Purpose: The aim of this study was to investigate the promoter hypermethylation as diagnostic markers to detect malignant prostate cells and as prognostic markers to predict the clinical recurrence of prostate cancer.

Experimental Design: DNA was isolated from prostate cancer and normal adjacent tissues. After bisulfite conversion, methylation of 14,495 genes was evaluated using the Methylation27 microarrays in 238 prostate tissues. We analyzed methylation profiles in four different groups: (i) tumor (n = 198) versus matched normal tissues (n = 40), (ii) recurrence (n = 123) versus nonrecurrence (n = 75), (iii) clinical recurrence (n = 80) versus biochemical recurrence (n = 43), and (iv) systemic recurrence (n = 36) versus local recurrence (n = 44). Group 1, 2, 3, and 4 genes signifying biomarkers for diagnosis, prediction of recurrence, clinical recurrence, and systemic progression were determined. Univariate and multivariate analyses were conducted to predict risk of recurrence. We validated the methylation of genes in 20 independent tissues representing each group by pyrosequencing.

Results: Microarray analysis revealed significant methylation of genes in four different groups of prostate cancer tissues. The sensitivity and specificity of methylation for 25 genes from 1, 2, and 4 groups and 7 from group 3 were shown. Validation of genes by pyrosequencing from group 1 (GSTP1, HIF3A, HAAO, and RARβ), group 2 (CRIP1, FLNC, RASGRF2, RUNX3, and HS3ST2), group 3 (PHLDA3, RASGRF2, and TNFRSF10D), and group 4 (BCL11B, POU3F3, and RASGRF2) confirmed the microarray results.

Conclusions: Our study provides a global assessment of DNA methylation in prostate cancer and identifies the significance of genes as diagnostic and progression biomarkers of prostate cancer. Clin Cancer Res; 18(10); 2882–95. ©2012 AACR.

This article is featured in Highlights of This Issue, p. 2721

Translational Relevance

We analyzed the methylation of 14,495 genes in four different sub groups of patients with prostate cancer: (i) tumor (n = 198) versus matched normal tissues (n = 40), (ii) recurrence (n = 123) versus nonrecurrence (n = 75); (iii) clinical recurrence (n = 80) versus biochemical recurrence (n = 43); and (iv) systemic recurrence (n = 36) versus local recurrence (n = 44). We confirmed the methylation of the genes by pyrosequencing in 20 independent tissues representing each group. The genes identified in group 1 represent the early detection biomarkers. These genes can increase the sensitivity and specificity of current screening and could have substantial impact to improve the cure rates when diagnosed at an early stage. Genes identified in group 2, 3, and 4 would be helpful in distinguishing indolent cancer from aggressive forms and could be used as biomarkers to identify patients whose cancer will likely recur and will need adjuvant treatments to reduce the mortality due to neoplasia.

Prostate cancer is the most frequently diagnosed cancer in men in Western countries (1). Current available therapies are surgery, radiotherapy, and androgen ablation. The response rate is as high nearly 90%; however, most of these recur or become refractory and androgen-independent (2). Existing approaches for diagnosing prostate cancer features digital rectal examination screening and serum prostate-specific antigen (PSA) determination (3). However, because of their limited sensitivity and specificity, these methods cannot reliably identify early-stage prostate cancer (4). The clinicopathologic features predictive of prostate cancer consist of Gleason score, tumor-node-metastasis (TNM) stage, surgical margin status, preoperative serum PSA levels, and GPSM (Gleason score, preoperative PSA, seminal vesicle involvement, and marginal status) score. GPSM score is a prognostic model using the weighed sum of the pathologic Gleason score, preoperative PSA, seminal vesicle involvement, and marginal status to predict biochemical progression after radical prostatectomy (5, 6). Several preoperative nomograms developed in the past decade have been based on clinicopathologic variables (6–8). These factors are helpful but far from perfect due to significant clinical heterogeneity of the disease.

Among the different types of biomarkers discovered (9), DNA methylation markers stand out for their potential to provide a unique combination of specificity, sensitivity, high information content, and applicability to a wide variety of clinical specimens (10). DNA promoter hypermethylation can lead to gene silencing (11); on the other hand, global hypomethylation, inducing genomic instability, contributes to cell transformation. Both events, either individually or in cooperation, result in the development and progression of cancer (12). Perhaps one of the most important features of DNA methylation profiling in cancer is that these profiles are both tissue- and tumor-type–specific (13). This can enable the identification of biomarkers for diagnosis, prognosis, and response to treatment and to identify potential biologic pathways which are disrupted in tumor development (14). GSTP1 is the most frequently investigated gene in prostate cancer epigenetics (15–18). Only a limited panel of genes has been identified so far, including those from our studies that are methylated at a high frequency in prostate cancer (16, 19–21). The limited value of established prognostic markers demands identification of new molecular parameters of interest in predicting the prognosis of patients with prostate cancer. We hypothesize that DNA methylation changes that occur very early during the development of cancer before the manifestation of clinical symptoms could have a substantial impact as reliable early detection biomarkers and those methylation changes that can distinguish the aggressive forms of prostate cancer which are life-threatening from indolent tumors could be established as new prognostic biomarkers for making effective treatment decisions to improve the cure rates and reduce the mortality due to neoplasia.

The objective of this study was to identify biologically and clinically relevant methylated genes as biomarkers characteristic of prostate cancer versus benign tissues, recurrent versus nonrecurrent tissues, clinical recurrent versus biochemical recurrent tissues, and systemic versus local recurrence using methylation microarrays for diagnosis and prediction of prostate cancer progression. The methylation profiles were generated from 36 systemic recurrence, 44 local recurrence, 43 biochemical recurrence, and 75 nonrecurrent patients. In addition, 40 matched normal prostatic tissues were also used. Methylation of genes that predict the risk of recurrence was analyzed by univariate and multivariate analyses in addition to the binary group comparisons. We further validated the methylation of genes in 20 independent cases of systemic recurrence, local recurrence, biochemical recurrence, and nonrecurrent patients.

Prostate tissue samples

Surgically resected prostate cancer tissue specimens were obtained from patients who had undergone radical prostatectomy for prostate cancer at Mayo Clinic, with Institutional Review Board approval. All the tissues were collected from nonpretreated patients and frozen at −80°C as described earlier (22). Hematoxylin and eosin–stained sections were prepared before and after slides were cut for DNA isolation, to ensure that tumor was present in the tissues used for analysis with minimal number of disturbing stromal cells. The pathologic status was confirmed before processing, and we included tumor samples with more than 70% cancer and excluded non-age–matched samples for methylation profiling. Cancerous regions were identified and marked in the sections. The sections were then macrodissected from the slide using a 21-gauge sterile needle to separate tumor from nontumor cells. For diagnosis of prostate cancer, case (tumor) and control (normal adjacent tissues) age-matched (birth year) samples were used. For prediction of progression, cases (tissues from recurrent patients within 5 years of prostatectomy) and controls (tissues from patients without any recurrence for 7 years following radical prostatectomy) matched on age, Gleason score, stage, and preoperative PSA were used. All patients with postoperative biochemical failure (men with initial PSA increase without further clinical progression for 5 years following radical prostatectomy, PSA level ≥ 0.2 ng/mL) were identified as PSA failure. Clinical recurrence is defined as men with local and systemic recurrence from 90 days to 5 years following the PSA increase. Local recurrence is defined on the basis of digital rectal examination, transrectal ultrasonography, and biopsy. Systemic progression is defined as evidence of metastatic disease on a bone scan. Clinical and pathologic information, including Gleason score, preoperative PSA, TNM stage, ploidy, extraprostatic extension, regional lymph node involvement, positive margins of resection, and GPSM score of patients were obtained from the Mayo SPORE prostate cancer tumor registry. Separately collected prostate tissue samples matched with the cancer tissues (obtained from the same patients) were used as matched normal controls.

DNA isolation and bisulfite conversion

Before processing for DNA isolation, quality control check of the samples was conducted to ensure that there was no DNA damage of the samples during the preservation and storage at −80°C. Genomic DNA was isolated from 10 tissue sections of 20-μm thickness using the ZR Genomic DNA II Kit (Zymo Research Corp.). Using a Zymo EZ DNA methylation kit (Zymo Research Corp.) 1.0 μg of the DNA was subjected to sodium bisulfite modification as described (16, 21, 22). Universal methylated human DNA and nonmethylated DNA (Zymo Research Corp.) were used as positive and negative controls for bisulfite modification. DNA quantity was then determined using both NanoDrop and the Agilent BioAnalyzer 2100 with DNA 1000 and 7500 chips.

Infinium methylation profiling

DNA methylation analysis was conducted using the Illumina Infinium Human Methylation27 BeadChip. Each HumanMethylation27 BeadChip consisted of 12 arrays and up to 4 bead chips were processed simultaneously. Cases and controls were randomly placed within the bead chips. The bead chip allowed the interrogation of 27,578 CpG sites representing 14,495 protein-coding gene promoters, which includes about 1,000 cancer-associated genes. The standard protocol provided by Illumina was used for DNA methylation analysis as described (23).

Statistical analysis of methylation

Raw data were imported into GenomeStudio (version 2009.1), and a methylation profile (including average β, detection P value, and signal intensities for signal A and B) was generated for each sample. The methylation status for each CpG site was measured by comparing fluorescent signal from the methylated allele with the sum of the fluorescent signal from both methylated and unmethylated alleles. Detailed quality assessment was conducted by examining the distribution of average β, number of reliably detected CpGs for each sample, principal components analysis plot, and unsupervised clustering. The differentially methylated CpGs were identified using one-way ANOVA with a batch-effect included in the model to adjust for any potential batch-effect. Analysis was first conducted between all tumors and normal samples and, subsequently, stratified analyses between tumors with recurrence and tumors without recurrence; tumors with systematic recurrence and tumors with local recurrence; and combined tumors with systematic and local recurrence and tumors with biochemical recurrence. Multiple testing was adjusted using false discovery rate q value as described (24). CpGs with a P value <0.05 and mean methylation difference between 2 contrast groups greater than 5% were initially selected as candidate markers. All statistical analyses were conducted using β-value as a continuous variable unless specified otherwise. Unsupervised and supervised hierarchical clustering analyses were conducted with the heatmap.2 function in the gplots library. Unsupervised clustering was used to characterize methylation patterns in an unbiased fashion, as conducted in other studies using methylation arrays. Supervised clustering analysis was used to further investigate methylation differences observed in unsupervised clustering. Additional evidence to support the delineation of clusters was obtained through unsupervised principal component analysis. The frequency and level of CpG methylation across different clusters were compared using a 2-sample proportion test based on both binarized and continuous β-values. The association of clinicopathologic and molecular variables with each cluster was analysed using continuous β-values and the 2-sample proportion t test. To estimate the HR of recurrence as a result of methylation change, we conducted a univariate analysis using Cox proportional hazard model. To evaluate whether the methylation can have an independent predictive value for recurrence, we incorporated Gleason score and pathologic stage into the Cox proportional model for multivariate analysis. All statistical analyses were conducted in R version 2.7.1 (The R Foundation for Statistical Computing) at 5% significance level unless otherwise stated. Where applicable, Bonferroni correction was applied to adjust for multiple testing. Differentially expressed genes were evaluated for biologic functions using Ingenuity Pathway Analysis (Ingenuity). Sensitivity and specificity of the genes were determined by receiver operator characteristic (ROC) curve analysis using the GraphPad Prism 4 Software. The area under the ROC curve and the methylation threshold yielding the optimal sensitivity and specificity were calculated for each DNA methylation marker.

