Skip to main content
The World Journal of Men's Health logoLink to The World Journal of Men's Health
. 2024 Apr 11;43(1):8–27. doi: 10.5534/wjmh.230386

Liquid Biomarkers in Prostate Cancer Diagnosis: Current Status and Emerging Prospects

Yutong Liu 1, Koji Hatano 1,, Norio Nonomura 1
PMCID: PMC11704174  PMID: 38772530

Abstract

Prostate cancer (PCa) is a major health concern that necessitates appropriate diagnostic approaches for timely intervention. This review critically evaluates the role of liquid biopsy techniques, focusing on blood- and urine-based biomarkers, in overcoming the limitations of conventional diagnostic methods. The 4Kscore test and Prostate Health Index have demonstrated efficacy in distinguishing PCa from benign conditions. Urinary biomarker tests such as PCa antigen 3, MyProstateScore, SelectMDx, and ExoDx Prostate IntelliScore test have revolutionized risk stratification and minimized unnecessary biopsies. Emerging biomarkers, including non-coding RNAs, circulating tumor DNA, and prostate-specific antigen (PSA) glycosylation, offer valuable insights into PCa biology, enabling personalized treatment strategies. Advancements in non-invasive liquid biomarkers for PCa diagnosis may facilitate the stratification of patients and avoid unnecessary biopsies, particularly when PSA is in the gray area of 4 to 10 ng/mL.

Keywords: Biomarkers, Diagnosis, Liquid biopsy, Prostate-specific antigen, Prostatic neoplasms

INTRODUCTION

Prostate cancer (PCa) remains a substantial global health challenge and is the second most prevalent non-cutaneous malignancy among men in the United States. PCa is the second leading cause of cancer-related deaths in men, with approximately 268,490 new cases reported annually, leading to 34,500 fatalities [1]. The accurate diagnosis and classification of PCa are imperative for treatment planning to optimize outcomes and survival rates in patients. Owing to the favorable survival rates following appropriate treatment of PCa, accurate diagnosis and staging have gained considerable attention [2].

Prostate-specific antigen (PSA), a protein produced by the prostate gland, is commonly detected in the blood. The effectiveness of PSA as a tumor marker for PCa has been substantiated by extensive evidence [3]. Notably, a large-scale randomized controlled trial conducted by the European Randomized Study of Screening for Prostate Cancer (ERSPC) revealed that mortality due to PCa was reduced by 25% in men who underwent at least one PSA screening [4]. Based on the recommendations against routine screening by the U. S. Preventive Services Task Force, PCa incidence increased by 3% annually from 2014 to 2019 after two decades of decline in the United States [5], highlighting the nuanced impact of screening guidelines on disease outcomes. However, PSA screening has inherent limitations in terms of sensitivity and specificity. Furthermore, PSA is not tumor-specific; elevated PSA levels are observed in various prostate conditions, including benign prostatic hyperplasia and infections [6]. These constraints often warrant unnecessary biopsies and contribute to the diagnosis of clinically insignificant cancers [7]. Understanding these limitations is crucial for a comprehensive evaluation of the benefits and risks of PSA screening for PCa diagnosis.

Transrectal ultrasound (TRUS)-guided needle biopsy, the gold standard for diagnosing PCa, is associated with risks such as pain, bleeding, and sepsis. Indications for biopsy included elevated serum PSA levels, abnormal findings on digital rectal examination (DRE), positive family history, and clinical symptoms [8]. Advancements in medical imaging have contributed to the diagnosis and treatment of PCa. Multiparametric magnetic resonance imaging (mpMRI) has improved the diagnostic accuracy for cancer detection [9,10]. Coupled with targeted biopsy techniques guided by mpMRI enabling precise and less invasive biopsies focused on suspicious areas [11]. Although early studies suggested that mpMRI with targeted biopsy and TRUS-guided full biopsies demonstrate similar diagnostic efficiency, current practice often combines mpMRI with both targeted and systematic biopsies [12]. Despite the diagnostic potential of mpMRI, owing to the possibility of a missed diagnosis for clinically significant cancers [13], mpMRI is not considered an alternative to biopsy [14].

To address the limitations of conventional diagnostic approaches, there is a growing consensus on non-invasive liquid biopsies to complement existing diagnostic approaches to stratify patients and avoid unnecessary biopsies [15]. The U.S. Food and Drug Administration (FDA) or Clinical Laboratory Improvement Amendments have recognized tests such as 4Kscore test, Prostate Health Index (PHI), prostate-specific antigen 3 (PCA3), MyProstateScore (MPS), SelectMDx (MDx Health) and ExoDx Prostate IntelliScore (EPI) test for clinical application, thereby offering a promising avenue for improved PCa diagnosis and was recommended by NCCN and American Urological Association/Society of Urologic Oncology guidelines [16,17,18]. Ongoing research exploring innovative techniques, such as the detection of non-coding RNA and PSA glycosylation, holds exciting potential for more accurate and personalized PCa diagnosis and treatment strategies. This review analyzes the current status and new perspectives on liquid biomarkers for PCa diagnosis.

BLOOD DIAGNOSTIC BIOMARKERS

1. 4Kscore test

The 4Kscore test, developed by OPKO Health, is a blood test approved by the FDA in 2015 that utilizes four kallikrein proteins to assess the risk of aggressive PCa in men with elevated PSA levels. The 4Kscore test was included in the NCCN guidelines for PCa early detection, combining total PSA, free PSA, intact PSA, and human kallikrein 2 with patient age, DRE results, and previous prostate biopsy results, expanding upon PSA testing for enhanced accuracy [17]. Compared to previously reported findings, the 4Kscore test offers remarkable precision over PSA, demonstrating a higher area under the curve (AUC) [19]. This considerably influences crucial clinical decisions in an attempt to provide patient-specific PCa diagnosis and management.

In a large-scale trial with 366 men, the 4Kscore test (AUC 0.81) accurately detected aggressive PCa, outperforming a base model (AUC 0.74) across diverse populations [20]. Another study on 1,012 participants validated its diagnostic ability with an AUC of 0.82, highlighting its potential to reduce unnecessary biopsies by 30% to 58% with minimal impact on delayed diagnosis (1.3%–4.7% of Gleason ≥7 PCa cases), demonstrating exceptional accuracy in detecting aggressive PCa [21]. For patients under active surveillance, the 4Kscore test accurately predicted tumor reclassification with 89% sensitivity and 29% specificity in a study of 137 low-risk PCa patients, reducing unnecessary biopsies by 27% and enabling informed decisions regarding interventions [22]. When integrated with mpMRI, the 4Kscore test substantially reduces unnecessary biopsies [23]. This ensures that biopsies are performed only when essential, thereby minimizing patient discomfort and healthcare costs.

In a comprehensive analysis of 20 studies, the 4Kscore test consistently demonstrated exceptional predictive accuracy for PCa across diverse clinical scenarios (Table 1) [19,21,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. Comparative analyses with existing models, such as the Rotterdam Prostate Cancer Risk Calculator (RPCRC), revealed that 4Kscore and RPCRC had similar high AUCs (0.88 vs. 0.87; p=0.41) [42]. Notably, the 4Kscore test discriminates between racial and ethnic groups. Darst et al [24] reported that the 4Kscore test has the highest discriminative ability in native Hawaii, followed by Latinos, Japanese, Whites, and African Americans. An AUC of 0.777 (95% confidence interval [CI]: 0.747–0.807) was observed for all ethnic groups in PCa with Gleason score ≥8 and 0.782 (95% CI: 0.756–0.808) for aggressive PCa, emphasizing its universal clinical utility. The AUC for overall PCa was the lowest for African Americans (0.686, 95% CI: 0.637–0.735) and highest for native Hawaiians (0.875, 95% CI: 0.804–0.942).

Table 1. Studies on diagnostic performance of the 4Kscore test.

Author Year Number of patients Endpoint Outcome
Vickers et al [25] 2010 2,914 Predictive accuracy for PCa AUC: 0.764 (95% CI: 0.739–0.788)
Gupta et al [26] 2010 925 Predictive accuracy for PCa in men with previous negative biopsy AUC: 0.68 (95% CI: 0.62–0.74) in any PCa; 0.87 (95% CI: 0.81–0.94) in HGPC
Benchikh et al [27] 2010 262 Predictive accuracy for PCa AUC: 0.782 (95% CI: 0.719–0.845) in any PCa; 0.870 (95% CI: 0.807–0.933) in HGPC
Vickers et al [28] 2010 1,501 Predictive accuracy for PCa AUC: 0.713 (95% CI: 0.682–0.743)
Vickers et al [29] 2010 1,241 Predictive accuracy for PCa AUC: 0.678 (95% CI: 0.643–0.714) in any PCa; 0.800 (95% CI: 0.732–0.859) in HGPC
Vickers et al [30] 2011 11,063 Predictive accuracy for PCa AUC: 0.751 (95% CI: 0.726–0.777) for any PCa; 0.803 (95% CI: 0.774–0.831) for clinical stage >T2 PCa; 0.824 (95% CI: 0.785–0.858) for clinical stage >T3 or has evidence of metastasis PCa
Vedder et al [33] 2014 708 Predictive value added by 4Kscore test to the ERSPC multivariable prediction models AUC: 0.78 (95% CI: 0.69–0.85)
Nordström et al [32] 2015 531 Predictive accuracy for PCa AUC: 0.718 (95% CI: 66.8–76.7)
Parekh et al [21] 2015 1,012 Predictive accuracy for Gleason ≥7 AUC: 0.821 (95% CI: 0.790–0.852)
Bryant et al [19] 2015 6,129 Predictive accuracy for PCa AUC: 0.719 (95% CI: 0.704–0.734) for any-grade PCa; 0.820 (95% CI: 0.802–0.838) for HGPC
Borque-Fernando et al [34] 2016 51 Predictive accuracy for PCa AUC: 0.794 (95% CI: 0.657–0.894)
Braun et al [36] 2016 749 Predictive accuracy for HGPC AUC: 0.683 (95% CI: 0.644–0.722) in any PCa; 0.777 (95% CI: 0.736–0.819) in HGPC
Kim et al [35] 2017 946 Predictive accuracy for HGPC AUC: 0.786 (95% CI: 0.748–0.816)
Lin et al [38] 2017 718 Predictive accuracy for HGPC AUC: 0.783 (95% CI: 0.691–0.871) in the initial surveillance biopsy; 0.754 (95% CI: 0.657–0.838) in reclassification in subsequent biopsies
Assel et al [37] 2019 1,632 Predictive accuracy for PCa AUC: 0.743 (95% CI: 0.722–0.763) for any PCa; 0.746 (95% CI: 0.717–0.774) for HGPC
Darst et al [24] 2020 2,358 Discriminative ability across racial/ethnic groups AUC: 0.777 (95% CI: 0.747–0.807) for Gleason 8+PCa; 0.782 (95% CI: 0.756–0.808) for aggressive PCa
Haese et al [39] 2020 2,330 Predictive accuracy for PCa AUC: 0.718 for biopsy GG 3+3 patients; 0.659 for biopsy GG 3+4 patients
Wysock et al [40] 2020 128 Ability to detect csPCa AUC: 0.830 (95% CI: 0.710–0.949)
Fredsøe et al [41] 2022 234 Predictive accuracy for PCa AUC: 0.763 (95% CI: 0.696–0.829)

AUC: area under the curve, CI: confidence interval, csPCa: clinically significant prostate cancer, ERSPC: European Randomized Study of Screening for Prostate Cancer, HGPC: high-grade prostate cancer, PCa: prostate cancer.

A systematic review comparing the 4Kscore test with other diagnostic methods demonstrated the superiority of the 4Kscore test in predicting biopsy histopathology and metastatic/aggressive disease, reducing unnecessary biopsies (ranging from 25% to 82%), and avoiding overtreatment (14%) [43]. Its integration promises personalized PCa treatment, decision optimization, and improved patient outcomes.

2. Prostate Health Index

PHI was introduced by Beckman Coulter as an FDA-approved automated immunoassay for PCa detection. The PHI, calculated from total PSA, free PSA, and [-2] proPSA values, demonstrates strong diagnostic potential across diverse patient groups. PHI is endorsed in NCCN guidelines for men aged ≥50 years with PSA levels between 4 and 10 µg/L and a non-suspicious DRE, following extensive studies [17,44]. PHI outperforms traditional markers, particularly in the PSA range of 2 to 10 µg/L, and efficiently distinguishes PCa from benign conditions thereby reducing unnecessary biopsies [45].

The diagnostic accuracy of PHI has been assessed across diverse populations (Table 2) [32,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78]. A study by Park et al (2018) [46] involving 246 Korean men demonstrated an AUC of 0.76 (95% CI: 0.69–0.84) in patients with total PSA 3.5 to 10 ng/mL and an impressive AUC of 0.97 (95% CI: 0.94–0.99) in Gleason score ≥7 PCa. A study by Ito et al [78] involving 363 men with PSA levels below 10 ng/mL demonstrated that PHI was superior to PSA for detecting clinically significant PCa, with an AUC of 0.789 (95% CI: 0.737–0.835) and 0.577 (95% CI: 0.518–0.637), respectively. Chiu et al (2019) [47], in their study on 2,488 patients that compared Asian and European populations, demonstrated an AUC of 0.71 (95% CI: 0.66–0.76) and 0.74 (95% CI: 0.70–0.79) for European and Asian patients, respectively.

Table 2. Studies on diagnostic performance of PHI.

