Journal List > J Korean Med Sci > v.34(Suppl 1) > 1123995

Kang, Park, Seo, Byun, Piao, Kim, Kim, Yun, Jang, Shon, Ryu, Lee, Kim, and Kim: Methylation Signature for Prediction of Progression Free Survival in Surgically Treated Clear Cell Renal Cell Carcinoma

Abstract

Background

Little is known about epigenetic silencing of genes by promoter hypermethylation in renal cell carcinoma (RCC). The aim of this study was to identify prognostic methylation markers in surgically treated clear cell RCC (ccRCC).

Methods

Methylation patterns were assayed using the Infinium HumanMethylation450 BeadChip array on pairs of ccRCC and normal tissue from 12 patients. Using quantitative PSQ analysis, tumor-specific hypermethylated genes were validated in 25 independent cohorts and their clinical relevance was also verified in 152 independent cohorts.

Results

Using genome-wide methylation array, Zinc finger protein 278 (ZNF278), Family with sequence similarity 155 member A (FAM155A) and Dipeptidyl peptidase 6 (DPP6) were selected for tumor-specific hypermethylated genes in primary ccRCC. The promoter methylation of these genes occurred more frequently in ccRCC than normal kidney in independent validation cohort. The hypermethylation of three genes were associated with advanced tumor stage and high grade tumor in ccRCC. During median follow-up of 39.2 (interquartile range, 15.4–79.1) months, 22 (14.5%) patients experienced distant metastasis. Multivariate analysis identified the methylation status of these three genes, either alone, or in a combined risk score as an independent predictor of distant metastasis.

Conclusion

The promoter methylation of ZNF278, FAM155A and DPP6 genes are associated with aggressive tumor phenotype and early development of distant metastasis in patients with surgically treated ccRCC. These potential methylation markers, either alone, or in combination, could provide novel targets for development of individualized therapeutic and prevention regimens.

Graphical Abstract

jkms-34-e144-abf001

INTRODUCTION

Renal cell carcinoma (RCC) affects more than 270,000 individuals annually worldwide, and is attributable for nearly 120,000 deaths each year.1 In Korea, age-standardized incidence rate and age-standardized mortality rate were 5.7 (men, 8.2; women, 3.4) and 1.0 (men, 1.7; women, 0.5) per 100,000 in 2015, respectively.2 Clear-cell RCC (ccRCC) is the most common type of RCC and accounts for 70% of cases.3 Although patients with small, localized (stage I–III) ccRCC treated surgically have excellent 5-year survival rates, around 30% of patients treated for localized disease will relapse.45 Similar to other tumor entities, ccRCC presents with significant clinical heterogeneity, which ranges from indolent to highly aggressive.6 The identification of accurate predictors of clinical outcome is imperative to determine individualized follow-up strategies and to facilitate counseling regarding adjuvant therapy. At present, prediction of tumor relapse for patients with surgically treated localized ccRCC is based on clinical and pathological features, including TNM stage, nuclear grade, which can be subject to inter-observer variability and might not entirely account for individual tumor biology.789 These limitations have led to much research aimed at identifying more accurate molecular markers that lead to better identification and counseling for patients with RCC. Our ability to better manage patients with RCC has been largely driven by a better understanding of the genetic and epigenetic modifications.101112 Epigenetic gene silencing is a molecular mechanism of silencing a gene by methylating its promoter region.13 Although previous epigenetic studies have identified a number of frequently methylated genes in RCC, few studies have determined the prognostic implications of DNA methylation profile in RCC.141516
The present study aims to identify clinical diagnostic, prognostic, and predictive methylation markers in surgically treated ccRCC. We performed comprehensive DNA methylation array analyses to detect differentially methylated genes in ccRCC and assessed the implications of novel candidate genes as prognostic methylation markers for surgically treated ccRCC.

