1
|
Kalampokis N, Zabaftis C, Spinos T, Karavitakis M, Leotsakos I, Katafigiotis I, van der Poel H, Grivas N, Mitropoulos D. Review on the Role of BRCA Mutations in Genomic Screening and Risk Stratification of Prostate Cancer. Curr Oncol 2024; 31:1162-1169. [PMID: 38534919 PMCID: PMC10969585 DOI: 10.3390/curroncol31030086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Somatic and germline alterations can be commonly found in prostate cancer (PCa) patients. The aim of our present study was to perform a comprehensive review of the current literature in order to examine the impact of BRCA mutations in the context of PCa as well as their significance as genetic biomarkers. (2) Methods: A narrative review of all the available literature was performed. Only "landmark" publications were included. (3) Results: Overall, the number of PCa patients who harbor a BRCA2 mutation range between 1.2% and 3.2%. However, BRCA2 and BRCA1 mutations are responsible for most cases of hereditary PCa, increasing the risk by 3-8.6 times and up to 4 times, respectively. These mutations are correlated with aggressive disease and poor prognosis. Gene testing should be offered to patients with metastatic PCa, those with 2-3 first-degree relatives with PCa, or those aged < 55 and with one close relative with breast (age ≤ 50 years) or invasive ovarian cancer. (4) Conclusions: The individualized assessment of BRCA mutations is an important tool for the risk stratification of PCa patients. It is also a population screening tool which can guide our risk assessment strategies and achieve better results for our patients and their families.
Collapse
Affiliation(s)
- Nikolaos Kalampokis
- Department of Urology, G. Hatzikosta General Hospital, 45001 Ioannina, Greece;
| | - Christos Zabaftis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Theodoros Spinos
- Department of Urology, University of Patras Hospital, 26504 Patras, Greece;
| | - Markos Karavitakis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Leotsakos
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Ioannis Katafigiotis
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
| | - Henk van der Poel
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Nikolaos Grivas
- Department of Laparoscopy and Endourology, Central Urology, Lefkos Stavros the Athens Clinic, PC 11528 Athens, Greece; (C.Z.); (M.K.); (I.L.); (I.K.)
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Dionysios Mitropoulos
- Department of Urology, Medical School, National & Kapodistrian University of Athens, 14122 Athens, Greece;
| |
Collapse
|
2
|
Brook MN, Ní Raghallaigh H, Kote-Jarai Z, Eeles RA. Reply to Kari Hemminki, Jianguang Ji, and Jan Sundquist's Letter to the Editor re: Mark N. Brook, Holly Ní Raghallaigh, Koveela Govindasami, et al. Family History of Prostate Cancer and Survival Outcomes in the UK Genetic Prostate Cancer Study. Eur Urol 2023;83:257-66. Eur Urol 2023:S0302-2838(23)02719-7. [PMID: 37061444 DOI: 10.1016/j.eururo.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 04/17/2023]
Affiliation(s)
| | | | | | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK; Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Hemminki K, Ji J, Sundquist J. Re: Mark N. Brook, Holly Ní Raghallaigh, Koveela Govindasami, et al. Family History of Prostate Cancer and Survival Outcomes in the UK Genetic Prostate Cancer Study. Eur Urol 2023;83:257-66. Eur Urol 2023:S0302-2838(23)02718-5. [PMID: 37061447 DOI: 10.1016/j.eururo.2023.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 04/17/2023]
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czechia; Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany; Center for Primary Health Care Research, Lund University, Malmö, Sweden.
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Community-based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan
| |
Collapse
|
4
|
Díaz-Fernández F, Celma A, Salazar A, Moreno O, López C, Cuadras M, Regis L, Planas J, Morote J, Trilla E. Systematic review of methods used to improve the efficacy of magnetic resonance in early detection of clinically significant prostate cancer. Actas Urol Esp 2023; 47:127-139. [PMID: 36462603 DOI: 10.1016/j.acuroe.2022.11.007] [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: 04/13/2021] [Accepted: 04/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND OBJECTIVE Prostate cancer (PC) is the malignant neoplasm with the highest incidence after lung cancer worldwide. The objective of this study is to review the literature on the methods that improve the efficacy of the current strategy for the early diagnosis of clinically significant PC (csPC), based on the performance of magnetic resonance imaging (RM) and targeted biopsies when suspicious lesions are detected, in addition to systematic biopsy. EVIDENCE ACQUISITION A systematic literature review was performed in PubMed, Web of Science and Cochrane according to the PRISMA criteria (Preferred Reporting Items for Systematic Reviews and Meta-Analyses), using the search terms: multiparametric magnetic resonance imaging, biparametric magnetic resonance imaging, biomarkers in prostate cancer, prostate cancer y early diagnosis. A total of 297 references were identified and, using the PICO selection criteria, 21 publications were finally selected to synthesize the evidence. EVIDENCE SYNTHESIS With the consolidation of MRI as the test of choice for the diagnosis of prostate cancer, the role of PSA density (PSAD) becomes relevant as a predictive tool included in prediction nomograms, without added cost. PSAD and diagnostic markers, combined with MRI, offer a high diagnostic power with an area under curve (AUC) above 0.7. Only the SHTLM3 model integrates markers in the creation of a nomogram. Prediction models also offer consistent efficacy with an AUC greater than 0.8 when associating MRI. CONCLUSIONS The efficacy of MRI in clinically significant prostate cancer detection can be improved with different parameters in order to generate predictive models that support decision making.
Collapse
Affiliation(s)
- F Díaz-Fernández
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
| | - A Celma
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - A Salazar
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - O Moreno
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - C López
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - M Cuadras
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - L Regis
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - J Planas
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - J Morote
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Universistat Autònoma de Barcelona, Barcelona, Spain
| | - E Trilla
- Departamento de Urología y Trasplante Renal, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Universistat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Nyberg T, Brook MN, Ficorella L, Lee A, Dennis J, Yang X, Wilcox N, Dadaev T, Govindasami K, Lush M, Leslie G, Lophatananon A, Muir K, Bancroft E, Easton DF, Tischkowitz M, Kote-Jarai Z, Eeles R, Antoniou AC. CanRisk-Prostate: A Comprehensive, Externally Validated Risk Model for the Prediction of Future Prostate Cancer. J Clin Oncol 2023; 41:1092-1104. [PMID: 36493335 PMCID: PMC9928632 DOI: 10.1200/jco.22.01453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/26/2022] [Accepted: 10/07/2022] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Prostate cancer (PCa) is highly heritable. No validated PCa risk model currently exists. We therefore sought to develop a genetic risk model that can provide personalized predicted PCa risks on the basis of known moderate- to high-risk pathogenic variants, low-risk common genetic variants, and explicit cancer family history, and to externally validate the model in an independent prospective cohort. MATERIALS AND METHODS We developed a risk model using a kin-cohort comprising individuals from 16,633 PCa families ascertained in the United Kingdom from 1993 to 2017 from the UK Genetic Prostate Cancer Study, and complex segregation analysis adjusting for ascertainment. The model was externally validated in 170,850 unaffected men (7,624 incident PCas) recruited from 2006 to 2010 to the independent UK Biobank prospective cohort study. RESULTS The most parsimonious model included the effects of pathogenic variants in BRCA2, HOXB13, and BRCA1, and a polygenic score on the basis of 268 common low-risk variants. Residual familial risk was modeled by a hypothetical recessively inherited variant and a polygenic component whose standard deviation decreased log-linearly with age. The model predicted familial risks that were consistent with those reported in previous observational studies. In the validation cohort, the model discriminated well between unaffected men and men with incident PCas within 5 years (C-index, 0.790; 95% CI, 0.783 to 0.797) and 10 years (C-index, 0.772; 95% CI, 0.768 to 0.777). The 50% of men with highest predicted risks captured 86.3% of PCa cases within 10 years. CONCLUSION To our knowledge, this is the first validated risk model offering personalized PCa risks. The model will assist in counseling men concerned about their risk and can facilitate future risk-stratified population screening approaches.
Collapse
Affiliation(s)
- Tommy Nyberg
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Mark N. Brook
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Lorenzo Ficorella
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Andrew Lee
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Xin Yang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Naomi Wilcox
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Tokhir Dadaev
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Koveela Govindasami
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Michael Lush
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Goska Leslie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Artitaya Lophatananon
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Kenneth Muir
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Elizabeth Bancroft
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Marc Tischkowitz
- Department of Medical Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Zsofia Kote-Jarai
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Rosalind Eeles
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Antonis C. Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Ugai T, Sasamoto N, Lee HY, Ando M, Song M, Tamimi RM, Kawachi I, Campbell PT, Giovannucci EL, Weiderpass E, Rebbeck TR, Ogino S. Is early-onset cancer an emerging global epidemic? Current evidence and future implications. Nat Rev Clin Oncol 2022; 19:656-673. [PMID: 36068272 PMCID: PMC9509459 DOI: 10.1038/s41571-022-00672-8] [Citation(s) in RCA: 159] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 02/07/2023]
Abstract
Over the past several decades, the incidence of early-onset cancers, often defined as cancers diagnosed in adults <50 years of age, in the breast, colorectum, endometrium, oesophagus, extrahepatic bile duct, gallbladder, head and neck, kidney, liver, bone marrow, pancreas, prostate, stomach and thyroid has increased in multiple countries. Increased use of screening programmes has contributed to this phenomenon to a certain extent, although a genuine increase in the incidence of early-onset forms of several cancer types also seems to have emerged. Evidence suggests an aetiological role of risk factor exposures in early life and young adulthood. Since the mid-20th century, substantial multigenerational changes in the exposome have occurred (including changes in diet, lifestyle, obesity, environment and the microbiome, all of which might interact with genomic and/or genetic susceptibilities). However, the effects of individual exposures remain largely unknown. To study early-life exposures and their implications for multiple cancer types will require prospective cohort studies with dedicated biobanking and data collection technologies. Raising awareness among both the public and health-care professionals will also be critical. In this Review, we describe changes in the incidence of early-onset cancers globally and suggest measures that are likely to reduce the burden of cancers and other chronic non-communicable diseases.
Collapse
Affiliation(s)
- Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Naoko Sasamoto
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Hwa-Young Lee
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Institute of Convergence Science, Convergence Science Academy, Yonsei University, Seoul, Republic of Korea
| | - Mariko Ando
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Rulla M Tamimi
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Ichiro Kawachi
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Zhu Family Center for Global Cancer Prevention, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
7
|
Revisión sistemática de los métodos para incrementar la eficacia de la resonancia magnética en el diagnóstico precoz de cáncer de próstata clínicamente significativo. Actas Urol Esp 2022. [DOI: 10.1016/j.acuro.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Clements MB, Vertosick EA, Guerrios-Rivera L, De Hoedt AM, Hernandez J, Liss MA, Leach RJ, Freedland SJ, Haese A, Montorsi F, Boorjian SA, Poyet C, Ankerst DP, Vickers AJ. Defining the Impact of Family History on Detection of High-grade Prostate Cancer in a Large Multi-institutional Cohort. Eur Urol 2021; 82:163-169. [PMID: 34980493 PMCID: PMC9243191 DOI: 10.1016/j.eururo.2021.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The risk of high-grade prostate cancer, given a family history of cancer, has been described in the general population, but not among men selected for prostate biopsy in an international cohort. OBJECTIVE To estimate the risk of high-grade prostate cancer on biopsy based on a family history of cancer. DESIGN, SETTING, AND PARTICIPANTS This is a multicenter study of men undergoing prostate biopsy from 2006 to 2019, including 12 sites in North America and Europe. All sites recorded first-degree prostate cancer family histories; four included more detailed data on the number of affected relatives, second-degree relatives with prostate cancer, and breast cancer family history. OUTCOMES MEASUREMENTS AND STATISTICAL ANALYSIS Multivariable logistic regressions evaluated odds of high-grade (Gleason grade group ≥2) prostate cancer. Separate models were fit for family history definitions, including first- and second-degree prostate cancer and breast cancer family histories. RESULTS AND LIMITATIONS A first-degree prostate cancer family history was available for 15 799 men, with a more detailed family history for 4617 (median age 65 yr, both cohorts). Adjusted odds of high-grade prostate cancer were 1.77 times greater (95% confidence interval [CI] 1.57-2.00, p < 0.001, risk ratio [RR] = 1.40) with first-degree prostate cancer, 1.38 (95% CI 1.07-1.77, p = 0.011, RR = 1.22) for second-degree prostate cancer, and 1.30 (95% CI 1.01-1.67, p = 0.040, RR = 1.18) for first-degree breast cancer family histories. Interaction terms revealed that the effect of a family history did not differ based on prostate-specific antigen but differed based on age. This study is limited by missing data on race and prior negative biopsy. CONCLUSIONS Men with indications for biopsy and a family history of prostate or breast cancer can be counseled that they have a moderately increased risk of high-grade prostate cancer, independent of other risk factors. PATIENT SUMMARY In a large international series of men selected for prostate biopsy, finding a high-grade prostate cancer was more likely in men with a family history of prostate or breast cancer.
