Skip to main content

Promoter Hypermethylation as a Biomarker in Prostate Adenocarcinoma

  • Protocol
  • First Online:
Cancer Epigenetics

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1238))

Abstract

There is increasing evidence that DNA methylation is a critical source of gene regulation. In addition, interindividual differences in DNA methylation status in cancer related genes are associated with risk and progression of prostate cancer. Therefore, DNA methylation in cancer related genes can be potential biomarkers and therapeutic targets for prostate cancer.

In this chapter, current information on frequently hypermethylated genes associated with the carcinogenesis and progression of prostate cancer was updated. The potential biological role of hypermethylated genes in prostate cancer is discussed. These findings may provide new information of the pathogenesis, the exciting potential to be predictive and to provide strategies for personalized treatment of prostate cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 89.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 119.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Siegel R, Naishadham D, Jemal A (2013) Cancer statistics, 2013. CA Cancer J Clin 63(1):11–30

    PubMed  Google Scholar 

  2. Baylin SB, Herman JG (2000) DNA hypermethylation in tumorigenesis: epigenetics joins genetics. Trends Genet 16(4):168–174

    CAS  PubMed  Google Scholar 

  3. Weichenhan D, Plass C (2013) The evolving epigenome. Hum Mol Genet 22(R1):R1–R6

    CAS  PubMed  Google Scholar 

  4. Smiraglia DJ, Plass C (2002) The study of aberrant methylation in cancer via restriction landmark genomic scanning. Oncogene 21(35):5414–5426

    CAS  PubMed  Google Scholar 

  5. Rush LJ et al (2001) Novel methylation targets in de novo acute myeloid leukemia with prevalence of chromosome 11 loci. Blood 97(10):3226–3233

    CAS  PubMed  Google Scholar 

  6. Costello JF et al (2000) Aberrant CpG-island methylation has non-random and tumour-type-specific patterns. Nat Genet 24(2):132–138

    CAS  PubMed  Google Scholar 

  7. Baylin SB et al (1998) Alterations in DNA methylation: a fundamental aspect of neoplasia. Adv Cancer Res 72:141–196

    CAS  PubMed  Google Scholar 

  8. Di Croce L et al (2002) Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science 295(5557):1079–1082

    PubMed  Google Scholar 

  9. Yan PS et al (2003) Differential distribution of DNA methylation within the RASSF1A CpG island in breast cancer. Cancer Res 63(19):6178–6186

    CAS  PubMed  Google Scholar 

  10. Graff JR et al (1997) Mapping patterns of CpG island methylation in normal and neoplastic cells implicates both upstream and downstream regions in de novo methylation. J Biol Chem 272(35):22322–22329

    CAS  PubMed  Google Scholar 

  11. Esteller M (2000) Epigenetic lesions causing genetic lesions in human cancer: promoter hypermethylation of DNA repair genes. Eur J Cancer 36(18):2294–2300

    CAS  PubMed  Google Scholar 

  12. Bachman KE et al (1999) Methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene suggest a suppressor role in kidney, brain, and other human cancers. Cancer Res 59(4):798–802

    CAS  PubMed  Google Scholar 

  13. Toyota M et al (2000) Distinct genetic profiles in colorectal tumors with or without the CpG island methylator phenotype. Proc Natl Acad Sci U S A 97(2):710–715

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Stirzaker C et al (1997) Extensive DNA methylation spanning the Rb promoter in retinoblastoma tumors. Cancer Res 57(11):2229–2237

    CAS  PubMed  Google Scholar 

  15. Deng G et al (1999) Methylation of CpG in a small region of the hMLH1 promoter invariably correlates with the absence of gene expression. Cancer Res 59(9):2029–2033

    CAS  PubMed  Google Scholar 

  16. Gonzalgo ML et al (1997) Low frequency of p16/CDKN2A methylation in sporadic melanoma: comparative approaches for methylation analysis of primary tumors. Cancer Res 57(23):5336–5347

    CAS  PubMed  Google Scholar 

  17. Gonzalez-Zulueta M et al (1995) Methylation of the 5′ CpG island of the p16/CDKN2 tumor suppressor gene in normal and transformed human tissues correlates with gene silencing. Cancer Res 55(20):4531–4535

    CAS  PubMed  Google Scholar 

  18. Consortium EP et al (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489(7414):57–74

    Google Scholar 

  19. Costa VL et al (2010) Epigenetic regulation of Wnt signaling pathway in urological cancer. Epigenetics 5(4):343–351

    CAS  PubMed  Google Scholar 

  20. Baylin SB, Ohm JE (2006) Epigenetic gene silencing in cancer—a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 6(2):107–116

    CAS  PubMed  Google Scholar 

  21. Lind GE et al (2004) A CpG island hypermethylation profile of primary colorectal carcinomas and colon cancer cell lines. Mol Cancer 3:28

