1
|
Shore N, Nielsen SM, Esplin ED, Antonarakis ES, Barata PC, Beer TM, Beltran H, Bryce A, Cookson MS, Crawford ED, Dorff TB, George DJ, Heath EI, Helfand BT, Hussain M, Mckay RR, Morgans AK, Morris MJ, Paller CJ, Ross AE, Sartor O, Shen J, Sieber P, Smith MR, Wise DR, Armstrong AJ. Implementation of Universal Germline Genetic Testing Into Standard of Care for Patients With Prostate Cancer: The Time Is Now. JCO Oncol Pract 2024:OP2400626. [PMID: 39700441 DOI: 10.1200/op-24-00626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Indications for and implications of germline genetic testing (GGT) in patients with prostate cancer have expanded over the past decade, particularly related to precision therapies and management. GGT has become the standard of care for many cancers such as breast, ovarian, colorectal, pancreatic, and metastatic prostate cancer, and it is imperative that patients be offered timely and equitable access to testing as it can inform patient-physician shared decision making for management of the current cancer as well as anticipatory guidance for disease progression. Additionally, GGT guides screening for and prevention of secondary malignancies for the patient and cascade testing for at-risk family members. Here, we present data supporting the notion that clinicians should offer all patients with prostate cancer the opportunity to undergo comprehensive GGT for pathogenic germline variants known to be associated with familial cancer and/or known to have implications for treatment and management.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | - Sarah M Nielsen
- Labcorp Genetics Inc (formerly Invitae Corp), San Francisco, CA
| | - Edward D Esplin
- Labcorp Genetics Inc (formerly Invitae Corp), San Francisco, CA
| | | | | | - Tomasz M Beer
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR
- Exact Sciences Corporation, Madison, WI
| | | | - Alan Bryce
- City of Hope Cancer Center, Goodyear, AZ
| | - Michael S Cookson
- Stephenson Cancer Center, OU Health, The University of Oklahoma, Oklahoma City, OK
| | | | | | | | | | - Brian T Helfand
- NorthShore University HealthSystem/Endeavor Health, Evanston, IL
| | - Maha Hussain
- Northwestern Feinberg School of Medicine, Chicago, IL
| | | | | | | | - Channing J Paller
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ashley E Ross
- Northwestern Feinberg School of Medicine, Chicago, IL
| | | | - John Shen
- UCLA David Geffen School of Medicine, Los Angeles, CA
| | | | | | - David R Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | | |
Collapse
|
2
|
Zayhowski K, Wang C, Nahorniak M, Loo S, Gignac G, Gunn C. A qualitative study of the experiences of patients with prostate cancer when receiving negative genetic results: "I still don't have a grasp of what it all means". J Genet Couns 2024. [PMID: 39611226 DOI: 10.1002/jgc4.2003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Germline genetic testing has been increasingly conducted for treatment implications in patients with prostate cancer due to the expansion of testing eligibility. Understanding patients' comprehension of genetic results is crucial for establishing effective result disclosure practices. This importance has grown due to the increasing prevalence of negative genetic results being conveyed via electronic communication and by providers without a genetics specialization. This study explores patients with prostate cancer's perceptions of genetic results communication. We analyzed 24 qualitative, semi-structured interviews with patients with prostate cancer at an urban safety-net hospital who had genetic results documented in their medical records. Interview questions focused on patient experiences with genetic referrals, genetic counseling, and genetic result disclosure. Audio recordings were professionally transcribed and analyzed by the study team utilizing an inductive thematic approach to generate themes from recurring codes. Of those who participated, 18 were interviewed in English, 5 in Spanish, and 1 in Haitian Creole. No participants reported having a pathogenic variant identified with genetic testing. Study participants identified a number of gaps in results communication which led to misconceptions regarding hereditary cancer risk. Three themes were generated: (1) Patients desired clear communication about the next steps after genetic testing, (2) Patients commonly experienced cognitive dissonance with negative genetic results given personal and family history of cancer, and (3) Patients felt reassurance from negative genetic results. This research suggests that maintaining conversations between patients and healthcare providers alongside the delivery of negative results assists in patient comprehension. Additionally, it is essential to evaluate the accessibility and appropriateness of notes and results sent to patients. Ultimately, understanding communication barriers in genetic results return is imperative in order to provide high-quality genetic care.
Collapse
Affiliation(s)
- Kimberly Zayhowski
- Department of Obstetrics and Gynecology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Catharine Wang
- Department of Community Health Sciences, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Mary Nahorniak
- Master's Program in Genetic Counseling, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Stephanie Loo
- Department of Health Law, Policy, and Management, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Gretchen Gignac
- Department of Hematology and Oncology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
- The Cancer Care Center, Boston Medical Center, Boston, Massachusetts, USA
| | - Christine Gunn
- Department of Health Law, Policy, and Management, Boston University School of Public Health, Boston, Massachusetts, USA
- Dartmouth Institute for Health Policy and Clinical Practice, Dartmouth Cancer Center, Lebanon, New Hampshire, USA
- Section of General Internal Medicine, Evans Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Russo P, Bizzarri FP, Filomena GB, Marino F, Iacovelli R, Ciccarese C, Boccuto L, Ragonese M, Gavi F, Rossi F, Savoia C, Suraci PP, Falabella R, Pandolfo SD, Napolitano L, Leoni C, Trevisan V, Palermo G, Racioppi M, Sacco E, Muselaers S, Foschi N. Relationship Between Loss of Y Chromosome and Urologic Cancers: New Future Perspectives. Cancers (Basel) 2024; 16:3766. [PMID: 39594721 PMCID: PMC11593089 DOI: 10.3390/cancers16223766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The Y chromosome (ChrY) is essential for male sex determination and spermatogenesis. However, recent studies have revealed its broader role in various physiological processes and disease susceptibility, including cancer. Methods: A comprehensive literature review was conducted using databases like MEDLINE, Scopus, Web of Science, and Google Scholar. The review included clinical and preclinical studies in animals and humans focusing on the role of LoY in urological tumors. Data on the frequency of LoY, its clinical implications, and underlying mechanisms were extracted and analyzed. Results: The evidence suggests that LoY is associated with an increased risk of urologic neoplasms, potentially serving as an early marker of genomic instability. Studies reveal that LoY in urologic cancers correlates with worse survival outcomes and may contribute to tumor progression. LoY may interfere with chromatin structure and epigenetic regulation, suggesting its role as a contributor to early tumorigenesis. Conclusions: LoY appears to be a structural aberration with unique biological and clinical relevance in urologic cancers, possibly serving as a biomarker for genomic instability. Further research is necessary to identify specific Y-linked genes affected by LoY, potentially informing targeted therapies and early diagnostic strategies for these cancers.
Collapse
Affiliation(s)
- Pierluigi Russo
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Pio Bizzarri
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
- Department of Urology, Ospedale Isola Tiberina—Gemelli Isola, 00168 Rome, Italy
- Department of Urology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Giovanni Battista Filomena
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
- Department of Urology, “F. Miulli” General Hospital, 70021 Acquaviva Delle Fonti, BA, Italy
| | - Filippo Marino
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
- Department of Urology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Roberto Iacovelli
- Department of Oncology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Chiara Ciccarese
- Department of Oncology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy
| | - Luigi Boccuto
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, SC 29634, USA
| | - Mauro Ragonese
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
| | - Filippo Gavi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Rossi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
- Department of Urology, Ospedale Isola Tiberina—Gemelli Isola, 00168 Rome, Italy
| | - Cosimo Savoia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Paolo Pietro Suraci
- Unit of Urology, Department of Medical-Surgical Sciences and Biotechnologies, Istituto Chirurgico Ortopedico Traumatologico Hospital, University of Rome Sapienza, Via F. Faggiana 1668, 04100 Latina, Italy
| | | | - Savio Domenico Pandolfo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80138 Naples, Italy
| | - Luigi Napolitano
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80138 Naples, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Valentina Trevisan
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe Palermo
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
| | - Marco Racioppi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Emilio Sacco
- Department of Urology, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
- Department of Urology, Ospedale Isola Tiberina—Gemelli Isola, 00168 Rome, Italy
| | - Stijn Muselaers
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Nazario Foschi
- Department of Urology, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, 00168 Rome, Italy or (P.R.); (G.P.); (N.F.)
| |
Collapse
|
4
|
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
|
5
|
Custodio-Cabello S, Pacheco-Barcia V, Palka-Kotlowska M, Fernández-Hernández L, Del Álamo JF, Oliveros-Acebes E, Cabezón-Gutiérrez L. Prognostic value of germline mutations in metastatic hormone-sensitive prostate cancer (mHSPC). Urol Oncol 2024; 42:331.e13-331.e24. [PMID: 38926076 DOI: 10.1016/j.urolonc.2024.05.010] [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: 01/28/2024] [Revised: 05/02/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND About 8% to 12% of patients presenting with mHSPC exhibit germline pathogenic variants (PV) in cancer predisposition genes. The aim of this study is to assess the presence of germline PV as a prognostic factor in the setting of mHSPC and to determine whether mutational status can predict rapid progression to castration resistance. METHODS Genetic analysis using a multigene next-generation sequencing (NGS) panel was performed on 34 patients diagnosed with mHSPC undergoing treatment. We assessed the prevalence of germline PV and examined differences based on clinical-pathological characteristics, family history (FH), prostate-specific antigen (PSA) response, impact on time to castration-resistant prostate cancer (TTCRPC), and overall survival (OS). RESULTS Germline PV were identified in 6 patients (17,6%). When comparing the clinical-pathological characteristics of PV carriers (n = 6) to noncarriers (n = 28), no significant associations were observed except for the presence of FH of hereditary breast and ovarian cancer (HBOC) syndrome and/or Lynch syndrome (P = 0.024). At a median follow-up of 33 months, significant differences in OS were observed based on the presence of PV (26 months in carriers vs. 74 months in noncarriers; P < 0.01). Patients who harbored a BRCA2 mutation (n = 3) showed a worse clinical outcome, presenting a shorter TTCRPC (7 months vs. 23 months; P = 0.005) and lower OS (7 months vs. 74 months; P < 0.001) compared to noncarriers (n = 31). CONCLUSION mHSPC germline PV carriers had a worse survival outcome. Furthermore, BRCA2 germline mutation was an independent poor prognostic factor for mHSPC disease, associated with earlier progression to castration-resistant prostate cancer, and shorter OS. These results highlight the importance of evaluating germline mutational status in patients with hormone-sensitive prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Luis Cabezón-Gutiérrez
- Medical Oncology, Hospital Universitario De Torrejón, Madrid, Spain; Faculty of Medicine, Francisco de Vitoria University, Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Huang J, Sun J, Wang K, Zheng L, Fan Y, Qian B. Causal relationship between prostatic diseases and prostate cancer: a mendelian randomization study. BMC Cancer 2024; 24:774. [PMID: 38937672 PMCID: PMC11210166 DOI: 10.1186/s12885-024-12551-9] [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: 10/29/2023] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Although it is thought that prostatitis or benign prostatic hyperplasia (BPH) is related to prostate cancer (PCa), the underlying causal effects of these diseases are unclear. METHODS We assessed the causal relationship between prostatitis or BPH and PCa using a two-sample Mendelian randomization (MR) approach. The data utilized in this study were sourced from genome-wide association study. The association of genetic variants from cohorts of prostatitis or BPH and PCa patients was determined using inverse-variance weighted and MR Egger regression techniques. The direction of chance was determined using independent genetic variants with genome-wide significance (P < 5 × 10-6). The accuracy of the results was confirmed using sensitivity analyses. RESULTS MR analysis showed that BPH had a significant causal effect on PCa (Odds Ratio = 1.209, 95% Confidence Interval: 0.098-0.281, P = 5.079 × 10- 5) while prostatitis had no significant causal effect on PCa (P > 0.05). Additionally, the pleiotropic test and leave-one-out analysis showed the two-sample MR analyses were valid and reliable. CONCLUSIONS This MR study supports that BPH has a positive causal effect on PCa, while genetically predicted prostatitis has no causal effect on PCa. Nonetheless, further studies should explore the underlying biochemical mechanism and potential therapeutic targets for the prevention of these diseases.
Collapse
Affiliation(s)
- Jiaguo Huang
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Ji Sun
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Kai Wang
- Department of Urology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Yi Fan
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China.