Author Year Number of patients Endpoint Outcome
Jansen et al [51] 2010 756 Diagnostic value of PCa AUC: 0.750 (95% CI: 0.704–0.791)
Ferro et al [52] 2012 251 Diagnostic value of PCa AUC: 0.77; sensitivity: 0.85 (95% CI: 0.74–0.94); specificity: 0.61 (95% CI: 0.49–0.72)
Guazzoni et al [54] 2012 350 Diagnostic performance of predicting pathologic outcomes of RP AUC: 0.740 in Gleason sum ≥7; AUC: 0.686 in Gleason sum upgrading from 6 to 7; 0.799 of tumor volume <5 mL
Lazzeri et al [55] 2012 222 Diagnostic value of PCa in men who accepted repeat biopsy AUC: 0.67 (95% CI: 0.61–0.73)
Loeb et al [53] 2013 892 Diagnostic value of PCa AUC: 0.704
Scattoni et al [56] 2013 211 Diagnostic value of PCa AUC: 0.57 (95% CI: 0.47–0.68) in initial biopsy; 0.63 (95% CI: 0.50–0.75) in repeat biopsy
Ferro et al [57] 2013 300 Diagnostic value of PCa AUC: 0.77 (95% CI: 0.72–0.83)
Lazzeri et al [58] 2013 1,026 Diagnostic value of PCa AUC: 0.73 (95% CI: 0.66–0.80)
Stephan et al [59] 2013 1,362 Diagnostic value of PCa AUC: 0.74 (95% CI: 0.68–0.80)
Filella et al [60] 2014 354 Predictive performance of aggressiveness of PCa AUC: 0.833 (95% CI: 0.775–0.892) in Gleason score ≥7 PCa; 0.866 (95% CI: 0.787–0.942) in PCa T2 or T3
Nordström et al [32] 2015 531 Diagnostic value of PCa AUC: 0.711 (95% CI: 0.66–0.762)
Loeb et al [61] 2015 658 Diagnostic value of PCa AUC: 0.708 (95% CI: 0.668–0.747) for any PCa; 0.698 (95% CI: 0.651–0.745) for HGPC
Abrate et al [62] 2015 965 Diagnostic value of PCa AUC: 0.687 (95% CI: 0.638–0.733) in patients BMI <25 kg/m2; 0.671 (95% CI: 0.625–0.715) in patients BMI 25–29.9 kg/m2; 0.839 (95% CI: 0.768–0.895) in patients BMI ≥30 kg/m2
de la Calle et al [63] 2015 956 Diagnostic value of PCa AUC: 0.815 (95% CI: 0.771–0.858) with aggressive PCa; 0.783 (95% CI: 0.734–0.832) with Gleason score ≥7 Pca
Cantiello et al [48] 2016 188 Diagnostic value added to the Epstein criteria (Model 1: PSA density, number of positive cores and % of core involvement) AUC: 0.92 (95% CI: 0.87–0.95) for PHI add to the Epstein criteria
Foley et al [65] 2016 250 Diagnostic value of HGPC AUC: 0.712 (95% CI: 0.646–0.777) for any PCa; 0.776 (95% CI: 0.710–0.841) for HGPC
Chiu et al [66] 2016 135 Diagnostic value of PCa in Chinese men AUC: 0.800 (95% CI: 0.725–0.875) in HGPC; 0.818 (95% CI: 0.746–0.890) for tumor volume >0.5 mL
Tosoian et al [64] 2017 345 Diagnostic value of PCa AUC: 0.722 (95% CI: 0.636–0.807) for any PCa; 0.767 (95% CI: 0.681–0.852) for GG2 (GS 3+4=7) PCa
Tosoian et al [79] 2017 118 Diagnostic value of csPCa AUC: 0.76 (95% CI: 0.68–0.85)
Sriplakich et al [67] 2018 101 Diagnostic value of PCa in men with a total PSA of 4–10 ng/mL and a negative DRE AUC: 0.758 (95% CI: 0.6–0.9)
Park et al [46] 2018 246 Diagnostic value of PCa in Korean men AUC: 0.76 (95% CI: 0.69–0.84) in patients with total PSA 3.5–10 ng/mL; 0.97 (95% CI: 0.94–0.99) in Gleason score ≥7 PCa
Hsieh et al [68] 2018 154 Diagnostic value of PCa AUC: 0.77 (95% CI: 0.68–0.87)
Tang et al [69] 2018 140 Predictive performance of predicting pathologic outcomes of RP AUC: 0.767 (95% CI: 0.685–0.849) with Gleason >6 PCa
Chiu et al [47] 2019 2,488 Diagnostic value of PCa between Asian and European AUC: 0.71 (95% CI: 0.66–0.76) for European; 0.74 (95% CI: 0.70–0.79) for Asian
Cheng et al [70] 2019 213 Diagnostic value of PCa 0.772 (95% CI: 0.678–0.867)
Ito et al [78] 2020 363 Diagnostic value of PCa AUC: 0.767 (95% CI: 0.717–0.813)
Nassir et al [50] 2020 194 Diagnostic value of PCa AUC: 0.87 (95% CI: 0.825–0.948) for discrimination between Pca and normal men; 0.733 (95% CI: 0.644–0.822) for discrimination between PCa and BPH
Fan et al [49] 2021 164 Diagnostic value of csPCa in MRI-TRUS fusion biopsy AUC: 0.792 (95% CI: 0.707–0.877) in patients with PI-RADS 4/5 lesions; AUC: 0.884 (95% CI: 0.792–0.976) in the patients with PI-RADS 3 lesions
Stephan et al [71] 2021 1,057 Diagnostic value of PCa AUC: 0.789 (95% CI: 0.76–0.82) in men with PSA values 1–8 ng/mL
Chiu et al [72] 2021 412 Predictive value csPCa AUC: 0.72 for PCa; 0.77 for csPCa
Babajide et al [73] 2021 293 Diagnostic value of PCa AUC: 0.63 (95% CI: 0.54–0.73)
Garrido et al [74] 2021 237 Diagnostic value of PCa from non-Beckman Coulter manufacturers (Roche and Abbott) AUC: 0.776 (95% CI: 0.716–0.836) with Beckman Coulter Access®; 0.785 (95% CI: 0.726–0.844) with Roche cobas®; 0.778 (95% CI: 0.718–0.838) with Abbott Architect®
Filella et al [75] 2022 455 Diagnostic value of PCa AUC: 0.766 (95% CI: 0.725–0.804)
Yáñez-Castillo et al [76] 2023 140 Diagnostic value of PCa AUC: 0.77 (95% CI: 0.73–0.81)
Rius Bilbao et al [77] 2023 559 Diagnostic value of PCa AUC: 0.78 (95% CI: 0.73–0.83) in overall PCa; 0.79 (95% CI: 0.7–0.87) in patients PSA >10 ng/mL

AUC: area under the curve, CI: confidence interval, csPCa: clinically significant prostate cancer, DRE: digital rectal examination, HGPC: high-grade prostate cancer, MRI: magnetic resonance imaging, PCa: prostate cancer, PHI: Prostate Health Index, PSA: prostate-specific antigen, RP: radical prostatectomy, TRUS: transrectal ultrasound, BPH: benign prostatic hyperplasia.

Moreover, integration of the PHI into multimodal approaches, such as MRI, and its combination with established clinical parameters has significantly enhanced its diagnostic utility [79,80]. Cantiello et al (2016) [48] demonstrated its ability to outperform traditional risk calculators, with an AUC of 0.92 (95% CI: 0.87–0.95) when added to the Epstein criteria, highlighting its clinical relevance. Furthermore, a study by Fan et al (2021) [49] on MRI-TRUS fusion biopsy revealed an AUC of 0.792 (95% CI: 0.707–0.877) in patients with Prostate Imaging Reporting and Data System (PIRADS) 4/5 lesions and an AUC of 0.884 (95% CI: 0.792–0.976) in those with PI-RADS 3 lesions.

Nassir et al [50] demonstrated that the PHI was more effective in discriminating between patients with PCa and healthy men (AUC 0.887, 95% CI: 0.825–0.948) than in differentiating between PCa and patients with benign prostatic hyperplasia (BPH) (AUC 0.639, 95% CI: 0.546–0.733), which is in concordance with the findings of Lazzeri et al [81]. Notably, the excellent discriminative ability of PHI between men with PCa and healthy men is beneficial for PCa diagnosis.

The PHI-based nomograms, incorporating clinical parameters, demonstrated robust predictive accuracy, outperforming traditional risk calculators [82]. Moreover, economic analyses underscore the cost-effectiveness of PHI, especially with optimized pricing [83]. The PHI has emerged as a critical diagnostic tool with high accuracy, broad applicability, and cost efficiency. Its integration into PCa management ensures the implementation of precise, efficient, and effective cost-reduction strategies.

URINE DIAGNOSTIC BIOMARKERS

1. Prostate-specific antigen 3

PCA3, a non-coding prostate-specific RNA, is notably overexpressed in PCa tissue and serves as a biomarker of PCa; the PCA3 test was approved by the FDA in 2012 [84,85]. This molecular test is groundbreaking as it helps determine the necessity for repeat prostate biopsies in individuals with previous negative biopsy results, and its inclusion in the NCCN guidelines further underscores its clinical importance. PCA3 offers a precise diagnostic target, with over 95% of primary prostate tumors exhibiting substantial overexpression that significantly outperforms PSA in terms of predictive value and specificity [86]. Studies elucidating the diagnostic accuracy and clinical utility of PCA3 is summarized in Table 3 [33,48,52,56,57,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110]. A prospective study by Tinzl et al [87] demonstrated excellent clinical performance with an 82% sensitivity, 76% specificity, and an AUC of 0.87 (95% CI: 0.81–0.92), indicating its high predictive value. Chevli et al [99] analyzed 3,073 men and showed that PCA3 is a better predictor of PCa than PSA, with an AUC of 0.697 vs. 0.599 (p<0.01).

Table 3. Studies on diagnostic performance of PCA3.

Author Year Number of patients Endpoint Outcome
Hessels et al [88] 2003 108 Ability to identify PCa AUC: 0.72 (95% CI: 0.58, 0.85)
Tinzl et al [87] 2004 201 Ability to identify PCa AUC: 0.87 (95% CI: 0.81, 0.92)
Groskopf et al [91] 2006 143 Ability to identify PCa AUC: 0.746 (95% CI, 0.574, 0.918)
Chun et al [92] 2009 1,206 Predictive value of HGPC AUC: 0.633 for PCA3 assay score cut-off threshold of 17; 0.624 for PCA3 cut-off of 24; 0.620 for PCA3 cut-off of 35
Roobol et al [90] 2010 721 Diagnostic value of PCa AUC: 0.635 (95% CI: 0.582, 0.689)
Pepe et al [93] 2012 118 Diagnosis ability in PCa with cut-off of 35 vs. 20 AUC: 0.678 for PCA3 ≥20; 0.634 for PCA3 ≥35
Klatte et al [94] 2012 205 Diagnostic value of PCa and impact of age AUC: 0.754 (95% CI: 0.684, 0.824) for PCA3 alone; 0.771 (95% CI: 0.702, 0.840) with age-specific PCA3 score
Wu et al [95] 2012 103 Diagnostic value of PCa AUC: 0.64 (95% CI: 0.53, 0.75)
Ferro et al [52] 2012 251 Diagnostic value of PCa AUC: 0.71; sensitivity: 0.81 (95% CI: 0.68, 0.91); specificity: 0.57 (95% CI: 0.45, 0.69)
Crawford et al [96] 2012 1,962 Diagnostic value of PCa AUC: 0.706 (95% CI: 0.673, 0.739)
Cornu et al [97] 2013 291 Diagnostic value of PCa AUC: 0.66 (95% CI: 0.60, 0.72)
Scattoni et al [56] 2013 211 Diagnostic value of PCa AUC: 0.69 (95% CI: 0.59, 0.79) in initial biopsy; 0.72 (95% CI: 0.59, 0.84) in repeat biopsy
Ruffion et al [98] 2013 601 Diagnostic value of PCa AUC: 0.780 (95% CI: 0.743, 0.818)
Ferro et al [57] 2013 300 Diagnostic value of PCa AUC: 0.73 (95% CI: 0.68, 0.79)
Ochiai et al [100] 2013 647 Diagnostic value of PCa in Asian men AUC: 0.742; sensitivity 66.5%; specificity 71.6%
Chevli et al [99] 2014 3,073 Diagnostic value of PCa AUC: 0.697 for all PCa; 0.682 for HGPC
Leyten et al [89] 2014 497 Diagnostic value of PCa AUC: 0.720 (95% CI: 0.67, 0.77) in all PCa; 0.53 (95% CI: 0.44, 0.61) in patients Gleason score ≥7; 0.48 (95% CI: 0.39, 0.56) in patients clinical tumor stage T3–T4
Vedder et al [33] 2014 708 Predictive value added to the ERSPC multivariable prediction models AUC: 0.62 (95% CI: 0.52, 0.73)
Wei et al [101] 2014 859 Diagnostic value of PCa PPV: 80% (95% CI: 72%, 86%); NPV: 88% (95% CI: 81%, 93%); AUC 0.79 for initial prostate biopsy of any cancer; 0.69 for repeat prostate biopsy of any cancer; 0.78 for initial prostate biopsy of high-grade cancer; 0.79 for repeat prostate biopsy of high-grade cancer AUC: 0.775
Capoluongo et al [102] 2014 734 Evaluation diagnostic performance of PCA3 AUC: 0.775
Merola et al [103] 2015 407 Diagnostic value of PCa AUC: 0.865
Zheng et al [104] 2015 52 Diagnostic value of PCa AUC: 0.831; sensitivity: 82.4%; specificity: 77.8%
Tomlins et al [105] 2016 1,244 Diagnostic value of HGPC AUC: 0.726
Feibus et al [106] 2016 304 Diagnostic value of PCa AUC: 0.804 (95% CI: 0.752, 0.855)
Cantiello et al [48] 2016 188 Diagnostic value added to the Epstein criteria (Model 1: PSA density, number of positive cores and % of core involvement) AUC: 0.77 (95% CI: 0.70, 0.83) for PCA3 add to the Epstein criteria
Cao et al [107] 2018 271 Diagnostic value of PCa AUC: 0.729 (95% CI: 0.65, 0.78)
Ankerst et al [109] 2019 1,225 Diagnostic value of PCa AUC: 0.713; 0.764 for updated for PCPTRC with PCA3 included: 0.771 for updated PCPTRC with both PCA3 and T2:ERG included
Newcomb et al [110] 2019 782 Diagnostic value of PCa AUC: 0.611 (95% CI: 0.553, 0.668)
Gan et al [108] 2022 124 Diagnostic value of PCa AUC: 0.7829 (95% CI: 0.7021, 0.8633)

AUC: area under the curve, CI: confidence interval, ERSPC: European Randomized Study of Screening for Prostate Cancer, HGPC: high-grade prostate cancer, NPV: negative predictive value, PCa: prostate cancer, PCA3: prostate cancer antigen 3, PCPTRC: Prostate Cancer Prevention Trial Risk Calculator, PPV: positive predictive value.

