1,126
Views
6
CrossRef citations to date
0
Altmetric
Reviews

Biomarkers and personalized risk stratification for patients with clinically localized prostate cancer

, , &

Abstract

Prostate cancer (PCa) is the most common neoplasia among men in developed countries and a leading cause of cancer-related morbidity and mortality. PCa is a very heterogeneous disease, both clinically and biologically. Currently, it is difficult to stratify patients into risk groups that entail different disease management. Therefore, a personalized view of this disease is mandatory, through the development of new and more accurate biomarkers that may help clinicians to stratify patients according to threat that PCa poses for each patient. Hence, this review focuses on recent developments of molecular and immunohistochemical biomarkers for PCa risk stratification that might enable a personalized approach to PCa patients. However, despite the increasing amount of available data, there is also an urgent need to translate the most promising biomarkers for clinical use through large multicenter validation trials. Ultimately, these will contribute for an improved clinical management of PCa patients.

Prostate cancer (PCa) is a major burden on human health worldwide. In 2014, this neoplasia is predicted to be the most incident cancer in men and the second leading cause of cancer-related death Citation[1]. PCa is well known for its heterogeneity, ranging from clinically indolent to extremely aggressive tumors Citation[2,3]. Such feature poses a challenge for clinicians concerning patient’s risk stratification Citation[4]. As PCa is mostly asymptomatic until extra-prostatic invasion occurs, there was an urge to develop and implement accurate screening methods on a population basis Citation[5]. Thus, serum prostate-specific antigen (PSA) testing and digital rectal examination have been commonly used in clinical practice to screen and assist clinicians on therapeutic decision Citation[5]. Consequently, international guidelines, regarding the application of these tests and precise actions to be taken depending on results, have been developed Citation[6,7]. Since the serum PSA test implementation in the 1980s, PCa incidence rates in the USA and elsewhere skyrocketed followed by a significant decrease Citation[8]. PSA is prostate-specific but not PCa-specific, thus leading to reduced accuracy in diagnosis of PCa, but also on the identification of clinically significant tumors. Ultimately, this lack of specificity has led not only to overdiagnosis and overtreatment of indolent tumors, on the one hand, but also to underdiagnosis and undertreatment of life-threatening PCa, on the other Citation[9]. Thus, an attentive look at PCa incidence and mortality rates worldwide clearly indicates that there is an urgent need for a more patient-directed approach for risk stratification. More important than to identify individuals with PCa is to determine how life threatening is that condition.

The current lack of optimal tools to accurately stratify PCa patients into risk groups might be overcome through the development of novel biomarkers. Biomarkers are, typically, molecules whose detection or quantitation provides additional insight on disease beyond clinical features observed by physicians Citation[10]. Proteins, metabolites, RNA transcripts, DNA and epigenetic alterations are all candidates to become disease biomarkers Citation[10]. Imaging techniques are under development but mostly for cancer localization and guidance of diagnostic procedures and are also limited due to their cost and availability Citation[10]. Moreover, biomarkers might fall into several categories according to their potential to assist in disease predisposition assessment, screening, diagnosis, prognostication, prediction of response to treatment, disease monitoring and pharmacogenomics Citation[10]. There are several established criteria that define an ideal biomarker: be harmless and preferably non-invasive, highly accurate (i.e., high specificity, sensitivity, positive and negative predictive values) for its intent and improve decision-making abilities together with clinicopathological data Citation[10]. Hence, it is extremely unlikely that a single molecule or alteration can meet all these features, and a panel of biomarkers might thus provide a superior and more reliable approach. In PCa, three major areas are left to be filled with reliable biomarkers: distinguish low PSA tumors from benign prostatic hyperplasia; discriminate indolent from aggressive disease and identify metastatic disease Citation[10–12].

Besides well-established technologies such as immunohistochemistry (IHC) and FISH analysis, recent developments have occurred on the characterization of epigenetic alterations in PCa as promising biomarkers. Due to a better comprehension and description of these alterations, data concerning their usefulness as biomarkers have been accumulating. Bearing in mind their stability, frequency, reversibility and accessibility in body fluids, epigenetic biomarkers might be considered at the top of the list for candidates to next-generation biomarkers that may answer most, if not all, the questions raised above Citation[13].

This review aims to compile and critically analyze the most relevant data on emerging biomarkers for PCa risk stratification, with a special emphasis on those related with epigenetic alterations. For that purpose, PubMed was searched for publications concerning PCa biomarkers, between 1998 and 2014. Reports were selected based on the detail of analysis and/or potential clinical applications and/or clinical translational studies . Biomarkers hereafter reported were selected based on substantial clinical translational studies or attempts to bring them to daily clinical routine.

Table 1. Best performing biomarkers for diagnosis and prognosis assessment of prostate cancer.

Immunohistochemical biomarkers

The ability of IHC to simultaneously evaluate protein expression and correlate with morphology was decisive to make it the most widely used ancillary tool in pathology, with a substantial impact in clinical practice. Therefore, development of new biomarkers based on IHC is a constantly evolving field of research, including for PCa Citation[14]. Notwithstanding the vast array of candidate IHC biomarkers, published to date, for PCa risk assessment, only Ki-67 has been extensively validated, displaying the potential to stratify patients according to disease aggressiveness Citation[14]. Ki-67 has been described as possessing prognostic value for PCa-related mortality in patients ascribed to watchful waiting cohorts Citation[15,16], biochemical relapse in PCa patients submitted to radical prostatectomy Citation[17,18] and for disease progression in patients undergoing radiation therapy Citation[19,20]. As Ki-67 is a marker of cell proliferation, it is not surprising that this biomarker carries independent prognostic value in most studies, adding important information to the Gleason score and serum PSA levels Citation[14]. However, differences in study design and cohort composition led to different results regarding Ki-67 usefulness as a prognostic biomarker for PCa. Unsolved issues relate with the method for assessing Ki-67 immunostain, including the minimum number of cells to be counted, how to count them and which cutoff value to be used for patient’s stratification. Therefore, additional studies, preferentially enrolling patients from multiple centers, are mandatory to determine the true relevance of Ki-67 in clinical practice.