Methylation analysis by pyrosequencing

Pyrosequencing assays were developed to confirm the DNA methylation of genes observed in the microarray profiling. We selected the genes from each group and validated the methylation levels in an independent set of 20 samples representing each group. One microgram of genomic DNA was bisulphite-converted as described above. For pyrosequencing, a 2-step PCR was carried out using specially designed forward and reverse primers using PyroMark Assay Design 2.0 software (Qiagen). The sequencing primers were designed to analyze the target CpG site used in the Infinium methylation microarrays. PCR primers, annealing temperature used for PCR amplification, and the sequencing primers are listed in Table 3. One of the 2 primers in the PCR amplification of the target region was biotinylated to further select the strand for pyrosequencing. PCR amplifications were carried out in a final volume of 25 μL using PyroMark PCR kit per manufacturer's procedure (25). For pyrosequencing analysis, 10 μL of the PCR product and 3.5 nmol of the target-specific sequencing primer, PyroMark Gold reagents, and the PyroMark MD instrument (Qiagen) were used following the supplier's protocol. A gene was considered as methylated if the methylation density was greater than 10%. The CpG site methylation data were obtained from 20 cases and 20 controls for each group. ROC curves were generated for each DNA methylation marker; these were summarized by area under the curve (AUC). Significance of methylation of the individual CpG site as a marker for the given group was analyzed using Student t test. P value >0.05 was considered statistically significant.

Methylation markers for diagnosis and progression of prostate cancer selected from profiling 192 prostate tissues

To determine the prevalence of methylation changes in a spectrum of prostate cancer disease ranging from indolent tumor to aggressive metastatic disease, we measured the methylation in a total of 238 samples (198 prostate cancer tissues and 40 matched normal tissues) using Illumina Infinium Methylation27 bead arrays. Clinicopathologic characteristics of 198 prostate cancer tissues are listed in Table 1. Each Human Methylation27 Bead Array enabled analysis of 12 samples. Investigation of the quantile-normalization at signal intensities of the total 238 samples visualized using a principal components analysis plot revealed 2 distinct sets of samples. Samples run on 4 microarrays (n = 46, which include 40 tumor and 6 normal) showed batch-effect from that of the other microarrays (n = 192, which include 158 tumor and 34 normal; ref. 26). The average β signal intensities from the 4 arrays were higher from the rest of the arrays. Therefore, we first analyzed the data obtained from 192 samples, then analyzed the results in the 46 samples with the batch-effect. In our first analysis, we grouped 192 prostate tissues on the basis of clinical outcome. For group 1, we compared tumor tissues (n = 158) versus normal tissues (n = 34) to identify the markers for early detection of cancer. For group 2, we compared tissues from patients with disease recurrence (n = 98) versus tissues from without disease recurrence patients (n = 60) to identify markers associated with recurrence of cancer. For group 3, we compared tissues from patients with clinical recurrence (n = 59) versus biochemical recurrence (n = 39) to distinguish increasing PSA without metastases or symptoms from metastasis. For group 4, we compared tissues from patients with systemic recurrence (n = 23) versus those with local recurrence (n = 36) to identify the markers associated with metastatic spread. The genes were initially selected using a P value <0.05 and a mean difference between 2 contrast groups greater than 5%. We further narrowed down the selection using a combination of P value and fold change in methylation between the 2 comparison groups. With a fold change cutoff of >2.0 and a P value of <8.67E-25, there are 147 genes in group 1. In groups 2, 3, and 4 with a fold change cutoff of >1.5 and with P values of <0.002, <0.05, and <0.01 respectively, there are 75, 16, and 68 genes significantly methylated. The list of genes and their major biologic functions are shown (Supplementary Table S1A–S1D). Methylation levels of 25 genes that showed increased frequency of methylation in cases compared with controls in the 1, 2, and 4 groups, and 7 genes for group 3, were depicted in the cluster diagram (Fig. 1A–D). To assess the potential use of hypermethylation of genes as molecular markers of prostate cancer, we determined the optimal sensitivity and specificity of the individual genes. The sensitivity and specificity of the 25 most differentially methylated genes for group 1, 2, and 4, and 7 genes for group 3, are shown in Table 2A–D. Genes listed in Table 2A were highly sensitive and specific to distinguishing prostate cancer from normal prostate tissues. Each of these markers had area under the ROC curve of more than 0.9 and sensitivities and specificities between 87% and 100%. Genes listed in Table 2B were methylated in prostate cancer tissues of patients with recurrence of the disease after primary therapy compared with those of patients without disease recurrence. These genes had an area under the ROC curve between 0.63 and 0.86, and sensitivities and specificities between 52% and 86%. There were fewer genes that were methylated in patients with clinical recurrence, compared with patients with only biochemical recurrence. The area under the ROC curve for these genes was between 0.66 and 0.70, with sensitivities and specificities between 47% and 84%. Finally, we assessed the genes that were methylated in patients with systemic recurrence compared with those with local recurrence. The genes with systemic recurrence showed an area under the ROC curve between 0.68 and 0.93, with sensitivities and specificities between 55% and 91%.

Table 1.

Prostate cancer tissues used for methylation microarray analysis

NonrecurrentBiochemical recurrenceLocal recurrenceSystemic recurrence
Number of patients 75 43 44 36 
Age, y 62.61 ± 8.04 64.04 ± 5.95 63.31 ± 6.03 64.41 ± 7.57 
Preoperative PSA, ng/mL 
 <4 17 (22.6) 1 (2.3) 2 (4.5) 3 (8.3) 
 4–10 45 (60) 22 (51.1) 19 (43.1) 13 (36.1) 
 >10 12 (16) 17 (39.5) 15 (34.09) 15 (13.8) 
 n.a. 19 (1.3) 3 (6.9) 8 (18.1) 5 (13.8) 
Gleason score 
 6 23 (30.6) 4 (9.3) 3 (6.8) 
 7 34 (45.3) 19 (44.1) 27 (61.3) 11 (30.5) 
 8 8 (10.6) 8 (18.6) 6 (13.6) 5 (13.8) 
 9 10 (13.3) 12 (27.9) 8 (18.1) 15 (41.6) 
 10 5 (13.8) 
TNM 
 T2a 19 (25.3) [4] 7 (16.2) [1] 11 (25) [3] 1 (2.7) 
 T2b 30 (40) [16] 14 (32.5) [3] 16 (36.3) [9] 6 (16.6) [4] 
 T3a 20 (26.6) [11] 10 (23.2) [7] 8 918.1) [4] 12 (33.3) [5] 
 T3b 6 (8) [3] 12 (27.9) [9] 7 (15.9) [3] 11 (30)[7] 
 T4 2 (4.5) [1] 6 (16.6) [4] 
Ploidy 
 Diploid 38 (50.6) 19 (44.1) 17 (38.6) 15 (41.6) 
 Tetraploid 29 (38.6) 19 (44.1) 22 (50) 13 (36.1) 
 Aneuploid 8 (10.6) 5 (11.6) 5 (11.3) 8 (22.2) 
GPSM score 
 <10 47 (62.2) 15 (34.8) 10 (22.7) 6 (16.6) 
 10–12 22 (29.3) 14 (32.5) 20 (45.4) 14 (38.8) 
 >12 5 (6.6) 11 (25.5) 6 (13.6) 12 (33.3) 
 n.a. 1 (1.3) 3 (6.9) 8 (18.2) 5 (13.8) 
PSA progression, y 2.33 ±1.77 0.77 ± 0.42 1.39 ± 1.54 
Local progression, y 2.31 ± 1.18 3.27 ± 2.99 
Systemic progression, y 2.59 ±3.41 
Follow-up, y 6.25 ± 1.48 5.87 ± 1.41 4.31 ± 1.69 4.43 ± 3.97 
Prostate death 
Other death 
NonrecurrentBiochemical recurrenceLocal recurrenceSystemic recurrence
Number of patients 75 43 44 36 
Age, y 62.61 ± 8.04 64.04 ± 5.95 63.31 ± 6.03 64.41 ± 7.57 
Preoperative PSA, ng/mL 
 <4 17 (22.6) 1 (2.3) 2 (4.5) 3 (8.3) 
 4–10 45 (60) 22 (51.1) 19 (43.1) 13 (36.1) 
 >10 12 (16) 17 (39.5) 15 (34.09) 15 (13.8) 
 n.a. 19 (1.3) 3 (6.9) 8 (18.1) 5 (13.8) 
Gleason score 
 6 23 (30.6) 4 (9.3) 3 (6.8) 
 7 34 (45.3) 19 (44.1) 27 (61.3) 11 (30.5) 
 8 8 (10.6) 8 (18.6) 6 (13.6) 5 (13.8) 
 9 10 (13.3) 12 (27.9) 8 (18.1) 15 (41.6) 
 10 5 (13.8) 
TNM 
 T2a 19 (25.3) [4] 7 (16.2) [1] 11 (25) [3] 1 (2.7) 
 T2b 30 (40) [16] 14 (32.5) [3] 16 (36.3) [9] 6 (16.6) [4] 
 T3a 20 (26.6) [11] 10 (23.2) [7] 8 918.1) [4] 12 (33.3) [5] 
 T3b 6 (8) [3] 12 (27.9) [9] 7 (15.9) [3] 11 (30)[7] 
 T4 2 (4.5) [1] 6 (16.6) [4] 
Ploidy 
 Diploid 38 (50.6) 19 (44.1) 17 (38.6) 15 (41.6) 
 Tetraploid 29 (38.6) 19 (44.1) 22 (50) 13 (36.1) 
 Aneuploid 8 (10.6) 5 (11.6) 5 (11.3) 8 (22.2) 
GPSM score 
 <10 47 (62.2) 15 (34.8) 10 (22.7) 6 (16.6) 
 10–12 22 (29.3) 14 (32.5) 20 (45.4) 14 (38.8) 
 >12 5 (6.6) 11 (25.5) 6 (13.6) 12 (33.3) 
 n.a. 1 (1.3) 3 (6.9) 8 (18.2) 5 (13.8) 
PSA progression, y 2.33 ±1.77 0.77 ± 0.42 1.39 ± 1.54 
Local progression, y 2.31 ± 1.18 3.27 ± 2.99 
Systemic progression, y 2.59 ±3.41 
Follow-up, y 6.25 ± 1.48 5.87 ± 1.41 4.31 ± 1.69 4.43 ± 3.97 
Prostate death 
Other death 

NOTE: Mean ± S.D are shown for age, years for PSA, local, and systemic progression and follow-up years of patients with prostate cancer. Value in parentheses indicates the percentage of cases representing the group. Value in square brackets indicates the node-positive cases. Matched normal tissues from 40 patients [age (mean ± SD) = 63.15 ± 7.28] were used.

Abbreviation: n.a., not available.

Table 2.

