1
|
Cirulli GO, Davis M, Stephens A, Chiarelli G, Finati M, Chase M, Tinsley S, Arora S, Sood A, Lughezzani G, Buffi N, Carrieri G, Salonia A, Briganti A, Montorsi F, Rogers C, Abdollah F. Midlife baseline prostate-specific antigen, velocity, and doubling time association with lethal prostate cancer and mortality. Cancer 2024. [PMID: 39377255 DOI: 10.1002/cncr.35563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Midlife baseline prostate-specific antigen (MB PSA), defined as a single PSA value measured between 40-59 years of age, has been proposed as a tool that can limit potential harms of PSA screening. This study aimed to examine the ability of MB PSA versus PSA doubling time (PSADT) and PSA velocity (PSAV) in assessing the likelihood of developing of lethal prostate cancer (PCa) in a diverse and contemporary North American population. METHODS Men 40-59 years old, who received their first PSA between the years 1995 and 2019, were included. For MB PSA values, the first PSA test result was included. For PSADT, the first two PSA test results were included. For PSAV, the first three PSA test results within 30 months were included. Selection criteria resulted in a total of 77,594 patients with at least two PSA test results and 11,634 patients with at least three PSA test results. Multivariable Fine-Gray regression was used to examine the impact of the value of the PSA testing methods on the development of lethal PCa (defined as death from PCa or development of metastatic disease either at diagnosis or during follow-up). Time-dependent receiver operating characteristic/area under the curve (AUC) at 5, 10, and 15 years were plotted. RESULTS In the main cohort, patients were most frequently in the 50-54 age category (32.8%), had a Charlson comorbidity index of 0 (70.5%), and were White (63.2%). Of these, 9.3% had the midlife baseline PSA in the top 10th percentile, and 0.4% had a PSADT 0-6 months. Lethal PCa was diagnosed in 593 (0.8%) patients. The median (interquartile range) time to lethal PCa was 8.6 (3.2-14.9) years. In the main cohort, MB PSA and PSADT showed significant associations with the occurrence of lethal PCa, with a hazard ratio (HR) of 6.10 (95% confidence interval [CI], 4.85-7.68) and HR of 2.20 (95% CI, 1.07-4.54) for patients in the top 10th percentile MB PSA group and in the PSADT between 0 to <6 months group, respectively. In patients with three PSA results available, MB PSA and PSAV showed significant associations with the occurrence of lethal PCa, with a HR of 3.95 (95% CI, 2.29-6.79) and 3.57 (95% CI, 2.17-5.86) for patients in the top 10th percentile MB PSA group and in the in the PSAV >0.4 ng/mL/year group, respectively. PSADT and PSAV did not exhibit higher AUCs than MB PSA in assessing the likelihood of lethal PCa. Specifically, they were 0.818 and 0.708 at 10 and 15 years, respectively, for the PSADT; 0.862 and 0.756 at 10 and 15 years, respectively, for the PSAV; and 0.868 and 0.762 at 10 and 15 years, respectively, for the MB PSA (all p > .05). CONCLUSIONS The study findings are that PSAV or PSADT were not superior to midlife baseline in assessing the likelihood of developing lethal PCa. This suggests that these variables may not have practical use in enhancing PSA screening strategies in a clinical setting.
Collapse
Affiliation(s)
- Giuseppe Ottone Cirulli
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Matthew Davis
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| | - Alex Stephens
- Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - Giuseppe Chiarelli
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
- Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Marco Finati
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
- Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| | - Morrison Chase
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| | - Shane Tinsley
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| | - Sohrab Arora
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| | - Akshay Sood
- Department of Urology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, US
| | - Giovanni Lughezzani
- Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Nicolo Buffi
- Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| | - Andrea Salonia
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Briganti
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Craig Rogers
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| | - Firas Abdollah
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
2
|
Jehle DVK, Nguyen N, Garza MA, Kim DK, Paul KK, Bilby NJ, Bogache WK, Chevli KK. PSA Levels and Mortality in Prostate Cancer Patients. Clin Genitourin Cancer 2024; 22:102162. [PMID: 39094287 DOI: 10.1016/j.clgc.2024.102162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Prostate cancer (PC) is the second most common cancer among men around the world. Several smaller studies have explored the relationship between elevated PSA and mortality, but results have been conflicting. Additionally, studies have shown that Black men are more likely to be diagnosed with PC at late-stages and may have a twofold increase in mortality risk. This study aims to evaluate the relationship between PSA levels and mortality in patients with PC and differences between Black versus White patients. METHODS In this retrospective study, the TriNetX database, was used to extract de-identified EMRs of 198,083 patients. Patients were included if they were diagnosed with PC and had obtained a PSA level (measured in ng/mL) within 6 months prior to diagnosis. Cohorts were separated into 7 groups based on intervals of PSA, ranging from < 2 to ≥ 500 and compared to a control cohort with a PSA of 4 to 20 for differing 2-year mortality rates. A subgroup analysis was performed to compare mortality differences between Black and White patients. A posthoc analysis evaluated 5- and 10-year mortality amongst all patients with PC. RESULTS After propensity matching, mortality risk was significantly lower for patients with PSA < 2 (5.9% vs. 7.5%; RR 0.784; P < .001) when compared to the control cohort. Mortality was significantly higher for all other subsequent PSA intervals > 20, with the lowest risk ratios at PSA 20-100 (24.1% vs. 10.0%; RR 2.419; P < .001) and highest at PSA 200 to 500 (50.4% vs. 10.8%; RR 4.673; P < .001). The sub-group analysis showed that when compared to White patients, Black patients with PSA < 20 had similar mortalities, but had significantly lower 2-year mortality rates at PSA levels ≥ 20. The posthoc analysis of PSA levels and 5- and 10-year mortality of all patients with PC showed similar trends to the 2-year outcomes. CONCLUSION This study found that prostate cancer patients with significantly elevated PSA levels have a greater mortality, and Black patients have lower 2-year mortality rates than their White counterparts when matched for PSA levels greater than 20.
Collapse
Affiliation(s)
| | - Nam Nguyen
- Department of Emergency Medicine, University of Texas Medical Branch, Galveston, TX
| | - Michael A Garza
- Department of Emergency Medicine, University of Texas Medical Branch, Galveston, TX
| | - Debora K Kim
- Department of Emergency Medicine, University of Texas Medical Branch, Galveston, TX
| | - Krishna K Paul
- Department of Emergency Medicine, University of Texas Medical Branch, Galveston, TX
| | - Nathaniel J Bilby
- Department of Emergency Medicine, University of Texas Medical Branch, Galveston, TX
| | - William K Bogache
- Department of Urology, Grand Strand Regional Medical Center, Myrtle Beach, SC
| | - K Kent Chevli
- Department of Urology, University of Buffalo, Buffalo, NY
| |
Collapse
|
3
|
Bratt O. Prostate Cancer Screening: Setting the Controls on the Mixing Board. Eur Urol 2024:S0302-2838(24)02598-3. [PMID: 39294046 DOI: 10.1016/j.eururo.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024]
Affiliation(s)
- Ola Bratt
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Urology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
4
|
Albers P, Becker N. [Prostate cancer screening? Only evidence-based, risk-adjusted, and organized!]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2024; 67:1047-1053. [PMID: 39102055 PMCID: PMC11349832 DOI: 10.1007/s00103-024-03916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 06/10/2024] [Indexed: 08/06/2024]
Abstract
In view of a recent recommendation of the European Commission to conceptualize novel screening approaches for lung, gastric, and prostate cancer, Germany is also invoked to revise its prostate early detection program. This discussion article provides an overview of new findings on prostate cancer screening, which suggest an organized and risk-adapted screening approach. Based on the German risk-adapted screening trial PROBASE, together with recently published data on organized screening programs in Europe, model projects should be established to determine the specific modalities for a new organized and risk-adapted prostate cancer screening program.
Collapse
Affiliation(s)
- Peter Albers
- Abteilung für Personalisierte Früherkennung des Prostatakarzinoms, Deutsches Krebsforschungszentrum (DKFZ) Heidelberg, C130, Im Neuenheimer Feld 581, 69120, Heidelberg, Deutschland.
- Klinik für Urologie, Universitätsklinikum Düsseldorf, Medizinische Fakultät, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Deutschland.
| | - Nikolaus Becker
- Abteilung für Personalisierte Früherkennung des Prostatakarzinoms, Deutsches Krebsforschungszentrum (DKFZ) Heidelberg, C130, Im Neuenheimer Feld 581, 69120, Heidelberg, Deutschland
| |
Collapse
|
5
|
Finati M, Davis M, Stephens A, Chiarelli G, Cirulli GO, Morrison C, Affas R, Sood A, Buffi N, Lughezzani G, Briganti A, Montorsi F, Carrieri G, Rogers C, Vickers AJ, Abdollah F. The Role of Baseline Prostate-specific Antigen Value Prior to Age 60 in Predicting Lethal Prostate Cancer: Analysis of a Contemporary North American Cohort. Eur Urol Oncol 2024:S2588-9311(24)00172-X. [PMID: 38991891 DOI: 10.1016/j.euo.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Studies evaluating the role of baseline midlife prostate-specific antigen (PSA) as a predictor of development and progression of prostate cancer relied predominately on cohorts from the pre-PSA screening introduction era. The aim of our study was to examine the role of baseline PSA prior to the age of 60 yr as a predictor of developing lethal prostate cancer using a contemporary North American cohort. METHODS Our cohort included all men aged 40-59 yr who received their first PSA through our health system between the years 1995 and 2019. Patients were divided into four categories based on age: 40-44, 45-49, 50-54, and 55-59 yr. Baseline PSA was the predictor of interest. Lethal disease was defined as death from prostate cancer or development of metastatic disease either at diagnosis or during follow-up. Cancer-specific mortality and overall mortality were obtained by linking our database to the Michigan Vital Records registry. Competing-risk regression was used to evaluate the association between PSA and lethal prostate cancer. KEY FINDINGS AND LIMITATIONS A total of 129067 men met the inclusion criteria during the study period. The median follow-up for patients free from cancer was 7.4 yr. For men aged 40-44, 45-49, 50-54, and 55-59 yr, the estimated rates of lethal prostate cancer at 20 yr were 0.02%, 0.14%, 0.33%, and 0.51% in men with PSA CONCLUSIONS AND CLINICAL IMPLICATIONS Baseline PSA is a very strong predictor of the subsequent risk of developing lethal prostate cancer in a large contemporary diverse North American cohort, which was exposed to opportunistic PSA screening. The association was far larger than that found for polygenic risk scores, confirming that baseline PSA prior to the age of 60 yr is the most effective tool for adjusting subsequent screening. Compared with studies of unscreened cohorts, there was a smaller difference in discrimination between incident and lethal disease, reflecting the influence of screening. PATIENT SUMMARY In this study, we found that a single baseline prostate-specific antigen (PSA) value is strongly predictive of the subsequent risk of developing metastatic prostate cancer, as well as the risk of dying from prostate cancer. The initial PSA level can therefore be used to adjust the frequency of subsequent PSA testing.
Collapse
Affiliation(s)
- Marco Finati
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA; Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| | - Matthew Davis
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA
| | - Alex Stephens
- Public Health Sciences, Henry Ford Health, Detroit, MI, USA
| | - Giuseppe Chiarelli
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA; Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Giuseppe Ottone Cirulli
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA; Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Chase Morrison
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA
| | - Rafe Affas
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA
| | - Akshay Sood
- Department of Urology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nicolò Buffi
- Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Giovanni Lughezzani
- Department of Urology, IRCCS Humanitas Research Hospital, Humanitas University, Milan, Italy
| | - Alberto Briganti
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Division of Oncology, Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, University of Foggia, Foggia, Italy
| | - Craig Rogers
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA; Henry Ford Health, Detroit, MI, USA
| | - Andrew Julian Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Firas Abdollah
- VUI Center for Outcomes Research, Analysis, and Evaluation, Henry Ford Health, Detroit, MI, USA; Henry Ford Health, Detroit, MI, USA.
| |
Collapse
|
6
|
Bjerner J, Bratt O, Aas K, Albertsen PC, Fosså SD, Kvåle R, Lilja H, Müller C, Müller S, Stensvold A, Thomas O, Røe OD, Vickers A, Walz J, Carlsson SV, Oldenburg J. Baseline Serum Prostate-specific Antigen Value Predicts the Risk of Subsequent Prostate Cancer Death-Results from the Norwegian Prostate Cancer Consortium. Eur Urol 2024; 86:20-26. [PMID: 37169639 PMCID: PMC10840440 DOI: 10.1016/j.eururo.2023.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/27/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Prostate-specific antigen (PSA) levels in midlife are strongly associated with the long-term risk of lethal prostate cancer in cohorts not subject to screening. This is the first study evaluating the association between PSA levels drawn as part of routine medical care in the Norwegian population and prostate cancer incidence and mortality. The objective of the study was to determine the association between midlife PSA levels <4.0 ng/ml, drawn aspart of routine medical care, and long-term risk of prostate cancer death. METHODS The Norwegian Prostate Cancer Consortium collected >8 million PSA results from >1 million Norwegian males (more than or equal to) 40 yr of age. We studied 176 099 men (predefined age strata: 40-54 and 55-69 yr) without a prior prostate cancer diagnosis who had a nonelevated baseline PSA level (<4.0 ng/ml) between January 1,1995 and December 31, 2005. We assessed the 16-yr risk of prostate cancer mortality. We calculated the discrimination (C-index) between predefined PSA strata (<0.5, 0.5-0.9, 1.0-1.9, 2.0-2.9, and 3.0-3.9 ng/ml) and subsequent prostate cancer death. Survival curves were plotted using the Kaplan-Meier method. KEY FINDINGS AND LIMITATIONS The median follow-up time of men who did not get prostate cancer was 17.9 yr. Overall, 84% of men had a baseline PSA level of <2.0 ng/ml and 1346 men died from prostate cancer, with 712 deaths (53%) occurring in the 16% of men with the highest baseline PSA of 2.0-3.9 ng/ml. Baseline PSA levels were associated with prostate cancer mortality (C-index 0.72 for both age groups, 40-54 and 55-69 yr). The fact that the reason for any given PSA measurement remains unknown represents a limitation. CONCLUSIONS AND CLINICAL IMPLICATIONS We replicated prior studies that baseline PSA at age 40-69 yr can be used to stratify a man's risk of dying from prostate cancer within the next 15-20 yr. PATIENT SUMMARY A prostate-specific antigen level obtained as part of routine medical care is strongly associated with a man's risk of dying from prostate cancer in the next two decades.