METHODS

Patients and tissue samples

Human kidney specimens from 12 pairs of ccRCC and normal tissue (array set) and 152 ccRCC including 25 pairs of ccRCC and matched normal tissue (validation set) from primary, histologically-proven ccRCC who underwent radical or partial nephrectomy were collected between September 1997 and December 2014 at our institution. The pathology samples were independently re-examined by a pathologist who was unaware of the use of the clinical data and confirmed the presence of tumor or normal tissue. To reduce confounding factors affecting the analyses, patients had to fulfill the following criteria to be enrolled in this study: 1) clinically and pathologically T1 to T4 without LN or distant metastasis; 2) histologic pure clear cell RCC; 3) minimum follow-up period of 3 months; 4) simple tumor enucleation and positive surgical margin at final pathology were excluded to avoid biasing the survival estimates.
Surgical procedures performed included pure laparoscopic, hand-assisted laparoscopic, robot-assisted and open approaches. Pathological staging was performed based on the 7th edition of the American Joint Committee on Cancer classification system, and histological differentiation was graded according to the Fuhrman nuclear grading system.1718 After surgery, each patient was monitored according to the standard guidelines.1920 Distant metastasis was defined as lymph-node and/or other organ (lung, brain, contralateral kidney, etc.) metastasis, but local recurrence was not included in this definition.

DNA methylation profiling

Twenty-four matched DNA samples (12 ccRCC and 12 matched normal-surrounding kidney tissues) were used for DNA methylation profiling. Twelve ccRCC tissue samples were randomly allocated with pathologic stage. Genomic DNA (gDNA) was extracted by standard methods by using the Wizard Genomic DNA Purification System (Promega, Madison, WI, USA). Bisulfite-modified gDNA was prepared using EZ DNA Methylation-Lightning kit (Zymo Research, Orange, CA, USA) according to the manufacturer's instructions. The methylation status was assayed using the Infinium HumanMethylation450 BeadChip array (Infinium Methylation 450K; Illumina Inc., San Diego, CA, USA), which enables interrogation of the methylation status of more than 480,000 CpG sites distributed over the whole genome. Fluorescence signals corresponding to C- or T-nucleotides were measured, and the data were used to assign a quantitative measure of methylation level of specific CpG islands.

Selection of candidate methylation-silenced genes

The β value represents a quantitative measure of the DNA methylation level of specific CpG islands and ranges from 0 (completely unmethylated) to 1 (completely methylated). Candidate methylation-silenced genes were selected with next criteria: 1) a difference in DNA methylation levels between ccRCC and matched normal kidney (∆β value) > 0.25; 2) a mean β value for matched normal kidney < 0.15.

Pyrosequencing analysis

The DNA methylation status of candidate methylation-silenced genes was specifically assessed by pyrosequencing analysis of 152 human kidney specimens using a PyroMark Q96 ID instrument (Qiagen, Valencia, CA, USA) according to the manufacturer's instructions. Each primer was designed using Pyrosequencing Assay Design Software v2.0 (Qiagen). PSQ primers were designed to encompass the CpG island loci assayed on the Illumina Infinium array. Primers were designed using NCBI Reference Sequences build version 37. PCR reactions were carried out in a volume of 20 µL with 20 ng or more converted gDNA, PCR premixture (Enzynomics, Daejeon, Korea), 1 µL of 10 pmole/µL forward primer, and 1 µL of 10 pmole/µL biotinylated reverse primer. The thermocycling parameters were as follows: denaturation at 95°C for 10 minutes; followed by 45 cycles of 95°C for 30 seconds, annealing at 59°C for 30 seconds, and 72°C for 30 seconds; and a final extension at 72°C for 5 minutes. The primer sequences are described in Supplementary Table 1.

Statistical analysis

The DNA methylation profile data were normalized using quantile normalization in the R language environment (version 2.10.0, available at http://www.r-project.org/).
The differences in continuous variables between groups were assessed using a two sample t-test or ANOVA. Receiver operating characteristic curves were constructed to estimate the capability of candidate markers for the prediction of metastasis, and to determine the optimal cut-off point for dividing patients into subgroups (hypomethylation or hypermethylation) with the highest combined sensitivity and specificity. To integrate the methylation status of the methylation markers with prognostic relevance, each patient's methylation score (M score) was calculated as the sum of the methylation levels of selected genes multiplied by the corresponding regression coefficients derived from the Cox regression analysis used to assess the predictive value of each gene for prediction of distant metastasis.21 The Kaplan-Meier curves were used to estimate time to metastasis according to methylation status, and differences were evaluated using log-rank tests. The prognostic value of methylation status was evaluated using the multivariate Cox proportional hazards regression analyses. Statistical analysis was performed using SPSS 21.0 software (IBM, Armonk, NY, USA). A P value < 0.05 was considered statistically significant.