Although a prognostic value has not been established for α-methylacyl-coenzyme A racemase (AMACR) expression in PCa, this enzyme is currently used as a diagnostic ancillary tool. Indeed, AMACR is frequently overexpressed in PCa Citation[21], displaying high sensitivity and specificity (>90%) when used as a diagnostic marker in tissue biopsy Citation[22]. AMACR, in conjunction with basal cell markers (e.g., P63) is reported to help avoiding unnecessary biopsies Citation[23]. Its value alone is, however, questionable because AMACR is significantly expressed also in PCa precursor lesions such as high-grade prostate intraepithelial neoplasia (HGPIN), prompting some authors to suggest AMACR as a marker for HGPIN and not PCa Citation[24]. It has not been elucidated whether AMACR expression levels might correlate with disease outcome or might identify HGPIN lesions more prone to progress to invasive adenocarcinoma.

Genetic biomarkers

Fusion genes

Over the last years, fusion genes have unexpectedly emerged as a common feature of PCa. Among these, the most common is TMPRSS2:ERG, which is PCa-specific, accounting for approximately 90% of all currently known fusion events Citation[10], and detected in nearly 50% of prostate adenocarcinomas Citation[25]. These characteristics suggest that TMPRSS2:ERG is an early, and probably founding, event in many PCa, a postulate that is also supported by the observation of this fusion gene in up to 25% of HGPIN lesions Citation[26,27].

In a cohort of PCa patients on watchful waiting, Demichelis et al. found that the presence of TMPRSS2:ERG fusion gene was associated with Gleason score >7 tumors, increased mortality rates due to PCa and progression to metastatic disease Citation[28]. Using real-time PCR, a different study found a positive correlation between fusion transcripts in urine after prostate massage and a high serum PSA, pathological stage and Gleason score Citation[29]. Despite its apparent prognostic utility, these results have been challenged by other researchers, which have not found any correlation between fusion genes and PCa aggressiveness Citation[30,31]. This is most likely due to PCa heterogeneity and differences in patients’ cohorts Citation[11]. Nevertheless, this lack of consistency did not discourage researchers from evaluating its detection in urine samples. A study found that TMPRSS2:ERG displayed 93% specificity and 94% positive predictive value in post-digital rectal examination urine samples, for prediction of PCa upon prostate biopsy Citation[32]. However, this fusion gene was present in only 50% of the cases, thus compromising sensitivity. Therefore, to augment the sensitivity of the assay, assessment of prostate cancer antigen 3 (PCA3) mRNA was added to the test (normalized to urine PSA mRNA), resulting in enhanced utility over PSA serum alone, both for PCa diagnosis and prediction of disease aggressiveness Citation[33]. Nonetheless, this test is useless when PSA mRNA is low or untraceable in urine Citation[11].

As PCa often display multiple tumor foci within the same gland, the distribution of neoplastic cells harboring the TMPRSS2:ERG fusion gene might be heterogeneous Citation[34]. This might limit its usefulness as a single biomarker and a combined approach (e.g., with PCA3) seems to be more efficient. Notwithstanding these constraints, published data on TMPRSS2:ERG suggest a possible role as PCa biomarker both for diagnosis and risk stratification.

Chromosome 8q amplification

A few studies have provided whole-genome information on PCa from material of prostate biopsy and that has been shown to be largely consistent with data derived from radical prostatectomy specimens Citation[35]. Among chromosome-level alterations, relative 8q gain has been proposed as a biomarker of aggressive disease, being detected in 12.8–54% of patients Citation[36]. A comparative genomic hybridization study, using a large series of prospectively collected prostate biopsies, found that patients with relative gains of 8q had worse prognosis Citation[37]. Moreover, when patients were stratified according to tumor grade or clinical stage, relative gains of 8q were able to discriminate those with poorer outcome Citation[37]. These results are in accordance with previous published studies that used FISH analysis to determine 8q status. In those studies, 8q gain was associated with disease-related mortality after an average 35 months of follow-up Citation[38] and also with lower disease-specific survival at 10 years of follow-up Citation[36]. The fact that 8q status might help to predict outcome in prostate biopsy material, that is, before any therapeutic option has been decided, is of key importance for its potential use in patients’ management and risk stratification.

Whole-transcriptome markers

The recent development of genome-wide characterization techniques prompted researchers to apply this knowledge to obtain specific transcript profiles of PCa patients that might correlate with disease aggressiveness and patient outcome. Several studies have been conducted to achieve this goal and all have identified new candidate biomarkers that could better stratify patients Citation[39–41]. There is, however, a lack of consistency in the data generated by those studies, which probably derives from differences in patient’s characteristics, sampling procedures, methodologies and data analysis Citation[14]. Therefore, more robust results and independent validation studies in larger cohorts are needed to ascertain whether a specific prognostic signature for PCa is likely to become a clinically useful tool.

Epigenetic biomarkers

Non-coding RNAs

PCA3

PCA3 has become the most prominent biomarker for PCa since the emergence of serum PSA in the 1990s Citation[10]. PCA3 is a long non-coding RNA that has been found to be highly expressed in 95% of prostate adenocarcinomas but only slightly expressed in normal prostate tissues, which has been estimated as a 66-fold upregulation in PCa compared with normal adjacent tissue Citation[11,42]. Importantly, PCA3 expression is independent of tumor size Citation[43]. These characteristics created great expectations about its usefulness as a biomarker, not only in tissue but also in urine samples, providing a non-invasive test for PCa Citation[44]. The most prominent role for PCA3 has been the evaluation of the chance for a man to have a positive repeat prostate biopsy Citation[45]. Consequently, the US FDA recently approved urine PCA3 expression levels as a diagnostic test for men suspected to have PCa but with a negative prostate biopsy, which still might be at risk and require a second intervention, that is, re-biopsy Citation[11].