Collapse
Affiliation(s)
- Matthew B Clements
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily A Vertosick
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lourdes Guerrios-Rivera
- Department of Surgery, Urology Section, Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, USA
| | - Amanda M De Hoedt
- Section of Urology, Durham Veterans Administration Health Care System, Durham, NC, USA
| | - Javier Hernandez
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael A Liss
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robin J Leach
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Stephen J Freedland
- Section of Urology, Durham Veterans Administration Health Care System, Durham, NC, USA; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexander Haese
- Martini-Clinic Prostate Cancer Center, University Clinic Eppendorf, Hamburg, Germany
| | - Francesco Montorsi
- Division of Oncology/Unit of Urology, URI, IRCCS Hospital San Raffaele, Milano, Italy; Department of Medicine, Vita-Salute San Raffaele University, Milano, Italy
| | | | - Cedric Poyet
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Donna P Ankerst
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Mathematics, Technical University of Munich, Garching, Munich, Germany
| | - Andrew J Vickers
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
9
|
Familial Risks and Proportions Describing Population Landscape of Familial Cancer. Cancers (Basel) 2021; 13:cancers13174385. [PMID: 34503195 PMCID: PMC8430802 DOI: 10.3390/cancers13174385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/10/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Familial cancer can be defined through the occurrence of the same cancer in two or more family members. Hereditary cancer is a narrower definition of high-risk familial aggregation through identified predisposing genes. The absence of correlation between spouses for risk of most cancers, particularly those not related to tobacco smoking or solar exposure, suggests that familial cancers are mainly due to genetic causes. The aim of the present study was to define the frequency and increased risk for familial cancer. Data on 31 of the most common cancers were obtained from the Swedish Family-Cancer Database and familial relative risks (SIRs) were estimated between persons with or without family history of the same cancer in first-degree relatives. Practically all cancers showed a familial risk, with an SIR most commonly around two, or a doubling of the risk because of family history. Abstract Background: Familial cancer can be defined through the occurrence of the same cancer in two or more family members. We describe a nationwide landscape of familial cancer, including its frequency and the risk that it conveys, by using the largest family database in the world with complete family structures and medically confirmed cancers. Patients/methods: We employed standardized incidence ratios (SIRs) to estimate familial risks for concordant cancer among first-degree relatives using the Swedish Cancer Registry from years 1958 through 2016. Results: Cancer risks in a 20–84 year old population conferred by affected parents or siblings were about two-fold compared to the risk for individuals with unaffected relatives. For small intestinal, testicular, thyroid and bone cancers and Hodgkin disease, risks were higher, five-to-eight-fold. Novel familial associations included adult bone, lip, pharyngeal, and connective tissue cancers. Familial cancers were found in 13.2% of families with cancer; for prostate cancer, the proportion was 26.4%. High-risk families accounted for 6.6% of all cancer families. Discussion/Conclusion: High-risk family history should be exceedingly considered for management, including targeted genetic testing. For the major proportion of familial clustering, where genetic testing may not be feasible, medical and behavioral intervention should be indicated for the patient and their family members, including screening recommendations and avoidance of carcinogenic exposure.
Collapse
|
10
|
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: 1.3] [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
|
11
|
Xu X, Kharazmi E, Tian Y, Mukama T, Sundquist K, Sundquist J, Brenner H, Fallah M. Risk of prostate cancer in relatives of prostate cancer patients in Sweden: A nationwide cohort study. PLoS Med 2021; 18:e1003616. [PMID: 34061847 PMCID: PMC8168897 DOI: 10.1371/journal.pmed.1003616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Evidence-based guidance for starting ages of screening for first-degree relatives (FDRs) of patients with prostate cancer (PCa) to prevent stage III/IV or fatal PCa is lacking in current PCa screening guidelines. We aimed to provide evidence for risk-adapted starting age of screening for relatives of patients with PCa. METHODS AND FINDINGS In this register-based nationwide cohort study, all men (aged 0 to 96 years at baseline) residing in Sweden who were born after 1931 along with their fathers were included. During the follow-up (1958 to 2015) of 6,343,727 men, 88,999 were diagnosed with stage III/IV PCa or died of PCa. The outcomes were defined as the diagnosis of stage III/IV PCa or death due to PCa, stratified by age at diagnosis. Using 10-year cumulative risk curves, we calculated risk-adapted starting ages of screening for men with different constellations of family history of PCa. The 10-year cumulative risk of stage III/IV or fatal PCa in men at age 50 in the general population (a common recommended starting age of screening) was 0.2%. Men with ≥2 FDRs diagnosed with PCa reached this screening level at age 41 (95% confidence interval (CI): 39 to 44), i.e., 9 years earlier, when the youngest one was diagnosed before age 60; at age 43 (41 to 47), i.e., 7 years earlier, when ≥2 FDRs were diagnosed after age 59, which was similar to that of men with 1 FDR diagnosed before age 60 (41 to 45); and at age 45 (44 to 46), when 1 FDR was diagnosed at age 60 to 69 and 47 (46 to 47), when 1 FDR was diagnosed after age 69. We also calculated risk-adapted starting ages for other benchmark screening ages, such as 45, 55, and 60 years, and compared our findings with those in the guidelines. Study limitations include the lack of genetic data, information on lifestyle, and external validation. CONCLUSIONS Our study provides practical information for risk-tailored starting ages of PCa screening based on nationwide cancer data with valid genealogical information. Our clinically relevant findings could be used for evidence-based personalized PCa screening guidance and supplement current PCa screening guidelines for relatives of patients with PCa.
Collapse
Affiliation(s)
- Xing Xu
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Elham Kharazmi
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Institute of Medical Biometry and Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Yu Tian
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Trasias Mukama
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
- Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Izumo, Japan
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
- Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Izumo, Japan
| | - Hermann Brenner
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mahdi Fallah
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- * E-mail:
| |
Collapse
|
12
|
Hunt TC, Ambrose JP, Haaland B, Kawamoto K, Dechet CB, Lowrance WT, Hanson HA, O'Neil BB. Decision fatigue in low-value prostate cancer screening. Cancer 2021; 127:3343-3353. [PMID: 34043813 DOI: 10.1002/cncr.33644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/22/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Low-value prostate-specific antigen (PSA) testing is common yet contributes substantial waste and downstream patient harm. Decision fatigue may represent an actionable target to reduce low-value urologic care. The objective of this study was to determine whether low-value PSA testing patterns by outpatient clinicians are consistent with decision fatigue. METHODS Outpatient appointments for adult men without prostate cancer were identified at a large academic health system from 2011 through 2018. The authors assessed the association of appointment time with the likelihood of PSA testing, stratified by patient age and appropriateness of testing based on clinical guidelines. Appointments included those scheduled between 8:00 am and 4:59 pm, with noon omitted. Urologists were examined separately from other clinicians. RESULTS In 1,581,826 outpatient appointments identified, the median patient age was 54 years (interquartile range, 37-66 years), 1,256,152 participants (79.4%) were White, and 133,693 (8.5%) had family history of prostate cancer. PSA testing would have been appropriate in 36.8% of appointments. Clinicians ordered testing in 3.6% of appropriate appointments and in 1.8% of low-value appointments. Appropriate testing was most likely at 8:00 am (reference group). PSA testing declined through 11:00 am (odds ratio [OR], 0.57; 95% CI, 0.50-0.64) and remained depressed through 4:00 pm (P < .001). Low-value testing was overall less likely (P < .001) and followed a similar trend, declining steadily from 8:00 am (OR, 0.48; 95% CI, 0.42-0.56) through 4:00 pm (P < .001; OR, 0.23; 95% CI, 0.18-0.30). Testing patterns in urologists were noticeably different. CONCLUSIONS Among most clinicians, outpatient PSA testing behaviors appear to be consistent with decision fatigue. These findings establish decision fatigue as a promising, actionable target for reducing wasteful and low-value practices in routine urologic care. LAY SUMMARY Decision fatigue causes poorer choices to be made with repetitive decision making. This study used medical records to investigate whether decision fatigue influenced clinicians' likelihood of ordering a low-value screening test (prostate-specific antigen [PSA]) for prostate cancer. In more than 1.5 million outpatient appointments by adult men without prostate cancer, the chances of both appropriate and low-value PSA testing declined as the clinic day progressed, with a larger decline for appropriate testing. Testing patterns in urologists were different from those reported by other clinicians. The authors conclude that outpatient PSA testing behaviors appear to be consistent with decision fatigue among most clinicians, and interventions may reduce wasteful testing and downstream patient harms.
Collapse
Affiliation(s)
- Trevor C Hunt
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jacob P Ambrose
- Population Sciences, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Benjamin Haaland
- Division of Biostatistics, Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Kensaku Kawamoto
- Department of Biomedical Informatics, University of Utah, Salt Lake City, Utah
| | - Christopher B Dechet
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - William T Lowrance
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Heidi A Hanson
- Population Sciences, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Brock B O'Neil
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
13
|
[Familial prostate cancer and genetic predisposition]. Urologe A 2021; 60:567-575. [PMID: 33721089 DOI: 10.1007/s00120-021-01491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Twenty percent of all prostate cancer patients have a positive family history (at least 1 first-degree relative with prostate cancer) and a part of these patients have a genetic predisposition. OBJECTIVES A literature search and analysis of studies investigating incidence, diagnosis, and clinical course of familial compared to sporadic prostate cancer as well as genetic predisposition was performed using PubMed and Embase. RESULTS Risk of prostate cancer depends on number, degree of relationship, and age of onset of affected men in the family. The incidence of familial prostate cancer is higher and the age of diagnosis lower compared to sporadic cases. The clinical course of the disease is comparable, but in individuals with a germline mutation, more intensive therapy is needed due to a more aggressive disease. CONCLUSIONS Crucial for risk assessment is a detailed family history, including creation of a pedigree with cancer family history if necessary. In high-risk families, genetic counselling and annual prostate-specific antigen (PSA) screening beginning at the age of 40 should be performed. Verification of a germline mutation requires more intensive therapy due to more aggressive disease.
Collapse
|
14
|
Choi HG, Bang WJ, Jo JK, Oh CY, Shim M, Cho JS. The association between family history of prostate cancer and development of prostate cancer among Korean population: A prospective cohort study using KoGES data. Medicine (Baltimore) 2021; 100:e24757. [PMID: 33607822 PMCID: PMC7899810 DOI: 10.1097/md.0000000000024757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/25/2021] [Indexed: 01/05/2023] Open
Abstract
This study aimed to assess the impact of family history (FH) on prostate cancer (PCa) development among a general Korean population. We conducted a prospective cohort study based on the registry records of 211,789 participants in the database of the Korean Genome and Epidemiology Study from 2001 to 2013. A total of 69,693 men with appropriate records were evaluated by being categorizing into 2 groups; a PCa group (100) and control group (69,593). FH of PCa was also categorized as FH of total, father, or brother. Odds ratios (ORs) of PCa development were calculated by using stratified logistic regression models. The adjusted OR of PCa history of father was 27.7 (95% confidence interval [CI] = 9.7-79.2, P < .001) in PCa patients compared to control, and that of PCa history of brother was 15.8 (95% CI = 3.6-69.6, P < .001). Among the adjusted variables, age (OR, 1.17; 95% CI, 1.14-1.21; P < .001), and hyperlipidemia (OR, 2.25; CI, 1.32-3.84; P = .003) were also identified as significant predictors of PCa development. There was no difference in the impact of FH on PCa development between different age groups at PCa diagnosis (<60 vs ≥60 years). To our knowledge, this study represents the first prospective cohort study based on the registry data of a Korean population showing the significance of FH on PCa development. Additionally, the effect of FH on the early onset of PCa has not been confirmed in our analysis.