Ethics statement

The study was carried out in agreement with applicable laws and regulations, good clinical practices, and ethical principles as described in the Declaration of Helsinki. The Institutional Review Board (IRB) of Chungbuk National University approved this protocol (IRB No. 2010-01-001), and written informed consent was obtained from each subject. Collection and analysis of all samples was approved by the IRB of Chungbuk National University.

RESULTS

Methylation profiling and identification of candidate silenced genes in primary ccRCC

The methylation profiles of 12 pairs of ccRCC and surrounding normal kidney were analyzed by the Infinium HumanMethylation450 array (Fig. 1). Using selection criteria (△ β value > 0.25 and mean β value of matched normal kidney < 0.15), 104 unique CpG island loci that were hypermethylated in ccRCC compared with the NCs were identified. Of these, pyrosequencing analyses of three out of top ranked 5 candidate genes, (Zinc finger protein 278 [ZNF278], Family with sequence similarity 155 member A [FAM155A] and Dipeptidyl peptidase 6 [DPP6]) were technically available (Table 1).
Fig. 1

Heat map for hierarchical clustering of human kidney specimens from 12 pairs of clear cell renal cell carcinoma and normal tissue.

jkms-34-e144-g001
Table 1

β-value differences between tumors and normal controls

jkms-34-e144-i001
Genes β-valuea Differential P value
Normal control ccRCC
ZFP28 0.0654 0.361 0.295 < 0.001
FAM155A 0.132 0.418 0.286 0.022
DPP6 0.133 0.388 0.255 < 0.001
ccRCC = clear cell renal cell carcinoma, ZFP28 = Zinc finger protein 28, FAM155A = family with sequence similarity 155, member A, DPP6 = dipeptidyl peptidase 6.
aThe β value represents a quantitative measure of the DNA methylation level of specific CpG islands and ranges from 0 (completely unmethylated) to 1 (completely methylated).

Validation of methylation array findings in an independent primary ccRCC

To validate methylation array findings, we used the independent validation cohort of 25 pairs of ccRCC and matched normal tissue with PSQ (Table 2). The methylation level of ZNF278, FAM155A and DPP6 was higher in ccRCC than normal kidney in independent validation cohort (each P < 0.001) (data not shown).
Table 2

Clinicopathologic characteristics of array and validation cohort

jkms-34-e144-i002
Variables Array cohort Validation cohort
Patient No. 12 152
Age, yr 57.3 ± 11.3 64.3 ± 13.8
Follow-up, mon, median (IQR) - 39.2 (15.4–79.1)
Gender, men 8 (66.7) 110 (72.4)
Pathologic T stage
T1 4 (33.3) 112 (73.7)
T2 4 (33.3) 14 (9.2)
T3 4 (33.3) 23 (15.1)
T4 - 3 (2.0)
Nuclear grade
1–2 9 (75.0) 102 (67.1)
3–4 3 (25.0) 50 (32.9)
Distant metastasis - 22 (14.5)
Data are presented as mean ± standard deviation or number (%).
IQR = interquartile range.

Association between methylation levels and tumor aggressiveness

To evaluate the relationship between methylation status of candidate genes and clinicopathological factors, such as pathologic T stage and Fuhrman grade. Clinicopathologic characteristics of independent primary ccRCC are listed in Table 2. The hypermethylation of three genes were associated with advanced tumor stage and higher tumor grade (all P < 0.05) (Table 3).
Table 3

Relationship between ZNF278, FAM155A and DPP6 methylation and clinicopathological parameters in surgically treated clear cell renal cell carcinoma

jkms-34-e144-i003
Variables ZNF278 FAM155A DPP6
Methylation, % P value Methylation, % P value Methylation, % P value
T stage 0.001 0.002 < 0.001
T1 18.30 ± 11.95 15.69 ± 10.10 19.41 ± 11.24
T2 26.56 ± 15.53 24.40 ± 17.39 25.39 ± 14.96
T3–4 28.19 ± 15.27 24.20 ± 19.15 32.61 ± 17.57
Fuhrman grade 0.022 0.039 0.001
1–2 18.85 ± 12.09 16.15 ± 10.89 19.21 ± 10.35
3–4 24.63 ± 15.32 21.61 ± 16.67 28.35 ± 17.42
Metastasis 0.002 < 0.001 < 0.001
No 19.38 ± 13.21 16.29 ± 12.13 20.44 ± 12.98
Yes 28.86 ± 12.36 27.72 ± 15.66 32.74 ± 13.61
P value calculated using Student's t-test.
ZFP28 = Zinc finger protein 28, FAM155A = family with sequence similarity 155, member A, DPP6 = dipeptidyl peptidase 6.