Besides this diagnostic utility, PCA3 has been also proposed as a prognostic tool since it might be able to forecast outcome after a first biopsy and to be correlated with PCa aggressiveness Citation[46,47]. Moreover, a correlation between PCA3 score, radical prostatectomy outcome and positive margins risk on prostatectomy specimens has been reported, as well Citation[48]. However, these data are still controversial because several studies neither found any correlation between PCA3 levels and clinicopathological data nor any prognostic value for this non-coding RNA Citation[25]. Nevertheless, PCA3 might be a good candidate biomarker both for distinguishing low PSA tumors from benign prostate hyperplasia and, eventually, to stratify patients according to their risk of progressive disease before and after radical prostatectomy, in a non-invasive manner.

miRNAs

miRNAs are small non-coding RNAs, of approximately 22 nucleotides in length, involved in gene expression regulation. They regulate several key cellular pathways and have been found to be deregulated in numerous diseases, including cancer Citation[49]. Development of more accurate technologies to evaluate miRNAs’ presence and expression levels led to enhanced interest in translating current knowledge about miRNAs into clinically useful cancer biomarkers. Recent research has focused on the identification of specific miRNAs signatures in PCa and on how they might correlate with clinicopathological variables, such as Gleason score, stage and metastatic spread. Assessment of miRNAs is also feasible in body fluids, such as urine and plasma/serum, which makes them potentially useful for non-invasive evaluation and stratification of PCa patients Citation[50]. Additionally, these molecules also show remarkable levels of stability in different clinical samples, as their small size and distinctive biochemical structure grant them resistance to RNase-mediated degradation Citation[51].

In a study using serum and plasma samples from PCa patients, as well as normal prostate tissues, miR-141 expression was found to be increased in metastatic PCa compared with localized disease. MiR-375 expression levels were also associated with higher Gleason score and positive lymph node status Citation[52]. Moreover, Schaefer et al. found that higher miR-96 expression was related to earlier biochemical relapse in PCa, using a training set/testing set methodology Citation[53]. In addition, miR-100, miR-145 and miR-191 were reported to bear prognostic value in PCa patients following radical prostatectomy Citation[54] and miR-221 downregulation has been associated with cancer progression in a cohort of 92 PCa patients Citation[55].

Furthermore, elevated blood levels of miR-141, miR-200b and miR-375 have been associated with castration-resistant metastatic PCa and high Gleason score, discriminating these high-risk patients from those with low-risk, clinically localized disease Citation[50]. Likewise, expression of miR-21 and miR-221 has been also associated with biochemical relapse Citation[56]. Expression of miR-21 in serum collected from PCa patients showed, also, an association with docetaxel resistance Citation[57]. Concerning miRNAs as biomarkers for PCa in urine, miR-107 and miR-574-3p were found to be upregulated in men with PCa compared with normal controls. Strikingly, a panel comprising both these miRNAs was even more accurate to identify PCa than PCA3 normalized to PSA Citation[50]. Despite being accurate biomarkers in serum and plasma samples, miR-141 and miR-375 were not able to distinguish between PCa patients and healthy donors in urine samples.

Taken together, these results suggest that miRNAs detection and quantitation in blood and urine might be useful for diagnosis and risk stratification of PCa patients. Nevertheless, it should be recalled that most of those studies were conducted in small cohorts, thus requiring validation in larger datasets, with standardization of protocols, biological material selection and patients’ characteristics Citation[58].

Histone modifications & chromatin remodeling

Histone marks

Histone modifications are very dynamic and, consequently, more difficult to scrutinize than the remaining epigenetic alterations. This class of alterations includes methylation, acetylation and phosphorylation of specific residues mostly on N-terminal tails of histones Citation[59,60], thus altering chromatin conformation and gene transcription Citation[59,60]. Currently, our knowledge about histone modifications and their role in gene expression regulation is still rather imperfect, impairing its use as potential cancer biomarkers.

Nevertheless, the first study suggesting histone modifications as potential cancer biomarkers was performed in PCa patients Citation[61]. It was found that global levels of H4K12Ac, H3K18Ac, H3K4Me2 and H4R3Me2, assessed by IHC, correlated with PCa stage, and that H3K18Ac and H3K4Me2 levels were associated with disease recurrence in low-grade PCa Citation[61]. Subsequently, global levels of these two marks were found to be independent predictors of PCa progression, irrespective of tumor grade Citation[62].

Another histone mark recognized as altered in PCa is H3K27Me3, which is overrepresented in metastatic tumors compared with non-malignant prostate tissue, using IHC analysis Citation[63]. This finding indicates a potential role for H3K27Me3 overexpression as a prognostic biomarker in PCa patients, which might be able to stratify patients according to their risk of developing PCa. Intriguingly, the only study that assessed H3K27Me3 levels in plasma samples of PCa patients, using ELISA, found a decrease in those levels in metastatic disease compared with localized disease Citation[64]. These controversial results emphasize the difficulties of using histone modification marks as potential PCa biomarkers. Therefore, more accurate techniques to evaluate histone marks in body fluids, IHC protocol standardization and testing in large cohorts of PCa are mandatory to effectively determine the role of histone marks as PCa biomarkers.

Histone modifiers

Enhancer of zeste homolog 2

The enhancer of zeste homolog 2 (EZH2) gene encodes for a polycomb group protein that is directly involved in transcription regulation, usually promoting gene silencing Citation[65,66]. EZH2 has been found overexpressed in several tumors, including PCa, in which it correlates with increased levels of H3K27Me3 Citation[12]. Because EZH2 has been pointed out as a mediator of E-cadherin silencing in cancer cells, it might constitute a potential biomarker for disease progression Citation[67]. Increased expression of both EZH2 mRNA and protein has been found in PCa tissues compared with benign prostate hyperplasia and HGPIN Citation[68]. Moreover, significantly higher EZH2 expression was observed in higher Gleason score tumors, correlating also with TNM stage and disease progression Citation[68]. In a different study, EZH2 performed better than serum PSA and Gleason score in the prediction of tumor progression Citation[65]. Furthermore, immunohistochemical assessment of EZH2 protein in PCa tissues was able to accurately predict tumor recurrence after therapy Citation[69]. Finally, higher EZH2 transcript levels have been associated with shorter time to PCa biochemical relapse, although it did not disclose independent prognostic value Citation[70].