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical College, Ganzhou, China.
| |
Collapse
|
9
|
Maekawa S, Takata R, Sekiguchi K, Kagabu M, Toyoshima M, Tamada S, Takahashi K, Ikarashi D, Matsuura T, Kato R, Kato Y, Kanehira M, Sugimura J, Abe T, Baba T, Obara W. Prevalence of germline BRCA1/2 pathogenic variants in Japanese patients treated with castration-resistant prostate cancer and efficacy of CRPC treatment in real-world clinical practice. Jpn J Clin Oncol 2024; 54:489-497. [PMID: 38157885 DOI: 10.1093/jjco/hyad185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE The companion diagnosis for olaparib, a poly (ADP-ribose) polymerase inhibitor for prostate cancer, aims to detect BRCA1/2 gene variants. In clinical practice, the frequency of germline BRCA1/2 variants in patients receiving castration-resistant prostate cancer treatment is unknown. We aimed to evaluate the prevalence of germline BRCA1/2 variants and their relationship to prognosis and treatment efficacy in castration-resistant prostate cancer. METHODS Between June 2021 and 2023, 92 patients receiving castration-resistant prostate cancer treatment were examined for germline BRCA1/2 variants using BRACAnalysis CDx®. Furthermore, the associations between BRCA1/2 pathogenic variants and clinical outcomes were assessed. RESULTS Of the 92 patients referred for genetic testing, 6 (6.5%) carried germline pathogenic variants in BRCA1/2. The BRCA2 variant was the most frequent (n = 5), followed by BRCA1 variant (n = 1). Among the five variants in BRCA2, the p.Asp427Thrfs*3 variant was identified for the first time in prostate cancer. Overall survival from castration-resistant prostate cancer for patients with BRCA1/2 variants was significantly shorter than for patients without BRCA1/2 variants (P = 0.043). Progression-free survival of androgen receptor signaling inhibitors for patients with BRCA1/2 variants was significantly shorter than for those without (P = 0.003). Progression-free survival of taxane chemotherapy was significantly shorter in patients with BRCA1/2 variants than in those without (P = 0.0149). CONCLUSIONS In clinical practice, 6.5% of patients treated with castration-resistant prostate cancer carried germline BRCA1/2 pathogenic variants. Japanese castration-resistant prostate cancer patients with germline BRCA1/2 mutants have a poor prognosis and may be less responsive to treatment with androgen receptor signaling inhibitors and taxane-based chemotherapy for castration-resistant prostate cancer.
Collapse
Affiliation(s)
| | - Ryo Takata
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Kie Sekiguchi
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Masahiro Kagabu
- Department of Obstetrics & Gynecology, Iwate Medical University, Iwate, Japan
| | - Moe Toyoshima
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Shinji Tamada
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Kenta Takahashi
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Daiki Ikarashi
- Department of Urology, Iwate Medical University, Iwate, Japan
| | | | - Renpei Kato
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Yoichiro Kato
- Department of Urology, Iwate Medical University, Iwate, Japan
| | | | - Jun Sugimura
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Takaya Abe
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Tsukasa Baba
- Department of Obstetrics & Gynecology, Iwate Medical University, Iwate, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University, Iwate, Japan
| |
Collapse
|
10
|
Sutherland DEK, Azad AA, Murphy DG, Eapen RS, Kostos L, Hofman MS. Role of FDG PET/CT in Management of Patients with Prostate Cancer. Semin Nucl Med 2024; 54:4-13. [PMID: 37400321 DOI: 10.1053/j.semnuclmed.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
Prostate cancer is the second most common cancer in men worldwide. [18F]FDG PET/CT imaging, a well-known and effective technique for detecting malignancies, has not been considered a useful tool for prostate cancer imaging by many because of its perceived low [18F]FDG uptake. Incidentally detected focal [18F]FDG uptake in the prostate is not uncommon, and typically benign. Imaging features that would increase concern for an underlying prostatic carcinoma, include focal uptake in the periphery near the gland margin without calcifications. [18F]FDG PET/CT imaging provides little value in the initial staging of prostate cancer, particularly in the era of prostate specific membrane antigen (PSMA) radiotracer. In cases of biochemical recurrence, the value of [18F]FDG PET/CT increases notably when Grade group 4 or 5 and elevated PSA levels are present. Active research is underway for theranostic approaches to prostate cancer, including [177Lu]Lu-PSMA therapy. Dual tracer staging using FDG and PSMA imaging significantly enhances the accuracy of disease site assessment. Specifically, the addition of [18F]FDG PET/CT imaging allows for the evaluation of discordant disease (PSMA negative/FDG positive). The maximal benefit from [177Lu]Lu-PSMA therapy relies on significant PSMA accumulation across all disease sites, and the identification of discordant disease suggests that these patients may derive less benefit from the treatment. The genuine value of [18F]FDG PET/CT imaging lies in advanced prostate cancer, PSMA-negative disease, as a prognostic biomarker, and the realm of new targeted theranostic agents.
Collapse
Affiliation(s)
- Duncan E K Sutherland
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Arun A Azad
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Renu S Eapen
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Kostos
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Al-Toubat M, Serrano S, Elshafei A, Koul K, Feibus AH, Balaji KC. Metastatic prostate cancer is associated with distinct higher frequency of genetic mutations at diagnosis. Urol Oncol 2023; 41:455.e7-455.e15. [PMID: 37838503 DOI: 10.1016/j.urolonc.2023.09.014] [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: 06/26/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/16/2023]
Abstract
INTRODUCTION AND OBJECTIVES We explored characteristic genetic mutations associated with metastatic prostate cancer (PCa) by comparing next generation sequencing (NGS) data between men with or without metastatic disease at diagnosis. METHODS We queried the American Association for Cancer Research Project Genomics Evidence Neoplasia Information Exchange (GENIE) registry for men diagnosed with PCa. Patients were categorized into with (M1) or without metastatic disease (M0) groups. The difference in the frequency of genetic mutations between the two groups and the prognostic significance of the mutations were analyzed using SPSS V28. We included frequency rate of > 5% and P values < 0.05 were considered statistically significant to maintain over 95% true positive detection rate. RESULTS Of a total of 10,580 patients with diagnosis of PCa in the dataset, we selected a study cohort of 1268 patients without missing data; 700 (55.2%) had nonmetastatic PCa, 421 (33.2%) and 147 (11.6%) patients had metastatic castration sensitive and resistant PCa respectively. The median age at diagnosis and serum prostate specific antigen (PSA) level for the entire cohort was 62.8 years (IQR 56.3-68.4) and 8.0 ng/ml (IQR 4.9-20.9) respectively. A vast majority of the cohort were of Caucasian ancestry (89.1%). Of a total of 561 genes sequenced, there were mutations in 79 genes (14.1%). The mutation frequency was significantly higher in M1PCa compared to M0PCa, 35.7% and 23.3%, respectively (P = <0.001). The median tumor mutational burden was also significantly higher in the samples from M1PCa (2.59 mut/MB) compared to M0PCa (1.96 mut/MB) (P < 0.001). Compared to M0PCa patients, M1PCa patients demonstrated significantly higher rate of genetic mutations; TP53 (38.73% vs. 17.71% P < 0.001), PTEN (25.70% vs. 11.71% P < 0.001), AR (17.25% vs. 1.43% P < 0.001), APC (11.8% vs. 4.43% P < 0.001), TMPRSS2 (31.5% vs. 11.14% P < 0.001), ERG (23.59% vs. 13.13% P < 0.001), FOXA1 (17.43% vs. 6.33% P < 0.001), MYC (8.45% vs. 2.29% P < 0.001), RB1 (10.39% vs. 2.43% P < 0.001) and CDK12 (8.45% vs. 1.31% P < 0.001). Of the various cellular signaling pathways, the androgen receptor signaling pathway was most often impacted. In the cohort with M1 disease, compared to men without genetic mutations the men with genetic mutations demonstrated worse survival (P = <0.001, log rank test). Compared to castration sensitive M1 patients, AR (57% vs. 4% P < 0.001), TP53 (50.7% vs. 34% P < 0.001), PTEN (35.2% vs. 22.1% P < 0.001), RB1(23.9% vs. 4.75% P < 0.001) were significantly more frequently mutated in castration resistant M1 patients. In contrast, mutations of SPOP (13.3% vs. 7.9% P < 0.001), FOXA1 (17.6% vs. 5.3% P < 0.001) and CDK12 (12% vs. 6.45% P < 0.001) were significantly more frequently found in castration sensitive M1 patients compared to castration resistant patients. CONCLUSION Patients with M1PCa demonstrated characteristic genetic mutations compared to M0PCa, which most often influenced androgen receptor signaling and is associated with worse survival. In addition, we identified distinct genetic mutations between castration sensitive and resistant M1PCa. These findings may be used to further our understanding and management of men with PCa.
Collapse
Affiliation(s)
- Mohammed Al-Toubat
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Samuel Serrano
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Ahmed Elshafei
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Kashyap Koul
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - Allison H Feibus
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL
| | - K C Balaji
- Department of Urology, University of Florida College of Medicine, Jacksonville, FL.
| |
Collapse
|
12
|
Hanson H, Astiazaran-Symonds E, Amendola LM, Balmaña J, Foulkes WD, James P, Klugman S, Ngeow J, Schmutzler R, Voian N, Wick MJ, Pal T, Tischkowitz M, Stewart DR. Management of individuals with germline pathogenic/likely pathogenic variants in CHEK2: A clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2023; 25:100870. [PMID: 37490054 PMCID: PMC10623578 DOI: 10.1016/j.gim.2023.100870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/26/2023] Open
Abstract
PURPOSE Although the role of CHEK2 germline pathogenic variants in cancer predisposition is well known, resources for managing CHEK2 heterozygotes in clinical practice are limited. METHODS An international workgroup developed guidance on clinical management of CHEK2 heterozygotes informed by peer-reviewed publications from PubMed. RESULTS Although CHEK2 is considered a moderate penetrance gene, cancer risks may be considered as a continuous variable, which are influenced by family history and other modifiers. Consequently, early cancer detection and prevention for CHEK2 heterozygotes should be guided by personalized risk estimates. Such estimates may result in both downgrading lifetime breast cancer risks to those similar to the general population or upgrading lifetime risk to a level at which CHEK2 heterozygotes are offered high-risk breast surveillance according to country-specific guidelines. Risk-reducing mastectomy should be guided by personalized risk estimates and shared decision making. Colorectal and prostate cancer surveillance should be considered based on assessment of family history. For CHEK2 heterozygotes who develop cancer, no specific targeted medical treatment is recommended at this time. CONCLUSION Systematic prospective data collection is needed to establish the spectrum of CHEK2-associated cancer risks and to determine yet-unanswered questions, such as the outcomes of surveillance, response to cancer treatment, and survival after cancer diagnosis.
Collapse
Affiliation(s)
- Helen Hanson
- Southwest Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Esteban Astiazaran-Symonds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; Department of Medicine, College of Medicine-Tucson, University of Arizona, Tucson, AZ
| | | | - Judith Balmaña
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medical Oncology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - William D Foulkes
- Departments of Human Genetics, Oncology and Medicine, McGill University, Montréal, QC, Canada
| | - Paul James
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia; Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Susan Klugman
- Division of Reproductive & Medical Genetics, Department of Obstetrics & Gynecology and Women's Health, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Joanne Ngeow
- Genomic Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Rita Schmutzler
- Center of Integrated Oncology (CIO), University of Cologne, Cologne, Germany; Center for Hereditary Breast and Ovarian Cancer, University Hospital of Cologne, Cologne, Germany
| | - Nicoleta Voian
- Providence Genetic Risk Clinic, Providence Cancer Institute, Portland, OR
| | - Myra J Wick
- Departments of Obstetrics and Gynecology and Clinical Genomics, Mayo Clinic, Rochester, MN
| | - Tuya Pal
- Department of Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
13
|
Shore N, Gazi M, Pieczonka C, Heron S, Modh R, Cahn D, Belkoff LH, Berger A, Mazzarella B, Veys J, Idom C, Morris D, Jayram G, Engelman A, Bukkapatnam R, Dato P, Bevan-Thomas R, Cornell R, Wise DR, Hardwick MK, Hernandez RD, Rojahn S, Layman P, Hatchell KE, Heald B, Nussbaum RL, Nielsen SM, Esplin ED. Efficacy of National Comprehensive Cancer Network Guidelines in Identifying Pathogenic Germline Variants Among Unselected Patients with Prostate Cancer: The PROCLAIM Trial. Eur Urol Oncol 2023; 6:477-483. [PMID: 37574391 DOI: 10.1016/j.euo.2023.07.008] [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: 03/16/2023] [Revised: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Prostate cancer (PCa) patients with pathogenic/likely pathogenic germline variants (PGVs) in cancer predisposition genes may be eligible for U.S. Food and Drug Administration-approved targeted therapies, clinical trials, or enhanced screening. Studies suggest that eligible patients are missing genetics-informed care due to restrictive testing criteria. OBJECTIVE To establish the prevalence of actionable PGVs among prospectively accrued, unselected PCa patients, stratified by their guideline eligibility. DESIGN, SETTING, AND PARTICIPANTS Consecutive, unselected PCa patients were enrolled at 15 sites in the USA from October 2019 to August 2021, and had multigene cancer panel testing. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Correlates between the prevalence of PGVs and clinician-reported demographic and clinical characteristics were examined. RESULTS AND LIMITATIONS Among 958 patients (median [quartiles] age at diagnosis 65 [60, 71] yr), 627 (65%) had low- or intermediate-risk disease (grade group 1, 2, or 3). A total of 77 PGVs in 17 genes were identified in 74 patients (7.7%, 95% confidence interval [CI] 6.2-9.6%). No significant difference was found in the prevalence of PGVs among patients who met the 2019 National Comprehensive Cancer Network Prostate criteria (8.8%, 43/486, 95% CI 6.6-12%) versus those who did not (6.6%, 31/472, 95% CI 4.6-9.2%; odds ratio 1.38, 95% CI 0.85-2.23), indicating that these criteria would miss 42% of patients (31/74, 95% CI 31-53%) with PGVs. The criteria were less effective at predicting PGVs in patients from under-represented populations. Most PGVs (81%, 60/74) were potentially clinically actionable. Limitations include the inability to stratify analyses based on individual ethnicity due to low numbers of non-White patients with PGVs. CONCLUSIONS Our results indicate that almost half of PCa patients with PGVs are missed by current testing guidelines. Comprehensive germline genetic testing should be offered to all patients with PCa. PATIENT SUMMARY One in 13 patients with prostate cancer carries an inherited variant that may be actionable for the patient's current care or prevention of future cancer, and could benefit from expanded testing criteria.