Methylation status as a predictor of distant metastasis

During median follow-up of 39.2 (interquartile range, 15.4–79.1) months, 22 (14.5%) patients experienced distant metastasis. Kaplan-Meier estimates identified significant differences in metastasis free survival according to methylation status of three candidate gene (log-rank test, all P < 0.001) (Fig. 2A-C). Integrate methylation score of three candidate genes, M score and adjusted-M score also showed identical results (log-rank test, all P < 0.001) (Fig. 2D and E). On multivariate analysis, hazard ratio (HR) was determined separately according to the methylation status of each gene and the clinicopathological factors. Multivariate analysis identified the methylation status of candidate three genes in each (ZNF278, HR, 7.012, P = 0.008; FAM155A, HR, 7.080, P = 0.008; DPP6, HR, 6.444, P = 0.011) (data not shown), or in a integrate methylation score of three candidate genes (M-score, HR, 3.804; P = 0.006) (Table 4) as an independent predictor of distant metastasis.
Fig. 2

Kaplan-Meier estimation for development of metastasis surgically treated clear cell renal cell carcinoma according to methylation status of three candidate gene. (A) ZNF278, (B) FAM155A, (C) DPP6, (D) M score and (E) M score with pathologic T stage adjusted.

jkms-34-e144-g002
Table 4

Multivariate Cox regression analysis for the prediction of metastasis in surgically treated clear cell renal cell carcinoma

jkms-34-e144-i004
Variables Univariate analysis Multivariate analysis
HR (95% CI) P value HR (95% CI) P value
Age, yr 1.016 (0.981–1.052) 0.373
Gender, women 0.779 (0.304–1.998) 0.603
HTN, yes 0.780 (0.317–1.919) 0.588
DM, yes 0.928 (0.314–2.744) 0.893
BMI, ≥ 23 kg/m2 0.371 (0.160–0.859) 0.021 0.270 (0.092–0.791) 0.017
Smoking history, yes 0.760 (0.273–2.114) 0.598
TNM stage
pT1 1 1
pT2 5.651 (1.711–18.662) 0.004 7.111 (1.776–28.464) 0.006
pT3-4 10.331 (3.811–28.011) < 0.001 6.567 (2.133–20.215) 0.001
Fuhrman grade, G3–4 5.385 (2.192–13.229) < 0.001 2.442 (0.841–7.091) 0.101
M score, ≥ 153.28a 7.166 (2.915–17.612) < 0.001 3.804 (1.477–9.800) 0.006
HR = hazard ratio, CI = confidence interval, HTN = hypertension, DM = diabetes mellitus, BMI = body mass index, TNM = tumor node metastasis.
aM score was calculated with the sum of the levels of methylation levels of three candidate genes multiplied by the corresponding regression coefficient derived from the Cox regression analysis.

