How Precisely Can Prostate Cancer Be Managed?

Article information

Int Neurourol J. 2016;20(Suppl 2):S120-130
Publication date (electronic) : 2016 November 22
doi : https://doi.org/10.5213/inj.1632724.362
1Department of Urology, Lahey Hospital and Medical Center, Tufts University School of Medicine, Burlington, MA, USA
2Urology Consultants, Ltd., St. Louis, MO, USA
Corresponding author: Liyan Zhuang http://orcid.org/0000-0003-2258-0176  Department of Urology, Lahey Hospital and Medical Center, Tufts University School of Medicine, 41 Mall Road, Burlington, MA 01805, USA  E-mail: liyanzhuang@yahoo.com / Tel: +1-603-594-0800 / Fax: +1-603-879-9329
Received 2016 September 23; Accepted 2016 October 17.

Abstract

Progress has been made in applying genetic information to disease management in the postgenomic era, and precision medicine is emerging in prostate cancer management. The prostate health index, the 4-kallikrein (4K) score, and the PCA3, TMPRSS2-ERG, and Prostarix tests have potential for refining prostate cancer screening in conjunction with traditional prostate-specific antigen testing. The Confirm MDx and PCA3 tests have shown promise in identifying men who need be rebiopsied after a primary negative biopsy. Oncotype DX, Prolaris, the biopsy-based Decipher prostate cancer test, and ProMark may improve predictive risk stratification in addition to the traditional Gleason score and tumor stage. Decipher and Prolaris may predict biochemical recurrence and metastasis after radical prostatectomy and possibly help identify patients who need adjuvant therapy. Androgen receptor splice variant 7 appears effective in guiding the selection of second hormonal manipulation with abiraterone or enzalutamide versus chemotherapy when treating metastatic castration-resistant prostate cancer.

INTRODUCTION

Precision medicine is an emerging approach for disease management that focuses on individual variability at the genomic, transcriptomic, proteomic, and metabolomic levels. Precision medicine has been used in cancer screening, stratification, guidance of treatment, and outcome prediction. Precision medicine has exciting and evolving applications in prostate cancer (PCa) management.

PCa is the most common cancer in United States (US) men and is the second leading cause of cancer mortality in men. In 2016, it is estimated that 180,890 men will be diagnosed with PCa, and 26,120 will die of PCa [1]. In 2016, the death rate in the US was found to have fallen to 50% of the peak rate. It is believed that early detection and improved treatment are important in decreasing cancer-related mortality [1]. Although the early detection and treatment of PCa improve survival, this must be balanced against overdiagnosis and overtreatment. In 2012, the United States Preventive Services Task Force recommended against prostate-specific antigen (PSA)-based screening for PCa (grade D recommendation), given concerns that the overdiagnosis and overtreatment of PCa may cause more harm than good [2]. However, this criticism was largely influenced by the US Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial, a randomized clinical trial with high PSA screening contamination in the control arm [3]. Regardless, the grade D recommendation obviously challenges the current less-than-perfect PCa management. To improve the accuracy of diagnosis, the selection of necessary curative treatments, appropriate adjuvant therapy approaches, and the individualized treatment of metastatic disease, the development of precision medicine in PCa is as imperative as it is promising.

This review focuses on how currently available precision medicine tests are applied to common and challenging scenarios encountered in the management of PCa patients. We will discuss the following questions: (1) Who needs to be biopsied? (2) Who needs to be rebiopsied? (3) Who should be offered curative treatment or active surveillance? (4) Who needs adjuvant therapy after radical prostatectomy? (5) Who is a good candidate for secondary hormonal manipulation in castration resistant PCa?

WHO NEEDS TO BE BIOPSIED?

Although the benefit of PCa screening has been recently debated, many providers believe that PSA-based screening is still appropriate for early PCa detection. The 2016 National Comprehensive Cancer Network (NCCN) guidelines for the early detection of PCa recommended a PSA cutoff of 3 ng/mL in select populations to consider prostate biopsy. Patients with a PSA of 3–10 ng/mL are in a gray zone for prostate biopsy. Based on several studies [3,4], only 25%–35% of men with a PSA of 4–10 ng/mL who undergo prostate biopsy will be diagnosed with PCa, and most of those tumors will be low-grade. As recommended by NCCN guidelines, free PSA (fPSA), the 4 kallikrein (4K) and prostate health index (PHI) tests are recommended as options to detect more clinically significant PCa in addition to prostate biopsy or follow-up in 6–12 months with PSA testing and a digital rectal examination (DRE).

Free PSA

In contrast to total PSA (tPSA) in serum, %fPSA is the proportion of unbound protein. After the identification of fPSA [5], in 1998, the Food and Drug Administration (FDA) approved its use to improve PCa detection in men with total PSA levels of 4–10 ng/mL, largely based on the study by Catalona et al. [6]. This study showed that %fPSA can reduce unnecessary biopsies by 20% with a 95% detection rate in patients with a %fPSA≤25%, a tPSA level of 4–10 ng/mL, and a normal DRE. The %fPSA significantly improved the predictive accuracy of PCa detection, with an area under the curve (AUC) of 0.72 vs. 0.53 for tPSA alone.

Although the above findings have been confirmed in other studies and reviews [7-9], %fPSA showed very little additional value in combination with the clinical application of tPSA for early detection in a prospective study [10]. This study included 246 consecutive men with prostate biopsies and showed that when the cutoff was set at 25%, the sensitivity for detecting cancer was 95%, but the specificity was only 20% for men with a PSA level between 4 and 10 ng/mL.