Collapse
Affiliation(s)
- Johan Bjerner
- Fürst Laboratories, Department of Clinical Chemistry, Oslo, Norway
| | - Ola Bratt
- Department of Urology, Institute of Clinical Sciences, Gothenburg University, Gothenburg, Sweden; Department of Urology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kirsti Aas
- Department of Urology, Oslo University Hospital, Oslo, Norway
| | - Peter C Albertsen
- Division of Urology, Department of Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | - Sophie D Fosså
- National Advisory Unit for Late Effects After Cancer Treatment, Oslo University Hospital, Oslo, Norway; Medical Faculty of University of Oslo, Oslo, Norway
| | - Rune Kvåle
- Department of Health Registry Research and Development, Norwegian Institute of Public Health, Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Hans Lilja
- Departments of Laboratory Medicine, Surgery and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Stig Müller
- Department of Urology, Akershus University Hospital, Lørenskog, Norway
| | | | - Owen Thomas
- Department of Biostatistics, Akershus University Hospital, Lørenskog, Norway
| | - Oluf D Røe
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, Levanger Hospital, Nord-Trøndelag Health Trust, Levanger, Norway
| | - Andrew Vickers
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jochen Walz
- Department of Urology, Institut Paoli-Calmettes Cancer Centre, Marseille, France
| | - Sigrid V Carlsson
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jan Oldenburg
- Medical Faculty of University of Oslo, Oslo, Norway; Department of Oncology, Akershus University Hospital, Lørenskog, Norway.
| |
Collapse
|
7
|
van Harten MJ, Roobol MJ, van Leeuwen PJ, Willemse PPM, van den Bergh RCN. Evolution of European prostate cancer screening protocols and summary of ongoing trials. BJU Int 2024; 134:31-42. [PMID: 38469728 DOI: 10.1111/bju.16311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Population-based organised repeated screening for prostate cancer has been found to reduce disease-specific mortality, but with substantial overdiagnosis leading to overtreatment. Although only very few countries have implemented a screening programme on a national level, individual prostate-specific antigen (PSA) testing is common. This opportunistic testing may have little favourable impact, while stressing the side-effects. The classic early detection protocols as were state-of-the-art in the 1990s applied a PSA and digital rectal examination threshold for sextant systematic prostate biopsy, with a fixed interval for re-testing, and limited indication for expectant management. In the three decades since these trials were started, different important improvements have become available in the cascade of screening, indication for biopsy, and treatment. The main developed aspects include: better identification of individuals at risk (using early/baseline PSA, family history, and/or genetic profile), individualised re-testing interval, optimised and individualised starting and stopping age, with gradual invitation at a fixed age rather than invitation of a wider range of age groups, risk stratification for biopsy (using PSA density, risk calculator, magnetic resonance imaging, serum and urine biomarkers, or combinations/sequences), targeted biopsy, transperineal biopsy approach, active surveillance for low-risk prostate cancer, and improved staging of disease. All these developments are suggested to decrease the side-effects of screening, while at least maintaining the advantages, but Level 1 evidence is lacking. The knowledge gained and new developments on early detection are being tested in different prospective screening trials throughout Europe. In addition, the European Union-funded PRostate cancer Awareness and Initiative for Screening in the European Union (PRAISE-U) project will compare and evaluate different screening pilots throughout Europe. Implementation and sustainability will also be addressed. Modern screening approaches may reduce the burden of the second most frequent cause of cancer-related death in European males, while minimising side-effects. Also, less efficacious opportunistic early detection may be indirectly reduced.
Collapse
Affiliation(s)
- Meike J van Harten
- Cancer Center, Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Monique J Roobol
- Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Peter-Paul M Willemse
- Cancer Center, Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roderick C N van den Bergh
- Cancer Institute, Erasmus University Medical Centre, Rotterdam, The Netherlands
- St Antonius Hospital, Utrecht, The Netherlands
| |
Collapse
|
8
|
Würnschimmel C, Menges D, Kwiatkowski M, Sigg S, Prause L, Mattei A, Engeler D, Eberli D, Seifert H, Valerio M, Rentsch CA, Mortezavi A. Prostate cancer screening in Switzerland: a literature review and consensus statement from the Swiss Society of Urology. Swiss Med Wkly 2024; 154:3626. [PMID: 38820236 DOI: 10.57187/s.3626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024] Open
Abstract
Over a decade ago, the United States Preventive Services Taskforce (USPSTF) recommended against prostate-specific antigen (PSA)-based screening for prostate cancer in all men, which considerably influenced prostate cancer screening policies worldwide after that. Consequently, the world has seen increasing numbers of advanced stages and prostate cancer deaths, which later led the USPSTF to withdraw its initial statement. Meanwhile, the European Union has elaborated a directive to address the problem of implementing prostate cancer screening in "Europe's Beating Cancer Plan". In Switzerland, concerned urologists formed an open Swiss Prostate Cancer Screening Group to improve the early detection of prostate cancer. On the 20th of September 2023, during the annual general assembly of the Swiss Society of Urology (SGU/SSU) in Lausanne, members positively voted for a stepwise approach to evaluate the feasibility of implementing organised prostate cancer screening programs in Switzerland. The following article will summarise the events and scientific advances in the last decade during which evidence and promising additional modalities to complement PSA-based prostate cancer screening have emerged. It also aims to provide an overview of contemporary strategies and their potential harms and benefits.
Collapse
Affiliation(s)
- Christoph Würnschimmel
- Department of Urology, Luzerner Kantonsspital, Lucerne, Switzerland
- Faculty of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Maciej Kwiatkowski
- Department of Urology, Kantonsspital Aarau, Aarau, Switzerland
- Faculty Member, University Hospital Basel, Basel, Switzerland
| | - Silvan Sigg
- Department of Urology, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Lukas Prause
- Department of Urology, Kantonsspital Aarau, Aarau, Switzerland
| | - Agostino Mattei
- Department of Urology, Luzerner Kantonsspital, Lucerne, Switzerland
- Faculty of Health Sciences and Medicine, University of Lucerne, Lucerne, Switzerland
| | - Daniel Engeler
- Department of Urology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Helge Seifert
- Department of Urology, University Hospital Basel, Basel, Switzerland
| | - Massimo Valerio
- Department of Urology, University Hospital Geneva, Geneva, Switzerland
| | - Cyrill A Rentsch
- Department of Urology, University Hospital Basel, Basel, Switzerland
| | - Ashkan Mortezavi
- Department of Urology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
9
|
Garraway IP, Carlsson SV, Nyame YA, Vassy JL, Chilov M, Fleming M, Frencher SK, George DJ, Kibel AS, King SA, Kittles R, Mahal BA, Pettaway CA, Rebbeck T, Rose B, Vince R, Winn RA, Yamoah K, Oh WK. Prostate Cancer Foundation Screening Guidelines for Black Men in the United States. NEJM EVIDENCE 2024; 3:EVIDoa2300289. [PMID: 38815168 DOI: 10.1056/evidoa2300289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
BACKGROUND In the United States, Black men are at highest risk for being diagnosed with and dying from prostate cancer. Given this disparity, we examined relevant data to establish clinical prostate-specific antigen (PSA) screening guidelines for Black men in the United States. METHODS A comprehensive literature search identified 1848 unique publications for screening. Of those screened, 287 studies were selected for full-text review, and 264 were considered relevant and form the basis for these guidelines. The numbers were reported according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. RESULTS Three randomized controlled trials provided Level 1 evidence that regular PSA screening of men 50 to 74 years of age of average risk reduced metastasis and prostate cancer death at 16 to 22 years of follow-up. The best available evidence specifically for Black men comes from observational and modeling studies that consider age to obtain a baseline PSA, frequency of testing, and age when screening should end. Cohort studies suggest that discussions about baseline PSA testing between Black men and their clinicians should begin in the early 40s, and data from modeling studies indicate prostate cancer develops 3 to 9 years earlier in Black men compared with non-Black men. Lowering the age for baseline PSA testing to 40 to 45 years of age from 50 to 55 years of age, followed by regular screening until 70 years of age (informed by PSA values and health factors), could reduce prostate cancer mortality in Black men (approximately 30% relative risk reduction) without substantially increasing overdiagnosis. CONCLUSIONS These guidelines recommend that Black men should obtain information about PSA screening for prostate cancer. Among Black men who elect screening, baseline PSA testing should occur between ages 40 and 45. Depending on PSA value and health status, annual screening should be strongly considered. (Supported by the Prostate Cancer Foundation.).
Collapse
Affiliation(s)
- Isla P Garraway
- Department of Urology, David Geffen School of Medicine, University of California and Department of Surgical and Perioperative Care, VA Greater Los Angeles Healthcare System, Los Angeles
| | - Sigrid V Carlsson
- Departments of Surgery and Epidemiology and Biostatistics, Urology Service, Memorial Sloan Kettering Cancer Center, New York
- Department of Urology, Sahlgrenska Academy at Gothenburg University, Gothenburg, and Department of Translational Medicine, Division of Urological Cancers, Medical Faculty, Lund University, Lund, Sweden
| | - Yaw A Nyame
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle
- Department of Urology, University of Washington, Seattle
| | - Jason L Vassy
- Center for Healthcare Organization and Implementation Research (CHOIR), Veterans Health Administration, Bedford and Boston
- Harvard Medical School and Brigham and Women's Hospital, Boston
| | - Marina Chilov
- Medical Library, Memorial Sloan Kettering Cancer Center, New York
| | - Mark Fleming
- Virginia Oncology Associates, US Oncology Network, Norfolk, VA
| | - Stanley K Frencher
- Martin Luther King Jr. Community Hospital and University of California, Los Angeles
| | - Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC
| | - Adam S Kibel
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Sherita A King
- Section of Urology, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA
| | - Rick Kittles
- Morehouse School of Medicine, Community Health and Preventive Medicine, Atlanta
| | - Brandon A Mahal
- Sylvester Comprehensive Cancer Center, Miami
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami
| | - Curtis A Pettaway
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston
| | - Timothy Rebbeck
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston
- Harvard T.H. Chan School of Public Health, Boston
| | - Brent Rose
- Department of Radiation Oncology, University of California, San Diego
- Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Randy Vince
- Department of Urology, University of Michigan, Ann Arbor
| | - Robert A Winn
- Massey Cancer Center, Virginia Commonwealth University, Richmond
- Department of Internal Medicine, Virginia Commonwealth University, Richmond
| | - Kosj Yamoah
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
- James A. Haley Veterans' Hospital, Tampa, FL
| | - William K Oh
- Prostate Cancer Foundation, Santa Monica, CA
- Division of Hematology and Medical Oncology, Tisch Cancer Institute at Mount Sinai, New York
| |
Collapse
|
10
|
Kensler KH, Johnson R, Morley F, Albrair M, Dickerman BA, Gulati R, Holt SK, Iyer HS, Kibel AS, Lee JR, Preston MA, Vassy JL, Wolff EM, Nyame YA, Etzioni R, Rebbeck TR. Prostate cancer screening in African American men: a review of the evidence. J Natl Cancer Inst 2024; 116:34-52. [PMID: 37713266 PMCID: PMC10777677 DOI: 10.1093/jnci/djad193] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Prostate cancer is the most diagnosed cancer in African American men, yet prostate cancer screening regimens in this group are poorly guided by existing evidence, given underrepresentation of African American men in prostate cancer screening trials. It is critical to optimize prostate cancer screening and early detection in this high-risk group because underdiagnosis may lead to later-stage cancers at diagnosis and higher mortality while overdiagnosis may lead to unnecessary treatment. METHODS We performed a review of the literature related to prostate cancer screening and early detection specific to African American men to summarize the existing evidence available to guide health-care practice. RESULTS Limited evidence from observational and modeling studies suggests that African American men should be screened for prostate cancer. Consideration should be given to initiating screening of African American men at younger ages (eg, 45-50 years) and at more frequent intervals relative to other racial groups in the United States. Screening intervals can be optimized by using a baseline prostate-specific antigen measurement in midlife. Finally, no evidence has indicated that African American men would benefit from screening beyond 75 years of age; in fact, this group may experience higher rates of overdiagnosis at older ages. CONCLUSIONS The evidence base for prostate cancer screening in African American men is limited by the lack of large, randomized studies. Our literature search supported the need for African American men to be screened for prostate cancer, for initiating screening at younger ages (45-50 years), and perhaps screening at more frequent intervals relative to men of other racial groups in the United States.