Significance of differentially methylated genes in prostate cancer tissues, analyzed using Infinium Methylation27 BeadChips

GeneSensitivity (95% CI)(95% CI)AUCt Test (P)Transcription start siteStrandCpG island location
A, Genes that are significantly methylated in prostate cancer tissues (n = 158) compared with normal prostate (n = 34)a 
 A0X1 96.2 (91.9–98.5) 100 (89.7–100) 0.992 <0.0001 41 2: 201,158,654–201,159,798 
 CYBA 95.5 (91.1–98.2) 97.1 (84.7–99.9) 0.988 <0.0001 83 − 16: 87,244,422–87,245,165 
 EDG3 93.0 (87.9–96.4) 97.1 (84.7 to 99.9) 0.97 <0.0001 363 9: 90,794,956–90,796,827 
 ELF4 88.6 (82.6–93.1) 100 (89.7–100) 0.994 <0.0001 600 − X: 129,071,186–129,073,330 
 EPB41L3 93.7 (88.7–96.9) 100 (89.7–100) 0.978 <0.0001 185 − 18: 5,532,741–5,534,303 
 FLJ12056 94.3 (89.5–97.4) 100 (89.7–100) 0.978 <0.0001 316 2: 71,058,922–71,060,108 
 FLJ90650 96.2 (91.9–98.6) 100 (89.7–100) 0.994 <0.0001 449 5: 115,324,903–115,327,577 
 FLT4 99.4 (96.5–99.9) 100 (89.7–100) 0.999 <0.0001 691 − 5: 180,008,152–180,010,059 
 GAS6 96.2 (91.9–98.6) 100 (89.7–100) 0.99 <0.0001 964 13: 113,546,578–113,548,278 
 GRASP 96.2 (91.9–98.6) 100 (89.7–100) 0.993 <0.0001 159 12: 50,686,496–50,688,060 
 GSTP1 91.8 (86.3–95.6) 100 (89.7–100) 0.995 <0.0001 755 11: 67,106,980–67,108,634 
 HAAO 93.0 (87.9–96.5) 100 (89.7–100) 0.979 <0.0001 454 − 2: 42,873,041–42,874,496 
 HIF3A 95.6 (91.1–98.2) 100 (89.7–100) 0.989 <0.0001 153 19: 51,491,754–51,492,473 
 HOXC11 89.2 (83.3–93.6) 97.1 (84.7–99.9) 0.985 <0.0001 20 12: 52,652,893–52,656,105 
 LEP 98.1 (94.6–99.6) 97.1 (84.7–99.9) 0.997 <0.0001 51 7: 127,667,928–127,668,724 
 MGC39606 95.6 (91.1–98.2) 100 (89.7–100) 0.992 <0.0001 634 X: 134,383,127–134,384,321 
 MOBKL2B 92.4 (87.1–96.0) 100 (89.7–100) 0.97 <0.0001 851 − 9: 27,517,915–27,520,017 
 RAB34 91.8 (86.3–95.6) 94.1 (80.3–99.3) 0.976 <0.0001 157 − 17: 24,068,248–24,069,280 
 RARβ 93.0 (87.9–96.5) 100 (89.7–100) 0.986 <0.0001 352 3: 25,444,208–25,445,101 
 RHCG 95.6 (91.1–98.2) 100 (89.7–100) 0.993 <0.0001 110 − 15: 87,840,274–87,841,084 
 RND2 93.0 (87.9–96.5) 100 (89.7–100) 0.996 <0.0001 180 17: 38,429,704–38,431,262 
 SLC34A2 91.1 (85.6–95.1) 94.1 (80.3–99.3) 0.966 <0.0001 44 4: 25,266,077–25,266,795 
 SPATA6 95.6 (91.1–98.2) 100 (89.7–100) 0.991 <0.0001 595 − 1: 48,709,694–48,710,842 
 TPM4 94.9 (90.3–97.8) 100 (89.7–100) 0.977 <0.0001 306 19: 16,047,744–16,049,758 
 ZNF154 97.5 (93.6–99.3) 94.1 (80.3–99.3) 0.99 <0.0001 100 − 19: 62,911,404–62,912,681 
B, Hypermethylated genes in tissues of patients with recurrent prostate cancer (n = 98) compared with nonrecurrent cancer (n = 60)b 
 ACTL6B 65.3 (55.1–74.6) 66.7 (53.3–78.3) 0.702 <0.0001 136 − 7: 100,091,590–100,092,362 
 AEBP1 63.3 (52.9–72.8) 60.0 (46.5–72.4) 0.646 <0.0021 38 7: 44,110,152–44,111,361 
 AMID 77.5 (68.0–85.4) 76.7 (64.0–86.6) 0.843 <0.0001 25 − 10: 71,562,005–71,562,897 
 CD8A 75.5 (65.8–83.6) 71.7 (58.6–82.5) 0.829 <0.0001 114 − 2: 86,869,360–86,871,837 
 CRIP1 61.2 (50.8–70.9) 60.0 (46.5–72.4) 0.638 <0.0037 93 14: 105,023,580–105,025,363 
 FLJ30934 80.6 (71.4–87.9) 78.3 (65.8–87.9) 0.862 <0.0001 82 11: 65,357,151–65,358,255 
 FLNC 79.6 (70.3–87.1) 81.7 (69.6–90.5) 0.864 <0.0001 131 7: 128,257,322–128,258,463 
 FMOD 76.5 (66.9–84.5) 68.3 (55.0–79.7) 0.748 <0.0001 97 − 1: 201,586,895–201,587,112 
 FOXE3 65.3 (55.0–74.6) 63.3 (49.9–75.4) 0.697 <0.0001 570 1: 47,654,351–47,655,785 
 GAS7 66.3 (56.1–75.6) 66.7 (53.3–78.3) 0.744 <0.0001 395 − 17: 10,041,659–10,043,517 
 GDPD5 70.4 (60.3–79.2) 65.4 (51.6–76.8) 0.713 <0.0001 408 − 11: 74,913,525–74,915,608 
 HS3ST2 65.3 (55.0–74.6) 75.0 (62.1–85.3) 0.798 <0.0001 257 16: 22,732,005–22,734,135 
 LOC349136 67.3 (57.1–76.5) 65.0 (51.6–76.9) 0.742 <0.0001 479 − 7: 150,736,827–150,739,238 
 NEUROG1 66.3 (56.1–75.6) 70.0 (56.8–81.2) 0.731 <0.0001 19 − 5: 134,898,284–134,900,130 
 PLTP 68.4 (58.2–77.4) 75.1 (62.1–85.3) 0.709 <0.0001 165 − 20: 43,972,342–43,974,350 
 PTGER2 66.3 (56.1–75.6) 66.7 (53.3–78.3) 0.739 <0.0001 199 14: 51,850,265–51,852,038 
 RASGRF2 69.4 (59.3–78.3) 71.7 (58.6–82.5) 0.763 <0.0001 106 5: 80,291,476–80,292,927 
 RUNX3 67.4 (57.1–76.5) 71.7 (58.6–82.6) 0.746 <0.0001 528 − 1: 25,127,692–25,131,906 
 SIX6 65.3 (55.0–74.6) 68.3 (55.0–79.7) 0.745 <0.0001 185 14: 60,045,112–60,046,647 
 SLC9A3 69.4 (59.3–78.3) 70.0 (56.8–81.2) 0.713 <0.0001 178 − 5: 576,269–577,990 
 SPSB4 62.2 (51.9–71.8) 61.7 (48.2–73.9) 0.658 <0.0008 91 3: 142,252,136–142,254,676 
 SRD5A2 70.4 (60.3–79.2) 71.7 (58.6–82.5) 0.748 <0.0001 125 − 2: 31,658,633–31,660,643 
 SUSD3 64.3 (54.0–73.7) 63.3 (49.9–75.4) 0.652 <0.0014 560 9: 94,860,370–94,861,840 
 SYT10 66.3 (56.1–75.6) 65.4 (51.6–76.8) 0.714 <0.0001 254 − 12: 33,482,438–33,484,467 
 TMEM74 66.3 (56.1–75.6) 68.3 (55.0–79.7) 0.664 <0.0005 14 − 8: 109,868,266–109,869,213 
C, Significance of genes methylated in prostate cancer tissues of patients with clinical recurrence (n = 59) compared with patients with only biochemical recurrence (n = 39)c 
 CHST7 69.5 (56.1–80.8) 64.1 (47.2–78.8) 0.668 <0.0050 255 X: 46,317,905–46,319,911 
 LMX1B 71.2 (57.9–82.2) 64.1 (47.2–78.8) 0.684 <0.0021 106 9: 128,412,539–128,418,074 
 PHLDA 3 61.0 (47.4–73.4) 59.0 (42.1–74.4) 0.606 <0.0772 348 − 1: 199,704,096–199,705,294 
 RAFTLIN 62.7 (49.1–75.0) 64.1 (47.2–78.8) 0.668 <0.0050 89 − 3: 16,529,156–16,530,542 
 RASGRF2 66.1 (52.6–77.9) 69.2 (52.4–82.9) 0.690 <0.0015 106 5: 80,291,476–80,292,927 
 TNFRSF10D 62.7 (49.2–74.9) 66.6 (49.7–80.9) 0.700 <0.0008 25 − 8: 23,076,422–23,077,641 
 ZNF135 59.3 (45.7–71.9) 61.5 (44.6–76.6) 0.664 <0.0061 144 19: 63,262,091–63,264,376 
D, Genes that are methylated in tissues of patients with systemic recurrence (n = 23) compared with local recurrence (n = 36)d 
 ALPL 65.2 (42.7–83.6) 63.8 (46.2–79.1) 0.683 <0.0182 269 1: 21,707,968–21,709,033 
 AMPH 65.2 (42.7–83.6) 75.0 (57.8–87.8) 0.77 <0.0005 35 − 7: 38,636,784–38,637,861 
 BCDIN3 78.2 (56.3–92.5) 83.3 (67.1–93.6) 0.861 <0.0001 873 7: 99,863,663–99,866,414 
 BCL11B 82.6 (61.2–95.0) 80.5 (63.9–91.8) 0.797 <0.0001 274 − 14: 98,806,507–98,807,587 
 BRD4 73.9 (51.6–89.7) 69.4 (51.8–83.6) 0.724 <0.0038 665 − 19: 15,252,811–15,253,031 
 C18orf34 78.2 (56.3–92.5) 77.7 (60.8–89.8) 0.806 <0.0001 174 − 18: 29,274,320–29,275,128 
 DCAMKL1 82.6 (61.2–95.5) 91.6 (77.5–98.2) 0.874 <0.0001 734 − 13: 35,602,437–35,603,802 
 FGF5 91.3 (71.9–98.9) 83.3 (67.1–93.6) 0.911 <0.0001 544 4: 81,405,716–81,407,598 
 FLJ42486 82.6 (61.2–95.0) 77.7 (60.8–89.8) 0.834 <0.0001 99 − 14: 104,141,516–104,142,673 
 JAM2 86.9 (66.4–97.2) 86.1 (70.5–95.3) 0.936 <0.0001 587 21: 25,933,195–25,934,679 
 LHX9 82.6 (61.2–95.0) 69.4 (51.8–83.6) 0.822 <0.0001 106 1: 196,146,152–196,148,467 
 LOC283537 78.2 (56.3–92.5) 75.0 (57.8–87.8) 0.811 <0.0001 363 − 13: 28,190,499–28,191,650 
 LRAT 65.2 (42.7–83.6) 72.2 (54.8–85.8) 0.727 <0.0034 462 4: 155,882,485–155,885,476 
 PDE4B 91.3 (71.9–98.9) 77.7 (60.8–89.8) 0.856 <0.0001 423 1: 66,030,397–66,031,906 
 POU3F3 91.3 (71.9–98.9) 75.2 (57.8–87.8) 0.865 <0.0001 248 2: 104,835,039–104,840,521 
 PTGS2 60.8 (38.5–80.2) 80.5 (63.9–91.8) 0.688 <0.0153 37 − 1: 184,915,751–184,916,808 
 RASGRF2 73.9 (51.6–89.7) 75.4 (57.8–87.8) 0.788 <0.0002 106 5: 80,291,476–80,292,927 
 SLC27A6 78.2 (56.3–92.5) 69.4 (51.8–83.6) 0.742 <0.0017 22 5: 128,328,505–128,329,501 
 SLC03A1 73.9 (51.6–89.7) 83.3 (67.1–93.3) 0.797 <0.0001 333 15: 90,196,981–90,198,831 
 SPSB4 82.6 (61.2–95.0) 80.6 (63.9–91.8) 0.824 <0.0001 91 3: 142,252,136–142,254,676 
 STAT3 69.5 (47.0–86.7) 69.4 (51.8–83.6) 0.744 <0.0017 757 − 17: 37,794,547–37,794,828 
 SYN2 73.9 (51.6–89.7) 80.5 (63.9–91.8) 0.809 <0.0001 406 3: 12,020,118–12,021,768 
 TACR3 69.5 (47.0–86.7) 72.2 (54.8–85.8) 0.704 <0.0086 346 − 4: 104,860,449–104,860,859 
 TIRAP 73.9 (51.6–89.7) 69.4 (51.9–83.6) 0.779 <0.0003 645 11: 125,657,511–125,658,777 