Prostate Health Index

The PHI is a blood test using total PSA, fPSA, and [-2] proPSA, a PCa-associated isoform of fPSA [11]. This serum-based test was developed at Beckman Coulter (Brea, CA, USA) by putting the values of the above 3 PSA forms into an equation to obtain a PHI score. In multicenter studies, the PHI has been found to detect PCa with a greater specificity than total PSA and %fPSA [11-17], and showed a sensitivity nearly double that of %fPSA for cancer detection in men with a PSA between 2 and 10 ng/mL. In addition, the PHI showed more power in discriminating high-grade (Gleason score≥7) cancer from low-grade cancer or prior negative biopsy (AUC of 0.815) [11,18]. The PHI is an FDA-approved test to refine the specificity of PCa detection in initial and repeat biopsies. The PHI is emerging as a more specific test to avoid unnecessary prostate biopsies, as approximately 36% of unnecessary biopsies would be avoided using the PHI, and only 2.5% of high-grade cancers would missed in patients with a tPSA range of 3–10 ng/mL.

4K Score

The 4K score is a serum based test of 4K markers: tPSA, fPSA, intact PSA, and human kallikrein 2. The test also takes the patient’s age, prior biopsy results, and DRE into consideration to predict the prostate biopsy outcome. Several studies have confirmed its accuracy in the detection of clinically significant cancer (Gleason score≥7) and its potential to reduce the number of unnecessary biopsies [19-21].

In a case-control study, 12,542 men were followed for >15 years. A test model based on 4K scores significantly enhanced the prediction of metastasis compared with PSA levels alone [22]. In a prospective multicenter US trial of 1,012 patients, a high discrimination value was reported (AUC=0.82). The study also showed that using 4K scores would avoid 58% of unnecessary biopsies while missing 4.7% of high-grade cancers [20].

Although the above 3 NCCN-recommended biomarkers are related to PSA derivatives, other markers may have potential to refine PCa detection in primary and secondary biopsies.

PCA3

PCA3 is a noncoding, prostate-specific RNA sequence that is overexpressed in PCa cells [23,24]. The PCA3 gene was first identified by Bussemakers et al. [25] in 1999 and mapped to chromosome 9q21-22. The contemporary PCA3 test is a urinary assay measuring PCA3 mRNA normalized with PSA in prostate cells [26]. The PCA3 score is calculated as the ratio of PCA3 mRNA to PSA mRNA multiplied by 1,000.

Currently, a PCA3 score cutoff of 25 for repeat biopsy is well accepted [27-29]. When a PCA3 score >25 was used for the choice of performing a repeat biopsy, the biopsy sensitivity was 78% and specificity was 57%.

Although PCA3 is specifically overexpressed in PCa cells, the PCA3 test appears most informative in the repeat prostate biopsy setting. Its value in primary biopsy is limited by missing more high-risk (Gleason score>6) cancers in primary biopsies than in repeat biopsies [30-32]. For example, the Early Detection Research Network (EDRN) validation study in 895 men from 11 centers showed if the lower limit was set at <20, up to 13% of high-risk cancers would be missed in primary biopsies, but only 3% in repeat biopsies [30]. Although the NCCN and FDA do not suggest using PCA3 in the initial biopsy setting, the EDRN study demonstrated a positive predictive value of 80% if the PCA3 score cutoff was set at 60. This may make the PCA3 test valuable as a marker in patients prescreened by PSA for biopsy, especially combined with other clinical factors, such as age, PSA, DRE, prostate volume, or nomograms.

Prostarix

Prostarix is based on the metabolomics of PCa cells and was developed by Metabolon Inc. (Durham, NC, USA) as a post-DRE urine test. The test is based on the quantitative measurement of metabolites in the urine (sarcosine, alanine, glycine, and glutamate), which are combined in a logistic regression algorithm to generate a Prostarix risk score.

The test has been used for initial cancer evaluation to determine which men are candidates for prostate biopsy as well as for repeat biopsy decision-making for men with prior negative biopsies.

Sarcosine, a glycine metabolite, has also recently been identified as a mechanistic marker of PCa progression [33,34]. However, the specificity of sarcosine in the urine of PCa patients has been recently challenged. In a study evaluating amino acids as PCa biomarkers, 50 urine samples from PCa and benign prostatic hyperplasia patients were tested. Arginine, homoserine, and proline were more abundant in the urine samples of cancer patients. However, sarcosine was not a definitive indicator of PCa when analyzed in urine samples collected either before or after prostate massage [35].

TMPRSS2-ERG

TMPRSS2-ERG is a urine-based post-DRE test. In 2005, the chromosomal rearrangement and fusion of the 5’ region of the androgen-regulated gene TMPRSS2 (transmembrane protease, serine 2) with ERG or ETV1 (erythroblastosis virus E26 transformation-specific transcription factor family members) was found to occur in PCa [36].

As a result of this rearrangement, the overexpression of TMPRSS2-ERG fusion products was found to be close to 100% specific for the presence of PCa in tissue-based studies, and TMPRSS2-ERG gene fusion products were identified in approximately 50% of PSA-screened PCa samples [36]. Subsequently, reverse transcription-polymerase chain reaction (RT-PCR)-based assays were developed to detect TMPRSS2-ERG mRNA in urine [37,38]. As reported, the test has a sensitivity of 37% and a specificity of 93% in predicting PCa, with a positive predictive value of 94% [39].

