CNBH Association Française d’Urologie Collège National de Biochimie des Hôpitaux Société Française de biologie Clinique Société française de médecine Nucléaire – Groupe de Biologie Spécialisée GROUPE DE BIOLOGIE DE LA PROSTATE Niveaux de preuves pour l’utilisation du PSA et des autres biomarqueurs dans la détection précoce du cancer de la prostate Review of evidence for the use of PSA and other biomarkers in the early detection of prostate cancer Coordonnateur du groupe de travail: Pierre-Jean Lamy1 pierre-jean.lamy@icm.unicancer.fr Et Anne-Sophie Gauchez2 asgauchez@chu-grenoble.fr Laurent Salomon 3 laurent.salomon@hmn.aphp.fr Margaret Haugh4 mhaugh@medicom-consult.com Jocelyn Ceraline5 jocelyn.ceraline@chru-strasbourg.fr Yvonne Fulla6 yvonne.fulla@gmail.com 1 Agnès Georges7 agnes.georges@chu-bordeaux.fr Stéphane Larré8 stephanelarre@yahoo.fr Sylvain Loric9 sylvain.loric@hmn.ap-hop-paris.fr Elisabeth Luporsi10 e.luporsi@nancy.unicancer.fr Pierre-Marie Martin11 pierre-marie.martin@mail.ap-hm.fr Catherine Mazerolles12 mazerolles.c@chu-toulouse.fr Vincent Molinié13 vincent.molinie@chu-fortdefrance.fr Pierre Mongiat-Artus14 pierre.mongiat-artus@sls.aphp.fr Jacques Piffret15 jacques.piffret@orange.fr François Thuillier16 thuillierfrancois@sfr.fr Paul Perrin17 paul.perrin@chu-lyon.fr Xavier Rebillard18 xavier.rebillard@wanadoo.fr, et le groupe de relecture du biologie de la prostate : Drs, David Azria, Maguy Bernard, Karim Chick, Cyril Clavel, Stéphane Culine, Olivier Cussenot, Alexandre de la Taille, Aurélien Descazeaud, David Guenet, François Iborra, Jacques Irani, Igor Latorzeff, Marie-Pierre Moineau, Didier Peiffert, Pierre Richaud, Jean Marc Riedinger, Pascal Rischmann, François Rozet, Corinne Sault, Virginie Vlaeminck-Guillem. 1 Biologie Spécialisée et Oncogénétique, CRLC Val d’Aurelle, Montpellier France 2 UMR-S INSERM 1039, Institut de Biologie et de Pathologie CHU Grenoble, France 3 Service d'Urologie, APHP CHU Henri Mondor, Créteil, France 4 MediCom Consult, Villeurbanne, France 5 UMR_S 1113, FMTS, Université de Strasbourg, Strasbourg, France 6 Société Française de Médecine Nucléaire, Paris, France 7 Médecine 8 Service 9 Nucléaire, CHU Bordeaux, France d'urologie, CHU de Reims, Reims, France Biochimie clinique et génétique, APHP CHU Henri Mondor Créteil, France 10 Service d’Oncologie, Centre Alexis Vautrin, Nancy, France 2 11 Laboratoire de Transfert d’Oncologie Biologie, APHM, Marseille, France 12 Laboratoire d'Anatomie et Cytologie Pathologiques, IUCT Oncopole 1, Toulouse, France 13 Laboratoire d'Anatomie et Cytologie Pathologiques CHU La Meynard Fort de France, France 14 Service d'urologie, APH, CHU Saint-Louis, Paris, France 15 Association Française d’Urologie, Paris, France 16 Société Française de Biologie Clinique, Collège National de Biochimie des Hôpitaux Paris, France 17 Service d'urologie, Hôpital Lyon-Sud, Hospices Civils de Lyon, Lyon 18 Clinique Beausoleil, Montpellier, France 3 RESUME Contexte: Malgré des preuves contradictoires sur le bénéfice de l'utilisation de l'antigène prostatique spécifique (PSA) pour la détection précoce du cancer de la prostate celui-ci est actuellement largement utilisé. De nouveaux biomarqueurs visant à améliorer la valeur prédictive du PSA sont également utilisés. Objectif: Examiner systématiquement les données scientifiques sur l'utilisation du PSA et d'autres biomarqueurs pour la détection précoce du cancer de la prostate. Acquisition des données: nous avons cherché dans PubMed les essais cliniques et des études publiés entre 2000 et mai 2013, évaluant le PSA et d'autres biomarqueurs pour la détection précoce du cancer de la prostate, étude qui comprenait plus de 200 sujets. Le niveau de preuve de l’utilité clinique a été évalué en utilisant un système d’évaluation spécifique aux marqueurs tumoraux. Un total de 84 publications, correspondant à 70 essais et études ont été sélectionnés pour cette revue. Synthèse: Six essais cliniques randomisés évaluant le PSA ont été identifiés, mais quatre présentaient des faiblesses méthodologiques. Bien que ces essais aient inclus un grand nombre de sujets avec un long suivi, leurs résultats présentent des limites qui sont dues à la contamination (par des dosages antérieurs du PSA) dans le groupe de contrôle, à la qualité du suivi de ce groupe et de la variabilité dans les méthodes. Malgré ces limites, nous avons attribué un niveau de preuve IA (le plus élevé) pour l’utilisation du PSA pour la détection précoce, mais nous ne recommandons pas son utilisation dans le dépistage de masse. Les biomarqueurs émergents ont été évalués dans les études cas-témoins et de cohorte prospectives: PCA3 (n = 3); kallicréines (n = 3); [-2] ProPSA n = 5); oncogènes de fusion (n = 2). Ces études ont utilisé les résultats des biopsies pour le cancer de la prostate pour 4 déterminer la spécificité et la sensibilité des tests, mais elles n’ont pas évalué l'effet sur la mortalité. Le niveau de preuve attribué était III-C, insuffisant pour une utilisation en clinique. Conclusions: Le PSA peut être utilisé pour la détection précoce du cancer de la prostate, mais le dépistage de masse n’est pas recommandé. Les études sur d'autres biomarqueurs suggèrent qu'ils pourraient être utilisés, individuellement ou en combinaison, pour améliorer la sélection des patients avec des niveaux élevés de PSA en vue de la réalisation d’une biopsie, mais des essais cliniques évaluant leur impact sur la gestion du cancer de la prostate et sur la mortalité sont nécessaires. ABSTRACT Context: Despite conflicting evidence for the benefit of using of prostate specific antigen (PSA) screening in the early detection of prostate cancer (PCa), is currently widely used. New biomarkers aiming to improve the predictive value of PSA are also used. Objective: To systematically review the evidence for the use of PSA and other biomarkers in the early detection of prostate cancer. Evidence acquisition: We searched PubMed for clinical trials and studies assessing PSA and other biomarkers in the early detection of prostate cancer, published between 2000 and May 2013 that included >200 subjects. The level of evidence (LOE) for clinical utility was evaluated using the tumor marker utility grading system. A total of 84 publications, corresponding to 70 trials and studies were selected for inclusion in this review. 5 Evidence synthesis: Six randomised controlled trials (RCTs) assessing PSA were identified but four were found to have methodological weaknesses. Although these trials included large numbers of subjects and long-term follow-up, their limitations include contamination in the control group, lower quality follow-up in this group and variability in methods. Despite these limitations, we attributed a level of evidence (LoE) of IA to PSA for early PCa detection, but we do not recommend its use in mass screening. Emerging biomarkers were assessed in prospective case-control and cohort studies: PCA3 (n=3); kallikreins (n=3); [-2]proPSA n=5); fusion oncogenes (n=2). These studies used biopsy results for prostate cancer to determine specificity and sensitivity, but they did not assess the effect on PCa mortality. The LoE attributed was III-C. Conclusions: PSA can be used for early prostate cancer detection but mass screening is not recommended. Studies on other biomarkers suggest that they could be used, individually or in combination, to improve the selection of patients with elevated PSA levels for biopsy, but RCTs assessing their impact on prostate cancer management and mortality are needed. 6 Message pour les patients: Dans cette étude nous avons recherché quelles sont les preuves scientifiques pour l’utilisation du PSA et des autres biomarqueurs pour détecter précocement un cancer de la prostate. Le PSA reste le marqueur diagnostique standard du cancer de la prostate. Plus le PSA est élevé plus le risque du cancer est grand. A des niveaux élevés (> 10ng/mL), il y a un intérêt non discutable de rechercher un cancer par biopsie. Nous n’avons pas de preuves permettant de valider un dépistage de masse utilisant le PSA. Pour les autres biomarqueurs, les données suggèrent un intérêt de leur utilisation dans la sélection des patients avec un PSA élevé qui auraient besoin d’une biopsie. Néanmoins cela doit être confirmé dans des essais cliniques dédiés. Patient summary: In this study, we reviewed evidence for the use of PSA and other biomarkers for the early detection of prostate cancer. PSA remains the standard diagnostic biomarker in prostate cancer. PSA level is a continuous risk factor of prostate cancer. At high levels (>10ng/mL) the need for further investigation, e.g. biopsy is generally accepted. We did not find any evidence to support mass PSA screening. Data for other biomarkers suggest that these may have a role to play in selecting subjects with elevated PSA levels for biopsy but well-designed RCTs assessing their impact on prostate cancer management are needed. 7 Introduction Prostate cancer is the second most frequently diagnosed cancer in men worldwide (899 000 new cases, 13.6% of the total) and the fifth most common cancer overall. More than 70% of the cases occur in developed countries (644 000 cases). The incidence of prostate cancer varies worldwide, with the highest rates observed in Australia/New Zealand (104.2 per 100,000), and the lowest in South-Central Asia (4.1 per 100 000). Incidence rates are relatively high in some developing regions such as the Caribbean islands, South America and sub-Saharan Africa. This variation can be partly explained by differences in prostate specific antigen (PSA) testing and subsequent biopsies that are more frequently performed in countries with the highest incidence. With an estimated 258 000 deaths in 2008, prostate cancer is the sixth cause of cancer death in men (6.1% of the total). Because PSA testing has a much greater effect on the incidence of prostate cancer than on its mortality rate, there is less variation in mortality rates worldwide (10-fold) than observed for incidence (25-fold). Prostate cancers, which occur mainly in older men, are often slow growing. The worldwide weighted mean of the age-specific rates (ASR (W)) for prostate cancer mortality in developed regions is only twice that in developing regions (10.5 vs 5.6, respectively). These rates are generally higher in predominantly black populations (Caribbean, 26.3 per 100,000 and sub-Saharan Africa, 18-19 per 100 000), and very low in Asia (e.g. 2.5 per 100 000 in Eastern Asia) and intermediate in Europe, North America and Oceania. In Europe the incidence of prostate cancer is 59.3 per 100 000, with a mortality rate of 12.0 per 100 000. In France and the UK the mortality rates are similar, 12.7 and 8 13.8 per 100 000, respectively but the incidence is almost twice as high (118.3 and 64.0 per 100 000, respectively). This difference in the apparent incidence of prostate cancer may be explained by the fact that in France 50% of men ≥50 years have undergone PSA testing, compared with 10% in the UK. The impact of PSA testing on the incidence of prostate cancer detection was demonstrated through the surveillance of the incidence of prostate cancer in the US [1]. The introduction of PSA testing in the US resulted in an increase in the incidence of prostate cancer from 140/100 000 in 1987 to 240/100 000 in 1993. After this peak, there was a reduction in the incidence of detected prostate cancer to 170/100 000, because there were no more ‘undetected’ cancers, just new cancers developing. This incidence remains, nevertheless, higher than that prior to the introduction of PSA testing in 1987. PSA is a glycoprotein that is produced mainly in the prostate epithelial cells. In healthy men, PSA is generally concentrated in prostatic tissue and serum PSA levels are very low. PSA is an organ-specific marker rather than tumour specific and serum levels can be elevated in the event of various non-malignant (e.g. benign prostatic hyperplasia (BPH), prostatitis, trauma) and malignant prostatic diseases. Increasing levels of serum PSA have been shown to be associated with a higher risk of prostate cancer [2]. However, there is no clear-cut point that can differentiate insignificant cancers, which are not likely to be life-threatening, from those that are significant and likely to be life-threatening [2]. Screening can help to detect chronic diseases and cancers in people before they develop symptoms with the aim of being able to offer treatment before it is too late. In 1968 the World Health Organisation (WHO) commissioned a report on screening which, at that time, was becoming an important but controversial topic [3]. The report 9 listed criteria for screening. Over the years, other criteria have been proposed but they are mainly based on the WHO criteria [4]. One of these recent criteria is that there should be strong scientific evidence that the screening program is effective. From a public health point-of-view, the aim of prostate cancer screening is to improve overall survival and from an individual point-of-view, the aim is to increase prostate cancer disease-free survival and decrease the prostate cancer-specific mortality rate. In the USA prostate cancer screening began in the eighties when the majority of patients were diagnosed with advanced prostate cancer and a poor survival rate. Screening resulted in prostate cancer being detected at earlier stages and therefore the patients had higher disease-free survival rates. In addition, the lower PSA threshold introduced and the increased number of biopsy cores resulted in the detection and treatment of a large number of patients with low-volume and low-grade tumours [5]. Prostate cancer screening is now widely used in the developed world, although the scientific evidence supporting its benefits is controversial. One of the controversies concerns the choice between mass screening, i.e. offering screening to all men after a certain age, or targeted or individual screening, i.e. proposing screening to men ‘at risk’, also known as ‘early detection'. Another controversy concerns the PSA cut-point since many studies have shown that there is a continuum of prostate cancer risk at all values of PSA and no cut-point with a simultaneously high specificity and high sensitivity [6, 7]. Even at low PSA levels, e.g. 2.1 ng/mL, the results from Prostate Cancer Prevention Trial (PCPT) showed a sensitivity of 52.6% and a specificity of 72.5%. It is possible that some of the emerging biomarkers could improve the specificity of PSA and other prognostic factors and thus help in the management decision. 10 As part of an overall review on the role of PSA and other emerging biomarkers in detection, diagnosis and treatment of prostate cancer, we present here a review of the literature on the use of PSA and other biomarkers in the early detection of prostate cancer with an assessment of the level of evidence [8]. Methods PubMed was searched using combinations of the terms given in Table 1 to identify publications in English or French and published between 01/01/2000 and 06/05/2013 that evaluated PSA and other biomarkers in prostate cancer. This search was for a global systematic review to cover the use of biomarkers for early detection, prognosis, and prediction. In this manuscript only the results for the use of biomarkers in the early detection of prostate cancer are presented. The titles and abstracts of the references were screened to identify potentially pertinent references for the global review. A second screen on the titles and abstracts of references selected in the first screen was performed to identify potentially pertinent references for the present analysis on early detection of prostate cancer. Full papers were then obtained and relevant papers were selected to correspond to the following categories: Early detection with PSA in randomised clinical trials (RCTs) Early detection with PSA in systematic reviews/meta-analyses Early detection with PSA in ‘real-life’ studies Early detection with PSA in prospective cohort studies Early detection with emerging biomarkers International guidelines for early detection of PSA 11 In addition, the co-authors were asked to provide details of studies they knew about that had not been detected in the PubMed search and reference lists of included publications were scanned to identify other studies. Studies were eligible if they included > 200 men as smaller studies would be underpowered. For PSA, only randomized clinical trials or prospective cohort studies were included, since we knew that large randomized clinical trials were available. For the other biomarkers, we included randomized clinical trials and prospective and retrospective cohort studies. We did not include studies assessing genetic testing since these require biopsy tissue and are, therefore, not used in early detection of prostate cancer. The study characteristics and results were extracted into tables and verified by at least two of the authors. Results A total of 5825 publications were retrieved from the PubMed search and 75 additional publications were identified either by the authors or in the reference lists of included studies (Figure 1). A total of 2256 publications were selected after the initial screen on titles and abstracts; these were then rescreened to select potential publications for this review on biomarkers in early detection. The PDFs were obtained for 254 articles for more detailed screening. Finally, 70 studies were selected for inclusion in this review (Figure 1). Total PSA in early detection of prostate cancer in RCTs Although we identified five randomized clinical trials, only two were retained for analysis because the other trials had major flaws in their design resulting in high risk of bias. In the Stockholm trial, the participants were not randomly selected and so are not representative of the population [9, 10]. In addition, this study was carried out 12 many years ago and used diagnostic methods that are no longer used. Both the Stockholm study and the Norrkoping study reported a 10-fold higher risk of prostate cancer than the other studies so that their results are not generalizable to other countries [9-11]. The results from the Norrkoping trial are not comparable with the other trials because the criteria used was cytology [11]. The study performed in Quebec was judged to have a risk of bias due to no concealment of allocation and results not given from an intention-to-treat analysis [12]. The characteristics, quality assessment and results of the two trials included, the American Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial and the European Randomized Study of Screening for Prostate Cancer (ERSPC) are summarized in Tables 2, 3 and 4 [13-40]. The individual countries in the ERSPC trial had their own specificities in terms of the methods used (Table 2). The PLCO trial did not compare early detection with no early detection as there was a very high rate of opportunistic screening in the ‘unscreened’ group. This can be considered as a comparison of intense screening vs. less intense screening [5, 41]. In contrast, the ERSPC trial, which was conducted in countries where PSA testing is generally far less prevalent, reported a reduction in prostate-cancer mortality. Over time, the size of the reduction has continued to increase; however, the number needed to screen and the number needed to detect to prevent one death from prostate cancer are 1,055 and 37, respectively [14]. An analysis of the data at 11 years suggest that for every 1000 men who have a PSA test, 35.1 more cancers will be detected and 1 additional prostate cancer death will be prevented. It was calculated that to detect these cancers in the screening group, 267 biopsies and 56 prostatectomies would have to be performed. 