Evidence Summary

Prostate Cancer: Screening

May 08, 2018

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

By Joshua J. Fenton, MD, MPH; Meghan S. Weyrich, MPH; Shauna Durbin, MPH; Yu Liu, MS; Heejung Bang, PhD; and Joy Melnikow, MD, MPH

The information in this article is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This article is intended as a reference and not as a substitute for clinical judgment.

This article may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied.

This article was first published in JAMA on May 8, 2018.

 

Return to Table of Contents

Importance: Prostate cancer is the second leading cause of cancer death among US men.

Objective: To systematically review evidence on prostate-specific antigen (PSA)–based prostate cancer screening, treatments for localized prostate cancer, and prebiopsy risk calculators to inform the US Preventive Services Task Force.

Data Sources: Searches of PubMed, EMBASE, Web of Science, and Cochrane Registries and Databases from July 1, 2011, through July 15, 2017, with a surveillance search on February 1, 2018.

Study Selection: English-language reports of randomized clinical trials (RCTs) of screening; cohort studies reporting harms; RCTs and cohort studies of active localized cancer treatments vs conservative approaches (eg, active surveillance, watchful waiting); external validations of prebiopsy risk calculators to identify aggressive cancers.

Data Extraction and Synthesis: One investigator abstracted data; a second checked accuracy. Two investigators independently rated study quality.

Main Outcomes and Measures: Prostate cancer and all-cause mortality; false-positive screening results, biopsy complications, overdiagnosis; adverse effects of active treatments. Random-effects meta-analyses were conducted for treatment harms.

Results: Sixty-three studies in 104 publications were included (N = 1,904,950). Randomization to PSA screening was not associated with reduced risk of prostate cancer mortality in either a US trial with substantial control group contamination (n = 76,683) or a UK trial with low adherence to a single PSA screen (n = 408,825) but was associated with significantly reduced prostate cancer mortality in a European trial (n = 162,243; relative risk [RR], 0.79 [95% CI, 0.69-0.91]; absolute risk reduction, 1.1 deaths per 10,000 person-years [95% CI, 0.5-1.8]). Of 61,604 men screened in the European trial, 17.8% received false-positive results. In 3 cohorts (n = 15,136), complications requiring hospitalization occurred in 0.5% to 1.6% of men undergoing biopsy after abnormal screening findings. Overdiagnosis was estimated to occur in 20.7% to 50.4% of screen-detected cancers. In an RCT of men with screen-detected prostate cancer (n = 1643), neither radical prostatectomy (hazard ratio [HR], 0.63 [95% CI, 0.21-1.93]) nor radiation therapy (HR, 0.51 [95% CI, 0.15-1.69]) were associated with significantly reduced prostate cancer mortality vs active monitoring, although each was associated with significantly lower risk of metastatic disease. Relative to conservative management, radical prostatectomy was associated with increased risk of urinary incontinence (pooled RR, 2.27 [95% CI, 1.82-2.84]; 3 trials; n = 1796) and erectile dysfunction (pooled RR, 1.82 [95% CI, 1.62-2.04]; 2 trials; n = 883). Relative to conservative management (8 cohort studies; n = 3066), radiation therapy was associated with increased risk of erectile dysfunction (pooled RR, 1.31 [95% CI, 1.20-1.42]).

Conclusions and Relevance: PSA screening may reduce prostate cancer mortality risk but is associated with false-positive results, biopsy complications, and overdiagnosis. Compared with conservative approaches, active treatments for screen-detected prostate cancer have unclear effects on long-term survival but are associated with sexual and urinary difficulties.

Return to Table of Contents

Prostate cancer is the most commonly diagnosed cancer in US men and the second leading cause of cancer death.1 It has been estimated that in 2018, approximately 165,000 US men will be diagnosed with prostate cancer and 29,000 men will die of prostate cancer.2 Prostate cancer incidence is 74% greater among African American than white men1 and is also relatively greater in men with vs men without a family history of prostate cancer.3

US prostate cancer incidence increased sharply with the dissemination of prostate-specific antigen (PSA)–based screening beginning in the late 1980s.4 In 2012, the US Preventive Services Task Force (USPSTF) recommended against PSA-based screening for prostate cancer, concluding that there was moderate certainty that the benefits of screening do not outweigh the harms (D recommendation). New evidence has since emerged from screening and treatment trials, and among US men prostate cancer is increasingly managed with active surveillance, in which treatment is deferred indefinitely unless evidence of progression is found during periodic physical examination, PSA–based testing, or repeat biopsy. This systematic review of screening and treatment benefits and harms and whether prebiopsy risk calculators can reliably detect higher-risk prostate cancers, along with a review of decision modeling studies,5 was conducted to inform the USPSTF in its update of the 2012 recommendation regarding PSA-based screening for prostate cancer.

Return to Table of Contents

Scope of Review

This review addressed 5 key questions (KQs) (Figure 1) encompassing the benefits and harms of PSA screening (KQ1 and KQ2), benefits and harms of treatments for localized prostate cancer (KQ3 and KQ4), and the utility of prebiopsy risk calculators to identify men with higher-risk prostate cancers (KQ5). Results addressing primary key questions are summarized here, while results for subquestions 2a, 3a, 4a, 4b, and 5a, as well as additional methodological details regarding search strategies, study inclusion criteria, quality assessment, excluded studies, and data analyses, are publicly available at https://www.uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/prostate-cancer-screening1.

Data Sources and Searches

MEDLINE, PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials were searched to locate studies informing the key questions that were published since the end of the search periods for the 2011 USPSTF reviews7,8 (July 1, 2011, through July 15, 2017). Database searches were supplemented with expert suggestions and by reviewing reference lists from relevant systematic reviews and prior USPSTF reports. KQ5, on prebiopsy risk calculators, was new to this review; a preliminary search revealed that the earliest article in this field was published in 2006, so databases were searched from January 1, 2006, through October 6, 2016, for KQ5. Since October 2016, ongoing surveillance continued through article alerts and targeted searches of high-impact journals to identify studies published that could affect the conclusions or the related USPSTF recommendation. The last surveillance search was conducted in February 2018; during ongoing surveillance, we identified extended follow-up from a treatment trial,9 2 cohort studies reporting longitudinal treatment outcomes,10,11 and 3 studies of multivariable risk calculators.12-14 We also included a recently published large screening trial.15

Study Selection

For KQ1, randomized clinical trials (RCTs) of asymptomatic men undergoing PSA screening vs no screening were included that assessed cancer incidence, prostate cancer–related morbidity, prostate cancer–specific mortality, or all-cause mortality. For KQ2, RCTs and cohort studies of asymptomatic men undergoing PSA screening or prostate biopsy after abnormal screening results were included that assessed the frequency of false-positive PSA screening, physical or psychological harms of screening or biopsy, or health-related quality of life. As has been performed for breast and lung cancer screening,16-19 extra-incidence data from trials were used to estimate the percentage of men diagnosed with cancer in the screening groups who were overdiagnosed. In the absence of overdiagnosis, the number of cases in the control group of a screening trial would be expected to eventually catch up with the number of cases diagnosed in the screening group; therefore, an excess of cases in the screening group with extended follow-up implies overdiagnosis.

For KQ3 and KQ4, RCTs and comparative cohort studies of men with localized prostate cancer (stages T1-T2) were included that reported prostate cancer–specific morbidity or mortality, all-cause mortality (KQ3), or physical or psychological harms of treatment, including adverse quality-of-life effects (KQ4). Studies were required to compare outcomes among men receiving active treatments (including radical prostatectomy, radiation therapy, androgen deprivation therapy, cryotherapy, or high-intensity focused ultrasound) and men receiving conservative management (ie, watchful waiting, active surveillance, observation, or no treatment). Because few studies were found for treatments other than radical prostatectomy and radiation therapy, only findings for radical prostatectomy and radiation therapy are summarized in this article; evidence on other treatment modalities is reviewed in the full evidence report. For KQ4 (treatment harms), uncontrolled observational studies with sample sizes of at least 100 men were also included.

For KQ5, external validation studies of multivariable risk calculators were included if they predicted the presence of “significant” prostate cancer using PSA testing in addition to patient variables routinely available before prostate biopsy (eg, patient characteristics, rectal examination results, PSA level). Significant prostate cancers were defined as either high grade (Gleason score ≥7) or clinical stage T2b or higher. Studies of novel serum biomarkers or imaging studies were excluded because results from these studies would not be routinely available before biopsy in most urology practices.

Data Extraction and Quality Assessment

Two reviewers independently appraised the quality of included articles using predefined criteria,6,20-23 with disagreements resolved by consensus or consultation with a third investigator. One reviewer extracted study-level data into standardized evidence tables; a second checked data accuracy. Included studies were limited to those published in English and were rated as fair or good quality using USPSTF quality rating standards. Poor-quality studies contained a fatal flaw or multiple significant limitations that could invalidate the results. Three poor-quality RCTs of screening PSA were excluded.24-26 Limitations of these studies included inadequate statistical power,24-26 faulty analysis,25 and potentially biased outcomes assessment.24-26

Data Synthesis and Analysis

For each key question, the study designs, population characteristics, screening and treatment details, and overall results were summarized using descriptive statistics. Pooled meta-analyses were not performed for outcomes of screening effectiveness (KQ1), screening harms (KQ2), or treatment effectiveness (KQ3), because data on these outcomes derived from few studies with variable populations and interventions. For KQ4, random-effects meta-analyses were performed using the method of DerSimonian and Laird to estimate pooled relative risks (RRs) of urinary incontinence (ie, daily use of a pad or worse) and erectile dysfunction (ie, erections insufficient for intercourse) among patients receiving radical prostatectomy or radiation therapy vs conservative management.

Statistical heterogeneity across studies was estimated using the I2 statistic. Meta-analyses were performed using Stata version 14.2 (StataCorp). Pooled RRs were used to estimate the number of patients needed to be treated for 1 patient to be harmed (number needed to harm); in these calculations, the absolute risk of the harm in actively treated patients was estimated as the product of the pooled RR and the absolute risk among all men included in the conservative management control groups of included studies. Statistical tests were 2-sided, with P < 0.05 indicating statistical significance.

Return to Table of Contents

Two reviewers independently assessed 4105 unique citations and 303 full-text articles for inclusion (Figure 2). Overall, 24 articles (3 studies) were included for KQ1, 15 articles (8 studies) were included for KQ2, 23 articles (13 studies) were included for KQ3, 38 articles (31 studies) were included for KQ4, and 14 articles (14 studies) were included for KQ5.