DISCUSSION

The present study aims to identify clinical diagnostic, prognostic, and predictive methylation markers in surgically treated ccRCC. We performed comprehensive DNA methylation array analyses and identified 3 novel methylated loci, ZNF278, FAM155A and DPP6. The validity of these candidate methylation markers was determined in a relatively large independent ccRCC. Notably, the methylation status of these candidate genes, either single or combined was closely related to not only tumor histology but also development of metastasis in surgically treated ccRCC. Our findings suggest the utility for the DNA methylation patterns in these genes as clinically useful surrogate markers, as well as new molecular pathways for further investigation as therapeutic targets in ccRCC.
Aberrant methylation of promoter CpG islands is an important inactivation mechanism of tumor suppressors and tumor-related genes.22 Methylation-induced silencing in early phases of tumorigenesis could potentially be used as markers for identifying individuals at increased risk of developing malignancy or for aiding in the diagnosis of early malignancy, whereas those genes undergoing methylation during the progression of malignancy could potentially be used as prognostic markers.2324 Genome-wide methylation profiling has been used as an approach for the identification of prognostic markers in different cancers evaluating thousands of CpG sites simultaneously.25 Although a number of hyper- or hypomethylated loci have been also identified for renal cell RCC, prognostic methylation markers of RCC that can provide useful information about survival and treatment options at diagnosis remains a major clinical challenge.1626
Candidate-gene approaches have disclosed several genes frequently methylated in ccRCC, including CDH1, APAF1, COL1A1, DKK2, DKK3, SFRPI, SFRP4, SFRP5, and WIF1.26 Genome-wide approaches have uncovered novel genes and pathways involved in ccRCC pathogenesis. McRonald and colleagues performed high-throughput epigenetic profiling using the Illumina Goldengate Methylation Array in 29 RCC from von Hippel-Lindau (VHL) disease and 20 sporadic ccRCC with wild type VHL and 13 sporadic papillary RCC to elucidate the differences in tumorigenesis mechanisms dependent on VHL gene status. They observed differing patterns of tumour-specific CpG methylation in VHL and non VHL ccRCC and papillary RCC.27 Morris et al.14 conducted the first whole-genome expression microarray study using methylated DNA immunoprecipitation (MeDIP) with whole-genome microarray analysis to identify differentially methylated regions in RCC. They identified a number of genes, including KLHL35, QPCT, SCUBE3, ZSCAN18, CCDC8, FBN2, ATP5G2, PCDH8 and CORO6 were frequently methylated in RCC and promoter hypermethylation of these genes resulted in significant reduction of their expression level. Recent study by Ricketts et al.15 have performed genome-wide methylation profiling of RCC using the HumanMethylation27 BeadChips in 38 sporadic RCC and 9 age-matched normal kidney controls. Eight novel ccRCC TSG candidates were identified, including OVOL1, DLEC1, BMP4, SST, TMPRSS2, TM6SF1, SLC34A2, and COL1A2, which demonstrated methylation in kidney cancer cell lines, re-expression in kidney cancer cell lines after 5-aza-20-deoxycytidine treatment, and tumor-specific methylation. Moreover, OVOL1 knockdown increased c-Myc mRNA levels which may lead to c-Myc pathway activation. We also performed the Illumina BeadArray technology to directly assay methylated CpG status, and identified 3 novel methylated loci, ZNF278, FAM155A, and DPP6. To the best of our knowledge, theses 3 loci have not previously been reported in RCC. From a prognostic point of view, the methylation status of these candidate genes, either single or combined was closely related to not only tumor histology but also development of metastasis in surgically treated ccRCC.
This study has several limitations and strengths. The β values of our three candidate genes are not high. We can't explain exactly the reasons, but this may be originated from the tumor characteristic of RCC or our relatively small number of methylation profiling. Second, there is currently only limited information available in the literature on the specific function of three candidate genes. ZNF278 also named POZ/BTB and AT-hook-containing zinc finger protein (PATZ), is a recently identified transcription factor with seven C2H2-type zinc fingers. Although the physiological role of ZNF278 is not clear, experimental evidence suggests that it is a potential transcription repressor. Previous study observed up-regulation of ZNF278 expression in human colorectal cancer tissues.28 The specific function of FAM155A has not been determined. DPP6 encodes protein dipeptidyl-peptidase 6, which binds to specific voltage-gated potassium channels and alters their expression and biophysical properties. A recent study on core signaling pathways in human pancreatic cancers suggested that DPP6 might play a crucial role in regulation of invasion of pancreatic cancer cells.29 Additional studies are needed to define the precise biological functions of these novel biomarkers on tumorigenesis and prognosis in ccRCC. Nonetheless, the present study represents an important step towards the clinical use of prognostic methylation markers in ccRCC.
In conclusion, the present study identified potential methylation markers which could represent the aggressive tumor phenotype and early development of distant metastasis in ccRCC. These potential methylation markers, either alone, or integrate methylation score, could offers a chance to identify cancer-specific diagnostic and prognostic tools, in addition to novel targets for development of individualized therapeutic and prevention regimens.

ACKNOWLEDGEMENTS

The specimens for this study were provided by Chungbuk National University Hospital, a member of the National Biobank of Korea, which is supported by the Ministry of Health, Welfare and Family Affairs. The authors wish to thank Ms. Eun-Ju Shim from the National Biobank of Korea at Chungbuk National University Hospital for sample preparation and technical assistance.