Furthermore, the strategic amalgamation of PCA3 with multiple biomarkers is promising for clinical application in a new era of precision PCa detection and characterization. Leyten et al (2014) [89] demonstrated that AUC increased from 0.799 (ERSPC risk calculator), to 0.833 (ERSPC risk calculator plus PCA3), to 0.842 (ERSPC risk calculator plus PCA3 plus TMPRSS2-ERG gene fusions) to predict PCa. In addition, TMPRSS2-ERG was useful in predicting prognosis. This prospective study supports the idea that a biomarker panel combining PCA3 and TMPRSS2-ERG could lead to a significant reduction in prostate biopsies.

2. MyProstateScore

MPS, previously named as Mi-Prostate Score, measures total serum PSA and post-DRE urine expression of PCA3 and the TMPRSS2:ERG fusion gene developed by Tomlins et al [105,111]. MPS represents a pivotal advancement in PCa risk assessment, aiming to enhance diagnostic precision, and is included in the NCCN guidelines to improve PCa diagnosis. TMPRSS2:ERG fusion-positive tumors correlated strongly with higher Gleason scores [112]. Studies demonstrating the diagnostic value of MPS, as well as the combination of PCA3 and TMPRSS2:ERG fusion gene, are summarized in Table 4 [89,97,105,106,109,111,113,114].

Table 4. Studies on diagnostic performance of MPS, including the studies combining PCA3 and TMPRSS2:ERG fusion gene.

Author Year Number of patients Endpoint Outcome patients
Tomlins et al [111] 2011 1,312 Diagnostic value of PCa AUC: 0.71
Salami et al [113] 2013 45 Diagnostic value of PCa AUC: 0.77 (95% CI: 0.61, 0.90)
Cornu et al [97] 2013 291 Diagnostic value of PCa AUC: 0.67 (95% CI: 0.61, 0.73)
Leyten et al [89] 2014 497 Diagnostic value of PCa AUC: 0.59 (95% CI: 0.53, 0.64) in all PCa; 0.64 (95% CI: 0.56, 0.71) in patients Gleason score ≥7; 0.60 (95% CI: 0.52, 0.68) in patients clinical tumor stage T3–T4
Tomlins et al [105] 2016 1244 Diagnostic value of PCa AUC: 0.751 for all group of PCa; 0.772 for HGPC (CI is not mentioned in paper)
Feibus et al [106] 2016 304 Diagnostic value of PCa AUC: 0.781 (95% CI: 0.730, 0.838)
Ankerst et al [109] 2019 1,225 Diagnostic value of PCa AUC: 0.628 for MPS; 0.771 for updated PCPTRC with both PCA3 and MPS included
Cani et al [114] 2022 126 Diagnostic value of PCa AUC: 0.80 (95% CI: 0.74, 0.87) for retrained MPS; 0.72 (95% CI: 0.60, 0.84) for Hi−grade MPS

AUC: area under the curve, CI: confidence interval, PCa: prostate cancer, HGPC: high-grade prostate cancer, MPS: MyProstateScore, PCA3: prostate cancer antigen 3, PCPTRC: Prostate Cancer Prevention Trial Risk Calculator.

Tomlins et al [111] reported that MPS outperformed conventional PSA markers, with an AUCs ranging from 0.71 to 0.79 in various cohorts, indicating its diagnostic efficiency. Notably, MPS demonstrates exceptional predictive accuracy, particularly for Gleason grade ≥2 cancer [105,106]. Despite its robust performance in diagnosing PCa, especially in distinguishing aggressive variants, TMPRSS2:ERG showed limitations in predicting recurrence and mortality in men who underwent radical prostatectomy. This suggests that MPS may have a limited prognostic impact on surgically treated patients [115].

Tosoian et al [116] reported that MPS test, with cut of value >40, prevented 67% of biopsies with a negative predictive value (NPV) of 95%, minimizing unnecessary biopsies. Tosoian et al [117] also reported that, in men (n=121) with equivocal mpMRI (PI-RADS 3), the AUC for predicting Gleason grade ≥2 PCa was 0.55 for PSA, 0.62 for PSA density, and 0.73 for MPS. Economically, MPS has greater value than MRI when used as a reflex test before prostate biopsy, especially in patients with intermediate PSA levels (4–10 ng/mL) [118]. By offering both diagnostic accuracy and economic feasibility, MPS is a valuable diagnostic tool.

3. SelectMDx

Leyten et al (2015) [119] showed that HOXC6, TDRD1 and DLX1 are overexpressed in urine samples from Gleason score ≥7 PCa and can be beneficial for identifying patients with aggressive PCa, regardless of PSA values. The urine-based test after DRE measures three biomarkers: DLX1 (progression gene), HOXC6 (cell proliferation gene), KLK3 (reference gene), and clinical risk factors (age, DRE, PSA, and prostate volume, which can be calculated from the TRUS measurements substituted into the formula: height×width×length×0.523); it was introduced as SelectMDx and was included in the 2018 European Association of Urology (EAU) clinical guidelines. The ability to integrate messenger RNA (mRNA) markers and clinical risk factors offers a comprehensive understanding of PCa risk [120]. Although SelectMDx is more sensitive but less specific than mpMRI for PCa detection [121], it optimizes healthcare resources through precise patient stratification, leading to a considerable reduction in unnecessary biopsies and marking a paradigm shift in diagnosis [122]. Studies demonstrating the diagnostic accuracy of SelectMDx are listed in Table 5 [40,121,122,123,124,125,126,127,128,129,130,131]. The ability of SelectMDx to accurately identify high-grade PCa (HGPC) is of potential concern, and its sensitivity, specificity, and predictive values have proven its efficacy across diverse patient populations [132]. Furthermore, SelectMDx is cost-efficient, alleviating the burden of unnecessary medical interventions on patients in European countries [133].

Table 5. Studies on diagnostic performance of SelectMDx.

Author Year Number of patients Endpoint Outcome
Hendriks et al [123] 2017 172 Predictive accuracy of PCa AUC: 0.83 (95% CI: 0.77–0.89)
Haese et al [124] 2019 1,955 Predictive accuracy of csPCa AUC 0.85 (95% CI: 0.83–0.88)
Roumiguié et al [126] 2020 117 Predictive accuracy of PCa AUC 0.73 (95% CI: 0.64–0.83)
Wysock et al [40] 2020 128 Predictive accuracy of csPCa AUC: 0.672 (95% CI: 0.517–0.828; p=0.036)
Pepe et al [127] 2020 125 Predictive accuracy of csPCa Sensitivity: 55.6%; Specificity: 65.8%; PPV: 27.8%; NPV: 87%
Maggi et al [121] 2021 310 Predictive accuracy of PCa and csPCa Predicting PCa: AUC: 0.80 (95% CI: 0.76–0.85); sensitivity: 86.5% (95% CI: 78.5–91.9); specificity: 73.8% (95% CI: 64.7–79.3); Predicting csPCa: AUC: 0.75 (95% CI: 0.70–0.81); sensitivity: 87.1% (95% CI: 76.2–93.5); specificity: 63.7% (95% CI: 57.5–69.4)
Hendriks et al [122] 2021 599 Predictive accuracy of HGPC NPV 92% (95% CI: 0.88–0.95); PPV 44% (95% CI: 0.39–0.50); Sensitivity, 90% (95% CI: 0.85–0.94)
Busetto et al [128] 2021 52 Predictive accuracy of PCa NPV 97.0% (95% CI: 0.911–1.000); PPV 84.2% (95% CI: 0.678–1.000); Sensitivity, 94.1% (95% CI: 0.707–1.000); Specificity, 91.4% (95% CI: 0.767–0.977)
Lendínez-Cano et al [129] 2021 163 Predictive accuracy of csPCa AUC: 0.63 (95% CI: 0.56–0.71); Sensitivity: 76.9% (95% CI: 63.2–87.5); Specificity: 49.6% (95% CI: 39.9–59.2); NPV: 82.09% (95% CI: 70.8–90.4); PPV: 41.67% (95% CI: 31.7–52.2)
Fiorella et al [130] 2021 86 Predictive accuracy for predicting pathological progression in PCa active surveillance AUC: 0.714 (95% CI: 0.603–0.825)
Katzendorn et al [131] 2022 74 Accuracy for detection of PCa AUC 0.76
Margolis et al [125] 2022 1,212 Predictive accuracy of PCa AUC 0.70 (95% CI: 0.67–0.73)

AUC: area under the curve, CI: confidence interval, csPCa: clinically significant prostate cancer, HGPC: high-grade prostate cancer, NPV: negative predictive value, PCa: prostate cancer, PPV: positive predictive value.

The predictive accuracy of SelectMDx has been substantiated by extensive research. Hendriks et al [123] demonstrated an AUC of 0.83 (95% CI: 0.77–0.89) for predicting PCa by SelectMDx. Haese et al [124] achieved an AUC of 0.85 (95% CI: 0.83–0.88) for predicting clinically significant PCa with 93% sensitivity, 47% specificity, and 95% NPV. Visser et al [134] reported that of 5157 patients from 10 European countries, SelectMDx results were negative in 40.72% of cases, indicating a substantial number of patients could avoid the need for biopsy. A prospective study demonstrated that the SelectMDx test, as a risk stratification tool for biopsy-naïve men, avoided 38% of unnecessary biopsies and missed only 10% of HGPC. If the use of mpMRI is limited or expensive, a good alternative strategy is to use mpMRI only for SelectMDx test-positive patients [122].

4. ExoDx Prostate IntelliScore test

Donovan et al [135] revealed that the sum of exosomal PCA3 and ERG RNA levels deribed from non-DRE urine had predictive value for PCa diagnosis. Based on this finding, McKiernan et al. developed the EPI test, a non-invasive urine exosome RNA-based assay that assesses the expression of three genes (PCA3, ERG, and SPDEF), which has emerged as a revolutionary tool for risk assessment and decision-making [136]. The robust diagnostic accuracy of the EPI test has been demonstrated, positioning it as a valuable tool for PCa risk assessment, particularly for ruling out cases and detecting HGPC (Table 6) [125,136,137,138,139]. In a study involving 229 men with previous negative results from biopsies, the EPI test with a cut point of 15.6 avoided 26% of unnecessary biopsies and missed only 2.1% of HGPC [137]. Clinical utility studies involving 1,094 patients and 72 urologists revealed a 30% increase in HGPC detection, emphasizing the impact of EPI on patient stratification [140]. Cost-effectiveness analyses revealed that the EPI test significantly improved quality-adjusted survival while reducing the number of unnecessary biopsies [141].

Table 6. Studies on diagnostic performance of ExoDx Prostate IntelliScore (EPI).

Author Year Number of patients Endpoint Outcome
McKiernan et al [136] 2016 499 Diagnostic accuracy of PCa AUC 0.77 (95% CI: 0.71–0.83)
McKiernan et al [139] 2018 503 Diagnostic accuracy of HGPC AUC (95% CI): 0.71 (0.66–0.76); Sensitivity: 90 (84.1–94.1); Specificity: 38.6 (33.4–43.9); NPV: 89.3 (83.1–93.7); PPV: 40.1 (35.0–45.4)
McKiernan et al [137] 2020 229 Diagnostic accuracy of PCa AUC: 0.66 (95% CI: 0.55–0.78); NPV: 92% (95% CI: 0.81–0.97) 82%; sensitivity: 92% (95% CI: 0.81–0.97)
Margolis et al [125] 2022 1,212 Diagnostic accuracy of PCa AUC 0.70 (95% CI: 0.67–0.73)
Kretschmer et al [138] 2022 2,066 Diagnostic accuracy of PCa AUC 0.84 (95% CI: 0.73–0.96)

AUC: area under the curve, CI: confidence interval, HGPC: high-grade prostate cancer, PPV: positive predictive value, NPV: negative predictive value, PCa: prostate cancer.

In summary, EPI has emerged as a powerful tool for PCa risk assessment, effective in ruling out cases and detecting HGPC. The following sections delve into the historical context, diagnostic utility, key research findings, and cost considerations of EPI and conclude with its overall significance in PCa diagnostics.