Collapse
Affiliation(s)
- Kevin H Kensler
- Department of Population Health Sciences, Weill Cornell Medical Center, New York, NY, USA
| | - Roman Johnson
- Center for Global Health, Massachusetts General Hospital, Boston, MA, USA
| | - Faith Morley
- Department of Population Health Sciences, Weill Cornell Medical Center, New York, NY, USA
| | - Mohamed Albrair
- Department of Global Health, University of Washington, Seattle, WA, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Barbra A Dickerman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sarah K Holt
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Hari S Iyer
- Section of Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Adam S Kibel
- Department of Urology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jenney R Lee
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Mark A Preston
- Department of Urology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jason L Vassy
- VA Boston Healthcare System, Boston, MA, USA
- Division of General Internal Medicine and Primary Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Erika M Wolff
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Yaw A Nyame
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
11
|
Cooperberg MR. Can early prostate cancer screening help address mortality disparities among Black men? J Natl Cancer Inst 2024; 116:9-11. [PMID: 37964676 DOI: 10.1093/jnci/djad217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Affiliation(s)
- Matthew R Cooperberg
- Departments of Urology and Epidemiology & Biostatistics, UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
12
|
Zhang Z, Tian A, Che J, Miao Y, Liu Y, Liu Y, Xu Y. Application and optimization of prostate-specific antigen screening strategy in the diagnosis of prostate cancer: a systematic review. Front Oncol 2024; 13:1320681. [PMID: 38264758 PMCID: PMC10803420 DOI: 10.3389/fonc.2023.1320681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
Currently, prostate cancer (PCa) poses a global risk to the well-being of males. Over the past few years, the utilization of prostate-specific antigen (PSA) screening has become prevalent in the identification and management of PCa, which has promoted a large number of patients with advanced PCa to receive timely treatment and reduce the mortality. Nevertheless, the utilization of PSA in PCa screening has sparked debate, and certain research has validated the potential for overdiagnosis and overtreatment associated with PSA screening. Hence, in order to decrease the mortality rate of PCa patients and prevent unnecessary diagnosis and treatment, it is crucial to carefully choose the suitable population and strategy for PSA screening in PCa. In this systematic review, the clinical studies on PSA screening for the diagnosis and treatment of PCa were thoroughly examined. The review also delved into the effects and mechanisms of PSA screening on the prognosis of PCa patients, examined the factors contributing to overdiagnosis and overtreatment, and put forth strategies for optimization. The objective of this research is to offer valuable recommendations regarding the utilization of PSA screening for the detection and management of PCa.
Collapse
Affiliation(s)
- Zhengchao Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Jizhong Che
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yandong Miao
- Department of Oncology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
13
|
Vickers AJ, Lilja H. Eight Misconceptions about Prostate-Specific Antigen. Clin Chem 2024; 70:13-16. [PMID: 38175588 DOI: 10.1093/clinchem/hvad138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/03/2023] [Indexed: 01/05/2024]
Affiliation(s)
- Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hans Lilja
- Departments of Pathology and Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
14
|
Remmers S, Bangma CH, Godtman RA, Carlsson SV, Auvinen A, Tammela TLJ, Denis LJ, Nelen V, Villers A, Rebillard X, Kwiatkowski M, Recker F, Wyler S, Zappa M, Puliti D, Gorini G, Paez A, Lujan M, Nieboer D, Schröder FH, Roobol MJ. Relationship Between Baseline Prostate-specific Antigen on Cancer Detection and Prostate Cancer Death: Long-term Follow-up from the European Randomized Study of Screening for Prostate Cancer. Eur Urol 2023; 84:503-509. [PMID: 37088597 PMCID: PMC10759255 DOI: 10.1016/j.eururo.2023.03.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND The European Association of Urology guidelines recommend a risk-based strategy for prostate cancer screening based on the first prostate-specific antigen (PSA) level and age. OBJECTIVE To analyze the impact of the first PSA level on prostate cancer (PCa) detection and PCa-specific mortality (PCSM) in a population-based screening trial (repeat screening every 2-4 yr). DESIGN, SETTING, AND PARTICIPANTS We evaluated 25589 men aged 55-59 yr, 16898 men aged 60-64 yr, and 12936 men aged 65-69 yr who attended at least one screening visit in the European Randomized Study of Screening for Prostate Cancer (ERSPC) trial (screening arm: repeat PSA testing every 2-4 yr and biopsy in cases with elevated PSA; control arm: no active screening offered) during 16-yr follow-up (FU). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We assessed the actuarial probability for any PCa and for clinically significant (cs)PCa (Gleason ≥7). Cox proportional-hazards regression was performed to assess whether the association between baseline PSA and PCSM was comparable for all age groups. A Lorenz curve was computed to assess the association between baseline PSA and PCSM for men aged 60-61 yr. RESULTS AND LIMITATIONS The overall actuarial probability at 16 yr ranged from 12% to 16% for any PCa and from 3.7% to 5.7% for csPCa across the age groups. The actuarial probability of csPCa at 16 yr ranged from 1.2-1.5% for men with PSA <1.0 ng/ml to 13.3-13.8% for men with PSA ≥3.0 ng/ml. The association between baseline PSA and PCSM differed marginally among the three age groups. A Lorenz curve for men aged 60-61 yr showed that 92% of lethal PCa cases occurred among those with PSA above the median (1.21 ng/ml). In addition, for men initially screened at age 60-61 yr with baseline PSA <2 ng/ml, further continuation of screening is unlikely to be beneficial after the age of 68-70 yr if PSA is still <2 ng/ml. No case of PCSM emerged in the subsequent 8 yr (up to age 76-78 yr). A limitation is that these results may not be generalizable to an opportunistic screening setting or to contemporary clinical practice. CONCLUSIONS In all age groups, baseline PSA can guide decisions on the repeat screening interval. Baseline PSA of <1.0 ng/ml for men aged 55-69 yr is a strong indicator to delay or stop further screening. PATIENT SUMMARY In prostate cancer screening, the patient's baseline PSA (prostate-specific antigen) level can be used to guide decisions on when to repeat screening. The PSA test when used according to current knowledge is valuable in helping to reduce the burden of prostate cancer.
Collapse
Affiliation(s)
- Sebastiaan Remmers
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands.
| | - Chris H Bangma
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Rebecka A Godtman
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at Goteborg University, Goteborg, Sweden
| | - Sigrid V Carlsson
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at Goteborg University, Goteborg, Sweden; Departments of Surgery (Urology Service) and Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anssi Auvinen
- School of Health Sciences, University of Tampere, Tampere, Finland
| | - Teuvo L J Tammela
- Department of Urology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Louis J Denis
- Department of Urology, Meeting Centre Antwerp, Antwerp, Belgium
| | - Vera Nelen
- Provincial Institute for Hygiene, Antwerp, Belgium
| | - Arnauld Villers
- Department of Urology, Université Lille Nord de France, Lille, France
| | - Xavier Rebillard
- Department of Urology, Clinique Beau Soleil, Montpellier, France
| | - Maciej Kwiatkowski
- Department of Urology, Kantonsspital Aarau, Aarau, Switzerland; Medical Faculty, University of Basel, Basel, Switzerland; Department of Urology, Academic Hospital Braunschweig, Braunschweig, Germany
| | | | - Stephen Wyler
- Department of Urology, Kantonsspital Aarau, Aarau, Switzerland; Medical Faculty, University of Basel, Basel, Switzerland
| | - Marco Zappa
- Oncologic Network, Prevention and Research Institute (ISPRO), Florence, Italy
| | - Donella Puliti
- Oncologic Network, Prevention and Research Institute (ISPRO), Florence, Italy
| | - Giuseppe Gorini
- Oncologic Network, Prevention and Research Institute (ISPRO), Florence, Italy
| | - Alvaro Paez
- Department of Urology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - Marcos Lujan
- Department of Urology, Hospital Infanta Cristina, Madrid, Spain
| | - Daan Nieboer
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands; Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Fritz H Schröder
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
15
|
Heijnsdijk EAM, de Koning HJ. Wise Prostate-specific Antigen Testing Means a Limited, Risk-adjusted, and Personal Approach. Eur Urol 2023; 84:359-360. [PMID: 37296041 DOI: 10.1016/j.eururo.2023.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023]
Affiliation(s)
- Eveline A M Heijnsdijk
- Department of Public Health, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
| | - Harry J de Koning
- Department of Public Health, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Ramalingam V, McCarthy CJ, Degerstedt S, Ahmed M. Image-Guided Prostate Cryoablation: State-of-the-Art. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1589. [PMID: 37763708 PMCID: PMC10535457 DOI: 10.3390/medicina59091589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023]
Abstract
Image-guided focal therapy has increased in popularity as a treatment option for patients with primary and locally recurrent prostate cancer. This review will cover the basic indications, evaluation, treatment algorithm, and follow-up for patients undergoing image-guided ablation of the prostate. Additionally, this paper will serve as an overview of some technical approaches to cases so that physicians can familiarize themselves with working in this space. While the focus of this paper is prostate cryoablation, readers will obtain a basic literature overview of some of the additional available image-guided treatment modalities for focal prostate therapy.
Collapse
Affiliation(s)
- Vijay Ramalingam
- Beth Israel Deaconess Medical Center, Division of Vascular and Interventional Radiology, Harvard Medical School, Deaconess Rd, Rosenburg 3, Boston, MA 02215, USA; (C.J.M.); (S.D.); (M.A.)
| | | | | | | |
Collapse
|
17
|
Wei JT, Barocas D, Carlsson S, Coakley F, Eggener S, Etzioni R, Fine SW, Han M, Kim SK, Kirkby E, Konety BR, Miner M, Moses K, Nissenberg MG, Pinto PA, Salami SS, Souter L, Thompson IM, Lin DW. Early Detection of Prostate Cancer: AUA/SUO Guideline Part I: Prostate Cancer Screening. J Urol 2023; 210:46-53. [PMID: 37096582 PMCID: PMC11060750 DOI: 10.1097/ju.0000000000003491] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE The summary presented herein covers recommendations on the early detection of prostate cancer and provides a framework to facilitate clinical decision-making in the implementation of prostate cancer screening, biopsy, and follow-up. This is Part I of a two-part series that focuses on prostate cancer screening. Please refer to Part II for discussion of initial and repeat biopsies as well as biopsy technique. MATERIALS AND METHODS The systematic review utilized to inform this guideline was conducted by an independent methodological consultant. The systematic review was based on searches in Ovid MEDLINE and Embase and Cochrane Database of Systematic Reviews (January 1, 2000-November 21, 2022). Searches were supplemented by reviewing reference lists of relevant articles. RESULTS The Early Detection of Prostate Cancer Panel developed evidence- and consensus-based guideline statements to provide guidance in prostate cancer screening, initial and repeat biopsy, and biopsy technique. CONCLUSIONS Prostate-specific antigen (PSA)-based prostate cancer screening in combination with shared decision-making (SDM) is recommended. Current data regarding risk from population-based cohorts provide a basis for longer screening intervals and tailored screening, and the use of available online risk calculators is encouraged.
Collapse
Affiliation(s)
- John T Wei
- University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | - Ruth Etzioni
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Samson W Fine
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Misop Han
- Johns Hopkins University, Baltimore, Maryland
| | - Sennett K Kim
- American Urological Association, Linthicum, Maryland
| | - Erin Kirkby
- American Urological Association, Linthicum, Maryland
| | | | | | | | - Merel G Nissenberg
- National Alliance of State Prostate Cancer Coalitions, Los Angeles, California
| | | | | | - Lesley Souter
- Nomadic EBM Methodology, Smithville, Ontario, Canada
| | | | | |
Collapse
|
18
|
Michael ZD, Kotamarti S, Arcot R, Morris K, Shah A, Anderson J, Armstrong AJ, Gupta RT, Patierno S, Barrett NJ, George DJ, Preminger GM, Moul JW, Oeffinger KC, Shah K, Polascik TJ. Initial Longitudinal Outcomes of Risk-Stratified Men in Their Forties Screened for Prostate Cancer Following Implementation of a Baseline Prostate-Specific Antigen. World J Mens Health 2023; 41:631-639. [PMID: 36047079 PMCID: PMC10307649 DOI: 10.5534/wjmh.220068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/27/2022] [Accepted: 07/21/2022] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Prostate cancer (PCa) screening can lead to potential over-diagnosis/over-treatment of indolent cancers. There is a need to optimize practices to better risk-stratify patients. We examined initial longitudinal outcomes of mid-life men with an elevated baseline prostate-specific antigen (PSA) following initiation of a novel screening program within a system-wide network. MATERIALS AND METHODS We assessed our primary care network patients ages 40 to 49 years with a PSA measured following implementation of an electronic health record screening algorithm from 2/2/2017-2/21/2018. The multidisciplinary algorithm was developed taking factors including age, race, family history, and PSA into consideration to provide a personalized approach to urology referral to be used with shared decision-making. Outcomes of men with PSA ≥1.5 ng/mL were evaluated through 7/2021. Statistical analyses identified factors associated with PCa detection. Clinically significant PCa (csPCa) was defined as Gleason Grade Group (GGG) ≥2 or GGG1 with PSA ≥10 ng/mL. RESULTS The study cohort contained 564 patients, with 330 (58.5%) referred to urology for elevated PSA. Forty-nine (8.7%) underwent biopsy; of these, 20 (40.8%) returned with PCa. Eleven (2.0% of total cohort and 55% of PCa diagnoses) had csPCa. Early referral timing (odds ratio [OR], 4.58) and higher PSA (OR, 1.07) were significantly associated with PCa at biopsy on multivariable analysis (both p<0.05), while other risk factors were not. Referred patients had higher mean PSAs (2.97 vs. 1.98, p=0.001). CONCLUSIONS Preliminary outcomes following implementation of a multidisciplinary screening algorithm identified PCa in a small, important percentage of men in their forties. These results provide insight into baseline PSA measurement to provide early risk stratification and detection of csPCa in patients with otherwise extended life expectancy. Further follow-up is needed to possibly determine the prognostic significance of such mid-life screening and optimize primary care physician-urologist coordination.