Thus, EZH2 stands as a potentially useful biomarker of aggressive PCa, amenable to be assessed in prostate biopsies using IHC but validation in large datasets has not been accomplished, yet. Moreover, the potential usefulness of EZH2 assessment in body fluids as PCa biomarker is still largely unexplored.

SMYD3

SMYD3 is a histone methyltransferase responsible for establishing several histone marks in lysine residues, such as H3K4, H4K5 and H4K20 Citation[71,72]. This enzyme was found overexpressed in multiple cancers, including those of colorectum, breast, hepatocellular and prostate Citation[70,73,74].

Recently, it has been reported that higher SMYD3 transcript levels in PCa tissue samples were associated with shorter time to relapse in a series of 150 patients with clinically localized disease submitted to radical prostatectomy Citation[70]. Importantly, SMYD3 levels retained their statistical significance as independent prognostic biomarker in multivariate analysis, in addition to Gleason score and pathological stage Citation[70]. These results require further validation to ascertain SMYD3 potential for risk stratification in PCa. Furthermore, the development of a test that might detect and quantify SMYD3 expression in body fluids might provide a novel non-invasive approach for PCa detection and assessment of disease aggressiveness.

DNA methylation

DNA methylation is the most frequently studied epigenetic alteration in cancer, including PCa. Therefore, the majority of candidate epigenetic cancer biomarkers are generally based on aberrant DNA methylation. Indeed, this is a very common, stable and accessible alteration that might be detected in tissues and body fluids of PCa patients. Global genome hypomethylation appears to be correlated with tumor recurrence in PCa patients Citation[75]. However, because the assessment of global hypomethylation is rather cumbersome, most research efforts have focused in DNA, and especially gene promoter, hypermethylation, instead.

In PCa, several gene promoters have been reported to be hypermethylated, but the best established as a biomarker is the glutathione-S-transferase P1 (GSTP1) gene. Several reasons account for its prominent role as PCa biomarker: GSTP1 methylation is more PCa specific than serum PSA Citation[76]; GSTP1 methylation levels can discriminate PCa from benign prostate lesions Citation[77–79]; increased GSTP1 methylation levels correlate with higher Gleason score and tumor stage Citation[80], and, importantly, it might be detected in body fluids, including urine and serum Citation[81]. Remarkably, quantitative GSTP1 methylation assays display 93–100% specificity and 21.4–38.9% sensitivity for PCa detection in urine samples Citation[77,82,83], which might be increased 75% by prostatic massage prior to urine collection Citation[84].

Although the specificity of GSTP1 methylation is very high, even in urine and serum, sensitivity is suboptimal Citation[85]. This might be overcome by the addition of other gene promoters also known to be frequently and specifically hypermethylated in PCa, thus constituting gene panels Citation[85]. Indeed, the combination of GSTP1 with APC increased sensitivity to 98.3%, keeping 100% specificity in tissue samples Citation[80]. Other gene panels were shown to accurately detect PCa in urine (GSTP1/ARF/CDKN2A/MGMT and GSTP1/APC/RARB2/RASSF1A) and serum (GSTP1/PTGS2/RPRM/TIG1), with 86% sensitivity and 89% specificity in urine and 42–47% sensitivity and 92% specificity in serum Citation[86–88]. These results have led to the development of a diagnostic-level assay, the Prostate Cancer Methylation (ProCaM) assay, which consists of the quantitation of promoter methylation levels of GSTP1, APC and RARB2, in urine. The test has been validated in multicenter prospective study, in which urine samples of men with serum PSA levels of 2.0–10.0 ng/ml were collected and analyzed with the ProCaM assay and its performance was compared with existing methods based on clinical workup and serum PSA levels Citation[89]. Remarkably, the ProCaM assay was able to more accurately detect PCa than serum PSA or any of its derived parameters Citation[89]. Importantly, a positive result in the ProCaM assay associated with increased risk of finding high Gleason score (≥7) PCa in prostate biopsy Citation[89]. Recently, a multicenter trial validated the performance GSTP1, APC and RASSF1 promoter methylation as an independent predictor of PCa risk to guide decision for biopsy repetition Citation[90]. This assay demonstrated an 88% negative predictive value and may, thus, contribute to avoid unnecessary repeat prostate biopsies Citation[90].

Concerning risk stratification, quantitative promoter methylation of GSTP1, APC and MDR1 was able to discriminate clinically localized from advanced disease with 72.1% sensitivity and 67.8% specificity Citation[91]. On the other hand, patients with hypermethylated GSTP1 in serum had a 4.4-fold increased risk of biochemical relapse in one study Citation[92], although this has not been confirmed in a different cohort Citation[93]. However, in the latter study, a ninefold increased risk for biochemical recurrence was determined for PCa patients with CD44 and PTGS2 promoter hypermethylation Citation[93]. Furthermore, a multicenter study of PCa patients submitted to radical prostatectomy found that PITX2 promoter methylation levels were of prognostic significance following treatment, both uni- and multivariate analysis Citation[94]. Indeed, higher levels of PITX2 promoter methylation associated with a fourfold increased risk for biochemical relapse Citation[94]. Finally, in a series of prospectively collected prostate biopsies, high APC promoter methylation levels were independently associated with worse disease-free and disease-specific survival, with superior prognostic performance compared with serum PSA and Gleason score Citation[95].

Thus, multigene DNA methylation-based assays might not only provide useful tools for PCa detection, but also for identification of clinically significant disease, thus helping clinicians in the stratification of risk for developing PCa and in the assessment of PCa aggressiveness on an individual basis.