13 The 95% confidence interval for prostate cancer mortality for both trials overlap (0.87–1.36 for PLCO; 0.68–0.91 for ERSPC); the overlap of the 95% confidence intervals, suggest that the trial results are not heterogeneous. - LOE for total PSA in early detection of Pca: Ia - Comments: there is a small effect or no effect on PCa mortality Total PSA in early detection of prostate cancer: systematic review / metaanalyses Two systematic reviews of RCTs assessing PSA in early detection have been published (Table 5) [42, 43]. The methods used in the trials included in these reviews differed and many methodological concerns were reported. No statistically significant reduction in prostate cancer-specific mortality was observed: RR=0.88 (95% CI=0.71 – 1.09) and RR=0.95 (95% CI=0.85 – 1.07). There was significant heterogeneity when all trials were analysed together; this disappeared when the trials judged to be at a high risk of bias were removed in one of the meta-analysis, but the reduction in prostate cancer-specific mortality remained non-significant: RR=0.89 (95%CI=0.77 – 1.04) [43]. The authors of one of the reviews concluded that there was no evidence from randomised clinical trials to support the routine use of screening for prostate cancer with PSA (with or without digital rectal examination (DRE) [42]. The conclusions on the other paper were that since any benefits from prostate cancer screening may take >10 years to accrue, men with a life expectancy of <10-15 years should be informed that screening for prostate cancer is not beneficial and can be harmful [43]. Total PSA in early detection of prostate cancer: ‘real-life’ studies 14 Three studies assessing PSA for early detection of prostate cancer in ‘real-life' settings were identified; one from the US, one from Austria and one from Japan (Tables 6 and 7). No benefit from PSA was observed in the American study, probably due to the high contamination rate in the control group [44, 45]. In contrast, in the Austrian study both the before/after comparison in the region where the screening was performed and the comparison with the whole country showed a reduction in prostate cancer-specific mortality; 0.70 (95% CI=0.57-0.87) and 0.92 (95% CI=0.870.97), respectively [46, 47]. In the Japanese study, 44% (1 224/2 775) of men diagnosed with prostate cancer lived in communities with mass screening and the prostate cancer-specific mortality rates were 32.9% and 56.4% in the communities with and without mass screening, respectively [48]. Total PSA in early detection of prostate cancer: prospective cohort studies We identified six prospective studies that assessed PSA for the early detection of prostate cancer (Tables 8 and 9) [6, 49-55]. These studies included between 300 and 39 213 participants (median: 12 215) and used different techniques for PSA testing (different tests and different thresholds) and different biopsy protocols. Most of these studies confirmed that the risk of prostate cancer increased with increasing PSA levels, although there was no consensus for the cut-point (Table 9). The authors of one study suggested that there was a continuum of risk and that this should be communicated to patients and healthcare professional [6, 50]. The smallest study reported interim results for 300 subjects with families with BRCA1/2 mutations and therefore, a genetic predisposition. The authors concluded that the results from the interim analysis suggested that screening men with a genetic predisposition can allow clinically significant prostate cancer to be detected. These results need to be 15 confirmed with the analysis of the whole cohort. In June 2013, the study was still recruiting; 2620 subjects had been recruited [56]. Summary of international guidelines for PSA in the early detection of prostate cancer The main recommendations from 13 international clinical practice guidelines, published from 2006 (one guideline) to 2012 (five guidelines) for PSA testing in the early detection of prostate cancer are summarised in Table 10 [57-70]. These guidelines were from the US (n=5), Canada (n=1), Japan (n=1), Europe (n=2), UK (n=2) France (n=1), and Belgium (n=1). None of the guidelines recommended mass screening, generally because the cost-benefit has not been demonstrated. The majority of the guidelines recommended individual testing for early detection in subjects aged <75 years old (with no comorbidities), with a life expectancy of more than 10 years or in at-risk subjects (family history, ethnic origins, e.g. Afro-West Indians, Afro-Americans). They also stress the need for the subjects to be informed before PSA testing. The recommendation for early PSA testing in men aged from 40 to 50 years old is becoming more widespread. The rationale for this is that it can help to evaluate and stratify the prostate cancer risk and therefore adapt the frequency of future testing based on the estimate risk of prostate cancer. A PSA level of <1 ng/mL seems to be generally accepted as a standard to define low risk of prostate cancer and therefore lower frequency or even stopping subsequent PSA testing. Summary of studies assessing PSA velocity (PSAV) and doubling time (PSADT) 16 A total of 1 RCT, 15 retrospective cohort studies and a systematic review were identified assessing the use of PSAV or PSADT for the early detection of prostate cancer were identified [50, 71-86]. Data for patients in the RCT (PCPT – Finasteride vs placebo) [50] suggested that PSAV was correlated with a higher risk of cancer at biopsy in both the placebo and treatment groups. In the placebo arm, the addition of PSAV in multivariate analysis that included PSA, led to a very small increase in the area under the curve of the receiving operating characteristic curve (AUROC) for predicting positive biopsy (0.702 to 0.709). Four of the retrospective cohort studies from the Rotterdam and Goteborg ERSPC study centers concluded that PSAV does not provide any advantage over PSA [7477]. The other 11 retrospective cohort studies concluded that PSAV was correlated with the risk of prostate cancer on biopsy and there was a correlation with the cancer stage [71-73, 78-85]. Three of the studies also concluded that there was no correlation between PSADT and the risk of prostate cancer on biopsy [83-85]. One systematic review [86] analyzed 87 articles. They concluded that here is little evidence that PSAV and PSADT provide additional information above that provided by total PSA (tPSA) alone. LOE for the use of PSAV for predicting positive biopsy: IIa (1 ancillary study of a RCT). Harms of PSA-based screening for prostate cancer Data about the harms of PSA-based screening for prostate cancer were reported by two of the randomized trials that assess the impact of PSA-based screening on prostate cancer mortality [14, 37]. A false positive was defined as a positive result and consequent workup with no histopathologic diagnosis of cancer within one year 17 of the screening test. In the entire ERPSC trial, 75.9% of men that underwent a biopsy because of an elevated PSA value had a false-positive result [14]. In the PLCO trial, after four PSA tests, men had a 12.9% cumulative risk of receiving at least one false-positive result (defined as a PSA level of ≥4.0 ng/mL and no prostate cancer diagnosis after 3 years) [37]. Neither of these trials reported data on the potential psychological harms of prostate cancer screening, such as anxiety, or its impact on health-related quality of life. Summary of studies assessing kallikrein 2 (hK2) in the early detection of prostate cancer We did not identify any randomised clinical trials assessing the use of kallikrein 2 (hK2) in the early detection of prostate cancer but we identified three prospective case-control studies (Table 11) [87-89]. Two of these studies included men who had been referred for biopsy, and the third included men referred because their PSA was ≥4 ng/mL or between, 3-4 ng/mL and they had an abnormal DRE. In this latter study the results from multivariate analyses showed that the risk of prostate cancer increased with increasing concentrations of hK2 and hK2/fPSA [87]. One of the other studies concluded that hK2 concentrations were statistically different in patients with biopsy-confirmed prostate cancer while the other did not find a statistically significant difference [88, 89]. In this latter study the ratios f/tPSAS, hK2/fPSA and hK2/(f/tPSA), but not hK2/tPSA were all statistically significantly different in patients with prostate cancer compared with those with BPH [89]. Thus, these studies, with the highest level of evidence identified, showed conflicting results for hK2 used alone. When used in combination with other measures of PSA there seemed to be some advantage, but the level of evidence remained low. 18 In addition, we identified 10 retrospective cohort studies and 3 retrospective casecontrol studies (Table 10). The results from some of these studies suggest that hK2 alone or in combination with other markers offers some advantages over PSA for selection men for biopsy. However, there are conflicting results from other studies. In addition, many of these studies assessed hK2 in populations with high rates of prostate cancer, which makes extrapolation to a population undergoing primary early detection difficult. LOE for kallikrein 2 (hK2) in the early detection of prostate cancer: conflicting data with LOE<II. Studies assessing emerging biomarkers in early detection of prostate cancer We identified 28 studies assessing other biomarkers: these studies were either prospective cohort or case-control studies (Table 12) [90-118]. The biomarkers assessed most often were PCA3 (prostate cancer antigen 3) and PHI (Prostate Health Index), a composite biomarker involving total PSA, free PSA and –(2)proPSA, an isoform of free PSA). Generally, the populations included in the studies had high rates of prostate cancer, which makes extrapolation to a population undergoing primary early detection difficult. Many studies included men who had already had a negative biopsy as well as those undergoing first biopsy. In addition, the biomarkers were often used in combination with different forms of PSA and included in a logistic regression model or an artificial neural network. The conclusions of the studies were heterogeneous. Sixteen studies assessed the PCA3 score in prostate cancer diagnosis in either prospective cohort or case-control studies using a variety of methods to determine the PCA3 concentration (Table 11) [101, 103, 104, 109, 111, 114, 116-118]. Almost 19 all studies suggest that PCA3 performs better than PSA. When compared with other emerging biomarkers, the results were contradictory for PCA3 being better or not. The ranges for the positive predictive value (PPV) and the negative predictive value (NPV) in those that reported these values were 50% to 70% and 35% to 90%, respectively. The studies used a wide range of PCA3 score cut-offs (Table 12). These studies do not provide evidence for the use of PCA3 for the early detection of prostate cancer but the results suggest there may be role for PCA3 in deciding which men with elevated PSA levels should undergo biopsy. LOE of PCA3 in patient selection for a second biopsy: II-b Five studies assessed pro-PSA (Table 12) [99, 102, 107, 112, 113]. These studies concluded that pro-PSA, in combination with other forms of PSA, could improve the detection of men more likely to have prostate cancer and thus reduce the number of unnecessary biopsies. However, the studies reported the AUROC and there were no clear cut-points. The men included in these studies had a high level of PSA and therefore had a high risk for prostate cancer; they were not representative of populations that would be targeted for early detection. LOE for PHI in patient selection for a second biopsy: II-b Five studies assessed the role of fusion proteins in men who had undergone PSA testing and/or biopsy [104-106, 108, 115]. One study reported a positive predictive value (PPV) of 0.94 and a negative predictive value (NPV) of 0.6 but 70% of the patients had prostate cancer [104]. Two studies reported that diagnostic accuracy increased when the fusion proteins were combined with other biomarkers [105, 115]. In one of the other studies all the patients had prostate cancer and in the other the negative controls were not necessarily biopsy-confirmed [106, 108]. These results do not support the use of fusion proteins in the early detection of prostate cancer, but 20 they suggest that they could be useful in selecting men with elevated PSA levels for biopsy. The men included in these studies were pre-selected and therefore the fusion proteins were not used in the context of early detection screening. LOE for fusions genes in patient selection for a second biopsy: II-b Discussion While the benefit of diagnosing cancer promptly in patients with symptoms with the hope of avoiding metastatic cancer and being able to provide effective treatment, is generally accepted, there is more controversy about detecting cancer in asymptomatic subjects. Cancer detection in asymptomatic subjects can be done either through mass screen programs in which all subjects are invited to participate, or through early detection programs in which subjects that may be at risk of developing a cancer are invited to participate. The discovery that higher PSA levels were associated with potentially lethal prostate cancer, which could be treated to increase the likelihood of disease-free survival revolutionized prostate cancer prognosis. Using lower thresholds results in improved prostate cancer diagnosis and the detection of earlier-stage tumours with a higher chance of cure when treated. Following the initial euphoria of this breakthrough, concerns increased about the over-detection and over-treatment of an increasing number of indolent prostate tumours that were similar to those found during autopsy of men who died from other diseases [119]. It has been estimated that the risk of having an indolent prostate cancer is approximately equal to the patient’s age minus 10, expressed as a percentage [120]. Thus in Europe, where male life expectancy in 2010 was 72.5 years, this risk is much higher than the lifetime risk of dying of prostate cancer, which is estimated to be 3% [5, 121]. In a long-term follow-up study of men with low-grade 21 prostate cancer 10-year prostate cancer survival was 97.2% supporting the low risk of these cancers[122]. In a setting of increased PSA testing, several randomized clinical trials were initiated; the two biggest were in the US (PLCO) and in Europe (ERSPC). In PLCO annual PSA testing for six years was accompanied by digital rectal examination (DRE) for the first four years; in ERSPC which was a ‘federation’ of multiple screening trials in different countries, the screening frequencies differed between the trials and this was not always associated with DRE. The results from PLCO did not show a reduction in prostate-cancer mortality although the rate of prostate cancer detection was higher in the screening group [123]. However, the control group was ‘contaminated’ by the high level of non-trial PSA testing in the community. The level of this contamination was lower in the countries included in the ERSPC study. The high level of prescreening would have reduced the number of men with undetected prostate cancer that could have been detected in the trials, and would therefore have lowered the statistical power to detect a reduction in mortality [123]. The latest results from the ERSPC, after 11 years of follow-up, showed a lower rate of prostate-cancer mortality in the screened group which continues to decrease over time. It was estimated that 1 055 men would have to be screened to prevent one prostate-cancer death and to detect 37 prostate cancers [14]. However, this would result in 267 extra biopsies, 56 prostatectomies and high levels of adverse events (e.g. incontinence, erectile dysfunction). For comparison, in two breast cancer screening trials in Sweden and the UK it was estimated that 8.8 and 5.7 breastcancer deaths, respectively would be avoided for every 1 000 women screened with 4.3 and 2.3 per 1000 women over-diagnosed, respectively [124]. This translates to between 2 and 2.5 lives saved for every over-diagnosed case. In addition, it has 22 been estimated that 12% to 13% of screened men had false-positive results after three to four screening rounds, and clinically important infections, bleeding, or urinary retention occurred after 0.5%–1.0% of prostate biopsies [123]. As PSA is expressed by the normal prostate gland, it is not cancer specific biomarker. PSA levels increase with age as the prostate gland naturally becomes bigger; levels are high in men with prostate cancer but they are also high in men with benign prostatic hyperplasia [125]. PSA values are continuous but to be useful in clinical practice a threshold value needs to be defined. When PSA levels are low, < 4ng/mL the risk of prostate cancer is relatively low and when the levels are high, > 10ng/mL the risk of prostate cancer is high and the clinical management of these patients is relatively clear. However, in the ‘grey zone’ between these levels there is uncertainty about the risk of prostate cancer. The positive predictive value (PPV) for a PSA between 4.0 ng/mL and 10 ng/mL is about 25%, i.e. less than one man in four will have prostate cancer detected on biopsy, whereas that for a PSA > 10 ng/mL is between 42% to 64% (refs). In addition almost three out of four cancers detected in the ‘grey zone’ are organ-confined with a good chance of being cured, whereas this is less than 50% of those detected with PSA levels >10 ng/mL (ref). Hence, in the grey zone, many unnecessary biopsies are performed because of the high false positive rate. Although the threshold used often is 4 ng/mL, some laboratories have started to use a threshold between 2 and 3 ng/mL, without strong evidence [126, 127]. It is generally accepted that a single test for total PSA with a unique threshold is not very informative and that it is better to assess its evolution over time, using the subject as their own control. This approach has been proposed for the early detection of ovarian cancer. Screening is based on patterns from serial CA-125 values to predict women at risk of preclinical ovarian cancer [128]. Compared with the use of 23 single fixed cut-off CA-125 measurement, the model was shown to improve sensitivity for the early detection of ovarian cancer. Although the World Health Organisation (WHO) has developed international standards for total and free PSA they are not used. For example, some assays use the Hybritech® standard that gives levels that are 20% to 30% higher than those obtained with the WHO international standard. Even with use of the international standard, quality control shows there is high intra- and inter-assay variability; these can be about 5% and up to 30%, respectively. In addition to the assay variability there are a number of factors that are known to affect the real level of PSA in samples, such as sport (cycling, jogging, horse-riding) sexual intercourse, manipulation of the prostate after biopsy, inflammatory infections of the urinary tract. Hence there are guidelines that should be respected to minimise the effect of these factors, including not to take samples within 48 hours of having practiced sport or having sexual intercourse and, above all, not to take samples from men with inflammatory conditions. The main concerns of the high false positive rate are the cost and the harms that arise from unnecessary biopsies. Indolent prostate cancers that are either subject to active surveillance or active treatment result in high costs, both medical and human. Active surveillance has economic implications for the healthcare system and the patient and has human costs for the patient and their family with an impact on their quality of life. The high rate of indolent prostate cancers found that are treated leads to significant side effects from the treatment and loss of quality of life for the patient and their partner [129]. The negative predictive value for PSA ≤4 ng/mL was estimated to be 85% in the PCPT [2]. Among the men with PSA ≤0.5 ng/mL who underwent biopsy, 6.6% 24 (32/449) had prostate cancer and 12.5% of these (4/32) had high grade cancer. The rate of prostate cancer and high-grade cancers increased up to 26.9% (52/449) and 25% (13/52), respectively in those with PSA between 3.1 and 4.0 ng/mL [2]. These data show that for all levels of PSA there is a risk of prostate cancer that increases with the PSA level. It seems reasonable to think that changes in PSA levels over time, (PSAV or PSADT) could be a better marker for more aggressive cancer compared with a single measure of total PSA. However, the results from the studies identified and the metaanalysis do not show they have any advantages