TMPRSS2-ERG was also associated with clinically significant PCa found through biopsy and prostatectomy [40]. Measuring TMPRSS2-ERG fusion transcripts is encouraging and may represent a new method of improving accuracy in detecting clinically significant PCa.

WHO NEEDS TO BE REBIOPSIED?

More than one million prostate biopsies are performed every year in the US. Furthermore, 40% of men who have had an initial prostate biopsy will undergo a repeat prostate biopsy, and more than 700,000 repeat biopsies are performed in the US annually [41]. Multiparametric magnetic resonance imaging (MRI) followed by ultrasound-MRI fusion biopsy is emerging as a promising approach to identify more clinically significant cancers [42-44]. The previously discussed molecular markers, including %fPSA, the PHI, the 4K score, and PCA3, can also be used to guide repeat biopsies. Eventually, the use of markers to help determine which biopsies are truly negative is imperative to avoid unnecessary biopsies.

Confirm MDx

Confirm MDx, developed by MDx Health Inc. (Irvine, CA, USA), is an epigenetic test that is used to predict the results of repeat prostate biopsy after an initial negative biopsy. A quantitative methylation-specific polymerase chain reaction assay is used to analyze methylation of the glutathione S-transferase p1 (GSTP1), adenomatous polyposis coli (APC), and Ras association domain family member 1 (RASSF1) genes from negative biopsy specimens. A negative result may help avoid unnecessary biopsies. The negative predictive value within 24 to 30 months of the initial biopsy was as high as 88%–90% [45,46].

WHO SHOULD BE OFFERED CURATIVE TREATMENT OR ACTIVE SURVEILLANCE?

After the diagnosis of PCa, the Gleason score is still the dominant factor in determining treatment. In the PSA era, most diagnosed cases of PCa (81%) involve organ-localized disease. However, given the heterogeneity of PCa in limited biopsy specimens, it is challenging to accurately predict the true risk level with biopsy specimens alone. Approximately 30% of low-risk PCa cases are upgraded or upstaged after radical prostatectomy [47-49]. Prospective studies have shown that curative management should be offered to patients with a good life expectancy and high-risk disease to maximize patients’ survival while minimizing side effects [50,51].

According to the 2016 NCCN guidelines, curative treatment is recommended for any man with PCa with a Gleason score of 4 or 5. However, the role of active surveillance in very low-risk or low-risk patients (Gleason 3+3, PSA<10, PSA density <0.15, <3 cores positive for PCa) can be challenging, as not all low-risk cases of PCa behave indolently. Identifying precise molecular markers to stratify potentially unappreciated high risk factors in seemingly low-risk cancers is imperative.

Oncotype DX

Oncotype DX introduced by Genomic Health Inc. (Redwood City, CA, USA), is a quantitative RT-PCR based test using small (1 mm) fixed paraffin-embedded tissue samples obtained by needle biopsy. This genomics-based test measures 12 cancer-related genes that represent 4 different biological pathways (androgen pathway, cellular organization, proliferation, and stromal response) and 5 reference genes, which are algorithmically combined to calculate the genomic prostate score (GPS) [52].

Together with the NCCN risk criteria, the GPS improves the risk discrimination of PCa by separating patients into very low, low, and modified-intermediate risk groups in order to help clinicians select appropriate candidates for active surveillance versus curative treatment [53,54].

Prolaris

The Prolaris test is a molecular test developed by Myriad Genetics Inc. (Salt Lake City, UT, USA). The test measures 31 cell cycle progression genes and 15 housekeeping genes selected for their correlation with PCa proliferation [55-58]. The test is performed in formalin-fixed paraffin-embedded tissue obtained by biopsy or prostatectomy. The results of the test are reported as 10-year PCa mortality, and stratify PCa patients into different risk categories. It is used to predict the level of risk in pretreatment PCa patients in order to choose between active surveillance versus curative treatment. It also may have a role in postprostatectomy patients with adverse pathological features (APFs) to guide the use of adjuvant treatment.

Decipher PCa Test

Decipher (GenomeDx Biosciences, San Diego, CA, USA) uses the expression of 22 selected RNA markers to predict cancer control outcomes in postradical prostatectomy tissue. In addition, several studies have recently shown that Decipher can be used in prostate biopsy specimens to predict the risk of cancer progression events, such as metastasis at 10 years, pelvic lymph node invasion, and APFs in the pretreatment setting.

Although biopsies yield a limited amount of tissue, the transcriptomic features detected in biopsy and prostatectomy tissues are highly correlated (r=0.96) [59]. The concordance between Decipher scores in prostatectomy tissue and biopsy tissue was reported to be as high as 86% in a recent study. This high concordance may allow the Decipher score in biopsy tissue to accurately predict pelvic lymph node invasion (0.78) and seminal vesical invasion (0.7) in prostatectomy tissues [60]. Biopsy-based Decipher scores have also been reported as an independent predictor of adverse pathology at surgery (c-index, 0.71) and clinical metastasis after surgery (c-index, 0.80) at 10 years, compared with 0.75 by NCCN risk stratification [61].

Based on the above studies, biopsy-based Decipher scores may represent another genomic tool to stratify biopsy-proven PCa patients to inform the choice of active surveillance versus curative treatment. It may also improve operative planning by indicating patients who require pelvic lymph node dissection or neoadjuvant therapy.