Collapse
Affiliation(s)
- Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA.
| | - Mukaram Gazi
- University Urology Associates of New Jersey, Hamilton, NJ, USA
| | | | - Sean Heron
- Advanced Urology Institute, St. Petersburg, FL, USA
| | - Rishi Modh
- Advanced Urology Institute, St. Petersburg, FL, USA
| | | | | | - Aaron Berger
- Associated Urological Specialists, Chicago Ridge, IL, USA
| | | | | | | | | | | | | | | | - Paul Dato
- Genesis Healthcare Partners, San Diego, CA, USA
| | | | | | - David R Wise
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Ryan D Hernandez
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Robert L Nussbaum
- Invitae Corporation, San Francisco, CA, USA; Volunteer Faculty, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
14
|
Valsecchi AA, Dionisio R, Panepinto O, Paparo J, Palicelli A, Vignani F, Di Maio M. Frequency of Germline and Somatic BRCA1 and BRCA2 Mutations in Prostate Cancer: An Updated Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:cancers15092435. [PMID: 37173901 PMCID: PMC10177599 DOI: 10.3390/cancers15092435] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
In prostate cancer (PC), the presence of BRCA somatic and/or germline mutation provides prognostic and predictive information. Meta-analysis aims to estimate the frequency of BRCA mutations in patients with PC (PCp). In November 2022, we reviewed literature searching for all articles testing the proportion of BRCA mutations in PCp, without explicit enrichment for familiar risk. The frequency of germline and somatic BRCA1 and/or BRCA2 mutations was described in three stage disease populations (any/metastatic/metastatic castration-resistant PC, mCRPC). Out of 2253 identified articles, 40 were eligible. Here, 0.73% and 1.20% of any stage PCp, 0.94% and 1.10% of metastatic PCp, and 1.21% and 1.10% of mCRPC patients carried germline and somatic BRCA1 mutation, respectively; 3.25% and 6.29% of any stage PCp, 4.51% and 10.26% of metastatic PCp, and 3.90% and 10.52% of mCRPC patients carried germline and somatic BRCA2 mutation, respectively; and 4.47% and 7.18% of any stage PCp, 5.84% and 10.94% of metastatic PCp, and 5.26% and 11.26% of mCRPC patients carried germline and somatic BRCA1/2 mutation, respectively. Somatic mutations are more common than germline and BRCA2 are more common than BRCA1 mutations; the frequency of mutations is higher in the metastatic setting. Despite that BRCA testing in PC is now standard in clinical practice, several open questions remain.
Collapse
Affiliation(s)
- Anna Amela Valsecchi
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Rossana Dionisio
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Olimpia Panepinto
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Jessica Paparo
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Andrea Palicelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Francesca Vignani
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, 10128 Turin, Italy
| |
Collapse
|
15
|
Mehra N, Kloots I, Vlaming M, Aluwini S, Dewulf E, Oprea-Lager DE, van der Poel H, Stoevelaar H, Yakar D, Bangma CH, Bekers E, van den Bergh R, Bergman AM, van den Berkmortel F, Boudewijns S, Dinjens WN, Fütterer J, van der Hulle T, Jenster G, Kroeze LI, van Kruchten M, van Leenders G, van Leeuwen PJ, de Leng WW, van Moorselaar RJA, Noordzij W, Oldenburg RA, van Oort IM, Oving I, Schalken JA, Schoots IG, Schuuring E, Smeenk RJ, Vanneste BG, Vegt E, Vis AN, de Vries K, Willemse PPM, Wondergem M, Ausems M. Genetic Aspects and Molecular Testing in Prostate Cancer: A Report from a Dutch Multidisciplinary Consensus Meeting. EUR UROL SUPPL 2023; 49:23-31. [PMID: 36874601 PMCID: PMC9975012 DOI: 10.1016/j.euros.2022.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/27/2023] Open
Abstract
Background Germline and tumour genetic testing in prostate cancer (PCa) is becoming more broadly accepted, but testing indications and clinical consequences for carriers in each disease stage are not yet well defined. Objective To determine the consensus of a Dutch multidisciplinary expert panel on the indication and application of germline and tumour genetic testing in PCa. Design setting and participants The panel consisted of 39 specialists involved in PCa management. We used a modified Delphi method consisting of two voting rounds and a virtual consensus meeting. Outcome measurements and statistical analysis Consensus was reached if ≥75% of the panellists chose the same option. Appropriateness was assessed by the RAND/UCLA appropriateness method. Results and limitations Of the multiple-choice questions, 44% reached consensus. For men without PCa having a relevant family history (familial PCa/BRCA-related hereditary cancer), follow-up by prostate-specific antigen was considered appropriate. For patients with low-risk localised PCa and a family history of PCa, active surveillance was considered appropriate, except in case of the patient being a BRCA2 germline pathogenic variant carrier. Germline and tumour genetic testing should not be done for nonmetastatic hormone-sensitive PCa in the absence of a relevant family history of cancer. Tumour genetic testing was deemed most appropriate for the identification of actionable variants, with uncertainty for germline testing. For tumour genetic testing in metastatic castration-resistant PCa, consensus was not reached for the timing and panel composition. The principal limitations are as follows: (1) a number of topics discussed lack scientific evidence, and therefore the recommendations are partly opinion based, and (2) there was a small number of experts per discipline. Conclusions The outcomes of this Dutch consensus meeting may provide further guidance on genetic counselling and molecular testing related to PCa. Patient summary A group of Dutch specialists discussed the use of germline and tumour genetic testing in prostate cancer (PCa) patients, indication of these tests (which patients and when), and impact of these tests on the management and treatment of PCa.
Collapse
Affiliation(s)
- Niven Mehra
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Iris Kloots
- Department of Medical Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Michiel Vlaming
- Division Laboratories, Pharmacy and biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Shafak Aluwini
- Department of Radiation Oncology, UMCG, Groningen, The Netherlands
| | - Els Dewulf
- Centre for Decision Analysis & Support, Ismar Healthcare NV, Lier, Belgium
| | - Daniela E. Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Henk van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Herman Stoevelaar
- Centre for Decision Analysis & Support, Ismar Healthcare NV, Lier, Belgium
| | - Derya Yakar
- Department of Radiology, UMCG, Groningen, The Netherlands
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Chris H. Bangma
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | - Elise Bekers
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Andries M. Bergman
- Department of Medical Oncology and Oncogenomics, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | - Steve Boudewijns
- Department of Medical Oncology, Bravis Hospital, Roosendaal, The Netherlands
| | | | - Jurgen Fütterer
- Department of Medical Imaging, Radboud UMC, Nijmegen, The Netherlands
| | - Tom van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Guido Jenster
- Department of Urology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Michel van Kruchten
- Department of Medical Oncology, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Pim J. van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | | | - Walter Noordzij
- Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Irma Oving
- Department of Internal Medicine, Ziekenhuis Groep Twente, Almelo, The Netherlands
| | | | - Ivo G. Schoots
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ed Schuuring
- Department of Pathology, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert J. Smeenk
- Department of Radiation Oncology, Radboud UMC, Nijmegen, The Netherlands
| | - Ben G.L. Vanneste
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht UMC, Maastricht, The Netherlands
- Department of Human Structure and Repair, Ghent University Hospital, Ghent, Belgium
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Erik Vegt
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Kim de Vries
- Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Maurits Wondergem
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Margreet Ausems
- Division Laboratories, Pharmacy and biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
16
|
Gerido LH, Griggs JJ, Resnicow K, Kidwell KM, Delacroix E, Austin S, Hanson EN, Bacon E, Koeppe E, Goodall S, Demerath M, Rizzo EA, Weiner S, Hawley ST, Uhlmann WR, Roberts JS, Stoffel EM. The Michigan Genetic Hereditary Testing (MiGHT) study's innovative approaches to promote uptake of clinical genetic testing among cancer patients: a study protocol for a 3-arm randomized controlled trial. Trials 2023; 24:105. [PMID: 36765432 PMCID: PMC9911941 DOI: 10.1186/s13063-023-07125-2] [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: 12/05/2022] [Accepted: 01/28/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Although most cancers are sporadic, germline genetic variants are implicated in 5-10% of cancer cases. Clinical genetic testing identifies pathogenic germline genetic variants for hereditary cancers. The Michigan Genetic Hereditary Testing (MiGHT) study is a three-arm randomized clinical trial that aims to test the efficacy of two patient-level behavioral interventions on uptake of cancer genetic testing. METHODS The two interventions being tested are (1) a virtual genetics navigator and (2) motivational interviewing by genetic health coaches. Eligible participants are adults with a diagnosis of breast, prostate, endometrial, ovarian, colorectal, or pancreatic cancer who meet the National Comprehensive Cancer Network (NCCN) criteria for genetic testing. Participants are recruited through community oncology practices affiliated with the Michigan Oncology Quality Consortium (MOQC) and have used the Family Health History Tool (FHHT) to determine testing eligibility. The recruitment goal is 759 participants, who will be randomized to usual care or to either the virtual genetics navigator or the motivational interviewing intervention arms. The primary outcome will be the proportion of individuals who complete germline genetic testing within 6 months. DISCUSSION This study addresses patient-level factors which are associated with the uptake of genetic testing. The study will test two different intervention approaches, both of which can help address the shortage of genetic counselors and improve access to care. TRIAL REGISTRATION This study has been approved by the Institutional Review Board of the University of Michigan Medical School (HUM00192898) and registered in ClinicalTrials.gov (NCT05162846).
Collapse
Affiliation(s)
| | - Jennifer J Griggs
- University of Michigan School of Public Health, Ann Arbor, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
- University of Michigan School of Medicine, Ann Arbor, USA
| | - Ken Resnicow
- University of Michigan School of Public Health, Ann Arbor, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
| | - Kelley M Kidwell
- University of Michigan School of Public Health, Ann Arbor, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
| | - Emerson Delacroix
- University of Michigan School of Public Health, Ann Arbor, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | - Sarah Austin
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
| | - Erika N Hanson
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | - Elizabeth Bacon
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | - Erika Koeppe
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | | | | | | | - Shayna Weiner
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
| | - Sarah T Hawley
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
| | - Wendy R Uhlmann
- University of Michigan School of Medicine, Ann Arbor, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | - J Scott Roberts
- University of Michigan School of Public Health, Ann Arbor, USA
| | - Elena M Stoffel
- University of Michigan Rogel Cancer Center, Ann Arbor, USA
- University of Michigan School of Medicine, Ann Arbor, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| |
Collapse
|
17
|
Finn CM, McCormick S, Peterson D, Niendorf KB, Rodgers LH. Motivation and family communication in hereditary prostate cancer genetic testing: Survey of patients from a US tertiary medical center. J Genet Couns 2023; 32:79-89. [PMID: 35941805 DOI: 10.1002/jgc4.1624] [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: 11/03/2021] [Revised: 07/08/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022]
Abstract
Identification of a hereditary prostate cancer in an affected individual can guide treatment and may also impact cancer screening and surveillance for patients and their relatives. This study aimed to determine the factors that are associated with the decision-making process of individuals with prostate cancer regarding whether to pursue genetic testing as well as how, why, and with whom genetic test results are shared. We surveyed 113 patients diagnosed with prostate cancer who received cancer genetic counseling through a United States tertiary medical center, inquiring about genetic testing motivations and family communication about results. Among those who pursued genetic testing, (1) learning about my family's possible cancer risk (98%), (2) learning information that may guide cancer treatment (93%), and (3) learning if I am at risk for future cancers (92%) were most frequently identified as slightly or very important factors in their decision. Participants shared their genetic test results in a higher proportion to male first-degree relatives than female first-degree relatives; however, no significant difference was found (p = 0.103). Our study may suggest sex differences related to family communication about genetic testing results. Such findings indicate a critical need for genetic counselors to clearly communicate the impact of genetic test results on both male and female relatives. Further research on motivation and family communication about genetic test results in diverse cohorts is needed.
