Molecular Diagnosis of Prostate Cancer: Are We Up to Age?
Transcription
Molecular Diagnosis of Prostate Cancer: Are We Up to Age?
Molecular Diagnosis of Prostate Cancer: Are We Up to Age? Tapan Bhavsar, Peter McCue, and Ruth Birbe Prostate cancer (PCa), a highly heterogeneous disease, is the one of the leading cause of morbidity and mortality in the developed countries. Historically used biomarkers such as prostatic acid phosphatase (PAP), serum prostate-specific antigen (PSA), and its precursor have not stood the challenge of sensitivity and specificity. At present, there is need to re-evaluate the approach to diagnose and monitor PCa. To this end, molecular markers that can accurately identify men with PCa at an early stage, and those who would benefit from early therapeutic intervention, are the need of the hour. There has been unprecedented progress in the development of new PCa biomarkers through advancements in proteomics, tissue DNA and protein/RNA microarray, identification of microRNA, isolation of circulating tumor cells, and tumor immunohistochemistry. This review will examine the current status of prostate cancer biomarkers with emphasis on emerging biomarkers by evaluating their diagnostic and prognostic potentials. Semin Oncol 40:259-275 & 2013 Elsevier Inc. All rights reserved. P rostate cancer (PCa) is the one of the leading causes of morbidity and mortality in developed countries. In the United States and Europe, PCa claims around 30,350 and 85,200 every year.1,2 As many as 232,090 men in the United States and 237,800 men in Europe are newly diagnosed with PCa annually.3 PCa is a highly heterogeneous disease that includes genetic aberrations, local invasion of extracellular matrix, micrometastatic cell extravasations to circulation, and invasion of secondary organ tissues followed by resistance to hormonal modulation. Historically, human prostatic acid phosphatase (PAP) (or serum acid phosphatase [AP]), the first serum biomarker for PCa, lacked sufficient sensitivity as a reliable biomarker for response to systemic therapy or recurrence. Later, serum prostate-specific antigen (PSA) was used for screening men with an existing diagnosis of PCa4 and was regarded as an ideal marker for identifying recurring disease subsequent to treatment. However, PCa has been detected in about 15% of men with normal (upper limit, 4.0 ng/mL) or very low levels of total PSA, thereby making it difficult to reliably rule out the possibility Department of Pathology, Thomas Jefferson University, Philadelphia, PA. Conflicts of interest: none. Address correspondence to Ruth Birbe, MD, Department of Pathology, Thomas Jefferson University, 285 Main Bldg, 132 South 10th St, Philadelphia, PA 19107. E-mail: [email protected] 0270-9295/ - see front matter & 2013 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1053/j.seminoncol.2013.04.002 Seminars in Oncology, Vol 40, No 3, June 2013, pp 259-275 of PCa at any PSA level.5 ProPSA, the precursor form of PSA, served as an additional indicator in differentiating PCa from benign processes.6 Unfortunately neither of these markers has stood the challenge of sensitivity and specificity. At the present, there is need to re-evaluate the approach to diagnose and monitor PCa. New biomarkers are clearly needed. To this end, in lieu of analyzing the PCa tumor cells from biopsy specimens, PCa was studied indirectly by analyzing blood, urine, and prostatic ejaculates for variations in proteins with the hope of developing a panel of biomarkers that would provide more diagnostic information than any one modality. Although the subject of ongoing debate, molecular markers that can accurately identify men with PCa at an early stage, and those who would benefit from early therapeutic intervention, are the need of the hour. There has been unprecedented progress in the development of new PCa biomarkers through advancements in proteomics, tissue DNA and protein/RNA microarray, identification of microRNA (miRNA), isolation of circulating tumor cells, and tumor immunohistochemistry (IHC). The contribution of Human Genome Sequencing Project and National Cancer Institute–sponsored PCa Specialized Program of Research Excellence (SPORE) cancer centers in the United States has enriched this field. These biomarkers, after clinical validation, would be sine qua non to guide clinicians in defining prognosis and monitoring patients, as well as in the assessment of treatment options of PCa. The biomarkers can be broadly classified into protein markers, including those detected on tissue, 259 260 urine, and other body fluids, DNA- and RNA-based markers, and novel markers identified through proteomic studies. This review will examine the current status of PCa biomarkers with emphasis on emerging biomarkers by evaluating their diagnostic and prognostic potentials. MOLECULAR MARKERS ON TISSUE Ki-67/MIB-1 Ki-67, a cell-proliferation associated protein marker with an expression pattern under cell cycle regulation,7 had been described as a promising biomarkers of PCa. IHC staining for Ki-67 was shown to discriminate latent PCa revealed by the presence of a decreased proportion of actively proliferating cells from clinical cancers with poorer prognosis.8–10 However, several studies show that it does not seem to be an independent prognostic marker for PCa recurrence after radical prostatectomy, although it may have some clinical utility in a subset of patients.11–13 p53 Nuclear accumulation of p53, a marker of growth control and apoptosis, has been associated with advanced stages of PCa,14 metastases,15 and an overall worse prognosis.16–22 Studies conducted on localized PCa have reported frequencies of p53 alteration ranging from 4%–60% and as high as 94% in more advanced castrate-resistant PCa (CRPC) Interestingly, DNA sequencing analysis has suggested a lower rate of p53 mutations than indicated by overexpression in IHC analysis.23 More recently, it has been found that focal p53 expression by IHC in primary PCa is predictive of cancer recurrence after radical prostatectomy. However, altered p53 occurs in many malignancies and it is not a specific marker for PCa. Bcl-2 Overexpression of Bcl-2 protein by IHC has been commonly detected in advanced CRPC. Bcl-2 protein expression in primary PCa, along with p53, is a predictor of cancer recurrence after radical prostatectomy and radiotherapy.24–26 Furthermore, the combination of p53 and Bcl-2 protein expression is an independent predictor of recurrence after surgery in radical prostatectomies. Curiously, this combination is not predictive of postoperative recurrence when evaluated in the pretreatment needle biopsy. Alpha-Methylacyl CoA Racemase Alpha-methylacyl CoA racemase (AMACR) is an enzyme involved in fat metabolism and functions as a growth-promoter independent of androgens in PCa.27,28 IHC staining using monoclonal antibodies T. Bhavsar, P. McCue, and R. Birbe to AMACR is now a common practice to assist in the diagnosis of PCa; a multi-institutional study has shown sensitivity and specificity of 97% and 92%, respectively.29 The specificity of AMACR in the detection of PCa in biopsy specimens is regarded as an improvement over the serum PSA test,30 in as much as one study has shown 100% specificity and 97% sensitivity.31,32 On the flip side, AMACR has been identified in prostatic intraepithelial neoplasia (PIN) and occasionally in benign conditions. To err on the side of caution, a PCa diagnosis should not be based solely on a positive AMACR stain, especially when the luminal staining is weak and/or noncircumferential. To circumvent this hurdle, in clinical practice, AMACR is used in combination with PCa-negative basal cell stains (p63, 34bE12, and cytokeratins 5 and 6).33,34 AMACR mRNA and protein levels also have been shown to be overexpressed in PCa, making it a highly specific tissue biomarker.29,32,35 The detection of AMACR in prostatic secretions36 and urine37 has shown promise, although this enzyme is found in other malignancies such as renal cell carcinoma. Prostate-Specific Membrane Antigen Prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein predominantly expressed in the prostate epithelium. Significantly increased expression of PSMA is observed in PCa, particularly in poorly differentiated, metastatic, and CRPC.38 Multiple studies have suggested that PSMA correlates well with Gleason score and/or the stage of disease. It can be used to monitor post-treatment disease progression.39–44 PSMA-specific reverse transcriptase–polymerase chain reaction (RT-PCR) has shown promise as a sensitive biomarker in identifying micrometastases in lymph nodes that are not detectable by light microscopy.45 PSMA also functions in the radiologic imaging of PCa. The immunoscintigraphic test, ProstaScint (Cytogen, Princeton, NJ) localizes PCa cells by targeting PSMA with an indium 111–labeled monoclonal antibody,46 and has been used for imaging soft tissue sites of metastatic PCa for presurgical staging, especially in high-risk patients. It may be of utility in the evaluation of biochemical relapse after local therapy. Osteopontin Splicing Isoforms The diagnosis and prediction of prognosis in PCa has been augmented by a number of alternatively spliced genes such as osteopontin (OPN), kallikreins, spliced variant of fibroblast growth factor receptor (FGF-R2), and truncated form of vascular endothelial growth factor receptor flt-4 (VEGFR-3).47 OPN is a glycophosphoprotein that acts as an important autocrine and paracrine signal affecting the growth and behavior of PCa cells.48,49 Its 261 Molecular diagnosis of prostate cancer expression has been correlated with stage, grade, and early tumor progression in PCa.49,50 Osteopontin splicing isoforms (OPN-SI) have been shown to be strongly associated with PCa occurrence and tumor cell differentiation.51–55 The three isoforms, OPNa, OPNb, and OPNc splice variants, are all overexpressed in PCa in relation to benign prostatic hyperplasia (BPH). OPNc has been shown to be the most upregulated isoform in PCa, outperforming the remaining isoforms and serum PSA serum levels in PCa diagnostic accuracy. BMP-6 Bone morphogenetic proteins (BMP) are involved in new bone formation and other organ development. The expression of both BMP-6 mRNA and protein is upregulated in PCa, in as much as BMP-6 mRNA expression has been demonstrated in 95%– 100% of primary PCa with