5. Emerging diagnostic biomarkers in liquid biopsy

Recently, novel biomarkers that offer promising avenues for early detection, precise prognosis, and tailored treatment strategies have been identified. In addition to established markers, emerging biomarkers have attracted considerable attention among researchers and clinicians. These innovative biomarkers, including non-coding RNAs, PSA glycosylation, and DNA methylation patterns, are at the forefront of PCa diagnosis, heralding a new era in personalized medicine.

6. Non-coding RNA

More than 80% of human genome sequencing results in diverse non-coding RNA transcripts categorized into long non-coding RNAs (LncRNAs) (>200 nucleotides) and small non-coding RNAs (below this threshold) based on their size differences [142]. In recent decades, molecular biomarker studies have primarily focused on coding genes. However, extensive research has revealed that mRNAs often display less specific expression patterns across different tissues and disease stages. In contrast, non-coding RNAs exhibit high tissue and stage specificity for diseases [143]. This distinct characteristic is a key factor in the proposal of non-coding RNAs as molecular biomarkers of cancer.

7. LncRNAs

LncRNAs have been implicated in several biological and pathological processes, including chromatin reprogramming, transcriptional regulation, cell proliferation, tumorigenesis, and malignant transformation [144]. Because of their critical role in cancer, which may explain their tissue- and tumor-specific expression, lncRNAs have potential as molecular biomarkers [145,146].

The differential expression of some lncRNAs has been analyzed in patients with PCa, and their potential as biomarkers has been explored. Among these, PCA3 is considered a PCa biomarker approved by the FDA.

The importance of lncRNAs, including PCAT1, PCGEM1, SChLAP1, and PCAT6, in PCa is evident. PCAT1, a recently discovered lncRNA, is a promising biomarker. The elevated expression of PCAT1 is strongly associated with cancer infiltration depth, lymph node metastasis, distant metastasis, and TNM stage, providing a detailed description of cancer progression [147]. PCGEM1, another lncRNA, delves into the realm of genetic variation, revealing polymorphisms in its genes that influence PCa risk and opening avenues for exploring the interplay between genetics and lncRNA function [148]. SChLAP1, an overexpressed lncRNA, is a critical determinant of PCa aggressiveness, orchestrating cancer cell invasiveness and metastasis via antagonistic interactions with the SWI/SNF chromatin-modifying complex [149,150]. PCAT6, revealed through high-throughput analyses, unravels the m6A-induced PCAT6/IGF2BP2/IGF1R axis, providing insights on the intricate mechanisms underlying PCa bone metastasis and tumor growth [151].

In addition to lncRNA expression levels, cancer risk is linked to the increased prevalence of single-nucleotide polymorphisms (SNPs) within lncRNA genes in PCa. Specifically, SNPs in PCAT19, PCGEM1 and PRNCR1 have been identified as potential contributors to PCa susceptibility [148,152].

8. Small non-coding RNAs

Owing to their remarkable stability in bodily fluids, miRNAs have become one of the most extensively studied small non-coding RNAs [153,154,155]. In addition to their diagnostic significance, certain miRNAs, including miR-21, miR-221, and miR-375, are dysregulated in PCa and linked to the prognosis of patients with castration-resistant PCa [156,157]. Moreover, certain miRNAs regulate cellular sensitivity to ionization radiation, indicating that miRNAs can be therapeutic biomarkers [158].

The role of small non-coding RNAs in PCa extends beyond that of miRNAs. Deep sequencing analyses have revealed significant differential expression of small nucleolar RNAs and transfer RNAs in PCa [157]. In addition, the analysis of urinary exosomal small non-coding RNAs has paved the way for the development of advanced diagnostic tools. The miR Sentinel PCa Tests based on urinary exosomal small non-coding RNAs have demonstrated exceptional sensitivity and specificity for diagnosing and classifying PCa [159].

These advances highlight the potential of small non-coding RNAs as robust biomarkers for diagnosing PCa. The integration of these innovative tests into clinical practice could revolutionize early detection, provide patients with timely and accurate prognostic information, and ultimately enhance PCa management.

9. PSA glycosylation

Recent advancements in glycobiology have revealed that aberrant glycosylation is fundamental to tumorigenesis, making glycoproteins with tumor-specific glycans potential cancer markers [160,161]. Proteins, including PSA, undergo diverse glycosylation patterns owing to alterations in their cellular pathways. PSA glycosylation is pivotal in PCa research and specifically focuses on α2,3-sialylated PSA and α1,6-fucosylated PSA [162,163,164,165,166,167]. Yoneyama et al [168] reported that the PCa detectability of α2,3-sialylated PSA density was superior to that of total PSA or PSA density (AUC: 0.7758 vs 0.6360 and 0.7509, respectively). Fujita et al [169] reported that the AUC for the detection of Gleason grade ≥2 by α1,6-fucosylated PSA index was 0.698 (95% CI: 0.600–0.796; p=0.0004), compared to 0.554 (95% CI: 0.445–0.664; p=0.33) for PSA. By concurrently analyzing α2,3-Sia-PSA and α1,6-Fuc-PSA (SF index), Hatano et al [170] demonstrated the superior diagnostic capabilities of the SF index for Gleason grade ≥2 PCa (AUC 0.842, 95% CI: 0.782–0.903), which outperformed single PSA tests. The ability of the SF index to complement conventional PSA tests enhances accuracy and reliability, underscoring its potential to revolutionize PCa diagnosis. PSA glycosylation represents a paradigm shift, offering a targeted approach for PCa detection and marking a new era in cancer biomarkers.

10. Cell-free DNA-derived diagnosis assay

Cell-free DNA facilitates real-time genomic profiling of cancer cells without tissue biopsy. In PCa, Cell-free DNA analysis enables the detection of crucial genomic alterations, including DNA methylation pattern changes, somatic mutations, copy number variants, and structural rearrangements [171]. The comprehensive characterization of these genomic changes within cell-free DNA not only establishes it as a potent diagnostic tool but also provides invaluable insights into the underlying biology of metastatic disease. By unraveling the intricate genomic landscape, a deeper understanding of the molecular intricacies that drive PCa progression can be obtained, enabling more precise and personalized therapeutic interventions.

11. DNA methylation biomarkers

Among the diverse array of emerging diagnostic biomarkers, DNA methylation patterns have attracted considerable attention. DNA methylation, an epigenetic modification involving the addition of a methyl group to cytosine residues, plays a pivotal role in the regulation of gene expression. Differences in methylation between normal and cancerous cells have been previously described [172]. In the context of PCa, aberrant DNA methylation patterns serve as potential diagnostic biomarkers [173].

The PCa genome has been extensively studied by identifying specific regions in which DNA methylation alterations are prevalent [174]. These changes in genes associated with crucial cellular processes provide valuable insights into disease progression and aggressiveness [175]. Substantial efforts have been made to develop non-invasive diagnostic assays by deciphering these methylation signatures. Liquid biopsy coupled with methylation-specific techniques allows the detection of epigenetic alterations in circulating tumor DNA or other bodily fluids, offering a minimally invasive yet highly accurate approach to diagnosing PCa [176].

The incorporation of DNA methylation biomarkers into liquid biopsy assays holds immense promise for tailoring personalized treatment strategies for PCa. These markers not only aid in early detection but also provide valuable information regarding disease prognosis and treatment response [177,178]. DNA methylation can distinguish PCa at different Gleason stages (Gleason 6 vs. Gleason 3+4: AUC=0.63; Gleason 6 vs. Gleason 4+3 and 8-10: AUC=0.87) [179]. Additionally, the noninvasive nature of liquid biopsy procedures reduces patient discomfort and offers a convenient avenue for regular monitoring, particularly in cases requiring active surveillance [180,181]. With further advancements, the identification of specific methylation profiles associated with different PCa subtypes may facilitate targeted therapies and revolutionize PCa management.

12. Genomic profiling

Integrated genomic profiling of prostate tumors has revealed a spectrum of clinically actionable molecular alterations, including modifications within the DNA damage repair pathway and PTEN/PI3K signaling. These genomic changes, which influence cell proliferation, tumorigenesis, and cell cycle pathways, are associated with adverse clinical outcomes in PCa. Advanced genomic profiling tests hold promise for the identification of innovative diagnostic, prognostic, and therapeutic molecular biomarkers [182]. Advanced genomic tests such as Oncotype Dx Prostate, Prolaris, and Decipher offer personalized insights for refining PCa diagnosis and treatment. These tests guide active surveillance and aid clinical decision-making [183]. However, their routine application is discouraged by the limited number of prospective studies evaluating their long-term impact on patient outcomes, including quality of life, treatment necessity, and survival.

CONCLUSIONS

The landscape of PCa diagnosis is evolving, emphasizing the need for an integrated, multimodal approach. The incorporation of various biomarkers and diagnostic tools, such as blood-based markers (e.g., 4Kscore test, PHI) and urine-based markers (e.g., SelectMDx and PCA3), offers a more comprehensive assessment of the disease. Ongoing optimization and standardization of assay thresholds will further expand their impact. Exciting emerging biomarkers such as non-coding RNAs, fucosylated PSA, and methylation signatures provide new avenues to unravel PCa biology in real time through liquid biopsy analysis.

However, validation in large cohorts, demonstrating added value over current standards, and assay consistency issues remain challenging. Addressing these gaps through collaborative research will accelerate clinical integration. With insights from basic science combined with technological innovations, liquid biopsy platforms can evolve beyond diagnosis to guide screening, monitoring, prognosis, and therapy selection for improved patient outcomes.

The integration of diverse biomarkers and diagnostic tools represents a shift in the diagnosis of PCa. Although promising, evidence for combined diagnostic approaches is lacking, highlighting a key area for future research. Continued research and collaborative efforts are essential to effectively integrating these diagnostics into personalized management strategies for PCa. Overcoming the current challenges is pivotal for realizing the potential benefits of this integrated approach with the goal of revolutionizing PCa management through early detection, informed risk stratification, and personalized treatment strategies.

Acknowledgements

None.

Footnotes

Conflict of Interest: The authors have nothing to disclose.

Funding: This research was supported by the Japan Society for the Promotion of Science KAKENHI [grant number 22K09447].

Author Contribution:
  • Conceptualization: YL, KH, NN.
  • Data curation: YL, KH.
  • Formal analysis: YL.
  • Funding acquisition: KH.
  • Investigation: KH.
  • Methodology: YL, KH.
  • Project administration: YL, KH, NN.
  • Resources: KH.
  • Software: YL.
  • Supervision: KH, NN.
  • Validation: YL, KH, NN.
  • Visualization: YL.
  • Writing – original draft: YL, KH.
  • Writing – review & editing: YL, KH, NN.