Notes

Funding This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2015R1D1A1A01057786 and 2018R1D1A1B07043906).

Disclosure The authors have no potential conflicts of interest to disclose.

Author Contributions

  • Conceptualization: Kang HW, Park H, Kim YJ.

  • Data curation: Seo SP, Kim SM, Jang W.

  • Formal analysis: Park H, Kim WT, Yun SJ.

  • Investigation: Byun YJ, Piao XM.

  • Methodology: Seo SP, Kim SM, Jang W.

  • Validation: Kim WT, Yun SJ, Shin HS.

  • Writing - original draft: Kang HW, Park H, Kim YJ.

  • Writing - review & editing: Shon HS, Ryu KH, Lee SC, Kim WJ, Kim YJ.

References

1. Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM. New strategies in renal cell carcinoma: targeting the genetic and metabolic basis of disease. Clin Cancer Res. 2015; 21(1):10–17.
crossref
2. Jung KW, Won YJ, Kong HJ, Lee ES. Community of Population-Based Regional Cancer Registries. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2015. Cancer Res Treat. 2018; 50(2):303–316.
crossref
3. Brugarolas J. Molecular genetics of clear-cell renal cell carcinoma. J Clin Oncol. 2014; 32(18):1968–1976.
crossref
4. Eggener SE, Yossepowitch O, Pettus JA, Snyder ME, Motzer RJ, Russo P. Renal cell carcinoma recurrence after nephrectomy for localized disease: predicting survival from time of recurrence. J Clin Oncol. 2006; 24(19):3101–3106.
crossref
5. Chung JS, Son NH, Lee SE, Hong SK, Jeong CW, Kwak C, et al. Partial versus radical nephrectomy for T1-T2 renal cell carcinoma in patients with chronic kidney disease stage III: a multiinstitutional analysis of kidney function and survival rate. J Korean Med Sci. 2018; 33(43):e277.
crossref
6. Gulati S, Martinez P, Joshi T, Birkbak NJ, Santos CR, Rowan AJ, et al. Systematic evaluation of the prognostic impact and intratumour heterogeneity of clear cell renal cell carcinoma biomarkers. Eur Urol. 2014; 66(5):936–948.
crossref
7. Al-Aynati M, Chen V, Salama S, Shuhaibar H, Treleaven D, Vincic L. Interobserver and intraobserver variability using the Fuhrman grading system for renal cell carcinoma. Arch Pathol Lab Med. 2003; 127(5):593–596.
crossref
8. Lang H, Lindner V, de Fromont M, Molinié V, Letourneux H, Meyer N, et al. Multicenter determination of optimal interobserver agreement using the Fuhrman grading system for renal cell carcinoma: assessment of 241 patients with > 15-year follow-up. Cancer. 2005; 103(3):625–629.
9. Kim TW, Lee JH, Shim KH, Choo SH, Choi JB, Ahn HS, et al. Prognostic significance of preoperative and follow-up neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in patients with non-metastatic clear cell renal cell carcinoma. Investig Clin Urol. 2019; 60(1):14–20.
crossref
10. Vasudev NS, Selby PJ, Banks RE. Renal cancer biomarkers: the promise of personalized care. BMC Med. 2012; 10(1):112.
crossref
11. Jonasch E, Futreal PA, Davis IJ, Bailey ST, Kim WY, Brugarolas J, et al. State of the science: an update on renal cell carcinoma. Mol Cancer Res. 2012; 10(7):859–880.
crossref
12. Dienstmann R, Rodon J, Barretina J, Tabernero J. Genomic medicine frontier in human solid tumors: prospects and challenges. J Clin Oncol. 2013; 31(15):1874–1884.
crossref
13. Baylin SB. DNA methylation and gene silencing in cancer. Nat Clin Pract Oncol. 2005; 2:Suppl 1. S4–S11.
crossref
14. Morris MR, Ricketts CJ, Gentle D, McRonald F, Carli N, Khalili H, et al. Genome-wide methylation analysis identifies epigenetically inactivated candidate tumour suppressor genes in renal cell carcinoma. Oncogene. 2011; 30(12):1390–1401.
crossref