over total PSA in the early detection of prostate cancer. Although we know from the pre-PSA testing era, when prostate cancer was mainly diagnosed by DRE, the lesions were frequently high-grade with high mortality rate. PSA testing has definitely enabled the detection of lower-grade, curable lesions. In the PSA era, the incidence of prostate cancer has drastically increased with no such increase in prostate-cancer mortality. Throughout the world, the incidence of prostate cancer varies widely. It is higher in countries where PSA testing occurs, while prostate-cancer mortality rates are more homogeneous. The challenge today is therefore to find a way to reduce the number of unnecessary biopsies and reduce the detection of indolent prostate cancer, while ensuring that patients with lesions that require treatment are identified early. In our review we found many studies that have attempted to increase the PPV by using other biomarkers for patients with PSA levels in this ‘grey zone’ before deciding to perform biopsy. However, to date, there have been no randomised controlled trials assessing these biomarkers. There have been many prospective cohort and case-control studies performed to assess the advantages of using hK2, fusion proteins, PCA3 and 25 pro-PSA (Tables 11 and 12). Although the results were not always concordant, many results suggest that these emerging biomarkers could reduce the number of unnecessary biopsies and improve the identification of men that should undergo biopsy. Some studies have evaluated the role of imaging for early detection of prostate cancer, but this was not the objective of this review. Although prostate ultrasound has not been shown to be useful, results from prostate magnetic resonance imaging (RMI) assessment studies look promising for the detection or differentiation of significant from insignificant prostate cancer [130]. In a setting where there is insufficient evidence to propose mass PSA screening, there does seem to be some merit to propose individual PSA testing [131]. To identify men who should be invited to undergo PSA testing, their risk factors should be taken into consideration [132]. In many guidelines, this approach to early prostate cancer detection is suggested for men aged between 50 and 70 years with a life expectancy of at least 10 years, with family history of prostate cancer, of a ‘high-risk’ ethnic and organochlorine pesticide exposure (e.g. chlordecone) (Table 10) [133]. Although not within the scope of this review, it will be important to have a management strategy for those men that are identified as being at risk of having prostate cancer in terms of biopsy or repeat PSA (and how regularly). Also outside the scope of this review but essential for the management of men who have positive biopsy results, we need to have evidence about who should be offered active surveillance and who should be offered active treatment. Conclusions 26 PSA has without doubt contributed to improve the prognostic of prostate cancer, enabling curable cancers to be identified, compared with the pre-PSA era. While it remains the standard biomarker in prostate cancer, PSA level is a continuous risk factor. At high levels (> 10ng/mL) the need for further investigation, e.g. biopsy is generally accepted. However, when the levels are lower, even below the traditional threshold of 4 ng/mL, other factors (presence of risk factors, results from DRE and possibly from imaging, such as RMI) need to be taken into consideration in the decision to propose biopsy with the aim of reducing over-diagnosis. In the current context where unorganized PSA testing in widely present and where various biomarkers are being developed for the early detection of prostate cancer, it would seem useful to suggest an individual management approach, integrating clinical and biological data in order to obtain a more specific diagnosis and risk assessment. This approach could be used to choose between prostate biopsy and active surveillance. Mass screening is not to be recommended currently; it would seem that the future lies with a personalised, multifactorial approach to early detection of prostate cancer. Acknowledgment: The authors are grateful to Julie Courraud for editing assistance. This work was supported by an unconditional grant from Beckman-Coulter and Hologic and by the scientific societies: Association Française d'Urologie, Collège national de Biochimie des Hôpitaux, Société Française de Biologie Clinique, Société Française de Médecine Nucléaire. 27 Identification Figure 1: Flow diagram of articles identified, screened and selected for inclusion in current review. A total of 70 trials and studies were included; these correspond to 84 articles as some had more than one publication Records identified through PubMed searching (n = 5825) Additional records identified through other sources (n = 75) Included Eligibility Screening Records after duplicates removed (n = 5900) 1st screen for all topics (n = 5900) Records excluded (n = 3644) 2nd screen for early detection studies (n = 2256) Records to be screened for other topics (n = 2002) Full-text articles assessed for eligibility (n = 254) Full-text articles excluded, with reasons (n = 174) Retrospective (for PSA) <200 men Not early detection Flaws in study design (n=4) Studies included in qualitative synthesis (n = 70) 28 Tables Table 1: PubMed search: terms used in combination as either MeSh terms or free text Prostatic Neoplasms / diagnosis / drug therapy / epidemiology / mortality / radiotherapy / surgery / therapy diagnosis / early diagnosis / prognosis randomized controlled trial / controlled clinical trial / clinical trial / meta-analysis / practice guideline / multivariate analysis / prognosis / prognostic / evidence-based medicine* PSA / pro-PSA / fusion oncogene / human PCA 3 / body mass index / prostatespecific antigen / tumor burden 29 Table 2: Characteristics of randomized clinical trials assessing PSA in the early detection of prostate cancer Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPC-global [13, 14, 16, 40] / Varied by center (see details below) 182,160 aged between 50 and 74 years (162,388 in core group: age: 55-69 years) in 8 European countries from 1993 to 2005 Serum PSA with Tandem-R/ Tandem-E Access assay (Hybritech) Screening interval = 4 years (Sweden = 2 years) External quality assessment (2 samples 6times/year) Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup Varied by center but in 2012 publication: 1995-1998: ≥3.4ng/mL 1999-2004: 2.9ng/mL After 2004: 2.5ng/mL 30 Sextant prostate biopsy; lateral sextant biopsies adopted in June 1996 Biopsy sample processing and storage and assessment Central guidelines [134] Placed in 10% neutralbuffered formalin and sent to lab in separatelynumbered containers Paraffinblocked and H&E stain Semiquantitative evaluation of amount of cancer Outcomes measured Primary: prostate cancer mortality Secondary: overall mortality; Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup ERSPCSweden [1720]/ 1st Invitation sent from January 1995December 1996 (Study became associate with ERSPC in 1996 – without any changes to protocol Follow-up to 31 December 2008 Randomisatio n before inclusion Men in Goteborg born between 1/1/30 and 31/12/44: N=9952 to screening and 109952 to control 76.0% screened >once All samples analysed in one laboratory Prostatus; Wallac Oy, Abo, Finland Screening every two years; in third round, men with <1 ng/mL found in second round were not invited Men ≥70 years no longer invited After clotting, serum obtained by centrifugation at 3000 g for 20 mins and stored frozen at -20°C within 3h from sampling PSA assay within 2 weeks of sampling and <3 of thawing ≥3ng/mL, lowered to 2.54ng/mL from the third round 31 Medical history; DRE; TRUS; laterally directed sextant biopsy Blind review of all medical records / pathology reports / autopsy protocols Biopsy sample processing and storage and assessment Central guidelines [134] Specimens stepsectioned in 4 mm increments Tumor areas measured with 1mm grid; volume estimated (multiplying the sum of tumor areas in consecutive sections – no correction for shrinkage) Outcomes measured Absolute and relative reduction in cumulative prostatecancer mortality Cumulative incidence of prostate cancer Proportion of screening attendees Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPCNetherlands [16, 17, 21-25] / Randomisatio n from Nov 1994-March 2000 Using population registries in Rotterdam and 12 neighbouring municipalities men aged 5574 years 21,210 to screening; 21,166 to control 94.6% screened >once All samples analysed in one laboratory (GP Lab) that covered Rotterdam and 7/12 of the municipalitie s (subjects from the remaining 5 were not included for PSA rates (only 11 tests) In 1st round: PSA-2 (Bayer); in 2nd round: Hybritech Tandem E (BeckmanCoulter) (after 2000 replaced with automated version Plus DRE and Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup Suspicious finding from any of the 3 diagnostic tests (PSA≥3ng/mL ) From November 1997 PSA threshold: ≥4ng/mL 32 laterally sextant needle biopsies Biopsy sample processing and storage and assessment Central guidelines [134] Placed in 10% neutralbuffered formalin and sent to lab in separatelynumbered containers Paraffinblocked and H&E stain Semiquantitative evaluation of amount of cancer in 4mm step sections Outcomes measured Primary outcome: prostate cancer mortality (blinded assessment by Cause of Death Committee) Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPC-Spain [17, 26, 27] / Recruitment from February 1996-June 1999 4278 men randomised (screening: 2416; control: 1862) aged between 4570 years in Madrid Mailed invitation (population registry list) 51.9% screened >once All samples analysed in one laboratory Screening interval: 4 years Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup 9 February 1996-12 May 1998: tPSA >4ng/mL; 12 May 19981 January 2002: tPSA >2.99ng/mL; From 1 January 2002present: tPSA >2.99ng/mL and tPSA 12.99ng/mL and f/tPSA ≤20% 33 TRUS-guided sextant prostate biopsy Biopsy sample processing and storage and assessment Central guidelines [134] Outcomes measured Primary: prostate cancer mortality Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPCBelgium [17, 28, 29] / Recruitment from June 1991December 2003 Follow-up until December 2007 (or up to 10 years) Population database of Antwerp city; men aged 5574 years (26.5% of invited participated) Randomisatio n after informed consent Screening: 5188; control: 5171 90.7% screened >once Initially all men had PSA, DRE and TRUS; from 1996 only PSA and DRE All screening done at one centre Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup 1992-94: suspicious finding from any of the 3 diagnostic tests (PSA≥10ng/m L) 1995-97: same with threshold at ≥4ng/mL 1999-2003 PSA and DRE with threshold: ≥3ng/mL 34 TRUS-guided prostate biopsy Biopsy sample processing and storage and assessment Central guidelines [134] Outcomes measured Prostate cancer mortality city population database; prostate cancer (national and local cancer registries) blinded assessment by Cause of Death Committee Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPC-Italy [17, 30] / October 1996October 2000 Randomisatio n before informed consent (Florence population registry) Age 55-69 years Screening: 7286; control: 7271 49.9% screened >once Screening interval: 4 years Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup PSA >4ng/mL; PSA ≥2.5ng/mL 35 Initially sextant random biopsy; currently: DRE and TRUS guided biopsy (transperineal ) Biopsy sample processing and storage and assessment Outcomes measured Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) ERSPCFrance [17] / Herault: June 2003-March 2005; Tarn: December 2000-June 2004 Randomisatio n before informed consent (health insurance list) in South-west France (Tarn and Herault) 1035 in each group Age 55-69 years Divided into previously screened/ no previous screen, then randomised within subgroup to screening or control 50.1% (Herault) and 49.0% (Tarn) screened >once Central testing (BeckmannHybritech) Every two years Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup PSA >3ng/mL 36 DRE and TRUS-guided sextant biopsy; recall of those with high-grade PIN, a negative biopsy with suspicious DRE/TRUS or PSA >10ng/mL Biopsy sample processing and storage and assessment Central guidelines [134] Outcomes measured Reduction of prostate cancer mortality Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup ERSPCFinland [17, 31-33] / January 1996January 1999 Randomisatio n before informed consent (mailed invitation) Men born in 1929-1944 in the metropolitan areas of Helsinki and Tampere Total 79,494 with 30,403 in screening group Mean age in both groups: 59.6 years 74.4% screened >once Central testing tPSA: Hybritech Tandem-E (Beckman Coulter) and Delfia (Wallac) assays until June 2001 then BeckmannCoulter assay f/tPSA: Wallac ProStatus (Wallac) Serum stored at -80°C 4 aliquots stored and since 1998 whole blood samples also stored PSA ≥4ng/mL PSA 3.03.9ng/mL : biopsy if suspicious DRE (199698) or if f/tPSA <0.16 (since 1999) 37 DRE, TRUS and TRUSguided sextant biopsy (in 2002 – 1012-core biopsy) from focal lesions, base, central and apex Biopsy sample processing and storage and assessment Central guidelines [134] Assessment done in four participating laboratories Outcomes measured Prostate cancer mortality Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup ERSPCSwitzerland [17, 34] / September 1998-August 2003 Mail invitation (55-70 years) in Canton Aargau Randomisatio n after informed consent 5150 in both groups 50.0% screened >once Screening interval: 4 years AxSym (Abbott) until June 2000 and Access (BeckmannCoulter Hybritech from July 2000 Serum frozen at -70°C within 2h of blood sampling Samples thawed immediately before PSA test within 2 weeks of sampling PSA >3ng/mL or PSA 13ng/mL and f/tPSA <20% 38 DRE and TRUS-guided transrectal sextant biopsy taken laterally from the peripheral zone of the prostate Biopsy sample processing and storage and assessment Central guidelines [134] Each biopsy separately processed and evaluated All positive and suspicious samples reviewed by an independent review pathologist Outcomes measured Study ID (ref) / Dates for randomisation and follow-up Population included (how selected, number, age) Detection / screening test(s) Serum processing and storage Definition of Protocol for positive for ‘positive’ further workup PLCO [35-39] From November 1993 to June 2001 Screening complete by October 2006 Follow-up: 31 December 2009 or 13 years from trial entry 76,693 aged between 55 and 74 years: 38,343 screen; 38,350 control in 10 centres across USA Annual PSA for 6 years and DRE for 4 years Single lab for testing using Tandem-R until Jan 1 2004 then Access Hybridtech PAS Centralised laboratory Serum frozen at -70°C or colder within 2-4 hours of blood collection Shipped weekly overnight on dry ice to central laboratory Spare sample stored at 70°C for future research PSA: ≥4ng/mL DRE: nodularity or induration, or investigator judgement of suspicion 39 No studyspecific protocol Local practice in each study center followed Certified tumor registrars ascertained stage, Gleason score and histology Biopsy sample processing and storage and assessment Outcomes measured Primary: prostate cancer mortality Secondary: incidence; staging; survival Table 3: PSA in randomized clinical trials: Trial design and quality characteristics Study ID (ref) Randomisatio n methods Sample size calculation ERSPC [13, 14, 16, 40] Italy, France, Finland and Sweden: randomisation before informed consent using population registries – Belgium, Netherlands, Spain and Switzerland: mailed invitation, then randomisation after informed consent Power 80% to detect 25% reduction in prostate cancer mortality with 10 years follow-up Number of screened/invit ed (%) Number of lost to followup Excluding France: 82.6% underwent screening at least once With France: 63.4% Median Blinding of follow-up time outcome (min and max) assessors Contaminatio n of control group Intention to screen analysis Median screen interval: 4.02 years Mean and median follow up (as of 2008) for core group excluding France: 10.5 and 11.0 – including France: 8.6 and 9.8 Results available for Spain and Netherlands (others are on-going) Spain: in general population aged 55-69 years – 86.8 per 1000 patient-years Netherlands: in population aged 55-69 years – 45 per 1000 patientyears Yes on core age group (55-69 years old) 40 Yes PLCO [35-38] Blocked with stratification on center, age and sex Power 90% (one-sided test) to detect 20% reduction in prostate cancer mortality with 10 years follow-up At 10 years, vital status known for 67% (although 23% had not been enrolled for 10 years) 11.5 years (7.2 to 14.8) 41 Cause of death from death certificate and medical records in an ‘unbiased’ manner PSA Yes screening: 40% in 1st year to 52% in 6th year DRE: from 41% to 46% Table 4: Results from randomized clinical trials assessing PSA in the early detection of prostate cancer Study ID (ref) Number of screening tests / screening rate Median screening interval / follow-up % positive Prostate cancer incidence (per 100 000) or rate (%) Screening group = 9.66 Control group = 5.95 Rate ratio = 1.63 (1.57 – 1.69) Prostate Overall cancer mortality rate mortality (n) (per 100 000) rate (per 100 000) ERSPC [14] mean=2.27/subj 4.02 years 16.7% Screening Screening ect (excluding group = 299; group = 18.2 France); rate 0.39 Control group 16.6% Control group = 18.5 (including = 462 rate = Rate ratio = France) 0.50 0.99 (0.97 – Rate ratio = 1.01) 0.79 (0.68 – 0.91) p=0.001 NNI = 1055 NND = 37 PLCO [35-39] Range from Every year / 7.9% At 7 years: At 7 years: 50 Screening 40% to 52% median=11.5 2820 vs 2322; vs 44; rate group = 2544 years (range rate ratio = ratio = 1.13 Control group 7.2-14.8) 1.22 (1.16 – (0.75 – 1.70) = 2596 1.29) At 13 years: Rate ratio = At 10 years: 158 vs 145; 0.97 (0.93 – 3452 vs 2974; rate ratio = 1.01) rate ratio = 1.09 (0.87 – 1.17 (1.11 – 1.36) 1.22) NNI = number of men needed to be invited to screening to prevent one death from prostate cancer; NND = number of cancers needed to be detected to prevent one death from prostate cancer 42 43 Table 5: Comparison of two systematic reviews of randomized clinical trials assessing PSA in the early detection of prostate cancer Djulbegovic et al 2010 [42] Ilic et al 2011 [43] Studies included (no. ERSPC (72 890 / 89 353) ERSPC (82 816 / 99 184) screened / no. control) PLCO (38 343 / 38 350) PLCO (38 343 / 38 350) Quebec (31 133 / 15 353) Quebec (31 133 / 15 353) Norrkoping (1 494 / 7 532) Norrkoping (1 494 / 7 532) French ERSPC (42 590 / 42 191) Stockholm (2 374 / 24 772) Gothenburg (9 952 / 9952) Total subjects screened / 153 812 / 160 540 156 160 / 185 191 All trials: RR=0.88 (95% CI=0.71-1.09) All trials: RR=0.95 (95% CI=0.85-1.07) control for prostate cancerspecific mortality Prostate cancer-specific Trials with a low risk of bias: RR=0.89 (95% CI=0.77-1.04) mortality results Trials with a high risk of bias: RR=1.05 (95% CI=0.871.25) 44 Djulbegovic et al 2010 [42] Prostate cancer diagnosis Ilic et al 2011 [43] RR=1.46 (95%CI=1.21-1.77) RR=1.35 (95%CI=1.06-1.72) ‘The existing evidence from RCTs does ‘Prostate cancer screening did not significantly decrease not support the routine use of screening prostate cancer-specific mortality’ for prostate cancer with PSA with or ‘Any benefits from prostate cancer screening may take without DRE’ >10 years to accrue, so men with a life expectance <10- results Conclusions 15 years should be informed that screening for prostate cancer is not beneficial and has harms’ 45 Table 6: Characteristics of ‘real-life’ studies on PSA in early detection Study ID Population included (how selected, number, age) Study design : data sources Outcomes measured SeattleConnecticut [44, 45] ● Two fixed cohorts from 1987-97 ● Population-based cohorts aged 65-79 (Medicare beneficiaries) ● Seattle: 94 900 Connecticut: 120 621 Observational cohort study Rate of PCa screening Medicare denominator and vital statistics databases, SEER-Medicare linked database, local SEER registry PCa mortality Tyrol [46, 47, 135] ● One region in Austria, Tyrol where PSA screening is free of charge to all aged 