Effectiveness of PSA-Based Screening

Key Question 1. Is there direct evidence that PSA-based screening for prostate cancer reduces short- or long-term prostate cancer morbidity and mortality and all-cause mortality?

Key Question 1a. Does the effectiveness of PSA-based screening vary by subpopulation or risk factor (eg, age, race/ethnicity, family history, or clinical risk assessment)?

Three fair-quality RCTs (n = 647,906) assessed the effect of PSA screening on prostate cancer morbidity and mortality and all-cause mortality: the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial,27 the European Randomized Study of Screening for Prostate Cancer (ERSPC),28 and the Cluster Randomized Trial of PSA Testing for Prostate Cancer (CAP)15 (Table 1). Since the previous USPSTF review, the ERSPC30 and PLCO29 trials each reported results of extended follow-up; recent reports from 4 ERSPC sites (Göteborg-Sweden,32 Finland,37 the Netherlands,35 and Spain34) were also included.

In the PLCO trial, 76,683 US men aged 55 to 74 years were recruited from 1993 to 2001 and randomized to either annual PSA screening for 6 years or usual care. When men had PSA levels of 4.0 ng/mL or greater, they and their clinicians were informed; community physicians coordinated diagnostic evaluations and treatments.The PLCO trial has been characterized as comparing the effectiveness of organized vs opportunistic screening,40 because during the screening phase of the trial, approximately 46% of control group participants received routine screening PSA testing from community physicians during each year, compared with approximately 85% of men in the screening group.41 PSA testing was also common in both trial groups during the 7 years after the screening phase.

In the ERSPC trial, 181,999 men aged 50 to 74 years were enrolled at 7 European centers from 1993 to 2003, although primary outcomes were reported among 162,243 men aged 55 to 69 years at randomization. The ERSPC screening interval was 4 years at all sites except in Sweden, which used a 2-year interval; threshold prompting biopsy recommendation was most commonly 3 ng/mL but varied across sites and over time within some sites. Contamination was not systematically assessed at all ERSPC sites, although 19.4% of control group participants had a screening PSA test during the screening phase at the Netherlands site.36 In the CAP trial, 408,825 UK men aged 50 to 69 years were randomized by primary care practice cluster (n = 573) to invitation (or no invitation) to a single PSA screen conducted from 2001 to 2009.15 After invitation, 34% of men obtained a valid PSA screening test. Although PSA screening was not routine in UK practice during the study period, from 2002 to 2011, UK men aged 45 to 69 years had a 39.2% cumulative risk over 10 years of receiving at least 1 PSA test for screening or diagnostic purposes.42

Prostate Cancer Incidence

Cumulative incidences of prostate cancer in the screening and control groups were 11.1% and 9.9%, respectively, at 13 years of median follow-up in the PLCO trial (RR, 1.12 [95% CI, 1.07-1.17]), 10.2% and 6.0% at 13 years of median follow-up in the ERSPC trial (RR, 1.57 [95% CI, 1.51-1.62]), and 4.3% and 3.6% at 10 years of median follow-up in the CAP trial (RR, 1.19 [95% CI, 1.14-1.25]) (Table 1). Observed risk differences in prostate cancer incidence indicate a number needed to invite of 84 men in the PLCO trial (95% CI, 59- 144), 26 men in the ERSPC trial (95% CI, 24-29), and 154 men in the CAP trial (95% CI, 128-192) for 1 additional man to be diagnosed with prostate cancer.15,30,39

Cumulative Incidence of Metastatic Disease

Within 4 ERSPC sites, the cumulative incidence of metastatic cancer (ie, metastases on imaging or a PSA level >100 ng/mL) at a median follow-up of 12.0 years was lower among men randomized to screening compared with control (RR, 0.70 [95% CI, 0.60 to 0.82]), and randomization to screening was associated with an absolute reduction in long-term risk of metastatic prostate cancer of 3.1 cases per 1000 men.43 After accounting for the 55.6% relative increase in prostate cancer incidence in the screening vs control groups at these sites, the number needed to invite to avoid 1 case of metastatic cancer was 323 (95% CI, 227-556), and the number needed to diagnose with prostate cancer through screening to avert 1 case of metastatic cancer was 12 (95% CI, 9-21).43

Prostate Cancer Mortality

At a median follow-up of 14.8 years in the PLCO trial, the prostate cancer–specific mortality rate was 4.8 per 10,000 person-years among men in the intervention group and 4.6 per 10,00 person-years among men in the control group (RR, 1.04 [95% CI, 0.87- 1.24]) (Table 1).29 In the CAP trial, at a median follow-up of 10 years, prostate cancer–specific mortality was 3.0 per 10,000 person-years among men invited to screening and 3.1 per 10,000 person-years among men in the control group (RR, 0.96 [95% CI, 0.85-1.08]). Among men in the ERSPC core age group after a median of 13 years of follow-up, prostate cancer–specific mortality was 4.3 per 10,000 person-years in the intervention group and 5.4 per 10,000 person-years in control group (RR, 0.79 [95% CI, 0.69-0.91]; P = 0.001).30 In the ERSPC trial, the absolute risk reduction in prostate cancer mortality associated with screening was 1.1 deaths per 10,000 person-years (95% CI, 0.5-1.8), or 1.3 fewer prostate cancer deaths per 1000 men; the number needed to invite to prevent 1 prostate cancer death was 781 (95% CI, 490-1929), and the number needed to diagnose was 27 (95% CI, 17-66). Across all ERSPC sites, prostate cancer mortality was statistically significantly reduced only at the sites in the Netherlands and Sweden, although point estimates were in favor of screening at all sites except Switzerland.

Randomization to screening was not associated with statistically significant reductions in prostate cancer mortality among men aged 65 to 74 years at baseline in the PLCO trial (RR, 1.02 [95% CI, 0.77-1.37])29 or among men aged 70 to 74 years at baseline in the ERSPC trial (RR, 1.17 [95% CI, 0.82-1.66]).30 No reports on differences in outcomes by race/ethnicity within the PLCO or ERSPC trials were found, and the sample size of 3370 non-Hispanic black men randomized in the PLCO trial (4.4% of the overall population) was relatively small. Among white men in the PLCO trial who reported of a family history of prostate cancer (n = 4833), the multivariable-adjusted hazard ratio for prostate cancer death among men randomized to screening was lower relative to control participants but was not statistically significant (HR, 0.49 [95% CI, 0.22-1.10]).44

All-Cause Mortality

After median follow-up periods ranging from 10 years in the CAP trial to 14.8 years in the PLCO trial, randomization to screening (relative to control) was not associated with statistically significantly reduced all-cause mortality in any of the 3 trials (CAP trial: RR, 0.99 [95% CI, 0.94-1.03]; ERSPC trial: RR, 1.00 [95% CI, 0.98-1.02]; PLCO trial: RR, 0.98 [95% CI, 0.95-1.00]) (Table 1).15,29,30

Harms of Screening

Key Question 2. What are the harms of PSA-based screening for prostate cancer and diagnostic follow-up?

Evidence on the harms of PSA screening and diagnostic follow-up derives from the PLCO, CAP, and ERSPC trials (n = 647,906), as well as 2 good-quality and 3 fair-quality cohort studies (n = 297,971).15,27,29,30,45-55

False-Positive Testing and Diagnostic Biopsies

Among men who underwent at least 1 PSA screen during the initial 4 (of 6) PLCO screening rounds (n = 32,576), 10.4% received at least 1 false-positive PSA screening result,56 compared with 17.8% of men who were screened at least once within 5 ERSPC centers (n = 61,604).50 Across all PLCO screening rounds, 12.6% of men randomized to screening underwent 1 or more biopsies, resulting in a total of 6295 biopsies (16.4 biopsies per 100 men randomized to screening). Within ERSPC, the rate of biopsies among men randomized to screening was higher (27.7 biopsies per 100 men). Of biopsies performed in the 3 trials, 67.7%, 75.8%, and 60.6% did not result in a prostate cancer diagnosis in the PLCO, ERSPC, and CAP trials, respectively.15,30,52

Among men undergoing a single round of PSA screening within the Veterans Affairs health system (n = 295,645), 8.5% had a PSA level of 4 ng/mL or greater; of these, 32.9% underwent subsequent prostate biopsy.55 Of biopsies performed, 37.2% did not result in a prostate cancer diagnosis.

Physical and Psychological Harms of Screening and Diagnostic Follow-Up

Among men who underwent biopsy in the PLCO trial (n = 4861), 2.0% experienced complications consisting predominately of infection, bleeding, or urinary difficulties, while 5.6% of 8313 men undergoing biopsy experienced similar complications in the Veterans Affairs cohort.55 Among men undergoing biopsy after abnormal PSA screening results, post-biopsy hospitalization ranged from 0.5% to 1.6% in 1 trial and 2 cohorts.53,55,57 Within approximately 1 month of biopsy within a UK cohort (n = 1147),53 7.3% of men experienced moderate or serious pain and 5.5% experienced moderate or severe fevers. Prostate biopsy has not been associated with statistically significantly increased risk of death.47,52,53 Within 3 cohorts (n = 1179),46,48,49,51 men who had abnormal PSA screening results followed by normal biopsy results had increased prostate cancer–specific worry up to 1 year after biopsy but no increase in depression or trait anxiety.

Overdiagnosis

The percentage of detected cancers that were overdiagnosed was estimated using the excess incidence method applied to screening trials data.18 When overdiagnosis was estimated as a percentage of all prostate cancers diagnosed, 16.4% of prostate cancers were overdiagnosed in the PLCO trial, 33.2% in the ERSPC trial, and 40.7% in the CAP trial. When estimated as a percentage of cancers detected by screening during the 2 trials reporting such data, 20.7% of cancers were overdiagnosed in the PLCO trial and 50.4% in the ERSPC trial. The extent of overdiagnosis varied across ERSPC sites, with higher rates of overdiagnosis occurring at 2 sites that also reported statistically significantly reduced prostate cancer mortality with screening.33,35

Benefits of Early Detection

Key Question 3. Is there evidence that various treatment approaches for early-stage or screen-detected prostate cancer reduce morbidity and mortality?

Three RCTs (n = 2524)9,58,59 and 7 cohort studies (n = 64,965)11,60-65 assessed morbidity and mortality with radical prostatectomy compared with conservative management, and 1 RCT (n = 1090)58 and 7 cohort studies (n = 60,585)11,60-65 compared radiation therapy with conservative management (Table 2). Conservative management in these studies consisted of observation without treatment, active surveillance, watchful waiting, or a combination of these approaches. Comparative studies of benefits and harms associated with other treatment modalities are summarized in the full evidence report.