ProMark

ProMark (Metamark Genetics Inc., Waltham, MA, USA) is a proteomic PCa prognostic test using biopsy tissues [62]. Eight protein biomarkers (CUL2, DERL1, FUS, HSPA9, PDSS2, pS6, SMAD4, and YBX1) are measured with a fully automated immunofluorescent imaging platform. The 8 biomarkers represent PCa aggressiveness and lethality and were selected from 160 candidate proteomic markers using a quantitative proteomic approach [63]. This test aims to stratify the aggressiveness of PCa in Gleason 3+3 and 3+4 biopsy specimens to guide treatment choices.

In an initial study [62], the risk score for favorable and unfavorable pathology was defined in a sample of 381 patient biopsies with matched prostatectomy pathology. At a risk score ≤0.33, the predictive values for favorable pathology in very low-risk and low-risk NCCN groups and low-risk D’Amico groups were 95%, 81.5%, and 87.2%, respectively, higher than for these current risk classification groups themselves (80.3%, 63.8%, and 70.6%, respectively). At a risk score >0.8, the predictive value for unfavorable pathology was 76.9% across all risk groups.

The ProMark assay may provide better individualized and independent prognostic information than current clinicopathology-based risk stratification systems, improving the precision of clinical decision-making following prostate biopsy in patients with Gleason score 3+3 or 3+4 disease.

WHO NEEDS ADJUVANT THERAPY AFTER RADICAL PROSTATECTOMY?

Radical prostatectomy is an effective curative treatment for intermediate and high-risk PCa in patients with a life expectancy of more than 10 years [50]. However, some patients have APFs, such as positive margins, extracapsular extension, and seminal vesicle or bladder neck involvement. Although biochemical recurrence rates are high for men with APFs at radical prostatectomy (up to 60% at 10 years following surgery), it remains unclear whether it is beneficial for all men to undergo adjuvant treatment immediately following surgery [64].

The natural history of metastasis was studied in 3,089 patients who underwent radical prostatectomy in the PSA era at Johns Hopkins [65]. Forty-three individuals (n=1,327) within the cohort had APFs at the time of radical prostatectomy. In the group with APFs, the metastasis rate was 7.5% at 10 years, while the cumulative incidence of metastasis was 6% at 10 years for all patients. Thus, treating all patients with AFPs after radical prostatectomy would result in overtreatment in more than 90% of patients.

Despite the available level I evidence favoring the treatment of APFs with adjuvant radiation therapy (aRT), the use of aRT in patients is still limited [66,67]. Surgeons are cautious to recommend aRT due to concerns of morbidity and overtreatment. Even without adjuvant treatment, almost 50% of patients with APFs do not develop biochemical recurrence [68-71].

Utilizing the genomic features of prostate tissue may offer more precise decision-making in response to the above concerns regarding postprostatectomy APFs.

Decipher

Decipher is a genomic test that serves as a prognostic marker to predict cancer control outcomes in postradical prostatectomy patients that has shown a very high discrimination in predicting clinical metastasis (0.75–0.83) and cancer-specific mortality (0.78) in external validation studies, outperforming all routinely available clinicopathologic characteristics [72-75]. In a large single institutional study with 260 patients, of whom 99 experienced metastasis, the Decipher score showed a strong correlation with the cumulative incidence of biochemical recurrence, metastasis, and PCa-specific mortality (P<0.01) [65].

Several reports have shown that Decipher test results had an important impact on decision-making in contemporary clinical practice. Among subspecialized urologists and radiation oncologists, the Decipher results changed aRT decisions in 30%–45% of cases, and observation was offered to nearly 80% of patients with a low Decipher score [76-79].

Although showing early promise, the long-term effects of Decipher-based testing need to be validated with prospective studies to incorporate the Decipher score into PCa therapy decision making.

Prolaris

The Prolaris test can be performed in post-prostatectomy specimens as a prognostic genomic marker. However, long-term guidance for adjuvant treatment requires future clinical trials.

WHO IS A GOOD CANDIDATE FOR SECONDARY HORMONAL MANIPULATION IN CASTRATION-RESISTANT PCa?

Androgen deprivation therapy is the standard treatment for metastatic PCa. However, metastatic PCa eventually becomes castration-resistant [80]. Six new therapies have shown benefits affecting survival in men with metastatic castration-resistant prostate cancer (mCRPC). These therapies include secondary androgen receptor (AR)-inhibiting therapies (abiraterone acetate [81] and enzalutamide [82]), taxane chemotherapies (docetaxel [83] and cabazitaxel [84]), immunotherapies (sipuleucel-T [85]), and bone-targeting radiopharmaceuticals (radium-223) [86]. Of these, the most widely used are secondary AR manipulation with abiraterone/enzalutamide and the chemotherapies. Currently, no molecular biomarkers have been identified to help guide optimal treatment choices in these patients.

Of the patients who receive abiraterone or enzalutamide for the treatment of mCRPC, 20%–40% have primary resistance; that is, no PSA response. Furthermore, the development of secondary resistance is common. Interestingly, the presence of AR splice variants (AR-Vs) may be a cause of AR therapy failure, as the expression of AR-Vs has been observed in xenograft-based models of resistance to abiraterone, and high AR-V levels were found in PCa cell lines resistant to enzalutamide [87,88].

However, although the presence of splicing variant AR-V7 has been implicated in primary resistance to abiraterone and enzalutamide [89], a recent study found that AR-V7 status in 36 men with mCRPC was not related to primary resistance to taxane chemotherapy [90].

These seminal findings regarding AR-Vs may represent a field of research with the promise of furthering precision medicine in selecting treatments for PCa and other cancers.

CONCLUSION

With better interpretation and understanding of genomics, proteomics, transcriptomics and metabolomics, precision medicine will be incorporated into PCa management to an increasing extent.