Collapse
Affiliation(s)
- Hyo Geun Choi
- Department of Otorhinolaryngology-Head & Neck Surgery
- Hallym Data Science Laboratory
| | - Woo Jin Bang
- Department of Urology, Hallym University College of Medicine, Anyang
| | - Jung Ki Jo
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Cheol Young Oh
- Department of Urology, Hallym University College of Medicine, Anyang
| | - Myungsun Shim
- Department of Urology, Hallym University College of Medicine, Anyang
| | - Jin Seon Cho
- Department of Urology, Hallym University College of Medicine, Anyang
| |
Collapse
|
15
|
Xu X, Fallah M, Tian Y, Mukama T, Sundquist K, Sundquist J, Brenner H, Kharazmi E. Risk of invasive prostate cancer and prostate cancer death in relatives of patients with prostatic borderline or in situ neoplasia: A nationwide cohort study. Cancer 2020; 126:4371-4378. [PMID: 32697345 DOI: 10.1002/cncr.33096] [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] [Received: 03/16/2020] [Revised: 06/14/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The question of whether having a family history of prostatic borderline or in situ neoplasia (PBISN) is associated with an increased risk of invasive prostate cancer (PCa) or death from PCa remains unanswered. The objective of the current study was to provide an evidence-based risk estimation for the relatives of patients with PBISN. METHODS Nationwide Swedish family cancer data sets were used for the current study, including data regarding all residents of Sweden who were born after 1931 and their parents. Standardized incidence ratios (SIRs), standardized mortality ratios (SMRs), and lifetime cumulative risks of PCa were calculated for men with different constellations of family history. Family history was defined as a dynamic (time-dependent) variable considering changes during follow-up (1958-2015). RESULTS Of the 6,343,727 men in the current study, a total of 238,961 developed invasive PCa and 5756 were diagnosed with PBISN during the follow-up. Men with 1 first-degree relative who was diagnosed with PBISN had a 70% increased risk of invasive PCa (SIR, 1.7; 95% confidence interval, 1.5-1.9) and PCa death (SMR, 1.7; 95% confidence interval, 1.3-2.2) compared with men with no family history of PBISN or invasive PCa. These were rather close to estimates in men with 1 first-degree relative diagnosed with invasive PCa (SIR, 2.1 and SMR, 1.8). A higher risk of PCa in family members was found among patients with a family history of PBISN and/or PCa diagnosed before age 60 years. The results in terms of cumulative risk resembled this trend. CONCLUSIONS A family history of PBISN appears to be as important as a family history of invasive PCa with regard to an increased risk of invasive PCa or PCa mortality. Such a history should not be overlooked in PCa screening recommendations or in future research regarding familial PCa.
Collapse
Affiliation(s)
- Xing Xu
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Mahdi Fallah
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmo, Sweden
| | - Yu Tian
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Trasias Mukama
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmo, Sweden.,Department of Family Medicine and Community Health, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York.,Center for Community-based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Izumo, Japan
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmo, Sweden.,Department of Family Medicine and Community Health, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York.,Center for Community-based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Izumo, Japan
| | - Hermann Brenner
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Elham Kharazmi
- Division of Preventive Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmo, Sweden.,Statistical Genetics Group, Institute of Medical Biometry and Informatics, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
16
|
Oswald N, Lin T, Haaland B, Flynn M, Kawamoto K, Cooney KA, Lowrance W, Hanson HA, O'Neil B. Factors associated with appropriate and low-value PSA testing. Cancer Epidemiol 2020; 66:101724. [PMID: 32361642 DOI: 10.1016/j.canep.2020.101724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/03/2020] [Accepted: 04/11/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Prostate-specific antigen (PSA) testing for early detection of prostate cancer is low-value when it is not indicated by guidelines and the harms outweigh the benefits. In this retrospective cohort study, we identify provider and patient factors associated with PSA testing, particularly in situations where testing would be low-value. METHODS We used electronic health record data from 2011 to 2018 representing 1,738,021 health system encounters in the United States. Using logistic generalized estimating equation models, we examined patient factors (age, comorbid illness, family history, race and prior PSA results), provider factors (gender, specialty, graduation year and medical school rank), and overall time trends associated with PSA testing in low-value and appropriate settings. RESULTS Comorbid illness (odds ratio (OR) 0.0 for 3+ conditions vs none) and no prior PSA testing (OR 0.2) were associated with a lower likelihood of PSA testing in low-value situations, while family history of prostate cancer (OR 1.6) and high prior PSA test results (OR 2.2 for PSA > 6 vs 0-1) were associated with a greater likelihood. Men aged 55-65 years were at greatest risk for PSA testing in low-value situations. The provider factor associated with PSA testing in low-value situations was specialty, with urologists being most likely (OR 2.3 versus advanced practice providers). Internal medicine physicians were more likely to perform PSA testing during low-value situations (OR 1.3 versus advanced practice providers) but much more likely to order a PSA test where appropriate (OR 2.2). All PSA testing decreased since 2011. CONCLUSION We identified several patient and provider factors associated with PSA testing in low-value settings. Some aspects suggest attention to relevant factors for PSA testing in low-value settings (e.g. comorbid illness), while others may encourage PSA testing in low-value settings (e.g. family history). The greatest likelihood of PSA testing in low-value settings is among men within the age range most commonly recommended by guidelines.
Collapse
Affiliation(s)
- Nathaniel Oswald
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Tengda Lin
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Benjamin Haaland
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Michael Flynn
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Kensaku Kawamoto
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Kathleen A Cooney
- Department of Medicine and Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - William Lowrance
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Heidi A Hanson
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | - Brock O'Neil
- Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
17
|
Meissner VH, Strüh JGH, Kron M, Liesenfeld LA, Kranz S, Gschwend JE, Herkommer K. The role of fatal family history and mode of inheritance in prostate cancer for long-term outcomes following radical prostatectomy. World J Urol 2020; 38:3091-3099. [PMID: 32161996 PMCID: PMC7716855 DOI: 10.1007/s00345-020-03147-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/26/2020] [Indexed: 01/07/2023] Open
Abstract
Purpose To determine whether fatal family history (FFH) or mode of inheritance in prostate cancer (PCa) has an impact on long-term outcomes following radical prostatectomy (RP). Methods 1076 PCa patients after RP with at least one deceased first-degree relative with PCa were included and stratified by FFH (four subgroups: fraternal, paternal, multiple, and none) and by mode of inheritance (two subgroups: male to male, non-male to male). We compared clinicopathological characteristics between subgroups with Fisher’s exact or Chi-square tests. Biochemical recurrence-free survival (BRFS) and cancer-specific survival (CSS) were analyzed using the method of Kaplan and Meier. Simple and multiple Cox regression with backward elimination were performed to select prognostic factors for BRFS and CSS. Results Median age at surgery was 63.3 (range 35.9–79.4) years. The overall Kaplan–Meier estimated BRFS rate at 10 and 15 years was 65.6% and 57.0%, respectively. The overall Kaplan–Meier estimated CSS rate at 10 and 15 years was 98.1% and 95.7%, respectively. Neither FFH nor mode of inheritance were factors associated with worse BRFS. However, in multiple Cox regression, paternal FFH was an important prognostic factor for a better CSS (HR 0.19, CI 0.05–0.71, p = 0.014) compared to non-FFH. Conclusion FFH and mode of inheritance do not seem to be prognostic factors of worse long-term outcomes following RP. Rather, a paternal FFH was associated with a better CSS; however, the reasons remain unclear. Nevertheless, patients after RP and FFH could be reassured that their own PCa diagnosis is not associated with a worse long-term outcome.
Collapse
Affiliation(s)
- Valentin H Meissner
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Jamila G H Strüh
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Martina Kron
- Institute of Epidemiology and Medical Biometrics, University of Ulm, Ulm, Germany
| | - Lea A Liesenfeld
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Stephanie Kranz
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Jürgen E Gschwend
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany
| | - Kathleen Herkommer
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Munich, Germany.
| |
Collapse
|
18
|
Nyberg T, Frost D, Barrowdale D, Evans DG, Bancroft E, Adlard J, Ahmed M, Barwell J, Brady AF, Brewer C, Cook J, Davidson R, Donaldson A, Eason J, Gregory H, Henderson A, Izatt L, Kennedy MJ, Miller C, Morrison PJ, Murray A, Ong KR, Porteous M, Pottinger C, Rogers MT, Side L, Snape K, Walker L, Tischkowitz M, Eeles R, Easton DF, Antoniou AC. Prostate Cancer Risks for Male BRCA1 and BRCA2 Mutation Carriers: A Prospective Cohort Study. Eur Urol 2020; 77:24-35. [PMID: 31495749 PMCID: PMC6926480 DOI: 10.1016/j.eururo.2019.08.025] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND BRCA1 and BRCA2 mutations have been associated with prostate cancer (PCa) risk but a wide range of risk estimates have been reported that are based on retrospective studies. OBJECTIVE To estimate relative and absolute PCa risks associated with BRCA1/2 mutations and to assess risk modification by age, family history, and mutation location. DESIGN, SETTING, AND PARTICIPANTS This was a prospective cohort study of male BRCA1 (n = 376) and BRCA2 carriers (n = 447) identified in clinical genetics centres in the UK and Ireland (median follow-up 5.9 and 5.3 yr, respectively). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Standardised incidence/mortality ratios (SIRs/SMRs) relative to population incidences or mortality rates, absolute risks, and hazard ratios (HRs) were estimated using cohort and survival analysis methods. RESULTS AND LIMITATIONS Sixteen BRCA1 and 26 BRCA2 carriers were diagnosed with PCa during follow-up. BRCA2 carriers had an SIR of 4.45 (95% confidence interval [CI] 2.99-6.61) and absolute PCa risk of 27% (95% CI 17-41%) and 60% (95% CI 43-78%) by ages 75 and 85 yr, respectively. For BRCA1 carriers, the overall SIR was 2.35 (95% CI 1.43-3.88); the corresponding SIR at age <65 yr was 3.57 (95% CI 1.68-7.58). However, the BRCA1 SIR varied between 0.74 and 2.83 in sensitivity analyses to assess potential screening effects. PCa risk for BRCA2 carriers increased with family history (HR per affected relative 1.68, 95% CI 0.99-2.85). BRCA2 mutations in the region bounded by positions c.2831 and c.6401 were associated with an SIR of 2.46 (95% CI 1.07-5.64) compared to population incidences, corresponding to lower PCa risk (HR 0.37, 95% CI 0.14-0.96) than for mutations outside the region. BRCA2 carriers had a stronger association with Gleason score ≥7 (SIR 5.07, 95% CI 3.20-8.02) than Gleason score ≤6 PCa (SIR 3.03, 95% CI 1.24-7.44), and a higher risk of death from PCa (SMR 3.85, 95% CI 1.44-10.3). Limitations include potential screening effects for these known mutation carriers; however, the BRCA2 results were robust to multiple sensitivity analyses. CONCLUSIONS The results substantiate PCa risk patterns indicated by retrospective analyses for BRCA2 carriers, including further evidence of association with aggressive PCa, and give some support for a weaker association in BRCA1 carriers. PATIENT SUMMARY In this study we followed unaffected men known to carry mutations in the BRCA1 and BRCA2 genes to investigate whether they are at higher risk of developing prostate cancer compared to the general population. We found that carriers of BRCA2 mutations have a high risk of developing prostate cancer, particularly more aggressive prostate cancer, and that this risk varies by family history of prostate cancer and the location of the mutation within the gene.