45-74 since 1988-89 ● Control: Austria without Tyrol where there is no free PSA screening ● In 1981, MS in 5/70 communities of Gunma (Japan); in 1986, MS in 22 Observational cohort study PCa mortality (by 5year categories) starting at 60 Gunma [48] Statistics Austria Mortality and Census data; Observational cohort study Cancer register 46 PCa diagnosis Table 7: Results from ‘real-life’ studies on PSA in early detection Study ID (ref) Followup Seattle15 Connecticut [44, years 45] PSA screening rates Prostate cancer mortality rate (per 100 000) Seattle: n= 94900 PSA / Seattle: 172.4 person year: 11 Connecticut: 171.1 858/325 873; rate per 100: 36.5 Rate ratio Seattle/Connecticut Connecticut: n= (95%CI°: 1.02 (0.96120621/ 1.09) PSA / person year: 136296/500 547 rate per 100: 27,2 Rate ratio Seattle/Connecticut (95%CI) = 1.35 (1.341.36) 47 Comments No benefit from PSA but in the US PSA is widely used, so that this non-randomised study is probably contaminated by the level of non-study PSA Study ID (ref) Followup PSA screening rates Prostate cancer mortality rate (per 100 000) Comments Tyrol [46, 47, 135] 18 years Estimated 75% ≥1 PSA test 2004-08 compared with rate in 1989-93: Tyrol: 0.70 (0.57-0.87) The observed reduction in mortality was considered to be too early and is more likely to be due to the role of PSA in the management of prostate cancer than the actual impact of early detection. Austria-Tyrol: 0.92 (0.87-0.97) These studies emphasise more the access to PSA testing and thus patient information in the screening Gunma [48] 15 years ND ND 1,224/2,775 (44%) of men with PCa lived in MS communities 48 Table 8: PSA in prospective cohort studies: study characteristics Study ID / design (ref) Rowe et al 2005/ prospective cohort study [49] Population included (how selected, number, age) ● Aged 50-65 years invited by GP letter to screening (6 practices in London, UK) ● 3652 contacted / 775 expressed interest / 773 accepted and gave a blood sample ● 20 min consulting and consent Detection / screening test(s) Sample processing and storage Single test with Serum Access assay for separated and tPSA and fPSA snap-frozen in (Beckman Coulter) liquid N2 detection limit 0,1 within 3h of ng/mL) blood sample STD: probably being drawn Hybritech 113/115 biopsies perform by one operator and all histology by one pathologist 49 Definition of positive If tPSA of 1.1-4.0 ng/mL and a f/tPSA of ≤20% tPSA ≥4ng/mL DRE and biopsy Protocol for ‘positive’ Outcomes measured Sextant biopsy for volume ≤30mL; two extra cores for volume 3040mL; ≥40mL six additional cores Primary: prostate cancer in men aged 50-65 with tPSA of 1.1-4.0 ng/mL and a f/tPSA of ≤20% Stat : Mann Whitney U test Comments Study ID / design (ref) Thompson 2007 / RCT) 2 prospective cohorts / [6, 50] Population included (how selected, number, age) ● 18,882 men aged ≥55 years, normal DRE, PSA<3, randomised to finasteride or placebo for 7 years. ● Annual PSA and DRE biopsy if DRE abnormal or PSA >4 ● All participants had end-ofstudy biopsy ● In this analysis: 4,400 from finasteride group and 5,587 from control group Detection / screening test(s) Central lab: Tandem E assay (Beckman Coulter) Sample processing and storage No details 50 Definition of positive PSA >4 or DRE abnormal Protocol for ‘positive’ Outcomes measured Comments Biopsy with ≥6 cores % positive biopsy; sensitivity, specificity, AUC, other risk factors in univariate and multivariate analyses Two reports, one each for ± finasteride Study ID / design (ref) Ishidoya et al 2008 / prospective cohort study [51] Population included (how selected, number, age) ● 2003 2004 ● Four centres of 11 communities in North Japan ● 5548 men aged 50-79 years enrolled ● No conditions for ineligibility Detection / screening test(s) PSA t and f measured 3-5h after collection (Architect, Abbott) STD: not specified (detection limit 0,1 ng/mL) Sample processing and storage Tested within 3-5h 51 Definition of positive tPSA: >4 ng/mL or tPSA: 2-4 ng/mL and f/tPSA: ≤12% for biopsy Protocol for ‘positive’ TRUS-guided 12-core biopsy (one pathologist made diagnosis based on biopsy specimen DRE only Gleason if biopsy evaluated Outcomes measured Comments Number of biopsies Number of positive biopsies Positive predictive value (PPV) No DRE in community 2001 2002 preliminary screening for cut off of ratio f/t 12% with sensitivity of 58,3 % and specificity of 77,8 % Stat: Stat view Study ID / design (ref) Antenor 2004 / Prospective longitudinal screening study [52] Population included (how selected, number, age) ● May 1991November 2001 ● 26,111 men 40++, mean 59 (40-96) ● 91 % White (90 % in table!!) ● 8 % black ● 2% others ineligible: ● prostatitis ● urinary infection ● anterior biopsy Detection / screening test(s) Sample processing and storage DRE and PSA at Limitations 6-month (if PSA (from authors) ‘suspicious’ ● only for or 1 year interval – white populati by on Tandem-E PSA ● inadeq (Hybritech) until uate May 2000, samplin (detection limit 0,5 g of ng/mL) then biopsy ACCESS analyser ● complia (Beckmann nce not Coulter) complet chemiluminescenc e of e subject + DRE(detection limit 0,1 ng/mL) STD? probably Hybritech < 20% WHO 52 Definition of positive Up to May 1995: >4.0 ng/mL or DRE findings– after 1995: >2.5ng/m L or DRE suspiciou s Protocol for ‘positive’ Outcomes measured TRUS-guided quadrant (sextant after May 1995) needle biopsy + additional for DRE or US suspicious zones Prostate cancer by initial PSA level No difference between 2 groups in insignificant cancer Stat: SPSS Comments Study ID / design (ref) IMPACT 2010 Ongoing, multicenter Prospective case/control study [53, 54] Population included (how selected, number, age) ● October 2005 to June 2008 ● 300 Men aged 40-69 recruited over 33 months in 20 centers in five countries ● Subjects recruited by postal invitation or approach of cancer genetics clinics in UK from families with mutations in BRCA1/2, ● Controls BRCA1/2 negative (but from families with mutations) ● Eligibility: 40 69 years, without PC and with BRCA1/2 ● Inegibility: PC <5 yrs, Detection / screening test(s) PSA annually (centralized in local laboratory) and analysed at a central reference laboratory (standardization and Quality assurance, methods not described) Central histology review, with a sample secondarily reviewed Screening: ● PSA < 3 :annual ● PSA > 3 :Biopsy if negative and PSA +50% other in 1 year ● If intraepitheli al or inconclusiv e other in 6 weeks Sample processing and storage Bio-repository for collection of blood, lympho urine and tissue For proteomic metabolomic and microarrays approaches Definition of positive PSA >3 ng/mL Protocol for ‘positive’ Outcomes measured Comments Prostate biopsy (10core recommended ) Positive biopsy rate Higher incidence of PC than in general population but limitations by lack of control group and few patients and the description of IMPACT STUDY if benign, PSA at 12 months re-biopsy if PSA >50% higher repeat biopsy after 6 weeks if inconclusive 53 Study ID / design (ref) Okihara 2008 / Prospective cohort (‘mass screening cohort’) [55] Population included (how selected, number, age) ● Otokuni district screening 39,213 male >55 years attending basic health check between 1995-2004 (10 years) ● Detection / screening test(s) First, Delfia PSA assay, cut off 4 (detection limit 1 ng/mL) second screening if PSA 4.1 to 10 Sample processing and storage Not indicated Definition of positive PSA 4.110 ng/mL Protocol for ‘positive’ First screening % of PCa PSA > 4 at biopsy Referral to central hospital for secondary screening: Biopsy if PSA > 10 or between 4-10 With DRE or TRUS + or both – with PSAD > 0.15. If PSAD < 0,15 again next year Biopsy: transperineal, systematic sextant; since 2002 – additional sample from far lateral region in both lobes (systematic octant biopsy) 54 Outcomes measured Comments Table 9: PSA in prospective cohort studies: study results Study ID (ref) Rowe 2005 [49] Thomps on 2007 [6, 50] Rate of PSA screening uptake ● 773/3,652 (21.2%) accepted ● 78.9 % Caucasian, 10.9 % afro Caribbean, 5% Asian, 3.4 % Middle Eastern, 1.8% Oriental ● age: mean 57.9 (50.1 – 64.9) ● Finasteride group: 4,440/5,676 (78%) PSA results Biopsy results Comments tPSA <4 ng/mL: 707/773 tPSA <1.1 ng/mL 352/773 tPSA=1.1-3.99 ng/mL: 355/773 eligible for free PSA f/tPSA of ≤20%: 137/355 115/137 agree for biopsy No. biopsy: 115/137 (84%) PCa detected: 13/115; rate 1 in 8.9 men (11.3%) At threshold of 3.5 ng/mL only 1/13 prostate cancer would be detected 4/13 had prostatectomy one was upgraded of Gleason 6 to 7 Finasteride PSA: Number (%) positive biopsy N=155 (6.2%) N=164 (15.3%) N=94 (24.9%) N=71 (35.9%) N=98 (52.7%) N=67 (57.8%) 0-0.5; N=2,489 0.6-1.0; N=1,073 1.1-1.5; N=378 16.-2.0; N=198 2.1-3.0; N=198 >3.0 55 Present method can reduce mortality PC particularly in 50 65 years men These men would have the widest range of locally curative treatment Not confirmed: overall incidence of PC if PSA between 1- 4 because men with ratio > 20 were not biopsied PSA level was a risk factor for PCa at biopsy in univariate: OR=3.49 (P<0.001) and multivariate: OR=3.48 (P<0.001) Study ID (ref) Rate of PSA screening uptake ● Placebo group: 5,587/8,575 (65%) PSA results Biopsy results Comments PSA > 4.0 ng/mL = normal DRE: N=577 (10.3%) PSA > 4.0 ng/mL = abnormal DRE: N=60 (1.1%) PSA ≤ 4.0 ng/mL = normal DRE: N=4,453 (79.7%) PSA ≤ 4.0 ng/mL = abnormal DRE: N=497 (8.9%) Sensitivity and specificity for PCa biopsy: Cut-point=4.1ng/mL 6.2% false positive rate, detect 20.5% of cancers. If lowered to 1.1 ng/mL would detect 83.4% of cancers but false positive rate of 61.1% In this study the authors recognised they had selected a healthier, more compliant population with generally low PSA levels – so could affect the generalizability to the general population 56 ‘Patients and healthcare professionals must be reeducated that there is a continuum of risk and no clearly defined cut-point for recommending biopsy’ Study ID (ref) Ishidoya 2008 [51] Rate of PSA screening uptake ● 5,548 subjects enrolled, 5079 years, no mean? ● biopsy in 6% of enrolled PSA results Biopsy results Comments ● tPSA >,337) ● tPSA 2-4ng/mL: 385/5,548 (6.9%); < 2ng/mL: 78.1% (44 ng/mL:14.9 % (826) with f/tPSA ≤12%: 100/5,548 (1.8% ) ● tPSA 4- 10 ng/mL: 6.9% (310) ● tPSA > 10 ng/mL: 1.4 % (75) Overall 138/332 (42.9%) tPSA >4ng/mL: 283/385 (73.5%) underwent biopsy 122/283 were PCa ● effected, % positive ● Overall: 332, 2.5% ● tPSA 2-4 ng/mL and f/tPSAt ≤12%: 16/49 (32.7% PPV) ● tPSA 4 10ng/mL: 218, 36.2% ● tPSA > 10ng/mL: 65, 66.2% f/tPSA ≤12%: detected PCa in men with PSA 2-4 ng/mL in 16 men / 49, 32.7% Gleason 3 or 4 T1c to T3, identical to 36.2% in 4-10 ng/mL tPSA difference in prevalence of PCa between Japanese and American or Europeans Limitations: sample size too small and follow up not long enough 57 Half of patients refused biopsy Men with PSA 4-10 ng/mL used to determine cut point of f/tPSA of 12% not yet verified future survey is expected Study ID (ref) Antenor 2004 [52] Rate of PSA screening uptake ● 26,111 enrolled ● mean followup: 51 months ( 0126) ● Age 40-50: 5% 51 60: 51 % 61 70: 33% >71: 12% ● DRE normal: 71% abnormal: 21% suspicious: 8% PSA results Biopsy results Comments tPSA <1.0 ng/mL:12,862 (50%) tPSA 1.1-2.5 ng/mL: 8,596 (33%) tPSA 2.6-4.0 ng/mL:2,416 (9%) tPSA %) 4.1-10.0 ng/mL: 1,866 (7%) tPSA >10.0 ng/mL: 351 (1%) Overall 2,122 (8%) diagnosed with prostate cancer PSA median at Ca diagnosis 4.3 ng/mL, to Ca diagnosis 20 ng/mL % clinically localized 99% % organ confined 72% % Gleason score > 7 biopsy: 15 % surgery: 26% RR PCa increased with PSA level 42% of those with PSA2.64.0 ng/mL increased >4.0 ng/mL vs. 2% and 12% for <1.0 ng/mL and 1.1-2.5 ng/mL, respectively Study demonstrates higher relative risk of cancer if PSA> 0.7 ng/mL (40 49 yrs ) or PSA >0.9 ng/mL (50 59) Initial screening PSA associated with risk of subsequent prostate cancer detection Authors’ Comments: Study useful for designing screening protocols Cost-benefit analysis would be required 58 Study ID (ref) IMPACT [53, 54] Rate of PSA screening uptake ● 99.7% PSA results Biopsy results Comments Year 1 PSA >3 ng/mL: BRAC1: 6/89; BRAC2: 11/116; Control: 5/95 Overall: 22/300 (7.3%) Year 2 PSA >3 ng/mL: BRAC1: 0; BRAC2: 5/51; Control: 1/42 Overall: 6/127 (4.7%) IMPA BR BR Con ER CT CA CA trols SP 1 1 2 C year Age 53 52 54 55 66 media n % 8.3 6.7 10. 7.4 20. PSA > 3 1 3 incide 3.3 4.5 3.4 2.1 5.3 nce of PC PPV 45.4 66. 36. 40.0 29. PSA 6 4 2 Year 1 BRAC1: 4/6; BRAC2: 4/11; control 2/4 Overall: 10/300 (3.3%) Year 2 BRAC1: 0; BRAC2: 1/4; control 0 Overall: 1/127 (0.8%) Interim results; recruitment to end December 2012 (expected n=1,700); all subjects will be screened for 5 years 59 Preliminary data from the IMPACT study show that there is a relatively low rate of biopsy 7% with PSA> 3 but PPV high 48%. Present study provides evidence that screening men with genetic predisposition detects clinically significant PC and support the rationale for continued screening in such men Comparison with ERSPC PSA ≥ 3ng/mL expected: recruitment to end December 2012 n=1,700; all subjects will be screened for 5 years in 32 centers through 11 countries Study ID (ref) Otokuni – Japan [55] Rate of PSA screening uptake ● From 1995 to 2004 more than two-fold increase in number of PSA ● 39,213 attended screening ● Between 1995-1998 could not detect first time primary screen, so only total number given, so estimated 65% uptake PSA results Biopsy results Comments PSA >4 ng/mL: 2,428 (6%) – 1,633 (67%) received secondary screening; 1,439 at central hospital 1st time secondary screen: biopsy recommended: 625/894 (70%) biopsy done: 606/625 (97%) prostate cancer: 183/606 (30%) PSA=4.1-10: biopsy recommended: 496/765 (65%) biopsy done: 482/496 (97%) prostate cancer: 118/482 (25%) PSA>10: biopsy recommended: 129/129 (100%) biopsy done. 124/129 (96%) prostate cancer 65/124 (52%) Prostate cancer detection rate 2x higher in those with PSA>10 ng/mL 60 Table 10: Summary of international guidelines for PSA in the early detection of prostate cancer Guideline / date updated (ref) Recommendations on early screening Comments European Association of Urology (EAU) / 2012 [63] ‘A baseline PSA determination at age 40 years has been suggested, upon which the subsequent screening interval may then be based (GR: B).’ ‘A screening interval of 8 years might be enough in men with initial PSA levels < 1 ng/mL.’ ‘PSA testing in men older than 75 years is not recommended because its early detection would not have any clinical impact’ European Society of Medical Oncology (ESMO) / 2010 [70] ‘Decisions on population screening await longer follow-up and the results of analyses of cost-effectiveness and quality of life [I, B]’ ‘Serum PSA should be measured and digital rectal examination (DRE) performed in appropriately counselled patients in whom there is clinical suspicion of prostate cancer or in those who wish to be screened.’ 61 Main conclusions on PSA for early detection 1. Not useful in those aged >75 years (although the rates of prostate cancerspecific mortality in France is highest in older men) 2. Possible advantage of PSA testing at 40 years old, since the PSA level is not ‘contaminated by BHP 3. If PSA < 1 ng/mL, no retest before 8 years PSA. This idea of PSA < 1 ng/mL is to be compared with the results from the study by Vickers et al (ref) showing that if a man aged 60 years has a PSA level < 1 ng/mL, the risk of prostate cancer-specific mortality is < 0.02%, therefore there is no need to perform another PSA test. The results from early detection studies currently correspond to a short follow-up and report only cancer-related outcomes. In terms of public health the important outcomes, are costeffectiveness and quality of life, for which there is very little data. It is essential that the patient is involved in the decision to undergo a PSA test; this test cannot be undertaken without pre-test information for the patient Canadian guidelines / 2011 [64] ‘the harms and benefits of PCa screening must be explained to each patient so they understand all the factors to be considered in the shared decision-making about screening’ ‘Prostate cancer screening should be offered to all men 50 years of age with at least a 10-year life expectancy.’ ‘Annual screening has been the standard; however, two screening studies demonstrate that screening is beneficial every 2 to 4 years.’ ‘If there is a higher risk of PCa, such as family history of PCa or if the patient is of African descent, screening should be offered at age 40 years.’ ‘there may be benefit in offering a baseline PSA for men 40 to 49 years of age to establish future PCa risk.’ ‘Initial screening should include DRE and PSA.’ 62 The Canadian guidelines are similar to the French guidelines: early detection is an individual decision; involves a DRE and a PSA assay; prior patient information is essential; is proposed to men aged ≥ 50 years (or 40 years, if an at-risk population); only in those with a life expectancy >10 years. The concept of an initial test at 40 years old to evaluate the risk of prostate cancer and therefore decide how regularly PSA testing should be done is beginning to seen. American Cancer Society / 2010 [57, 69] ‘The American Cancer Society recommends that men make an informed decision with their doctor about whether to be tested for prostate cancer.’ ‘Research has not yet proven that the potential benefits of testing outweigh the harms of testing and treatment. The American Cancer Society believes that men should not be tested without learning about what we know and don’t know about the risks and possible benefits of testing and treatment.’ ‘Starting at age 50, men should talk to a doctor about the pros and cons of testing so they can decide if testing is the right choice for them.’ ‘If they are African American or have a father or brother who had prostate cancer before age 65, men should have this talk with a doctor starting at age 45.’ ‘If men decide to be tested, they should have the PSA blood test with or without a rectal exam.’ ‘How often they are tested will depend on their PSA level.’ 63 No PSA testing without prior patient information and not in men under 50 years old, except in atrisk populations Testing based on the PSA level Frequency of testing dependent on the initial PSA level ‘In men with a life expectancy<10 years, it is recommended that general screening for prostate cancer with total PSA be discouraged, because harms seem to outweigh potential benefits.’ Type and strength of recommendation. Evidence based: strong. Strength of evidence. Moderate: based on five randomized clinical trials (RCTs) with intermediate to high risk of bias, moderate follow-up, and limited data on subgroup populations. ‘In men with a life expectancy>10 years, it is recommended that physicians discuss with their patients whether PSA testing for prostate cancer screening is appropriate for them. PSA testing may save lives but is associated with harms, including complications, from American Society unnecessary biopsy, surgery, or radiation treatment.’ of Clinical Type and strength of recommendation. Evidence based: Oncology (ASCO) / strong. 2012 [59] Strength of evidence. For benefit, moderate; for harm, strong: based on five RCTs (and several cohort studies) with intermediate to high risk of bias, moderate follow-up, indirect data, inconsistent results, and limited data on subgroup populations. ‘It is recommended that information written in lay language be available to clinicians and their patients to facilitate the discussion of the benefits and harms associated with PSA testing before the routine ordering of a PSA test.’ Type and strength of recommendation. Informal consensus: strong. Strength of evidence. Indeterminate: evidence was not systematically reviewed to inform this recommendation; however, randomized trials are available on the topic. 