 WNT11 65.22 (42.7–83.6) 63.9 (46.2–79.2) 0.753 <0.0011 221 − 11: 75,594,610–75,600,053 
GeneSensitivity (95% CI)(95% CI)AUCt Test (P)Transcription start siteStrandCpG island location
A, Genes that are significantly methylated in prostate cancer tissues (n = 158) compared with normal prostate (n = 34)a 
 A0X1 96.2 (91.9–98.5) 100 (89.7–100) 0.992 <0.0001 41 2: 201,158,654–201,159,798 
 CYBA 95.5 (91.1–98.2) 97.1 (84.7–99.9) 0.988 <0.0001 83 − 16: 87,244,422–87,245,165 
 EDG3 93.0 (87.9–96.4) 97.1 (84.7 to 99.9) 0.97 <0.0001 363 9: 90,794,956–90,796,827 
 ELF4 88.6 (82.6–93.1) 100 (89.7–100) 0.994 <0.0001 600 − X: 129,071,186–129,073,330 
 EPB41L3 93.7 (88.7–96.9) 100 (89.7–100) 0.978 <0.0001 185 − 18: 5,532,741–5,534,303 
 FLJ12056 94.3 (89.5–97.4) 100 (89.7–100) 0.978 <0.0001 316 2: 71,058,922–71,060,108 
 FLJ90650 96.2 (91.9–98.6) 100 (89.7–100) 0.994 <0.0001 449 5: 115,324,903–115,327,577 
 FLT4 99.4 (96.5–99.9) 100 (89.7–100) 0.999 <0.0001 691 − 5: 180,008,152–180,010,059 
 GAS6 96.2 (91.9–98.6) 100 (89.7–100) 0.99 <0.0001 964 13: 113,546,578–113,548,278 
 GRASP 96.2 (91.9–98.6) 100 (89.7–100) 0.993 <0.0001 159 12: 50,686,496–50,688,060 
 GSTP1 91.8 (86.3–95.6) 100 (89.7–100) 0.995 <0.0001 755 11: 67,106,980–67,108,634 
 HAAO 93.0 (87.9–96.5) 100 (89.7–100) 0.979 <0.0001 454 − 2: 42,873,041–42,874,496 
 HIF3A 95.6 (91.1–98.2) 100 (89.7–100) 0.989 <0.0001 153 19: 51,491,754–51,492,473 
 HOXC11 89.2 (83.3–93.6) 97.1 (84.7–99.9) 0.985 <0.0001 20 12: 52,652,893–52,656,105 
 LEP 98.1 (94.6–99.6) 97.1 (84.7–99.9) 0.997 <0.0001 51 7: 127,667,928–127,668,724 
 MGC39606 95.6 (91.1–98.2) 100 (89.7–100) 0.992 <0.0001 634 X: 134,383,127–134,384,321 
 MOBKL2B 92.4 (87.1–96.0) 100 (89.7–100) 0.97 <0.0001 851 − 9: 27,517,915–27,520,017 
 RAB34 91.8 (86.3–95.6) 94.1 (80.3–99.3) 0.976 <0.0001 157 − 17: 24,068,248–24,069,280 
 RARβ 93.0 (87.9–96.5) 100 (89.7–100) 0.986 <0.0001 352 3: 25,444,208–25,445,101 
 RHCG 95.6 (91.1–98.2) 100 (89.7–100) 0.993 <0.0001 110 − 15: 87,840,274–87,841,084 
 RND2 93.0 (87.9–96.5) 100 (89.7–100) 0.996 <0.0001 180 17: 38,429,704–38,431,262 
 SLC34A2 91.1 (85.6–95.1) 94.1 (80.3–99.3) 0.966 <0.0001 44 4: 25,266,077–25,266,795 
 SPATA6 95.6 (91.1–98.2) 100 (89.7–100) 0.991 <0.0001 595 − 1: 48,709,694–48,710,842 
 TPM4 94.9 (90.3–97.8) 100 (89.7–100) 0.977 <0.0001 306 19: 16,047,744–16,049,758 
 ZNF154 97.5 (93.6–99.3) 94.1 (80.3–99.3) 0.99 <0.0001 100 − 19: 62,911,404–62,912,681 
B, Hypermethylated genes in tissues of patients with recurrent prostate cancer (n = 98) compared with nonrecurrent cancer (n = 60)b 
 ACTL6B 65.3 (55.1–74.6) 66.7 (53.3–78.3) 0.702 <0.0001 136 − 7: 100,091,590–100,092,362 
 AEBP1 63.3 (52.9–72.8) 60.0 (46.5–72.4) 0.646 <0.0021 38 7: 44,110,152–44,111,361 
 AMID 77.5 (68.0–85.4) 76.7 (64.0–86.6) 0.843 <0.0001 25 − 10: 71,562,005–71,562,897 
 CD8A 75.5 (65.8–83.6) 71.7 (58.6–82.5) 0.829 <0.0001 114 − 2: 86,869,360–86,871,837 
 CRIP1 61.2 (50.8–70.9) 60.0 (46.5–72.4) 0.638 <0.0037 93 14: 105,023,580–105,025,363 
 FLJ30934 80.6 (71.4–87.9) 78.3 (65.8–87.9) 0.862 <0.0001 82 11: 65,357,151–65,358,255 
 FLNC 79.6 (70.3–87.1) 81.7 (69.6–90.5) 0.864 <0.0001 131 7: 128,257,322–128,258,463 
 FMOD 76.5 (66.9–84.5) 68.3 (55.0–79.7) 0.748 <0.0001 97 − 1: 201,586,895–201,587,112 
 FOXE3 65.3 (55.0–74.6) 63.3 (49.9–75.4) 0.697 <0.0001 570 1: 47,654,351–47,655,785 
 GAS7 66.3 (56.1–75.6) 66.7 (53.3–78.3) 0.744 <0.0001 395 − 17: 10,041,659–10,043,517 
 GDPD5 70.4 (60.3–79.2) 65.4 (51.6–76.8) 0.713 <0.0001 408 − 11: 74,913,525–74,915,608 
 HS3ST2 65.3 (55.0–74.6) 75.0 (62.1–85.3) 0.798 <0.0001 257 16: 22,732,005–22,734,135 
 LOC349136 67.3 (57.1–76.5) 65.0 (51.6–76.9) 0.742 <0.0001 479 − 7: 150,736,827–150,739,238 
 NEUROG1 66.3 (56.1–75.6) 70.0 (56.8–81.2) 0.731 <0.0001 19 − 5: 134,898,284–134,900,130 
 PLTP 68.4 (58.2–77.4) 75.1 (62.1–85.3) 0.709 <0.0001 165 − 20: 43,972,342–43,974,350 
 PTGER2 66.3 (56.1–75.6) 66.7 (53.3–78.3) 0.739 <0.0001 199 14: 51,850,265–51,852,038 
 RASGRF2 69.4 (59.3–78.3) 71.7 (58.6–82.5) 0.763 <0.0001 106 5: 80,291,476–80,292,927 
 RUNX3 67.4 (57.1–76.5) 71.7 (58.6–82.6) 0.746 <0.0001 528 − 1: 25,127,692–25,131,906 
 SIX6 65.3 (55.0–74.6) 68.3 (55.0–79.7) 0.745 <0.0001 185 14: 60,045,112–60,046,647 
 SLC9A3 69.4 (59.3–78.3) 70.0 (56.8–81.2) 0.713 <0.0001 178 − 5: 576,269–577,990 
 SPSB4 62.2 (51.9–71.8) 61.7 (48.2–73.9) 0.658 <0.0008 91 3: 142,252,136–142,254,676 
 SRD5A2 70.4 (60.3–79.2) 71.7 (58.6–82.5) 0.748 <0.0001 125 − 2: 31,658,633–31,660,643 
 SUSD3 64.3 (54.0–73.7) 63.3 (49.9–75.4) 0.652 <0.0014 560 9: 94,860,370–94,861,840 
 SYT10 66.3 (56.1–75.6) 65.4 (51.6–76.8) 0.714 <0.0001 254 − 12: 33,482,438–33,484,467 
 TMEM74 66.3 (56.1–75.6) 68.3 (55.0–79.7) 0.664 <0.0005 14 − 8: 109,868,266–109,869,213 
C, Significance of genes methylated in prostate cancer tissues of patients with clinical recurrence (n = 59) compared with patients with only biochemical recurrence (n = 39)c 
 CHST7 69.5 (56.1–80.8) 64.1 (47.2–78.8) 0.668 <0.0050 255 X: 46,317,905–46,319,911 
 LMX1B 71.2 (57.9–82.2) 64.1 (47.2–78.8) 0.684 <0.0021 106 9: 128,412,539–128,418,074 
 PHLDA 3 61.0 (47.4–73.4) 59.0 (42.1–74.4) 0.606 <0.0772 348 − 1: 199,704,096–199,705,294 
 RAFTLIN 62.7 (49.1–75.0) 64.1 (47.2–78.8) 0.668 <0.0050 89 − 3: 16,529,156–16,530,542 
 RASGRF2 66.1 (52.6–77.9) 69.2 (52.4–82.9) 0.690 <0.0015 106 5: 80,291,476–80,292,927 
 TNFRSF10D 62.7 (49.2–74.9) 66.6 (49.7–80.9) 0.700 <0.0008 25 − 8: 23,076,422–23,077,641 
 ZNF135 59.3 (45.7–71.9) 61.5 (44.6–76.6) 0.664 <0.0061 144 19: 63,262,091–63,264,376 
D, Genes that are methylated in tissues of patients with systemic recurrence (n = 23) compared with local recurrence (n = 36)d 
 ALPL 65.2 (42.7–83.6) 63.8 (46.2–79.1) 0.683 <0.0182 269 1: 21,707,968–21,709,033 
 AMPH 65.2 (42.7–83.6) 75.0 (57.8–87.8) 0.77 <0.0005 35 − 7: 38,636,784–38,637,861 
 BCDIN3 78.2 (56.3–92.5) 83.3 (67.1–93.6) 0.861 <0.0001 873 7: 99,863,663–99,866,414 
 BCL11B 82.6 (61.2–95.0) 80.5 (63.9–91.8) 0.797 <0.0001 274 − 14: 98,806,507–98,807,587 
 BRD4 73.9 (51.6–89.7) 69.4 (51.8–83.6) 0.724 <0.0038 665 − 19: 15,252,811–15,253,031 
 C18orf34 78.2 (56.3–92.5) 77.7 (60.8–89.8) 0.806 <0.0001 174 − 18: 29,274,320–29,275,128 
 DCAMKL1 82.6 (61.2–95.5) 91.6 (77.5–98.2) 0.874 <0.0001 734 − 13: 35,602,437–35,603,802 
 FGF5 91.3 (71.9–98.9) 83.3 (67.1–93.6) 0.911 <0.0001 544 4: 81,405,716–81,407,598 
 FLJ42486 82.6 (61.2–95.0) 77.7 (60.8–89.8) 0.834 <0.0001 99 − 14: 104,141,516–104,142,673 
 JAM2 86.9 (66.4–97.2) 86.1 (70.5–95.3) 0.936 <0.0001 587 21: 25,933,195–25,934,679 
 LHX9 82.6 (61.2–95.0) 69.4 (51.8–83.6) 0.822 <0.0001 106 1: 196,146,152–196,148,467 
 LOC283537 78.2 (56.3–92.5) 75.0 (57.8–87.8) 0.811 <0.0001 363 − 13: 28,190,499–28,191,650 
 LRAT 65.2 (42.7–83.6) 72.2 (54.8–85.8) 0.727 <0.0034 462 4: 155,882,485–155,885,476 
 PDE4B 91.3 (71.9–98.9) 77.7 (60.8–89.8) 0.856 <0.0001 423 1: 66,030,397–66,031,906 
 POU3F3 91.3 (71.9–98.9) 75.2 (57.8–87.8) 0.865 <0.0001 248 2: 104,835,039–104,840,521 
 PTGS2 60.8 (38.5–80.2) 80.5 (63.9–91.8) 0.688 <0.0153 37 − 1: 184,915,751–184,916,808 
 RASGRF2 73.9 (51.6–89.7) 75.4 (57.8–87.8) 0.788 <0.0002 106 5: 80,291,476–80,292,927 
 SLC27A6 78.2 (56.3–92.5) 69.4 (51.8–83.6) 0.742 <0.0017 22 5: 128,328,505–128,329,501 
 SLC03A1 73.9 (51.6–89.7) 83.3 (67.1–93.3) 0.797 <0.0001 333 15: 90,196,981–90,198,831 
 SPSB4 82.6 (61.2–95.0) 80.6 (63.9–91.8) 0.824 <0.0001 91 3: 142,252,136–142,254,676 
 STAT3 69.5 (47.0–86.7) 69.4 (51.8–83.6) 0.744 <0.0017 757 − 17: 37,794,547–37,794,828 
 SYN2 73.9 (51.6–89.7) 80.5 (63.9–91.8) 0.809 <0.0001 406 3: 12,020,118–12,021,768 
 TACR3 69.5 (47.0–86.7) 72.2 (54.8–85.8) 0.704 <0.0086 346 − 4: 104,860,449–104,860,859 
 TIRAP 73.9 (51.6–89.7) 69.4 (51.9–83.6) 0.779 <0.0003 645 11: 125,657,511–125,658,777 
 WNT11 65.22 (42.7–83.6) 63.9 (46.2–79.2) 0.753 <0.0011 221 − 11: 75,594,610–75,600,053 