References

  • 1.Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48. doi: 10.3322/caac.21763. [DOI] [PubMed] [Google Scholar]
  • 2.Narain V, Cher ML, Wood DP., Jr Prostate cancer diagnosis, staging and survival. Cancer Metastasis Rev. 2002;21:17–27. doi: 10.1023/a:1020104004588. [DOI] [PubMed] [Google Scholar]
  • 3.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
  • 4.Hugosson J, Roobol MJ, Månsson M, Tammela TLJ, Zappa M, Nelen V, et al. ERSPC Investigators. A 16-yr follow-up of the European randomized study of screening for prostate cancer. Eur Urol. 2019;76:43–51. doi: 10.1016/j.eururo.2019.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Desai MM, Cacciamani GE, Gill K, Zhang J, Liu L, Abreu A, et al. Trends in incidence of metastatic prostate cancer in the US. JAMA Netw Open. 2022;5:e222246. doi: 10.1001/jamanetworkopen.2022.2246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lilja H, Ulmert D, Vickers AJ. Prostate-specific antigen and prostate cancer: prediction, detection and monitoring. Nat Rev Cancer. 2008;8:268–278. doi: 10.1038/nrc2351. Erratum in: Nat Rev Cancer 2008;8:403. [DOI] [PubMed] [Google Scholar]
  • 7.Heijnsdijk EA, der Kinderen A, Wever EM, Draisma G, Roobol MJ, de Koning HJ. Overdetection, overtreatment and costs in prostate-specific antigen screening for prostate cancer. Br J Cancer. 2009;101:1833–1838. doi: 10.1038/sj.bjc.6605422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bruyère F, Malavaud S, Bertrand P, Decock A, Cariou G, Doublet JD, et al. Prosbiotate: a multicenter, prospective analysis of infectious complications after prostate biopsy. J Urol. 2015;193:145–150. doi: 10.1016/j.juro.2014.07.086. [DOI] [PubMed] [Google Scholar]
  • 9.Wu RC, Lebastchi AH, Hadaschik BA, Emberton M, Moore C, Laguna P, et al. Role of MRI for the detection of prostate cancer. World J Urol. 2021;39:637–649. doi: 10.1007/s00345-020-03530-3. [DOI] [PubMed] [Google Scholar]
  • 10.Eastham JA, Auffenberg GB, Barocas DA, Chou R, Crispino T, Davis JW, et al. Clinically localized prostate cancer: AUA/ASTRO guideline, part I: introduction, risk assessment, staging, and risk-based management. J Urol. 2022;208:10–18. doi: 10.1097/JU.0000000000002757. [DOI] [PubMed] [Google Scholar]
  • 11.Klotz L, Chin J, Black PC, Finelli A, Anidjar M, Bladou F, et al. Comparison of multiparametric magnetic resonance imaging-targeted biopsy with systematic transrectal ultrasonography biopsy for biopsy-naive men at risk for prostate cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2021;7:534–542. doi: 10.1001/jamaoncol.2020.7589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mottet N, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer-2020 update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2021;79:243–262. doi: 10.1016/j.eururo.2020.09.042. [DOI] [PubMed] [Google Scholar]
  • 13.Panebianco V, Barchetti G, Simone G, Del Monte M, Ciardi A, Grompone MD, et al. Negative multiparametric magnetic resonance imaging for prostate cancer: what's next? Eur Urol. 2018;74:48–54. doi: 10.1016/j.eururo.2018.03.007. [DOI] [PubMed] [Google Scholar]
  • 14.Hettiarachchi D, Geraghty R, Rice P, Sachdeva A, Nambiar A, Johnson M, et al. Can the use of serial multiparametric magnetic resonance imaging during active surveillance of prostate cancer avoid the need for prostate biopsies?-a systematic diagnostic test accuracy review. Eur Urol Oncol. 2021;4:426–436. doi: 10.1016/j.euo.2020.09.002. [DOI] [PubMed] [Google Scholar]
  • 15.Alix-Panabières C, Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov. 2021;11:858–873. doi: 10.1158/2159-8290.CD-20-1311. [DOI] [PubMed] [Google Scholar]
  • 16.Chang EK, Gadzinski AJ, Nyame YA. Blood and urine biomarkers in prostate cancer: are we ready for reflex testing in men with an elevated prostate-specific antigen? Asian J Urol. 2021;8:343–353. doi: 10.1016/j.ajur.2021.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Schaeffer EM, Srinivas S, Adra N, An Y, Barocas D, Bitting R, et al. Prostate cancer, version 4.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2023;21:1067–1096. doi: 10.6004/jnccn.2023.0050. [DOI] [PubMed] [Google Scholar]
  • 18.Wei JT, Barocas D, Carlsson S, Coakley F, Eggener S, Etzioni R, et al. Early detection of prostate cancer: AUA/SUO guideline part II: considerations for a prostate biopsy. J Urol. 2023;210:54–63. doi: 10.1097/JU.0000000000003492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bryant RJ, Sjoberg DD, Vickers AJ, Robinson MC, Kumar R, Marsden L, et al. Predicting high-grade cancer at ten-core prostate biopsy using four kallikrein markers measured in blood in the ProtecT study. J Natl Cancer Inst. 2015;107:djv095. doi: 10.1093/jnci/djv095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Punnen S, Freedland SJ, Polascik TJ, Loeb S, Risk MC, Savage S, et al. A multi-institutional prospective trial confirms noninvasive blood test maintains predictive value in African American men. J Urol. 2018;199:1459–1463. doi: 10.1016/j.juro.2017.11.113. [DOI] [PubMed] [Google Scholar]
  • 21.Parekh DJ, Punnen S, Sjoberg DD, Asroff SW, Bailen JL, Cochran JS, et al. A multi-institutional prospective trial in the USA confirms that the 4Kscore accurately identifies men with high-grade prostate cancer. Eur Urol. 2015;68:464–470. doi: 10.1016/j.eururo.2014.10.021. [DOI] [PubMed] [Google Scholar]
  • 22.Borque-Fernando Á, Rubio-Briones J, Esteban LM, Dong Y, Calatrava A, Gómez-Ferrer Á, et al. Role of the 4Kscore test as a predictor of reclassification in prostate cancer active surveillance. Prostate Cancer Prostatic Dis. 2019;22:84–90. doi: 10.1038/s41391-018-0074-5. [DOI] [PubMed] [Google Scholar]
  • 23.Punnen S, Nahar B, Soodana-Prakash N, Koru-Sengul T, Stoyanova R, Pollack A, et al. Optimizing patient's selection for prostate biopsy: a single institution experience with multi-parametric MRI and the 4Kscore test for the detection of aggressive prostate cancer. PLoS One. 2018;13:e0201384. doi: 10.1371/journal.pone.0201384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Darst BF, Chou A, Wan P, Pooler L, Sheng X, Vertosick EA, et al. The four-kallikrein panel is effective in identifying aggressive prostate cancer in a multiethnic population. Cancer Epidemiol Biomarkers Prev. 2020;29:1381–1388. doi: 10.1158/1055-9965.EPI-19-1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Vickers A, Cronin A, Roobol M, Savage C, Peltola M, Pettersson K, et al. Reducing unnecessary biopsy during prostate cancer screening using a four-kallikrein panel: an independent replication. J Clin Oncol. 2010;28:2493–2498. doi: 10.1200/JCO.2009.24.1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gupta A, Roobol MJ, Savage CJ, Peltola M, Pettersson K, Scardino PT, et al. A four-kallikrein panel for the prediction of repeat prostate biopsy: data from the European randomized study of prostate cancer screening in Rotterdam, Netherlands. Br J Cancer. 2010;103:708–714. doi: 10.1038/sj.bjc.6605815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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. doi: 10.1186/1471-2407-10-635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.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;16:3232–3239. doi: 10.1158/1078-0432.CCR-10-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Vickers AJ, Cronin AM, Aus G, Pihl CG, Becker C, Pettersson K, et al. Impact of recent screening on predicting the outcome of prostate cancer biopsy in men with elevated prostate-specific antigen: data from the European randomized study of prostate cancer screening in Gothenburg, Sweden. Cancer. 2010;116:2612–2620. doi: 10.1002/cncr.25010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.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 cohort followed for 15 years without screening. Cancer Epidemiol Biomarkers Prev. 2011;20:255–261. doi: 10.1158/1055-9965.EPI-10-1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Carlsson S, Maschino A, Schröder F, Bangma C, Steyerberg EW, van der Kwast T, et al. Predictive value of four kallikrein markers for pathologically insignificant compared with aggressive prostate cancer in radical prostatectomy specimens: results from the European randomized study of screening for prostate cancer section Rotterdam. Eur Urol. 2013;64:693–699. doi: 10.1016/j.eururo.2013.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Nordström T, Vickers A, Assel M, Lilja H, Grönberg H, Eklund M. Comparison between the four-kallikrein panel and prostate health index for predicting prostate cancer. Eur Urol. 2015;68:139–146. doi: 10.1016/j.eururo.2014.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vedder MM, de Bekker-Grob EW, Lilja HG, Vickers AJ, van Leenders GJ, Steyerberg EW, et al. The added value of percentage of free to total prostate-specific antigen, PCA3, and a kallikrein panel to the ERSPC risk calculator for prostate cancer in prescreened men. Eur Urol. 2014;66:1109–1115. doi: 10.1016/j.eururo.2014.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Borque-Fernando Á, Esteban-Escaño LM, Rubio-Briones J, Lou-Mercadé AC, García-Ruiz R, Tejero-Sánchez A, et al. A preliminary study of the ability of the 4Kscore test, the prostate cancer prevention trial-risk calculator and the European research screening prostate-risk calculator for predicting high-grade prostate cancer. Actas Urol Esp. 2016;40:155–163. doi: 10.1016/j.acuro.2015.09.006. [DOI] [PubMed] [Google Scholar]
  • 35.Kim EH, Andriole GL, Crawford ED, Sjoberg DD, Assel M, Vickers AJ, et al. Detection of high grade prostate cancer among PLCO participants using a prespecified 4-kallikrein marker panel. J Urol. 2017;197:1041–1047. doi: 10.1016/j.juro.2016.10.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Braun K, Sjoberg DD, Vickers AJ, Lilja H, Bjartell AS. A four-kallikrein panel predicts high-grade cancer on biopsy: independent validation in a community cohort. Eur Urol. 2016;69:505–511. doi: 10.1016/j.eururo.2015.04.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Assel M, Sjöblom L, Murtola TJ, Talala K, Kujala P, Stenman UH, et al. A four-kallikrein panel and β-microseminoprotein in predicting high-grade prostate cancer on biopsy: an independent replication from the Finnish section of the European randomized study of screening for prostate cancer. Eur Urol Focus. 2019;5:561–567. doi: 10.1016/j.euf.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lin DW, Newcomb LF, Brown MD, Sjoberg DD, Dong Y, Brooks JD, et al. Canary Prostate Active Surveillance Study Investigators. Evaluating the four kallikrein panel of the 4Kscore for prediction of high-grade prostate cancer in men in the canary prostate active surveillance study. Eur Urol. 2017;72:448–454. doi: 10.1016/j.eururo.2016.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Haese A, Tin AL, Carlsson SV, Sjoberg DD, Pehrke D, Steuber T, et al. A pre-specified model based on four kallikrein markers in blood improves predictions of adverse pathology and biochemical recurrence after radical prostatectomy. Br J Cancer. 2020;123:604–609. doi: 10.1038/s41416-020-0914-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wysock JS, Becher E, Persily J, Loeb S, Lepor H. Concordance and performance of 4Kscore and SelectMDx for informing decision to perform prostate biopsy and detection of prostate cancer. Urology. 2020;141:119–124. doi: 10.1016/j.urology.2020.02.032. [DOI] [PubMed] [Google Scholar]
  • 41.Fredsøe J, Rasmussen M, Tin AL, Vickers AJ, Borre M, Sørensen KD, et al. Predicting Grade group 2 or higher cancer at prostate biopsy by 4Kscore in blood and uCaP microRNA model in urine. Sci Rep. 2022;12:15193. doi: 10.1038/s41598-022-19460-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Verbeek JFM, Bangma CH, Kweldam CF, van der Kwast TH, Kümmerlin IP, van Leenders GJLH, et al. Reducing unnecessary biopsies while detecting clinically significant prostate cancer including cribriform growth with the ERSPC Rotterdam risk calculator and 4Kscore. Urol Oncol. 2019;37:138–144. doi: 10.1016/j.urolonc.2018.11.021. [DOI] [PubMed] [Google Scholar]
  • 43.Olleik G, Kassouf W, Aprikian A, Hu J, Vanhuyse M, Cury F, et al. Evaluation of new tests and interventions for prostate cancer management: a systematic review. J Natl Compr Canc Netw. 2018;16:1340–1351. doi: 10.6004/jnccn.2018.7055. [DOI] [PubMed] [Google Scholar]
  • 44.Ferro M, De Cobelli O, Lucarelli G, Porreca A, Busetto GM, Cantiello F, et al. Beyond PSA: the role of prostate health index (PHI) Int J Mol Sci. 2020;21:1184. doi: 10.3390/ijms21041184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Filella X, Giménez N. Evaluation of [-2] proPSA and prostate health index (PHI) for the detection of prostate cancer: a systematic review and meta-analysis. Clin Chem Lab Med. 2013;51:729–739. doi: 10.1515/cclm-2012-0410. [DOI] [PubMed] [Google Scholar]