Expert commentary

The boom of PCa detection following the widespread use of serum PSA testing as a screening tool has come to a halt when large cooperative studies demonstrated no gain in survival or limited increase in overall survival, but at the cost of increased and unnecessary morbidity and mortality Citation[96–98]. This has led to the recommendation of the US Preventive Services Task Force against the disseminated utilization of serum PSA for PCa screening Citation[99]. Thus, the focus was changed from PCa detection by itself to the assessment of PCa aggressiveness once it was diagnosed, to avoid overtreatment.

Because currently available tools (mostly clinical and pathological) are rather imperfect to predict the threat that a given PCa poses to the patient’s life, the development and validation of new biomarkers is mandatory. Concerning detection, the new assays have to compare favorably with serum PSA, adding sensitivity and specificity, as well as maintaining a low cost and none or minimal invasiveness for sample collection. Thus, urine-based assays are quite appealing and, in this regard, the PCA3 assay may be considered an important step forward. However, its usefulness is restricted and its cost is a limiting factor for generalized use. In this setting, epigenetic biomarkers, especially those based on DNA methylation, seem to be most promising candidates for a new generation of urine-based PCa biomarkers. They were shown not only to accurately detect PCa in clinical samples, but they may also identify the clinically significant tumors.

Probably, prostate biopsy will remain the gold-standard for PCa diagnosis and prognostication. However, to more efficiently predict PCa aggressiveness, histopathological evaluation must be coupled with biomarkers. These might be based on IHC assays, which have become routine in most pathology laboratories, permitting its disseminated application. Ki-67 is undoubtedly in the frontline in this respect, but other biomarkers such as histone posttranslational modifications and modifiers will be probably required to fine-tune the assessment of prognosis. Prostate biopsies might also be used for 8q amplification analysis by FISH and DNA extracted from these tissue samples might also be used for quantitation of promoter methylation of genes that have been associated with worse disease survival and more aggressive PCa, such as APC. Indeed, cancer tissue samples are the most informative source for the search of alterations at proteomic, genomic, epigenomic or other -omic level and they might be maximized to yield the most relevant information for patient management. However, a major challenge lies in the widely recognized heterogeneity of PCa, which might prevent accurate risk stratification based on non-representative samples of tumorous tissue.

Five-year view

PCa will remain a major health concern, not only in western countries, but also in developing countries as improved life conditions will be followed by increased longevity and, necessarily, growing cancer incidence. Because this will occur mostly in countries of low economical resources, cost-effective means to detect and diagnose PCa will be an obvious challenge that must be met through improved technology. On the other hand, the growing concern about PCa overdiagnosis and overtreatment will lead inexorably to the development of more accurate biomarkers for PCa. New biomarkers are needed to avoid unnecessary biopsies and radical prostatectomies to distinguish benign from malignant lesions and to better discriminate localized from advanced disease. These new biomarkers might be used in tissue or body fluid samples and are expected to guide clinicians toward a more accurate and personalized treatment of PCa.

The recent development of wide-spectrum technologies might also be of great utility because it will permit genome-wide analysis of a single tumor in a fast and relatively inexpensive manner. This will also allow for an exponential increase of data that might be generated from discovery or validation studies, providing more robust data with personalized profiles being generated. Bearing in mind that a multigene or target approach will probably lead to a more accurate management of this disease, there is still the need to identify individual biomarkers that would ultimately be part of more complex and accurate assays. Likewise, a multicenter perspective for validation of potential biomarkers is crucial to consolidate new findings that eventually show up. The development of next-generation and third-generation sequencing is expected to change the landscape of the assessment of genomic information from cancer patients within the next 5 years.

Key issues

  • Prostate cancer (PCa), one of the most incident tumors in men worldwide, is highly heterogeneous ranging from clinically indolent to extremely aggressive.

  • There is a current lack of optimal biomarkers to accurately stratify PCa patients according to their risk of developing clinically significant disease.

  • Prostate-specific antigen is prostate specific but not PCa-specific leading to low accuracy in diagnosis of PCa and also in the identification of clinically significant tumors. This lack of specificity has led to overdiagnosis and overtreatment of indolent tumors.

  • There is an urgent need to develop non-invasive, accurate biomarkers for PCa that may: distinguish low prostate-specific antigen tumors from benign prostatic hyperplasia; discriminate indolent from aggressive disease and identify metastatic disease.

  • Recent developments in wide-spectrum genomic techniques allowed for the identification of both genetic and epigenetic aberrant patterns that may characterize different subsets of PCa.

  • Several genetic and epigenetic alterations have been identified as promising biomarkers for PCa both in tissue and body fluids.

  • Most candidate genetic and epigenetic biomarkers require further validation in large patient cohorts.

  • A broader characterization of the prostate cancer genome and epigenome may contribute to more personalized risk stratification and an optimized clinical approach.