NOTE: Performance of the genes was analyzed by AUC of the genes for the 2 contrast groups. Sensitivity and specificity are reported for the threshold that minimized sensitivity–specificity. The P value indicates the statistical significance. Location of the CpG site used for analysis is given with respect to the distance from the transcription start site. The CpG island location on the respective chromosome is shown.

Abbreviation: CI, confidence interval.

at test P value is obtained by comparing tumor (n = 158) and benign tissues (n = 34).

bt test P value is obtained by comparing prostate cancer tissues of patients with recurrence (n = 98) and without recurrence (n = 60).

ct test P value is obtained by comparing prostate cancer tissues of patients with clinical recurrence (n = 59) and biochemical recurrence (n = 39).

dt test P value is obtained by comparing prostate cancer tissues of patients with systemic recurrence (n = 23) and local recurrence (n = 36).

Figure 1.

Methylation of genes in prostate cancer tissues, analyzed using Infinium Methylation27 BeadChips. Cluster diagram depicting genes that distinguish 2 contrast groups. A, genes that are significantly methylated in prostate cancer tissues (n = 158) compared with normal prostate (n = 34); normal (N), nonrecurrence (NR), biochemical recurrence (BR), local recurrence (LR), and systemic recurrence (SR) prostate cancer tissues. B, hypermethylated genes in tissues of patients with recurrent prostate cancer (n = 98) compared with nonrecurrent patients (n = 60). C, significance of genes methylated in prostate cancer tissues of patients with clinical recurrence (n = 59) compared with patients with only biochemical recurrence (n = 39). D, genes that are methylated in tissues of patients with systemic recurrence (n = 23) compared with local recurrence (n = 36). Each row represents a gene and each column a tissue sample. Methylation is color scaled from white to black such that white represents zero (no methylation detected) and black represents more than 99% (input DNA is methylated), respectively, relatively to the median of the reference pool. Color saturation is proportional to the magnitude of the difference from the mean. Each gene is labeled by its gene name.

Figure 1.

Methylation of genes in prostate cancer tissues, analyzed using Infinium Methylation27 BeadChips. Cluster diagram depicting genes that distinguish 2 contrast groups. A, genes that are significantly methylated in prostate cancer tissues (n = 158) compared with normal prostate (n = 34); normal (N), nonrecurrence (NR), biochemical recurrence (BR), local recurrence (LR), and systemic recurrence (SR) prostate cancer tissues. B, hypermethylated genes in tissues of patients with recurrent prostate cancer (n = 98) compared with nonrecurrent patients (n = 60). C, significance of genes methylated in prostate cancer tissues of patients with clinical recurrence (n = 59) compared with patients with only biochemical recurrence (n = 39). D, genes that are methylated in tissues of patients with systemic recurrence (n = 23) compared with local recurrence (n = 36). Each row represents a gene and each column a tissue sample. Methylation is color scaled from white to black such that white represents zero (no methylation detected) and black represents more than 99% (input DNA is methylated), respectively, relatively to the median of the reference pool. Color saturation is proportional to the magnitude of the difference from the mean. Each gene is labeled by its gene name.

Close modal

Risk of recurrence was estimated for methylation change by univariate analysis using Cox proportional hazard model by pooling all recurrence types [pooled (n = 98), which include biochemical (n = 39), local (n = 36), and systemic recurrence (n = 23); as the survival curves were almost similar for the three recurrence types]. The event was prostate cancer recurrence and those without the event (nonrecurrence, n = 60) were censored at the time of last follow-up. The methylation average β-values were transformed to the scale of 1 to 20 by multiplying with 20 so that each unit of increment is equivalent to an increase of 5% methylation ratio. With a P value of <9.0E-05, the top 128 genes associated with recurrence from the univariate analysis are shown (Supplementary Table S2). It is intriguing to find that there are 17 of 25 genes presented in the Table 2B from the binary recurrence versus nonrecurrence analysis that are among the 128 genes from the univariate Cox model of analysis. These findings suggest that our binary analysis includes most of the genes associated with recurrence. To determine whether these methylation changes could have an independent predictive value for recurrence, we incorporated Gleason score and pathologic stage into the above mentioned Cox model and conducted multivariate analysis. With a P value <9.0E-05, there are 183 genes that predict recurrence risk (Supplementary Table S3). Similar to the univariate analysis, we found that 16 of 25 genes presented in the Table 2B from the binary recurrence versus nonrecurrence analysis are among the 183 genes from the multivariate Cox model of analysis which indicates that these genes could be independent predictors of recurrence regardless of Gleason score and pathologic stage. Taken together, our data suggest that methylation of these genes could play a vital role in prostate cancer progression.

Analysis of the methylation markers in separate batch of 46 samples

Because of the batch-effects observed in the 4 microarrays containing 46 samples, we analyzed these separately. There were 6 matched normal, 15 nonrecurrent, 4 biochemical recurrent, 8 local recurrent, and 13 systemic recurrent cases in these samples. Because of the small number of tissues representing different groups, the data obtained from these 46 samples were used to analyze the sensitivity and specificity of the genes shown in Table 2A–D. These results were almost similar to those seen with the batch of the above 192 samples (Supplementary Table S4A–S4D]).

Validation of methylation in different groups by pyrosequencing in independent samples

Because of the impracticability of validating all genes identified in the microarray, we focused on 14 genes to confirm their methylation in different groups of prostate cancer tissues. These genes were selected because of their increased frequency of methylation in cases compared with the controls in the respective groups. To confirm the methylation data, we further validated the methylation of genes in an independent set of 20 samples representing each group by pyrosequencing method. These samples were matched to the samples used in microarray analysis based on the age, PSA levels, Gleason score, TNM stage, and GPSM score. Clinicopathologic characteristics of a total of 80 prostate cancer tissues, which included 20 from each group, are listed in Supplementary Table S5. The bisulfite-converted DNA was used for PCR amplification using specific primers, and the PCR product was analyzed by pyrosequencing using gene-specific sequencing primers (Table 3). We selected the genes HIF3A, HAAO, RARβ, and GSTP1 for group 1; CRIP1, RUNX3, HS3ST2, FLNC, and RASGRF2 for group 2; PHLDA3, TNFRSF10D, RASGRF2, and ZNF135 for group 3; and BCL11B, POU3F3, and RASGRF2 for group 4. The same CpG site of the gene used in the microarray experiments was selected as the target CpG site for all the genes except RUNX3 and FLNC, to design the PCR primers and sequencing primers for pyrosequencing using the PSQ Assay Design 2.0 Software. For the genes RUNX3 and FLNC, PyroMark CpG assays (Qiagen) were used because of the difficulty in designing the primers for the target site used in microarray experiments by the PSQ Assay Design. Pyrosequencing was conducted in triplicates for each sample, and the representative pyrogram of the genes from each group of cases was shown (Fig. 2A–D). We determined the optimal sensitivity and specificity of methylation by ROC analysis. This analysis revealed that HIF3A, HAAO, RARβ, and GSTP1 had ROC AUC of greater than 0.99, with sensitivities and specificities of 84% to 100% for detecting prostate cancer. CRIP1, RUNX3, HS3ST2, FLNC, and RASGRF2 had AUC of greater than 0.66, with sensitivities and specificities of 55% to 75% for prediction of recurrence. PHLDA3, RASGRF2, and TNFRSF10D had an AUC of greater than 0.69, with sensitivities and specificities of 60% to 75% for prediction of clinical recurrence, whereas the ZNF135 showed lower AUC of 0.55, and sensitivity and specificity of 50%. BCL11B, POU3F3, and RASGRF2 had an AUC of greater than 0.70, with sensitivities and specificities of 60% to 75% for prediction of systemic recurrence (Table 4). Except for the ZNF135, which is less consistent with the microarray data, validation of all other genes supports the findings obtained from the microarray experiments, although the magnitude of gene methylation was not always the same using these 2 different methods.