Collapse
Affiliation(s)
- Zoe D Michael
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| | - Srinath Kotamarti
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Rohith Arcot
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kostantinos Morris
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Anand Shah
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - John Anderson
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Family Medicine and Community Health, Duke University Medical Center, Durham, NC, USA
| | - Andrew J Armstrong
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Rajan T Gupta
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Steven Patierno
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Family Medicine and Community Health, Duke University Medical Center, Durham, NC, USA
| | - Nadine J Barrett
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Daniel J George
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Glenn M Preminger
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Judd W Moul
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kevin C Oeffinger
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Kevin Shah
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Thomas J Polascik
- The Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
- Division of Urologic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
19
|
Kachuri L, Hoffmann TJ, Jiang Y, Berndt SI, Shelley JP, Schaffer KR, Machiela MJ, Freedman ND, Huang WY, Li SA, Easterlin R, Goodman PJ, Till C, Thompson I, Lilja H, Van Den Eeden SK, Chanock SJ, Haiman CA, Conti DV, Klein RJ, Mosley JD, Graff RE, Witte JS. Genetically adjusted PSA levels for prostate cancer screening. Nat Med 2023; 29:1412-1423. [PMID: 37264206 PMCID: PMC10287565 DOI: 10.1038/s41591-023-02277-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 02/27/2023] [Indexed: 06/03/2023]
Abstract
Prostate-specific antigen (PSA) screening for prostate cancer remains controversial because it increases overdiagnosis and overtreatment of clinically insignificant tumors. Accounting for genetic determinants of constitutive, non-cancer-related PSA variation has potential to improve screening utility. In this study, we discovered 128 genome-wide significant associations (P < 5 × 10-8) in a multi-ancestry meta-analysis of 95,768 men and developed a PSA polygenic score (PGSPSA) that explains 9.61% of constitutive PSA variation. We found that, in men of European ancestry, using PGS-adjusted PSA would avoid up to 31% of negative prostate biopsies but also result in 12% fewer biopsies in patients with prostate cancer, mostly with Gleason score <7 tumors. Genetically adjusted PSA was more predictive of aggressive prostate cancer (odds ratio (OR) = 3.44, P = 6.2 × 10-14, area under the curve (AUC) = 0.755) than unadjusted PSA (OR = 3.31, P = 1.1 × 10-12, AUC = 0.738) in 106 cases and 23,667 controls. Compared to a prostate cancer PGS alone (AUC = 0.712), including genetically adjusted PSA improved detection of aggressive disease (AUC = 0.786, P = 7.2 × 10-4). Our findings highlight the potential utility of incorporating PGS for personalized biomarkers in prostate cancer screening.
Collapse
Affiliation(s)
- Linda Kachuri
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Hoffmann
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Yu Jiang
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - John P Shelley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Shengchao A Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Ryder Easterlin
- Biological and Medical Informatics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Cathee Till
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ian Thompson
- CHRISTUS Santa Rosa Medical Center Hospital, San Antonio, TX, USA
| | - Hans Lilja
- Departments of Laboratory Medicine, Surgery and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Population and Preventive Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David V Conti
- Center for Genetic Epidemiology, Department of Population and Preventive Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan D Mosley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca E Graff
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| | - John S Witte
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Biomedical Data Science and Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
20
|
Etzioni R, Castle PE. Shopping for New Cancer Screening Tests. J Clin Oncol 2023; 41:2471-2473. [PMID: 36862969 PMCID: PMC10414693 DOI: 10.1200/jco.23.00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
|
21
|
Kato M, Horiguchi G, Ueda T, Fujihara A, Hongo F, Okihara K, Marunaka Y, Teramukai S, Ukimura O. A big data-based prediction model for prostate cancer incidence in Japanese men. Sci Rep 2023; 13:6579. [PMID: 37085532 PMCID: PMC10121595 DOI: 10.1038/s41598-023-33725-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/18/2023] [Indexed: 04/23/2023] Open
Abstract
To define a normal range for PSA values (ng/mL) by age and create a prediction model for prostate cancer incidence. We conducted a retrospective analysis using 263,073 observations of PSA values in Japanese men aged 18-98 years (2007-2017), including healthy men and those diagnosed with prostate cancer. Percentiles for 262,639 PSA observations in healthy men aged 18-70 years were calculated and plotted to elucidate the normal fluctuation range for PSA values by age. Univariable and multivariable logistic regression analyses were performed to develop a predictive model for prostate cancer incidence. PSA levels and PSA velocity increased with age in healthy men. However, there was no difference in PSA velocity with age in men diagnosed with prostate cancer. Logistic regression analysis showed an increased risk of prostate cancer for PSA slopes ranging from 0.5 to 3.5 ng/mL/year. This study provides age-specific normal fluctuation ranges for PSA levels in men aged 18-75 years and presents a novel and personalized prediction model for prostate cancer incidence. We found that PSA slope values of > 3.5 ng/mL/year may indicate a rapid increase in PSA levels caused by pathological condition such as inflammation but are unlikely to indicate cancer risk.
Collapse
Affiliation(s)
- Mineyuki Kato
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, 602-8566, Japan
| | - Go Horiguchi
- Department of Biostatistics, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, Japan
| | - Takashi Ueda
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, 602-8566, Japan.
| | - Atsuko Fujihara
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, 602-8566, Japan
| | - Fumiya Hongo
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, 602-8566, Japan
| | - Koji Okihara
- Department of Urology, North Medical Center Kyoto Prefectural University of Medicine, Yosano-gun, Kyoto, Japan
| | - Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, Kyoto, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, Japan
| | - Osamu Ukimura
- Department of Urology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, 602-8566, Japan
| |
Collapse
|
22
|
Albers P. Re: Association of Prostate-specific Antigen Screening Rates with Subsequent Metastatic Prostate Cancer Incidence at US Veterans Health Administration Facilities: Prostate cancer screening needs not only to be well meant but also well done. Eur Urol 2023; 83:369-370. [PMID: 36631351 DOI: 10.1016/j.eururo.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023]
Affiliation(s)
- Peter Albers
- Department of Urology, Düsseldorf University, Düsseldorf, Germany; Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ) Heidelberg, Germany.
| |
Collapse
|
23
|
Krilaviciute A, Albers P, Lakes J, Radtke JP, Herkommer K, Gschwend J, Peters I, Kuczyk M, Koerber SA, Debus J, Kristiansen G, Schimmöller L, Antoch G, Makowski M, Wacker F, Schlemmer H, Benner A, Giesel F, Siener R, Arsov C, Hadaschik B, Becker N, Kaaks R. Adherence to a risk-adapted screening strategy for prostate cancer: First results of the PROBASE trial. Int J Cancer 2023; 152:854-864. [PMID: 36121664 DOI: 10.1002/ijc.34295] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 01/11/2023]
Abstract
PROBASE is a population-based, randomized trial of 46 495 German men recruited at age 45 to compare effects of risk-adapted prostate cancer (PCa) screening starting either immediately at age 45, or at a deferred age of 50 years. Based on prostate-specific antigen (PSA) levels, men are classified into risk groups with different screening intervals: low-risk (<1.5 ng/ml, 5-yearly screening), intermediate-risk (1.5-2.99 ng/ml, 2 yearly), and high risk (>3 ng/ml, recommendation for immediate biopsy). Over the first 6 years of study participation, attendance rates to scheduled screening visits varied from 70.5% to 79.4%, depending on the study arm and risk group allocation, in addition 11.2% to 25.4% of men reported self-initiated PSA tests outside the PROBASE protocol. 38.5% of participants had a history of digital rectal examination or PSA testing prior to recruitment to PROBASE, frequently associated with family history of PCa. These men showed higher rates (33% to 57%, depending on subgroups) of self-initiated PSA testing in-between PROBASE screening rounds. In the high-risk groups (both arms), the biopsy acceptance rate was 64% overall, but was higher among men with screening PSA ≥4 ng/ml (>71%) and with PIRADS ≥3 findings upon multiparameter magnetic resonance imaging (mpMRI) (>72%), compared with men with PSA ≥3 to 4 ng/ml (57%) or PIRADS score ≤ 2 (59%). Overall, PROBASE shows good acceptance of a risk-adapted PCa screening strategy in Germany. Implementation of such a strategy should be accompanied by a well-structured communication, to explain not only the benefits but also the harms of PSA screening.
Collapse
Affiliation(s)
- Agne Krilaviciute
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Albers
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jale Lakes
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jan Philipp Radtke
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Kathleen Herkommer
- Department of Urology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Munchen, Germany
| | - Jürgen Gschwend
- Department of Urology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Munchen, Germany
| | - Inga Peters
- Department of Urology, Medical University Hannover, Hannover, Germany.,Department of Urology, Krankenhaus Nordwest, Frankfurt am Main, Germany
| | - Markus Kuczyk
- Department of Urology, Medical University Hannover, Hannover, Germany
| | - Stefan A Koerber
- Department of Radiation Oncology, Heidelberg University Hospital, Ruprecht Karls University, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Ruprecht Karls University, Heidelberg, Germany
| | | | - Lars Schimmöller
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcus Makowski
- Institute of Diagnostic and Interventional Radiology, Technical University Munich, München, Germany
| | - Frank Wacker
- Institute of Diagnostic and Interventional Radiology, Medical University Hannover, Hannover, Germany
| | - Heinz Schlemmer
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, Medical Faculty, Duesseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Roswitha Siener
- Department of Radiation Oncology, Heidelberg University Hospital, Ruprecht Karls University, Heidelberg, Germany.,Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Christian Arsov
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Boris Hadaschik
- Department of Urology, Heidelberg University Hospital, Ruprecht Karls University, Heidelberg, Germany
| | - Nikolaus Becker
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Pagadala MS, Lynch J, Karunamuni R, Alba PR, Lee KM, Agiri FY, Anglin T, Carter H, Gaziano JM, Jasuja GK, Deka R, Rose BS, Panizzon MS, Hauger RL, Seibert TM. Polygenic risk of any, metastatic, and fatal prostate cancer in the Million Veteran Program. J Natl Cancer Inst 2023; 115:190-199. [PMID: 36305680 PMCID: PMC9905969 DOI: 10.1093/jnci/djac199] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 10/26/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Genetic scores may provide an objective measure of prostate cancer risk and thus inform screening decisions. We evaluated whether a polygenic hazard score based on 290 genetic variants (PHS290) is associated with prostate cancer risk in a diverse population, including Black men, who have higher average risk of prostate cancer death but are often treated as a homogeneously high-risk group. METHODS This was a retrospective analysis of the Million Veteran Program, a national, population-based cohort study of US military veterans conducted 2011-2021. Cox proportional hazards analyses tested for association of genetic and other risk factors (including self-reported race and ethnicity and family history) with age at death from prostate cancer, age at diagnosis of metastatic (nodal or distant) prostate cancer, and age at diagnosis of any prostate cancer. RESULTS A total of 590 750 male participants were included. Median age at last follow-up was 69 years. PHS290 was associated with fatal prostate cancer in the full cohort and for each racial and ethnic group (P < .001). Comparing men in the highest 20% of PHS290 with those in the lowest 20% (based on percentiles from an independent training cohort), the hazard ratio for fatal prostate cancer was 4.42 (95% confidence interval = 3.91 to 5.02). When accounting for guideline-recommended risk factors (family history, race, and ethnicity), PHS290 remained a strong independent predictor of any, metastatic, and fatal prostate cancer. CONCLUSIONS PHS290 stratified US veterans of diverse ancestry for lifetime risk of prostate cancer, including metastatic and fatal cancer. Predicting genetic risk of lethal prostate cancer with PHS290 might inform individualized decisions about prostate cancer screening.