Collapse
Affiliation(s)
- Caitlin M Finn
- MGH Institute of Health Professions Genetic Counseling Program, Boston, Massachusetts, USA.,Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA.,Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Shelley McCormick
- Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| | - Danielle Peterson
- MGH Institute of Health Professions Genetic Counseling Program, Boston, Massachusetts, USA
| | - Kristin B Niendorf
- MGH Institute of Health Professions Genetic Counseling Program, Boston, Massachusetts, USA
| | - Linda H Rodgers
- Massachusetts General Hospital, Cancer Center, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Nugroho PP, Ghozali SAS, Buchanan DD, Pisano MI, Reece JC. Risk of cancer in individuals with Lynch-like syndrome and their families: a systematic review. J Cancer Res Clin Oncol 2023; 149:25-46. [PMID: 36251064 PMCID: PMC9889410 DOI: 10.1007/s00432-022-04397-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/05/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Lynch-like syndrome (LLS) tumors have similar clinicopathological features to Lynch syndrome (LS) tumors but have no identifiable pathogenic germline mismatch repair gene variant. However, cancer risks in LLS patients and first-degree relatives (FDRs) are not well defined. METHODS To clarify LLS-associated cancer risks, a systematic review of all studies examining all cancer risks in LLS was performed. Searching of Medline, Embase, Pubmed, Cochrane and CINAHL databases and reference/citation checking identified relevant studies published between January 1, 1980 and February 11, 2021. Joanna Briggs Institute Appraisal Tools assessed the risk of bias. RESULTS Six studies (five cohort/one cross-sectional) were eligible for study inclusion. One study found no difference in colorectal cancer (CRC) incidence between LLS and LS patients or CRC risks at aged 70 years. Three studies found CRC incidence in LLS FDRs was higher than the general population but lower than LS FDRs. Two studies showed no difference in CRC diagnosis age between LLS patients and LS patients. Endometrial cancer risks in LLS patients were higher than the general population but lower than LS patients. CONCLUSION Evidence of elevated CRC risks in LLS patients and FDRs supports increased colonoscopy surveillance strategies for LLS patients and FDRs in line with current recommendations for LS. Due to heterogeneity amongst LLS populations, extended intervals between screening may be advised for low-risk families. Studies to resolve the molecular characterization and definition of LLS are needed to clarify cancer risks associated with LLS which in turn may individualize surveillance strategies for LLS patients and families.
Collapse
Affiliation(s)
- Pandu P Nugroho
- Faculty of Medicine, Universitas Indonesia, Depok, West Java, Indonesia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Siti Alyaa S Ghozali
- Faculty of Medicine, Universitas Indonesia, Depok, West Java, Indonesia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
- University of Melbourne Centre for Cancer Research, Parkville, VIC, Australia
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Mia I Pisano
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Jeanette C Reece
- Neuroepidemiology Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Level 3 207 Bouverie Street, Parkville, VIC, 3010, Australia.
| |
Collapse
|
19
|
What is the most effective way to ensure that patients successfully undergo germline testing for prostate cancer? Urol Oncol 2023; 41:49.e1-49.e6. [PMID: 36283931 DOI: 10.1016/j.urolonc.2022.09.002] [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: 04/22/2022] [Revised: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION We sought to identify the most effective way to refer patients with prostate cancer to germline testing. METHODS After IRB approval, we queried the electronic medical records (EMR) to identify patients (ages 18-89) with prostate cancer who were referred for or offered germline testing for prostate cancer from May 1, 2019 to February 24, 2021 through either telephone referral, EMR referral or in-office testing. The 3 cohorts were compared on receipt of testing and time to testing. Multivariate logistic regression and Cox regression evaluated the influence of referral cohort and reason for testing on receipt of testing and time to testing, respectively. RESULTS A total of 184 patients met study inclusion criteria; 47 were referred for germline testing via telephone, 70 were referred through the EMR and 67 were offered testing in the office. No significant demographic or clinical differences were observed. Telephone referral yielded the lowest response (17%; P < 0.001) with the longest time interval between referral and testing (103 days; P < 0.001); in-office testing yielded the highest response (66%). More patients were referred because of both family history and high risk characteristics in the EMR and in-office testing cohorts (21.4% and 25.4% respectively). Referral method was significantly (P < 0.001) associated with receipt of test, while reason for testing was not. Referral method was also independently related to time to testing (P < 0.001) while reason for referral was not. CONCLUSIONS Urologists should offer germline testing in the office for the most effective and expedient results.
Collapse
|
20
|
Kirchner K, Gamulin M, Kulis T, Sievers B, Kastelan Z, Lessel D. Comprehensive Clinical and Genetic Analysis of CHEK2 in Croatian Men with Prostate Cancer. Genes (Basel) 2022; 13:1955. [PMID: 36360192 PMCID: PMC9689475 DOI: 10.3390/genes13111955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2023] Open
Abstract
Germline pathogenic and likely pathogenic (P/LP) variants in CHEK2 have been associated with increased prostate cancer (PrCa) risk. Our objective was to analyze their occurrence in Croatian PrCa men and to evaluate the clinical characteristics of P/LP variant carriers. Therefore, we analyzed CHEK2 in 150 PrCa patients unselected for age of onset, family history of PrCa or clinical outcome, and the frequency of identified variants was compared to findings in 442 cancer-free men, of Croatian ancestry. We identified four PrCa cases harboring a P/LP variant in CHEK2 (4/150, 2.67%), which reached a statistical significance (p = 0.004) as compared to the control group. Patients with P/LP variants in CHEK2 developed PrCa almost 9 years earlier than individuals with CHEK2 wild-type alleles (8.9 years; p = 0.0198) and had an increased risk for lymph node involvement (p = 0.0047). No association was found between CHEK2 status and further clinical characteristics, including the Gleason score, occurrence of aggressive PrCa, the tumor or metastasis stage. However, carriers of the most common P/LP CHEK2 variant, the c.1100delC, p.Thr367Metfs15*, had a significantly higher Gleason score (p = 0.034), risk for lymph node involvement (p = 0.0001), and risk for developing aggressive PrCa (p = 0.027). Thus, in a Croatian population, CHEK2 P/LP variant carriers were associated with increased risk for early onset prostate cancer, and carriers of the c.1100delC, p.Thr367Metfs15* had increased risk for aggressive PrCa.
Collapse
Affiliation(s)
- Kira Kirchner
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marija Gamulin
- Department of Oncology, University Hospital Center Zagreb, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Tomislav Kulis
- Department of Urology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Bianca Sievers
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Zeljko Kastelan
- Department of Urology, University Hospital Centre Zagreb, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
21
|
Zhang D, Xu X, Wei Y, Chen X, Li G, Lu Z, Zhang X, Ren X, Wang S, Qin C. Prognostic Role of DNA Damage Response Genes Mutations and their Association With the Sensitivity of Olaparib in Prostate Cancer Patients. Cancer Control 2022; 29:10732748221129451. [PMID: 36283420 PMCID: PMC9608002 DOI: 10.1177/10732748221129451] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective Evidence shows that gene mutation is a significant proportion of genetic factors associated with prostate cancer. The DNA damage response (DDR) is a signal cascade network that aims to maintain genomic integrity in cells. This comprehensive study was performed to determine the link between different DNA damage response gene mutations and prostate cancer. Materials and methods A systematic literature search was performed using PubMed, Web of Science, and Embase. Papers published up to February 1, 2022 were retrieved. The DDR gene mutations associated with prostate cancer were identified by referring to relevant research and review articles. Data of prostate cancer patients from multiple PCa cohorts were obtained from cBioPortal. The OR or HR and 95% CIs were calculated using both fixed-effects models (FEMs) and random-effects models (REMs). Results Seventy-four studies were included in this research, and the frequency of 13 DDR genes was examined. Through the analysis of 33 articles that focused on the risk estimates of DDR genes between normal people and PCa patients, DDR genes were found to be more common in prostate cancer patients (OR = 3.6293 95% CI [2.4992; 5.2705]). Also, patients in the mutated group had a worse OS and DFS outcome than those in the unmutated group (P < .05). Of the 13 DDR genes, the frequency of 9 DDR genes in prostate cancer was less than 1%, and despite differences in race, BRCA2 was the potential gene with the highest frequency (REM Frequency = .0400, 95% CI .0324 - .0541). The findings suggest that mutations in genes such as ATR, BLM, and MLH1 in PCa patients may increase the sensitivity of Olaparib, a PARP inhibitor. Conclusion These results demonstrate that mutation in any DDR pathway results in a poor prognosis for PCa patients. Furthermore, mutations in ATR, BLM, and MLH1 or the expression of POLR2L, PMS1, FANCE, and other genes significantly influence Olaparib sensitivity, which may be underlying therapeutic targets in the future.
Collapse
Affiliation(s)
- Dong Zhang
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinchi Xu
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuang Wei
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinglin Chen
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Guangyao Li
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhongwen Lu
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xu Zhang
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaohan Ren
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shangqian Wang
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China,Chao Qin, The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. ; Shangqian Wang, The State Key Lab of Reproductive; Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Chao Qin
- The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China,Chao Qin, The State Key Lab of Reproductive, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China. ; Shangqian Wang, The State Key Lab of Reproductive; Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
22
|
Weise N, Shaya J, Javier-Desloges J, Cheng HH, Madlensky L, McKay RR. Disparities in germline testing among racial minorities with prostate cancer. Prostate Cancer Prostatic Dis 2022; 25:403-410. [PMID: 34775478 PMCID: PMC8590439 DOI: 10.1038/s41391-021-00469-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022]
Abstract
Germline testing is becoming increasingly relevant in prostate cancer (PCa) screening, prognosis, and management. A subset of patients with PCa harbor pathogenic/likely pathogenic variants (P/LPVs) in genes mediating DNA-repair processes, and these P/LPVs have implications for cancer screening, treatment, and cascade testing. As a result, it is recommended that all men with high-risk localized and metastatic PCa undergo routine germline testing. As more PCa patients undergo germline testing, it is important that clinicians and genetics experts recognize current disparities in germline testing rates among racial/ethnic minorities in the United States. The reasons for these disparities are multiple and require similarly manifold consideration to close the germline testing gap and reduce inequities in PCa screening, management, and treatment.
Collapse
Affiliation(s)
- Nicole Weise
- University of California San Diego, San Diego, CA, USA
| | - Justin Shaya
- University of California San Diego, San Diego, CA, USA
| | | | - Heather H Cheng
- University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Rana R McKay
- University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
23
|
Esplin ED, Nielsen SM, Bristow SL, Garber JE, Hampel H, Rana HQ, Samadder NJ, Shore ND, Nussbaum RL. Universal Germline Genetic Testing for Hereditary Cancer Syndromes in Patients With Solid Tumor Cancer. JCO Precis Oncol 2022; 6:e2100516. [PMID: 36108258 PMCID: PMC9489188 DOI: 10.1200/po.21.00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
| | | | | | | | - Heather Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology & Therapeutic Research, City of Hope National Cancer Center, Duarte, CA
| | | | - N Jewel Samadder
- Center for Individualized Medicine, Mayo Clinic, Phoenix, AZ.,Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, AZ.,Department of Clinical Genomics, Mayo Clinic, Phoenix, AZ
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | |
Collapse
|
24
|
Giri VN, Hartman R, Pritzlaff M, Horton C, Keith SW. Germline Variant Spectrum Among African American Men Undergoing Prostate Cancer Germline Testing: Need for Equity in Genetic Testing. JCO Precis Oncol 2022; 6:e2200234. [PMID: 35666082 PMCID: PMC9200399 DOI: 10.1200/po.22.00234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Guidelines for prostate cancer (PCA) germline testing (GT) have expanded, with impact on clinical management and hereditary cancer assessment. African American (AA) men have lower engagement in GT, with concern for widening disparities in genetically informed care. We evaluated the germline spectrum in a cohort of men with PCA enriched for AA men who underwent GT to inform tailored genetic evaluation strategies. METHODS Participants included AA and White men with PCA tested with a 14-gene PCA panel: ATM, BRCA1, BRCA2, CHEK2, EPCAM, HOXB13, MLH1, MSH2, MSH6, NBN, PALB2, PMS2, RAD51D, and TP53. Germline analysis was performed per standard clinical testing and variant classification protocols. Data were compared with Fisher's exact, chi-squared, or two sample t-tests, as appropriate. Multivariable analysis was conducted using Akaike's Information Criterion. The significance level was set a priori at .05. RESULTS The data set included 427 men all tested using the 14-gene PCA panel: AA (n = 237, 56%) and White (n = 190, 44%). Overall, the pathogenic/likely pathogenic (P/LP) variant rate was 8.2%, with AA men having lower rates of P/LP variants then White men (5.91% v 11.05%, respectively; P = .05). Borderline associations with P/LP variant status were observed by race (AA v White; odds ratio = 0.51; P = .07) and age (> 50 v ≤ 50 years; odds ratio = 0.48; P = .06). The P/LP spectrum was narrower in AA men (BRCA2, PALB2, ATM, and BRCA1) than White men (BRCA2, ATM, HOXB13, CHEK2, TP53, and NBN). A significant difference was noted in rates of variants of uncertain significance (VUSs) between AA men and White men overall (25.32% v 16.32%; P = .02) and for carrying multiple VUSs (5.1% v 0.53%, P = .008). CONCLUSION Germline evaluation in a cohort enriched for AA men highlights the narrower spectrum of germline contribution to PCA with significantly higher rates of multiple VUSs in DNA repair genes. These results underscore the imperative to engage AA men in GT, the need for larger panel testing in AA men, and the necessity to incorporate novel genomic technologies to clarify VUS to discern the germline contribution to PCA. Furthermore, tailored genetic counseling for AA men is important to ensure understanding of VUS and promote equitable genetics care delivery.