The contemporary molecular markers in prostate cancer management are summarized in Table 1.

Summary of the contemporary molecular markers in prostate cancer management

Notes

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

References

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7–30.
2. Moyer VA, ; U.S. Preventive Services Task Force. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2012;157:120–34.
3. Andriole GL, Crawford ED, Grubb RL 3rd, Buys SS, Chia D, Church TR, et al. Prostate cancer screening in the randomized Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial: mortality results after 13 years of follow-up. J Natl Cancer Inst 2012;104:125–32.
4. Lavallée LT, Binette A, Witiuk K, Cnossen S, Mallick R, Fergusson DA, et al. Reducing the harm of prostate cancer screening: repeated prostate-specific antigen testing. Mayo Clin Proc 2016;91:17–22.
5. Lilja H, Christensson A, Dahlén U, Matikainen MT, Nilsson O, Pettersson K, et al. Prostate-specific antigen in serum occurs predominantly in complex with alpha 1-antichymotrypsin. Clin Chem 1991;37:1618–25.
6. Catalona WJ, Partin AW, Slawin KM, Brawer MK, Flanigan RC, Patel A, et al. Use of the percentage of free prostate-specific antigen to enhance differentiation of prostate cancer from benign prostatic disease: a prospective multicenter clinical trial. JAMA 1998;279:1542–7.
7. Roddam AW, Duffy MJ, Hamdy FC, Ward AM, Patnick J, Price CP, et al. Use of prostate-specific antigen (PSA) isoforms for the detection of prostate cancer in men with a PSA level of 2-10 ng/ml: systematic review and meta-analysis. Eur Urol 2005;48:386–99.
8. Lee R, Localio AR, Armstrong K, Malkowicz SB, Schwartz JS, ; Free PSA Study Group. A meta-analysis of the performance characteristics of the free prostate-specific antigen test. Urology 2006;67:762–8.
9. Catalona WJ, Southwick PC, Slawin KM, Partin AW, Brawer MK, Flanigan RC, et al. Comparison of percent free PSA, PSA density, and age-specific PSA cutoffs for prostate cancer detection and staging. Urology 2000;56:255–60.
10. Partin AW, Brawer MK, Subong EN, Kelley CA, Cox JL, Bruzek DJ, et al. Prospective evaluation of percent free-PSA and complexed-PSA for early detection of prostate cancer. Prostate Cancer Prostatic Dis 1998;1:197–203.
11. Catalona WJ, Partin AW, Sanda MG, Wei JT, Klee GG, Bangma CH, et al. A multicenter study of [-2]pro-prostate specific antigen combined with prostate specific antigen and free prostate specific antigen for prostate cancer detection in the 2.0 to 10.0 ng/ml prostate specific antigen range. J Urol 2011;185:1650–5.
12. Sokoll LJ, Wang Y, Feng Z, Kagan J, Partin AW, Sanda MG, et al. [-2]proenzyme prostate specific antigen for prostate cancer detection: a national cancer institute early detection research network validation study. J Urol 2008;180:539–43.
13. Stephan C, Jung K, Semjonow A, Schulze-Forster K, Cammann H, Hu X, et al. Comparative assessment of urinary prostate cancer antigen 3 and TMPRSS2:ERG gene fusion with the serum [-2]proprostate-specific antigen-based prostate health index for detection of prostate cancer. Clin Chem 2013;59:280–8.
14. 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;51:729–39.
15. Bruzzese D, Mazzarella C, Ferro M, Perdonà S, Chiodini P, Perruolo G, et al. Prostate health index vs percent free prostate-specific antigen for prostate cancer detection in men with “gray” prostate-specific antigen levels at first biopsy: systematic review and meta-analysis. Transl Res 2014;164:444–51.
16. 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;115:537–45.
17. Lazzeri M, Haese A, de la Taille A, Palou Redorta J, McNicholas T, Lughezzani G, et al. Serum isoform [-2]proPSA derivatives significantly improve prediction of prostate cancer at initial biopsy in a total PSA range of 2-10 ng/ml: a multicentric European study. Eur Urol 2013;63:986–94.
18. 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.
19. 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;20:255–61.
20. 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;68:464–70.
21. 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;Apr. 107(7)pii: djv095.
22. Stattin P, Vickers AJ, Sjoberg DD, Johansson R, Granfors T, Johansson M, et al. Improving the specificity of screening for lethal prostate cancer using prostate-specific antigen and a panel of kallikrein markers: a nested case-control study. Eur Urol 2015;68:207–13.
23. 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.
24. 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;62:2695–8.
25. 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;59:5975–9.
26. 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;52:1089–95.
27. Auprich M, Bjartell A, Chun FK, de la Taille A, Freedland SJ, Haese A, et al. Contemporary role of prostate cancer antigen 3 in the management of prostate cancer. Eur Urol 2011;60:1045–54.
28. Aubin SM, Reid J, Sarno MJ, Blase A, Aussie J, Rittenhouse H, et al. PCA3 molecular urine test for predicting repeat prostate biopsy outcome in populations at risk: validation in the placebo arm of the dutasteride REDUCE trial. J Urol 2010;184:1947–52.
29. Gittelman MC, Hertzman B, Bailen J, Williams T, Koziol I, Henderson RJ, et al. PCA3 molecular urine test as a predictor of repeat prostate biopsy outcome in men with previous negative biopsies: a prospective multicenter clinical study. J Urol 2013;190:64–9.
30. 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;32:4066–72.
31. 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;58:475–81.
32. Bradley LA, Palomaki GE, Gutman S, Samson D, Aronson N. Comparative effectiveness review: prostate cancer antigen 3 testing for the diagnosis and management of prostate cancer. J Urol 2013;190:389–98.