Table 3.

Forward and reverse primers used for PCR amplification and the sequencing primer used for pyrosequencing reactions

GeneAnnealing temp, °CForward primerReverse primerSequencing primer
HIF3A 56.1 GGTAGGAGTTTTGGGAATTGG *-CCACCCCTACAATCCCTAA GGGTGAGAGTTAAGA 
HAAO 56.1 AGGTTTGGGGTTTATTTTGAAT *-TAAAAATCCCAACACCTAAAAACAC TTTTAGATGGGAAAGTTAAATT 
RARβ 56.1 GTGTTTTTAAAATGAGTAGGGGAGGA CCATTCTCTATTCTACAATTTAAAACTTAC TTATTTGAGGAGGTTTAGTTTGGAA 
GSTP1 57.5 *-AGGGAGTAAATAGATAGTAGGAAGAG CCTCTCCCCTACCCTATAAA AATCCCAACCATCTTAAAAAATTTC 
CRIP1 59.2 TGTTTTAAGTGTAATAAGGAGGTGTAT *-ACAAAAACCTCCACCCTC AATAAGGAGGTGTATT 
RUNX3 57.5 Hs_RUNX3_01_PM00000112 
HS3ST2 59.2 GATTATAGGGGAGGGA11111GGAGAA *-ACACTTACCTATATTACAACTTCCTATAC GGGA11111GGAGAAG 
FLNC 59.2 Hs_FLNC_03_PM00127330 
RASGRF2 56.1 *-AAGTAGAAGAGTATATTTTGGTAGAG AACACCCCCTATACCTAACCTTTCTAAC AAACCCCACTTCCTAAATAAAAAAA 
PHLDA3 57.5 GGGGTTTTATGGAGTGTTAAGTAATTAGT *-AACAACCCAAACCTTCTAACT TTTGAAGTTTAATATGTAATTGGTG 
TNFRSF10D 59.2 *-ATGGTGGTTAGGTTGGTTTTTAA CACTAAACTACCCCAAAAAAAATTCC AACCTCAAAAAACCAC 
ZNF135 57.5 GGI I I I I 1 AG 11 GAG 1A1 AG 1 GAG IGGG *-CCTAAAAAAACCCCTTTTTCTCC GTATAGTGAGTGGGG 
BCL11B 57.5 ATATTAATTAGGGTAGAGGAGGAGGT *-ATAATAAACTCCCTCTAAAACAAATACT AGATGTAGGGGTAATGTTT 
POU3F3 59.2 *-GGGGTAGGGTTGTGATTTATTG AAAACCCCTCCTCTATCAAAATAATCCAAA CCAAAACCACTTCATACAAAAAAC 
GeneAnnealing temp, °CForward primerReverse primerSequencing primer
HIF3A 56.1 GGTAGGAGTTTTGGGAATTGG *-CCACCCCTACAATCCCTAA GGGTGAGAGTTAAGA 
HAAO 56.1 AGGTTTGGGGTTTATTTTGAAT *-TAAAAATCCCAACACCTAAAAACAC TTTTAGATGGGAAAGTTAAATT 
RARβ 56.1 GTGTTTTTAAAATGAGTAGGGGAGGA CCATTCTCTATTCTACAATTTAAAACTTAC TTATTTGAGGAGGTTTAGTTTGGAA 
GSTP1 57.5 *-AGGGAGTAAATAGATAGTAGGAAGAG CCTCTCCCCTACCCTATAAA AATCCCAACCATCTTAAAAAATTTC 
CRIP1 59.2 TGTTTTAAGTGTAATAAGGAGGTGTAT *-ACAAAAACCTCCACCCTC AATAAGGAGGTGTATT 
RUNX3 57.5 Hs_RUNX3_01_PM00000112 
HS3ST2 59.2 GATTATAGGGGAGGGA11111GGAGAA *-ACACTTACCTATATTACAACTTCCTATAC GGGA11111GGAGAAG 
FLNC 59.2 Hs_FLNC_03_PM00127330 
RASGRF2 56.1 *-AAGTAGAAGAGTATATTTTGGTAGAG AACACCCCCTATACCTAACCTTTCTAAC AAACCCCACTTCCTAAATAAAAAAA 
PHLDA3 57.5 GGGGTTTTATGGAGTGTTAAGTAATTAGT *-AACAACCCAAACCTTCTAACT TTTGAAGTTTAATATGTAATTGGTG 
TNFRSF10D 59.2 *-ATGGTGGTTAGGTTGGTTTTTAA CACTAAACTACCCCAAAAAAAATTCC AACCTCAAAAAACCAC 
ZNF135 57.5 GGI I I I I 1 AG 11 GAG 1A1 AG 1 GAG IGGG *-CCTAAAAAAACCCCTTTTTCTCC GTATAGTGAGTGGGG 
BCL11B 57.5 ATATTAATTAGGGTAGAGGAGGAGGT *-ATAATAAACTCCCTCTAAAACAAATACT AGATGTAGGGGTAATGTTT 
POU3F3 59.2 *-GGGGTAGGGTTGTGATTTATTG AAAACCCCTCCTCTATCAAAATAATCCAAA CCAAAACCACTTCATACAAAAAAC 

NOTE: Annealing temperature used for PCR amplification is shown. RUNX3 and FLNC PyroMark assays were obtained from Qiagen.

*Indicates the PCR primer with biotinylation.

Table 4.

Validation of methylation by pyrosequencing in an independent batch tissue samples from 20 patients representing each group

GeneSensitivity (95% CI)Specificity (95% CI)AUCt test (P)
Group 1a 
 HIF3A 90.0 (68.3–98.77) 100 (83.1–100) 0.932 0.0001 
 HAAO 89.4 (66.8–98.7) 100 (83.1–100) 0.947 0.0001 
 RARβ 84.2 (60.4–96.6) 100 (82.3–100) 0.912 0.0001 
 GSTP1 94.7 (73.9–99.8) 100 (82.3–100) 0.977 0.0001 
Group 2b 
 CRIP1 65.0 (40.7–84.6) 65.6 (40.7–84.6) 0.727 0.0139 
 RUNX3 70.4 (45.7–88.1) 75.3 (50.9–91.3) 0.788 0.0018 
 HS3ST2 65.0 (40.8–84.6) 60.0 (36.0–80.9) 0.733 0.0115 
 FLNC 70.3 (45.7–88.1) 60.4 (36.0–80.8) 0.660 0.0835 
 RASGRF2 75.7 (50.9–91.3) 55.2 (31.5–76.9) 0.682 0.0515 
Group 3c 
 PHLDA3 65.6 (40.7–84.6) 65.0 (40.7–84.6) 0.73 0.0129 
 RASGRF2 75.4 (50.9–91.3) 60.3 (36.0–80.8) 0.761 0.0047 
 TNFRSF10D 60.8 (36.0–80.8) 75.6 (50.9–91.3) 0.692 0.0373 
 ZNF135 50.0 (27.2–72.8) 50.0 (27.2–72.8) 0.555 0.5518 
Group 4d 
 BCL11B 75.2 (50.9–91.3) 60.0 (36.0–80.8) 0.741 0.0091 
 POU3F3 65.5 (40.7–84.6) 70.7 (45.7–88.1) 0.701 0.0295 
 RASGRF2 70.6 (45.7–88.1) 75.4 (50.9–91.3) 0.748 0.0071 
GeneSensitivity (95% CI)Specificity (95% CI)AUCt test (P)
Group 1a 
 HIF3A 90.0 (68.3–98.77) 100 (83.1–100) 0.932 0.0001 
 HAAO 89.4 (66.8–98.7) 100 (83.1–100) 0.947 0.0001 
 RARβ 84.2 (60.4–96.6) 100 (82.3–100) 0.912 0.0001 
 GSTP1 94.7 (73.9–99.8) 100 (82.3–100) 0.977 0.0001 
Group 2b 
 CRIP1 65.0 (40.7–84.6) 65.6 (40.7–84.6) 0.727 0.0139 
 RUNX3 70.4 (45.7–88.1) 75.3 (50.9–91.3) 0.788 0.0018 
 HS3ST2 65.0 (40.8–84.6) 60.0 (36.0–80.9) 0.733 0.0115 
 FLNC 70.3 (45.7–88.1) 60.4 (36.0–80.8) 0.660 0.0835 
 RASGRF2 75.7 (50.9–91.3) 55.2 (31.5–76.9) 0.682 0.0515 
Group 3c 
 PHLDA3 65.6 (40.7–84.6) 65.0 (40.7–84.6) 0.73 0.0129 
 RASGRF2 75.4 (50.9–91.3) 60.3 (36.0–80.8) 0.761 0.0047 
 TNFRSF10D 60.8 (36.0–80.8) 75.6 (50.9–91.3) 0.692 0.0373 
 ZNF135 50.0 (27.2–72.8) 50.0 (27.2–72.8) 0.555 0.5518 
Group 4d 
 BCL11B 75.2 (50.9–91.3) 60.0 (36.0–80.8) 0.741 0.0091 
 POU3F3 65.5 (40.7–84.6) 70.7 (45.7–88.1) 0.701 0.0295 
 RASGRF2 70.6 (45.7–88.1) 75.4 (50.9–91.3) 0.748 0.0071 

NOTE: Performance of the genes was analyzed by AUC of the genes for the 2 contrast groups. Sensitivity and specificity are reported for the threshold that minimized sensitivity–specificity. The P value indicates the statistical significance. t test p value is obtained by comparing 20 tissues in each group.

Abbreviation: CI, confidence interval.

aGenes that are significantly methylated in prostate cancer tissues compared with normal prostate.

bHypermethylated genes in tissues of patients with recurrent prostate cancer compared with nonrecurrent tissues.

cSignificance of genes methylated in prostate cancer tissues of patients with clinical recurrence compared with patients with only biochemical recurrence.

dGenes that are methylated in tissues of patients with systemic recurrence compared with local recurrence.

Figure 2.