Collapse
Affiliation(s)
- Tommy Nyberg
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Daniel Barrowdale
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - D Gareth Evans
- Manchester Regional Genetics Service, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Elizabeth Bancroft
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Julian Adlard
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Munaza Ahmed
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Trust, London, UK
| | - Julian Barwell
- Leicestershire Clinical Genetics Service, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Angela F Brady
- North West Thames Regional Genetics Service, London North West University Healthcare NHS Trust, London, UK
| | - Carole Brewer
- Peninsula Clinical Genetics Service, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Jackie Cook
- North Trent Clinical Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, UK
| | - Rosemarie Davidson
- West of Scotland Regional Genetics Service, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Alan Donaldson
- South Western Regional Genetics Service, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Jacqueline Eason
- Nottingham Centre for Medical Genetics, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Helen Gregory
- North of Scotland Regional Genetics Service, NHS Grampian, Aberdeen, UK
| | - Alex Henderson
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - Louise Izatt
- South East Thames Regional Genetics Service, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - M John Kennedy
- St. James's Hospital, Dublin, Ireland; National Centre for Medical Genetics, Dublin, Ireland
| | - Claire Miller
- Merseyside and Cheshire Clinical Genetics Service, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Patrick J Morrison
- Northern Ireland Regional Genetics Service, Belfast Health and Social Care Trust, Belfast, UK
| | - Alex Murray
- Medical Genetics Services for Wales, Abertawe Bro Morgannwg University Health Board, Swansea, UK
| | - Kai-Ren Ong
- West Midlands Regional Genetics Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Mary Porteous
- South East of Scotland Regional Genetics Service, NHS Lothian, Edinburgh, UK
| | - Caroline Pottinger
- Medical Genetics Services for Wales, Betsi Cadwaladr University Health Board, Bodelwyddan, UK
| | - Mark T Rogers
- All Wales Medical Genetics Service, NHS Wales, Cardiff, UK
| | - Lucy Side
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Katie Snape
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Lisa Walker
- Oxford Regional Genetics Service, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, University of Cambridge, Cambridge, UK; East Anglian Regional Genetics Service, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Rosalind Eeles
- Oncogenetics Team, Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK; Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Meissner VH, Bittner R, Kron M, Schiele S, Schulwitz H, Gschwend JE, Herkommer K. Impact of a Changing Population Structure and Clustering of Cancer in Prostate Cancer Patients Depending on a First-Degree Family History. Urol Int 2019; 104:222-229. [PMID: 31865349 DOI: 10.1159/000504789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/13/2019] [Indexed: 02/03/2023]
Abstract
INTRODUCTION In the last century, there have been major changes within the population structure in Germany. The aim of this study was to determine the impact of a changing population structure on identification of familial prostate cancer (PCa), and to investigate how many and which types of other cancers have occurred in patients and their first-degree relatives. MATERIALS AND METHODS A total of 19,540 patients were evaluated in a prospectively collected PCa family database and divided into four birth cohorts: 1925-1934 (cohort A), 1935-1944 (cohort B), 1945-1954 (cohort C), and 1955-1964 (cohort D). Other primary cancers and cancers of first-degree relatives were evaluated. RESULTS The percentage of PCa patients with ≥2 sons declined (A: 28.9% to D: 21.6%). The percentage of patients whose fathers lived for ≥65 years increased (B: 64.2% to D: 73.0%). Malignancies of the skin, the urinary tract, and the lymphoid/hematopoietic tissue were more common in patients with a positive first-degree PCa family history and their first-degree relatives. Additionally, first-degree relatives reported more often neoplasms of respiratory/intrathoracic organs and the female breast. CONCLUSIONS A small family size, an early deceased father, and a high number of sporadic cases complicate the identification of familial PCa patients. Thus, a detailed family history should also include unaffected first-degree relatives to avoid any misclassification. Findings of other primary cancers in patients and their relatives warrant further investigation.
Collapse
Affiliation(s)
- Valentin Henri Meissner
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Robert Bittner
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martina Kron
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Stefan Schiele
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helga Schulwitz
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jürgen Erich Gschwend
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kathleen Herkommer
- Department of Urology, Faculty of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany,
| |
Collapse
|
20
|
Defining low-value PSA testing in a large retrospective cohort: Finding common ground between discordant guidelines. Cancer Epidemiol 2018; 56:112-117. [PMID: 30130683 DOI: 10.1016/j.canep.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/06/2018] [Accepted: 08/10/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Reports of low-value prostate-specific antigen (PSA) testing (testing in which the harms outweigh the benefits) generally employ population level data sources. While such results may be generalizable, they often lack the detail necessary to understand provider clinical decision making and guideline concordance. Using a retrospective study of PSA testing at our institution we intend to characterize the frequency and patterns associated with low-value PSA testing. METHODS We leveraged the electronic health record to determine guideline-defined low-value testing in our health system from 07/01/2012 to 06/30/2017. Secondarily, we measured the between-testing interval for repeat tests and the rates of prostate cancer risk factors and comorbidities among men receiving screening. RESULTS Overall, 21,145 PSA tests were performed on 12,303 men. The rate of low-value testing ranged from 23.4 to 56.8%, depending upon the specific guideline. For repeat tests, the median between-testing interval was 12.6 months. Risk factors for prostate cancer were uncommon, but more frequent in men age <55 years compared to men age 55-69 years (17.6% vs. 13.5%, p < 0.001). Screened older men (age >70 years) were more likely to have a Charlson Comorbidity Index ≥ 3, compared to the 55-69 reference group (31.4% vs. 17.3%, p < 0.001). CONCLUSION Low-value prostate cancer testing is prevalent. Between-testing intervals were often times shorter than recommended. Screening among younger men was frequent despite low rates of risk factors. High rates of comorbidity may limit life expectancy among older men receiving screening. These findings highlight the need for improved guidance with prostate cancer screening.
Collapse
|
21
|
Helfand BT, Chen H, Fantus RJ, Conran CA, Brendler CB, Zheng SL, Walsh PC, Isaacs WB, Xu J. Differences in inherited risk among relatives of hereditary prostate cancer patients using genetic risk score. Prostate 2018; 78:1063-1068. [PMID: 29923209 PMCID: PMC6773522 DOI: 10.1002/pros.23664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/29/2018] [Indexed: 11/12/2022]
Abstract
PURPOSE Family history assigns equivalent risk to all relatives based upon the degree of relationship. Recent genetic studies have identified single nucleotide polymorphisms (SNPs) that can be used to calculate a genetic risk score (GRS) to determine prostate cancer (PCa) risk. We sought to determine whether GRS can stratify PCa risk among individuals in families considered to be at higher risk due their family history of PCa. MATERIALS AND METHODS Family members with hereditary PCa were recruited and genotyped for 17 SNPs associated with PCa. A GRS was calculated for all subjects. Analyses compared the distribution of GRS values among affected and unaffected family members of varying relationship degrees. RESULTS Data was available for 789 family members of probands including 552 affected and 237 unaffected relatives. Median GRSs were higher among first-degree relatives compared to second- and third-degree relatives. In addition, GRS values among affected first- and second-degree relatives were significantly higher than unaffected relatives (P = 0.042 and P = 0.016, respectively). Multivariate analysis including GRS and degree of relationship demonstrated that GRS was a significant and independent predictor of PCa (OR 1.52, 95%CI 1.15-2.01). CONCLUSION GRS is an easy-to-interpret, objective measure that can be used to assess differences in PCa risk among family members of affected men. GRS allows for further differentiation among family members, providing better risk assessment. While prospective validation studies are required, this information can help guide relatives in regards to the time of initiation and frequency of PCa screening.
Collapse
Affiliation(s)
- Brian T Helfand
- Division of Urology, John and Carol Walter for Urologic Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Haitao Chen
- School of Public Health, Fudan University, Center for Genomic Translational Medicine and Prevention, Shanghai, P.R. China
| | - Richard J Fantus
- Department of Surgery, Section of Urology, University of Chicago Medical Center, University of Chicago, Chicago, Illinois
| | - Carly A Conran
- Division of Urology, John and Carol Walter for Urologic Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Charles B Brendler
- Division of Urology, John and Carol Walter for Urologic Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Siquan Lilly Zheng
- Division of Urology, John and Carol Walter for Urologic Health, NorthShore University HealthSystem, Evanston, Illinois
| | - Patrick C Walsh
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins, Baltimore, Maryland
| | - William B Isaacs
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins, Baltimore, Maryland
| | - Jianfeng Xu
- Division of Urology, John and Carol Walter for Urologic Health, NorthShore University HealthSystem, Evanston, Illinois
| |
Collapse
|
22
|
Valberg M, Stensrud MJ, Aalen OO. The surprising implications of familial association in disease risk. BMC Public Health 2018; 18:135. [PMID: 29334951 PMCID: PMC5769446 DOI: 10.1186/s12889-018-5033-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A wide range of diseases show some degree of clustering in families; family history is therefore an important aspect for clinicians when making risk predictions. Familial aggregation is often quantified in terms of a familial relative risk (FRR), and although at first glance this measure may seem simple and intuitive as an average risk prediction, its implications are not straightforward. METHODS We use two statistical models for the distribution of disease risk in a population: a dichotomous risk model that gives an intuitive understanding of the implication of a given FRR, and a continuous risk model that facilitates a more detailed computation of the inequalities in disease risk. Published estimates of FRRs are used to produce Lorenz curves and Gini indices that quantifies the inequalities in risk for a range of diseases. RESULTS We demonstrate that even a moderate familial association in disease risk implies a very large difference in risk between individuals in the population. We give examples of diseases for which this is likely to be true, and we further demonstrate the relationship between the point estimates of FRRs and the distribution of risk in the population. CONCLUSIONS The variation in risk for several severe diseases may be larger than the variation in income in many countries. The implications of familial risk estimates should be recognized by epidemiologists and clinicians.