  • 46.Park H, Lee SW, Song G, Kang TW, Jung JH, Chung HC, et al. Diagnostic performance of %[-2]proPSA and prostate health index for prostate cancer: prospective, multi-institutional study. J Korean Med Sci. 2018;33:e94. doi: 10.3346/jkms.2018.33.e94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chiu PK, Ng CF, Semjonow A, Zhu Y, Vincendeau S, Houlgatte A, et al. A multicentre evaluation of the role of the prostate health index (PHI) in regions with differing prevalence of prostate cancer: adjustment of PHI reference ranges is needed for European and Asian settings. Eur Urol. 2019;75:558–561. doi: 10.1016/j.eururo.2018.10.047. [DOI] [PubMed] [Google Scholar]
  • 48.Cantiello F, Russo GI, Cicione A, Ferro M, Cimino S, Favilla V, et al. PHI and PCA3 improve the prognostic performance of PRIAS and Epstein criteria in predicting insignificant prostate cancer in men eligible for active surveillance. World J Urol. 2016;34:485–493. doi: 10.1007/s00345-015-1643-z. [DOI] [PubMed] [Google Scholar]
  • 49.Fan YH, Pan PH, Cheng WM, Wang HK, Shen SH, Liu HT, et al. The prostate health index aids multi-parametric MRI in diagnosing significant prostate cancer. Sci Rep. 2021;11:1286. doi: 10.1038/s41598-020-78428-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Nassir AM, Kamel HFM. Explication of the roles of prostate health index (PHI) and urokinase plasminogen activator (uPA) as diagnostic and predictor tools for prostate cancer in equivocal PSA range of 4-10 ng/mL. Saudi J Biol Sci. 2020;27:1975–1984. doi: 10.1016/j.sjbs.2020.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jansen FH, van Schaik RH, Kurstjens J, Horninger W, Klocker H, Bektic J, et al. Prostate-specific antigen (PSA) isoform p2PSA in combination with total PSA and free PSA improves diagnostic accuracy in prostate cancer detection. Eur Urol. 2010;57:921–927. doi: 10.1016/j.eururo.2010.02.003. [DOI] [PubMed] [Google Scholar]
  • 52.Ferro M, Bruzzese D, Perdonà 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;413:1274–1278. doi: 10.1016/j.cca.2012.04.017. [DOI] [PubMed] [Google Scholar]
  • 53.Loeb S, Sokoll LJ, Broyles DL, Bangma CH, van Schaik RH, Klee GG, et al. Prospective multicenter evaluation of the Beckman Coulter prostate health index using WHO calibration. J Urol. 2013;189:1702–1706. doi: 10.1016/j.juro.2012.11.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Guazzoni G, Lazzeri M, Nava L, Lughezzani G, Larcher A, Scattoni V, et al. Preoperative prostate-specific antigen isoform p2PSA and its derivatives, %p2PSA and prostate health index, predict pathologic outcomes in patients undergoing radical prostatectomy for prostate cancer. Eur Urol. 2012;61:455–466. doi: 10.1016/j.eururo.2011.10.038. [DOI] [PubMed] [Google Scholar]
  • 55.Lazzeri M, Briganti A, Scattoni V, Lughezzani G, Larcher A, Gadda GM, et al. Serum index test %[-2]proPSA and prostate health index are more accurate than prostate specific antigen and %fPSA in predicting a positive repeat prostate biopsy. J Urol. 2012;188:1137–1143. doi: 10.1016/j.juro.2012.06.017. [DOI] [PubMed] [Google Scholar]
  • 56.Scattoni V, Lazzeri M, Lughezzani G, De Luca S, Passera R, Bollito E, et al. Head-to-head comparison of prostate health index and urinary PCA3 for predicting cancer at initial or repeat biopsy. J Urol. 2013;190:496–501. doi: 10.1016/j.juro.2013.02.3184. [DOI] [PubMed] [Google Scholar]
  • 57.Ferro M, Bruzzese D, Perdonà S, Marino A, Mazzarella C, Perruolo G, et al. Prostate health index (PHI) and prostate cancer antigen 3 (PCA3) significantly improve prostate cancer detection at initial biopsy in a total PSA range of 2-10 ng/ml. PLoS One. 2013;8:e67687. doi: 10.1371/journal.pone.0067687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lazzeri M, Haese A, Abrate A, de la Taille A, Redorta JP, McNicholas T, et al. Clinical performance of serum prostate-specific antigen isoform [-2]proPSA (p2PSA) and its derivatives,%p2PSA and the prostate health index (PHI), in men with a family history of prostate cancer: results from a multicentre European study, the PROMEtheuS project. BJU Int. 2013;112:313–321. doi: 10.1111/bju.12217. [DOI] [PubMed] [Google Scholar]
  • 59.Stephan C, Vincendeau S, Houlgatte A, Cammann H, Jung K, Semjonow A. Multicenter evaluation of [-2]proprostate-specific antigen and the prostate health index for detecting prostate cancer. Clin Chem. 2013;59:306–314. doi: 10.1373/clinchem.2012.195784. [DOI] [PubMed] [Google Scholar]
  • 60.Filella X, Foj L, Augé JM, Molina R, Alcover J. Clinical utility of %p2PSA and prostate health index in the detection of prostate cancer. Clin Chem Lab Med. 2014;52:1347–1355. doi: 10.1515/cclm-2014-0027. [DOI] [PubMed] [Google Scholar]
  • 61.Loeb S, Sanda MG, Broyles DL, Shin SS, Bangma CH, Wei JT, et al. The prostate health index selectively identifies clinically significant prostate cancer. J Urol. 2015;193:1163–1169. doi: 10.1016/j.juro.2014.10.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Abrate A, Lazzeri M, Lughezzani G, Buffi N, Bini V, Haese A, et al. Clinical performance of the prostate health index (PHI) for the prediction of prostate cancer in obese men: data from the PROMEtheuS project, a multicentre European prospective study. BJU Int. 2015;115:537–545. doi: 10.1111/bju.12907. [DOI] [PubMed] [Google Scholar]
  • 63.de la Calle C, Patil D, Wei JT, Scherr DS, Sokoll L, Chan DW, et al. Multicenter evaluation of the prostate health index to detect aggressive prostate cancer in biopsy naïve men. J Urol. 2015;194:65–72. doi: 10.1016/j.juro.2015.01.091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tosoian JJ, Druskin SC, Andreas D, Mullane P, Chappidi M, Joo S, et al. Use of the prostate health index for detection of prostate cancer: results from a large academic practice. Prostate Cancer Prostatic Dis. 2017;20:228–233. doi: 10.1038/pcan.2016.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Foley RW, Gorman L, Sharifi N, Murphy K, Moore H, Tuzova AV, et al. Improving multivariable prostate cancer risk assessment using the prostate health index. BJU Int. 2016;117:409–417. doi: 10.1111/bju.13143. [DOI] [PubMed] [Google Scholar]
  • 66.Chiu PK, Lai FM, Teoh JY, Lee WM, Yee CH, Chan ES, et al. Prostate health index and %p2PSA predict aggressive prostate cancer pathology in Chinese patients undergoing radical prostatectomy. Ann Surg Oncol. 2016;23:2707–2714. doi: 10.1245/s10434-016-5183-6. [DOI] [PubMed] [Google Scholar]
  • 67.Sriplakich S, Lojanapiwat B, Chongruksut W, Phuriyaphan S, Kitirattakarn P, Jun-Ou J, et al. Prospective performance of the prostate health index in prostate cancer detection in the first prostate biopsy of men with a total prostatic specific antigen of 4-10 ng/mL and negative digital rectal examination. Prostate Int. 2018;6:136–139. doi: 10.1016/j.prnil.2018.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hsieh PF, Chang CH, Yang CR, Huang CP, Chen WC, Yeh CC, et al. Prostate health index (PHI) improves prostate cancer detection at initial biopsy in Taiwanese men with PSA 4-10 ng/mL. Kaohsiung J Med Sci. 2018;34:461–466. doi: 10.1016/j.kjms.2018.02.007. [DOI] [PubMed] [Google Scholar]
  • 69.Tang B, Han CT, Lu XL, Wan FN, Zhang CZ, Zhu Y, et al. Preoperative prostate health index predicts poor pathologic outcomes of radical prostatectomy in patients with biopsy-detected low-risk patients prostate cancer: results from a Chinese prospective cohort. Prostate Cancer Prostatic Dis. 2018;21:64–70. doi: 10.1038/s41391-017-0002-0. [DOI] [PubMed] [Google Scholar]
  • 70.Cheng YT, Chiang CH, Pu YS, Liu SP, Lu YC, Chang YK, et al. The application of p2PSA% and prostate health index in prostate cancer detection: a prospective cohort in a tertiary medical center. J Formos Med Assoc. 2019;118(1 Pt 2):260–267. doi: 10.1016/j.jfma.2018.05.001. [DOI] [PubMed] [Google Scholar]
  • 71.Stephan C, Jung K, Lein M, Rochow H, Friedersdorff F, Maxeiner A. PHI density prospectively improves prostate cancer detection. World J Urol. 2021;39:3273–3279. doi: 10.1007/s00345-020-03585-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chiu ST, Cheng YT, Pu YS, Lu YC, Hong JH, Chung SD, et al. Prostate health index density outperforms prostate health index in clinically significant prostate cancer detection. Front Oncol. 2021;11:772182. doi: 10.3389/fonc.2021.772182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Babajide R, Carbunaru S, Nettey OS, Watson KS, Holloway-Beth A, McDowell T, et al. Performance of prostate health index in biopsy naïve Black men. J Urol. 2021;205:718–724. doi: 10.1097/JU.0000000000001453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Garrido MM, Ribeiro R, Pinheiro LC, Holdenrieder S, Guimarães JT. The prostate health index and the percentage of [-2]proPSA maintain their diagnostic performance when calculated with total and free PSA from different manufacturers. Clin Chem Lab Med. 2021;59:1869–1877. doi: 10.1515/cclm-2021-0554. [DOI] [PubMed] [Google Scholar]
  • 75.Filella X, Foj L, Wijngaard R, Luque P. Value of PHI and PHID in the detection of intermediate- and high-risk prostate cancer. Clin Chim Acta. 2022;531:277–282. doi: 10.1016/j.cca.2022.04.992. [DOI] [PubMed] [Google Scholar]
  • 76.Yáñez-Castillo YM, Melgarejo-Segura MT, Funes-Padilla C, Folgueral-Corral ME, García-Larios JV, Arrabal-Polo MA, et al. Prostate health index (PHI) as an accurate prostate cancer predictor. J Cancer Res Clin Oncol. 2023;149:9329–9335. doi: 10.1007/s00432-023-04860-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rius Bilbao L, Valladares Gomez C, Aguirre Larracoechea U, Pereira Arias JG, Arredondo Calvo P, Urdaneta Salegui LF, et al. Do PHI and PHI density improve detection of clinically significant prostate cancer only in the PSA gray zone? Clin Chim Acta. 2023;542:117270. doi: 10.1016/j.cca.2023.117270. [DOI] [PubMed] [Google Scholar]
  • 78.Ito K, Yokomizo A, Tokunaga S, Arai G, Sugimoto M, Akakura K, et al. Members of PROPHET. Diagnostic impacts of clinical laboratory based p2PSA indexes on any grade, gleason grade group 2 or greater, or 3 or greater prostate cancer and prostate specific antigen below 10 ng/ml. J Urol. 2020;203:83–91. doi: 10.1097/JU.0000000000000495. [DOI] [PubMed] [Google Scholar]
  • 79.Tosoian JJ, Druskin SC, Andreas D, Mullane P, Chappidi M, Joo S, et al. Prostate health index density improves detection of clinically significant prostate cancer. BJU Int. 2017;120:793–798. doi: 10.1111/bju.13762. [DOI] [PubMed] [Google Scholar]
  • 80.Schwen ZR, Mamawala M, Tosoian JJ, Druskin SC, Ross AE, Sokoll LJ, et al. Prostate health index and multiparametric magnetic resonance imaging to predict prostate cancer grade reclassification in active surveillance. BJU Int. 2020;126:373–378. doi: 10.1111/bju.15101. [DOI] [PubMed] [Google Scholar]
  • 81.Lazzeri M, Abrate A, Lughezzani G, Gadda GM, Freschi M, Mistretta F, et al. Relationship of chronic histologic prostatic inflammation in biopsy specimens with serum isoform [-2] proPSA (p2PSA), %p2PSA, and prostate health index in men with a total prostate-specific antigen of 4-10 ng/ml and normal digital rectal examination. Urology. 2014;83:606–612. doi: 10.1016/j.urology.2013.10.016. [DOI] [PubMed] [Google Scholar]
  • 82.Loeb S, Shin SS, Broyles DL, Wei JT, Sanda M, Klee G, et al. Prostate health index improves multivariable risk prediction of aggressive prostate cancer. BJU Int. 2017;120:61–68. doi: 10.1111/bju.13676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Mathieu R, Castelli C, Fardoun T, Peyronnet B, Shariat SF, Bensalah K, et al. Cost analysis of prostate cancer detection including the prostate health index (PHI) World J Urol. 2019;37:481–487. doi: 10.1007/s00345-018-2362-z. [DOI] [PubMed] [Google Scholar]
  • 84.Bussemakers MJ, van Bokhoven A, Verhaegh GW, Smit FP, Karthaus HF, Schalken JA, et al. DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res. 1999;59:5975–5979. [PubMed] [Google Scholar]
  • 85.de Kok JB, Verhaegh GW, Roelofs RW, Hessels D, Kiemeney LA, Aalders TW, et al. DD3(PCA3), a very sensitive and specific marker to detect prostate tumors. Cancer Res. 2002;62:2695–2698. [PubMed] [Google Scholar]
  • 86.Durand X, Moutereau S, Xylinas E, de la Taille A. Progensa™ PCA3 test for prostate cancer. Expert Rev Mol Diagn. 2011;11:137–144. doi: 10.1586/erm.10.122. [DOI] [PubMed] [Google Scholar]
  • 87.Tinzl M, Marberger M, Horvath S, Chypre C. DD3PCA3 RNA analysis in urine--a new perspective for detecting prostate cancer. Eur Urol. 2004;46:182–186. doi: 10.1016/j.eururo.2004.06.004. discussion 187. [DOI] [PubMed] [Google Scholar]
  • 88.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;44:8–15. doi: 10.1016/s0302-2838(03)00201-x. discussion 15-6. [DOI] [PubMed] [Google Scholar]
  • 89.Leyten GH, Hessels D, Jannink SA, Smit FP, de Jong H, Cornel EB, et al. Prospective multicentre evaluation of PCA3 and TMPRSS2-ERG gene fusions as diagnostic and prognostic urinary biomarkers for prostate cancer. Eur Urol. 2014;65:534–542. doi: 10.1016/j.eururo.2012.11.014. [DOI] [PubMed] [Google Scholar]