The ProtecT trial randomized men with localized, screen-detected prostate cancer to radical prostatectomy, radiation therapy (with neoadjuvant androgen deprivation therapy), or active surveillance (consisting of periodic PSA monitoring without surveillance biopsy).58 During a median follow-up of 10 years, 54.8% of men randomized to active surveillance received treatment, mostly commonly radical prostatectomy; prostate cancer survival was approximately 99% in each group, with no statistically significant differences in prostate cancer mortality or all-cause mortality.

In the Scandinavian Prostate Cancer Group Trial-4 (SPCG-4), which included men with clinically diagnosed, predominately palpable cancers, radical prostatectomy was associated with statistically significantly reduced prostate cancer mortality (RR, 0.56 [95% CI, 0.41-0.77]) and all-cause mortality (RR, 0.71 [95% CI, 0.59-0.86]), compared with watchful waiting.59 In the US Prostate Cancer Intervention vs Observation Trial (PIVOT), in which 50% of men had screen-detected rather than clinically detected cancers, radical prostatectomy did not significantly reduce either prostate cancer mortality (RR, 0.65 [95% CI, 0.41-1.03]) or all-cause mortality (RR, 0.92 [95% CI, 0.82-1.02]), compared with conservative management.9

In the ProtecT trial, incidence of metastatic disease among men randomized to active surveillance (6.3 cases per 1000 person-years) was higher than among men randomized to radical prostatectomy or radiation therapy (2.4 and 3.0 cases per 1000 person-years; P = 0.004 for overall difference across groups). Approximately 27 (95% CI, 16-77) and 32 (95% CI, 18-143) men with screen-detected localized cancer would need to be treated with radical prostatectomy and radiation therapy (rather than active surveillance), respectively, to prevent 1 man from progressing to metastatic disease within 10 years. In both the SPCG-4 and PIVOT trials, radical prostatectomy was also associated with reduced progression to metastatic or systemic disease (SPCG-4: RR, 0.57 [95% CI, 0.44- 0.75]; PIVOT: HR, 0.64 [95% CI, 0.42-0.97]).9,59

Compared with conservative management, radical prostatectomy and radiation therapy were associated with reduced prostate cancer mortality and all-cause mortality in several fair- to good-quality cohort studies (Table 2).60-65 However, cohort study estimates often diverged from RCT estimates, and cohort studies of treatments may be susceptible to residual confounding.

Harms of Treatment

Key Question 4. What are the harms of the various treatment approaches for early-stage or screen-detected prostate cancer?

Three RCTs (n = 2524)66-68 and 11 cohort studies (n = 8809)10,11,69-78 compared harms among men treated with radical prostatectomy compared with conservative management, and 6 uncontrolled studies of surgical harms related to radical prostatectomy were included.79-84 Two RCTs (n = 1198)66,67,85 and 12 cohort studies (n = 4762)10,11,69-78,86 compared harms among men treated with radiation therapy compared with conservative management, and 1 uncontrolled observational study of radiation therapy was included (n = 3180).87

Both urinary incontinence and erectile dysfunction occurred more commonly in men who underwent radical prostatectomy than in men receiving conservative management (Figure 3 and Figure 4). Pooled relative risks of incontinence with radical prostatectomy were 2.27 (95% CI, 1.82-2.84; I2 = 0.0%) in 3 RCTs and 2.75 (95% CI, 1.78-4.23; I2 = 63.0%) in 6 cohort studies. Based on risk differences estimated from RCT data, 7.9 men (95% CI, 5.4-12.2) would need to be treated with radical prostatectomy rather than conservative management for 1 additional man to develop incontinence.

In pooled meta-analyses of the 3 RCTs, there was marked statistical heterogeneity in the relative risk of erectile dysfunction with radical prostatectomy (I2 = 87.5%), which was attributable to a disparate outcome from the ProtecT trial, in which many men randomized to active surveillance received radical treatments during the comparatively long 6 years of median follow-up for harms (Figure 4).66 After excluding the ProtecT trial from the RCT meta-analysis, the pooled relative risk of erectile dysfunction associated with radical prostatectomy vs conservative management was 1.82 (95% CI, 1.62-2.04; I2 = 0.0%). Based on estimated risk differences from this pooled analysis, 2.7 men (95% CI, 2.2-3.6) need to be treated with radical prostatectomy rather than conservative management for 1 additional man to develop erectile dysfunction.

In 5 longitudinal studies,9-11,66,77 mean urinary and sexual function scores decreased to a nadir in the initial year after radical prostatectomy with some longer-term improvement, although urinary and sexual function typically remained statistically significantly lower than baseline up to 6 years after the procedure.

Data on surgical complications and perioperative mortality associated with radical prostatectomy are summarized in eTable 3 in the Supplement. In a cohort of US men undergoing radical prostatectomy, 1.7% experienced major medical complications (most commonly cardiac or pulmonary) and 5.3% experienced major surgical complications (requiring reintervention) within 30 days of surgery.82 Across 2 trials and 6 cohort studies, the median perioperative mortality with radical prostatectomy was 0.29% (range, 0.0%-0.52%).

There was marked variability across 8 studies comparing incidence of urinary incontinence with radiation and conservative management, so these were not meta-analyzed (Figure 5). In the ProtecT trial, the prevalence of erectile dysfunction at 6 years of median follow-up was similar among men randomized to radiation therapy vs active surveillance (39.8% vs 36.2%; RR, 0.91 [95% CI, 0.77-1.08]), likely attributable to the substantial crossover to radical treatment of men randomized to active surveillance.66 In contrast, in 8 cohort studies, the prevalence of erectile dysfunction was more common in men treated with radiation therapy compared with men receiving conservative management (pooled RR, 1.31 [95% CI, 1.20-1.42]; I2 = 22.1%) (Figure 6).10,11,69,73,75-77,86 Based on the cohort data, 6.9 men (95% CI, 5.1-10.7) would need to be treated with radiation therapy rather than conservative management for 1 additional man to develop erectile dysfunction. Although 2 US studies observed longitudinal improvement in adverse effects of radiation therapy on sexual function,10,11 initial decrements in sexual function associated with radiation therapy persisted throughout a 3-year follow-up period in an Australian cohort.77 In the ProtecT trial, 10.4% of men randomized to radiation therapy experienced bothersome bowel symptoms (eg, loose stools, fecal incontinence) at 6-month follow-up, which tended to diminish during the 6-year follow-up.66 Two US cohort studies suggest a similar pattern,10,11 in contrast to an Australian cohort study in which adverse bowel effects of radiation therapy persisted during 3 years of follow-up.77

In 3 trials comparing radical prostatectomy with conservative management,9,66,88 randomization to radical prostatectomy was not associated with differences in anxiety, depression, or physical or mental health status. Cohort studies11,70-72,74,75,77,78 also found no evidence of an adverse effect of radical prostatectomy on generic quality-of-life measures compared with conservative management. Similarly, 2 trials66,67 and 9 cohort studies11,70-73,75,77,78,86 suggest no substantive adverse effect of radiation therapy on general measures of physical, mental, or global health status compared with conservative management.

Accuracy of Risk Calculators

Key Question 5. Is there evidence that use of a prebiopsy prostate cancer risk calculator, in combination with PSA-based screening, accurately identifies men with clinically significant prostate cancer compared with PSA-based screening alone?

Fourteen articles (n = 48,234)12-14,89-99 provided evidence on discrimination or calibration of risk assessment tools for prediction of clinically significant prostate cancer (either Gleason score ≥7 or Stage 2b or higher) among men undergoing prostate biopsy (Table 3). All were external validation studies of either the Prostate Cancer Prevention Trial (PCPT) risk calculator or the ERSPC risk calculator. Most cohorts consisted of men referred to tertiary or academic centers and typically included both symptomatic men and asymptomatic men with abnormal screening results. No RCTs evaluated the clinical effect of risk calculator use on patient outcomes.

Both calculators usually discriminated better than PSA alone between men with and without clinically significant prostate cancer (median area under the curve [AUC] with PCPT calculator, 0.72 [range across 21 cohorts, 0.51-0.88]; median AUC with ERSPC calculator, 0.74 [range across 7 cohorts, 0.69-0.78]; median AUC with PSA alone, 0.68 [range across 16 cohorts, 0.59-0.82]). However, calibration was mixed with underestimation or overestimation of actual risk in several cohorts for each calculator.

Return to Table of Contents

Table 3 summarizes the evidence contained in this review. Direct evidence from 3 fair-quality trials demonstrates that PSA screening increases prostate cancer detection, especially of localized, less aggressive cancers,30,39 and evidence from 4 ERSPC sites suggests that screening can reduce the long-term incidence of metastatic disease.43 While the PLCO and CAP trials found no association between randomization to screening invitation and reduced prostate cancer mortality, the overall ERSPC trial found a relative risk of 0.79 for prostate cancer mortality with screening.30 Differences in trial outcomes may have been attributable to greater baseline exposure to PSA testing and contamination in the PLCO trial, low adherence to invitation to a single PSA screen in the CAP trial, and higher adherence to screening and biopsy in the ERSPC trial. A recent analysis of individual patient data from both the PLCO and the ERSPC trials suggests that results from both trials are consistent with approximately 25% to 30% relative reduction in the risk of prostate cancer death with screening vs no screening after accounting for differences in baseline risk, screening adherence, contamination, and the intensity of postscreening diagnostic evaluation.100 In the ERSPC trial, 27 men needed to be diagnosed (and potentially treated) to prevent 1 prostate cancer death, underscoring the potential for overdiagnosis and overtreatment with PSA screening.30 Evidence is limited on the benefit of screening among men older than 70 years or the differential benefits among African American men or men with a family history of prostate cancer.