Collapse
Affiliation(s)
- Morten Valberg
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, POB. 1122, Blindern, Oslo, N-0317 Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Mats Julius Stensrud
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, POB. 1122, Blindern, Oslo, N-0317 Norway
| | - Odd O. Aalen
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, POB. 1122, Blindern, Oslo, N-0317 Norway
| |
Collapse
|
23
|
Turner AR, Lane BR, Rogers D, Lipkus I, Weaver K, Danhauer SC, Zhang Z, Hsu FC, Noyes SL, Adams T, Toriello H, Monroe T, McKanna T, Young T, Rodarmer R, Kahnoski RJ, Tourojman M, Kader AK, Zheng SL, Baer W, Xu J. Randomized trial finds that prostate cancer genetic risk score feedback targets prostate-specific antigen screening among at-risk men. Cancer 2016; 122:3564-3575. [PMID: 27433786 PMCID: PMC5247411 DOI: 10.1002/cncr.30162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/16/2016] [Accepted: 04/21/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND Prostate-specific antigen (PSA) screening may reduce death due to prostate cancer but leads to the overdiagnosis of many cases of indolent cancer. Targeted use of PSA screening may reduce overdiagnosis. Multimarker genomic testing shows promise for risk assessment and could be used to target PSA screening. METHODS To test whether counseling based on the family history (FH) and counseling based on a genetic risk score (GRS) plus FH would differentially affect subsequent PSA screening at 3 months (primary outcome), a randomized trial of FH versus GRS plus FH was conducted with 700 whites aged 40 to 49 years without prior PSA screening. Secondary outcomes included anxiety, recall, physician discussion at 3 months, and PSA screening at 3 years. Pictographs versus numeric presentations of genetic risk were also evaluated. RESULTS At 3 months, no significant differences were observed in the rates of PSA screening between the FH arm (2.1%) and the GRS-FH arm (4.5% with GRS-FH vs. 2.1% with FH: χ2 = 3.13, P = .077); however, PSA screening rates at 3 months significantly increased with given risk in the GRS-FH arm (P = .013). Similar results were observed for discussions with physicians at 3 months and PSA screening at 3 years. Average anxiety levels decreased after the individual cancer risk was provided (P = .0007), with no differences between groups. Visual presentation by pictographs did not significantly alter comprehension or anxiety. CONCLUSIONS This is likely the first randomized trial of multimarker genomic testing to report genomic targeting of cancer screening. This study found little evidence of concern about excess anxiety or overuse/underuse of PSA screening when multimarker genetic risks were provided to patients. Cancer 2016;122:3564-3575. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Aubrey R. Turner
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian R. Lane
- Spectrum Health Hospital System, Grand Rapids, MI, 49546
- Michigan State University College of Human Medicine, Grand Rapids, MI 49546
| | - Dan Rogers
- Van Andel Research Institute, Grand Rapids, MI
| | | | - Kathryn Weaver
- Department of Social Sciences & Health Policy, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Suzanne C. Danhauer
- Department of Social Sciences & Health Policy, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Zheng Zhang
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Fang-Chi Hsu
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | | | - Tamara Adams
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Helga Toriello
- Spectrum Health Hospital System, Grand Rapids, MI, 49546
| | - Thomas Monroe
- Spectrum Health Hospital System, Grand Rapids, MI, 49546
| | - Trudy McKanna
- Spectrum Health Hospital System, Grand Rapids, MI, 49546
| | - Tracey Young
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Ryan Rodarmer
- Spectrum Health Hospital System, Grand Rapids, MI, 49546
| | | | | | - A. Karim Kader
- Department of Surgery, University of California San Diego, San Diego, CA
| | - S. Lilly Zheng
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - William Baer
- Grand Valley Medical Specialists, Grand Rapids, MI
| | - Jianfeng Xu
- Center for Cancer Genomics, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
24
|
Cremers RG, Aben KK, van Oort IM, Sedelaar JPM, Vasen HF, Vermeulen SH, Kiemeney LA. The clinical phenotype of hereditary versus sporadic prostate cancer: HPC definition revisited. Prostate 2016; 76:897-904. [PMID: 26989049 PMCID: PMC5069637 DOI: 10.1002/pros.23179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/25/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND The definition of hereditary prostate cancer (HPC) is based on family history and age at onset. Intuitively, HPC is a serious subtype of prostate cancer but there are only limited data on the clinical phenotype of HPC. Here, we aimed to compare the prognosis of HPC to the sporadic form of prostate cancer (SPC). METHODS HPC patients were identified through a national registry of HPC families in the Netherlands, selecting patients diagnosed from the year 2000 onward (n = 324). SPC patients were identified from the Netherlands Cancer Registry (NCR) between 2003 and 2006 for a population-based study into the genetic susceptibility of PC (n = 1,664). Detailed clinical data were collected by NCR-registrars, using a standardized registration form. Follow-up extended up to the end of 2013. Differences between the groups were evaluated by cross-tabulations and tested for statistical significance while accounting for familial dependency of observations by GEE. Differences in progression-free and overall survival were evaluated using χ(2) testing with GEE in a proportional-hazards model. RESULTS HPC patients were on average 3 years younger at diagnosis, had lower PSA values, lower Gleason scores, and more often locally confined disease. Of the HPC patients, 35% had high-risk disease (NICE-criteria) versus 51% of the SPC patients. HPC patients were less often treated with active surveillance. Kaplan-Meier 5-year progression-free survival after radical prostatectomy was comparable for HPC (78%) and SPC (74%; P = 0.30). The 5-year overall survival was 85% (95%CI 81-89%) for HPC versus 80% (95%CI 78-82%) for SPC (P = 0.03). CONCLUSIONS HPC has a favorable clinical phenotype but patients more often underwent radical treatment. The major limitation of HPC is the absence of a genetics-based definition of HPC, which may lead to over-diagnosis of PC in men with a family history of prostate cancer. The HPC definition should, therefore, be re-evaluated, aiming at a reduction of over-diagnosis and overtreatment among men with multiple relatives diagnosed with PC. Prostate 76:897-904, 2016. © 2016 The Authors. The Prostate published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ruben G Cremers
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- The Netherlands Foundation for the Detection of Hereditary Tumours, Leiden, The Netherlands
| | - Katja K Aben
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Inge M van Oort
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - J P Michiel Sedelaar
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Hans F Vasen
- The Netherlands Foundation for the Detection of Hereditary Tumours, Leiden, The Netherlands
| | - Sita H Vermeulen
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Lambertus A Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Bayne CE, Jarrett TW. Cancer of the Prostate: Incidence in the USA. Prostate Cancer 2016. [DOI: 10.1016/b978-0-12-800077-9.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
26
|
Nicolas E, Arora S, Zhou Y, Serebriiskii IG, Andrake MD, Handorf ED, Bodian DL, Vockley JG, Dunbrack RL, Ross EA, Egleston BL, Hall MJ, Golemis EA, Giri VN, Daly MB. Systematic evaluation of underlying defects in DNA repair as an approach to case-only assessment of familial prostate cancer. Oncotarget 2015; 6:39614-33. [PMID: 26485759 PMCID: PMC4741850 DOI: 10.18632/oncotarget.5554] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/02/2015] [Indexed: 01/03/2023] Open
Abstract
Risk assessment for prostate cancer is challenging due to its genetic heterogeneity. In this study, our goal was to develop an operational framework to select and evaluate gene variants that may contribute to familial prostate cancer risk. Drawing on orthogonal sources, we developed a candidate list of genes relevant to prostate cancer, then analyzed germline exomes from 12 case-only prostate cancer patients from high-risk families to identify patterns of protein-damaging gene variants. We described an average of 5 potentially disruptive variants in each individual and annotated them in the context of public databases representing human variation. Novel damaging variants were found in several genes of relevance to prostate cancer. Almost all patients had variants associated with defects in DNA damage response. Many also had variants linked to androgen signaling. Treatment of primary T-lymphocytes from these prostate cancer patients versus controls with DNA damaging agents showed elevated levels of the DNA double strand break (DSB) marker γH2AX (p < 0.05), supporting the idea of an underlying defect in DNA repair. This work suggests the value of focusing on underlying defects in DNA damage in familial prostate cancer risk assessment and demonstrates an operational framework for exome sequencing in case-only prostate cancer genetic evaluation.
Collapse
Affiliation(s)
| | - Sanjeevani Arora
- Programs in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yan Zhou
- Programs in Biostatistics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ilya G. Serebriiskii
- Programs in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA, USA
- Kazan Federal University, Kazan, Russia
| | - Mark D. Andrake
- Programs in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Dale L. Bodian
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA, USA
| | - Joseph G. Vockley
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA, USA
| | - Roland L. Dunbrack
- Programs in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eric A. Ross
- Programs in Biostatistics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brian L. Egleston
- Programs in Biostatistics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michael J. Hall
- Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Erica A. Golemis
- Programs in Molecular Therapeutics Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Veda N. Giri
- Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA, USA
| | - Mary B. Daly
- Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
27
|
[Familial prostate cancer research: meaningfulness of hereditary criteria in view of demographic change]. Urologe A 2015; 53:1779-85. [PMID: 25214313 DOI: 10.1007/s00120-014-3617-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND In view of demographic changes and previous research results, it is debatable whether hereditary criteria should be further applied when it comes to determining the status of prostate carcinoma (PC) patients. MATERIALS AND METHODS A total of 30,455 patients in the national research program "Familial Prostate Carcinoma" were divided into four groups based on birth year: I: 1920-1929 (n = 3.968), II: 1930-1939 (n = 14.110), III: 1940-1949 (n = 10.137), IV: 1950-1959 (n = 2.169). In all, 9.316 PC patients with a positive family history were analyzed with respect to their relatives (first degree). RESULTS The average number of siblings (group I: 3.3; IV: 2.2), brothers (I: 2.1; IV: 1.3), children (I: 2.2; IV: 1.7), and sons (I: 1.1; IV: 0.9) have decreased. The number of patients without a brother/son (I: 9.6/32.8%; IV: 27.6/37.5%) has increased. In the same period of time, the number of patients with two brothers/sons (I: 34.5/6.3%; IV: 14.8/2.2 %) has decreased. The number of fathers > 65 years has risen from 66.8% (I) to 71.1% (IV). CONCLUSION Hereditary criteria are less and less suited in order to determine the status of PC patients. However, positive family history remains one of the most crucial risk factors of prostate carcinoma and, therefore, detailed data ascertainment should be urged.
Collapse
|
28
|
Marcus PM, Freedman AN, Khoury MJ. Targeted Cancer Screening in Average-Risk Individuals. Am J Prev Med 2015; 49:765-771. [PMID: 26165196 PMCID: PMC4615467 DOI: 10.1016/j.amepre.2015.04.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/13/2015] [Accepted: 04/29/2015] [Indexed: 11/30/2022]
Abstract
Targeted cancer screening refers to use of disease risk information to identify those most likely to benefit from screening. Researchers have begun to explore the possibility of refining screening regimens for average-risk individuals using genetic and non-genetic risk factors and previous screening experience. Average-risk individuals are those not known to be at substantially elevated risk, including those without known inherited predisposition, without comorbidities known to increase cancer risk, and without previous diagnosis of cancer or pre-cancer. In this paper, we describe the goals of targeted cancer screening in average-risk individuals, present factors on which cancer screening has been targeted, discuss inclusion of targeting in screening guidelines issued by major U.S. professional organizations, and present evidence to support or question such inclusion. Screening guidelines for average-risk individuals currently target age; smoking (lung cancer only); and, in some instances, race; family history of cancer; and previous negative screening history (cervical cancer only). No guidelines include common genomic polymorphisms. RCTs suggest that targeting certain ages and smoking histories reduces disease-specific cancer mortality, although some guidelines extend ages and smoking histories based on statistical modeling. Guidelines that are based on modestly elevated disease risk typically have either no or little evidence of an ability to affect a mortality benefit. In time, targeted cancer screening is likely to include genetic factors and past screening experience as well as non-genetic factors other than age, smoking, and race, but it is of utmost importance that clinical implementation be evidence-based.
Collapse
Affiliation(s)
- Pamela M Marcus
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland.
| | - Andrew N Freedman
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| | - Muin J Khoury
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
29
|
Randazzo M, Müller A, Carlsson S, Eberli D, Huber A, Grobholz R, Manka L, Mortezavi A, Sulser T, Recker F, Kwiatkowski M. A positive family history as a risk factor for prostate cancer in a population-based study with organised prostate-specific antigen screening: results of the Swiss European Randomised Study of Screening for Prostate Cancer (ERSPC, Aarau). BJU Int 2015; 117:576-83. [PMID: 26332304 DOI: 10.1111/bju.13310] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To assess the value of a positive family history (FH) as a risk factor for prostate cancer incidence and grade among men undergoing organised prostate-specific antigen (PSA) screening in a population-based study. SUBJECTS AND METHODS The study cohort comprised all attendees of the Swiss arm of the European Randomised Study of Screening for Prostate Cancer (ERSPC) with systematic PSA level tests every 4 years. Men reporting first-degree relative(s) diagnosed with prostate cancer were considered to have a positive FH. Biopsy was exclusively PSA triggered at a PSA level threshold of 3 ng/mL. The primary endpoint was prostate cancer diagnosis. Kaplan-Meier and Cox regression analyses were used. RESULTS Of 4 932 attendees with a median (interquartile range, IQR) age of 60.9 (57.6-65.1) years, 334 (6.8%) reported a positive FH. The median (IQR) follow-up duration was 11.6 (10.3-13.3) years. Cumulative prostate cancer incidence was 60/334 (18%, positive FH) and 550/4 598 (12%, negative FH) [odds ratio 1.6, 95% confidence interval (CI) 1.2-2.2, P = 0.001). In both groups, most prostate cancer diagnosed was low grade. There were no significant differences in PSA level at diagnosis, biopsy Gleason score or Gleason score on pathological specimen among men who underwent radical prostatectomy between both groups. On multivariable analysis, age (hazard ratio [HR] 1.04, 95% CI 1.02-1.06), baseline PSA level (HR 1.13, 95% CI 1.12-1.14), and FH (HR 1.6, 95% CI 1.24-2.14) were independent predictors for overall prostate cancer incidence (all P < 0.001). Only baseline PSA level (HR 1.14, 95% CI 1.12-1.16, P < 0.001) was an independent predictor of Gleason score ≥7 prostate cancer on prostate biopsy. The proportion of interval prostate cancer diagnosed in-between the screening rounds was not significantly different. CONCLUSION Irrespective of the FH status, the current PSA-based screening setting detects the majority of aggressive prostate cancers and missed only a minority of interval cancers with a 4-year screening algorithm. Our results suggest that men with a positive FH are at increased risk of low-grade but not aggressive prostate cancer.