Collapse
Affiliation(s)
- Meghana S Pagadala
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
- Biomedical Science Program, University of California San Diego, La Jolla, CA, USA
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
| | - Patrick R Alba
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Fatai Y Agiri
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
| | - Tori Anglin
- VA Informatics and Computing Infrastructure, VA Salt Lake City Healthcare System (VINCI), Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hannah Carter
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Guneet Kaur Jasuja
- Center for Healthcare Organization and Implementation Research (CHOIR), VA Bedford Healthcare System, Bedford, MA, USA
- Section of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Rishi Deka
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Brent S Rose
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Section of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Urology, University of California San Diego, La Jolla, CA, USA
| | - Matthew S Panizzon
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Richard L Hauger
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavioral Genetics of Aging, University of California San Diego, La Jolla, CA, USA
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, USA
| | - Tyler M Seibert
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
25
|
Increasing the value of PSA through improved implementation. Urol Oncol 2023; 41:96-103. [PMID: 34750055 DOI: 10.1016/j.urolonc.2021.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/25/2021] [Indexed: 11/21/2022]
Abstract
Low-value testing and treatment contribute to billions of dollars in waste to the United States health care system annually. High frequency, low-cost testing, including prostate-specific antigen (PSA) testing, is a major contributor to this inefficient health care delivery. Despite decreasing mortality of prostate cancer over the last few decades, the reputation of prostate specific antigen (PSA) for prostate cancer screening has fluctuated over the last decade due to lack of clarity of the benefits of screening and high risk for overtreatment. The value of PSA could be improved by efficient implementation of smarter testing strategies that reduce the harms and increase the benefits.
Collapse
|
26
|
Carlsson SV, Arnsrud Godtman R, Pihl CG, Vickers A, Lilja H, Hugosson J, Månsson M. Young Age on Starting Prostate-specific Antigen Testing Is Associated with a Greater Reduction in Prostate Cancer Mortality: 24-Year Follow-up of the Göteborg Randomized Population-based Prostate Cancer Screening Trial. Eur Urol 2023; 83:103-109. [PMID: 36334968 PMCID: PMC10481420 DOI: 10.1016/j.eururo.2022.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/15/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND The risk of death from prostate cancer (PC) depends on age, but the age at which to start prostate-specific antigen (PSA) screening remains uncertain. OBJECTIVE To study the relationship between risk reduction for PC mortality and age at first PSA screening. DESIGN, SETTING, AND PARTICIPANTS The randomized Göteborg-1 trial invited men for biennial PSA screening between the ages of 50 and 70 yr (screening, n = 10 000) or no invitation but exposure to opportunistic PSA testing (control, n = 10 000). INTERVENTION Regular versus opportunistic PSA screening or no PSA. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We modeled the nonlinear association between starting age and the absolute risk reduction in PC mortality in three settings: (1) intention-to-screen (randomized arms); (2) historical control (screening group and 1990-1994 registry data); and (3) attendees only (screening attendees and matched controls). We tested whether the effect of screening on PC mortality depends on the age at starting screening by comparing survival models with and without an interaction between trial arm and age (intention-to-screen and attendees only). RESULTS AND LIMITATIONS Younger age on starting PSA testing was associated with a greater reduction in PC mortality. Starting screening at age 55 yr approximately halved the risk of PC death compared to first PSA at age 60 yr. The test of association between starting age and the effect of screening on PC mortality was slightly greater than the conventional level of statistical significance (p = 0.052) for the entire cohort, and statistically significant among attendees (p = 0.002). This study is limited by the low number of disease-specific deaths for men starting screening before age 55 yr and the difficulty in discriminating between the effect of starting age and screening duration. CONCLUSIONS Given that prior screening trials included men aged up to 70 yr on starting screening, our results suggest that the effect size reported in prior trials underestimates that of currently recommended programs starting at age 50-55 yr. PATIENT SUMMARY In this study from the Göteborg-1 trial, we looked at the effect of prostate-specific antigen (PSA) screening in reducing men's risk of dying from prostate cancer given the age at which they begin testing. Starting at a younger age reduced the risk of prostate cancer death by a greater amount. We recommend that PSA screening should start no later than at age 55 yr.
Collapse
Affiliation(s)
- Sigrid V Carlsson
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden
| | - Rebecka Arnsrud Godtman
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden; Department of Urology, Sahlgrenska University Hospital, Göteborg, Sweden
| | | | - Andrew Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Lilja
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Jonas Hugosson
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden; Department of Urology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Marianne Månsson
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Göteborg, Sweden.
| |
Collapse
|
27
|
Marima R, Mbeje M, Hull R, Demetriou D, Mtshali N, Dlamini Z. Prostate Cancer Disparities and Management in Southern Africa: Insights into Practices, Norms and Values. Cancer Manag Res 2022; 14:3567-3579. [PMID: 36597514 PMCID: PMC9805733 DOI: 10.2147/cmar.s382903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/01/2022] [Indexed: 12/29/2022] Open
Abstract
Prostate cancer (PCa) is a leading cause of mortality in men of African origin. While men of African descent in high-income countries (HICs) demonstrate poor prognosis compared to their European counterparts, African men on the African continent, particularly Southern Africa have shown even higher PCa mortality rates. Extrinsic factors such as the socioeconomic status, education level, income level, geographic location and race contribute to PCa patient outcome. These are further deepened by the African norms which are highly esteemed and may have detrimental effects on PCa patients' health. Insights into African cultures and social constructs have been identified as key elements towards improving men's health care seeking behaviour which will in turn improve PCa patients' outcome. Compared to Southern Africa, the Eastern, Western and Central African regions have lower PCa incidence rates but higher mortality rates. The availability of cancer medical equipment has also been reported to be disproportionate in Africa, with most cancer resources in Northern and Southern Africa. Even within Southern Africa, cancer management resources are unevenly available where one country must access PCa specialised care in the neighbouring countries. While PCa seems to be better managed in HICs, steps towards effective PCa management are urgently needed in Africa, as this continent represents a significant portion of low-middle-income countries (LMICs). Replacing African men in Africa with African American men may not optimally resolve PCa challenges in Africa. Adopting western PCa management practices can be optimised by integrating improved core-African norms. The aim of this review is to discuss PCa disparities in Africa, deliberate on the significance of integrating African norms around masculinity and discuss challenges and opportunities towards effective PCa care in Africa, particularly in Southern Africa.
Collapse
Affiliation(s)
- Rahaba Marima
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa
| | - Mandisa Mbeje
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa,Department of Medical Oncology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa
| | - Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa
| | - Nompumelelo Mtshali
- Department of Anatomical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa,Correspondence: Zodwa Dlamini, Tel +27 12 319 2614, Email
| |
Collapse
|
28
|
Chou A, Darst BF, Wilkens LR, Le Marchand L, Lilja H, Conti DV, Haiman CA. Association of Prostate-Specific Antigen Levels with Prostate Cancer Risk in a Multiethnic Population: Stability Over Time and Comparison with Polygenic Risk Score. Cancer Epidemiol Biomarkers Prev 2022; 31:2199-2207. [PMID: 36126957 PMCID: PMC9729398 DOI: 10.1158/1055-9965.epi-22-0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/01/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Studies in men of European ancestry suggest prostate-specific antigen (PSA) as a marker of early prostate cancer development that may help to risk-stratify men earlier in life. METHODS We examined PSA levels in men measured up to 10+ years before a prostate cancer diagnosis in association with prostate cancer risk in 2,245 cases and 2,203 controls of African American, Latino, Japanese, Native Hawaiian, and White men in the Multiethnic Cohort. We also compared the discriminative ability of PSA to polygenic risk score (PRS) for prostate cancer. RESULTS Excluding cases diagnosed within 2 and 10 years of blood draw, men with PSA above the median had a prostate cancer OR (95% CIs) of 9.12 (7.66-10.92) and 3.52 (2.50-5.03), respectively, compared with men with PSA below the median. A PSA level above the median identified 90% and 75% of cases diagnosed more than 2 and 10 years after blood draw, respectively. The associations were significantly greater for Gleason ≤7 versus 8+ disease. At 10+ years, the association of prostate cancer with PSA was comparable with that with the PRS [OR per SD increase: 1.88 (1.45-2.46) and 2.12 (1.55-2.93), respectively]. CONCLUSIONS We found PSA to be an informative marker of prostate cancer risk at least a decade before diagnosis across multiethnic populations. This association was diminished with increasing time, greater for low grade tumors, and comparable with a PRS when measured 10+ years before diagnosis. IMPACT Our multiethnic investigation suggests broad clinical implications on the utility of PSA and PRS for risk stratification in prostate cancer screening practices.
Collapse
Affiliation(s)
- Alisha Chou
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, US
| | - Burcu F. Darst
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, US,Present address: Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, U.S.A
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Hans Lilja
- Departments of Laboratory Medicine, Surgery, and Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, U.S.A.; and Department of Translational Medicine, Lund University, Malmö, Sweden
| | - David V. Conti
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, US
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, US
| |
Collapse
|
29
|
French AFU Cancer Committee Guidelines - Update 2022-2024: prostate cancer - Diagnosis and management of localised disease. Prog Urol 2022; 32:1275-1372. [DOI: 10.1016/j.purol.2022.07.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
|
30
|
Kanesvaran R, Castro E, Wong A, Fizazi K, Chua MLK, Zhu Y, Malhotra H, Miura Y, Lee JL, Chong FLT, Pu YS, Yen CC, Saad M, Lee HJ, Kitamura H, Prabhash K, Zou Q, Curigliano G, Poon E, Choo SP, Peters S, Lim E, Yoshino T, Pentheroudakis G. Pan-Asian adapted ESMO Clinical Practice Guidelines for the diagnosis, treatment and follow-up of patients with prostate cancer. ESMO Open 2022; 7:100518. [PMID: 35797737 PMCID: PMC9434138 DOI: 10.1016/j.esmoop.2022.100518] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/28/2022] [Accepted: 05/22/2022] [Indexed: 11/03/2022] Open
Abstract
The most recent version of the European Society for Medical Oncology (ESMO) Clinical Practice Guidelines for the diagnosis, treatment and follow-up of prostate cancer was published in 2020. It was therefore decided, by both the ESMO and the Singapore Society of Oncology (SSO), to convene a special, virtual guidelines meeting in November 2021 to adapt the ESMO 2020 guidelines to take into account the differences associated with the treatment of prostate cancer in Asia. These guidelines represent the consensus opinions reached by experts in the treatment of patients with prostate cancer representing the oncological societies of China (CSCO), India (ISMPO), Japan (JSMO), Korea (KSMO), Malaysia (MOS), Singapore (SSO) and Taiwan (TOS). The voting was based on scientific evidence and was independent of the current treatment practices and drug access restrictions in the different Asian countries. The latter were discussed when appropriate. The aim is to provide guidance for the optimisation and harmonisation of the management of patients with prostate cancer across the different regions of Asia.
Collapse
Affiliation(s)
- R Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore, Singapore.
| | - E Castro
- Department of Medical Oncology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Málaga, Malaga, Spain
| | - A Wong
- Division of Medical Oncology, National University Cancer Institute, Singapore, Singapore
| | - K Fizazi
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - M L K Chua
- Oncology Academic Programme, Duke-NUS Medical School, Singapore, Singapore; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Y Zhu
- Department of Urology, Fudan University, Shanghai Cancer Center, Shanghai, China
| | - H Malhotra
- Department of Medical Oncology, Sri Ram Cancer Center, Mahatma Gandhi Medical College Hospital, Mahatma Gandhi University of Medical Sciences & Technology, Jaipur, India
| | - Y Miura
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - J L Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - F L T Chong
- Department of Radiotherapy and Oncology, Sabah Women and Children's Hospital, Kota Kinabalu, Malaysia
| | - Y-S Pu
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - C-C Yen
- Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Medical Oncology, Center for Immuno-oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan; National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
| | - M Saad
- Department of Clinical Oncology, University of Malaya Medical Centre, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - H J Lee
- Department of Medical Oncology, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, South Korea
| | - H Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - K Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Q Zou
- Department of Urology, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - G Curigliano
- European Institute of Oncology, IRCCS and University of Milano, Milan, Italy
| | - E Poon
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - S P Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore; Medical Oncology, Curie Oncology, Singapore, Singapore
| | - S Peters
- Oncology Department, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - E Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | |
Collapse
|
31
|
Burns D, Anokian E, Saunders EJ, Bristow RG, Fraser M, Reimand J, Schlomm T, Sauter G, Brors B, Korbel J, Weischenfeldt J, Waszak SM, Corcoran NM, Jung CH, Pope BJ, Hovens CM, Cancel-Tassin G, Cussenot O, Loda M, Sander C, Hayes VM, Dalsgaard Sorensen K, Lu YJ, Hamdy FC, Foster CS, Gnanapragasam V, Butler A, Lynch AG, Massie CE, Woodcock DJ, Cooper CS, Wedge DC, Brewer DS, Kote-Jarai Z, Eeles RA. Rare Germline Variants Are Associated with Rapid Biochemical Recurrence After Radical Prostate Cancer Treatment: A Pan Prostate Cancer Group Study. Eur Urol 2022; 82:201-211. [PMID: 35659150 DOI: 10.1016/j.eururo.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Germline variants explain more than a third of prostate cancer (PrCa) risk, but very few associations have been identified between heritable factors and clinical progression. OBJECTIVE To find rare germline variants that predict time to biochemical recurrence (BCR) after radical treatment in men with PrCa and understand the genetic factors associated with such progression. DESIGN, SETTING, AND PARTICIPANTS Whole-genome sequencing data from blood DNA were analysed for 850 PrCa patients with radical treatment from the Pan Prostate Cancer Group (PPCG) consortium from the UK, Canada, Germany, Australia, and France. Findings were validated using 383 patients from The Cancer Genome Atlas (TCGA) dataset. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS A total of 15,822 rare (MAF <1%) predicted-deleterious coding germline mutations were identified. Optimal multifactor and univariate Cox regression models were built to predict time to BCR after radical treatment, using germline variants grouped by functionally annotated gene sets. Models were tested for robustness using bootstrap resampling. RESULTS AND LIMITATIONS Optimal Cox regression multifactor models showed that rare predicted-deleterious germline variants in "Hallmark" gene sets were consistently associated with altered time to BCR. Three gene sets had a statistically significant association with risk-elevated outcome when modelling all samples: PI3K/AKT/mTOR, Inflammatory response, and KRAS signalling (up). PI3K/AKT/mTOR and KRAS signalling (up) were also associated among patients with higher-grade cancer, as were Pancreas-beta cells, TNFA signalling via NKFB, and Hypoxia, the latter of which was validated in the independent TCGA dataset. CONCLUSIONS We demonstrate for the first time that rare deleterious coding germline variants robustly associate with time to BCR after radical treatment, including cohort-independent validation. Our findings suggest that germline testing at diagnosis could aid clinical decisions by stratifying patients for differential clinical management. PATIENT SUMMARY Prostate cancer patients with particular genetic mutations have a higher chance of relapsing after initial radical treatment, potentially providing opportunities to identify patients who might need additional treatments earlier.