Trial data demonstrate that abnormal PSA screening test results are common and that most men referred for biopsy after abnormal screening results will not have prostate cancer.15,30,52 Biopsy harms include pain, bleeding, and infection,53 but perhaps the most serious harm of prostate cancer screening is overdiagnosis, because overdiagnosis burdens men with the potential harms of diagnosis and treatment without improving life expectancy or quality of life. It was estimated that 16.4% to 50.4% of prostate cancers were overdiagnosed during the 3 trials, consistent with estimates based on ecological or modeling studies.4,101-104

This review assessed both the immediate harms of screening as well as the consequent adverse effects of prostate cancer treatments. Most men undergoing radical prostatectomy will experience long-term sexual difficulties and approximately 17% will experience urinary difficulties, while approximately 36% of men receiving radiation therapy experience erectile problems and many will experience adverse bowel symptoms.66 Nevertheless, compared with conservative management, active treatments for localized prostate cancer did not clearly compromise overall quality of life or global physical or mental health status, despite adverse sexual, urinary, and bowel effects.11,66,70-72,74,75,77,86

The ProtecT trial found similarly high prostate cancer survival among men with screen-detected, early-stage prostate cancer randomized to radical prostatectomy, radiation therapy, or active surveillance. Of men assigned to active surveillance, 45.2% remained under surveillance without receiving active treatment during the 10-year follow-up period, although an increase in the incidence of metastatic disease was observed in the active surveillance group (approximately 6% with active surveillance vs 2 to 3% with active treatment). In contrast to the ProtecT active surveillance protocol that consisted of periodic PSA monitoring, many active surveillance protocols include surveillance biopsy or imaging, which might reduce metastatic disease risk, albeit with added harms associated with repeated biopsy. In contemporary case series, prostate cancer–specific survival estimates for patients receiving active surveillance have been as high as 99% at 10 years of follow-up, although since relatively few men in these series have had extended follow-up, uncertainty remains about long-term outcomes.105

This review demonstrates that prebiopsy risk calculators can discriminate between men with and without high-risk cancer better than PSA screening alone, but net clinical benefit of routine calculator use in biopsy decisions is not established by existing evidence. Diagnostic or surveillance strategies based on serum or urine tests or multiparametric magnetic resonance imaging are under study, and some have been recommended by the National Comprehensive Cancer Network.106 Further research is needed to elucidate whether the use of adjunctive tests can improve the balance of benefits and harms of PSA-based prostate cancer screening in community settings.

Because the lead time for prostate cancer may be very long, current screening trials (with <15 years of median follow-up) might underestimate mortality benefits. Limited follow-up duration from trials may also exaggerate estimates of overdiagnosis based on extra incidence, although estimates based on longer-term follow-up may be influenced by posttrial PSA testing. In the PLCO trial, contamination among control group participants would be expected to bias trial results toward the null. The ERSPC trial, in contrast, was limited by unexplained stage-adjusted differences in prostate cancer treatments by study group that may have biased results in favor of screening.107 There was also variation across ERSPC sites in recruitment methods, screening intervals, use of ancillary testing, and PSA thresholds for biopsy referral. The CAP trial was limited by the low adherence of intervention participants to the invitation to a single PSA screen. Across all studies, relatively few men older than 70 years were enrolled, and there is limited evidence about the differential benefits or harms of screening for men at higher risk.

Of 4 randomized trials comparing the effectiveness and harms of treatments for localized prostate cancer, only the ProtecT trial exclusively enrolled men with screen-detected cancer, and prostate cancer–specific and all-cause mortality were extremely low in that study.58 Other treatment RCTs enrolled many or mostly men with clinically detected prostate cancer.59,68,85 Thus, uncertainty remains about the generalizability of treatment trial results to US men with early-stage prostate cancer detected by PSA screening.

Limitations

This review may be limited by language or publication biases. Aside from uncontrolled studies of treatment harms, found on comparative effectiveness of new or novel treatment-modalities, such as alternative surgical approaches (eg, nerve-sparing or robotic surgery), cryotherapy, or high-intensity focused ultrasound. Because of the limited use and variable definitions of active surveillance during the time periods of most included studies, active surveillance and watchful waiting were grouped in analyses, although outcomes may differ between these conservative approaches.

Return to Table of Contents

PSA screening may reduce prostate cancer mortality risk but is associated with false-positive results, biopsy complications, and overdiagnosis. Compared with conservative approaches, active treatments for screen-detected prostate cancer have unclear effects on long-term survival but are associated with sexual and urinary difficulties.

Return to Table of Contents

Source: This article was first published in the Journal of the American Medical Association on May 8, 2018 (JAMA. 2018;319(18):1914-1931. doi:10.1001/jama.2018.3712)

Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest and none were reported.

Funding/Support: This research was funded under contract HHSA-290-2012-00015-I, Task Order 6, from the Agency for Healthcare Research and Quality (AHRQ), US Department of Health and Human Services, under a contract to support the USPSTF.

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ had no role in study selection, quality assessment, or synthesis. AHRQ staff provided project oversight, reviewed the report to ensure that the analysis met methodological standards, and distributed the draft for peer review. Otherwise, AHRQ had no role in the conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript findings. The opinions expressed in this document are those of the authors and do not reflect the official position of AHRQ or the US Department of Health and Human Services.

Additional Contributions: We gratefully acknowledge the following individuals for their contributions to this project: David Meyers, MD, and Tracy Wolff, MD, MPH (AHRQ); current and former members of the US Preventive Services Task Force who contributed to topic deliberations; and Jennifer S. Lin, MD, MCR (Kaiser Permanente Research Affiliates Evidence-based Practice Center, Portland, Oregon), Russell Harris, MD, MPH (University of North Carolina), and Bruce Abbott, MLS (University of California, Davis). USPSTF members, peer reviewers, and federal partner reviewers did not receive financial compensation for their contributions.

Additional Information: A draft version of this evidence report underwent external peer review from 6 content experts (Otis Brawley, MD, American Cancer Society; Peter Carroll, MD, University of California, San Francisco; Phillip Dahm, MD, University of Minnesota; David Penson, MD, Vanderbilt-Ingram Cancer Center; Ian Thompson, MD, University of Texas Health Science Center at San Antonio; Andrew Vickers, DPhil, Memorial Sloan Kettering Cancer Cente;) and 8 federal partners from the National Institute on Minority Health and Health Disparities, National Cancer Institute, National Institute of Nursing Research, and the Centers for Disease Control and Prevention. Comments from reviewers were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Return to Table of Contents