64 Strong guideline for no PSA testing in men with a life expectancy of <10 years For men who are to be screened, prior oral and written information must be given to them. U.S. Preventive Services Task Force / 2012 [67] Japanese Urological Association (JUA) / 2010 [65] HAS (French) / February 2012 [62] ‘Prostate cancer is a serious health problem that affects thousands of men and their families. But before getting a PSA test, all men deserve to know what the science tells us about PSA screening: there is a very small potential benefit and significant potential harms. We encourage clinicians to consider this evidence and not screen their patients with a PSA test unless the individual being screened understands what is known about PSA screening and makes the personal decision that even a small possibility of benefit outweighs the known risk of harms’ ‘The Japanese Urological Association (JUA) recommends prostate-specific antigen (PSA) screening, which can reduce the risk of death as a result of prostate cancer, for men at risk of prostate cancer. The recommendation is based on fact sheets showing the benefits and drawbacks of screening for prostate cancer. The JUA provides the best available screening system for men who want to be screened.’ ‘the best available evidence does not support recommending mass PSA screening for prostate cancer in the general population’ ‘no evidence was found to support recommending PSA screening in men with a high risk for prostate cancer’ 65 The US Preventive Services Task Force guidelines state that prostate cancer is a public health problem but that early detection only provides a small benefit and should only be proposed after informing the subject about the benefits and risks The Japanese Urology Association recommends early detection, particularly in at-risk subjects. No recommendations can be formulated for organised screening in men ‘at-risk’ since there is no evidence; the two randomised trials (ERSPC and PCLO) were in the general population. However, in men ‘at-risk’ early detection of prostate cancer with a PSA test can be performed on an individual basis Conclusions from report by Rachid Salmi [136]: No reason to question the recommendations from HAS – the two studies (PLCO and ERSPC) are too different – the European study is too heterogeneous for a meta-analysis to be performed Centre Fédéral d’Expertise des Soins de Santé (KCE) [61]) National Comprehensive Cancer Network (NCCN) [66] UK National Screening Committee (UK NSC) [68] British Association of Urological Surgeons (BAUS) [60] American Urological Association (AUS) [58] Not recommended to install or develop PSA massscreening program Opportunistic screening for those requesting to be tested, after receiving sufficient information Produced in 2006 PSA testing is likely optimal when used for early detection in high-risk populations instead of general population screening Young, healthy, high-risk men will benefit from early testing Older men with competing comorbidities should be carefully selected to avoid over -detection The UK NSC does not recommend screening men for prostate cancer. “… support the idea that men should be aware prostate cancer can be diagnosed earlier by PSA testing and biopsy, and that this can save lives. Men should be able to request and receive prompt PSA testing and subsequent management.” Early detection and risk assessment of prostate cancer should be offered to asymptomatic men 40 years of age or older who wish to be screened with an estimated life expectancy of more than 10 years No mass screening, and individual screening needs to be evaluated (guidelines produced before the publication of the results of ERSPC et PCLO in 2009) PSA testing is useful for at-risk populations and for young patients, with comorbidities. No mass prostate cancer screening organised in the UK (waiting for the results from the UK ProTect study in 2014) However the British urologists recommend informing men about the benefits of PSA testing After providing information, PSA testing started in men >40 years can enable early detection of prostate cancer and to evaluate the risk of prostate cancer NB: NICE have a guideline for diagnosis and treatment of prostate cancer, but they do not cover early detection: in the UK, there is no organised screening program for prostate cancer but an informed choice program, Prostate Cancer Risk Management, has been introduced. (Cancer Screening NHS 2012) 66 Table 11: Kallikrein 2 (hK2): Study characteristics and results Population included (how Detection / screening selected, test(s) number, age) Prospective case-control studies Nam 2000, / Serum from hK2: time-resolved June 1998324/404 (80%) immunofluorometric January consecutive assay (mouse MAb) 1999 / patients, in a fPSA and tPSA: Toronto, single center Immulite Canada [87] referred for chemiluminescence PSA ≥4 ng/mL system (Diagnostics or between 3-4 Products Corporation) ng/mL with Biopsy: Sextant USabnormal DRE guided; 18-gauge No information spring-loaded biopsy on biopsy device protocol Study ID / place (ref) Sample processing and storage Plasma separated and stored at -70°C 67 Outcomes measured Results Histological Cases: Prostate presence of cancer: n=159 prostate Controls adenocarcinoma ● Normal at biopsy prostate: n=83 ● BPH: n=35 ● PIN: n=47 hK2 (ng/mL): 1.18 vs 0.53; p=0.0001 hK2/fPSA: 1.17 vs 0.62; p=0.0001 Multivariate analyses: OR for prostate cancer increased with hK2 and hK2/fPSA Comments Study ID / place (ref) Scorilas 2003 / January 1992December 1997 /Padova, Italy [88] Population included (how selected, number, age) 345 men undergoing biopsy Targeted population: BPH: n=174 PCa: n=171 Detection / screening test(s) hK2: time-resolved immunofluorometric assay fPSA and tPSA: Immulite chemiluminescence system (Diagnostics Products Corporation) No information on biopsy protocol Stephan 2005 / 19972001 / Berlin, Germany [89] 475 referred men for biopsy, Age: mean=64 (range=43-86) Targeted population: BPH: n=128 PCa: n=347 hK2: Toronto research assay fPSA and tPSA: Immulite chemiluminescence system (Diagnostics Products Corporation) Sample processing and storage Serum obtained under standardized conditions, specimens were residuals from routine testing, stored at 70°C until analysis ND No information on biopsy protocol 68 Outcomes measured ? Results Cases: Prostate cancer: n=171 Controls: BPH: n=174 hK2: statistically different between groups For all tPSA ranges, hK2 was not significantly different for prostate cancer or BPH Ratios f/tPSAS, hK2/fPSA and hK2/(f/tPSA) were all statistically significantly different for prostate cancer or BPH; hK2/tPSA was not Comments Population Study ID / included (how place (ref) selected, number, age) Retrospective cohort studies Becker serum samples 2000a / PCa: from: 1991-1993 / ● healthy Malmo, volunteers Sweden; (25 female BPH: 1994and 25 male) 1997 / ● 54 BPH Michigan, US patients [137] ● 57 with advanced PCa ● 136 with localised Pca ● No information on biopsy protocol Detection / screening test(s) hK2 immunofluorometric assay; analytical detection limit: 0.01ng/mL; functional sensitivity: 0.05 ng/mL antibodies from: Liljas Clin Chem 1991 and Lovgren, J, et al Production of recombinant PSA and hK2 and analysis of their immunologic crossreactivity. Biochem Biophys Res Comm, 213: 888, 1995 PSA-T and PSA-F: DELFIA Prostatus PSA F/T Dual Assay Sample processing and storage Blood samples taken before any treatment BPH and local PCa: sera stored at 4°C for max 3 days, then at -20°C for max 1 week and then at -80°C for max 3 years Advanced PCa: sera stored at -20°C for max 7 years Samples thawed max 3 times prior to test. 69 Outcomes measured Sensitivity and specificity to identify pts ± PCa Results Comments Results for BPH vs local cancer vs advanced cancer hK2 (ng/mL): 0.055 vs 0.085 vs 0.57 p<0.0001 for all comparisons; hK2/tPSA: 1.6 vs 1.3 vs 1.4; NS or slightly significant; hK2/fPSA: 8.8 vs 13 vs 12 NS or slightly significant; hK2 X tPSA/fPSA: 0.34 vs 0.91 vs 5.9; p<0.0001 for all comparisons; Single , patients not recruited specifically for this study hK2 X tPSA/fPSA: AUC=0.81 p=0.025 Study ID / place (ref) Becker 2000b / Goteborg, Sweden (ERSPC) [138] Population included (how selected, number, age) serum samples from a randomized PCa screening trial (Goteborg Screening Study): 5853 out of 9811 accepted to participate. This study involved samples from 604/611 (92%) men who had agreed to undergo sextant biopsy with tPSA ≥3 ng/mL 144/145 samples from men with PCa included Detection / screening test(s) hK2 immuno-fluorimetric assay; In-house research assay (functional sensitivity 0.030 ng/mL, crossreactivity with PSA less than 0.01%) tPSA and fPSA: DELFIA ProStatus PSA F/T Dual Assay pre-analysis conditions defined (in house hK2 dosage) Sample processing and storage Serum separated from blood cells within 3h; frozen & stored at 20°C; t-PSA/f-PSA assays within 2 weeks of sampling and 3 h of thawing; hK2 assay within 3 years, using aliquots from sample at 1st thawing – stored at 20°C for max 2 years, and then at -70°C without thawing 70 Outcomes measured Sensitivity and specificity of biomarker to predict presence of PCa at biopsy, retrospectively Results Comments 604 with tPSA≥3.0 ng/mL: 460 benign and 144 PC of which: 541 with tPSA 3.0 – 10.0 ng/mL; 439 benign and 102 PCa AUC hK2XPSAT/FPSA = 0.807 AUC PSAT = 0.696 Significant improvement for sensitivity and specificity for PCa by combining hK2 with tPSA/f-PSA Population Study ID / included (how place (ref) selected, number, age) Steuber 2007 Single center / 1999-2000 / study Hamburg, 355 men Germany referred for [139] TRUS-guided sextant biopsy due to PSA ≥4 ng/mL or abnormal DRE High number of men with PCa: n=234 (66%), with 50% men undergoing repeat biopsy Biased population? TRUS-guided bilateral sextant biopsy of the peripheral zone Detection / screening test(s) Sample processing and storage hK2: 3-step immunoassay (details Becker et al Clin Chem 2000;46:198-206): analytical detection limit: 0.003ng/mL; tPSA and fPSA: DELFIA ProStatus PSA F/T Dual Assay Blood taken before manipulation; serum collected and immediately stored at 80°C until analysis Outcomes measured Results Comments Cancer vs no cancer: hK2 (ng/mL): 0.074 vs 0.060 hK2 did not add diagnostic information Univariate OR: in all patients: 45.9 (1.25-16.87) p=0.036, (OR should be 4.59?? but no correction published) in patients with PSA 2.0-9.99 ng/mL: 0.68 (0.0218.9) hK2 was predictive in whole cohort but not in PSA grey zone (2.0-9.9 ng/mL) Full model recommended: fPSA, fPSA-N, uPAR(I), suPAR(I– III), suPAR(II–III) (full model) AUC 0.779 71 Study ID / place (ref) Lilja2007 (and Vickers 2007) [140, 141] Population included (how selected, number, age) Participants aged <50 years old in a cardiovascular prevention project (MPM) in Malmo, Sweden; participants were linked to the Swedish Cancer Registry to identify those who developed PCa before end 1999; 3 controls per case, matched by age and date of PSA blood sample No information on biopsy protocol Detection / screening test(s) Sample processing and storage hK2 measured with a research assay with a functional detection limit of 0.005 ng/mL and ≤0.01% cross-reaction with PSA; t-PSA and f-PSA: DELFIA Prostatus PSA F/T Dual Assay EDTA-anticoagulated blood, rapidly centrifuged and stored at -20°C until analysis (not previously thawed) 72 Outcomes measured Predictive value of various markers, including hK2 for PCa on biopsy Results Comments A tPSA increase of 1 ng/mL was associated with an increase in odds of cancer of 3.69 (95% CI, 2.99 to 4.56); (AUC) using 10fold cross validation for three models: tPSA, cPSA, and the combination of tPSA, fPSA, %fPSA, and hK2. The AUCs were 0.762 (tPSA), 0.763 (cPSA), and 0.759 (combined markers). hK2 not evaluated alone Malmö Cohort: 21,277 patients (3350 ans) 462/498 PCa (93%) and 1222 controls Authors’ stated : « additional markers added little or no discriminative accuracy [to tPSA]” Study ID / place (ref) Bangma 2004/ Rotterdam, Netherlands (ERSPC) [142] Population included (how selected, number, age) Randomly selected samples for men:150 patients/group 1) normal + tPSA= 0.99.3ng/mL; PV= 15-40mL (n=143) 2) BPH + tPSA= 2.49.6ng/mL; PV=40-199mL (n=142) 3) prostate cancer + tPSA= 1.410.2ng/mL; PV=15-122mL (n=146) Sextant biopsies (number of cores, unknown) Anderson grading (not Gleason score) was used Detection / screening test(s) hK2: Access hK2 test – detection limit= 0.008ng/mL tPSA and fPSA: Access immunoanalyser (Beckman-Coulter) Sample processing and storage Archived samples stored at 80°C after having been processed within 3-4h of sampling – thawed once 73 Outcomes measured Results Comments Group 1 vs 2 vs 3 hK2: 0.07 vs 0.05 vs 0.04 – all comparisons statistically significant hK2 X (tPSA/fPSA): 0.55 vs 0.44 vs 0.31 – all comparisons statistically significant Levels of (7,5)proPSA, hK2 and fPSA could be used to distinguish between BPH and cancer, but proPSA and hK2, alone or combined, did not improve the specificity of fPSA for discriminating between BPH and cancer Study ID / place (ref) Becker 2000a / 1995-1996 / Göteborg, Sweden (ERSPC) [138] Population included (how selected, number, age) 604 men who underwent biopsy for PSA ≥3 ng/mL Detection / screening test(s) hK2: in house research assay: detection limit= 0.030 ng/mL fPSA and tPSA: DELFIA Prostatus PSA F/T Dual Assay Sample processing and storage Archived samples, processed within 3-4h of sampling, stored at 20°C for up to 2 years, thawed and aliquoted once and frozen at 70°C until analysis within 3 years for hK2 74 Outcomes measured Results Median serum concentrations of hK2 and tPSA were higher in men with positive biopsy than in those with negative biopsy In multivariate analyses, tPSA, fPSA and hK2 all contributed significantly to prediction of cancer or benign results from biopsy: For benign vs cancer hK2 X (tPSA/fPSA): tPSA≥3 ng/mL: 0.37 vs 0.67 tPSA 3-10 ng/mL: 0.31 vs 0.57 Comments Study ID / place (ref) Vickers 2008 / 1995-2005 / Göteborg, Sweden (ERSPC) [143] Population included (how selected, number, age) 740 men who underwent biopsy for PSA ≥3 ng/mL after 1st round PSA screening in ERSPC: 192 (26%) with prostate cancer recruitment OK for PCa Sextant biopsies (number of cores, unknown) Detection / screening test(s) hK2: in house research assay: detection limit= 0.035ng/mL fPSA and tPSA: DELFIA Prostatus PSA F/T Dual Assay Sample processing and storage Archived samples, processed within 3-4h of sampling, stored at 20°C for up to 2 years, thawed and aliquoted once and frozen at 70°C until analysis 75 Outcomes measured Results Comments hK2 (and tPSA): significantly higher in men with cancer Multivariate analyses: AUC for full clinical and laboratory models better than basic models (age + PSA): Laboratory model: 0.68 vs 0.83 Clinical model: 0.72 vs 0.84 Developed nomogram using a training set and an evaluation set Study ID / place (ref) Benchikh 2010 / 20012005 / Tarn, France (ERSPC) [144] Population included (how selected, number, age) 262/629 men who underwent biopsy for PSA ≥3 ng/mL after 1st round PSA screening in ERSPC: 83 with prostate cancer 11 395 men between 2001 and 2005 in ERSPC: subpopulation 4 200 (37%) of whom 629 (15%) high PSA: 370 (59%) prostate biopsy Sextant 12core biopsies Detection / screening test(s) hK2 and iPSA(intact PSA): with F(ab’)2 fragments of MAbs – to reduce non-specific interference. fPSA and tPSA: DELFIA Prostatus PSA F/T Dual Assay Sample processing and storage Archived samples, processed within 3-4h of sampling, stored at 80°C shipped on dry ice to Memorial SloanKettering Cancer Centre (US) in 2008 for hK2 testing, then shipped to Wallenberg Research Labs (Sweden) in 2009 for fPSA, tPSA and iPSA 76 Outcomes measured Using model from Rotterdam ERSPC for previously unscreened men number of biopsies avoided Results Comments Biopsy threshold ≥20% cancer risk: ● 492 (49%) biopsies avoided ● 61 (19%) cancers missed ● 12 (4%) high-grade cancers missed Using the full kallikrein panel with a risk threshold of 20% would reduce biopsy rates by more than 50% for men with elevated PSA while missing only a small number of cancers (31 out of 152 low-grade and 3 out of 40 high-grade cancers).Net benefit reported for biopsy decision using an algorithm with all four markers Study ID / place (ref) Vickers 2010a / 1993-2000 / Rotterdam, Netherlands (ERSPC) [145] Population included (how selected, number, age) 2,914 previously unscreened men undergoing biopsy as a result of elevated PSA (>3 ng/mL) in the Rotterdam section of the ERSPC. Training set: n=728; with cancer = 202 (28%); highgrade disease = 74 (10%). Validation set: n=2,186; with cancer = 605 (28%); highgrade disease = 219 (10%). No details on biopsy protocol Detection / screening test(s) hK2: in-house research assay – detection limit= 0.035 ng/mL fPSA and tPSA: DELFIA ProStatus PSA F/T Dual Assay Sample processing and storage Archived samples stored at 80°C after having been processed within 3-4h of sampling 77 Outcomes measured Results Comments Full laboratory (age, PSA, f+iPSA, hK2) and full clinical (age, PSA, DRE, f+iPSA, hK2) models were statistically better than base models Model for every 1000 men with elevated PSA, 513 fewer biopsies, but 66 men with cancer (mostly low-stage, low-grade) would be advised against biopsy Laboratory models Base: age, tPSA 0.557 (0.524, 0.590) — Full: age, tPSA, fPSA, iPSA, hK2 0.713 (0.682, 0.743) <0.001 Clinical models Base: DRE, age, tPSA 0.585 (0.551, 0.619) Full: DRE, age, tPSA, fPSA, iPSA, hK2 0.711 (0.681, 0.741) a panel of four kallikreins can help predict the result of initial biopsy in previously screened men with elevated PSA who have no history of negative biopsy. (replicated study) Study ID / place (ref) Vickers 2010b / 1997-2006 / Rotterdam, Netherlands (ERSPC) [146] Population included (how selected, number, age) 1,501 men who had biopsy after 2nd or 3rd round screen; cancer = 388 (26%) Detection / screening test(s) hK2: in-house research assay – detection limit= 0.035 ng/mL fPSA and tPSA: DELFIA ProStatus PSA F/T Dual Assay No details on biopsy protocol Sample processing and storage Archived samples, processed within 3-4h of sampling, stored at 80°C shipped on dry ice to Malmö in 2005-2007 Retrospective case-control studies 78 Outcomes measured Validation of model developed for unscreened men in previously screened men Results Comments Full laboratory and full clinical models statistically significantly better than base model for predicting any cancer or highgrade cancer Model for every 1000 men with elevated PSA, 362 fewer biopsies, but 47 men with cancer would be advised against biopsy resulting in 4 high-grade being missed a panel of four kallikreins can predict the result of biopsy for prostate cancer in men with elevated PSA. Study ID / place (ref) Vickers 2007 / 1981-1982 / Malmo, Sweden [141] Population included (how selected, number, age) Participants in MPM study: 501 cases with cancer from Swedish registry (31/12/1999), 1292 controls (age + date of blood sample) Analysed for 44-50 (younger) and ~60 (older) Detection / screening test(s) Sample processing and storage hK2 measured with a research assay with a functional detection limit of 0.005 ng/mL and ≤0.01% cross-reaction with PSA; PSA-T and PSA-F: DELFIA Prostatus PSA F/T Dual Assay EDTA-anticoagulated blood, rapidly centrifuged and stored at -20°C until analysis (not previously thawed) No details on biopsy protocol 79 Outcomes measured Results Comments Cases vs controls: hK2 (ng/mL) ● younger: 0.038 vs .032 ● older: 0.065 vs 0.037 Univariate OR hK2 ● younger: 1.75 (95% CI: 1.452.11) ● older: 3.09 (95% CI: 1.97-4.85) Univariate OR tPSA ● younger: 6.86 (95% CI: 4.919.60) ● older: 3.27 (95% CI: 2.12-5.05) Multivariate AUC with tPSA + fPSA + f/tPSA + hK2 0.819 vs 0.758 (older vs younger) p=0.03 Additional biomarkers aid in discrimination cancer/non cancer more for older men than for younger Population Study ID / included (how place (ref) selected, number, age) Vickers 2011 Participants in / 1991-1996 / MDM study Malmo, with PSA >3.0 Sweden ng/mL: 474 Cohort cases with design based cancer from on caseSwedish control data registry [147] (21/12/2005) No details on biopsy protocol Detection / screening test(s) hK2 measured with a research assay with a functional detection limit of 0.005 ng/mL and ≤0.01% cross-reaction with PSA; PSA-T and PSA-F: DELFIA Prostatus PSA F/T Dual Assay with WHO 96/670 -(PSAWHO) and WHO 68/668 (fPSA-WHO) standards For remaining cohort: PSA level imputed using described methods Sample processing and storage EDTA-anticoagulated blood, 80 Outcomes measured Validation of a previously published model developed on ERSPC (Rotterdam) including tPSA, fPSA, intact PSA and hK2 + age Results Full