33. Sreekumar A, Poisson LM, Rajendiran TM, Khan AP, Cao Q, Yu J, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature 2009;457:910–4.
34. Khan AP, Rajendiran TM, Ateeq B, Asangani IA, Athanikar JN, Yocum AK, et al. The role of sarcosine metabolism in prostate cancer progression. Neoplasia 2013;15:491–501.
35. Sroka WD, Boughton BA, Reddy P, Roessner U, Słupski P, Jarzemski P, et al. Determination of amino acids in urine of patients with prostate cancer and benign prostate growth. Eur J Cancer Prev 2016;May. 24. [Epub]. http://doi.org/10.1097/CEJ.0000000000000248.
36. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 2005;310:644–8.
37. Laxman B, Tomlins SA, Mehra R, Morris DS, Wang L, Helgeson BE, et al. Noninvasive detection of TMPRSS2:ERG fusion transcripts in the urine of men with prostate cancer. Neoplasia 2006;8:885–8.
38. Laxman B, Morris DS, Yu J, Siddiqui J, Cao J, Mehra R, et al. A first-generation multiplex biomarker analysis of urine for the early detection of prostate cancer. Cancer Res 2008;68:645–9.
39. Hessels D, Smit FP, Verhaegh GW, Witjes JA, Cornel EB, Schalken JA. Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer. Clin Cancer Res 2007;13:5103–8.
40. 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.
41. Pinsky PF, Crawford ED, Kramer BS, Andriole GL, Gelmann EP, Grubb R, et al. Repeat prostate biopsy in the prostate, lung, colorectal and ovarian cancer screening trial. BJU Int 2007;99:775–9.
42. Rastinehad AR, Turkbey B, Salami SS, Yaskiv O, George AK, Fakhoury M, et al. Improving detection of clinically significant prostate cancer: magnetic resonance imaging/transrectal ultrasound fusion guided prostate biopsy. J Urol 2014;191:1749–54.
43. Mendhiratta N, Rosenkrantz AB, Meng X, Wysock JS, Fenstermaker M, Huang R, et al. Magnetic resonance imaging-ultrasound fusion targeted prostate biopsy in a consecutive cohort of men with no previous biopsy: reduction of over detection through improved risk stratification. J Urol 2015;194:1601–6.
44. Meng X, Rosenkrantz AB, Mendhiratta N, Fenstermaker M, Huang R, Wysock JS, et al. Relationship between prebiopsy multiparametric magnetic resonance imaging (MRI), biopsy indication, and MRI-ultrasound fusion-targeted prostate biopsy outcomes. Eur Urol 2016;69:512–7.
45. Stewart GD, Van Neste L, Delvenne P, Delrée P, Delga A, McNeill SA, et al. Clinical utility of an epigenetic assay to detect occult prostate cancer in histopathologically negative biopsies: results of the MATLOC study. J Urol 2013;189:1110–6.
46. Partin AW, Van Neste L, Klein EA, Marks LS, Gee JR, Troyer DA, et al. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol 2014;192:1081–7.
47. Truong M, Slezak JA, Lin CP, Iremashvili V, Sado M, Razmaria AA, et al. Development and multi-institutional validation of an upgrading risk tool for Gleason 6 prostate cancer. Cancer 2013;119:3992–4002.
48. Epstein JI, Feng Z, Trock BJ, Pierorazio PM. Upgrading and down-grading of prostate cancer from biopsy to radical prostatectomy: incidence and predictive factors using the modified Gleason grading system and factoring in tertiary grades. Eur Urol 2012;61:1019–24.
49. Pinthus JH, Witkos M, Fleshner NE, Sweet J, Evans A, Jewett MA, et al. Prostate cancers scored as Gleason 6 on prostate biopsy are frequently Gleason 7 tumors at radical prostatectomy: implication on outcome. J Urol 2006;176:979–84.
50. Bill-Axelson A, Holmberg L, Garmo H, Rider JR, Taari K, Busch C, et al. Radical prostatectomy or watchful waiting in early prostate cancer. N Engl J Med 2014;370:932–42.
51. Bolla M, Gonzalez D, Warde P, Dubois JB, Mirimanoff RO, Storme G, et al. Improved survival in patients with locally advanced prostate cancer treated with radiotherapy and goserelin. N Engl J Med 1997;337:295–300.
52. Knezevic D, Goddard AD, Natraj N, Cherbavaz DB, Clark-Langone KM, Snable J, et al. Analytical validation of the Oncotype DX prostate cancer assay - a clinical RT-PCR assay optimized for prostate needle biopsies. BMC Genomics 2013;14:690.
53. Klein EA, Cooperberg MR, Magi-Galluzzi C, Simko JP, Falzarano SM, Maddala T, et al. A 17-gene assay to predict prostate cancer aggressiveness in the context of Gleason grade heterogeneity, tumor multifocality, and biopsy undersampling. Eur Urol 2014;66:550–60.
54. Cullen J, Rosner IL, Brand TC, Zhang N, Tsiatis AC, Moncur J, et al. A Biopsy-based 17-gene genomic prostate score predicts recurrence after radical prostatectomy and adverse surgical pathology in a racially diverse population of men with clinically low- and intermediate-risk prostate cancer. Eur Urol 2015;68:123–31.
55. Cuzick J, Berney DM, Fisher G, Mesher D, Møller H, Reid JE, et al. Prognostic value of a cell cycle progression signature for prostate cancer death in a conservatively managed needle biopsy cohort. Br J Cancer 2012;106:1095–9.
56. Freedland SJ, Gerber L, Reid J, Welbourn W, Tikishvili E, Park J, et al. Prognostic utility of cell cycle progression score in men with prostate cancer after primary external beam radiation therapy. Int J Radiat Oncol Biol Phys 2013;86:848–53.
57. Cooperberg MR, Simko JP, Cowan JE, Reid JE, Djalilvand A, Bhatnagar S, et al. Validation of a cell-cycle progression gene panel to improve risk stratification in a contemporary prostatectomy cohort. J Clin Oncol 2013;31:1428–34.
58. Cuzick J, Stone S, Fisher G, Yang ZH, North BV, Berney DM, et al. Validation of an RNA cell cycle progression score for predicting death from prostate cancer in a conservatively managed needle biopsy cohort. Br J Cancer 2015;113:382–9.
59. Knudsen BS, Kim HL, Erho N, Shin H, Alshalalfa M, Lam LL, et al. Application of a clinical whole-transcriptome assay for staging and prognosis of prostate cancer diagnosed in needle core biopsy specimens. J Mol Diagn 2016;18:395–406.