Collapse
Affiliation(s)
- Marco Randazzo
- Department of Urology, University Hospital Zürich, Zürich, Switzerland.,Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Alexander Müller
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Sigrid Carlsson
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Urology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Daniel Eberli
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Andreas Huber
- Department of Laboratory Medicine, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Rainer Grobholz
- Department of Pathology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Lukas Manka
- Department of Urology, Academic Hospital Braunschweig, Braunschweig, Germany
| | - Ashkan Mortezavi
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Franz Recker
- Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Maciej Kwiatkowski
- Department of Urology, Cantonal Hospital Aarau, Aarau, Switzerland.,Department of Urology, Academic Hospital Braunschweig, Braunschweig, Germany
| |
Collapse
|
30
|
Hicks EM, Litwin MS, Maliski SL. Latino men and familial risk communication about prostate cancer. Oncol Nurs Forum 2015; 41:509-16. [PMID: 25158656 DOI: 10.1188/14.onf.509-516] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE/OBJECTIVES To investigate how familial communication about prostate cancer (PCa) risk and screening affects sons of men with PCa. RESEARCH APPROACH Qualitative grounded theory. SETTING Southern California. PARTICIPANTS 17 Latino sons of PCa survivors. METHODOLOGIC APPROACH The team conducted semistructured interviews and follow-up interviews. Therefore, the sample includes 25 transcripts. Data were analyzed with a mix of a priori topical codes and grounded theory techniques. FINDINGS Sons were in need of information about familial risk and screening options. They became sensitized to PCa, desired information, and held protective intentions. Hopeful intentions came up against cultural taboos around sex, reproductive health, and intimacy that limited discussions between fathers and sons. Fathers were a valued source of information but play various roles, which affect sons' screening intentions. Open communication between father and son promoted awareness of screening and familial risk. CONCLUSIONS Uncertainty about familial risk and screening options, especially early detection strategies, was exacerbated by cultural taboos around PCa. Fathers could have been primary and credible advocates for shared decision making, but sons had difficulty learning from their fathers' experience. INTERPRETATION FINDINGS from the study can help inform community-based interventions with Latino families, help to culturally tailor health messaging, and sensitize clinicians to a group that needs concerted counseling about PCa risk and screening.
Collapse
Affiliation(s)
| | - Mark S Litwin
- Department of Urology, University of California, Los Angeles
| | | |
Collapse
|
31
|
Albright F, Stephenson RA, Agarwal N, Teerlink CC, Lowrance WT, Farnham JM, Albright LAC. Prostate cancer risk prediction based on complete prostate cancer family history. Prostate 2015; 75:390-8. [PMID: 25408531 PMCID: PMC4293302 DOI: 10.1002/pros.22925] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/26/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostate cancer (PC) relative risks (RRs) are typically estimated based on status of close relatives or presence of any affected relatives. This study provides RR estimates using extensive and specific PC family history. METHODS A retrospective population-based study was undertaken to estimate RRs for PC based on complete family history of PC. A total of 635,443 males, all with ancestral genealogy data, were analyzed. RRs for PC were determined based upon PC rates estimated from males with no PC family history (without PC in first, second, or third degree relatives). RRs were determined for a variety of constellations, for example, number of first through third degree relatives; named (grandfather, father, uncle, cousins, brothers); maternal, paternal relationships, and age of onset. RESULTS In the 635,443 males analyzed, 18,105 had PC. First-degree RRs ranged from 2.46 (=1 first-degree relative affected, CI = 2.39-2.53) to 7.65 (=4 first-degree relatives affected, CI = 6.28-9.23). Second-degree RRs for probands with 0 affected first-degree relatives ranged from 1.51 (≥1 second-degree relative affected, CI = 1.47-1.56) to 3.09 (≥5 second-degree relatives affected, CI = 2.32-4.03). Third-degree RRs with 0 affected first- and 0 affected second-degree relatives ranged from 1.15 (≥1 affected third-degree relative, CI = 1.12-1.19) to 1.50 (≥5 affected third-degree relatives, CI = 1.35-1.66). RRs based on age at diagnosis were higher for earlier age at diagnoses; for example, RR = 5.54 for ≥1 first-degree relative diagnosed before age 50 years (CI = 1.12-1.19) and RR = 1.78 for >1 second-degree relative diagnosed before age 50 years, CI = 1.33, 2.33. RRs for equivalent maternal versus paternal family history were not significantly different. CONCLUSIONS A more complete PC family history using close and distant relatives and age at diagnosis results in a wider range of estimates of individual RR that are potentially more accurate than RRs estimated from summary family history. The presence of PC in second- and even third-degree relatives contributes significantly to risk. Maternal family history is just as significant as paternal family history. PC RRs based on a proband's complete constellation of affected relatives will allow patients and care providers to make more informed screening, monitoring, and treatment decisions.
Collapse
Affiliation(s)
- Frederick Albright
- Department of Pharmacotherapy, Pharmacotherapy Outcomes Research Center, College of Pharmacy, University of UtahSalt Lake City, Utah
| | - Robert A Stephenson
- Department of Surgery, Division of Urology, School of Medicine, University of UtahSalt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical CenterSalt Lake City, Utah
- Huntsman Cancer Institute, University of UtahSalt Lake City, Utah
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of UtahSalt Lake City, Utah
- Department of Medicine, Division of Medical Oncology, University of UtahSalt Lake City, Utah
| | - Craig C Teerlink
- Department of Internal Medicine, Division of Genetic Epidemiology, University of Utah School of MedicineSalt Lake City, Utah
| | - William T Lowrance
- Department of Surgery, Division of Urology, School of Medicine, University of UtahSalt Lake City, Utah
- George E. Wahlen Department of Veterans Affairs Medical CenterSalt Lake City, Utah
- Huntsman Cancer Institute, University of UtahSalt Lake City, Utah
| | - James M Farnham
- Department of Internal Medicine, Division of Genetic Epidemiology, University of Utah School of MedicineSalt Lake City, Utah
| | - Lisa A Cannon Albright
- George E. Wahlen Department of Veterans Affairs Medical CenterSalt Lake City, Utah
- Huntsman Cancer Institute, University of UtahSalt Lake City, Utah
- Department of Internal Medicine, Division of Genetic Epidemiology, University of Utah School of MedicineSalt Lake City, Utah
| |
Collapse
|
32
|
Salinas CA, Tsodikov A, Ishak-Howard M, Cooney KA. Prostate cancer in young men: an important clinical entity. Nat Rev Urol 2014; 11:317-23. [PMID: 24818853 PMCID: PMC4191828 DOI: 10.1038/nrurol.2014.91] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Prostate cancer is considered a disease of older men (aged >65 years), but today over 10% of new diagnoses in the USA occur in young men aged ≤55 years. Early-onset prostate cancer, that is prostate cancer diagnosed at age ≤55 years, differs from prostate cancer diagnosed at an older age in several ways. Firstly, among men with high-grade and advanced-stage prostate cancer, those diagnosed at a young age have a higher cause-specific mortality than men diagnosed at an older age, except those over age 80 years. This finding suggests that important biological differences exist between early-onset prostate cancer and late-onset disease. Secondly, early-onset prostate cancer has a strong genetic component, which indicates that young men with prostate cancer could benefit from evaluation of genetic risk. Furthermore, although the majority of men with early-onset prostate cancer are diagnosed with low-risk disease, the extended life expectancy of these patients exposes them to long-term effects of treatment-related morbidities and to long-term risk of disease progression leading to death from prostate cancer. For these reasons, patients with early-onset prostate cancer pose unique challenges, as well as opportunities, for both research and clinical communities. Current data suggest that early-onset prostate cancer is a distinct phenotype-from both an aetiological and clinical perspective-that deserves further attention.
Collapse
Affiliation(s)
- Claudia A. Salinas
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Alex Tsodikov
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Miriam Ishak-Howard
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kathleen A. Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Urology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
33
|
Influence of family history on psychosocial distress and perceived need for treatment in prostate cancer survivors. Fam Cancer 2014; 13:481-8. [DOI: 10.1007/s10689-014-9715-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Bratt O. The clinical impact of genetic susceptibility to prostate cancer. Eur Urol 2014; 66:500-1. [PMID: 24702982 DOI: 10.1016/j.eururo.2014.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Indexed: 10/25/2022]
Affiliation(s)
- Ola Bratt
- Department of Clinical Sciences, Malmö, Lund University, Helsingborg, Sweden; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Vertosick EA, Poon BY, Vickers AJ. Relative value of race, family history and prostate specific antigen as indications for early initiation of prostate cancer screening. J Urol 2014; 192:724-8. [PMID: 24641912 DOI: 10.1016/j.juro.2014.03.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2014] [Indexed: 11/15/2022]
Abstract
PURPOSE Many guidelines suggest earlier screening for prostate cancer in men at high risk, with risk defined in terms of race and family history. Recent evidence suggests that baseline prostate specific antigen is strongly predictive of the long-term risk of aggressive prostate cancer. We compared the usefulness of risk stratifying early screening by race, family history and prostate specific antigen at age 45 years. MATERIALS AND METHODS Using estimates from the literature we calculated the proportion of men targeted for early screening using family history, black race or prostate specific antigen as the criterion for high risk. We calculated the proportion of prostate cancer deaths that would occur in those men by age 75 years. RESULTS Screening based on family history involved 10% of men, accounting for 14% of prostate cancer deaths. Using black race as a risk criterion involved 13% of men, accounting for 28% of deaths. In contrast, 44% of prostate cancer deaths occurred in the 10% of men with the highest prostate specific antigen at age 45 years. In no sensitivity analysis for race and family history did the ratio of risk group size to number of prostate cancer deaths in that risk group approach that of prostate specific antigen. CONCLUSIONS Basing decisions for early screening on prostate specific antigen at age 45 years provided the best ratio between men screened and potential cancer deaths avoided. Given the lack of evidence that race or family history affects the relationship between prostate specific antigen and risk, prostate specific antigen based risk stratification would likely include any black men or men with a family history who are destined to experience aggressive disease. Differential screening based on risk should be informed by baseline prostate specific antigen.
Collapse
Affiliation(s)
- Emily A Vertosick
- Health Outcomes Research Group, Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Bing Ying Poon
- Health Outcomes Research Group, Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andrew J Vickers
- Health Outcomes Research Group, Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
36
|
Abstract
The purpose of this review is to identify clinical risk factors for prostate cancer and to assess the utility and limitations of our current tools for prostate cancer screening. Prostate-specific antigen is the single most important factor for identifying men at increased risk of prostate cancer but is best assessed in the context of other clinical factors; increasing age, race, and family history are well-established risk factors for the diagnosis of prostate cancer. In addition to clinical risk calculators, novel tools such as multiparametric imaging, serum or urinary biomarkers, and genetic profiling show promise in improving prostate cancer diagnosis and characterization. Optimal use of existing and future tools will help alleviate the problems of overdiagnosis and overtreatment of low-risk prostate cancer without reversing the substantial mortality declines that have been achieved in the screening era.