model was better than base model (tPSA + age); For prediction of a prostate cancer, the base model (age plus PSA) had a Cindex of 0.654 (95% CI: 0.621–0.683), which was significantly increased to 0.751 (95% CI: 0.726– 0.777) for the full model (age plus kallikrein panel). hK2 did not seem to have an important effect in the model Full model without hK2 0.752 (0.728– 0.782) Comments Population Study ID / included (how place (ref) selected, number, age) Becker Healthy 2000b / volunteers: 25 Goteborg, female and 25 Sweden and male Michigan, US BPH (negative [137] biopsy) = 54 localised cancer = 136 advanced cancer = 57 Sextant biopsies Detection / screening test(s) hK2: in-house research assay, detection limit = 0.05 ng/mL PSA-T and PSA-F: DELFIA Prostatus PSA F/T Dual Assay Sample processing and storage Blood taken before any treatment. Sera from BPH and local cancer patients stored at 4°C ≥3 days, then at -20°C for max 1 week and then -80°C for max 3 years. Sera from advanced cancer patients stored at 20°C for max 7 years. Sera thawed max 3 times 81 Outcomes measured Results Comments Sera from healthy volunteers all below hK2 detection limit. With the exception of hK2/tPSA and hK2/fPSA (and fPSA for BPH/local cancer) singly and combined the biomarkers were statistically significantly different for BPH/local, BPH/advanced and local/advanced Discrimination of men with and without PCa in a randomly selected population was improved by measuring hK2 in addition to tPSA and fPSA Table 12: Other biomarkers in cohort and case-control studies: study characteristics Study ID / design Population Detection / Sample Outcomes (ref) included (how screening processing measured selected, test(s) and storage number, age) 82 Results Comments Study ID / design (ref) Roobol 2010 / ERSPC Rotterdam rescreened cohort, prospective [95] Population included (how selected, number, age) ● Men invited for rescreening within ERSPC Rotterdam from September 2007 to February 2009 ● Single centre ● Men with PSA ≥3.0 ng/mL PSA or PCA3 score ≥10 were invited to undergo DRE, TRUS, and lateralised sextant biopsy ● n=965 including n=451 with 2 previous screens; n = 502 with 3 previous screen and n=12 with 4 previous screen ● 1/3 had Detection / screening test(s) PCA3: Progensa (in Radboud University, experimental laboratory) PSA: Hybridtech (in Erasmus University, clinical laboratory) Sample processing and storage No details 83 Outcomes measured Sensitivity, specificity to predict cancer; correlation between PSA and PCA3; ROC analysis Results Comments 721 men biopsied: 122 PCa (16.9%) PSA (>3.0 ng/mL): 35.2% with PCa and 69% without PCa PCA3 (>35): 68.0% with PCa and 55.7% without PCa; would have missed 39 PCa (5 serious) but avoided 48.3% biopsies Poor correlation between PSA and PCA3 Cannot extrapolate directly since men had been pre-screened with PSA low level of disease PCA3 as a 1st line screening test shows improved performance characteristics and identification of serious disease compared with PSA in a prescreened population. Study ID / design (ref) Crawford 2012 / prospective cohort study [91] Population included (how selected, number, age) 1 962 men with PSA (≥2.5ng/mL or abnormal DRE) in 50 centres referred for transrectal biopsy (>10 cores) 1913 informative samples Detection / screening test(s) PROGENSA PCA3 assay Sample processing and storage Post DRE urine sample before biopsy (first catch) Buffered samples shipped with frozen gel packs overnight to central lab samples analysed <48 hours after collection 84 Outcomes measured Correlation between PCA3 and histopathology and clinical outcomes Specificity of PSA (>2.5ng/mL) and PCA3 (>35 and >10) Results 802/1 913 had positive biopsy (42%) PCA3 >35: false +ve reduced from 1 089 to 249 (71%); false –ve increased from 17 to 413 (2 300% increase) PCA>10 false +ve decreased to 35.4%, false – ve increased 5.6% No correlation between PSA and PCA3 PCA3 but not PSA was relatively sensitive to suspicious changes found at biopsy PCA3 and PSA were positively correlated with increasing Gleason score AUC for PCA3 and PSA was better than PSA Comments PCA3 in conjunction with PSA could potentially significantly reduce the number of unnecessary biopsies Study ID / design (ref) Ferro 2012 / prospective cohort study [92] Population included (how selected, number, age) 151 men referred to one centre for 1st 18-core transrectal prostate biopsy with PSA between 2 and 20 ng/mL; DRE negative and no previous biopsy 50 yrs, no previous surgery, no treatment with 5alphareduct ase inhibitors, no prostatitis Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments PCA3: PROGENSA PSA: Access2 Immunoassa y System analyzer Serum stored at 80°C until assay First catch urine after attentive DRE, immediately before biopsy Comparison of Beckman coulter PHI: (p2PSA/fPSA)x tPSA and PCA score (PCA3 mRNA/PSA mRNA X 1000 in identification of PCa-ve, PCa+ve and HGPIN HGPIN: 24% PCa +ve: 32% (~90% significant) AUC: [-2]proPSA =0.73; PHI=0.77; PCA3=0.71 PHI=38.7: 85% sensitivity; 61% specificity) PCA3=32.5: 81%; sensitivity; 57% specificity [-2]proPSA , fPSA, PHI and PCA3 may be useful predictors of PCa at 1st biopsy; PCA3 seems to discriminate HGPIN from other noncancer conditions 85 Study ID / design (ref) Perdona 2013 / prospective cohort study [94] Population included (how selected, number, age) ● 160 men referred to one centre for 1st prostate biopsy (8-core TRUSguided) Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments Single laboratory PCA3: PROGENSA PSA: Access2 Immunoassa y System analyzer Serum stored at 80°C until assay First catch urine after attentive DRE, immediately before biopsy Diagnostic validity by ROC analyses for individual markers and for model-based scores PCa +ve: 29.4% (46.8% Gleason ≥7) [-2]proPSA , PHI and PCA3 significantly higher for PCa; fPSA significantly lower Univariable AUC: PHI=0.71; [2]proPSA =0.68; PCA3=0.66 (PHI and PCA3 not significantly different) Combined PHI and PCA3 better than single marker Individually PHI and PCA3 showed no difference in ability to predict PCa at 1st biopsy, but combined was better 86 Study ID / design (ref) Ochiai 2013 / prospective cohort study [93] Population included (how selected, number, age) ● 633/647 men with elevated PSA or abnormal DRE referred to four centers in Japan for 8core prostate biopsy (158 (24.4%) had previous negative biopsy) Detection / screening test(s) Sample processing and storage PCA3: PROGENSA f/tPSA in men with PSA 4-10 ng/mL; PV with US PSA density First voided predictive value urine of PCA3, PSA, sample after PV and PASD DRE 87 Outcomes measured Results Comments No relation between PCA3 and PSA PCa +ve: 41.7% PCA3 significantly higher in PCa +ve PCA3 <20: 16.0% PCa +ve PCA3≥50: 60.6% PCa +ve PCA3=35: sensitivity=66.5 %; specificity= 71.6% diagnostic accuracy=69.7% AUC: PCA3=0.748; PSAD=0.712; PV=0.706; PSA=0.583 PCA3 AUC significantly better than PSA but PSAD multivariate analysis PCA3, PSAD, PV and previous biopsy all independent predictors of Combined PSAD and PCA3 may be useful to avoid unnecessary biopsy Study ID / design (ref) Adam 2011 / prospective cohort [90] Population included (how selected, number, age) ● 105/107 men referred to two centres in South Africa for first (82%) or repeat 13core TRUS biopsy from July 2009 to February 2010 Detection / screening test(s) Sample processing and storage PSA: Beckman Coulter Access Hybridtech system PCA3: PROGENSA PV: TRUS Serum Predictive value within 24 of markers hours of biopsy First catch urine after DRE, within 24 hours of biopsy Blinded assessment by the same staff 88 Outcomes measured Results Comments PCa +ve: 42.9% Higher PCA3 in PCa +ve PCA3>35: sensitivity 77.7%; specificity=50%; NPV=75%; PPV=54% PCA3 independent of PV but PSA correlated with PV Sum of PCA3 and PSA was not better than PSA alone PCA3 performed best for PSA ‘grey zone’ (4-10 ng/mL) PCA3 was not superior to PSA; could be useful in men with higher PV and PSA between 4-10 ng/mL PCA3 score was higher and performed worse in Black patients Study ID / design (ref) Bollito 2012 / prospective cohort [98] Population included (how selected, number, age) ● 1,246 men with elevated PSA and negative DRE referred for first or repeat biopsy ● 3 centres Detection / screening test(s) Sample processing and storage PROGENSA 89 Outcomes measured Results Comments Unvariate and multivariate for PSA, fPSA and PCA3 to predict PCa +ve biopsy PCA3 cutoff 3950 best in repeat biopsy; >39 avoid 51.9% unnecessary biopsy – miss 7.8% cancers >50 avoid 56.5% unnecessary biopsy – miss 29 (10.3%) cancers (5 aggressive) Cannot be extrapolated since this was an observational study on 3,571 men who had PCA3 test. Were further selected for analysis 1,246 men who underwent prostate biopsy after PCA3 test (it is not a diagnostic study with systematic PCA3 and biopsies) PCA3 >39 useful in repeat biopsy; PCA3 not better than PSA in 1st biopsy Study ID / design (ref) Hessels 2007 / prospective cohort study [104] Population included (how selected, number, age) ● Men referred to one clinic (monocentrique)for biopsy for PSA ≥3 ng/mL or DRE ● n=108; age?NA PSA NA ● 72% PCa (nonrepresentative of the general population (30%) ● >8 biopsies (6 sextant peripheral zone and 2 transition zone + suspicious nodule by DRE) Detection / screening test(s) Sample processing and storage TMPRSS2ERG fusion transcript: rtPCR with SuperScript II RNase H Reverse Transcriptas e (Invitrogen) PCA3 as described in another publication: RT-PCR in sediments: normalizatio n by PSA First 30ml Diagnostic voided urine performance after DRE, immediately cooled on ice. Centrifuged at 4°C to obtain sediment – then snap frozen in liquid N2 and stored at -70°C 90 Outcomes measured Results Comments Fusion transcripts: Sensitivity:0.3 7 Specificity: 0.93 NPV: 0.36 PPV: 0.94 PCA3: (cutoff 58): Sensitivity: 0.62 Spe: 0.53 NPV: 0.35 PPV: 0.59 Combined with PCA3: Sensitivity: 73% 78/108 patients (72%) had positive biopsies ; this does not represent the typical patient population having a prostate biopsy based on elevated serum PSA levels Study ID / design (ref) Laxman 2006 / prospective cohort study [106] Population included (how selected, number, age) ● 19 Men with localized prostate cancer urine sampling prior to needle biopsy (n=11) or radical prostatectomy (n=8) ● n=19; mean age=60 years Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments TMPRSS2ERG fusion transcript, PCA3, RTPCR 30ml urine after DRE, collected in cups containing DNA/RNA preservatio n. Centrifuged to obtain sediment and stored at -20°C until RNA extraction Number positive 8/19 samples No comparison with a cohort of benign cases Correlation with the detection by FISH on tissue samples (biopsy or prostatectom y) 91 Study ID / design (ref) Tomlins 2011 / prospective cohorts (5) study [115] Population included (how selected, number, age) Men undergoing needle biopsy (2 cohorts) or radical prostatectomy (1 cohort) n= 1312; median age : 60 in RP cohort; 62 in academic cohort and 65 in community cohort (all baseline characteristic s are available in supplementar y material) 606/623 (97%) patients recruited in 2 centres: 269 PCa (44%) 187 patients had had radical prostatectomy 606 patients had positive biopsy No details on Detection / screening test(s) Sample processing and storage TMPRSS2:E RGa, PCA3 and PSA (to control for presence of prostate cells) using TMA Urine refrigerated immediately , and processed within 4h, mixed with urine transport medium (1:1) and stored at -70°C 92 Outcomes measured Results Comments Fusion in prostatectomy: +ve association with tumour volume, no. +ve cores, % cores with cancer, greatest cancer involvement in a single core; and maximum tumour dimension. Also difference in clinically significant / nonsignficant cancers Fusion in academic cohort: +ve association with no. +ve cores, % cores with cancer, greatest cancer and involvement in a single core. Also difference in cancer / no cancer Fusion in community cohort: Incorporation of TMPRSS2:ERG a and PCA3 into PCPT risk calculator improved clinical outcome for academic cohort but not community cohort No data available about diagnostic performances of the sole PCA3 score (only data about T2:ERG associated with PCA3 are available) Detection of TMPRSS2-ERG was correlated with tumor volume Study ID / design (ref) Laxman 2008b / prospective cohort study [105] Population included (how selected, number, age) ● Single centre ● total of 276 men undergoing needle biopsy (n=216) or radical prostatectomy (n=60) ● (but 257 informative samples), mean age = 62.8 (see supplementar y table S1) ; mean PSA = 7.9 ● analysis on 138 patients avec PCa (86 biopsies + et 52 radical prostatectomy ) and 96 patients with ve biopsies ● No details of biopsy protocol Detection / screening test(s) Sample processing and storage RNA and Transplex whole transcriptom e amplification qPCR for 7 biomarkers: PCA3, AMACR, GOLPH2, ERG, TMPRSS2:E RG, TFF3, SPINK1 First voided Association with urine after prostate cancer DRE, diagnosis collected in cups containing DNA/RNA preservative s. Centrifugati on to obtain urine sediments 93 Outcomes measured Results Comments Univariated analysis: OLPH2, SPINK1, PCA3 and TMPRSS2:ERG significantly associated with cancer Multivariated regression showed that a multiplex model with these biomarkers were better than serum PSA or PCA3 alone: ● Sensitivity: 0.659 ● Specificity: 0.760 ● PPV: 79.8% ● NPV: 60.8% PCA3 alone: ● Sensitivity: 0.75 ● Specificity: 0.56 ● PPV: 0.71 ● NPV: 0.61 ● AUC: 0.661 No data about inclusion criteria ; patients of the PR group were included in the cancer group when comparing to negative biopsy group Study ID / design (ref) Nguyen 2011 / prospective case control study [108] Population included (how selected, number, age) ● Single ● 3 groups: PCa-free (n=44); confirmed PCa (n=46); negative biopsy (n=11) ● total n=101 ● 46 PCa: ● -21 active surveillance, ● -11 Pretreatment, ● 4 metastatic ● No details of biopsy protocol Detection / screening test(s) Sample processing and storage Outcomes measured qPCR for a panel of TMPRSS2:E RG fusion markers First voided Number of urine (10-40 positives mL) after attentive DRE (females and postRP subjects were exempt from DRE as they have no prostate), collected in cups containing DNA/RNA preservative s Low-speed centrifuge at 4°C for sediment which was resuspende d in TRizol reagent for either immediate 94 RNA extraction or stored at -80°C until Results No PCa: 0/44 positive Confirmed PCa: 16/46 (35%) Negative biopsy: 2/11 Comments Study ID / design (ref) Groskopf 2006 / prospective cohort [101] Population included (how selected, number, age) ● Single centre ● patients scheduled for biopsy because PSA >2.5 and/or ‘other risk factors’ (n=70 but 68 informative samples; mean age=67); mean PSA = 7.7) ● PCa-free (n=52) ● Post-RP (n=21) ● Women (n=6) ● No details of biopsy protocol Detection / screening test(s) Sample processing and storage APTIMA PCA3 with probes for PCA3 and PAS First voided urine (20-30 mL) after an attentive DRE, kept on ice and processed with 4h, shipped overnight on cold packs and stored at 70°C for <8 months 95 Outcomes measured Results Cut-off: 50x10-3 Pre-biopsy: AUC=0.746; Sensitivity=69%, Specificity=79% PPV=0.5, NPV=0.89 (serum PSA specificity= 28%) Comments Study ID / design (ref) van Gils 2007 / prospective cohort study [117] Population included (how selected, number, age) ● Men undergoing biopsy in 5 centres with PSA between 3 and 15 ● N=583 but 534 informative samples, mean age=64.3 ● mean PSA=7.49 ● tPSA and fPSA determined as part of standard clinical practice – all other data collected prospectively ● standard TRUS biopsy: 8 biopsies (3 from each peripheral zones and 2 transition zone + suspicious nodule is Detection / screening test(s) Sample processing and storage Dual TRFbased qPCR for PCA3 From men, first voided urine after DRE; samples from community hospitals (4) cooled to 4°C; processed within 48h; samples at university hospital processed within 1h; centrifuged to obtain sediment, snap-frozen in liquid N2 and stored at -70°C 96 Outcomes measured Results PCA3: AUC=0.66 Serum PSA: AUC=0.57 Cut-point = 58: Sensitivity: 65% Specificity: 66% PPV:48% NPV: 80% PSA with same sensitivity: specificity=47% Higher PCA3 correlated with high probability of +ve biopsy Comments Study ID / design (ref) Hessels 2003 / prospective cohort study [103] Population included (how selected, number, age) ● 2 centres ● Consecutive men undergoing radical prostatectomy for PSA>3ng/mL ● n=108; NA ● Median PSA: 9.15 ng/mL ; mean PSA: 11.3 ng/mL ● Positive control group: 8 PR; Negative control group (cancer and non-prostate tissue) ● Validation on a cohort de 108 patients undergoing biopsy for PSA > 3ng/mL (24 with prostate cancer and 84 without. ● Radical prostatectomy samples. Detection / screening test(s) Sample processing and storage Outcomes measured Results DD3PCA3 transcripts using timeresolved florescencebased RTPCR Voided urine after DRE, cooled on ice, centrifuged for sediment, snap-frozen in liquid N2 and stored at -70°C ratio DD3PCA3/PSA AUC=0.72 cut point=200x10-3 Specificity=83% Sensitivity=67% NPV=90% PPV=53% AUC PSA:0.59 (calculated from raw data from Table 1) 97 Comments Study ID / design (ref) Deras 2008 / prospective cohort study [100] Population included (how selected, number, age) ● 4 centres ● Consecutive men undergoing biopsy for PSA≥2.5 ng/mL, DRE, family history or other risk factor ● N=570; mean and median age=64 ● Median PSA: 9.15 ng/mL ; mean PSA: 11.3 ng/mL ● TRUS guided biopsy with at least 10 cores taken Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments PCA3 and PSA mRNA with PROGENSA PCA3 Assay PSA assays performed at each centre (not centralised) Urine samples (first voiding; 2030 mL) after DRE, processed within 4 hours; mixed with equal volume of stabilisation buffer, stored at 70°C until tested Biopsy results PCA3 score <5: 14% biopsy positive PCA3 score >100: 69% biopsy positive AUC for whole population = 0.686 AUC for 1st biopsy = 0.703 Overall: 37%+ve biopsy: Sensitivity=54% specificity=74% PSA <4 ng/mL : 26%+ve biopsy: Sensitivity=50% specificity=77% PPV=54%, NPV=74% PSA 4-10 ng/mL : 38%+ve biopsy: Sensitivity=53% specificity=71% PSA>10 ng/mL: 48%+ve biopsy: Sensitivity=61% specificity=80% AUC PSA for whole population =0.547 280/570 had previous biopsy 98 Study ID / design (ref) Van Gils 2007 / Prospective cohort [116] Population included (how selected, number, age) ● Single centre ● Men undergoing TRUS-biopsy for high PSA or DRE ● N=67; mean age=64 ● Mean PSA: 8.73 ng/mL ● no information on biopsy protocol ● Detection / screening test(s) Sample processing and storage Quantitative APTIMA PCA3 test for PCA3 and PSA for mRNA Prostatic PCA3 and +ve fluid biopsy discharge during DRE collected in EDTA; 1st voided urine collected in two tubes with EDTA (sediments) Cooled to 4°C and mailed in cold packs to central lab for processing within 48h of collection 99 Outcomes measured Results 23/67 (34%) = +ve biopsy PCA3 scores for biopsy –negative vs. positive: Prostatic fluid: 18 vs 73; p<0.001; AUC=0.76 Urine: 19 vs 48; p=0.006; AUC=0.70 Cut off PCA3=66 in prostatic fluid: sensitivity =65% specificity =82% PPV=65% NPV=82% (compared with PSA: sensitivity =65%, specificity =64%) Cut off PCA3= 43 in urine: sensitivity =61% specificity =80% PPV=61% NPV=80%(comp ared with PSA: sensitivity =61%, specificity =64%) Comments Study ID / design (ref) Aubin 2011 samples from RCT (Dutasteride REDUCE) [96, 97] Population included (how selected, number, age) ● Men participating in the REDUCE RCT ● N=930 / 1072(dutaster ide/ placebo); median age: 62 in both groups ● Median PSA = 5.3ng/mL (dutasteride) and 5.4ng/mL(plac ebo) ● Ratio of the selected population representative of the overall population ● Initial negative biopsy (6 – 12 cores) ● Biopsies during surveillance of between 2 and 4 years (10 cores) Detection / screening test(s) Sample processing and storage APTIMA PCA3 test for PCA3 and PSA mRNA Urine samples obtained before year2 and year 4 biopsies after DRE; stored on ice and processed with 4h of sampling; equal volume of Gen-Probe transport medium, frozen at 70°C and shipped on dry ice to central lab Gen Probe recommend a maximum of 1h before mixing with the 100 transport medium Outcomes measured