Collapse
Affiliation(s)
| | | | | | - Robert G Bristow
- Manchester Cancer Research Centre and CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Michael Fraser
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, Canada; Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada; Department of Medical Biophysics & Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Guido Sauter
- University Medical Centre Hamburg - Eppendorf, Hamburg, Germany
| | - Benedikt Brors
- German Cancer Research Center (DKFZ), Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Jan Korbel
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Joachim Weischenfeldt
- Charité - Universitätsmedizin Berlin, Berlin, Germany; Biotech Research & Innovation Centre (BRIC) & Finsen Laboratory, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian M Waszak
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Niall M Corcoran
- Department of Surgery, The University of Melbourne, Grattan Street, Parkville, Victoria, Australia; Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia; Melbourne Bioinformatics, The University of Melbourne, Grattan Street, Victoria, Australia
| | - Chol-Hee Jung
- The University of Melbourne, Grattan Street, Parkville, Victoria, Australia
| | - Bernard J Pope
- Department of Surgery, The University of Melbourne, Grattan Street, Parkville, Victoria, Australia; Royal Melbourne Hospital, Melbourne, Parwille, Victoria, Australia
| | - Chris M Hovens
- Melbourne Bioinformatics, The University of Melbourne, Grattan Street, Victoria, Australia; The University of Melbourne, Grattan Street, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, The Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia
| | - Géraldine Cancel-Tassin
- CeRePP, Hopital Tenon, Paris, France; Sorbonne Universite, GRC n°5 Predictive Onco-Urology, APHP, Tenon Hospital, Paris, France
| | - Olivier Cussenot
- CeRePP, Hopital Tenon, Paris, France; Sorbonne Universite, GRC n°5 Predictive Onco-Urology, APHP, Tenon Hospital, Paris, France
| | - Massimo Loda
- Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Chris Sander
- cBio Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Vanessa M Hayes
- Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW, Australia; School of Medical Sciences, University of Sydney, Charles Perkins Centre, Camperdown, NSW, Australia
| | - Karina Dalsgaard Sorensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | - Yong-Jie Lu
- Centre for Biomarker and Therapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | - Adam Butler
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, UK
| | - Andy G Lynch
- School of Medicine, University of St Andrews, St Andrews, Fife, UK; School of Mathematics & Statistics, St Andrews, Fife, UK
| | - Charlie E Massie
- CRUK Cambridge Institute, Hutchison MRC Research Centre, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | -
- CR-UK/Prostate Cancer UK, ICGC, The Pan Prostate Cancer Group, UK
| | - Dan J Woodcock
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Colin S Cooper
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Daniel S Brewer
- Norwich Medical School, University of East Anglia, Norwich, UK; The Earlham Institute, Norwich Research Park, Norwich, UK
| | | | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
32
|
Prostate cancer polygenic risk score and prediction of lethal prostate cancer. NPJ Precis Oncol 2022; 6:25. [PMID: 35396534 PMCID: PMC8993880 DOI: 10.1038/s41698-022-00266-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/11/2022] [Indexed: 11/23/2022] Open
Abstract
Polygenic risk scores (PRS) for prostate cancer incidence have been proposed to optimize prostate cancer screening. Prediction of lethal prostate cancer is key to any stratified screening program to avoid excessive overdiagnosis. Herein, PRS for incident prostate cancer was evaluated in two population-based cohorts of unscreened middle-aged men linked to cancer and death registries: the Västerbotten Intervention Project (VIP) and the Malmö Diet and Cancer study (MDC). SNP genotypes were measured by genome-wide SNP genotyping by array followed by imputation or genotyping of selected SNPs using mass spectrometry. The ability of PRS to predict lethal prostate cancer was compared to PSA and a commercialized pre-specified model based on four kallikrein markers. The PRS was associated with incident prostate cancer, replicating previously reported relative risks, and was also associated with prostate cancer death. However, unlike PSA, the PRS did not show stronger association with lethal disease: the hazard ratio for prostate cancer incidence vs. prostate cancer metastasis and death was 1.69 vs. 1.65 in VIP and 1.25 vs. 1.25 in MDC. PSA was a much stronger predictor of prostate cancer metastasis or death with an area-under-the-curve of 0.78 versus 0.63 for the PRS. Importantly, addition of PRS to PSA did not contribute additional risk stratification for lethal prostate cancer. We have shown that a PRS that predicts prostate cancer incidence does not have utility above and beyond that of PSA measured at baseline when applied to the clinically relevant endpoint of prostate cancer death. These findings have implications for public health policies for delivery of prostate cancer screening. Focusing polygenic risk scores on clinically significant endpoints such as prostate cancer metastasis or death would likely improve clinical utility.
Collapse
|
33
|
Mason RJ, Marzouk K, Finelli A, Saad F, So AI, Violette PD, Breau RH, Rendon RA. UPDATE - 2022 Canadian Urological Association recommendations on prostate cancer screening and early diagnosis Endorsement of the 2021 Cancer Care Ontario guidelines on prostate multiparametric magnetic resonance imaging. Can Urol Assoc J 2022; 16:E184-E196. [PMID: 35358414 PMCID: PMC9054332 DOI: 10.5489/cuaj.7851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Ross J. Mason
- Department of Urology, Dalhousie University, Halifax, NS, Canada
| | - Karim Marzouk
- Windsor General Hospital, Windsor, ON; and Western University, London, ON, Canada
| | - Antonio Finelli
- Division of Urology, University of Toronto, Toronto, ON, Canada
| | - Fred Saad
- Department of Surgery (Urology), University of Montreal, Montreal, QC, Canada
| | - Alan I. So
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Philippe D. Violette
- Department of Surgery, Western University, London, ON, Canada
- Departments of Surgery and Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Rodney H. Breau
- Division of Urology, University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
34
|
Carlsson SV, Murata K, Danila DC, Lilja H. PSA: role in screening and monitoring patients with prostate cancer. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00001-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
35
|
Kendrick J, Francis R, Hassan GM, Rowshanfarzad P, Jeraj R, Kasisi C, Rusanov B, Ebert M. Radiomics for Identification and Prediction in Metastatic Prostate Cancer: A Review of Studies. Front Oncol 2021; 11:771787. [PMID: 34790581 PMCID: PMC8591174 DOI: 10.3389/fonc.2021.771787] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
Metastatic Prostate Cancer (mPCa) is associated with a poor patient prognosis. mPCa spreads throughout the body, often to bones, with spatial and temporal variations that make the clinical management of the disease difficult. The evolution of the disease leads to spatial heterogeneity that is extremely difficult to characterise with solid biopsies. Imaging provides the opportunity to quantify disease spread. Advanced image analytics methods, including radiomics, offer the opportunity to characterise heterogeneity beyond what can be achieved with simple assessment. Radiomics analysis has the potential to yield useful quantitative imaging biomarkers that can improve the early detection of mPCa, predict disease progression, assess response, and potentially inform the choice of treatment procedures. Traditional radiomics analysis involves modelling with hand-crafted features designed using significant domain knowledge. On the other hand, artificial intelligence techniques such as deep learning can facilitate end-to-end automated feature extraction and model generation with minimal human intervention. Radiomics models have the potential to become vital pieces in the oncology workflow, however, the current limitations of the field, such as limited reproducibility, are impeding their translation into clinical practice. This review provides an overview of the radiomics methodology, detailing critical aspects affecting the reproducibility of features, and providing examples of how artificial intelligence techniques can be incorporated into the workflow. The current landscape of publications utilising radiomics methods in the assessment and treatment of mPCa are surveyed and reviewed. Associated studies have incorporated information from multiple imaging modalities, including bone scintigraphy, CT, PET with varying tracers, multiparametric MRI together with clinical covariates, spanning the prediction of progression through to overall survival in varying cohorts. The methodological quality of each study is quantified using the radiomics quality score. Multiple deficits were identified, with the lack of prospective design and external validation highlighted as major impediments to clinical translation. These results inform some recommendations for future directions of the field.
Collapse
Affiliation(s)
- Jake Kendrick
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, WA, Australia
| | - Roslyn Francis
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Ghulam Mubashar Hassan
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, WA, Australia
| | - Pejman Rowshanfarzad
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, WA, Australia
| | - Robert Jeraj
- Department of Medical Physics, University of Wisconsin, Madison, WI, United States
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | - Collin Kasisi
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Branimir Rusanov
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, WA, Australia
| | - Martin Ebert
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, WA, Australia
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
- 5D Clinics, Claremont, WA, Australia
| |
Collapse
|
36
|
Van Poppel H, Hogenhout R, Albers P, van den Bergh RCN, Barentsz JO, Roobol MJ. A European Model for an Organised Risk-stratified Early Detection Programme for Prostate Cancer. Eur Urol Oncol 2021; 4:731-739. [PMID: 34364829 DOI: 10.1016/j.euo.2021.06.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022]
Abstract
CONTEXT Overdiagnosis as the argument to stop prostate cancer (PCa) screening is less valid since the introduction of new technologies such as risk calculators (RCs) and magnetic resonance imaging (MRI). These new technologies result in fewer unnecessary biopsy procedures and fewer cases of both overdiagnosis and underdetection. Therefore, we can now adequately respond to the growing and urgent need for a structured risk assessment to detect PCa early. OBJECTIVE To provide expert discussion on the existing evidence for a previously published risk-stratified strategy regarding an organised population-based early detection programme for PCa. EVIDENCE ACQUISITION The proposed algorithm for early detection of PCa emerged from expert consensus by the authors based on available evidence derived from a nonsystematic review of the current literature using Medline/PubMed, Cochrane Library database, ClinicalTrials.gov, ISRCTN Registry, and the European Association of Urology guidelines on PCa. EVIDENCE SYNTHESIS Although not confirmed by the highest level of evidence, current literature and guidelines point towards an algorithm for early detection of PCa that starts with risk-based prostate-specific antigen (PSA) testing, followed by multivariable risk stratification with RCs. All men who are classified to be at intermediate and high risk are then offered prostate MRI. The combined data from RCs and MRI results can be used to select men for prostate biopsy. Low-risk men return to a risk-based safety net that includes individualised PSA-interval tests and, if necessary, repeated MRI. Depending on local availability, the use of the different risk stratification tools may be adapted. CONCLUSIONS We present a risk-stratified algorithm for an organised population-based early detection programme for clinically significant PCa. Although the proposed strategy has not yet been analysed prospectively, it exploits and may even improve the most important available benefits of "PSA-only" screening studies, while at the same time reduces unnecessary biopsies and overdiagnosis by using new risk stratification tools. PATIENT SUMMARY This paper presents a personalised strategy that enables selective early detection of prostate cancer by combining prostate-specific antigen (interval) testing' prediction models (risk calculators), and magnetic resonance imaging scans. This will likely lead to reduced prostate cancer-related morbidity and mortality, while reducing the need for prostate biopsy and limiting overdiagnosis.
Collapse
Affiliation(s)
- Hendrik Van Poppel
- Department of Development and Regeneration, University Hospital KU Leuven, Leuven, Belgium.
| | - Renée Hogenhout
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter Albers
- Department of Urology, Heinrich-Heine University Medical Faculty, Düsseldorf, Germany; Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Jelle O Barentsz
- Department of Medical Imaging, Radboudumc, Nijmegen, The Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
37
|
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
|
38
|
King B, McHugh J, Snape K. A Case-Based Clinical Approach to the Investigation, Management and Screening of Families with BRCA2 Related Prostate Cancer. APPLICATION OF CLINICAL GENETICS 2021; 14:255-266. [PMID: 34295175 PMCID: PMC8290889 DOI: 10.2147/tacg.s261737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/02/2021] [Indexed: 12/02/2022]
Abstract
BRCA2 is the most commonly implicated DNA damage repair gene associated with inherited prostate cancer. BRCA2 deficient prostate cancer typically presents at a younger age, is more poorly differentiated, and is associated with worse survival outcomes than non-BRCA2 associated prostate cancer. Despite these unfavourable prognostic implications, poly-ADP ribose polymerase inhibitors and platinum-based chemotherapy have been identified as potent targeted therapeutic agents towards BRCA1/2 deficient cancer cells. This review article explores the literature surrounding BRCA2-related prostate cancer through a familial clinical scenario. The investigation, diagnosis and management of BRCA2 deficient prostate cancer will be explored, alongside the implications of the identification of a germline pathogenic BRCA2 variant within a family, cascade screening and prostate cancer surveillance in unaffected male BRCA2 carriers. A greater understanding of the molecular pathogenesis of DNA damage repair gene deficient prostate cancer, coupled with new treatment paradigms and widened access to both somatic and germline genetic analysis for prostate cancer patients and their families will hopefully enable the robust implementation of high quality evidence-based clinical pathways for both the management and identification of BRCA2 deficient prostate cancer and improved screening, early detection and prevention strategies for individuals at increased genetic risk of prostate cancer.