Collapse
Affiliation(s)
- Veda N. Giri
- Departments of Medical Oncology, Cancer Biology, and Urology, Cancer Risk Assessment and Clinical Cancer Genetics, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
- Division of Population Science, Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Rebecca Hartman
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA
| | | | | | - Scott W. Keith
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
25
|
Shore ND, Lenz L, Cogan ES, Iliev D, Spencer L, Flake DD, Meek S, Davis T, Copeland K, Finch R, Schiff W, Korman H, Rao M, Belkoff L, Jalkut M, Mariados N, D'Anna R, Mehlhaff B, Slavin TP, Cohen TD. Hereditary cancer risk assessment and genetic testing in the community urology practice setting. Prostate 2022; 82:850-857. [PMID: 35239202 DOI: 10.1002/pros.24327] [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: 11/15/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES To evaluate the feasibility of integrating a hereditary cancer risk assessment (HCRA) process in the community urology practice setting for patients with prostate cancer (PCa). METHODS In this prospective intervention, an HCRA process was implemented across six different community urology clinics between May 2019 and April 2020. The intervention included a process integration during which the workflow at each site was refined, a post-integration period during which HCRA was conducted in all patients with PCa, and a follow-up period during which healthcare providers and patients reported their satisfaction with the HCRA and genetic testing process. RESULTS Among patients who completed a family history assessment during the post-integration period, 23.6% met guideline criteria for genetic testing. Of all patients seen at the clinic during the post-integration period, 8.7% completed genetic testing; this was a twofold increase over the period immediately preceding process integration (4.2%), and a sevenfold increase over the same period 1 year prior (1.2%). The majority of providers reported that the HCRA was as important as other regularly performed assessments (61.0%) and planned to continue using the process in their practice (68.3%). Most patients believed that the genetic test results were important for their future cancer care (84.7%) and had already shared their test results with at least one family member (63.2%). CONCLUSIONS This study demonstrated that implementing an HCRA process in the community urology practice setting was feasible, generally favored by providers and patients, and resulted in an increase in the number of patients with PCa who completed genetic testing.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center/GenesisCare, Myrtle Beach, South Carolina, USA
| | - Lauren Lenz
- Myriad Genetics, Inc., Salt Lake City, Utah, USA
| | | | - Diana Iliev
- Myriad Genetics, Inc., Salt Lake City, Utah, USA
| | | | - Darl D Flake
- Myriad Genetics, Inc., Salt Lake City, Utah, USA
| | | | | | | | - Robert Finch
- Myriad Genetics, Inc., Salt Lake City, Utah, USA
| | - William Schiff
- Urology Associates of Central California, Fresno, California, USA
| | | | - Manoj Rao
- Urologic Specialists of Northwest Indiana, Merrillville, Indiana, USA
| | | | - Mark Jalkut
- Associated Urologists of North Carolina, Raleigh, North Carolina, USA
| | - Neil Mariados
- Associated Medical Professionals, Syracuse, New York, USA
| | | | | | | | - Todd D Cohen
- Myriad Genetics, Inc., Salt Lake City, Utah, USA
| |
Collapse
|
26
|
Abstract
Genetic testing for prostate cancer is rapidly growing and is increasingly being driven by precision medicine. Rates of germline pathogenic variants have been reported in up to 15% of men with prostate cancer, particularly in metastatic disease, and results of genetic testing could uncover options for precision therapy along with a spectrum of hereditary cancer-predisposition syndromes with unique clinical features that have complex management options. Thus, the pre-test discussion, whether delivered by genetic counsellors or by health-care professionals in hybrid models, involves information on hereditary cancer risk, extent of gene testing, purpose of testing, medical history and family history, potential types of results, additional cancer risks that might be uncovered, genetically based management and effect on families. Understanding precision medicine, personalized cancer risk management and syndrome-related cancer risk management is important in order to develop collaborative strategies with genetic counselling for optimal care of patients and their families. In this Review, Russo and Giri describe and discuss germline testing criteria, genetic testing strategies, genetically informed screening, precision management, delivery of genetic counselling or alternative genetic services and special considerations for men with prostate cancer. Germline (hereditary) genetic testing is rising in importance for treatment, screening and risk assessment of prostate cancer. Multiple hereditary cancer syndromes might be associated with prostate cancer, might confer risk of other cancerous and non-cancerous conditions, and can have hereditary cancer implications for family members. The rates of these syndromes can vary based upon the attributed genetic mutations. Multiple aspects of germline testing should be discussed in the pre-test setting for men to make an informed decision, including the purpose of genetic testing, the benefits and risks of testing, hereditary cancer risk, identification of additional cancer risks, familial implications and the state of genetic discrimination protections. Genetic evaluation can be conducted by genetic counsellors or a hybrid model can be employed, in which health-care providers deliver pre-test informed consent for testing, order testing and then determine referral to genetic counselling for appropriate patients. Precision medicine is increasingly driving decisions for germline testing. Poly(ADP-ribose) polymerase (PARP) inhibitors, immune checkpoint inhibitors and various other agents now in clinical trials have clinical activity in patients with certain hereditary cancer gene mutations, such as in DNA repair genes. Patients’ experiences with germline testing can be variable; taking the patient’s current experience into account, considering referral to genetic counselling when needed and offering germline testing for eligible men at repeated intervals if initially declined are important.
Collapse
Affiliation(s)
- Jessica Russo
- Cancer Risk Assessment and Clinical Cancer Genetics, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Veda N Giri
- Cancer Risk Assessment and Clinical Cancer Genetics, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA. .,Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Pensabene M, Von Arx C, De Laurentiis M. Male Breast Cancer: From Molecular Genetics to Clinical Management. Cancers (Basel) 2022; 14:2006. [PMID: 35454911 PMCID: PMC9030724 DOI: 10.3390/cancers14082006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/18/2022] Open
Abstract
MBC is a rare disease accounting for almost 1% of all cancers in men and less than 1% of breast cancer. Emerging data on the genetic drivers of predisposition for MBC are available and different risk factors have been associated with its pathogenesis. Genetic alterations, such as pathogenetic variants in BRCA1/2 and other moderate-/low-penetrance genes, along with non-genetic risk factors, have been recognized as pathogenic factors for MBC. Preventive and therapeutic implications could be related to the detection of alterations in predisposing genes, especially BRCA1/2, and to the identification of oncogenic drivers different from FBC. However, approved treatments for MBC remain the same as FBC. Cancer genetic counseling has to be considered in the diagnostic work-up of MBC with or without positive oncological family history. Here, we review the literature, reporting recent data about this malignancy with a specific focus on epidemiology, and genetic and non-genetic risk factors. We introduce the perspective of cancer genetic counseling for MBC patients and their healthy at-risk family members, with a focus on different hereditary cancer syndromes.
Collapse
Affiliation(s)
- Matilde Pensabene
- National Cancer Institute, IRCCS Fondazione G. Pascale, 80131 Naples, Italy; (C.V.A.); (M.D.L.)
| | | | | |
Collapse
|
28
|
The promising role of new molecular biomarkers in prostate cancer: from coding and non-coding genes to artificial intelligence approaches. Prostate Cancer Prostatic Dis 2022; 25:431-443. [PMID: 35422101 PMCID: PMC9385485 DOI: 10.1038/s41391-022-00537-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/24/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
Background Risk stratification or progression in prostate cancer is performed with the support of clinical-pathological data such as the sum of the Gleason score and serum levels PSA. For several decades, methods aimed at the early detection of prostate cancer have included the determination of PSA serum levels. The aim of this systematic review is to provide an overview about recent advances in the discovery of new molecular biomarkers through transcriptomics, genomics and artificial intelligence that are expected to improve clinical management of the prostate cancer patient. Methods An exhaustive search was conducted by Pubmed, Google Scholar and Connected Papers using keywords relating to the genetics, genomics and artificial intelligence in prostate cancer, it includes “biomarkers”, “non-coding RNAs”, “lncRNAs”, “microRNAs”, “repetitive sequence”, “prognosis”, “prediction”, “whole-genome sequencing”, “RNA-Seq”, “transcriptome”, “machine learning”, and “deep learning”. Results New advances, including the search for changes in novel biomarkers such as mRNAs, microRNAs, lncRNAs, and repetitive sequences, are expected to contribute to an earlier and accurate diagnosis for each patient in the context of precision medicine, thus improving the prognosis and quality of life of patients. We analyze several aspects that are relevant for prostate cancer including its new molecular markers associated with diagnosis, prognosis, and prediction to therapy and how bioinformatic approaches such as machine learning and deep learning can contribute to clinic. Furthermore, we also include current techniques that will allow an earlier diagnosis, such as Spatial Transcriptomics, Exome Sequencing, and Whole-Genome Sequencing. Conclusion Transcriptomic and genomic analysis have contributed to generate knowledge in the field of prostate carcinogenesis, new information about coding and non-coding genes as biomarkers has emerged. Synergies created by the implementation of artificial intelligence to analyze and understand sequencing data have allowed the development of clinical strategies that facilitate decision-making and improve personalized management in prostate cancer.
Collapse
|
29
|
Hassanin E, May P, Aldisi R, Spier I, Forstner AJ, Nöthen MM, Aretz S, Krawitz P, Bobbili DR, Maj C. Breast and prostate cancer risk: The interplay of polygenic risk, rare pathogenic germline variants, and family history. Genet Med 2022; 24:576-585. [PMID: 34906469 DOI: 10.1016/j.gim.2021.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/12/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022] Open
Abstract
PURPOSE We aimed to investigate to what extent polygenic risk scores (PRS), rare pathogenic germline variants (PVs), and family history jointly influence breast cancer and prostate cancer risk. METHODS A total of 200,643 individuals from the UK Biobank were categorized as follows: (1) heterozygotes or nonheterozygotes for PVs in moderate to high-risk cancer genes, (2) PRS strata, and (3) with or without a family history of cancer. Multivariable logistic regression and Cox proportional hazards models were used to compute the odds ratio across groups and the cumulative incidence through life. RESULTS Cumulative incidence by age 70 years among the nonheterozygotes across PRS strata ranged from 9% to 32% and from 9% to 35% for breast cancer and prostate cancer, respectively. Among the PV heterozygotes it ranged from 20% to 48% in moderate-risk genes and from 51% to 74% in high-risk genes for breast cancer, and it ranged from 30% to 59% in prostate cancer risk genes. Family history was always associated with an increased cancer odds ratio. CONCLUSION PRS alone provides a meaningful risk gradient leading to a cancer risk stratification comparable to PVs in moderate risk genes, whereas acts as a risk modifier when considering high-risk genes. Including family history along with PV and PRS further improves cancer risk stratification.
Collapse
Affiliation(s)
- Emadeldin Hassanin
- Institute for Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rana Aldisi
- Institute for Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Isabel Spier
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Centre for Human Genetics, Philipps-University Marburg, Marburg, Germany; Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Stefan Aretz
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany
| | - Dheeraj Reddy Bobbili
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Carlo Maj
- Institute for Genomic Statistics and Bioinformatics, University of Bonn, Bonn, Germany.
| |
Collapse
|
30
|
Pan E, Shaya J, Madlensky L, Randall JM, Millard FE, Rose B, Parsons JK, Nielsen SM, Hatchell KE, Esplin ED, Nussbaum RL, Weise N, Murphy J, Martinez ME, McKay RR. Germline alterations among Hispanic men with prostate cancer. Prostate Cancer Prostatic Dis 2022; 25:561-567. [DOI: 10.1038/s41391-022-00517-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 12/31/2022]
|
31
|
Heise M, Jarzemski P, Nowak D, Bąk A, Junkiert-Czarnecka A, Pilarska-Deltow M, Borysiak M, Pilarska B, Haus O. Clinical Significance of Gene Mutations and Polymorphic Variants and their Association with Prostate Cancer Risk in Polish Men. Cancer Control 2022; 29:10732748211062342. [PMID: 35638715 PMCID: PMC9160909 DOI: 10.1177/10732748211062342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives: We tested the association of germline variants in BRCA1, BRCA2, CHEK2, CDKN2A, CYP1B1, HOXB13, MLH1, NBS1, NOD2 andPALB2 genes, as well as in 8q24 region, with prostate cancer (PC) risk and estimated their impact on disease clinical course, including overall survival time in Polish men with localized PC qualified for radical treatment.Materials and Methods: DNA of 110 patients with localized prostate cancer treated with radical prostatectomy (RP), from each age group and with different stages of the disease. DNA samples of the control group consisted of 111 men, volunteers, without PC (age-matched to study group). Sanger sequencing, AS-PCR, RFLP-PCR, and multiplex-PCR were used for variants detection.Results: The percentage of men with ≥1 germline variant was higher in PC group (52.7%) than in healthy men (37.8%) (P = .03). The presence of ≥2 variants was associated with shorter survival than the presence of one or no variant in the PC group (P = .14, trend). The HOXB13 G84E was detected in 2.9% of PC men and in no healthy men (P = .19, trend, OR = 7.21). A CHEK2 truncating mutation (1100delC or IVS2+1G>A) was detected in 2/110 (1.8%) PC patients and in no healthy men (P = .29, OR=5.14). The NBS1 I171V was detected in 2/110 (1.8%) PC patients and in no men from the control group (OR=5.14, P = .29, NS).Conclusions: We conclude that the presence of more than 2 germline variants was probably associated with shorter survival of patients with localized prostate cancer qualified for radical treatment. The HOXB13 (G84E), CHEK2 (1100delC or IVS2+1G>A) truncating variants and NBS1 (I171V) are associated with PC and hereditary form of the disease. The HOXB13 (G84E) and NOD2 (3020insC) single variants are associated with shorter and CYP1B1 (48CC, 119GG) single genotypes with longer overall survival.