60. Lee HJ, Yousefi K, Haddad Z, Abdollah F, Lam LL, Shin H, et al. Evaluation of a genomic classifier in radical prostatectomy patients with lymph node metastasis. Res Rep Urol 2016;8:77–84.
61. Klein EA, Haddad Z, Yousefi K, Lam LL, Wang Q, Choeurng V, et al. Decipher genomic classifier measured on prostate biopsy predicts metastasis risk. Urology 2016;90:148–52.
62. Blume-Jensen P, Berman DM, Rimm DL, Shipitsin M, Putzi M, Nifong TP, et al. Development and clinical validation of an in situ biopsy-based multimarker assay for risk stratification in prostate cancer. Clin Cancer Res 2015;21:2591–600.
63. Shipitsin M, Small C, Choudhury S, Giladi E, Friedlander S, Nardone J, et al. Identification of proteomic biomarkers predicting prostate cancer aggressiveness and lethality despite biopsy-sampling error. Br J Cancer 2014;111:1201–12.
64. Trock BJ. Adjuvant radiation following radical prostatectomy: what are the known unknowns? Eur Urol 2014;66:251–2.
65. Ross AE, Yousefi K, Davicioni E, Ghadessi M, Johnson MH, Sundi D, et al. Utility of risk models in decision making after radical prostatectomy: lessons from a natural history cohort of intermediate-and high-risk men. Eur Urol 2016;69:496–504.
66. Maurice MJ, Zhu H, Abouassaly R. Low use of immediate and delayed postoperative radiation for prostate cancer with adverse pathological features. J Urol 2015;194:972–6.
67. Kalbasi A, Swisher-McClure S, Mitra N, Sunderland R, Smaldone MC, Uzzo RG, et al. Low rates of adjuvant radiation in patients with nonmetastatic prostate cancer with high-risk pathologic features. Cancer 2014;120:3089–96.
68. Thompson IM, Tangen CM, Paradelo J, Lucia MS, Miller G, Troyer D, et al. Adjuvant radiotherapy for pathological T3N0M0 prostate cancer significantly reduces risk of metastases and improves survival: long-term follow-up of a randomized clinical trial. J Urol 2009;181:956–62.
69. Wiegel T, Bottke D, Steiner U, Siegmann A, Golz R, Störkel S, et al. Phase III postoperative adjuvant radiotherapy after radical prostatectomy compared with radical prostatectomy alone in pT3 prostate cancer with postoperative undetectable prostate-specific antigen: ARO 96-02/AUO AP 09/95. J Clin Oncol 2009;27:2924–30.
70. Bolla M, van Poppel H, Tombal B, Vekemans K, Da Pozzo L, de Reijke TM, et al. Postoperative radiotherapy after radical prostatectomy for high-risk prostate cancer: long-term results of a randomised controlled trial (EORTC trial 22911). Lancet 2012;380:2018–27.
71. Abdollah F, Suardi N, Cozzarini C, Gallina A, Capitanio U, Bianchi M, et al. Selecting the optimal candidate for adjuvant radiotherapy after radical prostatectomy for prostate cancer: a long-term survival analysis. Eur Urol 2013;63:998–1008.
72. Erho N, Crisan A, Vergara IA, Mitra AP, Ghadessi M, Buerki C, et al. Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PLoS One 2013;8e66855.
73. Karnes RJ, Bergstralh EJ, Davicioni E, Ghadessi M, Buerki C, Mitra AP, et al. Validation of a genomic classifier that predicts metastasis following radical prostatectomy in an at risk patient population. J Urol 2013;190:2047–53.
74. Den RB, Feng FY, Showalter TN, Mishra MV, Trabulsi EJ, Lallas CD, et al. Genomic prostate cancer classifier predicts biochemical failure and metastases in patients after postoperative radiation therapy. Int J Radiat Oncol Biol Phys 2014;89:1038–46.
75. Klein EA, Yousefi K, Haddad Z, Choeurng V, Buerki C, Stephenson AJ, et al. A genomic classifier improves prediction of metastatic disease within 5 years after surgery in node-negative high-risk prostate cancer patients managed by radical prostatectomy without adjuvant therapy. Eur Urol 2015;67:778–86.
76. Badani K, Thompson DJ, Buerki C, Davicioni E, Garrison J, Ghadessi M, et al. Impact of a genomic classifier of metastatic risk on postoperative treatment recommendations for prostate cancer patients: a report from the DECIDE study group. Oncotarget 2013;4:600–9.
77. Michalopoulos SN, Kella N, Payne R, Yohannes P, Singh A, Hettinger C, et al. Influence of a genomic classifier on post-operative treatment decisions in high-risk prostate cancer patients: results from the PRO-ACT study. Curr Med Res Opin 2014;30:1547–56.
78. Badani KK, Thompson DJ, Brown G, Holmes D, Kella N, Albala D, et al. Effect of a genomic classifier test on clinical practice decisions for patients with high-risk prostate cancer after surgery. BJU Int 2015;115:419–29.
79. Nguyen PL, Shin H, Yousefi K, Thompson DJ, Hornberger J, Hyatt AS, et al. Impact of a genomic classifier of metastatic risk on postprostatectomy treatment recommendations by radiation oncologists and urologists. Urology 2015;86:35–40.
80. Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer 2001;1:34–45.
81. de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011;364:1995–2005.
82. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012;367:1187–97.
83. Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351:1502–12.
84. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376:1147–54.
85. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411–22.
86. Parker C, Nilsson S, Heinrich D, Helle SI, O’Sullivan JM, Fosså SD, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369:213–23.
87. Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KA, Dehm SM. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res 2013;73:483–9.
88. Mostaghel EA, Marck BT, Plymate SR, Vessella RL, Balk S, Matsumoto AM, et al. Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: induction of steroidogenesis and androgen receptor splice variants. Clin Cancer Res 2011;17:5913–25.
89. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 2014;371:1028–38.
90. Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Nakazawa M, et al. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol 2015;1:582–91.