Representative pyrogram of genes methylated in prostate cancer tissues. The sequence in the top part of each pyrogram represents the sequence under investigation. The sequence below the pyrogram indicates the sequentially added nucleotides. The gray regions highlight the analyzed C/T sites, with percentage values for the respective cytosine above them. A, pyrogram of genes HIF3A, HAAO, RARβ, and GSTP1 methylated in prostate cancer tissues. B, pyrogram of genes RUNX3, HS3ST2, CRIP1, and FLNC methylated in prostate cancer tissues of patients who had recurrence.

Figure 2.

Representative pyrogram of genes methylated in prostate cancer tissues. The sequence in the top part of each pyrogram represents the sequence under investigation. The sequence below the pyrogram indicates the sequentially added nucleotides. The gray regions highlight the analyzed C/T sites, with percentage values for the respective cytosine above them. A, pyrogram of genes HIF3A, HAAO, RARβ, and GSTP1 methylated in prostate cancer tissues. B, pyrogram of genes RUNX3, HS3ST2, CRIP1, and FLNC methylated in prostate cancer tissues of patients who had recurrence.

Close modal

We comprehensively examined the methylation of genes to identify biomarkers for detection of cancer and to predict clinical outcome of the disease in a total of 238 prostate tissues using Methylation27 BeadChips. Our methylation data from the Illumina Infinium microarray platform were subjected to batch-effects. The 46 samples ran on 4 beadchips had higher average detection P values (26), which had a significant impact on downstream analyses and result interpretation. Therefore, the data from these 46 samples were analyzed separately from the rest of 192 samples to remove the batch-effect on the analysis. Because of the low number of samples per group in the 46 samples, we used this data set to compare the methylation levels of genes selected from the 192 sample set. The results obtained from 46 samples were analogous to the 192 samples, although the power for statistical analysis was lower (Supplementary Table S4A–S4D).

To identify the genes for diagnosis of prostate cancer, we initially compared the prostate cancer tissues and normal adjacent tissues. There were 147 differentially hypermethylated genes in the tumor tissues with a P value <0.8.67E-25 and a fold change of greater than 2. To narrow down the list of genes as biomarkers for early detection, we concentrated on the top 25 hypermethylated genes (Fig. 1A), all of which show very high sensitivity and specificity for detection of prostate cancer (Table 2A). In agreement with earlier work for detection of prostate cancer, many of the genes found to be hypermethylated in our study were previously reported to be methylated in prostate cancer tissues (21, 27, 28). Many of these genes function are associated with protection of cells against oxidative damage. An increased vulnerability to genome-damaging stresses from electrophiles and oxidants could cause wide range of DNA lesions including base modification that attribute to inactivation of these genes by hypermethylation permitting prostate carcinogenesis (29). Several studies have shown PTGS2 methylation as a prognostic biomarker for prostate cancer (28, 30, 31). In our study, PTGS2 was significantly hypermethylated in tumor versus normal and systemic recurrence versus local recurrence groups with a P < 0.01 (Supplementary Table S6). Our data suggest that aberrant methylation of these genes occurred in the early stage of carcinogenesis and persisted even during the progression of cancer to metastasis. Because it is impractical to validate all genes identified in the current study, we selected GSTP1, RARβ, HIF3A, and HAAO genes from group 1 tumor tissues versus normal tissues for pyrosequencing. Many reports, including ours, have shown aberrant methylation of GSTP1 and RARβ in prostate cancer (21, 32). Our validation by pyrosequencing confirmed the methylation of GSTP1 and RARβ in prostate cancer. Because GSTP1 methylation is not specific to prostate cancer, we suggest that combination of multiple markers can be used to sensitively and uniquely identify prostate cancer. In exploratory analysis, we validated HIF3A, which plays a central role in the response of cells to hypoxia (33) and HAAO, which is associated with microsatellite stability (34). We found for the first time highly significant hypermethylation of HIF3A and HAAO genes (Table 2A) in prostate cancer tissues, which indicates that methylation of these genes could be an early event in prostate cancer development. Our results indicate that methylation of these genes is very homogeneous in different patients, consistently maintained during the progression to metastasis, and can be considered as effective biomarkers for prostate cancer detection.

Although many published studies have assessed the performance of candidate biomarkers in predicting time to relapse of prostate cancer following radical prostatectomy (35), very few markers were identified with limited sensitivity and specificity that can effectively identify those patients with prostate cancer with a high risk of early clinical progression or prostate cancer–specific mortality (15, 28, 36). Predicting the probability of recurrence will enable us to distinguish between patients who have an indolent cancer not requiring any form of treatment and patients with biochemical relapse who may have a more aggressive course requiring early intervention. To predict the relapse and identify the risk of progression, we compared all recurrence types with nonrecurrence. Genes found to be associated with risk of recurrence both by univariate and multivariate analyses (Supplementary Tables S2 and S3) were also significantly methylated in the recurrence versus nonrecurrence group comparison (Table 2B). Our univariate and binary analysis revealed similar set of genes associated with recurrence and that are further supported by multivariate analysis which suggests that these genes could be considered as independent predictive biomarkers of recurrence. We selected CRIP1, FLNC, RASGRF2, RUNX3, and HS3ST2 from group 2 for validation by pyrosequencing. In our previous study, we showed methylation of FLNC as markers for recurrence (21). FLNC is an actin cross-linking protein, our microarray analysis and pyrosequencing further confirm the methylation of FLNC as a biomarker for recurrence. RUNX3 (37) is reported to be methylated at 48% and 84% in different cohorts of patients with prostate cancer (15). HS3ST2 controls the rate of production of the critical active site 3-O-sulfate group on the antithrombin-binding site of anticoagulant heparin sulfate (38), CRIP1 is involved in intestinal zinc transport (39) and RASGRF2, is a tumor suppressor gene that plays a vital role in lymphocyte proliferation, T-cell signaling responses, and lymphomagenesis (40, 41). Tumor tissues of recurrent patients showed increase in the methylation of these genes. We show here for the first time that RASGRF2 is a significantly methylated gene in all 3 groups of recurrent prostate tissues. Our validation supports the findings from microarrays that there is an increase in the methylation of these genes in the prostate cancer tissues of patients who developed recurrence in contrast to patients with nonrecurrence. Because these genes are involved in signaling and transcription pathways, their inhibition by promoter methylation may plausibly have a role in prostate cancer progression.

To predict the outcome of hormone-refractory patients with prostate cancer who progress to clinical recurrence from PSA recurrence, we selected PHLDA3, RASGRF2, TNFRS10D, and ZNF135 from group 3, clinical recurrence versus biochemical recurrence patients for validation by pyrosequencing. TNFRSF10D is a TRAIL receptor with a truncated death domain, a biomarker to predict cancer invasion, inflammation progression (42), PHLDA3, a tumor suppressor protein is a p53-regulated repressor of AKT (43) and ZNF135 is a zinc finger protein located on 19q13 involved in cellular proliferation and differentiation (44). We have shown, for the first time, methylation of these genes in clinical recurrence cases. Except for ZNF135, which showed relatively low sensitivity, the other 3 genes showed significant increase in methylation in the patients with clinical recurrence, which supports the microarray data. Methylation of these genes could be considered as a supplementary approach to the histopathologic markers of cancerous lesions.

Local recurrence without systemic progression can be actively treated by salvage external beam radiotherapy (sEBRT; ref. 45). Systemic progression patients will not respond well to sEBRT. In our study, we have seen relatively high mortality in the systemic recurrence group. To confirm the genes associated with the risk of systemic progression (Table 2D), we validated BCL11B, POU3F3, and RASGRF2. BCL11B was repressed by methylation in chronic lymphocytic leukemia (46) and POU3F3 (47) was shown to be methylated in B-cell lymphoma (48). The sensitivity and specificity of RASGRF2 methylation by pyrosequencing are similar to that of microarray results. BCL11B and POU3F3 methylation is relatively low in the validation samples than in the microarray samples. This could be due to the heterogeneity of the metastatic prostate cancer tissues. For example, one previous study reported methylation of 16 genes in 91 metastatic prostate cancer tissues (28). The results from this study reveal that methylation of the genes does not correlate with the anatomic metastatic sites. The role of these genes in the metastatic progression need to be evaluated in future studies.

The data presented here are of significance because we provide a large unbiased list of potential targets of DNA methylation in a broad spectrum of prostate cancer tissues. It is possible that several of these genes could have potential tumor suppressor properties, and the study of these genes may provide further information on their roles in metastatic progression. It is therefore possible that systemic analysis of these genes may help in the development of new prognostic markers for prostate cancer and guide therapeutic timing in these patients. It is known that DNA methyltransferase inhibitors act, in part, through reversing aberrant DNA methylation and restoring gene expression. The genes identified in our study would be helpful in evaluating the outcome in the ongoing clinical trials with DNA methylation inhibitors (49).

In summary, our results revealed an expanded list of genes as biomarkers for diagnosis of prostate cancer. Although we validated a few genes from each group, future studies will evaluate the efficacy of other genes from each group, and a combination of multiple genes could serve as unique biomarkers for prediction of prostate cancer progression. We postulate that the alterations in methylation that occur in the early stages of tumor development (Table 2A) are very homogeneous and persist through the progression of the disease. These genes can be considered as the early detection biomarkers of cancer. Genes that were differentially methylated in the recurrent patients (Table 2B–D) appear to be more heterogeneous. During the metastatic progression, tumor cells acquire epigenetic changes that are distinct to each individual tumor. Taken together, these observations suggest that no single gene can likely predict the progression of all advanced prostate tumors. The altered methylation of genes associated with the recurrence of cancer can be considered as a supplementary approach to the histopathologic workup. A multidimensional approach, including clinicopathologic and molecular parameters, needs to be established for prediction of prostate cancer progression.

No potential conflicts of interests were disclosed.