Collapse
Affiliation(s)
- Bradley King
- Institute of Medical and Biomedical Education, St. George's, University of London, London, UK
| | - Jana McHugh
- Department of Oncogenomics, Institute of Cancer Research, London, UK
| | - Katie Snape
- Department of Clinical Genetics, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
39
|
da Silva MACN, Manhães VPR, Gasparotto Júnior L, Tsukumo DML, Lalli CA. Pancytopenia as an initial manifestation of prostate cancer: a case report. J Med Case Rep 2021; 15:247. [PMID: 34006332 PMCID: PMC8132390 DOI: 10.1186/s13256-021-02843-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/02/2021] [Indexed: 01/04/2023] Open
Abstract
Background Prostate adenocarcinoma is the most frequent cancer type among men, followed by skin cancer. Patients with prostate cancer usually present lower urinary tract symptoms due to tumor involvement. Bone marrow invasion is associated with prostate cancer metastasis and is common if blastic lesions in bones are present but is very rare without a large bone involvement and uncommon as initial presentation. Case presentation We present a case of an 86-year-old Caucasian man with bone marrow invasion of prostate cancer without urological or bone-related symptoms and without prostate nodules. His findings were dyspnea, fatigue, and tachycardia. We detail the complete investigation of the case until we found the accurate diagnosis. The patient started treatment, but he had no response and so the oncology team started palliative care. Conclusion Bone marrow invasion as an initial manifestation of prostate cancer is not common, especially if no prostatic lesions are found. This report is important to provide additional information about prostate cancer management.
Collapse
Affiliation(s)
- Marcos Antonio Custódio Neto da Silva
- Faculty of Medical Science, Medical Residency Program in Internal Medicine, Clinical Hospital from State University of Campinas, Rua Tessália Vieira de Camargo, 126. Cidade Universitária Zeferino Vaz. CEP, 13083-887, Campinas, Sao Paulo, Brasil.
| | - Vitor Pimentel Rodrigues Manhães
- Faculty of Medical Science, Medical Residency Program in Internal Medicine, Clinical Hospital from State University of Campinas, Rua Tessália Vieira de Camargo, 126. Cidade Universitária Zeferino Vaz. CEP, 13083-887, Campinas, Sao Paulo, Brasil
| | - Luadir Gasparotto Júnior
- Faculty of Medical Science, Medical Residency Program in Internal Medicine, Clinical Hospital from State University of Campinas, Rua Tessália Vieira de Camargo, 126. Cidade Universitária Zeferino Vaz. CEP, 13083-887, Campinas, Sao Paulo, Brasil
| | - Daniela Miti Lemos Tsukumo
- Faculty of Medical Science, Medical Residency Program in Internal Medicine, Clinical Hospital from State University of Campinas, Rua Tessália Vieira de Camargo, 126. Cidade Universitária Zeferino Vaz. CEP, 13083-887, Campinas, Sao Paulo, Brasil
| | - Cristina Alba Lalli
- Faculty of Medical Science, Medical Residency Program in Internal Medicine, Clinical Hospital from State University of Campinas, Rua Tessália Vieira de Camargo, 126. Cidade Universitária Zeferino Vaz. CEP, 13083-887, Campinas, Sao Paulo, Brasil
| |
Collapse
|
40
|
[Intelligent early prostate cancer detection in 2021: more benefit than harm]. Urologe A 2021; 60:602-609. [PMID: 33881554 DOI: 10.1007/s00120-021-01519-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
Prostate-specific antigen (PSA) is used for early detection of prostate cancer which represents the most frequent cancer diagnosed in men in Germany and Europe. Results of the largest screening trials revealed that PSA testing reduces the incidence of locally advanced and metastatic prostate cancer and shows an effect on cancer-specific mortality. However, since early diagnosis also results in overdiagnosis and overtreatment of insignificant cancers with associated morbidities, there is a need for a more individualized and risk-tailored modern strategy. The PSA at baseline is an important part of this strategy although the German Federal Joint Committee declined its financial coverage by health insurances. Available validated instruments should accompany the baseline PSA to optimize detection of clinically significant prostate cancer.
Collapse
|
41
|
Heijnsdijk EAM, Gulati R, Tsodikov A, Lange JM, Mariotto AB, Vickers AJ, Carlsson SV, Etzioni R. Lifetime Benefits and Harms of Prostate-Specific Antigen-Based Risk-Stratified Screening for Prostate Cancer. J Natl Cancer Inst 2021; 112:1013-1020. [PMID: 32067047 PMCID: PMC7566340 DOI: 10.1093/jnci/djaa001] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/23/2022] Open
Abstract
Background Studies conducted in Swedish populations have shown that men with lowest prostate-specific antigen (PSA) levels at ages 44–50 years and 60 years have very low risk of future distant metastasis or death from prostate cancer. This study investigates benefits and harms of screening strategies stratified by PSA levels. Methods PSA levels and diagnosis patterns from two microsimulation models of prostate cancer progression, detection, and mortality were compared against results of the Malmö Preventive Project, which stored serum and tracked subsequent prostate cancer diagnoses for 25 years. The models predicted the harms (tests and overdiagnoses) and benefits (lives saved and life-years gained) of PSA-stratified screening strategies compared with biennial screening from age 45 years to age 69 years. Results Compared with biennial screening for ages 45–69 years, lengthening screening intervals for men with PSA less than 1.0 ng/mL at age 45 years led to 46.8–47.0% fewer tests (range between models), 0.9–2.1% fewer overdiagnoses, and 3.1–3.8% fewer lives saved. Stopping screening when PSA was less than 1.0 ng/mL at age 60 years and older led to 12.8–16.0% fewer tests, 5.0–24.0% fewer overdiagnoses, and 5.0–13.1% fewer lives saved. Differences in model results can be partially explained by differences in assumptions about the link between PSA growth and the risk of disease progression. Conclusion Relative to a biennial screening strategy, PSA-stratified screening strategies investigated in this study substantially reduced the testing burden and modestly reduced overdiagnosis while preserving most lives saved. Further research is needed to clarify the link between PSA growth and disease progression.
Collapse
Affiliation(s)
- Eveline A M Heijnsdijk
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Institute, Seattle, WA, USA
| | - Alex Tsodikov
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jane M Lange
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Institute, Seattle, WA, USA
| | - Angela B Mariotto
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, MD, USA
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sigrid V Carlsson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Institute, Seattle, WA, USA
| |
Collapse
|
42
|
Al-Monajjed R, Arsov C, Albers P. [Risk-adapted prostate cancer screening-update 2021]. Urologe A 2021; 60:592-601. [PMID: 33792743 DOI: 10.1007/s00120-021-01505-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common cancer and the second leading cause of cancer death in men in industrialized countries. There is no commonly accepted prostate cancer screening strategy. Based on the experience of various international screening studies, the German Prostate Cancer Early Detection Study Based on a Baseline PSA Value in Young Men (PROBASE) was established in 2014. OBJECTIVE Based on the positive results of retrospective cohort analyses, the PROBASE study is designed to demonstrate that a screening strategy based on risk stratification by a baseline prostate-specific antigen (PSA) level at age 45 or 50 years may be an alternative to population-based screening. PROBASE is presented in the context of other risk-adapted screening studies. MATERIALS AND METHODS There are basically several approaches to improve the population-based screening of PCa. Known risk factors for prostate cancer are age, a certain genetic predisposition (BRCA 1/2) and other germline mutations as well as individual somatic mutations. RESULTS A total of 23,301 participants were randomized to study arm A. Baseline PSA testing in study arm A categorized 89.18% of participants into the low-risk group, 9.32% into the intermediate-risk group, and 1.48% into the high-risk group. Thus, the risk assignment exactly matched the previously reported distribution. DISCUSSION Baseline PSA-dependent, risk-adapted PSA screening has the potential to reduce the high incidence of overdiagnosis and ultimately overtreatment of insignificant prostate cancers of population-based screening through extended testing intervals in the low-risk group. In parallel with PROBASE, several risk-adapted screening strategies are currently being tested worldwide; the evaluation of which is also awaited in several years.
Collapse
Affiliation(s)
- R Al-Monajjed
- Medizinische Fakultät, Universitätsklinikum Düsseldorf, Klinik für Urologie, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland.
| | - C Arsov
- Medizinische Fakultät, Universitätsklinikum Düsseldorf, Klinik für Urologie, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - P Albers
- Medizinische Fakultät, Universitätsklinikum Düsseldorf, Klinik für Urologie, Heinrich-Heine-Universität Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| |
Collapse
|
43
|
Galosi AB, Palagonia E, Scarcella S, Cimadamore A, Lacetera V, Delle Fave RF, Antezza A, Dell'Atti L. Detection limits of significant prostate cancer using multiparametric MR and digital rectal examination in men with low serum PSA: Up-date of the Italian Society of Integrated Diagnostic in Urology. ACTA ACUST UNITED AC 2021; 93:92-100. [PMID: 33754619 DOI: 10.4081/aiua.2021.1.92] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 11/22/2022]
Abstract
Reasons why significant prostate cancer is still missed in early stage were investigated at the 22nd National SIEUN (Italian Society of integrated diagnostic in Urology, Andrology, Nephrology) congress took place from 30th November to 1st December 2020, in virtual modality. Even if multiparametric magnetic resonance (MR) has been introduced in the clinical practice several, limitations are emerging in patient with regular digital rectal examination (DRE) and serum prostate specific antigen (PSA) levels approaching the normal limits. The present paper summarizes highlights observed in those cases where significant prostate cancer may be missed by PSA or imaging and DRE. The issue of multidisciplinary interest had been subdivided and deepened under four main topics: biochemical, clinical, pathological and radiological point of view with a focus on PI-RADS 3 lesions.
Collapse
Affiliation(s)
- Andrea B Galosi
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Erika Palagonia
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Simone Scarcella
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Alessia Cimadamore
- Division of Pathology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Vito Lacetera
- Division of Urology, Azienda Ospedaliera Marche Nord, Pesaro.
| | - Rocco F Delle Fave
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Angelo Antezza
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| | - Lucio Dell'Atti
- Division of Urology, School of Medicine, Università Politecnica delle Marche, Ancona.