Collapse
|
37
|
Early Detection, PSA Screening, and Management of Overdiagnosis. Hematol Oncol Clin North Am 2013; 27:1091-110, vii. [DOI: 10.1016/j.hoc.2013.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Birmingham WC, Agarwal N, Kohlmann W, Aspinwall LG, Wang M, Bishoff J, Dechet C, Kinney AY. Patient and provider attitudes toward genomic testing for prostate cancer susceptibility: a mixed method study. BMC Health Serv Res 2013; 13:279. [PMID: 23870420 PMCID: PMC3750463 DOI: 10.1186/1472-6963-13-279] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 06/27/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The strong association between family history and prostate cancer (PCa) suggests a significant genetic contribution, yet specific highly penetrant PCa susceptibility genes have not been identified. Certain single-nucleotide-polymorphisms have been found to correlate with PCa risk; however uncertainty remains regarding their clinical utility and how to best incorporate this information into clinical decision-making. Genetic testing is available directly to consumers and both patients and healthcare providers are becoming more aware of this technology. Purchasing online allows patients to bypass their healthcare provider yet patients may have difficulty interpreting test results and providers may be called upon to interpret results. Determining optimal ways to educate both patients and providers, and strategies for appropriately incorporating this information into clinical decision-making are needed. METHODS A mixed-method study was conducted in Utah between October 2011 and December 2011. Eleven focus group discussions were held and surveys were administered to 23 first-degree relatives of PCa patients living in Utah and 24 primary-care physicians and urologists practicing in Utah to present specific information about these assessments and determine knowledge and attitudes regarding health implications of using these assessments. RESULTS Data was independently coded by two researchers (relative Kappa = .88; provider Kappa = .77) and analyzed using a grounded theory approach. Results indicated differences in attitudes and behavioral intentions between patient and provider. Despite the test's limitations relatives indicated interest in genetic testing (52%) while most providers indicated they would not recommend the test for their patients (79%). Relatives expected providers to interpret genetic test results and use results to provide personalized healthcare recommendations while the majority of providers did not think the information would be useful in patient care (92%) and indicated low-levels of genetic self-efficacy. CONCLUSIONS Although similarities exist, discordance between provider and patient attitudes may influence the effective translation of novel genomic tests into clinical practice suggesting both patient and provider perceptions and expectations be considered in development of clinical decision-support tools.
Collapse
Affiliation(s)
- Wendy C Birmingham
- Department of Psychology, Brigham Young University, 1054 SWKT, Provo, UT 84602, USA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
- Department of Internal Medicine, University of Utah, 30 North 1900 East, Room 4C104, Salt Lake City, UT 84132, USA
| | - Wendy Kohlmann
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Lisa G Aspinwall
- Department of Psychology, University of Utah, 380 South 1530 East, BEHS 502, Salt Lake City, UT 84112, USA
| | - Mary Wang
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Jay Bishoff
- Intermountain Health Care, 5169 Cottonwood St Ste 420, Murray, UT 84107, USA
| | - Christopher Dechet
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
- Department of Urology, University of Utah, 30 North 1900 East, Salt Lake City, 84132 UT, USA
| | - Anita Y Kinney
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
- Department of Internal Medicine, University of Utah, 30 North 1900 East, Room 4C104, Salt Lake City, UT 84132, USA
| |
Collapse
|
39
|
Abstract
This chapter addresses issues relevant to prostate cancer overdiagnosis. Factors promoting the overdiagnosis of prostate cancer are reviewed. First is the existence of a relatively large, silent reservoir of this disease, as can be seen by evaluating autopsy studies and histologic step-sectioning results of prostates removed for other causes. The second main factor responsible for prostate cancer overdiagnosis is fairly widespread prostate-specific antigen and digital rectal examination-based screening, which has been fairly widely practiced in the United States for the past 20 years among heterogeneous groups of men. This has resulted in the identification of many men from this reservoir who otherwise may never have been diagnosed with symptomatic prostate cancer and is substantially responsible for the current annual incidence to mortality ratio for prostate cancer of approximately 6 to 1. Finally, the relatively indolent natural history and limited cancer-specific mortality as reported in a variety of contemporary randomized screening and treatment trials is reviewed. We attempt to quantitate the proportion of newly diagnosed prostate cancers that are overdiagnosed using various trial results and models. We explore the impact of prostate cancer overdiagnosis in terms of patient anxiety and the potential for overtreatment, with its attendant morbidity. We explore strategies to minimize overdiagnosis by targeting screening and biopsy only to men at high risk for aggressive prostate cancer and by considering the use of agents such as 5-alpha reductase inhibitors. Future prospects to prevent overtreatment, including better biopsy and molecular characterization of newly diagnosed cancer and the role of active surveillance, are discussed.
Collapse
Affiliation(s)
- Gurdarshan S Sandhu
- Division of Urologic Surgery, Washington University School of Medicine, 4960 Children's Place, St. Louis, MO 63110, USA
| | | |
Collapse
|
40
|
Lavery HJ, Droller MJ. Do Gleason patterns 3 and 4 prostate cancer represent separate disease states? J Urol 2012; 188:1667-75. [PMID: 22998919 DOI: 10.1016/j.juro.2012.07.055] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Indexed: 11/26/2022]
Abstract
PURPOSE The Gleason scoring system has been the traditional basis for studies on the assessment and treatment of prostate cancer. Recent reports of long-term prostate cancer outcomes stratified by Gleason score based on the 2005 ISUP (International Society of Urological Pathology) update suggest that important aspects of the biology of prostate cancer correlate with commonly available histopathological information. In this review we present a conceptual framework for the possible existence of distinct but interrelated developmental pathways in the context of the Gleason score in considering various biological and clinical aspects of prostate cancer. This may be useful in characterizing prostate cancer as an indolent condition in some and an aggressive disease in others, in decision making for treatment, and in the interpretation of the biological course and treatment outcomes. MATERIALS AND METHODS A comprehensive review of clinical, pathological and investigational biological literature on this topic was conducted. In addition, the biological behavior of prostate cancer as interpreted from this survey was compared to that of other solid neoplasms in developing a schema for characterizing the pathogenesis of various forms of the disease. RESULTS The Gleason scoring system has been found to have fundamental value in predicting the behavior of prostate cancer and assessing outcomes of its treatment. Increasingly, the proportion of Gleason pattern 4 in a prostatectomy specimen is being recognized as a critical factor in predicting the rates of biochemical recurrence and prostate cancer specific mortality. Under the current Gleason classification, a Gleason 3 + 3 = 6 cancer carries a minimal long-term risk of progression or mortality. Risk of biochemical recurrence and prostate cancer specific mortality increases with increasing proportions of the Gleason 4 component in the prostatectomy specimen, from 3 + 3 = 6 with tertiary 4 (ie less than 5% of a 4 component) to 3 + 4 = 7, 4 + 3 = 7 and 4 + 4 = 8. Assuming that the Gleason 4 component increases in volume more rapidly with time than well differentiated components, it can be inferred that a smaller proportion of Gleason 4 could mean that the cancer has been identified at an earlier phase in the natural history of the disease. This could explain the improved prognosis on the basis of length and lead time biases, and conceivably on the basis of a decreased likelihood of cancer cells having metastasized. Correspondingly, increasing amounts of Gleason 4 cancer in a prostate specimen might be explained in 2 ways, as the preferential growth of a single clone of Gleason 4 cells, possibly with intraprostatic spread, or the evolution of Gleason 3 cancer cells to become Gleason 4. These hypotheses have been examined by genetic analysis of metastatic deposits and by comparisons of multiple foci of cancer within individual prostates. The clinical significance of these concepts in regard to disease status at diagnosis, treatment selection, outcomes of treatment, and implications for future research on the basis of clinical and molecular observations are the basis of the developmental schemata we propose. CONCLUSIONS Given the relatively benign nature of homogeneous, low volume Gleason 3 tumors, and the progressive risk of biochemical recurrence and prostate cancer specific mortality with increasing quantities of Gleason 4 components, we propose that Gleason 4 (and 5) cancers constitute cancer diatheses distinct from that of Gleason 3 cancer. This distinction may contribute to the understanding of the prognosis intrinsic to these biological behavioral patterns, and help guide the translation of findings at molecular and histological levels to a more precise selection of treatments.
Collapse
Affiliation(s)
- Hugh J Lavery
- Department of Urology, Mount Sinai Medical Center, New York, New York 10022, USA.
| | | |
Collapse
|
41
|
Abstract
One hundred years ago, decades before the discovery of the structure of DNA, debate raged regarding how human traits were passed from one generation to the next. Phenotypes, including risk of disease, had long been recognized as having a familial component. Yet it was difficult to reconcile genetic segregation as described by Mendel with observations exhaustively documented by Karl Pearson and others regarding the normal distribution of human characteristics. In 1918, R. A. Fisher published his landmark article, "The Correlation Between Relatives on the Supposition of Mendelian Inheritance," bridging this divide and demonstrating that multiple alleles, all individually obeying Mendel's laws, account for the phenotypic variation observed in nature.Since that time, geneticists have sought to identify the link between genotype and phenotype. Trait-associated alleles vary in their frequency and degree of penetrance. Some minor alleles may approach a frequency of 50% in the human population, whereas others are present within only a few individuals. The spectrum for penetrance is similarly wide. These characteristics jointly determine the segregation pattern of a given trait, which, in turn, determine the method used to map the trait. Until recently, identification of rare, highly penetrant alleles was most practical. Revolutionary studies in genomics reported over the past decade have made interrogation of most of the spectrum of genetic variation feasible.The following article reviews recent discoveries in the genetic basis of inherited cancer risk and how these discoveries inform cancer biology and patient management. Although this article focuses on prostate cancer, the principles are generic for any cancer and, indeed, for any trait.
Collapse
|
42
|
Cremers R, van Asperen C, Kil P, Vasen H, Wiersma T, van Oort I, Kiemeney L. Urologists' and GPs' knowledge of hereditary prostate cancer is suboptimal for prostate cancer counseling: a nation-wide survey in The Netherlands. Fam Cancer 2012; 11:195-200. [PMID: 22160565 PMCID: PMC3365231 DOI: 10.1007/s10689-011-9500-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A family history of prostate cancer (PCa) is an established risk factor for PCa. In case of a positive family history, the balance between positive and adverse effects of prostate-specific antigen (PSA) testing might be different from the general population, for which the European Randomized Study of Screening for Prostate Cancer (ERSPC) showed a beneficial effect on mortality. This, however, went at the cost of considerable overtreatment. This study assessed Dutch physicians' knowledge of heredity and PCa and their 'post-ERSPC' attitude towards PCa testing, including consideration of family history. In January 2010, all Dutch urologists and clinical geneticists (CGs) and 300 general practitioners (GPs) were invited by email to complete an anonymous online survey, which contained questions about hereditary PCa and their attitudes towards PCa case-finding and screening. 109 urologists (31%), 69 GPs (23%) and 46 CGs (31%) completed the survey. CGs had the most accurate knowledge of hereditary PCa. All but 1 CG mentioned at least one inherited trait with PCa, compared to only 25% of urologists and 9% of GPs. CGs hardly ever counseled men about PCa testing. Most urologists and GPs discuss possible risks and benefits before testing for PCa with PSA. Remarkably, 35-40% of them do not take family history into consideration. Knowledge of urologists and GPs about heredity and PCa is suboptimal. Hence, PCa counseling might not be optimal for men with a positive family history. Multidisciplinary guidelines on this topic should be developed to optimize personalized counseling.