  • 90.Roobol MJ, Schröder 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;58:475–481. doi: 10.1016/j.eururo.2010.06.039. [DOI] [PubMed] [Google Scholar]
  • 91.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;52:1089–1095. doi: 10.1373/clinchem.2005.063289. [DOI] [PubMed] [Google Scholar]
  • 92.Chun FK, de la Taille A, van Poppel H, Marberger M, Stenzl A, Mulders PF, et al. Prostate cancer gene 3 (PCA3): development and internal validation of a novel biopsy nomogram. Eur Urol. 2009;56:659–667. doi: 10.1016/j.eururo.2009.03.029. [DOI] [PubMed] [Google Scholar]
  • 93.Pepe P, Fraggetta F, Galia A, Skonieczny G, Aragona F. PCA3 score and prostate cancer diagnosis at repeated saturation biopsy. Which cut-off: 20 or 35? Int Braz J Urol. 2012;38:489–495. doi: 10.1590/s1677-55382012000400008. [DOI] [PubMed] [Google Scholar]
  • 94.Klatte T, Waldert M, de Martino M, Schatzl G, Mannhalter C, Remzi M. Age-specific PCA3 score reference values for diagnosis of prostate cancer. World J Urol. 2012;30:405–410. doi: 10.1007/s00345-011-0749-1. [DOI] [PubMed] [Google Scholar]
  • 95.Wu AK, Reese AC, Cooperberg MR, Sadetsky N, Shinohara K. Utility of PCA3 in patients undergoing repeat biopsy for prostate cancer. Prostate Cancer Prostatic Dis. 2012;15:100–105. doi: 10.1038/pcan.2011.52. [DOI] [PubMed] [Google Scholar]
  • 96.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;188:1726–1731. doi: 10.1016/j.juro.2012.07.023. [DOI] [PubMed] [Google Scholar]
  • 97.Cornu JN, Cancel-Tassin G, Egrot C, Gaffory C, Haab F, Cussenot O. Urine TMPRSS2:ERG fusion transcript integrated with PCA3 score, genotyping, and biological features are correlated to the results of prostatic biopsies in men at risk of prostate cancer. Prostate. 2013;73:242–249. doi: 10.1002/pros.22563. [DOI] [PubMed] [Google Scholar]
  • 98.Ruffion A, Devonec M, Champetier D, Decaussin-Petrucci M, Rodriguez-Lafrasse C, Paparel P, et al. PCA3 and PCA3-based nomograms improve diagnostic accuracy in patients undergoing first prostate biopsy. Int J Mol Sci. 2013;14:17767–17780. doi: 10.3390/ijms140917767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chevli KK, Duff M, Walter P, Yu C, Capuder B, Elshafei A, et al. Urinary PCA3 as a predictor of prostate cancer in a cohort of 3,073 men undergoing initial prostate biopsy. J Urol. 2014;191:1743–1748. doi: 10.1016/j.juro.2013.12.005. [DOI] [PubMed] [Google Scholar]
  • 100.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;111:928–933. doi: 10.1111/j.1464-410X.2012.11683.x. [DOI] [PubMed] [Google Scholar]
  • 101.Wei JT, Feng Z, Partin AW, Brown E, Thompson I, Sokoll L, et al. Can urinary PCA3 supplement PSA in the early detection of prostate cancer? J Clin Oncol. 2014;32:4066–4072. doi: 10.1200/JCO.2013.52.8505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Capoluongo E, Zambon CF, Basso D, Boccia S, Rocchetti S, Leoncini E, et al. PCA3 score of 20 could improve prostate cancer detection: results obtained on 734 Italian individuals. Clin Chim Acta. 2014;429:46–50. doi: 10.1016/j.cca.2013.10.022. [DOI] [PubMed] [Google Scholar]
  • 103.Merola R, Tomao L, Antenucci A, Sperduti I, Sentinelli S, Masi S, et al. PCA3 in prostate cancer and tumor aggressiveness detection on 407 high-risk patients: a National Cancer Institute experience. J Exp Clin Cancer Res. 2015;34:15. doi: 10.1186/s13046-015-0127-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zheng K, Dou Y, He L, Li H, Zhang Z, Chen Y, et al. Improved sensitivity and specificity for prostate cancer diagnosis based on the urine PCA3/PSA ratio acquired by sequence-specific RNA capture. Oncol Rep. 2015;34:2439–2444. doi: 10.3892/or.2015.4266. [DOI] [PubMed] [Google Scholar]
  • 105.Tomlins SA, Day JR, Lonigro RJ, Hovelson DH, Siddiqui J, Kunju LP, et al. Urine TMPRSS2:ERG Plus PCA3 for individualized prostate cancer risk assessment. Eur Urol. 2016;70:45–53. doi: 10.1016/j.eururo.2015.04.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Feibus AH, Sartor O, Moparty K, Chagin K, Kattan MW, Ledet E, et al. Clinical use of PCA3 and TMPRSS2:ERG urinary biomarkers in African-American men undergoing prostate biopsy. J Urol. 2016;196:1053–1060. doi: 10.1016/j.juro.2016.04.075. [DOI] [PubMed] [Google Scholar]
  • 107.Cao L, Lee CH, Ning J, Handy BC, Wagar EA, Meng QH. Combination of prostate cancer antigen 3 and prostate-specific antigen improves diagnostic accuracy in men at risk of prostate cancer. Arch Pathol Lab Med. 2018;142:1106–1112. doi: 10.5858/arpa.2017-0185-OA. [DOI] [PubMed] [Google Scholar]
  • 108.Gan J, Zeng X, Wang X, Wu Y, Lei P, Wang Z, et al. Effective diagnosis of prostate cancer based on mRNAs from urinary exosomes. Front Med (Lausanne) 2022;9:736110. doi: 10.3389/fmed.2022.736110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ankerst DP, Goros M, Tomlins SA, Patil D, Feng Z, Wei JT, et al. Incorporation of urinary prostate cancer antigen 3 and TMPRSS2:ERG into prostate cancer prevention trial risk calculator. Eur Urol Focus. 2019;5:54–61. doi: 10.1016/j.euf.2018.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Newcomb LF, Zheng Y, Faino AV, Bianchi-Frias D, Cooperberg MR, Brown MD, et al. Performance of PCA3 and TMPRSS2:ERG urinary biomarkers in prediction of biopsy outcome in the Canary Prostate Active Surveillance Study (PASS) Prostate Cancer Prostatic Dis. 2019;22:438–445. doi: 10.1038/s41391-018-0124-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.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;3:94ra72. doi: 10.1126/scitranslmed.3001970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Demichelis F, Fall K, Perner S, Andrén O, Schmidt F, Setlur SR, et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene. 2007;26:4596–4599. doi: 10.1038/sj.onc.1210237. Erratum in: Oncogene 2007;26:5692. [DOI] [PubMed] [Google Scholar]
  • 113.Salami SS, Schmidt F, Laxman B, Regan MM, Rickman DS, Scherr D, et al. Combining urinary detection of TMPRSS2:ERG and PCA3 with serum PSA to predict diagnosis of prostate cancer. Urol Oncol. 2013;31:566–571. doi: 10.1016/j.urolonc.2011.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cani AK, Hu K, Liu CJ, Siddiqui J, Zheng Y, Han S, et al. Development of a whole-urine, multiplexed, next-generation RNA-sequencing assay for early detection of aggressive prostate cancer. Eur Urol Oncol. 2022;5:430–439. doi: 10.1016/j.euo.2021.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Pettersson A, Graff RE, Bauer SR, Pitt MJ, Lis RT, Stack EC, et al. The TMPRSS2:ERG rearrangement, ERG expression, and prostate cancer outcomes: a cohort study and meta-analysis. Cancer Epidemiol Biomarkers Prev. 2012;21:1497–1509. doi: 10.1158/1055-9965.EPI-12-0042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tosoian JJ, Sessine MS, Trock BJ, Ross AE, Xie C, Zheng Y, et al. MyProstateScore in men considering repeat biopsy: validation of a simple testing approach. Prostate Cancer Prostatic Dis. 2023;26:563–567. doi: 10.1038/s41391-022-00633-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Tosoian JJ, Singhal U, Davenport MS, Wei JT, Montgomery JS, George AK, et al. Urinary MyProstateScore (MPS) to rule out clinically-significant cancer in men with equivocal (PI-RADS 3) multiparametric MRI: addressing an unmet clinical need. Urology. 2022;164:184–190. doi: 10.1016/j.urology.2021.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Jiao B, Gulati R, Hendrix N, Gore JL, Rais-Bahrami S, Morgan TM, et al. Economic evaluation of urine-based or magnetic resonance imaging reflex tests in men with intermediate prostate-specific antigen levels in the United States. Value Health. 2021;24:1111–1117. doi: 10.1016/j.jval.2021.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Leyten GH, Hessels D, Smit FP, Jannink SA, de Jong H, Melchers WJ, et al. Identification of a candidate gene panel for the early diagnosis of prostate cancer. Clin Cancer Res. 2015;21:3061–3070. doi: 10.1158/1078-0432.CCR-14-3334. [DOI] [PubMed] [Google Scholar]
  • 120.Sari Motlagh R, Yanagisawa T, Kawada T, Laukhtina E, Rajwa P, Aydh A, et al. Accuracy of SelectMDx compared to mpMRI in the diagnosis of prostate cancer: a systematic review and diagnostic meta-analysis. Prostate Cancer Prostatic Dis. 2022;25:187–198. doi: 10.1038/s41391-022-00538-1. [DOI] [PubMed] [Google Scholar]
  • 121.Maggi M, Del Giudice F, Falagario UG, Cocci A, Russo GI, Di Mauro M, et al. SelectMDx and multiparametric magnetic resonance imaging of the prostate for men undergoing primary prostate biopsy: a prospective assessment in a multi-institutional study. Cancers (Basel) 2021;13:2047. doi: 10.3390/cancers13092047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Hendriks RJ, van der Leest MMG, Israël B, Hannink G, YantiSetiasti A, Cornel EB, et al. Clinical use of the SelectMDx urinary-biomarker test with or without mpMRI in prostate cancer diagnosis: a prospective, multicenter study in biopsy-naïve men. Prostate Cancer Prostatic Dis. 2021;24:1110–1119. doi: 10.1038/s41391-021-00367-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hendriks RJ, van der Leest MMG, Dijkstra S, Barentsz JO, Van Criekinge W, Hulsbergen-van de Kaa CA, et al. A urinary biomarker-based risk score correlates with multiparametric MRI for prostate cancer detection. Prostate. 2017;77:1401–1407. doi: 10.1002/pros.23401. [DOI] [PubMed] [Google Scholar]
  • 124.Haese A, Trooskens G, Steyaert S, Hessels D, Brawer M, Vlaeminck-Guillem V, et al. Multicenter optimization and validation of a 2-gene mRNA urine test for detection of clinically significant prostate cancer before initial prostate biopsy. J Urol. 2019;202:256–263. doi: 10.1097/JU.0000000000000293. [DOI] [PubMed] [Google Scholar]
  • 125.Margolis E, Brown G, Partin A, Carter B, McKiernan J, Tutrone R, et al. Predicting high-grade prostate cancer at initial biopsy: clinical performance of the ExoDx (EPI) Prostate Intelliscore test in three independent prospective studies. Prostate Cancer Prostatic Dis. 2022;25:296–301. doi: 10.1038/s41391-021-00456-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Roumiguié M, Ploussard G, Nogueira L, Bruguière E, Meyrignac O, Lesourd M, et al. Independent evaluation of the respective predictive values for high-grade prostate cancer of clinical information and RNA biomarkers after upfront MRI and image-guided biopsies. Cancers (Basel) 2020;12:285. doi: 10.3390/cancers12020285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Pepe P, Dibenedetto G, Pepe L, Pennisi M. Multiparametric MRI versus SelectMDx accuracy in the diagnosis of clinically significant PCa in men enrolled in active surveillance. In Vivo. 2020;34:393–396. doi: 10.21873/invivo.11786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Busetto GM, Del Giudice F, Maggi M, De Marco F, Porreca A, Sperduti I, et al. Prospective assessment of two-gene urinary test with multiparametric magnetic resonance imaging of the prostate for men undergoing primary prostate biopsy. World J Urol. 2021;39:1869–1877. doi: 10.1007/s00345-020-03359-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lendínez-Cano G, Ojeda-Claro AV, Gómez-Gómez E, Morales Jimenez P, Flores Martin J, Dominguez JF, et al. AEU-PIEM/2018/000 Investigators. Prospective study of diagnostic accuracy in the detection of high-grade prostate cancer in biopsy-naïve patients with clinical suspicion of prostate cancer who underwent the Select MDx test. Prostate. 2021;81:857–865. doi: 10.1002/pros.24182. [DOI] [PubMed] [Google Scholar]
  • 130.Fiorella D, Marenco JL, Mascarós JM, Borque-Fernando Á, Esteban LM, Calatrava A, et al. Role of PCA3 and SelectMDx in the optimization of active surveillance in prostate cancer. Actas Urol Esp (Engl Ed) 2021;45:439–446. doi: 10.1016/j.acuroe.2020.10.013. [DOI] [PubMed] [Google Scholar]
  • 131.Katzendorn O, von Klot CAJ, Mahjoub S, Faraj Tabrizi P, Harke NN, Tezval H, et al. Combination of PI-RADS score and mRNA urine test-a novel scoring system for improved detection of prostate cancer. PLoS One. 2022;17:e0271981. doi: 10.1371/journal.pone.0271981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Henning GM, Andriole GL, Kim EH. Liquid biomarkers for early detection of prostate cancer and summary of available data for their use in African-American men. Prostate Cancer Prostatic Dis. 2022;25:180–186. doi: 10.1038/s41391-022-00507-8. [DOI] [PubMed] [Google Scholar]
  • 133.Govers TM, Hessels D, Vlaeminck-Guillem V, Schmitz-Dräger BJ, Stief CG, Martinez-Ballesteros C, et al. Cost-effectiveness of SelectMDx for prostate cancer in four European countries: a comparative modeling study. Prostate Cancer Prostatic Dis. 2019;22:101–109. doi: 10.1038/s41391-018-0076-3. [DOI] [PubMed] [Google Scholar]