This work supported by the grants: American Cancer Society RSG-09-175-01-CCE and U.S. Department of Defense W81XWH-09-1-0216.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jemal
A
,
Siegel
R
,
Xu
J
,
Ward
E
. 
Cancer statistics, 2010
.
CA Cancer J Clin
2010
;
60
:
277
300
.
2.
Damber
JE
,
Aus
G
. 
Prostate cancer
.
Lancet
2008
;
371
:
1710
21
.
3.
Sciarra
A
,
Cattarino
S
,
Gentilucci
A
,
Salciccia
S
,
Alfarone
A
,
Mariotti
G
, et al
Update on screening in prostate cancer based on recent clinical trials
.
Rev Recent Clin Trials
2011
;
6
:
7
15
.
4.
Tenke
P
,
Horti
J
,
Balint
P
,
Kovacs
B
. 
Prostate cancer screening
.
Recent Results Cancer Res
2007
;
175
:
65
81
.
5.
Karnes
RJ
,
Cheville
JC
,
Ida
CM
,
Sebo
TJ
,
Nair
AA
,
Tang
H
, et al
The ability of biomarkers to predict systemic progression in men with high-risk prostate cancer treated surgically is dependent on ERG status
.
Cancer Res
2010
;
70
:
8994
9002
.
6.
Stephenson
AJ
,
Scardino
PT
,
Eastham
JA
,
Bianco
FJ
 Jr
,
Dotan
ZA
,
Fearn
PA
, et al
Preoperative nomogram predicting the 10-year probability of prostate cancer recurrence after radical prostatectomy
.
J Natl Cancer Inst
2006
;
98
:
715
7
.
7.
Blute
ML
,
Bergstralh
EJ
,
Iocca
A
,
Scherer
B
,
Zincke
H
. 
Use of Gleason score, prostate specific antigen, seminal vesicle and margin status to predict biochemical failure after radical prostatectomy
.
J Urol
2001
;
165
:
119
25
.
8.
Briganti
A
,
Gallina
A
,
Suardi
N
,
Chun
FK
,
Walz
J
,
Heuer
R
, et al
A nomogram is more accurate than a regression tree in predicting lymph node invasion in prostate cancer
.
BJU Int
2008
;
101
:
556
60
.
9.
Madu
CO
,
Lu
Y
. 
Novel diagnostic biomarkers for prostate cancer
.
J Cancer
2010
;
1
:
150
77
.
10.
Brait
M
,
Sidransky
D
. 
Cancer epigenetics: above and beyond
.
Toxicol Mech Methods
2011
;
21
:
275
88
.
11.
Rodriguez-Paredes
M
,
Esteller
M
. 
Cancer epigenetics reaches mainstream oncology
.
Nat Med
2011
;
17
:
330
9
.
12.
Kulis
M
,
Esteller
M
. 
DNA methylation and cancer
.
Adv Genet
2010
;
70
:
27
56
.
13.
Watanabe
Y
,
Maekawa
M
. 
Methylation of DNA in cancer
.
Adv Clin Chem
2010
;
52
:
145
67
.
14.
Park
JY
. 
Promoter hypermethylation in prostate cancer
.
Cancer Control
2010
;
17
:
245
55
.
15.
Richiardi
L
,
Fiano
V
,
Vizzini
L
,
De Marco
L
,
Delsedime
L
,
Akre
O
, et al
Promoter methylation in APC, RUNX3, and GSTP1 and mortality in prostate cancer patients
.
J Clin Oncol
2009
;
27
:
3161
8
.
16.
Vanaja
DK
,
Ballman
KV
,
Morlan
BW
,
Cheville
JC
,
Neumann
RM
,
Lieber
MM
, et al
PDLIM4 repression by hypermethylation as a potential biomarker for prostate cancer
.
Clin Cancer Res
2006
;
12
:
1128
36
.
17.
Vanaja
DK
,
Grossmann
ME
,
Cheville
JC
,
Gazi
MH
,
Gong
A
,
Zhang
JS
, et al
PDLIM4, an actin binding protein, suppresses prostate cancer cell growth
.
Cancer Invest
2009
;
27
:
264
72
.
18.
Woodson
K
,
Hayes
R
,
Wideroff
L
,
Villaruz
L
,
Tangrea
J
. 
Hypermethylation of GSTP1, CD44, and E-cadherin genes in prostate cancer among US Blacks and Whites
.
Prostate
2003
;
55
:
199
205
.
19.
Maliukova
AV
,
Loginov
VI
,
Khodyrev
DS
,
Kadyrova
EL
,
Pronina
IV
,
Ivanova
TA
, et al
Methylation of the putative tumor suppressor gene, RASSF1A, in primary cervical tumors
.
Mol Biol (Mosk)
2004
;
38
:
1005
13
.
20.
Li
LC
,
Okino
ST
,
Dahiya
R
. 
DNA methylation in prostate cancer
.
Biochim Biophys Acta
2004
;
1704
:
87
102
.
21.
Vanaja
DK
,
Ehrich
M
,
Van den Boom
D
,
Cheville
JC
,
Karnes
RJ
,
Tindall
DJ
, et al
Hypermethylation of genes for diagnosis and risk stratification of prostate cancer
.
Cancer Invest
2009
;
27
:
549
60
.
22.
Vanaja
DK
,
Cheville
JC
,
Iturria
SJ
,
Young
CY
. 
Transcriptional silencing of zinc finger protein 185 identified by expression profiling is associated with prostate cancer progression
.
Cancer Res
2003
;
63
:
3877
82
.
23.
Chowdhury
S
,
Erickson
SW
,
MacLeod
SL
,
Cleves
MA
,
Hu
P
,
Karim
MA
, et al
Maternal genome-wide DNA methylation patterns and congenital heart defects
.
PLoS One
2011
;
6
:
e16506
.
24.
Storey
JD
,
Tibshirani
R
. 
Statistical significance for genomewide studies
.
Proc Natl Acad Sci U S A
2003
;
100
:
9440
5
.
25.
White
HE
,
Durston
VJ
,
Harvey
JF
,
Cross
NC
. 
Quantitative analysis of SNRPN(correction of SRNPN) gene methylation by pyrosequencing as a diagnostic test for Prader-Willi syndrome and Angelman syndrome
.
Clin Chem
2006
;
52
:
1005
13
.
26.
Sun
Z
,
Chai
HS
,
Wu
Y
,
White
WM
,
Donkena
KV
,
Klein
CJ
, et al
Batch effect correction for genome-wide methylation data with Illumina Infinium platform
.
BMC Med Genomics
2011
;
4
:
84
.
27.
Li
LC
,
Carroll
PR
,
Dahiya
R
. 
Epigenetic changes in prostate cancer: implication for diagnosis and treatment
.
J Natl Cancer Inst
2005
;
97
:
103
15
.
28.
Yegnasubramanian
S
,
Kowalski
J
,
Gonzalgo
ML
,
Zahurak
M
,
Piantadosi
S
,
Walsh
PC
, et al
Hypermethylation of CpG islands in primary and metastatic human prostate cancer
.
Cancer Res
2004
;
64
:
1975
86
.
29.
Donkena
KV
,
Young
CY
,
Tindall
DJ
. 
Oxidative stress and DNA methylation in prostate cancer
.
Obstet Gynecol Int
2010
;
2010:302051
.
30.
Bastian
PJ
,
Palapattu
GS
,
Yegnasubramanian
S
,
Lin
X
,
Rogers
CG
,
Mangold
LA
, et al
Prognostic value of preoperative serum cell-free circulating DNA in men with prostate cancer undergoing radical prostatectomy
.
Clin Cancer Res
2007
;
13
:
5361
7
.
31.
Woodson
K
,
O'Reilly
KJ
,
Ward
DE
,
Walter
J
,
Hanson
J
,
Walk
EL
, et al
CD44 and PTGS2 methylation are independent prognostic markers for biochemical recurrence among prostate cancer patients with clinically localized disease
.
Epigenetics
2006
;
1
:
183
6
.
32.
Rosenbaum
E
,
Hoque
MO
,
Cohen
Y
,
Zahurak
M
,
Eisenberger
MA
,
Epstein
JI
, et al
Promoter hypermethylation as an independent prognostic factor for relapse in patients with prostate cancer following radical prostatectomy
.
Clin Cancer Res
2005
;
11
:
8321
5
.
33.
Pasanen
A
,
Heikkila
M
,
Rautavuoma
K
,
Hirsila
M
,
Kivirikko
KI
,
Myllyharju
J
. 
Hypoxia-inducible factor (HIF)-3alpha is subject to extensive alternative splicing in human tissues and cancer cells and is regulated by HIF-1 but not HIF-2
.
Int J Biochem Cell Biol
2010
;
42
:
1189
200
.
34.
Huang
YW
,
Luo
J
,
Weng
YI
,
Mutch
DG
,
Goodfellow
PJ
,
Miller
DS
, et al
Promoter hypermethylation of CIDEA, HAAO and RXFP3 associated with microsatellite instability in endometrial carcinomas
.
Gynecol Oncol
2010
;
117
:
239
47
.
35.
Quinn
DI
,
Henshall
SM
,
Sutherland
RL
. 
Molecular markers of prostate cancer outcome
.
Eur J Cancer
2005
;
41
:
858
87
.
36.
Weiss
G
,
Cottrell
S
,
Distler
J
,
Schatz
P
,
Kristiansen
G
,
Ittmann
M
, 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
:
1678
85
.
37.
Ito
K
. 
RUNX3 in oncogenic and anti-oncogenic signaling in gastrointestinal cancers
.
J Cell Biochem
2011
;
112
:
1243
9
.
38.
Shworak
NW
,
Liu
J
,
Fritze
LM
,
Schwartz
JJ
,
Zhang
L
,
Logeart
D
, et al
Molecular cloning and expression of mouse and human cDNAs encoding heparan sulfate D-glucosaminyl 3-O-sulfotransferase
.
J Biol Chem
1997
;
272
:
28008
19
.
39.
Hempe
JM
,
Cousins
RJ
. 
Cysteine-rich intestinal protein binds zinc during transmucosal zinc transport
.
Proc Natl Acad Sci U S A
1991
;
88
:
9671
4
.
40.
Ruiz
S
,
Santos
E
,
Bustelo
XR
. 
RasGRF2, a guanosine nucleotide exchange factor for Ras GTPases, participates in T-cell signaling responses
.
Mol Cell Biol
2007
;
27
:
8127
42
.
41.
Fernandez-Medarde
A
,
Santos
E
. 
The RasGrf family of mammalian guanine nucleotide exchange factors
.
Biochim Biophys Acta
2011
;
1815
:
170
88
.
42.
Lin
WW
,
Hsieh
SL
. 
Decoy receptor 3: a pleiotropic immunomodulator and biomarker for inflammatory diseases, autoimmune diseases and cancer
.
Biochem Pharmacol
2011
;
81
:
838
47
.
43.
Kawase
T
,
Ohki
R
,
Shibata
T
,
Tsutsumi
S
,
Kamimura
N
,
Inazawa
J
, et al
PH domain-only protein PHLDA3 is a p53-regulated repressor of Akt
.
Cell
2009
;
136
:
535
50
.
44.
Tommerup
N
,
Vissing
H
. 
Isolation and fine mapping of 16 novel human zinc finger-encoding cDNAs identify putative candidate genes for developmental and malignant disorders
.
Genomics
1995
;
27
:
259
64
.
45.
Raldow
A
,
Hamstra
DA
,
Kim
S
,
Yu
JB
. 
Salvage external beam radiotherapy for prostate cancer after radical prostatectomy: current status and controversy
.
Oncology (Williston Park)
2010
;
24
:
692
700
, 2.
46.
Tong
WG
,
Wierda
WG
,
Lin
E
,
Kuang
SQ
,
Bekele
BN
,
Estrov
Z
, et al
Genome-wide DNA methylation profiling of chronic lymphocytic leukemia allows identification of epigenetically repressed molecular pathways with clinical impact
.
Epigenetics
2010
;
5
:
499
508
.
47.
Schreiber
J
,
Enderich
J
,
Sock
E
,
Schmidt
C
,
Richter-Landsberg
C
,
Wegner
M
. 
Redundancy of class III POU proteins in the oligodendrocyte lineage
.
J Biol Chem
1997
;
272
:
32286
93
.
48.
Rahmatpanah
FB
,
Carstens
S
,
Guo
J
,
Sjahputera
O
,
Taylor
KH
,
Duff
D
, et al
Differential DNA methylation patterns of small B-cell lymphoma subclasses with different clinical behavior
.
Leukemia
2006
;
20
:
1855
62
.
49.
Boumber
Y
,
Issa
JP
. 
Epigenetics in cancer: what's the future?
Oncology (Williston Park)
2011
;
25
:
220
6
,
228
.

Supplementary data