Financial & competing interests disclosure

This work was supported by Federal funds through Programa Operacional Temático Factores de Competitividade (COMPETE) with co-participation from the European Community Fund (FEDER) and by national funds through Fundação para a Ciência e Tecnología (FCT) under the projects EXPL/BIM-ONC/0556/2012 (CJ) and Project Ciência 2008 (CJ). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin 2014;64(1):9-29
  • Hricak H, Scardino PT. Prostate cancer. Cambridge University Press; UK: 2009
  • Balistreri CR, Candore G, Lio D, Carruba G. Prostate cancer: from the pathophysiologic implications of some genetic risk factors to translation in personalized cancer treatments. Cancer Gene Ther 2014;21(1):2-11
  • Roobol MJ, Carlsson SV. Risk stratification in prostate cancer screening. Nat Rev Urol 2013;10(1):38-48
  • Kirby RS, Fitzpatrick JM, Irani J. Prostate cancer diagnosis in the new millennium: strengths and weaknesses of prostate-specific antigen and the discovery and clinical evaluation of prostate cancer gene 3 (PCA3). BJU Int 2009;103(4):441-5
  • Wolf AM, Wender RC, Etzioni RB, et al. American Cancer Society guideline for the early detection of prostate cancer: update 2010. CA Cancer J Clin 2010;60(2):70-98
  • Heidenreich A, Bellmunt J, Bolla M, et al. EAU guidelines on prostate cancer. Part 1: screening, diagnosis, and treatment of clinically localised disease. Eur Urol 2011;59(1):61-71
  • Barry MJ. Clinical practice. Prostate-specific-antigen testing for early diagnosis of prostate cancer. N Engl J Med 2001;344(18):1373-7
  • Duffy MJ. Prostate-specific antigen: does the current evidence support its use in prostate cancer screening? Ann Clin Biochem 2011;48:310-16. Available from: http://acb.rsmjournals.com/content/48/4/310.abstract
  • Prensner JR, Rubin MA, Wei JT, Chinnaiyan AM. Beyond PSA: the next generation of prostate cancer biomarkers. Sci Transl Med 2012;4(127):127rv3
  • Sardana G, Diamandis EP. Biomarkers for the diagnosis of new and recurrent prostate cancer. Biomarkers Med 2012;6(5):587-96
  • Dijkstra S, Hamid AR, Leyten GH, Schalken JA. Personalized management in low-risk prostate cancer: the role of biomarkers. Prostate Cancer 2012;2012:327104
  • Van Neste L, Herman JG, Otto G, et al. The epigenetic promise for prostate cancer diagnosis. Prostate 2012;72(11):1248-61
  • Kristiansen G. Diagnostic and prognostic molecular biomarkers for prostate cancer. Histopathology 2012;60(1):125-41
  • Borre M, Bentzen SM, Nerstrom B, Overgaard J. Tumor cell proliferation and survival in patients with prostate cancer followed expectantly. J Urol 1998;159(5):1609-14
  • Berney DM, Gopalan A, Kudahetti S, et al. Ki-67 and outcome in clinically localised prostate cancer: analysis of conservatively treated prostate cancer patients from the Trans-Atlantic Prostate Group study. Br J Cancer 2009;100(6):888-93
  • Gunia S, Albrecht K, Koch S, et al. Ki67 staining index and neuroendocrine differentiation aggravate adverse prognostic parameters in prostate cancer and are characterized by negligible inter-observer variability. World J Urol 2008;26(3):243-50
  • Miyake H, Muramaki M, Kurahashi T, et al. Expression of potential molecular markers in prostate cancer: correlation with clinicopathological outcomes in patients undergoing radical prostatectomy. Urol Oncol 2010;28(2):145-51
  • Pollack A, DeSilvio M, Khor LY, et al. Ki-67 staining is a strong predictor of distant metastasis and mortality for men with prostate cancer treated with radiotherapy plus androgen deprivation: radiation Therapy Oncology Group Trial 92-02. J Clin Oncol 2004;22(11):2133-40
  • Khor LY, Bae K, Paulus R, et al. MDM2 and Ki-67 predict for distant metastasis and mortality in men treated with radiotherapy and androgen deprivation for prostate cancer: RTOG 92-02. J Clin Oncol 2009;27(19):3177-84
  • Jiang Z, Fanger GR, Woda BA, et al. Expression of alpha-methylacyl-CoA racemase (P504s) in various malignant neoplasms and normal tissues: a study of 761 cases. Hum Pathol 2003;34(8):792-6
  • Rubin MA, Zhou M, Dhanasekaran SM, et al. Alpha-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer. JAMA 2002;287(13):1662-70
  • Carswell BM, Woda BA, Wang X, et al. Detection of prostate cancer by alpha-methylacyl CoA racemase (P504S) in needle biopsy specimens previously reported as negative for malignancy. Histopathology 2006;48(6):668-73
  • Gologan A, Bastacky S, McHale T, et al. Age-associated changes in alpha-methyl CoA racemase (AMACR) expression in nonneoplastic prostatic tissues. Am J Surg Pathol 2005;29(11):1435-41
  • Dijkstra S, Mulders PF, Schalken JA. Clinical use of novel urine and blood based prostate cancer biomarkers: a review. Clin Biochem 2014;47(10-11):889-96
  • Cerveira N, Ribeiro FR, Peixoto A, et al. TMPRSS2-ERG gene fusion causing ERG overexpression precedes chromosome copy number changes in prostate carcinomas and paired HGPIN lesions. Neoplasia 2006;8(10):826-32
  • Han B, Mehra R, Lonigro RJ, et al. Fluorescence in situ hybridization study shows association of PTEN deletion with ERG rearrangement during prostate cancer progression. Mod Pathol 2009;22(8):1083-93
  • Demichelis F, Fall K, Perner S, et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 2007;26(31):4596-9
  • Rostad K, Hellwinkel OJ, Haukaas SA, et al. TMPRSS2:ERG fusion transcripts in urine from prostate cancer patients correlate with a less favorable prognosis. APMIS 2009;117(8):575-82
  • Gopalan A, Leversha MA, Satagopan JM, et al. TMPRSS2-ERG gene fusion is not associated with outcome in patients treated by prostatectomy. Cancer Res 2009;69(4):1400-6
  • Fine SW, Gopalan A, Leversha MA, et al. TMPRSS2-ERG gene fusion is associated with low Gleason scores and not with high-grade morphological features. Mod Pathol 2010;23(10):1325-33