1. National Cancer Institute. SEER Cancer Statistics Factsheets: prostate cancer. https://seer.cancer.gov/statfacts/html/prost.html. Accessed July 24, 2017.
2. American Cancer Society. Cancer Facts & Figures 2018. Atlanta: American Cancer Society; 2018.
3. Kiciński M, Vangronsveld J, Nawrot TS. An epidemiological reappraisal of the familial aggregation of prostate cancer: a meta-analysis. PLoS One. 2011;6(10):e27130.
4. Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after the introduction of prostate-specific antigen screening: 1986-2005. J Natl Cancer Inst. 2009;101(19):1325-1329.
5. Lin JS, Petitti DB, Burda BU. Overview of Prostate Cancer Screening Decision Models: A Contextual Review for the U.S. Preventive Services Task Force. Rockville, MD: Agency for Healthcare Research and Quality; 2018. AHRQ publication 17-05229-EF-2.
6. US Preventive Services Task Force. US Preventive Services Task Force Procedure Manual. https://www.uspreventiveservicestaskforce.org/Home/GetFile/6/7/procedure-manual_2015/pdf. 2015. Accessed May 24, 2016.
7. Chou R, Croswell JM, Dana T, et al. Screening for prostate cancer: a review of the evidence for the U.S. Preventive Services Task Force. Ann Intern Med. 2011;155(11):762-771.
8. Lin K, Croswell JM, Koenig H, Lam C, Maltz A. Prostate-Specific Antigen–Based Screening for Prostate Cancer: An Evidence Update for the U.S. Preventive Services Task Force. Rockville, MD: Agency for Healthcare Research and Quality; 2011. AHRQ publication 12-05160-EF-1.
9. Wilt TJ, Jones KM, Barry MJ, et al. Follow-up of prostatectomy versus observation for early prostate cancer. N Engl J Med. 2017;377(2):132-142.
10. Chen RC, Basak R, Meyer AM, et al. Association between choice of radical prostatectomy, external beam radiotherapy, brachytherapy, or active surveillance and patient-reported quality of life among men with localized prostate cancer. JAMA. 2017;317(11):1141-1150.
11. Barocas DA, Alvarez J, Resnick MJ, et al. Association between radiation therapy, surgery, or observation for localized prostate cancer and patient-reported outcomes after 3 years. JAMA. 2017;317(11):1126-1140.
12. Gómez-Gómez E, Carrasco-Valiente J, Blanca-Pedregosa A, et al. European Randomized Study of Screening for Prostate Cancer risk calculator: external validation, variability, and clinical significance. Urology. 2017;102:85-91.
13. Maruf M, Fascelli M, George AK, et al. The prostate cancer prevention trial risk calculator 2.0 performs equally for standard biopsy and MRI/US fusion-guided biopsy. Prostate Cancer Prostatic Dis. 2017;20(2):179-185.
14. Park JY, Yoon S, Park MS, et al. Development and external validation of the Korean prostate cancer risk calculator for high-grade prostate cancer: comparison with two Western risk calculators in an Asian cohort. PLoS One. 2017;12(1):e0168917.
15. Martin RM, Donovan JL, Turner EL, et al; CAP Trial Group. Effect of a low-intensity PSA-based screening intervention on prostate cancer mortality: the CAP randomized clinical trial. JAMA. 2018;319(9):883-895.
16. Miller AB, Wall C, Baines CJ, Sun P, To T, Narod SA. Twenty five year follow-up for breast cancer incidence and mortality of the Canadian National Breast Screening Study: randomised screening trial. BMJ. 2014;348:g366.
17. Zackrisson S, Andersson I, Janzon L, Manjer J, Garne JP. Rate of over-diagnosis of breast cancer 15 years after end of Malmö mammographic screening trial: follow-up study. BMJ. 2006;332 (7543):689-692.
18. Carter JL, Coletti RJ, Harris RP. Quantifying and monitoring overdiagnosis in cancer screening: a systematic review of methods. BMJ. 2015;350:g7773.
19. Patz EF Jr, Pinsky P, Gatsonis C, et al; NLST Overdiagnosis Manuscript Writing Team. Overdiagnosis in low-dose computed tomography screening for lung cancer. JAMA Intern Med. 2014; 174(2):269-274.
20. Wells GA, Shea B, O’Connell D, et al. The Newcastle-Ottowa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analysis. http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp. 2016. Accessed August 16, 2016.
21. National Institute for Health and Care Excellence. The Guidelines Manual. London, United Kingdom: National Institute for Health and Care Excellence; 2006.
22. Moons KG, de Groot JA, Bouwmeester W, et al. Critical appraisal and data extraction for systematic reviews of prediction modelling studies: the CHARMS checklist. PLoS Med. 2014;11(10):e1001744.
23. Collins GS, Reitsma JB, Altman DG, Moons KG. Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD): the TRIPOD statement. Ann Intern Med. 2015;162(1):55-63.
24. Kjellman A, Akre O, Norming U, Törnblom M, Gustafsson O. 15-year followup of a population based prostate cancer screening study. J Urol. 2009;181(4):1615-1621.
25. Labrie F, Candas B, Cusan L, et al. Screening decreases prostate cancer mortality: 11-year follow-up of the 1988 Quebec prospective randomized controlled trial. Prostate. 2004;59(3):311-318.
26. Sandblom G, Varenhorst E, Rosell J, Löfman O, Carlsson P. Randomised prostate cancer screening trial: 20 year follow-up. BMJ. 2011;342:d1539.
27. Andriole GL, Crawford ED, Grubb RL III, et al; PLCO Project Team. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med. 2009;360(13):1310-1319.
28. Schröder FH, Hugosson J, Roobol MJ, et al; ERSPC Investigators. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med. 2009;360(13):1320-1328.
29. Pinsky PF, Prorok PC, Yu K, et al. Extended mortality results for prostate cancer screening in the PLCO trial with median follow-up of 15 years. Cancer. 2017;123(4):592-599.
30. Schröder FH, Hugosson J, Roobol MJ, et al; ERSPC Investigators. Screening and prostate cancer mortality: results of the European Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years of follow-up. Lancet. 2014;384(9959):2027-2035.
31. Ciatto S, Zappa M, Villers A, Paez A, Otto SJ, Auvinen A. Contamination by opportunistic screening in the European Randomized Study of Prostate Cancer Screening. BJU Int. 2003;92(suppl 2):97-100.
32. Arnsrud Godtman R, Holmberg E, Lilja H, Stranne J, Hugosson J. Opportunistic testing versus organized prostate-specific antigen screening: outcome after 18 years in the Göteborg randomized population-based prostate cancer screening trial. Eur Urol. 2015;68(3):354-360.
33. Hugosson J, Carlsson S, Aus G, et al. Mortality results from the Göteborg randomised population-based prostate-cancer screening trial. Lancet Oncol. 2010;11(8):725-732.
34. Luján M, Páez A, Angulo JC, et al. Prostate cancer incidence and mortality in the Spanish section of the European Randomized Study of Screening for Prostate Cancer (ERSPC). Prostate Cancer Prostatic Dis. 2014;17(2):187-191.
35. Roobol MJ, Kranse R, Bangma CH, et al; ERSPC Rotterdam Study Group. Screening for prostate cancer: results of the Rotterdam section of the European randomized study of screening for prostate cancer. Eur Urol. 2013;64(4):530-539.
36. Bokhorst LP, Bangma CH, van Leenders GJ, et al. Prostate-specific antigen-based prostate cancer screening: reduction of prostate cancer mortality after correction for nonattendance and contamination in the Rotterdam section of the European Randomized Study of Screening for Prostate Cancer. Eur Urol. 2014;65(2):329-336.
37. Kilpeläinen TP, Tammela TL, Malila N, et al. Prostate cancer mortality in the Finnish randomized screening trial. J Natl Cancer Inst. 2013;105(10):719-725.
38. Kilpeläinen TP, Pogodin-Hannolainen D, Kemppainen K, et al. Estimate of opportunistic prostate-specific antigen testing in the Finnish Randomized Study of Screening for Prostate Cancer. J Urol. 2017;198(1):50-57.
39. Andriole GL, Crawford ED, Grubb RL III, et al; PLCO Project Team. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: mortality results after 13 years of follow-up. J Natl Cancer Inst. 2012;104(2):125-132.
40. Cuzick J, Thorat MA, Andriole G, et al. Prevention and early detection of prostate cancer. Lancet Oncol. 2014;15(11):e484-e492.
41. Pinsky PF, Blacka A, Kramer BS, Miller A, Prorok PC, Berg C. Assessing contamination and compliance in the prostate component of the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. Clin Trials. 2010;7(4):303-311.
42. Young GJ, Harrison S, Turner EL, et al. Prostate-specific antigen (PSA) testing of men in UK general practice: a 10-year longitudinal cohort study. BMJ Open. 2017;7(10):e017729.
43. Schröder FH, Hugosson J, Carlsson S, et al. Screening for prostate cancer decreases the risk of developingmetastatic disease: findings from the European Randomized Study of Screening for Prostate Cancer (ERSPC). Eur Urol. 2012;62(5):745-752.
44. Liss MA, Chen H, Hemal S, et al. Impact of family history on prostate cancer mortality in white men undergoing prostate specific antigen based screening. J Urol. 2015;193(1):75-79.
45. Bokhorst LP, Zhu X, Bul M, Bangma CH, Schröder FH, Roobol MJ. Positive predictive value of prostate biopsy indicated by prostate-specific antigen-based prostate cancer screening: trends over time in a European randomized trial. BJU Int. 2012;110(11):1654-1660.
46. Brindle LA, Oliver SE, Dedman D, et al. Measuring the psychosocial impact of population-based prostate-specific antigen testing for prostate cancer in the UK. BJU Int. 2006;98(4):777-782.
47. Carlsson SV, Holmberg E, Moss SM, et al. No excess mortality after prostate biopsy: results from the European Randomized Study of Screening for Prostate Cancer. BJU Int. 2011;107(12):1912-1917.
48. Fowler FJ Jr, Barry MJ, Walker-Corkery B, et al. The impact of a suspicious prostate biopsy on patients’ psychological, socio-behavioral, and medical care outcomes. J Gen Intern Med. 2006;21(7):715-721.
49. Katz DA, Jarrard DF, McHorney CA, Hillis SL, Wiebe DA, Fryback DG. Health perceptions in patients who undergo screening and workup for prostate cancer. Urology. 2007;69(2):215-220.
50. Kilpeläinen TP, Tammela TL, Roobol M, et al. False-positive screening results in the European randomized study of screening for prostate cancer. Eur J Cancer. 2011;47(18):2698-2705.
51. McNaughton-Collins M, Fowler FJ Jr, Caubet JF, et al. Psychological effects of a suspicious prostate cancer screening test followed by a benign biopsy result. Am J Med. 2004;117(10):719-725.
52. Pinsky PF, Parnes HL, Andriole G. Mortality and complications after prostate biopsy in the Prostate, Lung, Colorectal and Ovarian Cancer Screening (PLCO) trial. BJU Int. 2014;113(2):254-259.
53. Rosario DJ, Lane JA, Metcalfe C, et al. Short term outcomes of prostate biopsy in men tested for cancer by prostate specific antigen: prospective evaluation within ProtecT study. BMJ. 2012;344:d7894.
54. Vasarainen H, Malmi H, Määttänen L, et al. Effects of prostate cancer screening on health-related quality of life: results of the Finnish arm of the European Randomized Screening Trial (ERSPC). Acta Oncol. 2013;52(8):1615-1621.
55. Walter LC, Fung KZ, Kirby KA, et al. Five-year downstream outcomes following prostate-specific antigen screening in older men. JAMA Intern Med. 2013;173(10):866-873.
56. Croswell JM, Kramer BS, Kreimer AR, et al. Cumulative incidence of false-positive results in repeated, multimodal cancer screening. Ann Fam Med. 2009;7(3):212-222.
57. Raaijmakers R, Kirkels WJ, Roobol MJ, Wildhagen MF, Schrder FH. Complication rates and risk factors of 5802 transrectal ultrasound-guided sextant biopsies of the prostate within a population-based screening program. Urology. 2002;60(5):826-830.
58. Hamdy FC, Donovan JL, Lane JA, et al; ProtecT Study Group. 10-Year outcomes after monitoring, surgery, or radiotherapy for localized prostate cancer. N Engl J Med. 2016;375(15):1415-1424.
59. Bill-Axelson A, Holmberg L, Garmo H, et al. Radical prostatectomy or watchful waiting in early prostate cancer. N Engl J Med. 2014;370(10):932-942.
60. Ladjevardi S, Sandblom G, Berglund A, Varenhorst E. Tumour grade, treatment, and relative survival in a population-based cohort of men with potentially curable prostate cancer. Eur Urol. 2010;57(4):631-638.
61. Schymura MJ, Kahn AR, German RR, et al. Factors associated with initial treatment and survival for clinically localized prostate cancer: results from the CDC-NPCR Patterns of Care Study (PoC1). BMC Cancer. 2010;10:152.
62. Stattin P, Holmberg E, Johansson JE, Holmberg L, Adolfsson J, Hugosson J; National Prostate Cancer Register (NPCR) of Sweden. Outcomes in localized prostate cancer: National Prostate Cancer Register of Sweden follow-up study. J Natl Cancer Inst. 2010;102(13):950-958.
63. Zhou EH, Ellis RJ, Cherullo E, et al. Radiotherapy and survival in prostate cancer patients: a population-based study. Int J Radiat Oncol Biol Phys. 2009;73(1):15-23.
64. Albertsen PC, Hanley JA, Penson DF, Barrows G, Fine J. 13-year outcomes following treatment for clinically localized prostate cancer in a population based cohort. J Urol. 2007;177(3):932-936.
65. Wong YN, Mitra N, Hudes G, et al. Survival associated with treatment vs observation of localized prostate cancer in elderly men. JAMA. 2006;296(22):2683-2693.
66. Donovan JL, Hamdy FC, Lane JA, et al; ProtecT Study Group. Patient-reported outcomes after monitoring, surgery, or radiotherapy for prostate cancer. N Engl J Med. 2016;375(15):1425-1437.
67. Fransson P, Damber JE, Widmark A. Health-related quality of life 10 years after external beam radiotherapy or watchful waiting in patients with localized prostate cancer. Scand J Urol Nephrol. 2009;43(2):119-126.
68. Wilt TJ, Brawer MK, Jones KM, et al; Prostate Cancer Intervention versus Observation Trial (PIVOT) Study Group. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med. 2012;367(3):203-213.
69. Hoffman RM, Hunt WC, Gilliland FD, Stephenson RA, Potosky AL. Patient satisfaction with treatment decisions for clinically localized prostate carcinoma: results from the Prostate Cancer Outcomes Study. Cancer. 2003;97(7):1653-1662.
70. Litwin MS, Lubeck DP, Spitalny GM, Henning JM, Carroll PR. Mental health in men treated for early stage prostate carcinoma: a posttreatment, longitudinal quality of life analysis from the Cancer of the Prostate Strategic Urologic Research Endeavor. Cancer. 2002;95(1):54-60.
71. Bacon CG, Giovannucci E, Testa M, Kawachi I. The impact of cancer treatment on quality of life outcomes for patients with localized prostate cancer. J Urol. 2001;166(5):1804-1810.
72. Lubeck DP, Litwin MS, Henning JM, Stoddard ML, Flanders SC, Carroll PR. Changes in health-related quality of life in the first year after treatment for prostate cancer: results from CaPSURE. Urology. 1999;53(1):180-186.
73. Litwin MS. Health-related quality of life after treatment for localized prostate cancer. Cancer. 1995;75(S7):2000-2003.
74. Litwin MS, Hays RD, Fink A, et al. Quality-of-life outcomes in men treated for localized prostate cancer. JAMA. 1995;273(2):129-135.
75. Schapira MM, Lawrence WF, Katz DA, McAuliffe TL, Nattinger AB. Effect of treatment on quality of life among men with clinically localized prostate cancer. Med Care. 2001;39(3):243-253.
76. Siegel T, Moul JW, Spevak M, Alvord WG, Costabile RA. The development of erectile dysfunction in men treated for prostate cancer. J Urol. 2001;165(2):430-435.
77. Smith DP, King MT, Egger S, et al. Quality of life three years after diagnosis of localised prostate cancer: population based cohort study. BMJ. 2009;339:b4817.
78. Smith DS, Carvalhal GF, Schneider K, Krygiel J, Yan Y, Catalona WJ. Quality-of-life outcomes for men with prostate carcinoma detected by screening. Cancer. 2000;88(6):1454-1463.
79. Alibhai SM, Leach M, Tomlinson G, et al. 30-day mortality and major complications after radical prostatectomy: influence of age and comorbidity. J Natl Cancer Inst. 2005;97(20):1525-1532.
80. Augustin H, Hammerer P, Graefen M, et al. Intraoperative and perioperative morbidity of contemporary radical retropubic prostatectomy in a consecutive series of 1243 patients: results of a single center between 1999 and 2002. Eur Urol. 2003;43(2):113-118.
81. Björklund J, Folkvaljon Y, Cole A, et al. Postoperative mortality 90 days after robot-assisted laparoscopic prostatectomy and retropubic radical prostatectomy: a nationwide population-based study. BJU Int. 2016;118(2):302-306.
82. Rabbani F, Yunis LH, Pinochet R, et al. Comprehensive standardized report of complications of retropubic and laparoscopic radical prostatectomy. Eur Urol. 2010;57(3):371-386.
83. Walz J, Montorsi F, Jeldres C, et al. The effect of surgical volume, age and comorbidities on 30-day mortality after radical prostatectomy: a population-based analysis of 9208 consecutive cases. BJU Int. 2008;101(7):826-832.
84. Yao SL, Lu-Yao G. Population-based study of relationships between hospital volume of prostatectomies, patient outcomes, and length of hospital stay. J Natl Cancer Inst. 1999;91(22):1950-1956.
85. Fransson P, Damber JE, Tomic R, Modig H, Nyberg G, Widmark A. Quality of life and symptoms in a randomized trial of radiotherapy versus deferred treatment of localized prostate carcinoma. Cancer. 2001;92(12):3111-3119.
86. ThongMS, Mols F, Kil PJ, Korfage IJ, van de Poll-Franse LV. Prostate cancer survivors who would be eligible for active surveillance but were either treated with radiotherapy or managed expectantly: comparisons on long-term quality of life and symptom burden. BJU Int. 2010;105(5):652-658.
87. Shah C, Jones PM, Wallace M, et al. Differences in disease presentation, treatment outcomes, and toxicities in African American patients treated with radiation therapy for prostate cancer. Am J Clin Oncol. 2012;35(6):566-571.
88. Johansson E, Steineck G, Holmberg L, et al; SPCG-4 Investigators. Long-term quality-of-life outcomes after radical prostatectomy or watchful waiting: the Scandinavian Prostate Cancer Group-4 randomised trial. Lancet Oncol. 2011;12(9):891-899.
89. Foley RW, Maweni RM, Gorman L, et al. European Randomised Study of Screening for Prostate Cancer (ERSPC) risk calculators significantly outperform the Prostate Cancer Prevention Trial (PCPT) 2.0 in the prediction of prostate cancer: a multi-institutional study. BJU Int. 2016;118(5):706-713.
90. Poyet C, Nieboer D, Bhindi B, et al. Prostate cancer risk prediction using the novel versions of the European Randomised Study for Screening of Prostate Cancer (ERSPC) and Prostate Cancer Prevention Trial (PCPT) risk calculators: independent validation and comparison in a contemporary European cohort. BJU Int. 2016;117(3):401-408.
91. Lundon DJ, Kelly BD, Foley R, et al. Prostate cancer risk assessment tools in an unscreened population. World J Urol. 2015;33(6):827-832.
92. Ankerst DP, Hoefler J, Bock S, et al. Prostate Cancer Prevention Trial risk calculator 2.0 for the prediction of low- vs high-grade prostate cancer. Urology. 2014;83(6):1362-1367.
93. Ankerst DP, Boeck A, Freedland SJ, et al. Evaluating the Prostate Cancer Prevention Trial High Grade Prostate Cancer Risk Calculator in 10 international biopsy cohorts: results from the Prostate Biopsy Collaborative Group. World J Urol. 2014;32(1):185-191.
94. Roobol MJ, van Vugt HA, Loeb S, et al. Prediction of prostate cancer risk: the role of prostate volume and digital rectal examination in the ERSPC risk calculators. Eur Urol. 2012;61(3):577-583.
95. Nam RK, Kattan MW, Chin JL, et al. Prospective multi-institutional study evaluating the performance of prostate cancer risk calculators. J Clin Oncol. 2011;29(22):2959-2964.
96. Ngo TC, Turnbull BB, Lavori PW, Presti JC Jr. The prostate cancer risk calculator from the Prostate Cancer Prevention Trial underestimates the risk of high grade cancer in contemporary referral patients. J Urol. 2011;185(2):483-487.
97. Trottier G, Roobol MJ, Lawrentschuk N, et al. Comparison of risk calculators from the Prostate Cancer Prevention Trial and the European Randomized Study of Screening for Prostate Cancer in a contemporary Canadian cohort. BJU Int. 2011;108(8, pt 2):E237-E244.
98. Nguyen CT, Yu C, Moussa A, Kattan MW, Jones JS. Performance of prostate cancer prevention trial risk calculator in a contemporary cohort screened for prostate cancer and diagnosed by extended prostate biopsy. J Urol. 2010;183(2):529-533.
99. Hernandez DJ, Han M, Humphreys EB, et al. Predicting the outcome of prostate biopsy: comparison of a novel logistic regression-based model, the prostate cancer risk calculator, and prostate-specific antigen level alone. BJU Int. 2009;103(5):609-614.
100. Tsodikov A, Gulati R, Heijnsdijk EA, et al. Reconciling the effects of screening on prostate cancer mortality in the ERSPC and PLCO trials. Ann Intern Med. 2017;167(7):449-455.
101. Zappa M, Ciatto S, Bonardi R, Mazzotta A. Overdiagnosis of prostate carcinoma by screening: an estimate based on the results of the Florence Screening Pilot Study. Ann Oncol. 1998;9(12):1297-1300.
102. Draisma G, Boer R, Otto SJ, et al. Lead times and overdetection due to prostate-specific antigen screening: estimates from the European Randomized Study of Screening for Prostate Cancer. J Natl Cancer Inst. 2003;95(12):868-878.
103. Draisma G, Etzioni R, Tsodikov A, et al. Lead time and overdiagnosis in prostate-specific antigen screening: importance of methods and context. J Natl Cancer Inst. 2009;101(6):374-383.
104. Etzioni R, Penson DF, Legler JM, et al. Overdiagnosis due to prostate-specific antigen screening: lessons from U.S. prostate cancer incidence trends. J Natl Cancer Inst. 2002;94(13):981-990.
105. Tosoian JJ, Carter HB, Lepor A, Loeb S. Active surveillance for prostate cancer: current evidence and contemporary state of practice. Nat Rev Urol. 2016;13(4):205-215.
106. Carroll PR, Parsons JK, Andriole G, et al. NCCN Guidelines Insights: prostate cancer early detection, version 2.2016. J Natl Compr Canc Netw. 2016;14(5):509-519.
107. Wolters T, Roobol MJ, Steyerberg EW, et al. The effect of study arm on prostate cancer treatment in the large screening trial ERSPC. Int J Cancer. 2010;126(10):2387-2393.