solid organ metastases, 18%–36% with organ-confined disease, and 85% of bone metastases.56,57 Taking a step forward, CpG demethylation of the BMP-6 promoter has been shown to be responsible for the high expression in primary and secondary sites of advanced PCa.58 Interestingly, co-expression of the proteins BMP-6, bone sialoprotein, and thymidine phosphorylase was observed in 90% of the radical prostatectomy specimens with bone metastases and only in 29% of men with curative treatment,59 making these proteins useful markers to identify at-risk patients for disease progression after treatment. PTEN PTEN is a tumor-suppressor gene that encodes for a protein that dephosphorylates phosphatidylinositol-3phosphate. The deletion of the PTEN gene has a frequency of about 40% in the western population, and a mere 7.6% in those of Chinese origin.60 Deletions of PTEN have been associated with Gleason scores 47, biochemical relapse, and nodal metastasis. Nuclear Factor-κB Nuclear factor-κB (NF-κB) activation has been shown to occur in the transition from a preneoplastic state to PCa. The overexpression of NF-κB has been shown to have a positive correlation with advanced PCa tumor stage and independently predicts a high risk of biochemical relapse of PCa.61 This is especially relevant in patients with positive surgical margins and lymph node metastasis. Those identified include somatic cytochrome c (CYCS), intestinal cell kinase (ICK), inhibitor of NF-κB kinase subunit beta (IKBKB), and glutamate decarboxylase 1 (GAD1).62,63 In a similar vein, CD10 has proved to be an important biomarker whose loss or decreased expression is thought to be an early (and frequent) event in human PCa.64 Syndecan-1 overexpression, as analyzed by tissue microarray, predicts early recurrence and is significantly associated with tumor-specific survival, high Gleason score, Ki-67, and Bcl-2 overexpression.65 A significant decrease in protein expression of annexins 1 and 7 in hormone-refractory PCa has been correlated with tumor progression.66 MOLECULAR MARKERS IN SERUM The clinical utility of serum markers for PCa diagnosis and disease progression is obvious. Much effort has been applied to identify efficacious candidates. Crisp-3/SGP28 Cysteine-rich secretory protein 3 (CRISP-3) is a specific granule protein of 28 kd (SGP28), purified from human neutrophils. The expression of CRISP-3/ SGP28 mRNA is increased 20- to 20,000-fold in Gleason grade 3 PCa. A quantitative enzyme-linked immunosorbent assay (ELISA)-based assay is under development to study the diagnostic potential of CRISP-3/SGP28 in plasma and body fluids.67 hK2 Using a quantitative RT-PCR, hK2 protein expression is found to be increased in high-grade PIN, PCa, and lymph node metastases. A highly sensitive hK2specific immunoassay shows promise in diagnostic applicability over free PSA ratio in the “gray zone” to distinguish PCa from BPH. Moreover, serum hK2 alone or in combination with total PSA and free PSA may improve the detection of extraprostatic or advanced disease.68–71 Osteoprotegerin Serum levels of osteoprotegerin (OPG) have been found to be significantly higher in patients with advanced PCa compared to patients at other stages of prostatic disease. An increased serum OPG concentration may be embraced as a marker of early relapse from androgen ablation therapy and as a monitor in patients with bone metastases.72,73 Novel Tissue Markers Chromogranin A The use of prostatic tissue microarrays has enriched the field with a varitey of new PCa markers. Chromogranin A (CGA or GRN-A), a member of the granin family of proteins, is an acidic protein 262 T. Bhavsar, P. McCue, and R. Birbe identified in all neuroendocrine cell types. CGA has clinical potential as a biomarker for early, progressive, and recurrent PCa.74 Several studies have suggested a role for CGA in the initial detection of PCa, particularly when used in combination with free-total PSA ratio.75 Conversely, not all PCa exhibit neuroendocrine differentiation, limiting the overall efficacy of chromogranin A in diagnosing and monitoring PCa patients. Vascular Endothelial Growth Factor Human Glandular Kallikrein 2 Other Serum Markers Human glandular kallikrein 2 is closely related to PSA and is expressed at high levels in prostate tissue.76,77 It has been used in various combinations with free and total PSA to statistically improve the sensitivity and specificity of identifying PCa.78,79 The methodology of DNA microarray emerges as an important innovation in the search of serum markers with the aim of selecting differentially expressed genes. Using the approach of antibody microarray, five proteins have been identified, including von Willebrand factor, immunoglobulin M, alpha-1 chymotrypsin, immunoglobulin G, and vilin, with significantly higher serum levels in PCa patients.95 In addition, serum levels of hK2/fPSA, Hepsin, AMACR, neuronspecific enolase, urokinase-type plasminogen activator system, β-catenin, and hK11 have been shown to be promising markers in the diagnosis of PCa. Transforming Growth Factor-β and Interleukin-6 Elevated serum levels of transforming growth factor-β (TGF-β1) and interleukin-6 (IL-6), via an increased p21 expression through Smad and STAT3 signaling, respectively, have been associated with poor prognosis. Additionally, elevated serum TGF-β1 levels have been associated with an increased risk of metastatic disease.80–82 Increased serum levels of IL-6, along with IL-8 and IL-11, have been demonstrated in patients with metastatic and CRPC.83-86 The measurement of preoperative levels of both TGF-β1 and IL-6 makes an effective contributor in improving the accuracy of predicting regional lymph node metastases, disease progression, and biochemical recurrence.82,87 Caveolin-1 Caveolin-1 (Cav-1) is an integral membrane protein and a main component of caveolae membranes in vivo. Abundant stromal Cav-1 immunostaining is present in almost all patients with BPH. A small subset of samples of primary PCa has been shown to have significantly decreased levels of stromal Cav-1.88 E-cadherin E-cadherin, a marker of cell adherence, has been studied extensively in prostatectomy specimens. In general, an inverse correlation with tumor grade and stage is found. Loss of E-cadherin expression is a promising marker of poor prognosis in localized PCa.89 EGFR Family, c-erbB-1/c-erbB-2 Both the epithelial growth factor receptor (EGFR) and the oncogene c-erbB2/HER-2 have been found to be overexpressed in PCa. There is an increasing level of expression from localized to metastatic disease.90 Serum levels of the ligand VEGF were found to be significantly higher in those PCa patients with metastatic disease.91 Increased plasma VEGF level is an independent prognostic factor in PCa.92,93 Moreover, it also has been demonstrated that the related microvessel density (MVD) count was significantly higher in PCa patients with metastatic disease when compared with those without it.94 MOLECULAR MARKERS IN URINE Urine biomarkers are promising for PCa detection as they are readily measurable and can be obtained noninvasively. TMPRSS2-ERG Gene Fusion Rearrangement Transmembrane protease serine 2 (TMPRSS2) is an androgen-regulated, type II transmembranebound serine protease overexpressed in PCa. It plays a role in tumor metastasis through activation of protease-activated receptor-2. Using Cancer Outlier Profile Analysis (COPA), an open source program that is designed to evaluate variance in a data using the median instead of the mean to maintain the peaks of outliers, it was found that the overexpression of two genes ERG and ETV1 was highly correlated with TMPRSS2. A fusion of TMPRSS2 (21q22.3) with the transcription factor genes ERG (21q22.2) and ETV1 (7p21.1) has been shown96,97 to be one of the earliest events in PCa. TMPRSS2-ERG is the most frequent oncogenic gene fusion rearrangement in PCa.87 It is detected in almost half of PCa patients and in about a quarter of patients with PIN.98 Several studies suggest that TMPRSS2:ERG fusion contributes to a more aggressive cancer phenotype that is associated with a higher tumor stage and PCaspecific death. Parenthetically, while the frequency of TMPRSS2-ERG rearrangement constitutes approximately 50% of the cases in Westerners, it is a mere 2.5% in the Chinese population. 263 Molecular diagnosis of prostate cancer The TMPRSS2-ERG fusion can be detected in the urine after digital rectal examination (DRE) with 93% specificity99 and 94% positive predictive value.100 The addition of TMPRSS2–ERG detection can increase the sensitivity of the urine PCA3 test (see below) from 62% to 73%99 and will increase the prediction of tumors with a higher Gleason score.101 The combination of TMPRSS2-ERG detection with PCA3 may become the preferred screening methodology for PCa.102,103 Recently, a multiplex urine based assay combining PCA3, TMPRSS2:ERG, annexin A3, sarcosine, and PSA was shown to maximize PCa detection compared with that of any single biomarker.104 The presence of PCA3 and TMPRSS2-ERG fusion has been detected in urine exosomes from PCa patients.105,106 PCA3/DD3 The prostate cancer antigen 3 (PCA3) gene encodes a prostate-specific mRNA that is overexpressed in PCa tissue.107,108 DD3 (differential display code 3)/PCA3 is the most PCa-specific gene (along with aforementioned TMPRSS2:ERG gene fusion) that has been described to date,109 showing overexpression in 495% of primary PCa specimens and in metastatic deposits. In biopsy specimens, PCA3/ DD3 expression has shown 72% sensitivity, 83% specificity, and 88% negative predictive value for the detection of PCa.110–112 A DD3/PCA3-based RT-PCR assay has been developed to quantify PCA3 mRNA levels in urine collected following DRE. This assay is advantageous as the specimen processing is relatively simple and uses clean catch urine. The fraction of specimens yielding sufficient RNA for analysis is 495% and the assay is readily adaptable to a clinical laboratory setting using available instrumentation.113 The assay also could be applicable in the detection of malignant prostate cells in blood or ejaculates. Following normalization to the total amount of prostate RNA, the resultant ratio, known as PCA3 score has shown 67%–69% sensitivity and 79%–83% specificity.110,113 This PCA3 score is independent of prostate volume, the number of prior biopsies, patient’s age, pharmacotherapy (including