Article information Continued

Table 1.

Summary of the contemporary molecular markers in prostate cancer management

Marker Description Sampling specimen Features
Who needs to be biopsied? PSA Protein biomarker used to screen prostate cancer Serum Lack of ideal sensitivity or specificity in prostate cancer early detection
%fPSA Unbound protein form of PSA Serum Limited improvement in prediction accuracy of PCa detection in men with PSA 2-10 ng/mL
PHI tPSA, fPSA, and [-2]proPSA incorporated into an equation Serum I ncreases the power to detect PCa, especially high risk (Gleason≥7) PCa
4K score 4 Kallikrein markers: total PSA, free PSA, intact PSA, and human kallikrein 2, incorporated with patients age, DRE prior biopsy results Serum B etter accuracy in the detection of clinically significant cancer (Gleason score≥7) and potential to reduce the number of unnecessary biopsies
PCA3 A noncoding, prostate specific RNA Urine It is not recommended for use in primary biopsy
If the PCA3 score cutoff is set at 60, the positive predict value can be at 80%
Prostar ix Measurement of metabolites in the urine: sarcosine, alanine, glycine and glutamate Urine Need future validation
TMPRSS2-ERG RT-PCR assay to detect TMPRSS2:ERG fusion transcripts Reported in 50% of PSA screened PCa Urine May represent a new method to increase accuracy to detect clinically significant PCa
Who should be rebiopsied? PSA Serum
%fPSA Serum
PHI Serum
4K score As described prior Serum As described prior
PCA3 Serum
Prostar ix Serum
TMPRSS2-ERG Serum
Confirm MDx A quantitative methylation specific polymerase chain reaction assay to analysis methylation of 3 genes from negative biopsy specimen Tissue High negative predictive values to reduce unnecessary repeat prostate biopsies
Who should be offered curative treatment or surveillance? Oncotype DX A genomic based tissue test that measures 12 cancer-related genes representing 4 different biological pathways Tissue Improves risk discrimination of PCa into very low, low and modified intermediate risk in order to help clinicians select appropriate candidates for active surveillance vs curative treatment
Prolaris Measures 31 cell cycle progression genes and 15 housekeeping genes selected by their correlation with PCa proliferation Tissue Reports as 10-year PCa mortality to predict grade of risk in p re treatment PCa patients to guide management plan
Decipher prostate test (biopsy based test) Uses the expression of 22 selected RNA markers to predict adverse pathology and metastasis after surgery Tissue A potential genomic tool to stratify biopsy positive PCa patients in treatment planning
ProMaik 8 Biomarkers representing PCa aggressiveness and lethality were selected from 160 candidate proteomic markers using a quantitative proteomic approach Tissue Aims to stratify aggressiveness of PCa in Gleason scores of 3+3 and 3+4 in biopsy specimen to guide treatment
Who needs adjuvant therapy after radical prostatectomy? Decipher prostate test As described prior Tissue Shows strong correlation with increased cumulative incidence of biochemical recurrence, metastasis, and PCa-specific mortality
Application is limited by a lack of prospective studies
Prolaris As described prior Tissue Prolaris test can be performed in post-prostatectomy specimen as prognostic genomic markers Long term guidance of adjuvant treatment still need future clinical trials to elucidate
Who is a good candidate for secondary hormonal manipulation in castration resistant PCa? AR-V7 Measurement of an AR-V7 Circulating cells Avoid treatment with abiraterone and enzalutaniide in AR-V7 positive CRPC patients.
Earlier chemotherapy would be beneficial

PSA, prostate-specific antigen; fPSA, free PSA; PHI, prostate health index; tPSA, total PSA; PCa, prostate cancer; DRE, digital rectal examination; RT-PCR, reverse transcription-polymerase chain reaction; AR-V7, androgen-receptor splice variant 7; CRPC, castration resistant prostate cancer.