Return to Table of Contents

Source No. of
Participants
Age
Range, y
PSA
Screening
Threshold,
ng/mL
Screening
Interval, y
(Planned
No. of
Screening
Rounds
Median
Follow-Up, y
Prostate Cancer Incidence, No. (%) No. of Prostate Cancer Deaths RR (95% CI)
Intervention Control Intervention Control Per 10,000 Men Per 10,000 Person-Years
Intervention Control Intervention Control Prostate Cancer
Mortality
All-Cancer Mortality
CAP,15 2018
(United Kingdom)
189,386 219,439 50-69 3.0 1 (1) 10.0 8054 (4.3) 7853 (3.6) 29.0 29.5 3.0 3.1 0.96
(0.85-1.08)
0.99
(0.94-1.03)
PLCO,27,29 2016
(United States)
38,340 38,343 55-74 4.0 1 (6) 13.0, 14.8b 4250 (11.1) 3815 (9.9) 66.5 63.6 4.8 4.6 1.04
(0.87-1.24)
0.98
(0.95-1.00)
ERSPC,30,31 2014c
(Belgium, Finland, France, Italy, the Netherlands, Spain, Sweden, Switzerland)d
72,891 89,352 55-69e 2.5-4.0f 2-4 (3-10)f 13.0 7408 (10.2) 6107 (6.0) 48.7 61.0 4.3 5.4 0.79
(0.69-0.91)
1.00
(0.98-1.02)
ERSPC–Göteborg,32,33 2015
(Sweden)
10,000 10,000 50-64 2.5-3.0 2 (10) 14.0, 18.0g 1396 (14.0) 962 (9.6) 79.0 122.0 NR NR 0.58
(0.46-0.72)
Intervention: 19.8%
Control: 19.8%
ERSPC–Spain,34 2014
(Spain)
2415 1861 45-70 3.0 4 (3) 15.2 161 (6.7) 80 (4.3) 20.7 26.9 4.8 4.6 0.76
(0.22-2.62)
0.92
(0.78-1.08)
ERSPC–Rotterdam,35,36 2013
(the Netherlands)
20,985 20,917 55-74 3.0-4.0 4 (5) 12.8 2674 (12.7) 1430 (6.8) 72.0 89.9 NR NR 0.80
(0.65-0.99)
Intervention: 2.8%
Control: 2.3%
ERSPC–Finland,37,38 2013
(Finland)
31,866 48,278 55-67 4.0 4 (3) 12.0 2883 (9.0) 3337 (6.9) 46.8 55.1 NR NR 0.85
(0.69-1.04)h
0.99
(0.96-1.02)h