Results Comments PCA3 outperformed PSA, improved diagnostic accuracy when combined with PSA and other variables for men in both groups Placebo: PCA3 associated with +ve biopsy (p<0.0001); multivariate model – AUC = 0.753 (without PCA3 = 0.717) Dutasteride: multivariate model – AUC = 0.712 (without PCA3 = 0.660) PCA3 assay was better than PSA for cancer detection in men undergoing dutasteride treatment and improved the diagnostic accuracy when combined with the PSA level and other clinical variables. No adjustment needed for men in placebo group Study ID / design (ref) Wang 2009 / Prospective cohort [118] Population included (how selected, number, age) ● Single centre ● Men undergoing TRUS 12core sextant biopsy because of high PSA, history of HGPIN and/or %fPSA<15% ● N= 192 but 187 informative samples mean age: 62 ● Mean PSA=8.7ng/m L Detection / screening test(s) Sample processing and storage Outcomes measured Results Gen-Probe assay Initial void after DRE, processed with GenProbe transport medium and transferred to central lab <30°C correlation with +ve biopsy Overall sensitivity and specificity of PCA3>35 for +ve biopsy was 52.9% and 80%; PPV=69.7; NPV=66.1% Logistic regression after adjustment PCA3 independently associated with +ve biopsy (p=0.003) No. +ve biopsy increased with higher PCA3 score range (p<0.0001) 101 Comments Study ID / design (ref) Shappell 2009 / prospective cohort [110] Ouyang 2009 / prospective cohort [109] Population included (how selected, number, age) ● Single centre ● Men undergoing PCA3 testing and biopsy within a larger cohort ● N=35 ● No details of biopsy protocol Detection / screening test(s) Sample processing and storage TMA platform (Gen-Probe) Initial void after DRE, processed with GenProbe transport medium and transferred to central lab on cold packs ● Single centre ● Men undergoing biopsy ● N=106 but 92 informative samples; age NA ● 43 with; 49 without ● No details of biopsy protocol Quantitative RT-PCR Initial void correlation with after DRE, +ve biopsy urine centrifuged immediately and sediment homogenise d in TRIzol, transported to central lab, stored at -80°C 102 Outcomes measured Results Comments 9 (38%) had previous biopsy PCA3 sensitivity and specificity (without ASAP/HGPIN): 72.7% and 84.2% Including ASAP/HGPIN: 72.7% and 79.2%; PPV=62%; NPV=86% Cut-point: 19.9; AUC=0.67 (for PSA; AUC=0.59) Sensitivity=72%; specificity=59%; PPV=61%; NPV=71% High % of men with previous biopsies Study ID / design (ref) Tinzl 2004 / prospective cohort [114] Population included (how selected, number, age) ● Single ● Men referred for biopsy for elevated PSA or DRE ● N=201 but 158 informative samples; age 66 (64.8 for PSA mRNA positive urine) ● No details of biopsy protocol Detection / screening test(s) Sample processing and storage uPM3™ Initial void assay for after DRE, PCA3 mRNA immediately stabilised, stored at 4°C and processed within 48h. Centrifugati on to obtain sediments Cells were harvested and lysed, then snapfrozen, transported to central laboratory on dry ice 103 Outcomes measured Results Comments uPM3 (cutoff = 0.5): sensitivity: 82%; specificity: 76%; PPV: 69% NPV: 87%; AUC=0.87 tPSA (cut point=4 ng/mL): sensitivity: 87%; specificity: 16%; PPV: 40% NPV: 65% PCA3 with PSA 4-10 ng/mL : sensitivity: 84%; specificity: 80%; PPV: 67% NPV: 93% PCA3 with PSA <4 ng/mL: sensitivity: 73%; specificity: 61%; PPV: 45% NPV: 75% PCA3 with PSA >10 ng/mLmL : sensitivity: 84%; specificity: 70%; PPV: 83% NPV: 71% Several inconsistencies between tables and text or between tables Max PSA in the cohort = 1486! Study ID / design (ref) Sokoll 2008 / prospective cohort [111] Guazzoni 2011 / prospective cohort study [102] Population included (how selected, number, age) ● 2 laboratories ● 72 men with known biopsy outcomes ● N=72; median age: 61.6; median PSA=7 ● ≥10 biopsies Detection / screening test(s) Sample processing and storage Outcomes measured Gen-Probe assay for PSA mRNA and PCA3 mRNA at two sites Specimens stored at 70°C for up to 12months Correlation with +ve biopsy ● Single centre ● Men referred for biopsy with PSA 2.010 ng/mL and negative DRE ● n=268 (107 (39.9%) with cancer; age: 63.3 ● 12 to 22 biopsies ● Centralised analyses of biopsy samples Access tPSA fPSA, and 2ProPSA assays Blood sample before any prostatic manipulatio n 104 Results PCA3, continuous score: AUC = 0.706 in site 1 & 0.703 in site 2 cut point=35: 68,1% correctly classified Regression line slope (after log transformation) = 0.9677 Diagnostic %pPSA OR accuracy of (compared with p2PSA, [base model) = 2]proPSA and 4.889 (2.507Beckman Coulter 9.534); p<0.001 PHI vs (multivariate established analysis) predictors AUC = 0.83 (0.78-.89) Gain in predictivity = 0.11 (0.06-0.17) Comments Additionally they demonstrated a significant relation between Gleason score, [-2]proPSA (p = 0.002), and PHI levels( p < 0.001) Study ID / design (ref) Sokoll 2010 / prospective cohort study [112] Population included (how selected, number, age) ● Prior to prostate biopsy, in 4 US cancer centres ● n=566 (245 with cancer) age: 63.3 vs 60.5 (cancer vs noncancer) ● ≥10 biopsies Detection / screening test(s) Sample processing and storage Outcomes measured Analysed in 1 laboratory using access2 Immunoassa y System analyser for fPSA, tPSA (commercial Abs) and [2]proPSA Beckman Coulter Access p2PSA ( 1st study with a completely automatic test, PSAT access FPSA access See [111] Blood sampled before manipulatio ns, processed and serum stored at 80°C Sensitivity of %[- %[-2]proPSA 2]proPSA at fixed and tPSA were specificity significantly higher in cancer patients; %fPSA was lower AUC was higher when log tPAS, log %fPSA and log %[-2]proPSA were included for all patients and in the subgroups of PSA 2-4 ng/mL, 4-10 ng/mL and 2-10 ng/mL. 105 Results Comments %[−2]proPSA increased with increasing Gleason score (P < 0.001) and was higher in aggressive cancers (P = 0.03). Study ID / design (ref) Stephan 2009 / prospective cohort study [113] Population included (how selected, number, age) ● Men referred for biopsy in Berlin, Germany 2002-2006 ● n=586 (311 with cancer); age: 62.1 vs 67.2 (cancer vs noncancer) ● 8 to 12 biopsies Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments Access2 Immunoassa y System analyser for fPSA, tPSA (Hybritech PSA and free PSA kits) and [2]proPSA (RUO p2PSA kit) Test used automated Access test Blood sampled before manipulatio ns, and at least 3-4 weeks after, processed and serum stored at 80°C After thawing at RT, samples analysed within 3 hours ANN model AUC 0.85 (0.81–0.88) Sup to logistic regression p2PSA was higher in cancer patients (10.78 vs 9.71 pg/mL; p=0.021) PSA, %fPSA, [2]proPSA and p2PSA/%fPSA were consistently different between cancer and non-cancer patients for all tPSA range and for tPSA 2-10 ng/mL [-2]proPSA and p2PSA/ %fPSA showed their superiority to preferentially detect aggressive PCa (P<0.0001– 0.0008). Gleason The new marker p2PSA significantly differentiates between PCa and NEM in the 0–30 ng/mL tPSA range while the ratios [-2]proPSA and p2PSA/%fPSA discriminate also in the 2–10 ng/mL tPSA range. RAS 106 Study ID / design (ref) Catalona 2011 / multicentre, prospective doubleblind, case control trial [99] Population included (how selected, number, age) ● 1372 men enrolled in 8 centres from Oct 2003 to June 2009 ● Only 892 were analysed who had no history of prostate cancer, normal DRE, PSA 2 to 10 ng/mL and 6core or greater ● Prostate biopsy in a prospective multiinstitutional trial. ● 121 prospectively enrolled; 743 prospectively enrolled under separate protocols; 28 retrospective samples ● 98% had ≥10 biopsies and Detection / screening test(s) Sample processing and storage Outcomes measured Results Comments Access2 Immunoassa y System analyser for fPSA, tPSA and p2PSA Duplicate assay for p2PSA (showed good correlation r= 0.9985) Frozen at 70°C <8 h after sampling (which allowed accurate PHI measureme nt) Validation of prostate health index (PHI) in patients with PSA 2-10 ng/mL PHI= p2PSA/fPSA X PSA1/2 PHI and p2PSA were significantly higher in patients with cancer PHI use may avoid unnecessary biopsies An increasing prostate health index was associated with a 4.7-fold increased risk of prostate cancer and a 1.61-fold increased risk of Gleason score ≥ to 4+3=7 disease on biopsy 107 Study ID / design (ref) Liang 2011 / casecontrol study nested in the SABOR cohort [107] Population included (how selected, number, age) ● Single centre 474/500 (95%) patients analysed ● Controls matched for age and ethnic distribution with >5 years follow-up with no prostate cancer detected (not necessarily biopsyconfirmed) ● Cases with PSA measured with 2.5 years before diagnosis ● n=474 (227 with cancer); age: 64.1 ● 12 biopsies Detection / screening test(s) Sample processing and storage Test Access Beckman Coulter Quality control: 20 blinded duplicate samples randomly mixed among the samples + PHI After clotting for 30 min at RT and centrifugatio n, serum stored at 80°C Samples thawed and distributed into smaller aliquots 108 Outcomes measured Results Comments AUC (95% CI) fPSA: 0.76 (0.71..080) p2PSA: 0.72 (0.67-0.76) %fPSA: 0.76 (0.72-0.80) [-2]proPSA : 0.73 (0.68-0.77) The AUC for PSA, 0.84 (95% CI 0.81–0.88), was higher than typically reported because many cancer cases were referred to biopsy due to high PSA (sampling bias) and not all controls were biopsy confirmed. The AUCs for the other markers decreased for PSA between 2 to 10 ng/mL vs the entire PSA range, although the study was not powered to conduct this secondary subgroup analyses in this PSA range Of the new markers freePSA and %freePSA Only fPSA retained independent diagnostic usefulness compared with traditional risk factors, but there were some biases REFERENCES 1. Brawley OW. Prostate cancer epidemiology in the United States. World J Urol 2012;30(2):195-200. 2. Thompson IM, Pauler DK, Goodman PJ, Tangen CM, Lucia MS, Parnes HL, et al. Prevalence of prostate cancer among men with a prostate-specific antigen level < or =4.0 ng per milliliter. N Engl J Med 2004;350(22):2239-46. 3. Wilson JM, Jungner YG. [Principles and practice of mass screening for disease]. Bol Oficina Sanit Panam 1968;65(4):281-393. 4. Andermann A, Blancquaert I, Beauchamp S, Déry V. Revisiting Wilson and Jungner in the genomic age: a review of screening criteria over the past 40 years. Bull World Health Organ 2008;86(4):317-19. 5. Tangen CM, Goodman PJ, Thompson IM, Jr. Survival in the prostate cancer prevention trial. N Engl J Med 2013;369(20):1968. 6. Thompson IM, Ankerst DP, Chi C, Lucia MS, Goodman PJ, Crowley JJ, et al. Operating characteristics of prostate-specific antigen in men with an initial PSA level of 3.0 ng/ml or lower. JAMA 2005;294(1):66-70. 7. Connolly D, Black A, Gavin A, Keane PF, Murray LJ. Baseline prostate-specific antigen level and risk of prostate cancer and prostate-specific mortality: diagnosis is dependent on the intensity of investigation. Cancer Epidemiol Biomarkers Prev 2008;17(2):271-78. 8. Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst 2009;101(21):144652. 109 9. Kjellman A, Akre O, Norming U, Tornblom M, Gustafsson O. Dihydrotestosterone levels and survival in screening-detected prostate cancer: a 15-yr follow-up study. Eur Urol 2008;53(1):106-11. 10. Kjellman A, Akre O, Norming U, Tornblom M, Gustafsson O. 15-year followup of a population based prostate cancer screening study. J Urol 2009;181(4):161521; discussion 21. 11. Sandblom G, Varenhorst E, Lofman O, Rosell J, Carlsson P. Clinical consequences of screening for prostate cancer: 15 years follow-up of a randomised controlled trial in Sweden. Eur Urol 2004;46(6):717-23; discussion 24. 12. Labrie F, Candas B, Cusan L, Gomez JL, Belanger A, Brousseau G, et al. Screening decreases prostate cancer mortality: 11-year follow-up of the 1988 Quebec prospective randomized controlled trial. Prostate 2004;59(3):311-8. 13. Schroder FH, Hugosson J, Roobol MJ, Tammela TL, Ciatto S, Nelen V, et al. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 2009;360(13):1320-8. 14. Schroder FH, Hugosson J, Roobol MJ, Tammela TL, Ciatto S, Nelen V, et al. Prostate-cancer mortality at 11 years of follow-up. N Engl J Med 2012;366(11):981-90. 15. Blijenberg BG, Lilja H, Neels H, Stenman UH. Quality assessment for prostatespecific antigen (PSA) in relation to ERSPC: report of the PSA committee. BJU Int 2003;92 Suppl 2:66-70. 16. ERSPC. ERSPC Study book (available as supplementary material with Schroder 2012 NEJM), 1998. 110 17. ERSPC. Study web page: http://www.erspc-media.org/. Accessed: 10 August 2012 18. Hugosson J, Aus G, Bergdahl S, Fernlund P, Frosing R, Lodding P, et al. Population-based screening for prostate cancer by measuring free and total serum prostate-specific antigen in Sweden. BJU Int 2003;92 Suppl 2:39-43. 19. Hugosson J, Aus G, Lilja H, Lodding P, Pihl CG. Results of a randomized, population-based study of biennial screening using serum prostate-specific antigen measurement to detect prostate carcinoma. Cancer 2004;100(7):1397-405. 20. Hugosson J, Carlsson S, Aus G, Bergdahl S, Khatami A, Lodding P, et al. Mortality results from the Goteborg randomised population-based prostatecancer screening trial. Lancet Oncol 2010;11(8):725-32. 21. Otto SJ, van der Cruijsen IW, Liem MK, Korfage IJ, Lous JJ, Schroder FH, et al. Effective PSA contamination in the Rotterdam section of the European Randomized Study of Screening for Prostate Cancer. Int J Cancer 2003;105(3):394-9. 22. Otto SJ, van Leeuwen PJ, Hoekstra JW, Merckelbach JW, Blom JH, Schroder FH, et al. Blinded and uniform causes of death verification in cancer screening: a major influence on the outcome of a prostate cancer screening trial? Eur J Cancer 2010;46(17):3061-7. 23. Roobol MJ, Kirkels WJ, Schroder FH. Features and preliminary results of the Dutch centre of the ERSPC (Rotterdam, the Netherlands). BJU Int 2003;92 Suppl 2:48-54. 111 24. Schroder FH, Roobol-Bouts M, Vis AN, van der Kwast T, Kranse R. Prostatespecific antigen-based early detection of prostate cancer--validation of screening without rectal examination. Urology 2001;57(1):83-90. 25. Schroder FH, van der Cruijsen-Koeter I, de Koning HJ, Vis AN, Hoedemaeker RF, Kranse R. Prostate cancer detection at low prostate specific antigen. J Urol 2000;163(3):806-12. 26. Berenguer A, Lujan M, Paez A, Santonja C, Pascual T. The Spanish contribution to the European Randomized Study of Screening for Prostate Cancer. BJU Int 2003;92 Suppl 2:33-8. 27. Lujan M, Paez A, Pascual C, Angulo J, Miravalles E, Berenguer A. Extent of prostate-specific antigen contamination in the Spanish section of the European Randomized Study of Screening for Prostate Cancer (ERSPC). Eur Urol 2006;50(6):1234-40; discussion 39-40. 28. Nelen V, Thys G, Hermans A, D'Hooge K, Dourcy-Belle-Rose B, Coebergh JW, et al. Interval cancers in the Antwerp European randomised study of screening for prostate cancer study, using a 6 year screening interval. Eur J Cancer 2010;46(17):3090-4. 29. Nelen V, Thys G, Neels H, Neetens I, Coebergh JW, Dourcy-Belle-Rose B, et al. ERSPC: features and preliminary results from the Antwerp study centre. BJU Int 2003;92 Suppl 2:17-21. 30. Ciatto S, Gervasi G, Frullini P, Zendron P, Zappa M. Specific features of the Italian section of the ERSPC. BJU Int 2003;92 Suppl 2:30-2. 31. Kilpelainen TP, Tammela TL, Maattanen L, Kujala P, Stenman UH, Ala-Opas M, et al. False-positive screening results in the Finnish prostate cancer screening trial. Br J Cancer 2010;102(3):469-74. 112 32. Finne P, Stenman UH, Maattanen L, Makinen T, Tammela TL, Martikainen P, et al. The Finnish trial of prostate cancer screening: where are we now? BJU Int 2003;92 Suppl 2:22-6. 33. Makinen T, Tammela TL, Stenman UH, Maattanen L, Aro J, Juusela H, et al. Second round results of the Finnish population-based prostate cancer screening trial. Clin Cancer Res 2004;10(7):2231-6. 34. Kwiatkowski M, Huber A, Stamm B, Lehmann K, Wernli M, Hafeli A, et al. Features and preliminary results of prostate cancer screening in Canton Aargau, Switzerland. BJU Int 2003;92 Suppl 2:44-7. 35. Grubb RL, 3rd, Pinsky PF, Greenlee RT, Izmirlian G, Miller AB, Hickey TP, et al. Prostate cancer screening in the Prostate, Lung, Colorectal and Ovarian cancer screening trial: update on findings from the initial four rounds of screening in a randomized trial. BJU Int 2008;102(11):1524-30. 36. Andriole GL, Crawford ED, Grubb RL, 3rd, Buys SS, Chia D, Church TR, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med 2009;360(13):1310-9. 37. Andriole GL, Crawford ED, Grubb RL, 3rd, Buys SS, Chia D, Church TR, et al. 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):125-32. 38. Prorok PC, Andriole GL, Bresalier RS, Buys SS, Chia D, Crawford ED, et al. Design of the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. Control Clin Trials 2000;21(6 Suppl):273S-309S. 39. PLCO. https://www.plcostars.com/Default.aspx?projectid=2c74312f-2a93-4d34ba81-55604d8ebe50. Accessed: 9 August 2012 113 40. Schroder F. Supplementary Appendix. N Engl J Med 2012;366(11):981-90. 41. Goodman PJ, Thompson IM, Tangen CM, Parnes HL, Godley PA, Ford LG. Long-term survival of subjects in the prostate cancer prevention trial. J Clin Oncol (Meeting Abstracts), 2013:Abstract #10. 42. Djulbegovic M, Beyth RJ, Neuberger MM, Stoffs TL, Vieweg J, Djulbegovic B, et al. Screening for prostate cancer: systematic review and meta-analysis of randomised controlled trials. BMJ 2010;341:c4543. 43. Ilic D, O'Connor D, Green S, Wilt TJ. Screening for prostate cancer: an updated Cochrane systematic review. BJU Int 2011;107(6):882-91. 44. Lu-Yao G, Albertsen PC, Stanford JL, Stukel TA, Walker-Corkery E, Barry MJ. Screening, treatment, and prostate cancer mortality in the Seattle area and Connecticut: fifteen-year follow-up. J Gen Intern Med 2008;23(11):1809-14. 45. Lu-Yao G, Albertsen PC, Stanford JL, Stukel TA, Walker-Corkery ES, Barry MJ. Natural experiment examining impact of aggressive screening and treatment on prostate cancer mortality in two fixed cohorts from Seattle area and Connecticut. BMJ 2002;325(7367):740. 46. Oberaigner W, Horninger W, Klocker H, Schonitzer D, Stuhlinger W, Bartsch G. Reduction of prostate cancer mortality in Tyrol, Austria, after introduction of prostate-specific antigen testing. Am J Epidemiol 2006;164(4):376-84. 47. Oberaigner W, Siebert U, Horninger W, Klocker H, Bektic J, Schafer G, et al. Prostate-specific antigen testing in Tyrol, Austria: prostate cancer mortality reduction was supported by an update with mortality data up to 2008. Int J Public Health 2012;57(1):57-62. 114 48. Kubota Y, Ito K, Imai K, Yamanaka H. Effectiveness of mass screening for the prognosis of prostate cancer patients in Japanese communities. Prostate 2002;50(4):262-9. 49. Rowe EW, Laniado ME, Walker MM, Patel A. Prostate cancer detection in men with a 'normal' total prostate-specific antigen (PSA) level using percentage free PSA: a prospective screening study. BJU Int 2005;95(9):1249-52. 50. Thompson IM, Pauler Ankerst D, Chi C, Goodman PJ, Tangen CM, Lippman SM, et al. Prediction of prostate cancer for patients receiving finasteride: results from the Prostate Cancer Prevention Trial. J Clin Oncol 2007;25(21):3076-81. 51. Ishidoya S, Ito A, Orikasa K, Kawamura S, Tochigi T, Kuwahara M, et al. The outcome of prostate cancer screening in a normal Japanese population with PSA of 2-4 ng/ml and the free/total PSA under 12%. Jpn J Clin Oncol 2008;38(12):844-8. 52. Antenor JA, Han M, Roehl KA, Nadler RB, Catalona WJ. Relationship between initial prostate specific antigen level and subsequent prostate cancer detection in a longitudinal screening study. J Urol 2004;172(1):90-3. 53. Mitra AV, Bancroft EK, Barbachano Y, Page EC, Foster CS, Jameson C, et al. Targeted prostate cancer screening in men with mutations in BRCA1 and BRCA2 detects aggressive prostate cancer: preliminary analysis of the results of the IMPACT study. BJU Int 2011;107(1):28-39. 54. Mitra AV, Bancroft EK, Eeles RA. A review of targeted screening for prostate cancer: introducing the IMPACT study. BJU Int 2007;99(6):1350-5. 55. Okihara K, Kitamura K, Okada K, Mikami K, Ukimura O, Miki T. Ten year trend in prostate cancer screening with high prostate-specific antigen exposure rate in Japan. Int J Urol 2008;15(2):156-60; discussion 61. 