15. Ricketts CJ, Morris MR, Gentle D, Brown M, Wake N, Woodward ER, et al. Genome-wide CpG island methylation analysis implicates novel genes in the pathogenesis of renal cell carcinoma. Epigenetics. 2012; 7(3):278–290.
crossref
16. Arai E, Chiku S, Mori T, Gotoh M, Nakagawa T, Fujimoto H, et al. Single-CpG-resolution methylome analysis identifies clinicopathologically aggressive CpG island methylator phenotype clear cell renal cell carcinomas. Carcinogenesis. 2012; 33(8):1487–1493.
crossref
17. Greene FL. The American Joint Committee on Cancer: updating the strategies in cancer staging. Bull Am Coll Surg. 2002; 87(7):13–15.
18. Fuhrman SA, Lasky LC, Limas C. Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol. 1982; 6(7):655–663.
crossref
19. Escudier B, Eisen T, Porta C, Patard JJ, Khoo V, Algaba F, et al. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012; 23:Suppl 7. vii65–vii71.
crossref
20. Ljungberg B, Bensalah K, Canfield S, Dabestani S, Hofmann F, Hora M, et al. EAU guidelines on renal cell carcinoma: 2014 update. Eur Urol. 2015; 67(5):913–924.
crossref
21. Kim YJ, Yoon HY, Kim JS, Kang HW, Min BD, Kim SK, et al. HOXA9, ISL1 and ALDH1A3 methylation patterns as prognostic markers for nonmuscle invasive bladder cancer: array-based DNA methylation and expression profiling. Int J Cancer. 2013; 133(5):1135–1142.
22. Ghavifekr Fakhr M, Farshdousti Hagh M, Shanehbandi D, Baradaran B.. DNA methylation pattern as important epigenetic criterion in cancer. Genet Res Int. 2013; 2013:317569.
crossref
23. Mulero-Navarro S, Esteller M. Epigenetic biomarkers for human cancer: the time is now. Crit Rev Oncol Hematol. 2008; 68(1):1–11.
crossref
24. How Kit A, Nielsen HM, Tost J. DNA methylation based biomarkers: practical considerations and applications. Biochimie. 2012; 94(11):2314–2337.
crossref
25. Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer. 2003; 3(4):253–266.
crossref
26. Rydzanicz M, Wrzesiński T, Bluyssen HA, Wesoły J. Genomics and epigenomics of clear cell renal cell carcinoma: recent developments and potential applications. Cancer Lett. 2013; 341(2):111–126.
crossref
27. McRonald FE, Morris MR, Gentle D, Winchester L, Baban D, Ragoussis J, et al. CpG methylation profiling in VHL related and VHL unrelated renal cell carcinoma. Mol Cancer. 2009; 8(1):31.
crossref
28. Tian X, Sun D, Zhang Y, Zhao S, Xiong H, Fang J. Zinc finger protein 278, a potential oncogene in human colorectal cancer. Acta Biochim Biophys Sin (Shanghai). 2008; 40(4):289–296.
crossref
29. Low SK, Kuchiba A, Zembutsu H, Saito A, Takahashi A, Kubo M, et al. Genome-wide association study of pancreatic cancer in Japanese population. PLoS One. 2010; 5(7):e11824.
crossref

SUPPLEMENTARY MATERIAL

Supplementary Table 1

Primer for pyrosequencing
TOOLS
ORCID iDs

Ho Won Kang
https://orcid.org/0000-0002-8164-4427

Hongyong Park
https://orcid.org/0000-0001-6183-8059

Sung Pil Seo
https://orcid.org/0000-0001-6179-7031

Young Joon Byun
https://orcid.org/0000-0002-9449-478X

Xuan-Mei Piao
https://orcid.org/0000-0001-6847-7743

Sung Min Kim
https://orcid.org/0000-0002-2374-020X

Won Tae Kim
https://orcid.org/0000-0002-7482-4550

Seok-Joong Yun
https://orcid.org/0000-0001-7737-4746

Wooyeong Jang
https://orcid.org/0000-0002-6688-0333

Ho Sun Shon
https://orcid.org/0000-0002-6717-7869

Keun Ho Ryu
https://orcid.org/0000-0003-0394-9054

Sang-Cheol Lee
https://orcid.org/0000-0002-4163-2210

Wun-Jae Kim
https://orcid.org/0000-0002-8060-8926

Yong-June Kim
https://orcid.org/0000-0001-7638-7174

Similar articles