide a basis for longer screening intervals and tailored screening, and the use of available online risk calculators is encouraged.
Collapse
Affiliation(s)
- John T Wei
- University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | - Ruth Etzioni
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Samson W Fine
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Misop Han
- Johns Hopkins University, Baltimore, Maryland
| | - Sennett K Kim
- American Urological Association, Linthicum, Maryland
| | - Erin Kirkby
- American Urological Association, Linthicum, Maryland
| | | | | | | | - Merel G Nissenberg
- National Alliance of State Prostate Cancer Coalitions, Los Angeles, California
| | | | | | - Lesley Souter
- Nomadic EBM Methodology, Smithville, Ontario, Canada
| | | | | |
Collapse
|
50
|
Lukashchuk N, Barnicle A, Adelman CA, Armenia J, Kang J, Barrett JC, Harrington EA. Impact of DNA damage repair alterations on prostate cancer progression and metastasis. Front Oncol 2023; 13:1162644. [PMID: 37434977 PMCID: PMC10331135 DOI: 10.3389/fonc.2023.1162644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
Prostate cancer is among the most common diseases worldwide. Despite recent progress with treatments, patients with advanced prostate cancer have poor outcomes and there is a high unmet need in this population. Understanding molecular determinants underlying prostate cancer and the aggressive phenotype of disease can help with design of better clinical trials and improve treatments for these patients. One of the pathways often altered in advanced prostate cancer is DNA damage response (DDR), including alterations in BRCA1/2 and other homologous recombination repair (HRR) genes. Alterations in the DDR pathway are particularly prevalent in metastatic prostate cancer. In this review, we summarise the prevalence of DDR alterations in primary and advanced prostate cancer and discuss the impact of alterations in the DDR pathway on aggressive disease phenotype, prognosis and the association of germline pathogenic alterations in DDR genes with risk of developing prostate cancer.
Collapse
Affiliation(s)
- Natalia Lukashchuk
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Alan Barnicle
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Carrie A. Adelman
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Joshua Armenia
- Oncology Data Science, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Jinyu Kang
- Global Medicines Development, Oncology Research and Development (R&D), AstraZeneca, Gaithersburg, MD, United States
| | - J. Carl Barrett
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Waltham, MA, United States
| | - Elizabeth A. Harrington
- Translational Medicine, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|