  • Hessels D, Smit FP, Verhaegh GW, et al. Detection of TMPRSS2-ERG fusion transcripts and prostate cancer antigen 3 in urinary sediments may improve diagnosis of prostate cancer. Clin Cancer Res 2007;13(17):5103-8
  • Tomlins SA, Aubin SM, Siddiqui J, et al. Urine TMPRSS2:ERG fusion transcript stratifies prostate cancer risk in men with elevated serum PSA. Sci Transl Med 2011;3(94):94ra72
  • Tomlins SA. Urine PCA3 and TMPRSS2:ERG using cancer-specific markers to detect cancer. Eur Urol 2014;65(3):543-5
  • Teixeira MR, Ribeiro FR, Eknaes M, et al. Genomic analysis of prostate carcinoma specimens obtained via ultrasound-guided needle biopsy may be of use in preoperative decision-making. Cancer 2004;101(8):1786-93
  • Barros-Silva JD, Ribeiro FR, Rodrigues A, et al. Relative 8q gain predicts disease-specific survival irrespective of the TMPRSS2-ERG fusion status in diagnostic biopsies of prostate cancer. Genes Chromosomes Cancer 2011;50(8):662-71
  • Ribeiro FR, Jeronimo C, Henrique R, et al. 8q gain is an independent predictor of poor survival in diagnostic needle biopsies from prostate cancer suspects. Clin Cancer Res 2006;12(13):3961-70
  • DeMarzo AM, Nelson WG, Isaacs WB, Epstein JI. Pathological and molecular aspects of prostate cancer. Lancet 2003;361(9361):955-64
  • Henshall SM, Afar DE, Hiller J, et al. Survival analysis of genome-wide gene expression profiles of prostate cancers identifies new prognostic targets of disease relapse. Cancer Res 2003;63(14):4196-203
  • Lapointe J, Li C, Higgins JP, et al. Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci USA 2004;101(3):811-16
  • Kristiansen G, Pilarsky C, Wissmann C, et al. Expression profiling of microdissected matched prostate cancer samples reveals CD166/MEMD and CD24 as new prognostic markers for patient survival. J Pathol 2005;205(3):359-76
  • Martin SK, Vaughan TB, Atkinson T, et al. Emerging biomarkers of prostate cancer (review). Oncol Rep 2012;28(2):409-17
  • Haese A, de la Taille A, van Poppel H, et al. Clinical utility of the PCA3 urine assay in European men scheduled for repeat biopsy. Eur Urol 2008;54(5):1081-8
  • Hessels D, Schalken JA. The use of PCA3 in the diagnosis of prostate cancer. Nat Rev Urol 2009;6(5):255-61
  • Hessels D, van Gils MP, van Hooij O, et al. Predictive value of PCA3 in urinary sediments in determining clinico-pathological characteristics of prostate cancer. Prostate 2010;70(1):10-16
  • Nakanishi H, Groskopf J, Fritsche HA, et al. PCA3 molecular urine assay correlates with prostate cancer tumor volume: implication in selecting candidates for active surveillance. J Urol 2008;179(5):1804-9. discussion 9-10
  • Whitman EJ, Groskopf J, Ali A, et al. PCA3 score before radical prostatectomy predicts extracapsular extension and tumor volume. J Urol 2008;180(5):1975-8. discussion 8-9
  • Durand X, Xylinas E, Radulescu C, et al. The value of urinary prostate cancer gene 3 (PCA3) scores in predicting pathological features at radical prostatectomy. BJU Int 2012;110(1):43-9
  • Chen PS, Su JL, Hung MC. Dysregulation of microRNAs in cancer. J Biomed Sci 2012;19:90
  • Bryant RJ, Pawlowski T, Catto JW, et al. Changes in circulating microRNA levels associated with prostate cancer. Br J Cancer 2012;106(4):768-74
  • Gordanpour A, Nam RK, Sugar L, Seth A. MicroRNAs in prostate cancer: from biomarkers to molecularly-based therapeutics. Prostate Cancer Prostatic Dis 2012;15(4):314-19
  • Kuner R, Brase JC, Sultmann H, Wuttig D. microRNA biomarkers in body fluids of prostate cancer patients. Methods 2013;59(1):132-7
  • Schaefer A, Jung M, Mollenkopf HJ, et al. Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma. Int J Cancer 2010;126(5):1166-76
  • Leite KR, Tomiyama A, Reis ST, et al. MicroRNA-100 expression is independently related to biochemical recurrence of prostate cancer. J Urol 2011;185(3):1118-22
  • Spahn M, Kneitz S, Scholz CJ, et al. Expression of microRNA-221 is progressively reduced in aggressive prostate cancer and metastasis and predicts clinical recurrence. Int J Cancer 2010;127(2):394-403
  • Selth LA, Townley S, Gillis JL, et al. Discovery of circulating microRNAs associated with human prostate cancer using a mouse model of disease. Int J Cancer 2012;131(3):652-61
  • Zhang F, Liu J, Lin B, et al. Increase in docetaxel-resistance of ovarian carcinoma-derived RMG-1 cells with enhanced expression of Lewis Y antigen. Int J Mol Sci 2011;12(11):7323-34
  • Sapre N, Selth LA. Circulating microRNAs as biomarkers of prostate cancer: the state of play. Prostate Cancer 2013;2013:539680
  • Leader JE, Wang C, Fu M, Pestell RG. Epigenetic regulation of nuclear steroid receptors. Biochem Pharmacol 2006;72(11):1589-96
  • Nightingale KP, O’Neill LP, Turner BM. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Curr Opin Genet Dev 2006;16(2):125-36
  • Seligson DB, Horvath S, Shi T, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature 2005;435(7046):1262-6
  • Bianco-Miotto T, Chiam K, Buchanan G, et al. Global levels of specific histone modifications and an epigenetic gene signature predict prostate cancer progression and development. Cancer Epidemiol Biomarkers Prev 2010;19(10):2611-22
  • Ellinger J, Kahl P, von der Gathen J, et al. Global histone H3K27 methylation levels are different in localized and metastatic prostate cancer. Cancer Invest 2012;30(2):92-7
  • Deligezer U, Yaman F, Darendeliler E, et al. Post-treatment circulating plasma BMP6 mRNA and H3K27 methylation levels discriminate metastatic prostate cancer from localized disease. Clin Chim Acta 2010;411(19-20):1452-6
  • Varambally S, Dhanasekaran SM, Zhou M, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 2002;419(6907):624-9
  • Chen H, Tu SW, Hsieh JT. Down-regulation of human DAB2IP gene expression mediated by polycomb Ezh2 complex and histone deacetylase in prostate cancer. J Biol Chem 2005;280(23):22437-44