type I and II 5 alpha-reductase inhibitors), and inflammatory conditions. The PCA3 score is useful in detecting PCa in men with commonly observed alternative causes of increased PSA such as prostatitis and in patients with normal or low PSA. Along the same lines, in the detection of high-grade PIN, PCA3 score has been demonstrated to be 16% higher than in the tissue of men without this lesion.114 Moreover, the PCA3 scores are higher in patients with larger, more invasive tumors, thereby assisting in determining if patients would require aggressive treatment or active surveillance. In a semi-quantitative method, transcripts of five different biomarkers, including PCA3, AR, SRD5A2, KLK2, and PSMA, were analyzed by multiplex RT-PCR assay in tissue samples from patients with PCa and BPH. It was shown that PCA3 detection, along with associated serum PSA levels, had a 28-fold higher chance for PCa detection with 92% specificity and 94% positive predictive value. uPM3 Test The uPM3 test is a nucleic acid amplification assay detecting the relative expression of PSA mRNA and PCA3/DD3 RNA in the urine using specific beacons as probes in a thermostatted spectrofluorometer. This test has demonstrated a sensitivity range of 46.9%–82.3%, specificity range of 56.3%–89%, and positive predictive range of 59.4%–97.4%. It has a negative predictive value range of 87.8%–98%115 in PCa detection, especially in the low PSA ranges. Duplex Assay for Urinary AMACR and PCA3 As single marker assays, the urinary AMACR and PCA3 tests have shown similar sensitivities, while the AMACR assay had a higher specificity. A duplex assay based on the use of the AMACR and PCA3 scores greatly improves the accuracy and performance of each methodology. Duplex testing may serve as an adjunct assay to the serum PSA test for monitoring elderly patients with repeat negative biopsies. Survivin Survivin is a member of the inhibitor of apoptosis (IAP) family that functions to prevent apoptosis. In locally advanced PCa, cytoplasmic overexpression of survivin predicts local progression. Interestingly, a nuclear expression is associated with improved survival.116 The evaluation of the expression of survivin mRNA in voided urine from patients with bladder, prostate, and renal cancers has shown 100% specificity but no sensitivity.117 Telomerase The use of telomerase activity for the detection of PCa cells obtained after prostatic massage has shown 58% sensitivity and 100% specificity.118 A high telomerase activity has been found in 90% of PCa tissue and in 100% of epithelial cells recovered from urine by immuno-captivation.119 Thymosin b15 Thymosin beta 15 (Tb15), an actin-binding protein, was found to be upregulated at both the mRNA 264 and protein level in high-grade PCa. Using a quantitative high-throughput ELISA, elevated concentrations in urine samples were found in patients preceding PCa recurrence.120,121 Hepsin Hepsin is a trypsin-like transmembrane serine protease. It has been shown to be overexpressed in up to 90% of PCa,122–127 particularly in advanced stage tumors and in tumors with Gleason scores 47. Although there are currently no known serum or urine tests for hepsin detection, the consistent overexpression of hepsin suggests the possibility of a potentially useful biomarker. Bradeion The detection of bradeion using monoclonal antibody-based immunochromatographic membrane strip tests on urine samples has been shown to detect 70% of prostate, renal, and bladder cancers with no false-positive results.128 Mini-Chromosome Maintenance-5 Urinary mini-chromosome maintenance-5 (MCM5) detection is a simple, accurate, and noninvasive method for identifying patients with PCa with sensitivity of 82% and specificity of 73%–93%.129 An increased urinary concentration of 8-OHdG, a biomarker of generalized cellular oxidative stress, was detected by ELISA in the urine of patients with prostate and bladder cancer.130 However, some studies cast doubt on its potential as a biomarker.131 Urinary 5a-reductase type 2 has shown same utility as urinary markers in the diagnosis of PCa, but it is not well studied.132 MOLECULAR MARKERS IN BLOOD Early Prostate Cancer Antigen Early prostate cancer antigen (EPCA) is a PCaassociated nuclear structural protein. An increased expression has been shown in PCa and adjacent areas but is absent in normal prostate tissue obtained from healthy donors and BPH samples.133 EPCA has shown promising results as a clinically useful PCa biomarker as shown by studies demonstrating 84%–94% sensitivity and 85%–100% specificity for PCa by IHC and blood ELISA and in precursor PIN and inflammatory prostatic lesions.134–136 Moreover, what makes it an intriguing biomarker is that positive EPCA staining in an initial PCa negative biopsy may precede the diagnosis of PCa by as much as 5 years. T. Bhavsar, P. McCue, and R. Birbe Prostate Stem Cell Antigen Prostate stem cell antigen (PSCA) is a prostatespecific glycosyl phosphatidylinositol-anchored glycoprotein. PSCA is shown to have strong expression in approximately 85% of PCa, and this expression has been correlated with Gleason score, stage, progression, and the presence of bone, liver, and lymph node metastases.137–141 This glycoprotein is expressed in approximately 73% of high-grade PIN and 22% of low-grade PIN.139 The prognostic value of PSCA has been shown in patients with extraprostatic disease; the presence of PSCA mRNA via peripheral blood RT-PCR is associated with a lower progression-free survival.141 EPIGENETIC BIOMARKERS IN PROSTATE CANCER Glutathione-S-Transferase P1 Glutathione-S-transferases (GSTs) are a ubiquitous family of multifunctional enzymes that conjugate reactive substrates with reduced glutathione (GSH) and are involved in detoxification. Epigenetic alterations are frequent in PCa and are thought to contribute to disease initiation and progression.142,143 Promoter hypermethylation of the GSTP1 gene is a highly specific but insensitive marker of PCa. The methylation of the 5V region of the GSTP1 gene is the most common epigenetic alteration in PCa.144–146 Hypermethylation of the GSTP1 promoter occurs in more than 90% of PCa and has been significantly associated with Gleason score 47 and the presence of a single Gleason 4 and/or 5 grade.144,147 Promoter hypermethylation is associated with high-grade PIN and can be used to distinguish PCa and PIN from BPH.148–151 GSTP1 promoter hypermethylation has been found in 72% of plasma or serum samples, 50% of ejaculates, and 36% of urine samples. GSTP1 methylation in cells derived from serum and urine, if successfully validated, may possess a significant clinical potential for early detection of PCa and posttreatment monitoring. CpG island hypermethylation of GSTP1 promoter also may serve as a useful molecular biomarker for the detection and diagnosis of PCa.149,152 Peripheral blood, urine, ejaculate, and prostatic secretions can be tested for CpG island hypermethylation and may become a useful adjunct to serum screening tests and digital rectal examination. The combinations of GSTP1 along with APC, RASSF1a, PTGS2, and MDR1 CpG island hypermethylation can distinguish primary PCa from benign prostate tissue with a sensitivity of 97.3%– 100% and a specificity of 92%–100%.153 The methylation profile of a panel of 10 hypermethylated genes including GSTP1, RASSF1A, CDH1, APC, 265 Molecular diagnosis of prostate cancer DAPK, MGMT, p14, p16INK4a, RARb2, and TIMP3 was assessed, in which all genes were significantly hypermethylated in the urine sediments of PCa except p14 and p16INK4a.154 Similarly, the methylation profile of a four-gene panel, GSTP1, RASSF1A, RARb2, and APC, was able to discriminate cancers from controls at 86% sensitivity and 89% specificity. Two clinical trials are investigating a panel of hypermethylated genes in urine and serum as an early detection marker (NCT00340717 and NCT01441687) and there is a single trial investigating the association of a miRNA expression profile as a prognostic biomarker (NCT01220427). Various other hypermethylated genes have been proposed as prognostic markers for PCa. The gene encoding for COX2, PTGS2, has been found to be methylated in 88% of cases and has been correlated with an increased risk of PSA recurrence independently of Gleason score or pathological stage.154 A correlation between hypermethylation at APC, RASSF1a, and RUNX3 with PSA value and Gleason score has been shown to have a poor prognosis.155 Interestingly, studies have demonstrated global hypomethylation associated with advanced metastatic PCa.156–159 An IHC study on human PCa showed a significant decrease in the global levels of 5-methylcytosine in patients with recurrent PCa.157 Retrotransposon elements such as LINE-1 and Alu repeats also have been found to be hypomethylated in PCa.158,159 For various reasons, not the least of which is complexity, the diagnostic or prognostic value of global DNA methylation levels needs further evaluation. HISTONE MODIFICATIONS AS BIOMARKERS IN PROSTATE CANCER The global levels of specific histone modifications, H3K9Ac, H3K18Ac, H4K12Ac, H3K4Me2, and H4R3Me2, using IHC have been analyzed in primary PCa. It has been evident that an overexpression of H3K27Me3 global levels in metastatic PCa is associated with biochemical recurrence.161–164 The combination of specific histone modifications, H3K4Me2 and H3K18Ac, has been shown to be indicative of PCa recurrence in low-grade PCa.165 A significant decrease in H3K27Me3 has been found in metastatic disease compared to localized disease using ELISA on serum samples.166 A CHIP-sequencing strategy may be used to profile concurrent histone modifications to investigate potential epigenetic biomarkers for PCa. miRNAs AS BIOMARKERS IN PROSTATE CANCER miRNAs are attractive as potential diagnostic and prognostic biomarkers for PCa due to easy detection in body fluids like blood and serum. These moieties are highly stable and tend to be tissue- and tumorspecific.167,168 A total of 15 differentially expressed miRNAs have been shown to aid in the discrimination between PCa and benign tissue with 82% accuracy using miRNA microarray analysis and RTPCR validation.169 In particular, miR-375, miR-141, and miR-200b are significantly associated with pathological stage and Gleason score using Taqman miRNA microarray analysis.170 GENETIC POLYMORPHISMS AND MUTATIONS MOLECULAR GENE-BASED TESTS DNA-based