  • 134.Visser WCH, de Jong H, Steyaert S, Melchers WJG, Mulders PFA, Schalken JA. Clinical use of the mRNA urinary biomarker SelectMDx test for prostate cancer. Prostate Cancer Prostatic Dis. 2022;25:583–589. doi: 10.1038/s41391-022-00562-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Donovan MJ, Noerholm M, Bentink S, Belzer S, Skog J, O'Neill V, et al. A molecular signature of PCA3 and ERG exosomal RNA from non-DRE urine is predictive of initial prostate biopsy result. Prostate Cancer Prostatic Dis. 2015;18:370–375. doi: 10.1038/pcan.2015.40. [DOI] [PubMed] [Google Scholar]
  • 136.McKiernan J, Donovan MJ, O'Neill V, Bentink S, Noerholm M, Belzer S, et al. A novel urine exosome gene expression assay to predict high-grade prostate cancer at initial biopsy. JAMA Oncol. 2016;2:882–889. doi: 10.1001/jamaoncol.2016.0097. [DOI] [PubMed] [Google Scholar]
  • 137.McKiernan J, Noerholm M, Tadigotla V, Kumar S, Torkler P, Sant G, et al. A urine-based Exosomal gene expression test stratifies risk of high-grade prostate cancer in men with prior negative prostate biopsy undergoing repeat biopsy. BMC Urol. 2020;20:138. doi: 10.1186/s12894-020-00712-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kretschmer A, Tutrone R, Alter J, Berg E, Fischer C, Kumar S, et al. Pre-diagnosis urine exosomal RNA (ExoDx EPI score) is associated with post-prostatectomy pathology outcome. World J Urol. 2022;40:983–989. doi: 10.1007/s00345-022-03937-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.McKiernan J, Donovan MJ, Margolis E, Partin A, Carter B, Brown G, et al. A prospective adaptive utility trial to validate performance of a novel urine exosome gene expression assay to predict high-grade prostate cancer in patients with prostate-specific antigen 2-10ng/ml at initial biopsy. Eur Urol. 2018;74:731–738. doi: 10.1016/j.eururo.2018.08.019. [DOI] [PubMed] [Google Scholar]
  • 140.Tutrone R, Donovan MJ, Torkler P, Tadigotla V, McLain T, Noerholm M, et al. Clinical utility of the exosome based ExoDx Prostate(IntelliScore) EPI test in men presenting for initial biopsy with a PSA 2-10 ng/mL. Prostate Cancer Prostatic Dis. 2020;23:607–614. doi: 10.1038/s41391-020-0237-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sathianathen NJ, Kuntz KM, Alarid-Escudero F, Lawrentschuk NL, Bolton DM, Murphy DG, et al. Incorporating biomarkers into the primary prostate biopsy setting: a cost-effectiveness analysis. J Urol. 2018;200:1215–1220. doi: 10.1016/j.juro.2018.06.016. [DOI] [PubMed] [Google Scholar]
  • 142.Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Singh N, Ramnarine VR, Song JH, Pandey R, Padi SKR, Nouri M, et al. The long noncoding RNA H19 regulates tumor plasticity in neuroendocrine prostate cancer. Nat Commun. 2021;12:7349. doi: 10.1038/s41467-021-26901-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Mirzaei S, Paskeh MDA, Okina E, Gholami MH, Hushmandi K, Hashemi M, et al. Molecular landscape of LncRNAs in prostate cancer: a focus on pathways and therapeutic targets for intervention. J Exp Clin Cancer Res. 2022;41:214. doi: 10.1186/s13046-022-02406-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bhan A, Soleimani M, Mandal SS. Long noncoding RNA and cancer: a new paradigm. Cancer Res. 2017;77:3965–3981. doi: 10.1158/0008-5472.CAN-16-2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136:629–641. doi: 10.1016/j.cell.2009.02.006. [DOI] [PubMed] [Google Scholar]
  • 147.Liang C, Qi Z, Ge H, Liang C, Zhang Y, Wang Z, et al. Long non-coding RNA PCAT-1 in human cancers: a meta-analysis. Clin Chim Acta. 2018;480:47–55. doi: 10.1016/j.cca.2018.01.043. [DOI] [PubMed] [Google Scholar]
  • 148.Xue Y, Wang M, Kang M, Wang Q, Wu B, Chu H, et al. Association between lncrna PCGEM1 polymorphisms and prostate cancer risk. Prostate Cancer Prostatic Dis. 2013;16:139–144. S1. doi: 10.1038/pcan.2013.6. [DOI] [PubMed] [Google Scholar]
  • 149.Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, et al. RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol. 2014;15:1469–1480. doi: 10.1016/S1470-2045(14)71113-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Prensner JR, Iyer MK, Sahu A, Asangani IA, Cao Q, Patel L, et al. The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. Nat Genet. 2013;45:1392–1398. doi: 10.1038/ng.2771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Mehra R, Udager AM, Ahearn TU, Cao X, Feng FY, Loda M, et al. Overexpression of the long non-coding RNA SChLAP1 independently predicts lethal prostate cancer. Eur Urol. 2016;70:549–552. doi: 10.1016/j.eururo.2015.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Hua JT, Ahmed M, Guo H, Zhang Y, Chen S, Soares F, et al. Risk SNP-mediated promoter-enhancer switching drives prostate cancer through lncRNA PCAT19. Cell. 2018;174:564–575.e18. doi: 10.1016/j.cell.2018.06.014. [DOI] [PubMed] [Google Scholar]
  • 153.O'Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne) 2018;9:402. doi: 10.3389/fendo.2018.00402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Sharova E, Grassi A, Marcer A, Ruggero K, Pinto F, Bassi P, et al. A circulating miRNA assay as a first-line test for prostate cancer screening. Br J Cancer. 2016;114:1362–1366. doi: 10.1038/bjc.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Matsuzaki K, Fujita K, Tomiyama E, Hatano K, Hayashi Y, Wang C, et al. MiR-30b-3p and miR-126-3p of urinary extracellular vesicles could be new biomarkers for prostate cancer. Transl Androl Urol. 2021;10:1918–1927. doi: 10.21037/tau-20-421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Bautista-Sánchez D, Arriaga-Canon C, Pedroza-Torres A, De La Rosa-Velázquez IA, González-Barrios R, Contreras-Espinosa L, et al. The promising role of miR-21 as a cancer biomarker and its importance in RNA-based therapeutics. Mol Ther Nucleic Acids. 2020;20:409–420. doi: 10.1016/j.omtn.2020.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Martens-Uzunova ES, Jalava SE, Dits NF, van Leenders GJ, Møller S, Trapman J, et al. Diagnostic and prognostic signatures from the small non-coding RNA transcriptome in prostate cancer. Oncogene. 2012;31:978–991. doi: 10.1038/onc.2011.304. [DOI] [PubMed] [Google Scholar]
  • 158.Hatano K, Kumar B, Zhang Y, Coulter JB, Hedayati M, Mears B, et al. A functional screen identifies miRNAs that inhibit DNA repair and sensitize prostate cancer cells to ionizing radiation. Nucleic Acids Res. 2015;43:4075–4086. doi: 10.1093/nar/gkv273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Wang WW, Sorokin I, Aleksic I, Fisher H, Kaufman RP, Jr, Winer A, et al. Expression of small noncoding RNAs in urinary exosomes classifies prostate cancer into indolent and aggressive disease. J Urol. 2020;204:466–475. doi: 10.1097/JU.0000000000001020. [DOI] [PubMed] [Google Scholar]
  • 160.Munkley J, Mills IG, Elliott DJ. The role of glycans in the development and progression of prostate cancer. Nat Rev Urol. 2016;13:324–333. doi: 10.1038/nrurol.2016.65. [DOI] [PubMed] [Google Scholar]
  • 161.Fujita K, Shimomura M, Uemura M, Nakata W, Sato M, Nagahara A, et al. Serum fucosylated haptoglobin as a novel prognostic biomarker predicting high-Gleason prostate cancer. Prostate. 2014;74:1052–1058. doi: 10.1002/pros.22824. [DOI] [PubMed] [Google Scholar]
  • 162.Llop E, Ferrer-Batallé M, Barrabés S, Guerrero PE, Ramírez M, Saldova R, et al. Improvement of prostate cancer diagnosis by detecting PSA glycosylation-specific changes. Theranostics. 2016;6:1190–1204. doi: 10.7150/thno.15226. Erratum in: Theranostics 2018;8:746-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Marek KW, Vijay IK, Marth JD. A recessive deletion in the GlcNAc-1-phosphotransferase gene results in peri-implantation embryonic lethality. Glycobiology. 1999;9:1263–1271. doi: 10.1093/glycob/9.11.1263. [DOI] [PubMed] [Google Scholar]
  • 164.Freeze HH. Genetic defects in the human glycome. Nat Rev Genet. 2006;7:537–551. doi: 10.1038/nrg1894. Erratum in: Nat Rev Genet 2006;7:660. [DOI] [PubMed] [Google Scholar]
  • 165.Gilgunn S, Conroy PJ, Saldova R, Rudd PM, O'Kennedy RJ. Aberrant PSA glycosylation--a sweet predictor of prostate cancer. Nat Rev Urol. 2013;10:99–107. doi: 10.1038/nrurol.2012.258. [DOI] [PubMed] [Google Scholar]
  • 166.Saldova R, Fan Y, Fitzpatrick JM, Watson RW, Rudd PM. Core fucosylation and alpha2-3 sialylation in serum N-glycome is significantly increased in prostate cancer comparing to benign prostate hyperplasia. Glycobiology. 2011;21:195–205. doi: 10.1093/glycob/cwq147. [DOI] [PubMed] [Google Scholar]
  • 167.Ohyama C, Hosono M, Nitta K, Oh-eda M, Yoshikawa K, Habuchi T, et al. Carbohydrate structure and differential binding of prostate specific antigen to Maackia amurensis lectin between prostate cancer and benign prostate hypertrophy. Glycobiology. 2004;14:671–679. doi: 10.1093/glycob/cwh071. [DOI] [PubMed] [Google Scholar]
  • 168.Yoneyama T, Yamamoto H, Sutoh Yoneyama M, Tobisawa Y, Hatakeyama S, Narita T, et al. Characteristics of α2,3-sialyl N-glycosylated PSA as a biomarker for clinically significant prostate cancer in men with elevated PSA level. Prostate. 2021;81:1411–1427. doi: 10.1002/pros.24239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Fujita K, Hatano K, Tomiyama E, Hayashi Y, Matsushita M, Tsuchiya M, et al. Serum core-type fucosylated prostate-specific antigen index for the detection of high-risk prostate cancer. Int J Cancer. 2021;148:3111–3118. doi: 10.1002/ijc.33517. [DOI] [PubMed] [Google Scholar]
  • 170.Hatano K, Yoneyama T, Hatakeyama S, Tomiyama E, Tsuchiya M, Nishimoto M, et al. Simultaneous analysis of serum α2,3-linked sialylation and core-type fucosylation of prostate-specific antigen for the detection of high-grade prostate cancer. Br J Cancer. 2022;126:764–770. doi: 10.1038/s41416-021-01637-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Herberts C, Wyatt AW. Technical and biological constraints on ctDNA-based genotyping. Trends Cancer. 2021;7:995–1009. doi: 10.1016/j.trecan.2021.06.001. [DOI] [PubMed] [Google Scholar]
  • 172.Feinberg AP, Vogelstein B. Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature. 1983;301:89–92. doi: 10.1038/301089a0. [DOI] [PubMed] [Google Scholar]
  • 173.Schulz WA, Hatina J. Epigenetics of prostate cancer: beyond DNA methylation. J Cell Mol Med. 2006;10:100–125. doi: 10.1111/j.1582-4934.2006.tb00293.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Serenaite I, Daniunaite K, Jankevicius F, Laurinavicius A, Petroska D, Lazutka JR, et al. Heterogeneity of DNA methylation in multifocal prostate cancer. Virchows Arch. 2015;466:53–59. doi: 10.1007/s00428-014-1678-3. [DOI] [PubMed] [Google Scholar]
  • 175.Steiner I, Jung K, Schatz P, Horns T, Wittschieber D, Lein M, et al. Gene promoter methylation and its potential relevance in early prostate cancer diagnosis. Pathobiology. 2010;77:260–266. doi: 10.1159/000318017. [DOI] [PubMed] [Google Scholar]
  • 176.Bjerre MT, Nørgaard M, Larsen OH, Jensen SØ, Strand SH, Østergren P, et al. Epigenetic analysis of circulating tumor DNA in localized and metastatic prostate cancer: evaluation of clinical biomarker potential. Cells. 2020;9:1362. doi: 10.3390/cells9061362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Ylitalo EB, Thysell E, Landfors M, Brattsand M, Jernberg E, Crnalic S, et al. A novel DNA methylation signature is associated with androgen receptor activity and patient prognosis in bone metastatic prostate cancer. Clin Epigenetics. 2021;13:133. doi: 10.1186/s13148-021-01119-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Constâncio V, Nunes SP, Henrique R, Jerónimo C. DNA methylation-based testing in liquid biopsies as detection and prognostic biomarkers for the four major cancer types. Cells. 2020;9:624. doi: 10.3390/cells9030624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Pu Y, Li C, Yuan H, Wang X. Identification of prostate cancer specific methylation biomarkers from a multi-cancer analysis. BMC Bioinformatics. 2021;22:492. doi: 10.1186/s12859-021-04416-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Mundbjerg K, Chopra S, Alemozaffar M, Duymich C, Lakshminarasimhan R, Nichols PW, et al. Identifying aggressive prostate cancer foci using a DNA methylation classifier. Genome Biol. 2017;18:3. doi: 10.1186/s13059-016-1129-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Zhao F, Olkhov-Mitsel E, van der Kwast T, Sykes J, Zdravic D, Venkateswaran V, et al. Urinary DNA methylation biomarkers for noninvasive prediction of aggressive disease in patients with prostate cancer on active surveillance. J Urol. 2017;197:335–341. doi: 10.1016/j.juro.2016.08.081. [DOI] [PubMed] [Google Scholar]
  • 182.Hatano K, Nonomura N. Genomic profiling of prostate cancer: an updated review. World J Mens Health. 2022;40:368–379. doi: 10.5534/wjmh.210072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Eggener SE, Rumble RB, Armstrong AJ, Morgan TM, Crispino T, Cornford P, et al. Molecular biomarkers in localized prostate cancer: ASCO guideline. J Clin Oncol. 2020;38:1474–1494. doi: 10.1200/JCO.19.02768. [DOI] [PubMed] [Google Scholar]

Articles from The World Journal of Men's Health are provided here courtesy of Korean Society for Sexual Medicine and Andrology

RESOURCES