Collapse
Affiliation(s)
- Marta Heise
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Piotr Jarzemski
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Dagmara Nowak
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Aneta Bąk
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Anna Junkiert-Czarnecka
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Maria Pilarska-Deltow
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| | - Maciej Borysiak
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Beata Pilarska
- Faculty of Health Sciences, Department of Urology, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Jan Biziel University Hospital in Bydgoszcz, Poland
| | - Olga Haus
- Faculty of Medicine, Department of Clinical Genetics, Collegium Medicum in Bydgoszcz, 49604Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
32
|
Constantin T, Savu DA, Bucur Ș, Predoiu G, Constantin MM, Jinga V. The Role and Significance of Bioumoral Markers in Prostate Cancer. Cancers (Basel) 2021; 13:5932. [PMID: 34885045 PMCID: PMC8656561 DOI: 10.3390/cancers13235932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/14/2021] [Accepted: 11/22/2021] [Indexed: 11/17/2022] Open
Abstract
The prostate is one of the most clinically accessible internal organs of the genitourinary tract in men. For decades, the only method of screening for prostate cancer (PCa) has been digital rectal examination of 1990s significantly increased the incidence and prevalence of PCa and consequently the morbidity and mortality associated with this disease. In addition, the different types of oncology treatment methods have been linked to specific complications and side effects, which would affect the patient's quality of life. In the first two decades of the 21st century, over-detection and over-treatment of PCa patients has generated enormous costs for health systems, especially in Europe and the United States. The Prostate Specific Antigen (PSA) is still the most common and accessible screening blood test for PCa, but with low sensibility and specificity at lower values (<10 ng/mL). Therefore, in order to avoid unnecessary biopsies, several screening tests (blood, urine, or genetic) have been developed. This review analyzes the most used bioumoral markers for PCa screening and also those that could predict the evolution of metastases of patients diagnosed with PCa.
Collapse
Affiliation(s)
- Traian Constantin
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.C.); (G.P.); (V.J.)
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050659 Bucharest, Romania
| | - Diana Alexandra Savu
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050659 Bucharest, Romania
| | - Ștefana Bucur
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.C.); (G.P.); (V.J.)
- IInd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Gabriel Predoiu
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.C.); (G.P.); (V.J.)
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050659 Bucharest, Romania
| | - Maria Magdalena Constantin
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.C.); (G.P.); (V.J.)
- IInd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Viorel Jinga
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.C.); (G.P.); (V.J.)
- Department of Urology, “Prof. Dr. Theodor Burghele” Hospital, 050659 Bucharest, Romania
| |
Collapse
|
33
|
Lee DJ, Hausler R, Le AN, Kelly G, Powers J, Ding J, Feld E, Desai H, Morrison C, Doucette A, Gabriel P, Genetics Center R, Judy RL, Weaver J, Kember R, Damrauer SM, Rader DJ, Domchek SM, Narayan V, Schwartz LE, Maxwell KN. Association of Inherited Mutations in DNA Repair Genes with Localized Prostate Cancer. Eur Urol 2021; 81:559-567. [PMID: 34711450 PMCID: PMC9035481 DOI: 10.1016/j.eururo.2021.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022]
Abstract
Background: Identification of germline mutations in DNA repair genes has significant implications for the personalized treatment of individuals with prostate cancer (PrCa). Objective: To determine DNA repair genes associated with localized PrCa in a diverse academic biobank and to determine genetic testing burden. Design, setting, and participants: A cross-sectional study of 2391 localized PrCa patients was carried out. Outcome measurements and statistical analysis: Genetic ancestry and mutation rates (excluding somatic interference) in 17 DNA repair genes were determined in 1588 localized PrCa patients and 3273 cancer-free males. Burden testing within individuals of genetically determined European (EUR) and African (AFR) ancestry was performed between biobank PrCa cases and cancer-free biobank and gnomAD males. Results and limitations: AFR individuals with localized PrCa had lower DNA repair gene mutation rates than EUR individuals (1.4% vs 4.0%, p = 0.02). Mutation rates in localized PrCa patients were similar to those in biobank and gnomAD controls (EUR: 4.0% vs 2.8%, p = 0.15, vs 3.1%, p = 0.04; AFR: 1.4% vs 1.8%, p = 0.8, vs 2.1%, p = 0.5). Gene-based rare variant association testing revealed that only BRCA2 mutations were significantly enriched compared with gnomAD controls of EUR ancestry (1.0% vs 0.28%, p = 0.03). Of the participants, 21% and 11% met high-risk and very-high-risk criteria; of them, 3.7% and 6.2% had any germline genetic mutation and 1.0% and 2.5% had a BRCA2 mutation, respectively. Limitations of this study include an analysis of a relatively small, single-institution cohort. Conclusions: DNA repair gene germline mutation rates are low in an academic biobank cohort of localized PrCa patients, particularly among individuals of AFR genetic ancestry. Mutation rates in genes with published evidence of association with PrCa exceed 2.5% only in high-risk, very-high-risk localized, and node-positive PrCa patients. These findings highlight the importance of risk stratification in localized PrCa patients to identify appropriate patients for germline genetic testing. Patient summary: In the majority of patients who develop localized prostate cancer, germline genetic testing is unlikely to reveal an inherited DNA repair mutation, regardless of race. High-risk features increase the possibility of a germline DNA repair mutation.
Collapse
Affiliation(s)
- Daniel J Lee
- Department of Surgery, Division of Urology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Hausler
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anh N Le
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory Kelly
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacquelyn Powers
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James Ding
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Feld
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heena Desai
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Casey Morrison
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail Doucette
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Gabriel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Renae L Judy
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joellen Weaver
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Kember
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott M Damrauer
- Department of Surgery, Division of Vascular Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Domchek
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vivek Narayan
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren E Schwartz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kara N Maxwell
- Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Ward M, Elder B, Habtemariam M. Current Testing Guidelines: A Retrospective Analysis of a Community-Based Hereditary Cancer Program. J Adv Pract Oncol 2021; 12:693-701. [PMID: 34671499 PMCID: PMC8504926 DOI: 10.6004/jadpro.2021.12.7.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It is estimated that 5% to 10% of all cancers are related to a hereditary cancer syndrome. However, specific cancers, such as pancreatic and ovarian cancers, are related to hereditary cancer syndromes 15% to 20% of the time. Genetic testing guidelines for hereditary cancer syndromes are frequently reviewed and updated by the National Comprehensive Cancer Network (NCCN). The purpose of this retrospective analysis is to identify carriers of pathogenic variants or hereditary cancer syndrome who do not meet NCCN criteria for testing and compare the results with previous studies. The data obtained can be used to provide recommendations to assess current guidelines for testing and evaluate the benefit of comprehensive panel testing vs. standard testing for specific hereditary cancer syndromes. This project is a retrospective review of clinical histories of patients who had multigene panel testing between September 2015 and February 2019 through a cancer outreach and risk assessment (CORA) program. Frequencies analyses were performed to analyze results. A total of 233 individuals were included in the analysis: 171 met BRCA1/2 testing criteria, 66 met Lynch syndrome criteria, and 4 met polyposis criteria. Of the individuals meeting established criteria for testing, 39 were identified with pathogenic variants. However, only 10 of these individuals were identified with a pathogenic variant associated with the criteria for which they met. Genetic testing that is limited to only those patients with genes associated with hereditary cancer syndromes may lead to exclusion of other potentially actionable genes, which may impair a patient's ability to receive additional screening or preventative measures.
Collapse
|
35
|
Jiang L, Bai Z, Zhu S, Zhao T, Yang Y, Li Z, Chen D, Wu Z, Wang Y, Zhou F, Li Y. A novel germline BRCA2 mutation in a Chinese patient with prostate cancer sensitive to platinum chemotherapy: a case report. BMC Urol 2021; 21:114. [PMID: 34425813 PMCID: PMC8381549 DOI: 10.1186/s12894-021-00879-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 08/06/2021] [Indexed: 11/10/2022] Open
Abstract
Background Germline BRCA2 mutation is associated with an aggressive prostate cancer phenotype and indicates higher risk for hereditary cancer. Recently, numerous studies have attempted to identify the genomic landscape of prostate cancer to better understand the genomic drivers of this disease and look for the molecular targets to guide treatment selection. Case presentation We report a 67-year-old patient diagnosed with prostate cancer who experienced rapid disease progression after androgen deprivation therapy and subsequent docetaxel treatment. The patient had a strong family history of malignancy as his mother was diagnosed with breast cancer and his father was died of lung cancer. Next generation sequencing demonstrated a novel pathogenic germline BRCA2 mutation (p.Gly2181Glufs*10) in the patient. His mother with breast cancer and his son were found to have the same BRCA2 mutation. The patient experienced impressive and durable responses to carboplatin treatment. Conclusions This case demonstrated that the carboplatin could have a dramatic antitumor effect on patients with prostate cancer with germline BRCA2 mutations and family history will help to ensure that patients and their families can be provided with proper genetic counseling.
Collapse
Affiliation(s)
- Lijuan Jiang
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zunguang Bai
- Department of Urology, The Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510000, China
| | - Shoulun Zhu
- Department of Urology, The Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510000, China
| | - Tingting Zhao
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, 200120, China
| | - Yining Yang
- GloriousMed Clinical Laboratory (Shanghai) Co., Ltd., Shanghai, 200120, China
| | - Zhiyong Li
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Dong Chen
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhiming Wu
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yanjun Wang
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yonghong Li
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
36
|
Stuttgen Finn K, Pacyna JE, Tsou C, Samadder NJ, Sharp RR. Factors that Influence Intent to Share Genetic Information Related to Cancer Risk with Family Members. JOURNAL OF HEALTH COMMUNICATION 2021; 26:545-552. [PMID: 34473010 PMCID: PMC10869109 DOI: 10.1080/10810730.2021.1968078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We describe factors influencing patient decisions to share positive cancer genetic test results with family members. We focused on patients who were diagnosed with several different cancer types but did not have a family history that was suggestive of an inherited risk. Participants were recruited from Mayo Clinic and had been recently diagnosed with cancer. An 80+ gene panel was performed. Before receiving genetic test results, patients completed a 49-item survey on their intent to share their results with relatives. 1,721 (57.7%) of 2,984 individuals who elected to pursue genetic testing completed the survey. Most patients planned to share cancer-related genetic results with a spouse or partner (97.0%), at least one adult child (92.2%), at least one sibling (86.2%), and with at least one parent (70.3%). Familial support scores and familial communication scores were predictive of intent to share cancer-related genetic test results. Our data highlight differences in family communication capacity and support that are important for clinicians to consider when supporting patients who wish to share cancer-related genetic test results with family members. Our data point to several potential interventional strategies that might increase the likelihood of cancer-related genetic test results being shared with family members at risk.
Collapse
Affiliation(s)
- Kelsey Stuttgen Finn
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, Minnesota, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Joel E Pacyna
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, Minnesota, USA
| | - Cindy Tsou
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Niloy J Samadder
- Center for Individualized Medicine, Mayo Clinic, Phoenix, Arizona, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, Arizona, USA
- Department of Clinical Genomics, Mayo Clinic, Phoenix, Arizona, USA
| | - Richard R Sharp
- Biomedical Ethics Research Program, Mayo Clinic, Rochester, Minnesota, USA
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
37
|
Chandrasekar T, Kelly WK, Gomella LG. Overview of Prostate Cancer Genetic Testing. Urol Clin North Am 2021; 48:279-282. [PMID: 34210484 DOI: 10.1016/j.ucl.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Thenappan Chandrasekar
- Department of Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1025 Walnut Street, Suite 1100, Philadelphia, PA 19107, USA.
| | - William K Kelly
- Medical Oncology and Urology, Division of Solid Tumor Oncology, Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Sidney Kimmel Cancer Center, 1025 Walnut Street, Suite 700, Philadelphia, PA 19107, USA
| | - Leonard G Gomella
- Department of Urology, Thomas Jefferson University and Hospital, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1025 Walnut Street, Suite 1100, Philadelphia, PA 19107, USA. https://twitter.com/LeonardGomella
| |
Collapse
|
38
|
Hyatt C, McDougall C, Miller-Samuel S, Russo J. Genetic Counseling for Men with Prostate Cancer. Urol Clin North Am 2021; 48:323-337. [PMID: 34210488 DOI: 10.1016/j.ucl.2021.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Germline genetic testing is becoming more prevalent in urology clinics because of precision medicine for prostate cancer treatment. Genetic testing results can also influence cancer screening discussions for patients and/or their families. An important part of germline genetic testing is genetic counseling. This article provides an overview of the historical aspects of genetic counseling, discusses the components needed to provide proper genetic counseling, summarizes genes related to hereditary prostate cancer risk, and reviews genetic privacy and genetic discrimination concerns related to germline genetic testing.