tests can be used for the detection of microsatellite alterations in PCa. However, these tests are prone to artifact. They require at least 20% of the analyzed genomic DNA from isolated PCa tumor cells. Methylation-specific PCR (MSP) has been shown to be useful for the detection of epigenetic modifications (eg, promoter hypermethylation) as discussed above. The latter has an advantage as it requires a tumor-to-normal ratio of only 0.1%–0.001%.31 RNA-based gene tests to quantify the mRNA of interest is advantageous over DNA due to commercially available US Food and Drug Administration (FDA)-approved technologies such as RTPCR, nucleic acid sequence-based amplification (NASBA), and transcription-mediated amplification (TMA). Of course, the robustness of the sample collection, storage and transport are crucial to maintain the sensitivity of RNA-based tests.160 The genetic polymorphisms examined to date for PCa have exhibited a relatively low penetrance and have not been validated in subsequent studies. A number of loci have been shown to have a positive correlation with PCa risk, including HPC1, HPC2, HPCX, HPC20, CAPB, PCAP, and an unnamed locus at 8p22–23.171 A large-scale association study of 425,000 SNPs within 16,000 genes using 368 matched cases and controls has provided evidence that genetic variants of ICAM5 are associated with a positive risk of PCa. Likewise, the BRCA1 mutation also has been shown to correlate with a positive risk of PCa. GENE EXPRESSION MARKERS IN PROSTATE CANCER Some of the important susceptibility genes for developing PCa include androgen receptor (AR) CAG repeats, MSR1 (macrophage scavenger receptor 1), 266 ELAC2 mutation carriers, BRCA2, ETV6, and AMACR/P504S. Expression levels of the KLK2 and DD3 genes in prostate biopsies may be used for diagnostic discrimination of PCa and benign hyperplasia, especially in patients with low serum levels of PSA. B7-H3, a member of the B7 family, is the first immune molecule that possibly participates in the development of PCa and in predicting recurrence and progression. Hypermethylation of the PDLIM4 gene has been shown to be a sensitive molecular tool in detecting prostate tumorigenesis.172 Increased expression of Golgi phosphoprotein 2 (GOLPH2), a gene that codes for type II Golgi membrane antigen GOLPH2/GP73, has been shown in PCa tissue in a comparative study of GOLPH2 protein, basal cell marker p63, and AMACR in benign and malignant prostate lesions. The abnormal methylation in the promoter region of the DAB2IP gene has been reported to be responsible for transcriptional silencing and a significant role in PCa progression.173 A link between a genetic variation in DAB2IP and the risk of aggressive PCa also has been shown.174 PAR-2, a gene that encodes for a G protein-coupled receptor activated by specific serine proteases, is associated with PCa metastasis175 and is overexpressed in approximately 40% of PCa.176 A markedly increased expression level of EZH2, a member of the polycomb group of proteins, has been demonstrated by gene expression profiling in patients who succumbed to metastatic disease. Interestingly, EZH2 has been found to be a better predictor for clinical progression than surgical margin status, maximum tumor dimension, Gleason score, or preoperative PSA.162 The expression of SPINK-1 a protein that inhibits pancreatic secretion of trypsin, has been detected in urine samples and correlates with the biochemical progression-free interval in PCa. Other genes, such as HEPSIN (TMPRSS1) and Pim-1, have been associated with PCa. Overexpression of CD44 variant 9 is associated with invasion177 and decreased fractions of the v3 variant predict biochemical recurrence.178 BIOMARKERS OF CELL CYCLE PROGRESSION AND SIGNALING PATHWAYS The use of advanced diagnostic techniques to evaluate tumors according to the molecular alterations they carry may lead to the discovery of new markers and targeted treatments of individual tumors. Cyclin D1 overexpression has been associated with poor prognosis after prostatectomy.179,180 Specific inactivation of p14 (ARF) in PCa occurs either through mutations or promoter hypermethylation153,181 and is a rare event. Amplification of the c-myc gene on 8q24 is found in more than 11%–21% of T. Bhavsar, P. McCue, and R. Birbe advanced PCa182,183 and is associated with hormone independence184 and poor prognosis.185 Overexpression of c-myc seems to be involved in tumor progression and hormone resistance rather than tumor development. High levels of p16 in prostatectomy specimens predict a rapid treatment failure,186 whereas decreased expression after radiotherapy predicts poor prognosis.187 Low p16 expression is often found in metastatic PCa lesions.188 The inhibitory role of retinoblastoma (Rb) in the E2F transcription factor complex has been suggested to play a role in PCa. Inactivation of Rb in mouse models has shown a phenotype of PIN.188,189 Loss of hMSH2 expression, a member of the MMR protein family, has been associated with lower recurrence rates after prostatectomy.190,191 Decreased IHC expression of p27 is associated with a high Gleason grade, seminal vesicle invasion, and positive surgical margin. In highgrade PIN, there is an inverse relation between PTEN and p27 expression.192 For markers of alternative growth signaling pathways, growth factors associated with PCa such as IGF-1,193,194 scatter factor/hepatocyte growth factor (HGF),195 and platelet-derived growth factor (PDGF)196 have been found to activate the PI3kinase/AKT/mTOR signaling cascade. NF-κB and subunits are involved in transcriptional regulation of gene expression of multiple growth factors including Bcl-2, cyclin D1, MMP-9, VEGF,197 and PSA.198 These are all found to be elevated in PCa.199 NEW TARGETS AND BIOMARKERS FOR PROSTATE CANCER THERAPY The F77 antigen, serine peptidase inhibitor Kazal type 1 (SPINK1) and survivin (see urine molecular markers above) are some of the new biomarkers for PCa. The F77 antigen, termed prostate cancer lipid antigen (PCLA), holds significant value as a biomarker due to its glycolipid properties. It is expressed on both androgen-dependent and androgen-independent PCa, making it a target for early diagnosis and treatment of advanced diseases. In tissue microarray studies, F77 was shown to stain 112/166 primary and 29/34 metastatic PCa specimens.200 Another biomarker, SPINK1, is specific to a subset of aggressive PCa that does not carry genetic rearrangement of the ETS transcription factors into the TMPRSS2 promoter region, and promotes prostate tumor growth through EGFR.201 NOVEL PROTEOMIC BIOMARKERS FOR PROSTATE CANCER Proteomic and Metabolic Profiling in PCa Proteomic profiling analyzes the subtle changes in proteins in tissue, serum, or urine, and can be used 267 Molecular diagnosis of prostate cancer to differentiate PCa from benign conditions. Using mass spectrometry protein profiling with bioinformatics, microRNA (miR-128) has been implicated for PCa progression.202 The use of metabolite profiling has identified the expression of sarcosine during progression from benign prostate tissue to metastatic PCa.203 Using proteomics expression profiles of androgen-stimulated PCa cells generated by twodimensional electrophoresis (2-DE) and spectrometric analysis, a metastasis-suppressor gene NDKA/ nm23 was identified that may explain a marked reduction in metastatic potential when these cells retain a functional androgen receptor pathway.204 Another proteomic study, which mapped the differences in protein expression profiles between normal and malignant prostate tissues, identified 20 proteins that were lost in malignant transformation, including the novel finding of NEDD8, calponin, and the follistatin-related proteins.205 Proteomic Profiling With Mass Spectrometry Mass spectrometry (MS) has been used to identify new individual biomarkers, such as PCa-24 in the serum206 and calgranulin B/MRP-14 in voided urine after prostatic massage.207 High-resolution 2-DE have shown absence of proteins A and B in urine from PCa and BPH patients, while protein F was highly expressed in BPH but not in PCa.208 The use of proteomic patterns from MS has shown 95% sensitivity and 78% specificity in identifying PCa.209 Enhanced results have been demonstrated using a boosted decision tree algorithm to analyze the surface-enhanced laser desorption/ionization (SELDI)/ time of flight (TOF) data, showing a sensitivity and specificity of 97%.210 Moreover, SELDI has been coupled with an artificial intelligence learning algorithm to identify biomarkers for early detection of PCa with a high sensitivity (83%), specificity (97%), and a positive predictive value (96%). The results of these study show the potential of SELDI proteomics in the early detection and diagnosis of PCa. Proteomic Techniques for PCa Biomarkers Proteomic approaches have been used in tissues and cancer cell lines to identify PCa biomarkers. Potential biomarkers for high grade PCa identification include nucleoside diphosphate kinase 1, chromobox protein, 39S ribosomal protein L12, cytosol aminopeptidase, endopeptidase C1p, inorganic pyrophosphatase, metaxin 2, GST-pi, acyl-CpA dehydrogenase, lysophospholipase, NADHubiquinone oxidoreductase, 60- and 70-kd GRP-78, b-actin, cytokeratins 7, 8, and 18, and stomatin-like protein 2, a-actin, and mutant desmin using 2DGE coupled with MS.211 Another four novel potential biomarkers—follistatin, chemokine (C-X-C motif) ligand 16, pentraxin 3, and spondin 2—have been identified by examining conditioned media from PCa cell lines using strong cation exchange highperformance liquid chromatography (HPLC) and HPLC-tandem MS.212 Unfortunately, there have been only a few PCa biomarkers on biological fluids using proteomic studies as the concentration of potential biomarkers varies in different human body fluids. Technically, it is difficult to identify lowabundance proteins.213,214 Proteomic PCa Biomarkers in Sera MS-based mass profiling (MP) combined with multivariate analysis has detected a protein at m/z 7771, identified as platelet factor 4 in PCa patients.215 It was significantly decreased in the sera of all the patients with metastatic PCa but not in those with localized PCa or healthy individuals. Similarly, another potential PCa biomarker protein at m/z at 8946, Apolipoprotein A-II, was identified using HPLC, reverse-phase chromatography, sosium dodecyl