Abbreviations: ERSPC, European Randomized Study of Screening for Prostate Cancer; NR, not reported; PLCO, Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial; PSA, prostate-specific antigen; RR, relative risk.
a The comparator for each randomized clinical trial was a control group that was not offered prostate cancer screening. All studies in this table were fair quality, as assessed using criteria from the US Preventive Services Task Force.6
b PLCO incidence data are based on median follow-up of 13 years,39 and mortality data are based on median follow-up of 14.8 years.29
c Results for the overall ERSPC trial are inclusive of all study locations, including those that have published site-specific results (Sweden, Spain, the Netherlands, Finland).
d France was excluded from all published ERSPC analyses of prostate cancer mortality because of incomplete follow-up.30 Data presented are for all sites except France.
e ERSPC recruited men aged 50 to 74 years; however, most ERSPC reports, including the most recent update,30 emphasize outcomes among men in the “core” age group of 55 to 69 years at randomization.
f The Belgium site initially used a PSA threshold of 10.0 ng/mL and a screening interval of 7 years but transitioned to a threshold of 4.0 ng/mL and a 4-year interval by the end of the study.
g ERSPC-Göteborg reported longest (rather than median) follow-up, which was 14.0 years for incidence and all-cause mortality and 18.0 years for prostate cancer mortality.
h ERSPC-Finland reported hazard ratio rather than relative risk.

 

Return to Table of Contents

Source Study Design
(Quality)b
No. of
Participants
Age
Range, y
Median
Follow-Up, y
Prostate Cancer Mortality All-Cause Mortality
Intervention Control Deaths, No. (%) Deaths, No. (%)
Intervention Control RR (95% CI) Intervention Control RR (95% CI)
Radical Prostatectomy
ProtecT,58 2016
(United Kingdom)
RCT (good) 553 545 49-69 10.0 5 (0.9) 8 (1.5) 0.63 (0.21-1.93)c 55 (9.9) 59 (10.8) 0.93 (0.65-1.35)c
SPCG-4,59 2014
(Sweden, Finland, Iceland)
RCT (good) 347 348 NR 13.4 63 (17.7) 99 (28.7) 0.56 (0.41-0.77) 200 (56.1) 247 (68.9) 0.71 (0.59-0.86)
PIVOT,9 2017
(United States)
RCT (good) 364 367 40-75 12.7 27 (7.4) 42 (11.4) 0.65 (0.41-1.03) 223 (61.3) 245 (66.8) 0.92 (0.82-1.02)
Barocas et al,11 2017
(United States)
Cohort (good) 1523 429 <80 3.3 1 (0.1) 0 NR 18 (1.3) 12 (2.9) NR
Ladjevardi et al,60 2010
(Sweden)
Cohort (fair) 12,950 12,645 <75 4.4 NR NR NR NR NR 0.36 (0.32-0.40)c
Schymura et al,61 2010
(United States)
Cohort (fair) 1310 614 NR 5.0d NR NR NR 6.3e 24.5e NR
Stattin et al,62 2010
(Sweden)
Cohort (fair) 3399 2021 <70 8.2 56 (1.7) 58 (2.9) 0.49 (0.34-0.71) 286 (8.4) 413 (20.4) 0.49 (0.41-0.57)
Zhou et al,63 2009
(United States)
Cohort (fair) 936 2306 ≥65 7.0d NR NR 0.25 (0.13-0.48)c NR NR 0.32 (0.25-0.41)c
Albertsen et al,64 2007
(United States)
Cohort (fair) 802 114 ≤75 13.3 64 (8.0) 18 (16.0) 0.29 (0.17-0.52)c 216 (27.0) 65 (57.0) NR
Wong et al,65 2006
(United States)
Cohort (good) 13,292 12,608 65-80 12.0d NR NR NR NR NR 0.50 (0.47-0.53)
Radiation Therapy
ProtecT,58 2016
(United Kingdom)
RCT (good) 545 545 49-69 10.0 4 (0.7) 8 (1.5) 0.51 (0.15-1.69)c 55 (10.1) 59 (10.8) 0.94 (0.65-1.36)c
Barocas et al,11 2017
(United States)
Cohort (good) 598 429 <80 3.3 2 (0.3) 0 NR 21 (3.9) 12 (2.9) NR
Ladjevardi et al,60 2010
(Sweden)
Cohort (fair) 6308 12,645 <75 4.4 NR NR NR NR NR 0.54 (0.49-0.59)c
Schymura et al,61 2010
(United States)
Cohort (fair) 1037 614 NR 5.0d NR NR NR 14.0e 24.5e NR
Stattin et al,62 2010
(Sweden)
Cohort (fair) 1429 2021 <70 8.2 40 (2.8) 58 (2.9) 0.70 (0.45-1.09) 196 (13.7) 413 (20.4) 0.68 (0.57-0.82)
Zhou et al,63 2009
(United States)
Cohort (fair) 879 2306 ≥65 7.0d NR NR 0.66 (0.41-1.04)c NR NR 0.63 (0.53-0.75)c
Albertsen et al,64 2007
(United States)
Cohort (fair) 702 114 ≤75 13.3 126 (18.0) 18 (16.0) 0.65c 393 (55.0) 65 (57.0) 0.83 (0.67-1.11)c
Wong et al,65 2006
(United States)
Cohort (good) 18,249 12,608 65-80 12.0d NR NR NR NR NR 0.81 (0.78-0.85)c

Abbreviations: CM, conservative management; NR, not reported; PIVOT, Prostate Cancer Intervention vs Observation Trial; ProtecT, Prostate Testing for Cancer and Treatment trial; RCT, randomized clinical trial; RR, relative risk.
a The comparator for each study was a control group receiving conservative management (ie, active surveillance, watchful waiting, deferred treatment or no treatment).
b Assessed using criteria from the US Preventive Services Task Force.6
c Study reported only hazard ratios (HRs). Confidence intervals are excluded when not reported.
d Maximum follow-up (ie, duration of study period).
e Study reported only percentages.

Return to Table of Contents

Test or Treatment Study Design No. of Studies (No.
of Participants)
Summary of Findings Body of Evidence Limitations Applicability Overall Quality
Key Question 1: Effectiveness of Screening
PSA-based
screening
RCT 3 (647,906) RR = 0.79 for prostate-specific mortality in ERSPC; no significant reduction in PLCO or CAP trials.

No trial observed significant reduction in all-cause mortality.

Within 4 ERSPC sites, screening associated with 3.1 fewer cases of metastatic disease per 1000 men at 12-y follow-up.

Contamination in PLCO control group, and low adherence with single PSA screening in CAP.

Variable protocols across ERSPC sites, and ERSPC screening group enrollees with cancer more likely to be treated with radical prostatectomy than similar controls.

Uncertain applicability of ERSPC results to US practice because of lower rate of baseline screening, lower biopsy thresholds (usually PSA ≥3 ng/mL), and higher rate of biopsy than is usual in US practice. Fair
Key Question 2: Harms of Screening
PSA-based
screening
RCT 3 (647,906) Cumulative incidence of false-positive results, 10.4%-17.8% among screened men.

Among biopsied men, 60.6%-75.8% of biopsies were negative for cancer.

In trials, 20.7%-50.4% of screen-detected cancers were overdiagnosed. In PLCO, 20.2 complications per 1000 biopsies (mostly infectious); no increase in mortality with biopsy.

ERSPC sites with variable PSA thresholds for positive test results.

Lower biopsy rate after positive screen result in PLCO vs ERSPC (44.8% vs 85.6%).

Limited follow-up in the trials (median, 10-13 y) may exaggerate overdiagnosis estimates.

Small sample sizes inhibit ability to detect increased mortality after biopsy.

Results from the PLCO trial generalizable to US practice.

Rates of biopsy in US practice may be lower than reported by PLCO or ERSPC, as many men may undergo monitoring (eg, repeat PSA testing) instead of biopsy.

Fair
Cohort 5 (297,971) Within 7 d of biopsy, 20.0%, 5.7%, and 4.0% of men reported moderate or severe hematospermia, pain, or fevers, respectively; 0.5% to 1.6% of men were hospitalized after biopsy; 10.4% sought outpatient care.

Men with abnormal PSA screen results have increased prostate cancer–specific worry up to 1 y; no increase in state anxiety or depression.

Key Question 3: Effectiveness of Treatments
Radical
prostatectomy
RCT 3 (2524) 1 of 3 trials reported statistically significant reductions in prostate cancer mortality with radical prostatectomy relative to conservative management, and this trial enrolled strictly men with clinically detected, palpable cancers.

All 3 trials reported significantly reduced risk of progression to metastatic disease with radical prostatectomy relative to conservative management.

Only the ProtecT trial exclusively randomized men with screen-detected cancer; PIVOT and SPCG-4 included men with higher-risk, clinically detected cancers.

Low event rate in ProtecT led to imprecise estimates for prostate cancer and all-cause mortality.

Cohort studies vulnerable to confounding by unmeasured patient characteristics.

Active surveillance in ProtecT consisted chiefly of PSA monitoring, which may not be applicable to US settings, where surveillance biopsy may be routine in most active surveillance protocols. Fair to good
Cohort 7 (64,965) Radical prostatectomy associated with statistically significantly reduced prostate cancer mortality in 3 cohorts; 4 of 7 cohorts reported reduced all-cause mortality with radical prostatectomy relative to conservative management.
Radiation therapy RCT 1 (1090) ProtecT trial found no significant reductions in prostate cancer or all-cause mortality with radiation therapy vs active surveillance.

Radiation therapy associated with significantly reduced risk of progression to metastatic disease (3.0 vs 6.3 per 1000 person-years with radiation therapy and conservative management, respectively).

Only 1 trial (ProtecT) assessed the effectiveness of radiation therapy and was not powered to detect differences in all-cause mortality.