Collapse
Affiliation(s)
- Colette Hyatt
- Familial Cancer Program, The University of Vermont Medical Center, Main Campus, East Pavilion, Level 2, 111 Colchester Avenue, Burlington, VT 05401, USA.
| | - Carey McDougall
- Sidney Kimmel Cancer Center, Clinical Cancer Genetics, 1100 Walnut Street, Suite 602, Philadelphia, PA 19107, USA
| | - Susan Miller-Samuel
- Sidney Kimmel Cancer Center, Clinical Cancer Genetics, 1100 Walnut Street, Suite 602, Philadelphia, PA 19107, USA
| | - Jessica Russo
- Sidney Kimmel Cancer Center, Clinical Cancer Genetics, 1100 Walnut Street, Suite 602, Philadelphia, PA 19107, USA
| |
Collapse
|
39
|
Helfand BT, Xu J. Germline Testing for Prostate Cancer Prognosis: Implications for Active Surveillance. Urol Clin North Am 2021; 48:401-409. [PMID: 34210494 DOI: 10.1016/j.ucl.2021.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Available evidence supports routine implementation of germline genetic testing for many aspects of prostate cancer (PCa) decision making. The purpose of obtaining genetic testing for newly diagnosed men would be focused on identifying mutations that predispose to aggressive PCa. Based on an evidence-based review, the authors review germline rare pathogenic mutations in several genes that are significantly associated with aggressiveness, metastases, and mortality. Then recent studies of these germline mutations in predicting tumor grade reclassification among patients undergoing active surveillance are discussed. Single nucleotide polymorphisms-based polygenic risk scores in differentiating PCa aggressiveness and prognosis are reviewed.
Collapse
Affiliation(s)
- Brian T Helfand
- Program for Personalized Cancer Care, Division of Urology, NorthShore University HealthSystem, 1001 University Place, Evanston, IL 60201, USA.
| | - Jianfeng Xu
- Program for Personalized Cancer Care, Division of Urology, NorthShore University HealthSystem, 1001 University Place, Evanston, IL 60201, USA
| |
Collapse
|
40
|
Exploring the most stable aptamer/target molecule complex by the stochastic tunnelling-basin hopping-discrete molecular dynamics method. Sci Rep 2021; 11:11406. [PMID: 34075115 PMCID: PMC8169667 DOI: 10.1038/s41598-021-90907-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 05/07/2021] [Indexed: 11/29/2022] Open
Abstract
The stochastic tunnelling-basin hopping-discrete molecular dynamics (STUN-BH-DMD) method was applied to the search for the most stable biomolecular complexes in water by using the MARTINI coarse-grained (CG) model. The epithelial cell adhesion molecule (EpCAM, PDB code: 4MZV) was used as an EpCAM adaptor for an EpA (AptEpA) benchmark target molecule. The effects of two adsorption positions on the EpCAM were analysed, and it is found that the AptEpA adsorption configuration located within the EpCAM pocket-like structure is more stable and the energy barrier is lower due to the interaction with water. By the root mean square deviation (RMSD), the configuration of EpCAM in water is more conservative when the AptEpA binds to EpCAM by attaching to the pocket space of the EpCAM dimer. For AptEpA, the root mean square fluctuation (RMSF) analysis result indicates Nucleobase 1 and Nucleobase 2 display higher flexibility during the CGMD simulation. Finally, from the binding energy contour maps and histogram plots of EpCAM and each AptEpA nucleobase, it is clear that the binding energy adsorbed to the pocket-like structure is more continuous than that energy not adsorbed to the pocket-like structure. This study has proposed a new numerical process for applying the STUN-BH-DMD with the CG model, which can reduce computational details and directly find a more stable AptEpA/EpCAM complex in water.
Collapse
|
41
|
Sabol RA, Ledet EM, Jaeger E, Hatton W, Moses M, Lankford A, Zaheria A, Barata P, Layton JL, Lewis BE, Sartor O. Family history and pathogenic/likely pathogenic germline variants in prostate cancer patients. Prostate 2021; 81:427-432. [PMID: 33760238 DOI: 10.1002/pros.24120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recent literature highlights the importance of germline genetic testing in prostate cancer (PCa) patients. Surprisingly, a literature review indicates that family history (FH) records are incomplete in the major published studies from prostate cancer patients. METHODS Prospective family history data were gathered from 496 men in a single institution with a personal history of PCa who underwent germline genetic testing using a panel of at least 79 genes. Comprehensive first degree FH were obtained in all PCa of patients and analysis of prevalent FH was assessed at the time of sample collection. RESULTS Pathogenic/likely pathogenic variants (PV/LPVs) were not associated with age at diagnosis, race, or presence of metastasis. One or more first degree relatives (FDR) with any cancer was not predictive for germline PV/LPVs for men with PCa (p = .96). Separate analysis of patients with one or more FDR with breast, prostate, ovarian, or pancreatic cancer revealed that only FDR with breast or ovarian cancer was predictive for PV/LPVs (p = .028, p = .015 respectively). Patients with a FDR with breast cancer had 1.8 increased risk of PV/LPVs, and patients with a FDR with ovarian cancer had 2.9 increased risk of PV/LPV. CONCLUSION In men with a personal history of PCa, germline PV/LPVs were associated with a FDR with breast or ovarian cancer. Notably having FDRs with PCa does not predict for PV/LPVs. These data emphasize the contribution of FH in a data set with complete ascertainment of FH.
Collapse
Affiliation(s)
- Rachel A Sabol
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elisa M Ledet
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Ellen Jaeger
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Whitley Hatton
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Marcus Moses
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Anjali Lankford
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Alexa Zaheria
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Pedro Barata
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jodi L Layton
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Brian E Lewis
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Department of Urology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
42
|
Bruneau M, Milliron BJ, Sinclair E, Obeid E, Gross L, Bealin L, Smaltz C, Butryn M, Giri VN. Physical activity assessment among men undergoing genetic counseling for inherited prostate cancer: a teachable moment for improved survivorship. Support Care Cancer 2021; 29:2145-2151. [PMID: 32876733 PMCID: PMC7897228 DOI: 10.1007/s00520-020-05667-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/31/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Genetic counseling (GC) presents an opportunity to address modifiable cancer risk factors, such as obesity, which is impacted by non-adherence to physical activity (PA) guidelines. Adherence to PA guidelines has not been assessed among men undergoing GC for prostate cancer (PCA). We conducted a targeted analysis of men undergoing PCA GC to assess adherence to PA recommendations. METHODS Using a cross-sectional design, a total of 158 men from the Genetic Evaluation of Men (GEM) study at two academic cancer centers with a diagnosis or at risk for PCA completed a structured lifestyle survey, including questions about the number of days and intensity of PA over the past year. One-sample t tests assessed adherence of participants to PA recommendations. Chi-square analyses compared differences in PA adherence by PCA status, aggressiveness, family history, and body mass index. Logistic regression analyses identified predictors of PA adherence. RESULTS High proportions of GEM participants were overweight (44.9%) or obese (38.0%, p = 0.002). Men with PCA engaged in less moderate (p = 0.019) and vigorous (p = 0.005) aerobic activity than men without PCA. Higher education was predictive of adherence to light (p = 0.008), moderate (p = 0.019), and vigorous (p = 0.002) intensity PA. Older age (p = 0.015) and higher education (p = 0.001) were predictive of adherence to strength-based recommendations. CONCLUSIONS High proportions of men receiving PCA GC were overweight/obese and lacked adherence to PA recommendations. GC represents a teachable moment to address PA to reduce cancer risk and promote cancer survivorship.
Collapse
Affiliation(s)
- Michael Bruneau
- Department of Health Sciences, College of Nursing and Health Professions, Drexel University, Philadelphia, PA, USA
| | - Brandy-Joe Milliron
- Department of Nutrition Sciences and Center for Family Intervention Science, College of Nursing and Health Professions, Drexel University, Philadelphia, PA, USA
| | - Elizabeth Sinclair
- Department of Health Sciences, College of Nursing and Health Professions, Drexel University, Philadelphia, PA, USA
| | - Elias Obeid
- Department of Clinical Genetics, Temple-Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Laura Gross
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lisa Bealin
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christa Smaltz
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Meghan Butryn
- Department of Psychology, College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Veda N Giri
- Cancer Risk Assessment and Clinical Cancer Genetics Program, Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Rare Germline Pathogenic Variants Identified by Multigene Panel Testing and the Risk of Aggressive Prostate Cancer. Cancers (Basel) 2021; 13:cancers13071495. [PMID: 33804961 PMCID: PMC8036662 DOI: 10.3390/cancers13071495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Identifying which men at the time of prostate cancer diagnosis have, or will progress to, an aggressive fatal disease will allow clinicians to assist men in making better informed treatment decisions. This will not only be important for those men whose disease is likely to remain indolent and who are currently undergoing unnecessary treatment procedures, but also for those who may need to be targeted with immediate and potentially life-saving therapy. Our case-control study confirms that men who carry BRCA1, BRCA2 and ATM germline pathogenic variants are at increased risk of aggressive disease and provides risk estimates that will be used by clinicians to improve counselling. Abstract While gene panel sequencing is becoming widely used for cancer risk prediction, its clinical utility with respect to predicting aggressive prostate cancer (PrCa) is limited by our current understanding of the genetic risk factors associated with predisposition to this potentially lethal disease phenotype. This study included 837 men diagnosed with aggressive PrCa and 7261 controls (unaffected men and men who did not meet criteria for aggressive PrCa). Rare germline pathogenic variants (including likely pathogenic variants) were identified by targeted sequencing of 26 known or putative cancer predisposition genes. We found that 85 (10%) men with aggressive PrCa and 265 (4%) controls carried a pathogenic variant (p < 0.0001). Aggressive PrCa odds ratios (ORs) were estimated using unconditional logistic regression. Increased risk of aggressive PrCa (OR (95% confidence interval)) was identified for pathogenic variants in BRCA2 (5.8 (2.7–12.4)), BRCA1 (5.5 (1.8–16.6)), and ATM (3.8 (1.6–9.1)). Our study provides further evidence that rare germline pathogenic variants in these genes are associated with increased risk of this aggressive, clinically relevant subset of PrCa. These rare genetic variants could be incorporated into risk prediction models to improve their precision to identify men at highest risk of aggressive prostate cancer and be used to identify men with newly diagnosed prostate cancer who require urgent treatment.
Collapse
|
44
|
Greenberg SE, Hunt TC, Ambrose JP, Lowrance WT, Dechet CB, O'Neil BB, Tward JD. Clinical Germline Testing Results of Men With Prostate Cancer: Patient-Level Factors and Implications of NCCN Guideline Expansion. JCO Precis Oncol 2021; 5:PO.20.00432. [PMID: 34250421 PMCID: PMC8232879 DOI: 10.1200/po.20.00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
Germline likely pathogenic or pathogenic variants (PVs) have been identified in up to 17% of men with prostate cancer (PC) and may drive disease severity or be targetable by novel therapies. National Comprehensive Cancer Network (NCCN) guidelines encouraging germline testing in metastatic PC were recently expanded to include all men with high-risk, very high-risk, or regional PC. Our aim was to assess the impact of expanded NCCN guidelines on the detection rate of germline PVs and to determine patient-level factors associated with a PV germline testing result. PATIENTS AND METHODS Men with PC underwent multigene germline genetic testing for PVs from June 2016 to December 2018, and trends were compared. The association of patient-level factors with a PV germline testing result, where ≥ 1 PV was identified, was assessed using analysis of variance and univariate logistic regression. Sensitivity analyses were limited to clinically actionable variants and those associated with disease severity or progression (BRCA1/2 and ATM). RESULTS Of 408 men undergoing germline testing, 42 (10.3%) men had PVs and 366 (89.7%) men did not have PVs identified. The proportion of men identified with a germline PV remained stable following testing criteria expansion (9.4% v 10.6%, P = .73). No patient-level factors were significantly associated with increased odds of a PV germline testing result, including age at diagnosis, race, pretreatment prostate-specific antigen, Gleason grade group, NCCN risk group, and family history of cancer (breast and/or ovarian, prostate, or any cancer). CONCLUSION This study demonstrated a stable PV detection rate in men with PC using expanded criteria aligned to the updated NCCN testing guidelines. However, we did not find strong evidence to suggest that patient-level factors are associated with PV germline testing results. These findings support the recent expansion of NCCN germline testing guidelines in PC.