  • Cao Q, Yu J, Dhanasekaran SM, et al. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene 2008;27(58):7274-84
  • Li J, Fan QH, Fan XS, et al. [EZH2 expression in human prostate cancer and its clinicopathologic significance]. Zhonghua Nan Ke Xue 2010;16(2):123-8
  • Rhodes DR, Sanda MG, Otte AP, et al. Multiplex biomarker approach for determining risk of prostate-specific antigen-defined recurrence of prostate cancer. J Natl Cancer Inst 2003;95(9):661-8
  • Vieira FQ, Costa-Pinheiro P, Ramalho-Carvalho J, et al. Deregulated expression of selected histone methylases and demethylases in prostate carcinoma. Endocr Relat Cancer 2014;21(1):51-61
  • Foreman KW, Brown M, Park F, et al. Structural and functional profiling of the human histone methyltransferase SMYD3. PLoS One 2011;6(7):e22290
  • Van Aller GS, Reynoird N, Barbash O, et al. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics 2012;7(4):340-3
  • Hamamoto R, Furukawa Y, Morita M, et al. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol 2004;6(8):731-40
  • Hamamoto R, Silva FP, Tsuge M, et al. Enhanced SMYD3 expression is essential for the growth of breast cancer cells. Cancer Sci 2006;97(2):113-18
  • Brothman AR, Swanson G, Maxwell TM, et al. Global hypomethylation is common in prostate cancer cells: a quantitative predictor for clinical outcome? Cancer Genet Cytogenet 2005;156(1):31-6
  • Wu T, Giovannucci E, Welge J, et al. Measurement of GSTP1 promoter methylation in body fluids may complement PSA screening: a meta-analysis. Br J Cancer 2011;105(1):65-73
  • Jeronimo C, Usadel H, Henrique R, et al. Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ-confined prostate adenocarcinoma. J Natl Cancer Inst 2001;93(22):1747-52
  • Harden SV, Sanderson H, Goodman SN, et al. Quantitative GSTP1 methylation and the detection of prostate adenocarcinoma in sextant biopsies. J Natl Cancer Inst 2003;95(21):1634-7
  • Nakayama M, Bennett CJ, Hicks JL, et al. Hypermethylation of the human glutathione S-transferase-pi gene (GSTP1) CpG island is present in a subset of proliferative inflammatory atrophy lesions but not in normal or hyperplastic epithelium of the prostate: a detailed study using laser-capture microdissection. Am J Pathol 2003;163(3):923-33
  • Jeronimo C, Henrique R, Hoque MO, et al. A quantitative promoter methylation profile of prostate cancer. Clin Cancer Res 2004;10(24):8472-8
  • Jeronimo C, Usadel H, Henrique R, et al. Quantitative GSTP1 hypermethylation in bodily fluids of patients with prostate cancer. Urology 2002;60(6):1131-5
  • Cairns P, Esteller M, Herman JG, et al. Molecular detection of prostate cancer in urine by GSTP1 hypermethylation. Clin Cancer Res 2001;7(9):2727-30
  • Gonzalgo ML, Pavlovich CP, Lee SM, Nelson WG. Prostate cancer detection by GSTP1 methylation analysis of postbiopsy urine specimens. Clin Cancer Res 2003;9(7):2673-7
  • Woodson K, O’Reilly KJ, Hanson JC, et al. The usefulness of the detection of GSTP1 methylation in urine as a biomarker in the diagnosis of prostate cancer. J Urol 2008;179(2):508-11. discussion 11-2
  • Henrique R, Jerónimo C. GSTP1 hypermethylation for prostate cancer detection. Prostate Cancer Screening. Springer; 2009. p. 279-88
  • Hoque MO, Topaloglu O, Begum S, et al. Quantitative methylation-specific polymerase chain reaction gene patterns in urine sediment distinguish prostate cancer patients from control subjects. J Clin Oncol 2005;23(27):6569-75
  • Roupret M, Hupertan V, Yates DR, et al. Molecular detection of localized prostate cancer using quantitative methylation-specific PCR on urinary cells obtained following prostate massage. Clin Cancer Res 2007;13(6):1720-5
  • Ellinger J, Haan K, Heukamp LC, et al. CpG island hypermethylation in cell-free serum DNA identifies patients with localized prostate cancer. Prostate 2008;68(1):42-9
  • Baden J, Adams S, Astacio T, et al. Predicting prostate biopsy result in men with prostate specific antigen 2.0 to 10.0 ng/ml using an investigational prostate cancer methylation assay. J Urol 2011;186(5):2101-6
  • Partin AW, Van Neste L, Klein EA, et al. Clinical validation of an epigenetic assay to predict negative histopathological results in repeat prostate biopsies. J Urol 2014. [Epub ahead of print]
  • Enokida H, Shiina H, Urakami S, et al. Multigene methylation analysis for detection and staging of prostate cancer. Clin Cancer Res 2005;11(18):6582-8
  • Bastian PJ, Palapattu GS, Lin X, et al. Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy. Clin Cancer Res 2005;11(11):4037-43
  • Woodson K, O’Reilly KJ, Ward DE, et al. CD44 and PTGS2 methylation are independent prognostic markers for biochemical recurrence among prostate cancer patients with clinically localized disease. Epigenetics 2006;1(4):183-6
  • Weiss G, Cottrell S, Distler J, et al. DNA methylation of the PITX2 gene promoter region is a strong independent prognostic marker of biochemical recurrence in patients with prostate cancer after radical prostatectomy. J Urol 2009;181(4):1678-85
  • Henrique R, Ribeiro FR, Fonseca D, et al. High promoter methylation levels of APC predict poor prognosis in sextant biopsies from prostate cancer patients. Clin Cancer Res 2007;13(20):6122-9
  • Andriole GL, Crawford ED, Grubb RL 3rd, et al. Mortality results from a randomized prostate-cancer screening trial. N Engl J Med 2009;360(13):1310-19
  • Schroder FH, Hugosson J, Roobol MJ, et al. Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 2009;360(13):1320-8
  • Wilt TJ, Brawer MK, Jones KM, et al. Radical prostatectomy versus observation for localized prostate cancer. N Engl J Med 2012;367(3):203-13
  • Moyer VA. Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med 2012;157(2):120-34

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.