Only the ProtecT triala assessed effect of radiation therapy on metastatic disease.

Cohort studies vulnerable to confounding by unmeasured patient characteristics.

Active surveillance in ProtecT consisted chiefly of PSA monitoring, which may not be applicable to US settings, where surveillance biopsy may be routine in most active surveillance protocols. Fair to good
Cohort 7 (60,585) Radiation therapy was associated with significant reductions in all-cause mortality in 4 of 7 cohorts.
Androgen deprivation therapy Cohort 3 (103,382) In instrumental variable analysis, ADT not associated with reduced prostate cancer mortality vs conservative management (HR, 1.01 [95% CI, 0.90-1.14]) or all-cause mortality (HR, 1.04 [95% CI, 0.99-1.09]).

Mortality results from other cohorts were mixed.

Small number of cohort studies available.

Potential confounding by indication in cohort studies of treatment effectiveness.

Potential bias in study estimates may limit applicability to US clinical practice. Fair
Key Question 4: Harms of Treatment
Radical prostatectomy RCT 3 (2524) Pooled RR of urinary incontinence with radical prostatectomy vs conservative management, 2.3 (95% CI, 1.8-2.8; I2 = 0.0%).

Pooled RR of erectile dysfunction, 1.6 (95% CI, 1.2-2.1; I2 = 87.5%).

No adverse effect of radical prostatectomy on physical or mental health status, anxiety, or depression relative to conservative management.

Inclusion of ProtecT in meta-analysis of erectile dysfunction may have increased heterogeneity because of (1) relatively long 6-y follow-up; and (2) crossover of >50% of men from conservative management to active treatment.

Studies had varying outcome definitions, inconsistent timing of follow-up measurements.

Many studies included predominately retropubic rather than robot-assisted radical prostatectomies.

Likely generalizable to US clinical practice. Fair to good
Cohort 11 (8809) Pooled RR of urinary incontinence with radical prostatectomy vs conservative management, 2.75 in 6 cohorts (95% CI, 1.78-4.23; I2 = 63.0%).

Pooled RR of erectile dysfunction, 1.49 in 7 cohorts (95% CI, 1.34-1.65; I2 = 59.2%).

Radical prostatectomy not associated with decrement in generic measures of quality of life vs conservative management.

RCT or uncontrolled
observational
2 RCTs and 6 cohorts (150,001) Median perioperative mortality after radical prostatectomy, 0.29% (range across 8 studies, including cohort studies and RCTs, 0.0% to 0.52%).

Thromboembolic or cardiovascular complications of radical prostatectomy ranged from 0.4%-9.0%.

Surgical complications required reintervention in ≈5% (US cohort).

Radiation
therapy
RCT 2 (1198) Radiation therapy not clearly associated urinary incontinence across 2 RCTs.

In ProtecT, radiation therapy not associated with erectile dysfunction vs active surveillance (RR, 0.9 [95% CI, 0.8-1.1]).

Bowel dysfunction more common with radiation therapy than conservative management in both trials.

No adverse effect of radiation therapy on physical or mental health status, anxiety, or depression, compared with conservative management (1 trial).

54.8% of men in ProtecT who were randomized to conservative management crossed over to active treatment, which may have increased rate of adverse effects in conservative management group.

Studies had varying outcome definitions, inconsistent timing of follow-up measurements.

Most studies of external-beam radiation therapy; relatively few studies of brachytherapy.

Likely generalizable to US clinical practice. Fair to good
Cohort 12 (4762) Marked variability across studies in estimates of risk of urinary incontinence with radiation therapy vs conservative management.

Pooled RR of erectile dysfunction with radiation therapy vs conservative management, 1.31 in 8 cohorts (95% CI, 1.20-1.42; I2 = 22.1%).

Bowel dysfunction more common with radiation therapy vs conservative management (5 cohorts).

Radiation therapy not associated with decrement in generic measures of quality of life vs conservative management.

Androgen deprivation
therapy
Cohort 3 (4662) Increased risk of erectile dysfunction with ADT (RR, range in RR, 1.6-2.9).

Worse function across several SF-36 domains (eg, physical function, vitality, emotional role).

Few studies; small sample sizes; varying outcome definitions; inconsistent timing of follow-up measurements. Likely generalizable to US clinical practice. Fair
High-intensity focused ultrasound Uncontrolled observational 7 (2239) Grade 2 urinary incontinence (ie, leaking with mild activity) or worse ranged from 0.0%-7.3%.

Among men with baseline potency, 37.3%-52.7% had erectile dysfunction after treatment (3 studies).

No studies compared HIFU with conservative management; only 1 study had a sample size exceeding 1000 men. Likely generalizable to US clinical practice. Fair
Key Question 5: Prebiopsy Risk Calculators to Predict Significant Prostate Cancer
PCPT and ERSPC risk calculators Cohort 14 (total 28 cohorts of men)
(48,234 biopsies)
Median AUC of PCTP, 0.72 (range, 0.51-0.88 across 21 cohorts) and 0.74 (range, 0.69-0.78 across 7 cohorts) for the ERSPC calculator, vs median AUC of 0.68 with PSA alone (range, 0.59-0.82 across 6 cohorts).

Calibration and decision curve analyses mixed for each calculator.

Most cohorts included many symptomatic men in addition to men referred for biopsy after abnormal PSA screening result.

ERSPC calculator derived with prostate volume determined by ultrasound rather than digital rectal examination.

No RCTs of calculator use.

Results may not generalize to men referred for biopsy after abnormal PSA screening result.

Unclear how risk information from calculators would influence biopsy decisions by actual physicians and patients.

Effects on long-term prostate cancer outcomes unknown.

Fair

Abbreviations: ADT, androgen deprivation therapy; AUC, area under the curve; HIFU, high-intensity focused ultrasound; HR, hazard ratio; PCPT, Prostate Cancer Prevention Trial; PSA, prostate-specific antigen; RCT, randomized clinical trial; RR, relative risk; SF-36, 36-Item Medical Outcomes Study Short Form Health Survey.
a For evidence pertaining to subgroups (eg, age, race/ethnicity, etc.) and general and disease-specific quality of life, please refer to the full evidence synthesis.

Return to Table of Contents

Figure 1 is the analytic framework that depicts the five Key Questions that guided the systematic evidence review. In general, the figure illustrates how PSA-based screening for prostate cancer in asymptomatic men may identify men with potentially early-stage prostate cancer who need to undergo additional diagnostic followup (i.e., needle biopsy of the prostate), the effectiveness and performance of prebiopsy risk calculators, and how screening may lead to improved outcomes or potential harms. In addition, the framework examines the efficacy of the range of treatments and any potential harms of those treatments

Evidence reviews for the US Preventive Services Task Force (USPSTF) use an analytic framework to visually display the key questions that the review will address to allow the USPSTF to evaluate the effectiveness and safety of a preventive service. The questions are depicted by linkages that relate interventions and outcomes. Refer to the USPSTF Procedure Manual for further details.6

Return to Table of Contents

Figure 2 is a flow chart that summarizes the search and selection of articles. There were 4,498 citations identified through literature databases and an additional 150 citations were identified from outside sources, such as reference lists and suggestions from peer reviewers. After duplicates were removed, 4,148 unique citations were screened at the title/abstract stage. The full text of 302 citations were examined for inclusion for one or more of the five Key Questions. The following number of articles were included for Key Question 1 (n=23), Key Question 2 (n=14), Key Question 3 (n=23), Key Question 4 (n=38), and Key Question 5 (n=13).

KQ indicates key question; USPSTF, US Preventive Services Task Force.
a Reasons for article exclusion: Relevance: study aim not relevant to key question. Setting: study not conducted in a country relevant to US practice. Intervention: study of excluded screening or treatment approach. Comparator: study lacked comparator group receiving either no screening (KQ1 and KQ2) or conservative management (KQ3 and KQ4). Population: study not conducted in a population at average risk for prostate cancer. Outcomes: study did not report relevant outcomes. Study design: design did not meet inclusion criteria for key question. Language: study published in non-English language. Quality: study did not meet criteria for fair or good quality. Unable to locate: full-text article could not be located.
b There were 17 new articles since the 2011 USPSTF review for KQ1, 9 for KQ2, 11 for KQ3, 15 for KQ4, and 14 for KQ5.

Return to Table of Contents

Figure 3 is a forest plot depicting the relative risk of urinary incontinence after radical prostatectomy for treatment of localized prostate cancer compared with conservative management approaches (i.e., watchful waiting or active surveillance).

ARD indicates absolute risk difference; RCT, randomized clinical trial; RR, relative risk.
a Conservative management encompasses management strategies in which active treatment is deferred or postponed indefinitely (ie, active surveillance, watchful waiting, or no treatment).

Return to Table of Contents

Figure 4 is a forest plot depicting the relative risk of erectile dysfunction after radical prostatectomy for treatment of localized prostate cancer compared with conservative management approaches (i.e., watchful waiting or active surveillance).

ARD indicates absolute risk difference; RCT, randomized clinical trial; RR, relative risk.
a Conservative management encompasses management strategies in which active treatment is deferred or postponed indefinitely (ie, active surveillance, watchful waiting, or no treatment).
b Data from Donovan et al66 are the source of marked heterogeneity, potentially because of the crossover of many active surveillance patients to radical treatments during the 6 years of median follow-up. Excluding those data and pooling only data from Wilt et al68 and Johansson et al88 results in RR = 1.82 (95% CI, 1.62-2.04; I2 = 0.0%).

Return to Table of Contents

Figure 5 is a forest plot depicting the relative risk of urinary incontinence after radiation therapy for treatment of localized prostate cancer compared with conservative management approaches (i.e., watchful waiting or active surveillance).

ARD indicates absolute risk difference; RCT, randomized clinical trial; RR, relative risk.
a Conservative management encompasses management strategies in which active treatment is deferred or postponed indefinitely (ie, active surveillance, watchful waiting, or no treatment).
b Meta-analysis not performed because of observed variability in the plots; no pooled estimates available.

Return to Table of Contents

Figure 6 is a forest plot depicting the relative risk of erectile dysfunction after radiation therapy for treatment of localized prostate cancer compared with conservative management approaches (i.e., watchful waiting or active surveillance).

ARD indicates absolute risk difference; RCT, randomized clinical trial; RR, relative risk.
a Conservative management encompasses management strategies in which active treatment is deferred or postponed indefinitely (ie, active surveillance, watchful waiting, or no treatment).

Return to Table of Contents