Collapse
Affiliation(s)
- Samantha E. Greenberg
- Genetic Counseling Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Trevor C. Hunt
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jacob P. Ambrose
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - William T. Lowrance
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Christopher B. Dechet
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Brock B. O'Neil
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jonathan D. Tward
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
45
|
Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Goggins M, Hutton ML, Karlan BY, Khan S, Klein C, Kohlmann W, Kurian AW, Laronga C, Litton JK, Mak JS, Menendez CS, Merajver SD, Norquist BS, Offit K, Pederson HJ, Reiser G, Senter-Jamieson L, Shannon KM, Shatsky R, Visvanathan K, Weitzel JN, Wick MJ, Wisinski KB, Yurgelun MB, Darlow SD, Dwyer MA. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021; 19:77-102. [DOI: 10.6004/jnccn.2021.0001] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Guidelines for Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic focus primarily on assessment of pathogenic or likely pathogenic variants associated with increased risk of breast, ovarian, and pancreatic cancer and recommended approaches to genetic testing/counseling and management strategies in individuals with these pathogenic or likely pathogenic variants. This manuscript focuses on cancer risk and risk management for BRCA-related breast/ovarian cancer syndrome and Li-Fraumeni syndrome. Carriers of a BRCA1/2 pathogenic or likely pathogenic variant have an excessive risk for both breast and ovarian cancer that warrants consideration of more intensive screening and preventive strategies. There is also evidence that risks of prostate cancer and pancreatic cancer are elevated in these carriers. Li-Fraumeni syndrome is a highly penetrant cancer syndrome associated with a high lifetime risk for cancer, including soft tissue sarcomas, osteosarcomas, premenopausal breast cancer, colon cancer, gastric cancer, adrenocortical carcinoma, and brain tumors.
Collapse
Affiliation(s)
| | - Tuya Pal
- 2Vanderbilt-Ingram Cancer Center
| | - Michael P. Berry
- 3St. Jude Children’s Research Hospital/The University of Tennessee Health Science Center
| | | | - Patricia Dickson
- 5Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Michael Goggins
- 9The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Seema Khan
- 12Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | | | | | | | | | | | | | - Holly J. Pederson
- 22Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Borque-Fernando A, Espílez R, Miramar D, Corbatón D, Rodríguez A, Castro E, Mateo J, Rello L, Méndez A, Gil Sanz MJ. Genetic counseling in prostate cancer: How to implement it in daily clinical practice? Actas Urol Esp 2021; 45:8-20. [PMID: 33059945 DOI: 10.1016/j.acuro.2020.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/22/2020] [Indexed: 11/16/2022]
Abstract
Prostate cancer plays an undeniably prominent role in public health in our days and health systems. Its epidemiological impact is quantitatively very close to that of other tumors such as colon cancer and breast cancer, in which genetic counseling is part of their routine clinical practice, both in the initial evaluation and in the selection of therapeutic strategies. Hereditary cancer syndromes, breast/ovarian and Lynch syndrome are part of genetic counseling in these tumors. Currently, we also know that they can be associated to prostate cancer. The time has come to implement genetic counseling in prostate cancer from the earliest stages of its approach, from initial suspicion to the most advanced tumors. We present an updated review carried out by our interdisciplinary working group on scientific literature, clinical practice guidelines and consensus documents, aimed at the creation and drafting of a'Protocol for genetic counseling in prostate cancer' for the study of germline, with easy application in different healthcare settings. This protocol is currently being implemented in our routine practice and provides answers to 3 specific questions: Who should receive genetic counseling for prostate cancer? Which gene panel should be analyzed? How should counseling be done according to the results obtained? Other aspects about who should perform genetic counseling, ethical considerations and regulations are also collected.
Collapse
Affiliation(s)
- A Borque-Fernando
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España.
| | - R Espílez
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - D Miramar
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - D Corbatón
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - A Rodríguez
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - E Castro
- Departamento de Oncología Médica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Málaga, España
| | - J Mateo
- Instituto de Oncología Vall d'Hebron y Hospital Universitario Vall d'Hebron, Barcelona, España
| | - L Rello
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - A Méndez
- Servicio de Oncología Radioterápica, Hospital Universitario Miguel Servet, Zaragoza, España
| | - M J Gil Sanz
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| |
Collapse
|
47
|
Loeb S, Giri VN. Clinical Implications of Germline Testing in Newly Diagnosed Prostate Cancer. Eur Urol Oncol 2020; 4:1-9. [PMID: 33390340 DOI: 10.1016/j.euo.2020.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/28/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022]
Abstract
CONTEXT Germline testing (GT) is increasingly impacting prostate cancer (PCa) management and screening, with direct effects in urology, medical oncology, and radiation oncology. The majority of testing indications and recommendations center on men with metastatic disease, although guidelines now encompass newly diagnosed, early-stage PCa and entail assessment of personal history, pathologic features, and family history to determine eligibility for testing. OBJECTIVE To describe current guidelines on GT for men with PCa and the impact on management. An additional objective was to review the literature on current uptake of GT across practice settings. EVIDENCE ACQUISITION A nonsystematic review was performed of current guidelines on GT in PCa from professional societies and consensus conferences, detailing supporting evidence for these recommendations. This was supplemented by a literature review of uptake of GT and precision medicine in practice. EVIDENCE SYNTHESIS Multiple guidelines and consensus panels recommend GT for men with metastatic PCa. Guidelines endorse BRCA2 testing in metastatic PCa because of strong evidence for PCa risk, aggressiveness, and PARP inhibitor candidacy. Testing of additional DNA repair genes in metastatic disease is also endorsed across guidelines. Immunotherapy with pembrolizumab is an option in some guidelines for men with DNA mismatch repair deficiency. In localized disease, GT is recommended on the basis of histologic features and family history; criteria vary between guidelines. GT for localized disease informs hereditary cancer risk and will probably impact future PCa management. Practice gaps exist regarding utilization of GT. CONCLUSIONS Germline evaluation is increasingly important in the management of men with metastatic PCa and may also affect the prognosis for men with localized disease. The presence of germline mutations has important hereditary cancer implications for men and their families. Uptake of germline evaluation may be underutilized in some practice settings, so strategies for optimization are required. PATIENT SUMMARY Patients with prostate cancer should talk to their doctor about the pros and cons of genetic testing, with attention to family history and cancer features. Genetic testing can have important implications for treatment, cancer screening, and family cancer risk.
Collapse
Affiliation(s)
- Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, New York, NY, USA
| | - Veda N Giri
- Cancer Risk Assessment and Clinical Cancer Genetics, Departments of Medical Oncology, Cancer Biology, and Urology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Stolarova L, Kleiblova P, Janatova M, Soukupova J, Zemankova P, Macurek L, Kleibl Z. CHEK2 Germline Variants in Cancer Predisposition: Stalemate Rather than Checkmate. Cells 2020; 9:cells9122675. [PMID: 33322746 PMCID: PMC7763663 DOI: 10.3390/cells9122675] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Germline alterations in many genes coding for proteins regulating DNA repair and DNA damage response (DDR) to DNA double-strand breaks (DDSB) have been recognized as pathogenic factors in hereditary cancer predisposition. The ATM-CHEK2-p53 axis has been documented as a backbone for DDR and hypothesized as a barrier against cancer initiation. However, although CHK2 kinase coded by the CHEK2 gene expedites the DDR signal, its function in activation of p53-dependent cell cycle arrest is dispensable. CHEK2 mutations rank among the most frequent germline alterations revealed by germline genetic testing for various hereditary cancer predispositions, but their interpretation is not trivial. From the perspective of interpretation of germline CHEK2 variants, we review the current knowledge related to the structure of the CHEK2 gene, the function of CHK2 kinase, and the clinical significance of CHEK2 germline mutations in patients with hereditary breast, prostate, kidney, thyroid, and colon cancers.
Collapse
Affiliation(s)
- Lenka Stolarova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic; (L.S.); (M.J.); (J.S.); (P.Z.)
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Petra Kleiblova
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, 12800 Prague, Czech Republic;
| | - Marketa Janatova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic; (L.S.); (M.J.); (J.S.); (P.Z.)
| | - Jana Soukupova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic; (L.S.); (M.J.); (J.S.); (P.Z.)
| | - Petra Zemankova
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic; (L.S.); (M.J.); (J.S.); (P.Z.)
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Zdenek Kleibl
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 12800 Prague, Czech Republic; (L.S.); (M.J.); (J.S.); (P.Z.)
- Correspondence: ; Tel.: +420-22496-745
| |
Collapse
|
49
|
Black MH, Li S, LaDuca H, Lo M, Chen J, Hoiness R, Gutierrez S, Tippin‐Davis B, Lu H, Gielzak M, Wiley K, Shi Z, Wei J, Zheng SL, Helfand BT, Isaacs W, Xu J. Validation of a prostate cancer polygenic risk score. Prostate 2020; 80:1314-1321. [PMID: 33258481 PMCID: PMC7590110 DOI: 10.1002/pros.24058] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/31/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Genome-wide association studies have identified over 100 single-nucleotide polymorphisms (SNPs) associated with prostate cancer (PrCa), and polygenic risk scores (PRS) based on their combined genotypes have been developed for risk stratification. We aimed to assess the contribution of PRS to PrCa risk in a large multisite study. METHODS The sample included 1972 PrCa cases and 1919 unaffected controls. Next-generation sequencing was used to assess pathogenic variants in 14 PrCa-susceptibility genes and 72 validated PrCa-associated SNPs. We constructed a population-standardized PRS and tested its association with PrCa using logistic regression adjusted for age and family history of PrCa. RESULTS The mean age of PrCa cases at diagnosis and age of controls at testing/last clinic visit was 59.5 ± 7.2 and 57.2 ± 13.0 years, respectively. Among 1740 cases with pathology data, 57.4% had Gleason score ≤ 6, while 42.6% had Gleason score ≥ 8. In addition, 39.6% cases and 20.1% controls had a family history of PrCa. The PRS was significantly higher in cases than controls (mean ± SD: 1.42 ± 1.11 vs 1.02 ± 0.76; P < .0001). Compared with men in the 1st quartile of age-adjusted PRS, those in the 2nd, 3rd, and 4th quartile were 1.58 (95% confidence interval [CI]: 1.31-1.90), 2.36 (95% CI: 1.96-2.84), and 3.98 (95% CI: 3.29-4.82) times as likely to have PrCa (all P < .0001). Adjustment for family history yielded similar results. PRS predictive performance was consistent with prior literature (area under the receiver operating curve = 0.64; 95% CI: 0.62-0.66). CONCLUSIONS These data suggest that a 72-SNP PRS is predictive of PrCa, supporting its potential use in clinical risk assessment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Marta Gielzak
- Department of Urology, The James Buchanan Brady Urologic InstituteJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Kathleen Wiley
- Department of Urology, The James Buchanan Brady Urologic InstituteJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Zhuqing Shi
- Program for Personalized Cancer CareNorthShore University Health SystemEvanstonIllinois
| | - Jun Wei
- Program for Personalized Cancer CareNorthShore University Health SystemEvanstonIllinois
| | - Siqun Lilly Zheng
- Program for Personalized Cancer CareNorthShore University Health SystemEvanstonIllinois
| | - Brian T. Helfand
- Program for Personalized Cancer CareNorthShore University Health SystemEvanstonIllinois
| | - William Isaacs
- Department of Urology, The James Buchanan Brady Urologic InstituteJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Jianfeng Xu
- Program for Personalized Cancer CareNorthShore University Health SystemEvanstonIllinois
| |
Collapse
|
50
|
Qian K, Wang G, Ju L, Liu J, Luo Y, Wang Y, Peng T, Chen F, Zhang Y, Xiao Y, Wang X. A novel germline EGFR variant p.R831H causes predisposition to familial CDK12-mutant prostate cancer with tandem duplicator phenotype. Oncogene 2020; 39:6871-6878. [PMID: 32978518 DOI: 10.1038/s41388-020-01476-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 09/02/2020] [Accepted: 09/15/2020] [Indexed: 02/05/2023]
Abstract
5-10% of total prostate cancer (PCa) cases are hereditary. Particularly, immune checkpoint inhibitor-sensitive tandem duplicator phenotype (TDP) accounts for 6.9% of PCa cases, whereas genetic susceptibility genes remain completely unknown. We identified a Chinese family with two PCa patients, in which the PCa phenotype co-segregated with a rare germline variant EGFRR831H. Patient-derived conditionally reprogrammed cells (CRC) exhibited increased EGFR and AKT phosphorylation, and a sensitivity to EGFR antagonist Afatinib in migration assays, suggesting the EGFR allele was constitutively active. Both EGFRR831H-mutant tumours contained biallelic CDK12 inactivation, together with prominent tandem duplication across the genome. These somatic mutations could be detected in urine before surgery. Analysis of public databases showed a significant correlation between the mutation status of EGFR and CDK12. Taken together, our genetic and functional analyses identified a previously undescribed link between EGFR and PCa.
Collapse
Affiliation(s)
- Kaiyu Qian
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yongwen Luo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yejinpeng Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianchen Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fangjin Chen
- Center of Life Sciences, Peking University, Beijing, China
| | - Yi Zhang
- Center of Life Sciences, Peking University, Beijing, China.
- Euler Technology, Beijing, China.
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Medical Research Institute, Wuhan University, Wuhan, China.
| |
Collapse
|