115 56. www.ClinicalTrials.gov. The IMPACT Study - Identification of Men With a Genetic Predisposition to ProstAte Cancer: http://clinicaltrials.gov/show/NCT00261456. Accessed: 13 May 57. American Cancer Society. Prostate cancer: early detection, 2010. 58. American Urological Association (AUA). Prostate-specific antigen best practice statement: 2009 update: http://www.auanet.org/content/clinical-practice- guidelines/clinical-guidelines.cfm#2. Accessed: 24 September 2012 59. Basch E, Oliver TK, Vickers A, Thompson I, Kantoff P, Parnes H, et al. Screening for prostate cancer with prostate-specific antigen testing: american society of clinical oncology provisional clinical opinion. J Clin Oncol 2012;30(24):3020-5. 60. British Association of Urological Surgeons. BAUS Response to the UK NSC consultation on prostate cancer screening: http://www.baus.org.uk/AboutBAUS/publications/prostate-screening. Accessed: 24 September 2012 61. Centre Fédéral d'Expertise des Soins de Santé. Health Technology Assessment: prostate-specific-antigen (PSA) for prostate cancer screening: https://kce.fgov.be/publication/report/prostate-specific-antigen-psa-for-thescreening-of-prostate-cancer. Accessed: 24/09/2012 62. HAS. Rapport d'orientation. Cancer de la prostate : identification des facteurs de risque et pertinence d’un dépistage par dosage de l’antigène spécifique prostatique (PSA) de populations d’hommes à haut risque ? Paris: Haute Autorité de Santé, 2012:79. 63. Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, Mason MD, et al. EAU Guidelines on prostate cancer. Arnhem, The Netherlands: European Association of Urology (EAU), 2012:163. 116 64. Izawa JI, Klotz L, Siemens DR, Kassouf W, So A, Jordan J, et al. Prostate cancer screening: Canadian guidelines 2011. Can Urol Assoc J 2011;5(4):235-40. 65. Japanese Urological Association. Updated Japanese Urological Association Guidelines on prostate-specific antigen-based screening for prostate cancer in 2010. Int J Urol 2010;17(10):830-8. 66. National Comprehensive Cancer Network. Prostate cancer early detection: http://www.nccn.org/professionals/physician_gls/f_guidelines.asp#detection. Accessed: 24 September 2012 67. U.S. Preventive Services Task Force. Screening for prostate cancer: current recommendation: http://www.uspreventiveservicestaskforce.org/prostatecancerscreening.htm. Accessed: 24 August 2012 68. UK National Screening Committee. http://www.screening.nhs.uk/prostatecancer. Accessed: 24 September 2012 69. Wolf AM, Wender RC, Etzioni RB, Thompson IM, D'Amico AV, Volk RJ, et al. American Cancer Society guideline for the early detection of prostate cancer: update 2010. CA Cancer J Clin 2010;60(2):70-98. 70. Horwich A, Parker C, Bangma C, Kataja V. Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2010;21 Suppl 5:v129-33. 71. Loeb S, Sutherland DE, D'Amico AV, Roehl KA, Catalona WJ. PSA velocity is associated with Gleason score in radical prostatectomy specimen: marker for prostate cancer aggressiveness. Urology 2008;72(5):1116-20; discussion 20. 72. Martinez CA, Dall'Oglio M, Nesrallah L, Leite KM, Ortiz V, Srougi M. Predictive value of PSA velocity over early clinical and pathological parameters in 117 patients with localized prostate cancer who undergo radical retropubic prostatectomy. Int Braz J Urol 2004;30(1):12-7. 73. Sun L, Moul JW, Hotaling JM, Rampersaud E, Dahm P, Robertson C, et al. Prostate-specific antigen (PSA) and PSA velocity for prostate cancer detection in men aged <50 years. BJU Int 2007;99(4):753-7. 74. Roobol MJ, Kranse R, de Koning HJ, Schroder FH. Prostate-specific antigen velocity at low prostate-specific antigen levels as screening tool for prostate cancer: results of second screening round of ERSPC (ROTTERDAM). Urology 2004;63(2):309-13; discussion 13-5. 75. Schroder FH, Roobol MJ, van der Kwast TH, Kranse R, Bangma CH. Does PSA velocity predict prostate cancer in pre-screened populations? Eur Urol 2006;49(3):460-5; discussion 65. 76. Vickers AJ, Wolters T, Savage CJ, Cronin AM, O'Brien MF, Pettersson K, et al. Prostate-specific antigen velocity for early detection of prostate cancer: result from a large, representative, population-based cohort. Eur Urol 2009;56(5):753-60. 77. Wolters T, Roobol MJ, Bangma CH, Schroder FH. Is prostate-specific antigen velocity selective for clinically significant prostate cancer in screening? European Randomized Study of Screening for Prostate Cancer (Rotterdam). Eur Urol 2009;55(2):385-92. 78. Benecchi L, Pieri AM, Destro Pastizzaro C, Potenzoni M. Optimal measure of PSA kinetics to identify prostate cancer. Urology 2008;71(3):390-4. 79. Benecchi L, Pieri AM, Pastizzaro CD, Potenzoni M. Evaluation of prostate specific antigen acceleration for 2011;185(3):821-6. 118 prostate cancer diagnosis. J Urol 80. Loeb S, Kettermann A, Ferrucci L, Landis P, Metter EJ, Carter HB. PSA doubling time versus PSA velocity to predict high-risk prostate cancer: data from the Baltimore Longitudinal Study of Aging. Eur Urol 2008;54(5):1073-80. 81. Berger AP, Deibl M, Strasak A, Bektic J, Pelzer AE, Klocker H, et al. Large-scale study of clinical impact of PSA velocity: long-term PSA kinetics as method of differentiating men with from those without prostate cancer. Urology 2007;69(1):134-8. 82. Carter HB, Ferrucci L, Kettermann A, Landis P, Wright EJ, Epstein JI, et al. Detection of life-threatening prostate cancer with prostate-specific antigen velocity during a window of curability. J Natl Cancer Inst 2006;98(21):1521-7. 83. Connolly D, Black A, Murray LJ, Napolitano G, Gavin A, Keane PF. Methods of calculating prostate-specific antigen velocity. Eur Urol 2007;52(4):1044-50. 84. Krejcarek SC, Chen MH, Renshaw AA, Loffredo M, Sussman B, D'Amico AV. Prediagnostic prostate-specific antigen velocity and probability of detecting high-grade prostate cancer. Urology 2007;69(3):515-9. 85. Loeb S, Roehl KA, Catalona WJ, Nadler RB. Prostate specific antigen velocity threshold for predicting prostate cancer in young men. J Urol 2007;177(3):899902. 86. Vickers AJ, Savage C, O'Brien MF, Lilja H. Systematic review of pretreatment prostate-specific antigen velocity and doubling time as predictors for prostate cancer. J Clin Oncol 2009;27(3):398-403. 87. Nam RK, Diamandis EP, Toi A, Trachtenberg J, Magklara A, Scorilas A, et al. Serum human glandular kallikrein-2 protease levels predict the presence of prostate cancer among men with elevated prostate-specific antigen. J Clin Oncol 2000;18(5):1036-42. 119 88. Scorilas A, Plebani M, Mazza S, Basso D, Soosaipillai AR, Katsaros N, et al. Serum human glandular kallikrein (hK2) and insulin-like growth factor 1 (IGF1) improve the discrimination between prostate cancer and benign prostatic hyperplasia in combination with total and %free PSA. Prostate 2003;54(3):220-9. 89. Stephan C, Jung K, Soosaipillai A, Yousef GM, Cammann H, Meyer H, et al. Clinical utility of human glandular kallikrein 2 within a neural network for prostate cancer detection. BJU Int 2005;96(4):521-7. 90. Adam A, Engelbrecht MJ, Bornman MS, Manda SO, Moshokoa E, Feilat RA. The role of the PCA3 assay in predicting prostate biopsy outcome in a South African setting. BJU Int 2011;108(11):1728-33. 91. Crawford ED, Rove KO, Trabulsi EJ, Qian J, Drewnowska KP, Kaminetsky JC, et al. Diagnostic performance of PCA3 to detect prostate cancer in men with increased prostate specific antigen: a prospective study of 1,962 cases. J Urol 2012;188(5):1726-31. 92. Ferro M, Bruzzese D, Perdona S, Mazzarella C, Marino A, Sorrentino A, et al. Predicting prostate biopsy outcome: prostate health index (phi) and prostate cancer antigen 3 (PCA3) are useful biomarkers. Clin Chim Acta 2012;413(1516):1274-8. 93. Ochiai A, Okihara K, Kamoi K, Oikawa T, Shimazui T, Murayama S, et al. Clinical utility of the prostate cancer gene 3 (PCA3) urine assay in Japanese men undergoing prostate biopsy. BJU Int 2013;111(6):928-33. 94. Perdona S, Bruzzese D, Ferro M, Autorino R, Marino A, Mazzarella C, et al. Prostate health index (phi) and prostate cancer antigen 3 (PCA3) significantly 120 improve diagnostic accuracy in patients undergoing prostate biopsy. Prostate 2013;73(3):227-35. 95. Roobol MJ, Schroder FH, van Leeuwen P, Wolters T, van den Bergh RC, van Leenders GJ, et al. Performance of the prostate cancer antigen 3 (PCA3) gene and prostate-specific antigen in prescreened men: exploring the value of PCA3 for a first-line diagnostic test. Eur Urol 2010;58(4):475-81. 96. Aubin SM, Reid J, Sarno MJ, Blase A, Aussie J, Rittenhouse H, et al. PCA3 molecular urine test for predicting repeat prostate biopsy outcome in populations at risk: validation in the placebo arm of the dutasteride REDUCE trial. J Urol 2010;184(5):1947-52. 97. Aubin SM, Reid J, Sarno MJ, Blase A, Aussie J, Rittenhouse H, et al. Prostate cancer gene 3 score predicts prostate biopsy outcome in men receiving dutasteride for prevention of prostate cancer: results from the REDUCE trial. Urology 2011;78(2):380-5. 98. Bollito E, De Luca S, Cicilano M, Passera R, Grande S, Maccagnano C, et al. Prostate cancer gene 3 urine assay cutoff in diagnosis of prostate cancer: a validation study on an Italian patient population undergoing first and repeat biopsy. Anal Quant Cytol Histol 2012;34(2):96-104. 99. Catalona WJ, Partin AW, Sanda MG, Wei JT, Klee GG, Bangma CH, et al. A multicenter study of [-2]pro-prostate specific antigen combined with prostate specific antigen and free prostate specific antigen for prostate cancer detection in the 2.0 to 10.0 ng/ml prostate specific antigen range. J Urol 2011;185(5):1650-5. 121 100. Deras IL, Aubin SM, Blase A, Day JR, Koo S, Partin AW, et al. PCA3: a molecular urine assay for predicting prostate biopsy outcome. J Urol 2008;179(4):1587-92. 101. Groskopf J, Aubin SM, Deras IL, Blase A, Bodrug S, Clark C, et al. APTIMA PCA3 molecular urine test: development of a method to aid in the diagnosis of prostate cancer. Clin Chem 2006;52(6):1089-95. 102. Guazzoni G, Nava L, Lazzeri M, Scattoni V, Lughezzani G, Maccagnano C, et al. Prostate-specific antigen (PSA) isoform p2PSA significantly improves the prediction of prostate cancer at initial extended prostate biopsies in patients with total PSA between 2.0 and 10 ng/ml: results of a prospective study in a clinical setting. Eur Urol 2011;60(2):214-22. 103. Hessels D, Klein Gunnewiek JM, van Oort I, Karthaus HF, van Leenders GJ, van Balken B, et al. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol 2003;44(1):8-15; discussion 15-6. 104. Hessels D, Smit FP, Verhaegh GW, Witjes JA, Cornel EB, Schalken JA. Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer. Clin Cancer Res 2007;13(17):5103-8. 105. Laxman B, Morris DS, Yu J, Siddiqui J, Cao J, Mehra R, et al. A first-generation multiplex biomarker analysis of urine for the early detection of prostate cancer. Cancer Res 2008;68(3):645-9. 106. Laxman B, Tomlins SA, Mehra R, Morris DS, Wang L, Helgeson BE, et al. Noninvasive detection of TMPRSS2:ERG fusion transcripts in the urine of men with prostate cancer. Neoplasia 2006;8(10):885-8. 122 107. Liang Y, Ankerst DP, Ketchum NS, Ercole B, Shah G, Shaughnessy JD, Jr., et al. Prospective evaluation of operating characteristics of prostate cancer detection biomarkers. J Urol 2011;185(1):104-10. 108. Nguyen PN, Violette P, Chan S, Tanguay S, Kassouf W, Aprikian A, et al. A panel of TMPRSS2:ERG fusion transcript markers for urine-based prostate cancer detection with high specificity and sensitivity. Eur Urol 2011;59(3):40714. 109. Ouyang B, Bracken B, Burke B, Chung E, Liang J, Ho SM. A duplex quantitative polymerase chain reaction assay based on quantification of alpha-methylacylCoA racemase transcripts and prostate cancer antigen 3 in urine sediments improved diagnostic accuracy for prostate cancer. J Urol 2009;181(6):250813; discussion 13-4. 110. Shappell SB, Fulmer J, Arguello D, Wright BS, Oppenheimer JR, Putzi MJ. PCA3 urine mRNA testing for prostate carcinoma: patterns of use by community urologists and assay performance in reference laboratory setting. Urology 2009;73(2):363-8. 111. Sokoll LJ, Ellis W, Lange P, Noteboom J, Elliott DJ, Deras IL, et al. A multicenter evaluation of the PCA3 molecular urine test: pre-analytical effects, analytical performance, and diagnostic accuracy. Clin Chim Acta 2008;389(1-2):1-6. 112. Sokoll LJ, Sanda MG, Feng Z, Kagan J, Mizrahi IA, Broyles DL, et al. A prospective, multicenter, National Cancer Institute Early Detection Research Network study of [-2]proPSA: improving prostate cancer detection and correlating with cancer aggressiveness. Cancer Epidemiol Biomarkers Prev 2010;19(5):1193-200. 123 113. Stephan C, Kahrs AM, Cammann H, Lein M, Schrader M, Deger S, et al. A [2]proPSA-based artificial neural network significantly improves differentiation between prostate cancer and benign prostatic diseases. Prostate 2009;69(2):198-207. 114. Tinzl M, Marberger M, Horvath S, Chypre C. DD3PCA3 RNA analysis in urine--a new perspective for detecting prostate cancer. Eur Urol 2004;46(2):182-6; discussion 87. 115. Tomlins SA, Aubin SM, Siddiqui J, Lonigro RJ, Sefton-Miller L, Miick S, et al. Urine TMPRSS2:ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA. Sci Transl Med 2011;3(94):94ra72. 116. van Gils MP, Cornel EB, Hessels D, Peelen WP, Witjes JA, Mulders PF, et al. Molecular PCA3 diagnostics on prostatic fluid. Prostate 2007;67(8):881-7. 117. van Gils MP, Hessels D, van Hooij O, Jannink SA, Peelen WP, Hanssen SL, et al. The time-resolved fluorescence-based PCA3 test on urinary sediments after digital rectal examination; a Dutch multicenter validation of the diagnostic performance. Clin Cancer Res 2007;13(3):939-43. 118. Wang R, Chinnaiyan AM, Dunn RL, Wojno KJ, Wei JT. Rational approach to implementation of prostate cancer antigen 3 into clinical care. Cancer 2009;115(17):3879-86. 119. Dorr VJ, Williamson SK, Stephens RL. An evaluation of prostate-specific antigen as a screening test for prostate cancer. Archives of internal medicine 1993;153(22):2529-37. 120. Delongchamps NB, Singh A, Haas GP. The role of prevalence in the diagnosis of prostate cancer. Cancer Control 2006;13(3):158-68. 124 121. Ferlay J, Shin H-R, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. International journal of cancer 2010;127(12):2893-917. 122. Klotz L, Zhang L, Lam A, Nam R, Mamedov A, Loblaw A. Clinical results of longterm follow-up of a large, active surveillance cohort with localized prostate cancer. J Clin Oncol 2010;28(1):126-31. 123. 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. Evidence Synthesis No. 90. AHRQ Publication No. 1205160-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; October 2011: http://www.uspreventiveservicestaskforce.org/prostatecancerscreening/prcasc res.pdf. Accessed: 23 June 2014 124. Duffy SW, Tabar L, Olsen AH, Vitak B, Allgood PC, Chen THH, et al. Absolute numbers of lives saved and overdiagnosis in breast cancer screening, from a randomized trial and from the Breast Screening Programme in England. Journal of medical screening 2010;17(1):25-30. 125. Hoffman RM. Screening for prostate cancer: http://www.uptodate.com/contents/screening-for-prostatecancer?source=search_result&search=psa&selectedTitle=2~91. Accessed: 23 June 2014 126. Thompson IM, Ankerst DP, Chi C, Goodman PJ, Tangen CM, Lucia MS, et al. Assessing prostate cancer risk: results from the Prostate Cancer Prevention Trial. J Natl Cancer Inst 2006;98(8):529-34. 125 127. Catalona WJ, Hudson MA, Scardino PT, Richie JP, Ahmann FR, Flanigan RC, et al. Selection of optimal prostate specific antigen cutoffs for early detection of prostate cancer: receiver operating characteristic curves. J Urol 1994;152(6 Pt 1):2037-42. 128. Skates SJ, Menon U, MacDonald N, Rosenthal AN, Oram DH, Knapp RC, et al. Calculation of the risk of ovarian cancer from serial CA-125 values for preclinical detection in postmenopausal women. J Clin Oncol 2003;21(10 Suppl):206s-10s. 129. Sanda MG, Dunn RL, Michalski J, Sandler HM, Northouse L, Hembroff L, et al. Quality of life and satisfaction with outcome among prostate-cancer survivors. The New England journal of medicine 2008;358(12):1250-61. 130. Sciarra A, Barentsz J, Bjartell A, Eastham J, Hricak H, Panebianco V, et al. Advances in magnetic resonance imaging: how they are changing the management of prostate cancer. European urology 2011;59(6):962-77. 131. Thompson IM, Ankerst DP. Editorial for special issue on epidemiology and prevention of prostate cancer. World J Urol 2012;30(2):129-30. 132. Thompson IM, Ankerst DP. The benefits of risk assessment tools for prostate cancer. Eur Urol 2012;61(4):662-3. 133. Multigner L, Ndong JR, Giusti A, Romana M, Delacroix-Maillard H, Cordier S, et al. Chlordecone exposure and risk of prostate cancer. J Clin Oncol 2010;28(21):3457-62. 134. van der Kwast TH, Lopes C, Santonja C, Pihl CG, Neetens I, Martikainen P, et al. Guidelines for processing and reporting of prostatic needle biopsies. J Clin Pathol 2003;56(5):336-40. 126 135. Schroder FH, Zappa M. Prostate-specific antigen testing in Tyrol, Austria: prostate cancer mortality reduction was supported by an update with mortality data up to 2008. Int J Public Health 2012;57(1):45-7. 136. Salami LR. Prostate cancer screening: critical review of articles published in the NEJM in March 2009 [Dépistage du cancer de la prostate : analyse critique des articles publiés dans le New England Journal of Medicine en mars 2009]: http://www.has-sante.fr/portail/upload/docs/application/pdf/201006/depistage_du_cancer_de_la_prostate__analyse_des_nouvelles_donnees_rapport.pdf. Accessed: 22 June 2014 137. Becker C, Piironen T, Pettersson K, Bjork T, Wojno KJ, Oesterling JE, et al. Discrimination of men with prostate cancer from those with benign disease by measurements of human glandular kallikrein 2 (HK2) in serum. J Urol 2000;163(1):311-6. 138. Becker C, Piironen T, Pettersson K, Hugosson J, Lilja H. Clinical value of human glandular kallikrein 2 and free and total prostate-specific antigen in serum from a population of men with prostate-specific antigen levels 3.0 ng/mL or greater. Urology 2000;55(5):694-9. 139. Steuber T, Vickers A, Haese A, Kattan MW, Eastham JA, Scardino PT, et al. Free PSA isoforms and intact and cleaved forms of urokinase plasminogen activator receptor in serum improve selection of patients for prostate cancer biopsy. Int J Cancer 2007;120(7):1499-504. 140. Lilja H, Ulmert D, Bjork T, Becker C, Serio AM, Nilsson JA, et al. Long-term prediction of prostate cancer up to 25 years before diagnosis of prostate cancer using prostate kallikreins measured at age 44 to 50 years. J Clin Oncol 2007;25(4):431-6. 127 141. Vickers AJ, Ulmert D, Serio AM, Bjork T, Scardino PT, Eastham JA, et al. The predictive value of prostate cancer biomarkers depends on age and time to diagnosis: towards a biologically-based screening strategy. Int J Cancer 2007;121(10):2212-7. 142. Bangma CH, Wildhagen MF, Yurdakul G, Schroder FH, Blijenberg BG. The value of (-7, -5)pro-prostate-specific antigen and human kallikrein-2 as serum markers for grading prostate cancer. BJU Int 2004;93(6):720-4. 143. Vickers AJ, Cronin AM, Aus G, Pihl CG, Becker C, Pettersson K, et al. A panel of kallikrein markers can reduce unnecessary biopsy for prostate cancer: data from the European Randomized Study of Prostate Cancer Screening in Goteborg, Sweden. BMC Med 2008;6:19. 144. Benchikh A, Savage C, Cronin A, Salama G, Villers A, Lilja H, et al. A panel of kallikrein markers can predict outcome of prostate biopsy following clinical work-up: an independent validation study from the European Randomized Study of Prostate Cancer screening, France. BMC Cancer 2010;10:635. 145. Vickers A, Cronin A, Roobol M, Savage C, Peltola M, Pettersson K, et al. Reducing unnecessary biopsy during prostate cancer screening using a fourkallikrein panel: an independent replication. J Clin Oncol 2010;28(15):2493-8. 146. Vickers AJ, Cronin AM, Roobol MJ, Savage CJ, Peltola M, Pettersson K, et al. A four-kallikrein panel predicts prostate cancer in men with recent screening: data from the European Randomized Study of Screening for Prostate Cancer, Rotterdam. Clin Cancer Res 2010;16(12):3232-9. 147. Vickers AJ, Gupta A, Savage CJ, Pettersson K, Dahlin A, Bjartell A, et al. A panel of kallikrein marker predicts prostate cancer in a large, population-based 128 cohort followed for 15 years without screening. Cancer Epidemiol Biomarkers Prev 2011;20(2):255-61. 129 130