Collapse
Affiliation(s)
- Ruben Cremers
- Department of Epidemiology, Biostatistics and HTA (133), Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, The Netherlands
| | - Christi van Asperen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul Kil
- Department of Urology, St. Elisabeth Hospital, Tilburg, The Netherlands
| | - Hans Vasen
- Netherlands Foundation for the Detection of Hereditary Tumors, Leiden, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tjerk Wiersma
- Dutch College of General Practitioners, Utrecht, The Netherlands
| | - Inge van Oort
- Department of Urology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Lambertus Kiemeney
- Department of Epidemiology, Biostatistics and HTA (133), Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
- Department of Urology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
- Comprehensive Cancer Centre The Netherlands, Nijmegen, The Netherlands
| |
Collapse
|
43
|
Prostate cancer epidemiology in the United States. World J Urol 2012; 30:195-200. [PMID: 22476558 DOI: 10.1007/s00345-012-0824-2] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/04/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Prostate cancer is a significant public health issue in the United States. It is the most commonly diagnosed non-skin cancer and the second leading cause of cancer death. The American Cancer Society estimates that in 2011, 240,890 men were diagnosed with prostate cancer and 33,720 men died of it. METHODS A review of the peer-reviewed literature was conducted: American Cancer Society, National Cancer Institute Surveillance, Epidemiology and End Results. Program data were assessed to describe trends in incidence, mortality, and survival rates and look at other predictors of risk of prostate cancer diagnosis and death. RESULTS Since 1985, there have been significant changing trends in prostate cancer incidence, mortality, and survival rates, as well as changes in the age distribution of the population diagnosed and even in the distribution of pathologies at diagnosis. Major risk factors for diagnosis include age, family history, race, and screening behavior. CONCLUSION While prostate cancer remains largely a disease diagnosed in older men (over age 65), screening has increased risk of diagnosis among men in their 40s and 50s. The incidence rates and 5-year survival rates are heavily influenced by the introduction of serum prostate-specific antigen (PSA) and widespread screening. The effects of PSA usage and screening on mortality rates are less certain. Outcome studies among men treated with radical prostatectomy show that greater than 30% relapse rates are common. This suggests that many men who are diagnosed with "localized early stage disease" actually have "apparently localized early stage disease," which is really low volume metastatic disease.
Collapse
|
44
|
Yassa M, Hijal T, Giraud P. Prostate cancer and androgenic alopecia. Expert Rev Endocrinol Metab 2012; 7:169-173. [PMID: 30764008 DOI: 10.1586/eem.11.97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prostate cancer is a burden on society. Its prevalence can reach up to 80% in males aged 70 years and older. Current screening programs based on prostate-specific antigen testing lead to overdiagnosis and overtreatment with uncertain benefits on survival. Androgenic alopecia is also highly prevalent in elderly males. Observational studies have found that androgenic alopecia is linked to prostate cancer, but studies have been conflictual. Further research should focus on finding the exact mechanism linking these two pathologies. This should help clinicians improve screening programs and guide research into novel molecules to help in the prevention and treatment of both androgenic alopecia and prostate cancer.
Collapse
Affiliation(s)
- Michael Yassa
- a Department of Radiation Oncology, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, Canada.
| | - Tarek Hijal
- b Department of Radiation Oncology, McGill University Hospital Center, Montreal, Canada
| | - Philippe Giraud
- c Department of Radiation Oncology, European Georges Pompidou Hospital, Paris Descartes University, Paris, France
| |
Collapse
|
45
|
Jansson KF, Akre O, Garmo H, Bill-Axelson A, Adolfsson J, Stattin P, Bratt O. Concordance of tumor differentiation among brothers with prostate cancer. Eur Urol 2012; 62:656-61. [PMID: 22386193 DOI: 10.1016/j.eururo.2012.02.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/14/2012] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genetic factors seem to be of greater importance in prostate cancer than in other forms of cancer. Studies have suggested familial concordance in survival, but the extent to which that is due to tumor characteristics is not known. OBJECTIVE We hypothesized that a brother of an index case with prostate cancer is at particularly increased risk of prostate cancer with the same tumor differentiation as the index case. DESIGN, SETTING AND PARTICIPANTS We identified 21,930 brothers of index cases with prostate cancer in the Prostate Cancer Data Base Sweden and followed them up for incidence of prostate cancer. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The relative risk of Gleason score-specific prostate cancer in the cohort of brothers was estimated by using the standardized incidence ratio (SIR) stratified by Gleason score of the index case. We estimated 95% confidence intervals (CIs) assuming a Poisson distribution. RESULTS AND LIMITATIONS Among brothers of index cases with Gleason score 8-10 cancer, the SIR was 2.53 (95% CI, 1.97-3.21) for a Gleason score 2-6 cancer and 4.00 (95% CI, 2.63-5.82) for a Gleason score 8-10 cancer. SIR for Gleason score 2-6 cancer among brothers decreased with time since the date of the index cases' diagnoses, whereas the risk of Gleason 8-10 cancer increased over time for brothers of index cases with Gleason 8-10 cancer (p for trend = 0.009). CONCLUSIONS Brothers of men with high-grade prostate cancer are at particularly increased risk of high-grade prostate cancer. Likewise, there is a concordance of less malignant prostate cancers within families. These findings may have direct clinical relevance for counseling men with a family history of prostate cancer.
Collapse
Affiliation(s)
- K Fredrik Jansson
- Urology Unit, Department of Surgery and Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
46
|
Brandt A, Sundquist J, Hemminki K. Risk for incident and fatal prostate cancer in men with a family history of any incident and fatal cancer. Ann Oncol 2012; 23:251-256. [PMID: 21467126 DOI: 10.1093/annonc/mdr056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Familial clustering of incident prostate cancer and some cancers at other discordant sites has been reported. Less is known about familial clustering of fatal prostate cancer with any fatal discordant cancers. Estimates on familial aggregation based on mortality are free from bias of overdiagnosis. PATIENTS AND METHODS We used the nationwide Swedish Family-Cancer Database to calculate standardized incidence ratios (SIRs) for incident prostate cancer for relatives of patients with any common cancer and standardized mortality ratios (SMRs) for death in prostate cancer for relatives of individuals who died from cancer. Similar risks were determined for any common cancer when relatives were affected by prostate cancer. RESULTS We observed familial aggregation of incident and fatal prostate cancers. Familial clustering (SIRs increased) of prostate cancer and of cancers at discordant sites was found for breast, ovarian, and kidney cancers and melanoma. Also, fatal prostate cancer clustered with these and cervical cancers (SMRs increased). CONCLUSIONS Our findings demonstrate that familial aggregation of prostate and breast cancers are not due to shared screening habits. The data on the association of cancers at discordant sites might be useful for clinical counseling and for mechanistic studies searching explanations to the familial clustering between discordant cancers.
Collapse
Affiliation(s)
- A Brandt
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - J Sundquist
- Center for Primary Care Research, Lund University, Malmö, Sweden; Stanford Prevention Research Center, Stanford University School of Medicine, Palo Alto, USA
| | - K Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany; Center for Primary Care Research, Lund University, Malmö, Sweden.
| |
Collapse
|
47
|
Familial risk and familial survival in prostate cancer. World J Urol 2011; 30:143-8. [DOI: 10.1007/s00345-011-0801-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 11/12/2011] [Indexed: 02/07/2023] Open
|
48
|
Zhu X, Albertsen PC, Andriole GL, Roobol MJ, Schröder FH, Vickers AJ. Risk-based prostate cancer screening. Eur Urol 2011; 61:652-61. [PMID: 22134009 DOI: 10.1016/j.eururo.2011.11.029] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 11/15/2011] [Indexed: 11/30/2022]
Abstract
CONTEXT Widespread mass screening of prostate cancer (PCa) is not recommended because the balance between benefits and harms is still not well established. The achieved mortality reduction comes with considerable harm such as unnecessary biopsies, overdiagnoses, and overtreatment. Therefore, patient stratification with regard to PCa risk and aggressiveness is necessary to identify those men who are at risk and may actually benefit from early detection. OBJECTIVE This review critically examines the current evidence regarding risk-based PCa screening. EVIDENCE ACQUISITION A search of the literature was performed using the Medline database. Further studies were selected based on manual searches of reference lists and review articles. EVIDENCE SYNTHESIS Prostate-specific antigen (PSA) has been shown to be the single most significant predictive factor for identifying men at increased risk of developing PCa. Especially in men with no additional risk factors, PSA alone provides an appropriate marker up to 30 yr into the future. After assessment of an early PSA test, the screening frequency may be determined based on individualized risk. A limited list of additional factors such as age, comorbidity, prostate volume, family history, ethnicity, and previous biopsy status have been identified to modify risk and are important for consideration in routine practice. In men with a known PSA, risk calculators may hold the promise of identifying those who are at increased risk of having PCa and are therefore candidates for biopsy. CONCLUSIONS PSA testing may serve as the foundation for a more risk-based assessment. However, the decision to undergo early PSA testing should be a shared one between the patient and his physician based on information balancing its advantages and disadvantages.
Collapse
Affiliation(s)
- Xiaoye Zhu
- Department of Urology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
49
|
Vickers AJ, Lilja H. Predicting prostate cancer many years before diagnosis: how and why? World J Urol 2011; 30:131-5. [PMID: 22101902 DOI: 10.1007/s00345-011-0795-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 11/01/2011] [Indexed: 11/26/2022] Open
Abstract
Evidence of reduced prostate cancer mortality from randomized trials in Europe supports early detection of prostate cancer with prostate-specific antigen (PSA). Yet PSA screening has generated considerable controversy: it is far from clear that the benefits outweigh risks, in terms of overdiagnosis and overtreatment. One way to shift the ratio of benefits to harm is to focus on men at highest risk, who have more to benefit than average. Neither family history nor any of the currently identified genomic markers offer sufficient risk stratification for practical use. However, there is considerable evidence that the levels of PSA in blood are strongly prognostic of the long-term risk of aggressive prostate cancer. Specifically, it is difficult to justify continuing to screen men aged 60 or older if they have a PSA less than 1 or 2 ng/ml; for men 45-60, intervals between PSA tests can be based on PSA levels, with 2-4-year retesting interval for men with PSA of 1 ng/ml or higher, and tests every 6-8 years for men with PSA <1 ng/ml. Men with the top 10% of PSAs at a young age (PSA ~1.5 ng/ml or higher below 50) are at particularly high risk and should be subject to intensive monitoring.
Collapse
Affiliation(s)
- Andrew J Vickers
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | |
Collapse
|
50
|
Kiciński M, Vangronsveld J, Nawrot TS. An epidemiological reappraisal of the familial aggregation of prostate cancer: a meta-analysis. PLoS One 2011; 6:e27130. [PMID: 22073129 PMCID: PMC3205054 DOI: 10.1371/journal.pone.0027130] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 10/11/2011] [Indexed: 11/19/2022] Open
Abstract
Studies on familial aggregation of cancer may suggest an overall contribution of inherited genes or a shared environment in the development of malignant disease. We performed a meta-analysis on familial clustering of prostate cancer. Out of 74 studies reporting data on familial aggregation of prostate cancer in unselected populations retrieved by a Pubmed search and browsing references, 33 independent studies meeting the inclusion criteria were used in the analysis performed with the random effects model. The pooled rate ratio (RR) for first-degree family history, i.e. affected father or brother, is 2.48 (95% confidence interval: 2.25-2.74). The incidence rate for men who have a brother who got prostate cancer increases 3.14 times (CI:2.37-4.15), and for those with affected father 2.35 times (CI:2.02-2.72). The pooled estimate of RR for two or more affected first-degree family members relative to no history in father and in brother is 4.39 (CI:2.61-7.39). First-degree family history appears to increase the incidence rate of prostate cancer more in men under 65 (RR:2.87, CI:2.21-3.74), than in men aged 65 and older (RR:1.92, CI:1.49-2.47), p for interaction = 0.002. The attributable fraction among those having an affected first-degree relative equals to 59.7% (CI:55.6-63.5%) for men at all ages, 65.2% (CI:57.7-71.4%) for men younger than 65 and 47.9% (CI:37.1-56.8%) for men aged 65 or older. For those with a family history in 2 or more first-degree family members 77.2% (CI:65.4-85.0%) of prostate cancer incidence can be attributed to the familial clustering. Our combined estimates show strong familial clustering and a significant effect-modification by age meaning that familial aggregation was associated with earlier disease onset (before age 65).
Collapse
Affiliation(s)
- Michał Kiciński
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium.
| | | | | |
Collapse
|