sulfate–polyacrylamide gel electropheresis (SDS-PAGE), HPLC–MS/MS, and SELDI/TOF-MS.216 Other potential PCa serum protein biomarkers that have been identified are collective variant forms of serum amyloid A (SAAs) using SELDI/TOF217 and squamous cell carcinoma antigen 1 (SCCA1), calgranulin B, and haptoglobin-related protein using anion displacement liquid chromate-focusing chromatography for serum fractionation, two-dimensional differential in-gel electrophoresis (2D-DIGE).218 FUTURE DIRECTIONS AND CONCLUSIONS Since PCa is a heterogeneous disease, it is clear that a defined set of markers will be important in the early diagnosis, monitoring, and prognoses of PCa. A wide array of molecular markers discussed here may be used in the near future as adjuncts to currently established prognostic parameters. That said, the practicality of the use of many molecular biomarkers is questionable, and it will take time for these markers to find their way into the clinical arena. The field of fusion genes is promising at the clinical and translational levels. The high incidence and the connection with the androgenic signaling pathway of TMPRSS2-ERG make it a biomarker with much potential at the diagnostic and prognostic levels. Microarray studies using exon-specific target elements can identify specific splice variants that may be more correlative with PCa phenotypes. PCA3 has shown improved positive predictive value and sensitivity for detection of PCa in the low-PSA ranges after previous negative biopsies. Further studies may lead to the application of GSTP1 hypermethylation in augmenting standard histopathology 268 for the diagnosis of PCa. Fluorescence-based quantitative RT-PCR assay for the determination of DD3/ PCA3 transcripts shows great promise as a noninvasive diagnostic tool and will have great impact for the reduction of the number of unnecessary biopsies. A limited number of molecular markers in PCa tissue are of clinical use in predicting outcome or response to therapy. They include p53, Bcl-2, p16INK4A, p27Kip1, c-Myc, AR, E-cadherin, and VEGF. With the advancement of new technologies such as next-generation sequencing and with the development of platforms for global epigenome analyses, epigenetic biomarkers may be used as powerful tools for determining diagnosis, prognosis, and therapy response in PCa. Modern advances in the field of proteomic techniques such as 2D-DIGE and SELDI/TOF-MS hold the promise of identifying novel PCa biomarkers for early diagnosis and prognosis. We close this truncated summary on a practical note that while extensive clinical validation of these novel PCa biomarkers remains one of the most significant challenges, overcoming this impediment will by no means eliminate the difficulty in their identification and development. REFERENCES 1. Jemal A, Murray T, Ward E, et al. Cancer statistics. CA Cancer J Clin. 2005;55:10–30. 2. Boyle P, Ferlay J. Cancer incidence and mortality in Europe. Ann Oncol. 2005;16:481–8. 3. Franceschi S, La Vecchia C. Cancer epidemiology in the elderly. Crit Rev Oncol Hematol. 2001;39: 219–26. 4. Ercole CJ, Lange PH, Mathisen M, et al. Prostatic specific antigen and prostatic acid phosphatase in the monitoring and staging of patients with prostatic cancer. J Urol. 1987;138:1181–4. 5. Thompson IM, Pauler DK, Goodman PJ, et al. Prevalence of prostate cancer among men with a prostate-specific antigen level o or ¼ 4.0 ng per milliliter. N Engl J Med. 2004;350:2239–46. 6. Oh WK, Hurwitz M, D'Amico AV, et al. Neoplasms of the prostate. In: Kufe DW, Pollock RE, Weichselbaum RR, et al, editors. Cancer medicine. Hamilton, Canada: BC Decker; 2003. 7. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182: 311–22. 8. Chang BL, Isaacs SD, Wiley KE, et al. Genome-wide screen for prostate cancer susceptibility genes in men with clinically significant disease. Prostate. 2005;64:356–61. 9. Rubio J, Ramos D, Lopez-Guerrero JA, et al. Immunohistochemical expression of Ki-67 antigen,COX-2 and Bax/Bcl-2 in prostate cancer; prognostic value in biopsies and radical prostatectomy specimens. Eur Urol. 2005;48:745–51. T. Bhavsar, P. McCue, and R. Birbe 10. Revelos K, Petraki C, Gregorakis A, et al. p27(kip1) and Ki-67 (MIB1) immunohistochemical expression in radical prostatectomy specimens of patients with clinically localized prostate cancer. In Vivo. 2005;19: 911–20. 11. Khatami A, Hugosson J, Wang W, et al. Ki-67 in screen-detected, low-grade, low-stage prostate cancer, relation to prostate-specific antigen doubling time, Gleason score and prostate-specific antigen relapse after radical prostatectomy. Scand J Urol Nephrol. 2009;43:12–8. 12. 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;100:888–93. 13. 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;27:3177–84. 14. Bookstein R, MacGrogan D, Hilsenbeck SG, Sharkey F, Allred DC. p53 is mutated in a subset of advancedstage prostate cancers. Cancer Res. 1993;53:3369–73. 15. Navone NM, Troncoso P, Pisters LL, et al. p53 protein accumulation and gene mutation in the progression of human prostate carcinoma. J Natl Cancer Inst. 1993;85:1657–69. 16. Bauer JJ, Sesterhenn IA, Mostofi FK, McLeod DG, Srivastava S, Moul JW. Elevated levels of apoptosis regulator proteins p53 and bcl-2 are independent prognostic biomarkers in surgically treated clinically localized prostate cancer. J Urol. 1996;156:1511–6. 17. Stricker HJ, Jay JK, Linden MD, Tamboli P, Amin MB. Determining prognosis of clinically localized prostate cancer by immunohistochemical detection of mutant p53. Urology. 1996;47:366–9. 18. Visakorpi T, Kallioniemi OP, Heikkinen A, Koivula T, Isola J. Small subgroup of aggressive, highly proliferative prostatic carcinomas defined by p53 accumulation. J Natl Cancer Inst. 1992;84:883–7. 19. Osman I, Drobnjak M, Fazzari M, Ferrara J, Scher HI, Cordon-Cardo C. Inactivation of the p53 pathway in prostate cancer: impact on tumor progression. Clin Cancer Res. 1999;5:2082–8. 20. Grignon DJ, Caplan R, Sarkar FH, et al. p53 status and prognosis of locally advanced prostatic adenocarcinoma: a study based on RTOG 8610. J Natl Cancer Inst. 1997;89:158–65. 21. Kuczyk MA, Serth J, Bokemeyer C, et al. The prognostic value of p53 for long-term and recurrence-free survival following radical prostatectomy. Eur J Cancer. 1998;34:679–86. 22. Inoue T, Segawa T, Shiraishi T, et al. Androgen receptor, Ki67, and p53 expression in radical prostatectomy specimens predict treatment failure in Japanese population. Urology. 2005;66:332–7. 23. Shi XB, Gandour-Edwards R, Beckett LA, Deitch AD, Vere White RW. A modified yeast assay used on archival samples of localized prostate cancer tissue improves the detection of p53 abnormalities and 269 Molecular diagnosis of prostate cancer 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. increases their predictive value. BJU Int. 2004;94: 996–1002. Heidenberg HB, Sesterhenn IA, Gaddipati JP, et al. Alteration of the tumor suppressor gene p53 in a high fraction of hormone refractory prostate cancer. J Urol. 1995;154:414–21. Raffo AJ, Perlman H, Chen MW, Day ML, Streitman JS, Buttyan R. Overexpression of bcl-2 protects prostate cancer cells from apoptosis in vitro and confers resistance to androgen depletion in vivo. Cancer Res. 1995;55:4438–45. Apakama I, Robinson MC, Walter NM, et al. bcl-2 overexpression combined with p53 protein accumulation correlates with hormone refractory prostate cancer. Br J Cancer. 1996;74:1258–62. Kuefer R, Varambally S, Zhou M, et al. Alphamethylacyl-CoA racemase: expression levels of this novel cancer biomarker depend on tumor differentiation. Am J Pathol. 2002;161:841–8. Zha S, Ferdinandusse S, Denis S, et al. Alpha-methylacylCoA racemase as an androgen-independent growth modifier in prostate cancer. Cancer Res. 2003;63: 7365–76. Jiang Z, Wu CL, Woda BA, et al. Alpha-methylacylCoA racemase: a multi-institutional study of a new prostate cancer marker. Histopathology. 2004;45: 218–25. Jiang Z, Woda BA, Wu CL, Yang XJ. Discovery and clinical application of a novel prostate cancer marker: alpha-methylacyl CoA racemase (P504S). Am J Clin Pathol. 2004;122:275–89. Hessels D, Verhaegh GW, Schalken JA, Witjes JA. Applicability of biomarkers in the early diagnosis of prostate cancer. Expert Rev Mol Diagn. 2004;4:513–26. Rubin MA, Zhou M, Dhanasekaran SM, et al. alphaMethylacyl coenzyme A racemase as a tissue biomarker for prostate cancer. JAMA. 2002;287:1662–70. Fleshman RL, MacLennan GT. Immunohistochemical markers in the diagnosis of prostate cancer. J Urol. 2005;173:1759. Jiang Z, Li C, Fischer A, Dresser K, Woda BA. Using an AMACR (P504S)/34betaE12/p63 cocktail for the detection of small focal prostate carcinoma in needle biopsy specimens. Am J Clin Pathol. 2005;123: 231–6. Jiang Z, Wu CL, Woda BA, et al. P504S/alphamethylacyl-CoA racemase: a useful marker for diagnosis of small foci of prostatic carcinoma on needle biopsy. Am J Surg Pathol. 2002;26:1169–74. Zielie PJ, Mobley JA, Ebb RG, et al. A novel diagnostic test for prostate cancer emerges from the determination of alpha-methylacyl-coenzyme a racemase in prostatic secretions. J Urol. 2004;172:1130–3. Rogers CG, Yan G, Zha S, et al. Prostate cancer detection on urinalysis for alpha methylacyl coenzyme a racemase protein. J Urol. 2004;172:1501–3. Gregorakis AK, Holmes EH, Murphy GP. Prostatespecific membrane antigen: current and future utility. Semin Urol Oncol. 1998;16:2–12. Murphy GP, Kenny GM, Ragde H, et al. Measurement of serum prostate-specific membrane antigen, a new 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. prognostic marker for prostate cancer. Urology. 1998;51:89–97. Murphy GP, Barren RJ, Erickson SJ, et al. Evaluation and comparison of two new prostate carcinoma markers. Free-prostate specific antigen and prostate specific membrane antigen. Cancer. 1996;78: 809–18. Marchal C, Redondo M, Padilla M, et al. Expression of prostate specific membrane antigen (PSMA) in prostatic adenocarcinoma and prostatic intraepithelial neoplasia. Histol Histopathol. 2004;19:715–8. Ren JQ, Chen ZQ, Zheng L, et al. Correlation study of expression levels of prostate-specific membrane antigen and prostate-specific antigen with Gleason score of prostate carcinoma [in Chinese]. Zhonghua Zhong Liu Za Zhi. 2004;26:735–8. Murphy G, Ragde H, Kenny G, et al. Comparison of prostate specific membrane antigen, and prostate specific antigen levels in prostatic cancer patients. Anticancer Res. 1995;15:1473–9. Xu T, Chen X, Wang XF, et al. Study of PSA, PSMA and hK2 mRNA in peripheral blood of prostate cancer patients and its clinical implications. Beijing Da Xue Xue Bao. 2004;36:164–8. Su SL, Boynton AL, Holmes EH, et al. Detection of extraprostatic prostate cells utilizing reverse transcription-polymerase chain reaction. Semin Surg Oncol. 