| |
Collapse
|
44
|
Shah A, Polascik TJ, George DJ, Anderson J, Hyslop T, Ellis AM, Armstrong AJ, Ferrandino M, Preminger GM, Gupta RT, Lee WR, Barrett NJ, Ragsdale J, Mills C, Check DK, Aminsharifi A, Schulman A, Sze C, Tsivian E, Tay KJ, Patierno S, Oeffinger KC, Shah K. Implementation and Impact of a Risk-Stratified Prostate Cancer Screening Algorithm as a Clinical Decision Support Tool in a Primary Care Network. J Gen Intern Med 2021; 36:92-99. [PMID: 32875501 PMCID: PMC7858708 DOI: 10.1007/s11606-020-06124-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/07/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Implementation methods of risk-stratified cancer screening guidance throughout a health care system remains understudied. OBJECTIVE Conduct a preliminary analysis of the implementation of a risk-stratified prostate cancer screening algorithm in a single health care system. DESIGN Comparison of men seen pre-implementation (2/1/2016-2/1/2017) vs. post-implementation (2/2/2017-2/21/2018). PARTICIPANTS Men, aged 40-75 years, without a history of prostate cancer, who were seen by a primary care provider. INTERVENTIONS The algorithm was integrated into two components in the electronic health record (EHR): in Health Maintenance as a personalized screening reminder and in tailored messages to providers that accompanied prostate-specific antigen (PSA) results. MAIN MEASURES Primary outcomes: percent of men who met screening algorithm criteria; percent of men with a PSA result. Logistic repeated measures mixed models were used to test for differences in the proportion of individuals that met screening criteria in the pre- and post-implementation periods with age, race, family history, and PSA level included as covariates. KEY RESULTS During the pre- and post-implementation periods, 49,053 and 49,980 men, respectively, were seen across 26 clinics (20.6% African American). The proportion of men who met screening algorithm criteria increased from 49.3% (pre-implementation) to 68.0% (post-implementation) (p < 0.001); this increase was observed across all races, age groups, and primary care clinics. Importantly, the percent of men who had a PSA did not change: 55.3% pre-implementation, 55.0% post-implementation. The adjusted odds of meeting algorithm-based screening was 6.5-times higher in the post-implementation period than in the pre-implementation period (95% confidence interval, 5.97 to 7.05). CONCLUSIONS In this preliminary analysis, following implementation of an EHR-based algorithm, we observed a rapid change in practice with an increase in screening in higher-risk groups balanced with a decrease in screening in low-risk groups. Future efforts will evaluate costs and downstream outcomes of this strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ariel Schulman
- Duke University, Durham, NC, USA.,Maimonides Medical Center, New York, NY, USA
| | - Christina Sze
- Duke University, Durham, NC, USA.,Weill Cornell Medical College, New York, NY, USA
| | | | - Kae Jack Tay
- Duke University, Durham, NC, USA.,SingHealth, Duke-NUS, Singapore, Singapore
| | | | | | | |
Collapse
|
45
|
Zavala VA, Bracci PM, Carethers JM, Carvajal-Carmona L, Coggins NB, Cruz-Correa MR, Davis M, de Smith AJ, Dutil J, Figueiredo JC, Fox R, Graves KD, Gomez SL, Llera A, Neuhausen SL, Newman L, Nguyen T, Palmer JR, Palmer NR, Pérez-Stable EJ, Piawah S, Rodriquez EJ, Sanabria-Salas MC, Schmit SL, Serrano-Gomez SJ, Stern MC, Weitzel J, Yang JJ, Zabaleta J, Ziv E, Fejerman L. Cancer health disparities in racial/ethnic minorities in the United States. Br J Cancer 2021; 124:315-332. [PMID: 32901135 PMCID: PMC7852513 DOI: 10.1038/s41416-020-01038-6] [Citation(s) in RCA: 488] [Impact Index Per Article: 162.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 07/16/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
There are well-established disparities in cancer incidence and outcomes by race/ethnicity that result from the interplay between structural, socioeconomic, socio-environmental, behavioural and biological factors. However, large research studies designed to investigate factors contributing to cancer aetiology and progression have mainly focused on populations of European origin. The limitations in clinicopathological and genetic data, as well as the reduced availability of biospecimens from diverse populations, contribute to the knowledge gap and have the potential to widen cancer health disparities. In this review, we summarise reported disparities and associated factors in the United States of America (USA) for the most common cancers (breast, prostate, lung and colon), and for a subset of other cancers that highlight the complexity of disparities (gastric, liver, pancreas and leukaemia). We focus on populations commonly identified and referred to as racial/ethnic minorities in the USA-African Americans/Blacks, American Indians and Alaska Natives, Asians, Native Hawaiians/other Pacific Islanders and Hispanics/Latinos. We conclude that even though substantial progress has been made in understanding the factors underlying cancer health disparities, marked inequities persist. Additional efforts are needed to include participants from diverse populations in the research of cancer aetiology, biology and treatment. Furthermore, to eliminate cancer health disparities, it will be necessary to facilitate access to, and utilisation of, health services to all individuals, and to address structural inequities, including racism, that disproportionally affect racial/ethnic minorities in the USA.
Collapse
Affiliation(s)
- Valentina A Zavala
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - John M Carethers
- Departments of Internal Medicine and Human Genetics, and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Luis Carvajal-Carmona
- University of California Davis Comprehensive Cancer Center and Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
- Genome Center, University of California Davis, Davis, CA, USA
| | | | - Marcia R Cruz-Correa
- Department of Cancer Biology, University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Melissa Davis
- Division of Breast Surgery, Department of Surgery, NewYork-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | - Adam J de Smith
- Center for Genetic Epidemiology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Julie Dutil
- Cancer Biology Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Jane C Figueiredo
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rena Fox
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kristi D Graves
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Scarlett Lin Gomez
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Andrea Llera
- Laboratorio de Terapia Molecular y Celular, IIBBA, Fundación Instituto Leloir, CONICET, Buenos Aires, Argentina
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Lisa Newman
- Division of Breast Surgery, Department of Surgery, NewYork-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
- Interdisciplinary Breast Program, New York-Presbyterian/Weill Cornell Medical Center, New York, NY, USA
| | - Tung Nguyen
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, Boston, MA, USA
| | - Nynikka R Palmer
- Department of Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, San Francisco, CA, USA
| | - Eliseo J Pérez-Stable
- Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Office of the Director, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Sorbarikor Piawah
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Erik J Rodriquez
- Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Stephanie L Schmit
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Silvia J Serrano-Gomez
- Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Mariana C Stern
- Departments of Preventive Medicine and Urology, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey Weitzel
- Department of Population Sciences, Beckman Research Institute of City of Hope, Duarte, CA, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Jun J Yang
- Department of Pharmaceutical Sciences, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center LSUHSC, New Orleans, LA, USA
| | - Elad Ziv
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Laura Fejerman
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
46
|
Borque-Fernando A, Espílez R, Miramar D, Corbatón D, Rodríguez A, Castro E, Mateo J, Rello L, Méndez A, Gil Sanz MJ. Genetic counseling in prostate cancer: How to implement it in daily clinical practice? Actas Urol Esp 2021; 45:8-20. [PMID: 33059945 DOI: 10.1016/j.acuro.2020.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/22/2020] [Indexed: 11/16/2022]
Abstract
Prostate cancer plays an undeniably prominent role in public health in our days and health systems. Its epidemiological impact is quantitatively very close to that of other tumors such as colon cancer and breast cancer, in which genetic counseling is part of their routine clinical practice, both in the initial evaluation and in the selection of therapeutic strategies. Hereditary cancer syndromes, breast/ovarian and Lynch syndrome are part of genetic counseling in these tumors. Currently, we also know that they can be associated to prostate cancer. The time has come to implement genetic counseling in prostate cancer from the earliest stages of its approach, from initial suspicion to the most advanced tumors. We present an updated review carried out by our interdisciplinary working group on scientific literature, clinical practice guidelines and consensus documents, aimed at the creation and drafting of a'Protocol for genetic counseling in prostate cancer' for the study of germline, with easy application in different healthcare settings. This protocol is currently being implemented in our routine practice and provides answers to 3 specific questions: Who should receive genetic counseling for prostate cancer? Which gene panel should be analyzed? How should counseling be done according to the results obtained? Other aspects about who should perform genetic counseling, ethical considerations and regulations are also collected.
Collapse
Affiliation(s)
- A Borque-Fernando
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España.
| | - R Espílez
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - D Miramar
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - D Corbatón
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| | - A Rodríguez
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - E Castro
- Departamento de Oncología Médica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga, Málaga, España
| | - J Mateo
- Instituto de Oncología Vall d'Hebron y Hospital Universitario Vall d'Hebron, Barcelona, España
| | - L Rello
- Servicio de Bioquímica, Unidad de Genética, Hospital Universitario Miguel Servet, Zaragoza, España
| | - A Méndez
- Servicio de Oncología Radioterápica, Hospital Universitario Miguel Servet, Zaragoza, España
| | - M J Gil Sanz
- Servicio de Urología, Hospital Universitario Miguel Servet, IIS-Aragón, Zaragoza, España
| |
Collapse
|
47
|
Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, Grivas N, Grummet J, Henry AM, van der Kwast TH, Lam TB, Lardas M, Liew M, Mason MD, Moris L, Oprea-Lager DE, van der Poel HG, Rouvière O, Schoots IG, Tilki D, Wiegel T, Willemse PPM, Cornford P. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer-2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur Urol 2020; 79:243-262. [PMID: 33172724 DOI: 10.1016/j.eururo.2020.09.042] [Citation(s) in RCA: 1578] [Impact Index Per Article: 394.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To present a summary of the 2020 version of the European Association of Urology (EAU)-European Association of Nuclear Medicine (EANM)-European Society for Radiotherapy and Oncology (ESTRO)-European Society of Urogenital Radiology (ESUR)-International Society of Geriatric Oncology (SIOG) guidelines on screening, diagnosis, and local treatment of clinically localised prostate cancer (PCa). EVIDENCE ACQUISITION The panel performed a literature review of new data, covering the time frame between 2016 and 2020. The guidelines were updated and a strength rating for each recommendation was added based on a systematic review of the evidence. EVIDENCE SYNTHESIS A risk-adapted strategy for identifying men who may develop PCa is advised, generally commencing at 50 yr of age and based on individualised life expectancy. Risk-adapted screening should be offered to men at increased risk from the age of 45 yr and to breast cancer susceptibility gene (BRCA) mutation carriers, who have been confirmed to be at risk of early and aggressive disease (mainly BRAC2), from around 40 yr of age. The use of multiparametric magnetic resonance imaging in order to avoid unnecessary biopsies is recommended. When a biopsy is performed, a combination of targeted and systematic biopsies must be offered. There is currently no place for the routine use of tissue-based biomarkers. Whilst prostate-specific membrane antigen positron emission tomography computed tomography is the most sensitive staging procedure, the lack of outcome benefit remains a major limitation. Active surveillance (AS) should always be discussed with low-risk patients, as well as with selected intermediate-risk patients with favourable International Society of Urological Pathology (ISUP) 2 lesions. Local therapies are addressed, as well as the AS journey and the management of persistent prostate-specific antigen after surgery. A strong recommendation to consider moderate hypofractionation in intermediate-risk patients is provided. Patients with cN1 PCa should be offered a local treatment combined with long-term hormonal treatment. CONCLUSIONS The evidence in the field of diagnosis, staging, and treatment of localised PCa is evolving rapidly. The 2020 EAU-EANM-ESTRO-ESUR-SIOG guidelines on PCa summarise the most recent findings and advice for their use in clinical practice. These PCa guidelines reflect the multidisciplinary nature of PCa management. PATIENT SUMMARY Updated prostate cancer guidelines are presented, addressing screening, diagnosis, and local treatment with curative intent. These guidelines rely on the available scientific evidence, and new insights will need to be considered and included on a regular basis. In some cases, the supporting evidence for new treatment options is not yet strong enough to provide a recommendation, which is why continuous updating is important. Patients must be fully informed of all relevant options and, together with their treating physicians, decide on the most optimal management for them.
Collapse
Affiliation(s)
- Nicolas Mottet
- Department of Urology, University Hospital, St. Etienne, France.
| | | | | | | | | | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Stefano Fanti
- Department of Nuclear Medicine, Policlinico S. Orsola, University of Bologna, Italy
| | - Nicola Fossati
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Giorgio Gandaglia
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Nikos Grivas
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeremy Grummet
- Department of Surgery, Central Clinical School, Monash University, Caulfield North, Victoria, Australia
| | - Ann M Henry
- Leeds Cancer Centre, St. James's University Hospital and University of Leeds, Leeds, UK
| | | | - Thomas B Lam
- Academic Urology Unit, University of Aberdeen, Aberdeen, UK; Department of Urology, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Michael Lardas
- Department of Urology, Metropolitan General Hospital, Athens, Greece
| | - Matthew Liew
- Department of Urology, Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, UK
| | - Malcolm D Mason
- Division of Cancer and Genetics, School of Medicine Cardiff University, Velindre Cancer Centre, Cardiff, UK
| | - Lisa Moris
- Department of Urology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU Medical Center, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Olivier Rouvière
- Hospices Civils de Lyon, Department of Urinary and Vascular Imaging, Hôpital Edouard Herriot, Lyon, France; Faculté de Médecine Lyon Est, Université de Lyon, Université Lyon 1, Lyon, France
| | - Ivo G Schoots
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
| | - Peter-Paul M Willemse
- Department of Urology, Cancer Center University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
48
|
Abstract
This article gives an overview of the current state of the evidence for prostate cancer early detection with prostate-specific antigen (PSA) and summarizes current recommendations from guideline groups. The article reviews the global public health burden and risk factors for prostate cancer with clinical implications as screening tools. Screening studies, novel biomarkers, and MRI are discussed. The article outlines 7 key practice points for primary care physicians and provides a simple schema for facilitating shared decision-making conversations.
Collapse
Affiliation(s)
- Sigrid V Carlsson
- Department of Surgery (Urology Service), Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Urology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Andrew J Vickers
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, New York, NY 10065, USA.
| |
Collapse
|
49
|
Recommandations françaises du Comité de cancérologie de l’AFU – actualisation 2020–2022 : cancer de la prostate. Prog Urol 2020; 30:S136-S251. [DOI: 10.1016/s1166-7087(20)30752-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
French WW, Wallen EM. Advances in the diagnostic options for prostate cancer. Postgrad Med 2020; 132:52-62. [PMID: 32900250 DOI: 10.1080/00325481.2020.1822067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Over the past decade, despite the controversies surrounding prostate cancer screening, significant refinements have improved its application. PSA screening, although it has been questioned, appears to confer a mortality benefit and remains the most effective way to identify the possible presence of prostate cancer. Methods to improve the specificity of PSA screening and limit overdiagnosis of indolent cancers, including risk-stratified screening regimens, are currently being utilized. Certain imaging modalities, such as multiparametric MRI, have proven to be excellent adjuncts providing improved risk stratification and the ability for targeted biopsies; however, concerns over variability in interpretation and generalizability persist. A number of novel biomarkers have become available with nearly all demonstrating the ability to improve upon the specificity of PSA screening; however, optimal timing, direct comparisons, and usefulness in conjunction with imaging modalities remain to be elucidated. With the improvement in testing options and recognition of the risk/benefit ratio for men undergoing screening for prostate cancer, the increasing role of shared decision making in the process is emphasized.
Collapse
Affiliation(s)
- William W French
- Department of Urology, University of North Carolina Medical Center , Chapel Hill, NC, United States
| | - Eric M Wallen
- Department of Urology, University of North Carolina Medical Center , Chapel Hill, NC, United States
| |
Collapse
|