2000;18:17–28. Lopes AD, Davis WL, Rosenstraus MJ, et al. Immunohistochemical and pharmacokinetic characterization of the site-specific immunoconjugate CYT-356 derived from antiprostate monoclonal antibody 7E11-C5. Cancer Res. 1990;50:6423–9. Cho SY, Choi K, Jeon JH, et al. Differential alternative splicing of human transglutaminase 4 in benign prostate hyperplasia and prostate cancer. Exp Mol Med. 2010;42:310–8. Angelucci A, Festuccia C, Gravina GL, et al. Osteopontin enhances the cell proliferation induced bythe epidermal growth factor in human prostate cancer cells. Prostate. 2004;59:157–66. Robertson BW, Bonsal L, Chellaiah MA. Regulation of Erk1/2 activation by osteopontin in PC3 human prostate cancer cells. Mol Cancer. 2010;9:260–70. Weber GF, Lett GS, Haubein NC. Categorical metaanalysis of osteopontin as a clinical cancer marker. Oncol Rep. 2011;25:433–41. Angelucci A, Festuccia C, D'Andrea G, Teti A. Bologna M. Osteopontin modulates prostate carcinoma invasive capacity through RGD-dependent upregulation of plasminogen activators. Biol Chem. 2002;383:229–34. Khodavirdi AC, Song Z, Yang S, et al. Increased expression of osteopontin contributes to the progression of prostate cancer. Cancer Res. 2006;66: 883–8. Liu W, Gong J, Hu J, et al. Quantitative assessment of AKAP12 promoter methylation in human prostate cancer using methylation-sensitive high-resolution melting: correlation with Gleason Score. Urology. 2011;77(1006):e1–7. 270 54. Ramankulov A, Lein M, Kristiansen G, Loening SA, Jung K. Plasma osteopontin in comparison with bone markers as indicator of bone metastasis and survival outcome in patients with prostate cancer. Prostate. 2007;67:330–40. 55. Zhang Y, Forootan SS, Kamalian L, et al. Suppressing tumourigenicity of prostate cancer cells by inhibiting osteopontin expression. Int J Oncol. 2011;38: 1083–91. 56. Hamdy FC, Autzen P, Robinson MC, Horne CH, Neal DE, Robson CN. Immunolocalization and messenger RNA expression of bone morphogenetic protein-6 in human benign and malignant prostatic tissue. Cancer Res. 1997;57:4427–31. 57. Autzen P, Robson CN, Bjartell A, et al. Bone morphogenetic protein 6 in skeletal metastases from prostate cancer and other common human malignancies. Br J Cancer. 1998;78:1219–23. 58. Tamada H, Kitazawa R, Gohji K, Kitazawa S. Epigenetic regulation of human bone morphogenetic protein 6 gene expression in prostate cancer. J Bone Miner Res. 2001;16:487–96. 59. De Pinieux G, Flam T, Zerbib M, et al. Bone sialoprotein, bone morphogenetic protein 6 and thymidine phosphorylase expression in localized human prostatic adenocarcinoma as predictors of clinical outcome: a clinicopathological and immunohistochemical study of 43 cases. J Urol. 2001;166: 1924–30. 60. Mao X, Yu Y, Boyd LK, et al. Distinct genomic alterations in prostate cancers in Chinese and western populations suggest alternative pathways of prostate carcinogenesis. Cancer Res. 2010;70:5207–12. 61. Domingo-Domenech J, Mellado B, Ferrer B, et al. Activation of nuclear factor-κB in human prostate carcinogenesis and association to biochemical relapse. Br J Cancer. 2005;93:1285–94. 62. Jaraj SJ, Augsten M, Ha¨ggarth L, et al. GAD1 is a biomarker for benign and malignant prostatic tissue. Scand J Urol Nephrol. 2011;45:39–45. 63. Haggarth L, Hagglo FC, Jaraj SJ, et al. Diagnostic biomarkers of prostate cancer. Scand J Urol Nephrol. 2011;45:60–7. 64. Freedland SJ, Seligson DB, Liu AY, et al. Loss of CD10 (neutral endopeptidase) is a frequent and early event in human prostate cancer. Prostate. 2003;55:71–80. 65. Zellweger T, Ninck C, Mirlacher M, et al. Tissue microarray analysis reveals prognostic significance of syndecan-1 expression in prostate cancer. Prostate. 2003;55:20–9. 66. Xin W, Rhodes DR, Ingold C, Chinnaiyan AM, Rubin MA. Dysregulation of the annexin family protein family is associated with prostate cancer progression. Am J Pathol. 2003;162:255–61. 67. Udby L, Cowland JB, Johnsen AH, Sorensen OE, Borregaard N, Kjeldsen L. An ELISA for SGP28/ CRISP-3, a cysteine-rich secretory protein in human neutrophils, plasma, and exocrine secretions. J Immunol Methods. 2002;263:43–55. 68. Stephan C, Jung K, Lein M, Sinha P, Schnorr D, Loening SA. Molecular forms of prostate-specific antigen and human kallikrein 2 as promising tools T. Bhavsar, P. McCue, and R. Birbe 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. for early diagnosis of prostate cancer. Cancer Epidemiol Biomarkers Prev. 2000;9:1133–47. Klee GG, Goodmanson MK, Jacobsen SJ, et al. Highly sensitive automated chemiluminometric assay for measuring free human glandular kallikrein-2. Clin Chem. 1999;45:800–6. Haese A, Vaisanen V, Finlay JA, et al. Standardization of two immunoassays for human glandular kallikrein 2. Clin Chem. 2003;49:601–10. Vaisanen V, Eriksson S, Ivaska KK, Lilja H, Nurmi M, Pettersson K. Development of sensitive immunoassays for free and total human glandular kallikrein 2. Clin Chem. 2004;50:1607–17. Brown JM, Vessella RL, Kostenuik PJ, Dunstan CR, Lange PH, Corey E. Serum osteoprotegerin levels are increased in patients with advanced prostate cancer. Clin Cancer Res. 2001;7:2977–83. Eaton CL, Wells JM, Holen I, Croucher PI, Hamdy FC. Serum osteoprotegerin (OPG) levels are associated with disease progression and response to androgen ablation in patients with prostate cancer. Prostate. 2004;59:304–10. Deftos LJ, Abrahamsson PA. Granins and prostate cancer. Urology. 1998;51:141–5. Fracalanza S, Prayer-Galetti T, Pinto F, et al. Plasma chromogranin A in patients with prostate cancer improves the diagnostic efficacy of free/total prostate-specific antigen determination. Urol Int. 2005;75:57–61. Chapdelaine P, Paradis G, Tremblay RR, et al. High level of expression in the prostate of a human glandular kallikrein mRNA related to prostatespecific antigen. FEBS Lett. 1988;236:205–8. Schedlich LJ, Bennetts BH, Morris BJ. Primary structure of a human glandular kallikrein gene. DNA. 1987;6:429–37. Nam RK, Diamandis EP, Toi A, et al. Serum human glandular kallikrein-2 protease levels predict the presence of prostate cancer among men with elevated prostate-specific antigen. J Clin Oncol. 2000; 18:1036–42. Becker C, Piironen T, Pettersson K, et al. Clinical value of human glandular kallikrein 2 and free and total prostate-specific antigen in serum from a population of men with prostate-specific antigen levels 3.0 ng/mL or greater. Urology. 2000;55:694–9. George DJ, Halabi S, Shepard TF, et al. The prognostic significance of plasma interleukin-6 levels in patients with metastatic hormone refractory prostate cancer: results from Cancer and Leukemia Group B 9480. Clin Cancer Res. 2005;11:1815–20. Barre B, Avril S, Coqueret O. Opposite regulation of myc and p21waf1 transcription by STAT3 proteins. J Biol Chem. 2003;278:2990–6. Shariat SF, Kattan MW, Traxel E, et al. Association of pre- and postoperative plasma levels of transforming growth factor beta(1) and interleukin 6 and its soluble receptor with prostate cancer progression. Clin Cancer Res. 2004;10:1992–9. Drachenberg DE, Elgamal AA, Rowbotham R, et al. Circulating levels of interleukin-6 in patients with Molecular diagnosis of prostate cancer 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. hormone refractory prostate cancer. Prostate. 1999; 41:127–33. Michalaki V, Syrigos K, Charles P, et al. Serum levels of IL-6 and TNF-alpha correlate with clinicopathological features and patient survival in patients with prostate cancer. Br J Cancer. 2004;90:2312–6. Furuya Y, Nishio R, Junicho A, et al. Serum interleukin-11 in patients with benign prostatic hyperplasia and prostate cancer. Int Urol Nephrol. 2005;37:69–71. Lehrer S, Diamond EJ, Mamkine B, et al. Serum interleukin-8 is elevated in men with prostate cancer and bone metastases. Technol Cancer Res Treat. 2004;3:411. Kattan MW, Shariat SF, Andrews B, et al. The addition of interleukin-6 soluble receptor and transforming growth factor beta1 improves a preoperative nomogram for predicting biochemical progression in patients with clinically localized prostate cancer. J Clin Oncol. 2003;21:3573–9. Di Vizio D, Morello M, Sotgia F, et al. An absence of stromal caveolin-1 is associated with advanced prostate cancer, metastatic disease and epithelial Akt activation. Cell Cycle. 2009;8:2420–4. Musiał J, Sporny S, Nowicki A. Prognostic significance of E-cadherin and Ezrin immunohistochemical expression in prostate cancer. Pol J Pathol. 2007; 58:235–43. Di Lorenzo G, Tortora G, D’Armiento FP, et al. Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res. 2002;8:3438–44. Duque JL, Loughlin KR, Adam RM, Kantoff PW, Zurakowski D, Freeman MR. Plasma levels of vascular endothelial growth factor are increased in patients with metastatic prostate cancer. Urology. 1999;54:523–7. George DJ, Halabi S, Shepard TF, et al. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin Cancer Res. 2001;7:1932–6. El-Gohary YM, Silverman JF, Olson PR, et al. Endoglin (CD105) and vascular endothelial growth factor as prognostic markers in prostatic adenocarcinoma. Am J Clin Pathol. 2007;127:572–9. Weidner N, Carroll PR, Flax J, Blumenfeld W, Folkman J. Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma. Am J Pathol. 1993;143:401–9. Miller JC, Zhou H, Kweke J, et al. Antibody microarray profiling of human prostate cancer sera: antibody screening and identification of potential biomarkers. Proteomics. 2003;3:56–63. Kumar-Sinha C, Tomlins SA, Chinnaiyan AM. Recurrent gene fusions in prostate cancer. Nat Rev Cancer. 2008;8:497–511. Tomlins SA, Rhodes DR, Perner S, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–8. 271 98. Gerdes J, Schwab U, Lemke H, et al. Production of a mouse monoclonal antibody reactive with a human nuclear antigen associated with cell proliferation. Int J Cancer. 1983;31:13–20. 99. Mosquera JM, Mehra R, Regan MM, et al. Prevalence of TMPRSS2-ERG fusion prostate cancer among men undergoing prostate biopsy in the United States. Clin Cancer Res. 2009;15:4706–11. 100. 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: 5103. 101. Yoshimoto M, Joshua AM, Cunha IW, et al. Absence of TMPRSS2: ERG fusions and PTEN losses in prostate cancer is associated with a favorable outcome. Mod Pathol. 2008;21:1451–60. 102. Mwamukonda K, Chen Y, Ravindranath L, et al. Quantitative expression of TMPRSS2 transcript in prostate tumor cells reflects TMPRSS2-ERG fusion status. Prostate Cancer Prostatic Dis. 2010;13 47-5. 103. Fradet Y. Biomarkers in prostate cancer diagnosis and prognosis: beyond prostate-specific antigen. Curr Opin Urol. 2009;19:243–6. 104. Cao DL, Ye DW, Zhang HL, et al. A multiplex model of combining gene-based, protein-based, and metabolitebased with positive and negative markers in urine for the early diagnosis of prostate cancer. Prostate. 2010;71:700. 105. Nilsson J, Skog J, Nordstrand A, et al. Prostate cancerderived urine exosomes: a novel approach to biomarkers for prostate cancer. Br J Cancer. 2009;100: 1603–7. 106. Mitchell PJ, Welton J, Staffurth J, Court J, et al. Can urinary exosomes act as treatment response markers in prostate cancer? J Transl Med. 2009;12:4. 107. Bussemakers MJ, van Bokhoven A, Verhaegh GW, et al. DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res. 1999;59:5975–9. 108. de Kok JB, Verhaegh GW, Roelofs RW, et al. DD3 (PCA3), a very sensitive and specific marker to detect prostate tumors. Cancer Res. 2002;62:2695–8. 109. Ploussard G, de la Taille A. Urine biomarkers in prostate cancer. Nat Rev Urol. 2010;7:101–9. 110. Hessels D, Klein Gunnewiek JM, van Oort I, et al. DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer. Eur Urol. 2003;44:8–15. 111. Tinzl M, Marberger M, Horvath S. Chypre C. DD3PCA3 RNA analysis in urine-a new perspective for detecting prostate cancer. Eur Urol. 2004;46:182–6. 112. Fradet Y, Saad F, Aprikian A, et al. uPM3, a new molecular urine test for the detection of prostate cancer. Urology. 2004;64:311–5. 113. Groskopf J, Aubin SM, Deras IL, 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. 114. 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;54:1081. 272 115. Ruiz-Aragon J, Marquez-Pelaez S. Assessment of the PCA3 test for prostate cancer diagnosis: a systematic review and meta-analysis. Actas Urol Esp. 2010;34: 346–55. 116. Zhang M, Hammond EH, Suzuki Y, et al. Prognostic value of survivin in locally advanced prostate cancer: study based on RTOG 8610. Int J Radiat Oncol Biol Phys. 2009;73:1033–42. 117. Wang H, Xi X, Kong X, Huang G, Ge G. The expression and significance of survivin mRNA in urinary bladder carcinomas. J Cancer Res Clin Oncol. 2004;130:487–90. 118. Meid FH, Gygi CM, Leisinger HJ, Bosman FT, Benhattar J. The use of telomerase activity for the detection of prostatic cancer cells after prostatic massage. J Urol. 2001;165:1802–5. 119. Botchkina GI, Kim RH, Botchkina IL, Kirshenbaum A, Frischer Z, Adler HL. Noninvasive detection of prostate cancer by D. Hessels et al. / EAU Update Series 3 (2005). 200-213 211 quantitative analysis of telomerase activity. Clin Cancer Res. 2005;11:3243–9. 120. Hutchinson LM, Chang EL, Becker CM, et al. Development of a sensitive and specific enzyme linked immunosorbent assay for thymosin beta15, a urinary biomarker of human prostate cancer. Clin Biochem. 2005;38:558–71. 121. Hutchinson LM, Chang EL, Becker CM, et al. Use of thymosin beta15 as a urinary biomarker in human prostate cancer. Prostate. 2005;16:116–27. 122. Dhanasekaran SM, Barrette TR, Ghosh D, et al. Delineation of prognostic biomarkers in prostate cancer. Nature. 2001;412:822–6. 123. Luo J, Duggan DJ, Chen Y, et al. Human prostate cancer and benign prostatic hyperplasia: molecular dissection by gene expression profiling. Cancer Res. 2001;61:4683–8. 124. Magee JA, Araki T, Patil S, et al. Expression profiling reveals hepsin overexpression in prostate cancer. Cancer Res. 2001;61:5692–6. 125. Rhodes DR, Barrette TR, Rubin MA, et al. Metaanalysis of microarrays: Interstudy validation of gene expression profiles reveals pathway dysregulation in prostate cancer. Cancer Res. 2002;62: 4427–33. 126. Stamey TA, Warrington JA, Caldwell MC, et al. Molecular genetic profiling of Gleason grade 4/5 prostate cancers compared to benign prostatic hyperplasia. J Urol. 2001;166:2171–7. 127. Welsh JB, Sapinoso LM, Su AI, et al. Analysis of gene expression identifies candidate markers and pharmacological targets in prostate cancer. Cancer Res. 2001;61:5974–8. 128. Tanaka M, Tanaka T, Matsuzaki S, et al. Rapid and quantitative detection of human septin family Bradeion as a practical diagnostic method of colorectal and urologic cancers. Med Sci Monit. 2003;9:MT61–8. 129. Dudderidge TJ, Kelly JD, Wollenschlaeger A, et al. Diagnosis of prostate cancer by detection of minichromosome maintenance 5 protein in urine sediments. Br J Cancer. 2010;103:701–7. T. Bhavsar, P. McCue, and R. Birbe 130. Jeronimo C, Nomoto S, Caballero OL, et al. Mitochondrial mutations in early stage prostate cancer and bodily fluids. Oncogene. 2001;20:5195–8. 131. Richardson T, McCanse W, Casale GP. Tissue-based quantification of 8-hydroxy-2’-deoxyguanosine in human prostate biopsies using quantitative fluorescence imaging analysis. Urology. 2009;74:1174–9. 132. Lombardo ME, Hudson PB. Preliminary evaluation of 5a-reductase type 2 in urine as potential marker for prostate disease. Steroids. 1997;62:682–5. 133. Partin AW, Getzenberg RH, CarMichael MJ, et al. Nuclear matrix protein patterns in human benign prostatic hyperplasia and prostate cancer. Cancer Res. 1993;53:744–6. 134. Dhir R, Vietmeier B, Arlotti J, et al. Early identification of individuals with prostate cancer in negative biopsies. J Urol. 2004;171:1419–23. 135. Uetsuki H, Tsunemori H, Taoka R, et al. Expression of a novel biomarker, EPCA, in adenocarcinomas and precancerous lesions in the prostate. J Urol. 2005;174:514–8. 136. Paul B, Dhir R, Landsittel D, et al. Detection of prostate cancer with a blood-based assay for early prostate cancer antigen. Cancer Res. 2005;65: 4097–100. 137. Han KR, Seligson DB, Liu X, et al. Prostate stem cell antigen expression is associated with Gleason score, seminal vesicle invasion and capsular invasion in prostate cancer. J Urol. 2004;171:1117–21. 138. Lam JS, Yamashiro J, Shintaku IP, et al. Prostate stem cell antigen is overexpressed in prostate cancer metastases. Clin Cancer Res. 2005;11:2591–6. 139. Zhigang Z, Wenlv S. Prostate stem cell antigen (PSCA) expression in human prostate cancer tissues: Implications for prostate carcinogenesis and progression of prostate cancer. Jpn J Clin Oncol. 2004;34: 414–9. 140. Gu Z, Thomas G, Yamashiro J, et al. Prostate stem cell antigen (PSCA) expression increases with high Gleason score, advanced stage and bone metastasis in prostate cancer. Oncogene. 2000;19:1288–96. 141. Hara N, Kasahara T, Kawasaki T, et al. Reverse transcription polymerase chain reaction detection of prostate-specific antigen, prostate-specific membrane antigen, and prostate stem cell antigen in one milliliter of peripheral blood: value for the staging of prostate cancer. Clin Cancer Res. 2002;8:1794–9. 142. Schulz WA, Hatina J. Epigenetics of prostate cancer: beyond DNA methylation. J Cell Mol Med. 2006; 10:100–25. 143. Li LC, Carroll PR, Dahiya R. Epigenetic changes in prostate cancer: implication for diagnosis and treatment. J Natl Cancer Inst. 2005;97:103–15. 144. Lee W, Morton RA, Epstein JL, Brooks JD, Campbell PA, Bova GS. Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A.. 1994;91:11733–7. 145. Lee WH, Isaacs WB, Bova GS, Nelson WGCG. island methylation changes near the GSTP1 gene in prostatic carcinoma cells detected using the polymerase Molecular diagnosis of prostate cancer 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. 156. 157. 158. 159. chain reaction: a new prostate cancer biomarker. Cancer Epidemiol Biomark Prev. 1997;6:433–50. Esteller M, Corn PG, Urena JM, Gabrielson E, Baylin SB, Herman JG. Inactivation of glutathione S-transferase P1 gene by promoter hypermethylation in human neoplasia. Cancer Res. 1999;58:4515–8. Bernardini S, Miano R, Iori R, et al. Hypermethylation of the CpG islands in the promoter region of the GSTP1 gene in prostate cancer: a useful diagnostic and prognostic marker? Clin Chim Acta. 2004;350: 181–8. 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;93:1747–52. Chu DC, Chuang CK, Fu JB, et al. The use of real-time quantitative polymerase chain reaction to detect hypermethylation of the CpG islands in the promoter region flanking the GSTP1 gene to diagnose prostate carcinoma. J Urol. 2002;167:1854–8. Brooks JD, Weinstein M, Lin X, et al. CG island methylation changes near the GSTP1 gene in prostatic intraepithelial neoplasia. Cancer Epidemiol Biomark Prev. 1998;7:531–6. Cairns P, Esteller M, Herman JG, et al. Molecular detection of prostate cancer in urine by GSTP1 hypermethylation. Clin Cancer Res. 2001;7:2727–30. 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;95:1634. Yegnasubramanian S, Kowalski J, Gonzalgo ML, et al. Hypermethylation of CpG islands in primary and metastatic human prostate cancer. Cancer Res. 2004;64:1975–86. Roupret M, Hupertan M, Yates, DR V, et al. Molecular detection of localized prostate cancer using quantitative methylation-specific PCR on urinary cells obtained following prostate massage. Clin Cancer Res. 2007;13:1720–5. Kang GH, Lee S, Lee HJ, Hwang KS. Aberrant CpG island hypermethylation of multiple genes in prostate cancer and prostatic intraepithelial neoplasia. J Pathol. 2004;202:233. Yegnasubramanian S, Haffner MC, Zhang Y, et al. DNA hypomethylation arises later in prostate cancer progression than CpG island hypermethylation and contributes to metastatic tumor heterogeneity. Cancer Res. 2008;68:8954–67. 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;156:31–6. Santourlidis S, Florl A, Ackermann R, Wirtz HC, Schulz WA. High frequency of alterations in DNA methylation in adenocarcinoma of the prostate. Prostate. 1999;39:166–74. Schulz WA, Elo JP, Florl AR, et al. Genomewide DNA hypomethylation is associated with alterations on chromosome 8 in prostate carcinoma. Genes Chromosomes Cancer. 2002;35:58–65. 273 160. Neumaier M, Braun A, Wagener C. Fundamentals of quality assessment of molecular amplification methods in clinical diagnostics. International Federation of Clinical Chemistry Scientific Division Committee on Molecular Biology Techniques. Clin Chem. 1998; 44:12–26. 161. Rhodes DR, Sanda MG, Otte AP, Chinnaiyan AM, Rubin MA. Multiplex biomarker approach for determining risk of prostate-specific antigen-defined recurrence of prostate cancer. J Natl Cancer Inst. 2003;95:661–8. 162. Hoffmann MJ, Engers R, Florl AR, Otte AP, Muller M, Schulz WA. Expression changes in EZH2, but not in BMI-1, SIRT1, DNMT1 or DNMT3B are associated with DNA methylation changes in prostate cancer. Cancer Biol Ther. 2007;6:1403–12. 163. Varambally S, Dhanasekaran SM, Zhou M, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624–9. 164. van Leenders GJ, Dukers D, Hessels D, et al. Polycombgroup oncogenes EZH2, BMI1, and RING1 are overexpressed in prostate cancer with adverse pathologic and clinical features. Eur Urol. 2007;52: 455–63. 165. Seligson DB, Horvath S, Shi T, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature. 2005;435:1262–6. 166. Deligezer U, Yaman F, Darendeliler E, et al. Posttreatment circulating plasma BMP6 mRNA and H3K27 methylation levels discriminate metastatic prostate cancer from localized disease. Clin Chim Acta. 2010;411:1452–6. 167. Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435: 834–8. 168. Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105:10513–8. 169. Schaefer A, Jung M, Mollenkopf HJ, et al. Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma. Int J Cancer. 2010;126:1166–76. 170. Brase JC, Johannes M, Schlomm T, et al. Circulating miRNAs are correlated with tumor progression in prostate cancer. Int J Cancer J. 2011;128:608–16. 171. Hsing AW, Chokkalingam AP. Prostate cancer epidemiology. Front Biosci. 2006;11:1388–413. 172. Vanaja DK, Ballman KV, Morlan BW, et al. PDLIM4 repression by hypermethylation as a potential biomarker for prostate cancer. Clin Cancer Res. 2006;12:1128–36. 173. Chen H, Toyooka S, Gazdar AF, et al. Epigenetic regulation of a novel tumor suppressor gene (hDAB2IP) in prostate cancer cell lines. J Biol Chem. 2003;278:3121–30. 174. Duggan D, Zheng SL, Knowlton M, et al. Two genome-wide association studies of aggressive prostate cancer implicate put ative prostate tumor suppressor gene DAB2IP. J Natl Cancer Inst. 2007;99: 1836–44. 175. Wilson S, Greer B, Hooper J, et al. The membraneanchored serine protease, TMPRSS2, activates PAR-2 in prostate cancer cells. Biochem J. 2005;388:967–72. 274 176. Black PC, Mize GJ, Karlin P, et al. Overexpression of protease-activated receptors-1,-2, and-4 (PAR-1, -2, and -4) in prostate cancer. Prostate. 2007;67:743–56. 177. Omara-Opyene AL, Qiu J, Shah GV, Iczkowski KA. Prostate cancer invasion is influenced more by expression of a CD44 isoform including variant 9 than by Muc18. Lab Invest. 2004;84:894–907. 178. Aaltomaa S, Lipponen P, Viitanen J, Kankkunen JP, Ala-Opas M, Kosma VM. Prognostic value of CD44 standard, variant isoforms 3 and 6 and -catenin expression in local prostate cancer treated by radical prostatectomy. Eur Urol. 2000;38:555–62. 179. Aaltomaa S, Eskelinen M, Lipponen P. Expression of cyclin A and D proteins in prostate cancer and their relation to clinopathological variables and patient survival. Prostate. 1999;38:175–82. 180. Kallakury BV, Sheehan CE, Ambros RA, Fisher HA, Kaufman RP Jr, Ross JS. The prognostic significance of p34cdc2 and cyclinD1 protein expression in prostate adenocarcinoma. Cancer. 1997;80:753–63. 181. Nguyen TT, Nguyen CT, Gonzales FA, Nichols PW, Yu MC, Jones PA. Analysis of cyclin-dependent kinase inhibitor expression and methylation patterns in human prostate cancers. Prostate. 2000;43:233–42. 182. Bubendorf L, Kononen J, Koivisto P, et al. Survey of gene amplifications during prostate cancer progression by high-throughout fluorescence in situ hybridization on tissue microarrays. Cancer Res. 1999;59: 803–6. 183. Jenkins RB, Qian J, Lieber MM, Bostwick DG. Detection of c-myc oncogene amplification and chromosomal anomalies in metastatic prostatic carcinoma by fluorescence in situ hybridization. Cancer Res. 1997;57:524–31. 184. Bernard D, Pourtier-Manzanedo A, Gil J, Beach DH. Myc confers androgen-independent prostate cancer cell growth. J Clin Invest. 2003;112:1724–31. 185. Sato K, Qian J, Slezak JM, et al. Clinical significance of alterations of chromosome 8 in high-grade, advanced, nonmetastatic prostate carcinoma. J Natl Cancer Inst. 1999;91:1574–80. 186. Jarrard DF, Modder J, Fadden P, et al. Alterations in the p16/pRb cell cycle checkpoint occur commonly in primary and metastatic human prostate cancer. Cancer Lett. 2002;185:191–9. 187. Chakravarti A, Heydon K, Wu CL, et al. Loss of p16 expression is of prognostic significance in locally advanced prostate cancer: an analysis from the Radiation Therapy Oncology Group protocol 86-10. J Clin Oncol. 2003;21:3328–34. 188. Maddison LA, Sutherland BW, Barrios RJ, Greenberg NM. Conditional deletion of Rb causes early stage prostate cancer. Cancer Res. 2004;64:6018–25. 189. Theodorescu D, Broder SR, Boyd JC, Mills SE, Frierson HF Jr. p53, bcl-2 and retinoblastoma proteins as long-term prognostic markers in localized carcinoma of the prostate. J Urol. 1997;158:131–7. 190. Prtilo A, Leach FS, Markwalder R, et al. Tissue microarray analysis of hMSH2 expression predicts outcome in men with prostate cancer. J Urol. 2005;174:1814–8. 191. Velasco A, Albert PS, Rosenberg H, Martinez C, Leach FS. Clinicopathologic implications of hMSH2 gene T. Bhavsar, P. McCue, and R. Birbe 192. 193. 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. 205. expression and microsatellite instability in prostate cancer. Cancer Biol Ther. 2002;1:362–7. Fenic I, Franke F, Failing K, Steger K, Woenckhaus J. Expression of PTEN in malignant and non-malignant human prostate tissues: comparison with p27 protein expression. J Pathol. 2004;203:559–66. Grimberg A. Mechanisms by which IGF-I may promote cancer. Cancer Biol Ther. 2003;2:630–5. Renehan AG, Zwahlen M, Minder C, O’Dwyer ST, Shalet SM, Egger M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis. Lancet. 2004; 363:1346–53. Xiao GH, Jeffers M, Bellacosa A, Mitsuuchi Y, Vande Woude GF, Testa JR. Anti-apoptotic signaling by hepatocyte growth factor/Met via the phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase pathways. Proc Natl Acad Sci USA. 2001;98: 247–52. Dolloff NG, Shulby SS, Nelson AV, et al. Bonemetastatic potential of human prostate cancer cells correlates with Akt/PKB activation by alpha plateletderived growth factor receptor. Oncogene. 2005;24: 6848–54. Shukla S, MacLennan GT, Fu P, et al. Nuclear factorkappaB/p65 (Rel A) is constitutively activated in human prostate adenocarcinoma and correlates with disease progression. Neoplasia. 2004;6:390–400. Chen CD, Sawyers CL. NF-kappa B activates prostatespecific antigen expression and is upregulated in androgen-independent prostate cancer. Mol Cell Biol. 2002;22:2862–70. Lessard L, Begin LR, Gleave ME, Mes-Masson AM, Saad F. Nuclear localisation of nuclear factor-kappaB transcription factors in prostate cancer: an immunohistochemical study. Br J Cancer. 2005;93:1019–23. Zhang G, Zhang H, Wang Q, et al. Suppression of human prostate tumor growth by a unique prostatespecific monoclonal antibody F77 targeting a glycolipid marker. Proc Natl Acad Sci U S A. 2010;107: 732–7. Ozaki N, Ohmuraya M, Hirota M, et al. Serine protease inhibitor Kazal type 1 promotes proliferation of pancreatic cancer cells through the epidermal growth factor receptor. Mol Cancer Res. 2009;7: 1572–81. Khan AP, Poisson LM, Bhat VB, et al. Quantitative proteomic profiling of prostate cancer reveals a role for miR-128 in prostate cancer. Mol Cell Proteomics. 2010;9:298. Sreekumar A, Poisson LM, Rajendiran TM, et al. Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature. 2009;457:910. Nelson PS, Han D, Rochon Y, et al. Comprehensive analyses of prostate gene expression: convergence of expressed sequence tag databases, transcript profiling and proteomics. Electrophoresis. 2000;21: 1823–31. Meehan KL, Holland JW, Dawkins HJ. Proteomic analysis of normal and malignant prostate tissue to 275 Molecular diagnosis of prostate cancer 206. 207. 208. 209. 210. 211. 212. identify novel proteins lost in cancer. Prostate. 2002;50:54–6. Zheng Y, Xu Y, Ye B, et al. Prostate carcinoma tissue proteomics for biomarker discovery. Cancer. 2003; 98:2576–82. Rehman I, Azzouzi AR, Catto JW, et al. Proteomic analysis of voided urine after prostatic massage from patients with prostate cancer: a pilot study. Urology. 2004;64:1238–43. Grover PK, Resnick MI. High resolution twodimensional electrophoretic analysis of urinary proteins of patients with prostatic cancer. Electrophoresis. 1997;18:814–8. Petricoin EF III, Ornstein DK, Paweletz CP, et al. Serum proteomic patterns for detection of prostate cancer. J Natl Cancer Inst. 2002;94:1576–8. Qu Y, Adam BL, Yasui Y, et al. Boosted decision tree analysis of surface-enhanced laser desorption/ionization mass spectral serum profiles discriminates prostate cancer from noncancer patients. Clin Chem. 2002;48:1835–43. Lexander H, Palmberg C, Hellman U, et al. Correlation of protein expression, Gleason score and DNA ploidy in prostate cancer. Proteomics. 2006;6:4370–80. Sardana G, Jung K, Stephan C, Diamandis EP. Proteomic analysis of conditioned media from the PC3, LNCaP, and 22Rv1 prostate cancer cell lines: 213. 214. 215. 216. 217. 218. discovery and validation of candidate prostate cancer biomarkers. J Proteome Res. 2008. Rabilloud T. Two-dimensional gel electrophoresis in proteomics: old, old fashioned, but it still climbs up the mountains. Proteomics. 2002;2:3–10. Moritz RL, Ji H, Schutz F, et al. A proteome strategy for fractionating proteins and peptides using continuous free-flow electrophoresis coupled off-line to reversed-phase high-performance liquid chromatography. Anal Chem. 2004;76:4811–24. Lam YW, Mobley JA, Evans JE, et al. Mass profilingdirected isolation and identification of a stage-specific serologic protein biomarker of advanced prostate cancer. Proteomics. 2005;5:2927–38. Malik G, Ward MD, Gupta SK, et al. Serum levels of an isoform of apolipoprotein A-II as a potential marker for prostate cancer. Clin Cancer Res. 2005;11:1073–85. Le L, Chi K, Tyldesley S, et al. Identification of serum amyloid A as a biomarker to distinguish prostate cancer patients with bone lesions. Clin Chem. 2005;51:695–707. Qin S, Ferdinand AS, Richie JP, et al. Chromatofocusing fractionation and two-dimensional difference gel electrophoresis for low abundance serum proteins. Proteomics. 2005;5:3183–92.