    PubMed Central  PubMed  Google Scholar 

  22. Yegnasubramanian S et al (2004) Hypermethylation of CpG islands in primary and metastatic human prostate cancer. Cancer Res 64(6):1975–1986

    CAS  PubMed  Google Scholar 

  23. Jeronimo C et al (2004) A quantitative promoter methylation profile of prostate cancer. Clin Cancer Res 10(24):8472–8478

    CAS  PubMed  Google Scholar 

  24. Kang GH et al (2004) Aberrant CpG island hypermethylation of multiple genes in prostate cancer and prostatic intraepithelial neoplasia. J Pathol 202(2):233–240

    CAS  PubMed  Google Scholar 

  25. Cho NY et al (2007) Hypermethylation of CpG island loci and hypomethylation of LINE-1 and Alu repeats in prostate adenocarcinoma and their relationship to clinicopathological features. J Pathol 211(3):269–277

    CAS  PubMed  Google Scholar 

  26. Rosenbaum E et al (2005) Promoter hypermethylation as an independent prognostic factor for relapse in patients with prostate cancer following radical prostatectomy. Clin Cancer Res 11(23):8321–8325

    CAS  PubMed  Google Scholar 

  27. Henrique R et al (2007) High promoter methylation levels of APC predict poor prognosis in sextant biopsies from prostate cancer patients. Clin Cancer Res 13(20):6122–6129

    CAS  PubMed  Google Scholar 

  28. Tokumaru Y et al (2004) Optimal use of a panel of methylation markers with GSTP1 hypermethylation in the diagnosis of prostate adenocarcinoma. Clin Cancer Res 10(16):5518–5522

    CAS  PubMed  Google Scholar 

  29. Maruyama R et al (2002) Aberrant promoter methylation profile of prostate cancers and its relationship to clinicopathological features. Clin Cancer Res 8(2):514–519

    CAS  PubMed  Google Scholar 

  30. Enokida H et al (2005) Multigene methylation analysis for detection and staging of prostate cancer. Clin Cancer Res 11(18):6582–6588

    CAS  PubMed  Google Scholar 

  31. Bastian PJ et al (2007) Prognostic value of preoperative serum cell-free circulating DNA in men with prostate cancer undergoing radical prostatectomy. Clin Cancer Res 13(18 Pt 1):5361–5367

    CAS  PubMed  Google Scholar 

  32. Bastian PJ et al (2004) GSTP1 hypermethylation as a molecular marker in the diagnosis of prostatic cancer: is there a correlation with clinical stage, Gleason grade, PSA value or age? Eur J Med Res 9(11):523–527

    CAS  PubMed  Google Scholar 

  33. Florl AR et al (2004) Coordinate hypermethylation at specific genes in prostate carcinoma precedes LINE-1 hypomethylation. Br J Cancer 91(5):985–994

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Cho NY et al (2009) Genomic hypomethylation and CpG island hypermethylation in prostatic intraepithelial neoplasm. Virchows Arch 454(1):17–23

    CAS  PubMed  Google Scholar 

  35. Bastian PJ et al (2005) Diagnostic and prognostic information in prostate cancer with the help of a small set of hypermethylated gene loci. Clin Cancer Res 11(11):4097–4106

    CAS  PubMed  Google Scholar 

  36. Henrique R et al (2006) Epigenetic heterogeneity of high-grade prostatic intraepithelial neoplasia: clues for clonal progression in prostate carcinogenesis. Mol Cancer Res 4(1):1–8

    CAS  PubMed  Google Scholar 

  37. Bastian PJ et al (2007) Prognostic value of CpG island hypermethylation at PTGS2, RAR-beta, EDNRB, and other gene loci in patients undergoing radical prostatectomy. Eur Urol 51(3):665–674, discussion 674

    CAS  PubMed  Google Scholar 

  38. Ellinger J et al (2008) CpG island hypermethylation at multiple gene sites in diagnosis and prognosis of prostate cancer. Urology 71(1):161–167

    PubMed  Google Scholar 

  39. Richiardi L et al (2009) Promoter methylation in APC, RUNX3, and GSTP1 and mortality in prostate cancer patients. J Clin Oncol 27(19):3161–3168

    CAS  PubMed  Google Scholar 

  40. Steiner I et al (2010) Gene promoter methylation and its potential relevance in early prostate cancer diagnosis. Pathobiology 77(5):260–266

    CAS  PubMed  Google Scholar 

  41. Vasiljevic N et al (2011) Absolute quantitation of DNA methylation of 28 candidate genes in prostate cancer using pyrosequencing. Dis Markers 30(4):151–161

    CAS  PubMed  Google Scholar 

  42. Yaqinuddin A et al (2013) Frequent DNA hypermethylation at the RASSF1A and APC gene loci in prostate cancer patients of Pakistani origin. ISRN Urol 2013:627249

    PubMed Central  PubMed  Google Scholar 

  43. Moritz R et al (2013) DNA hypermethylation as a predictor of PSA recurrence in patients with low- and intermediate-grade prostate cancer. Anticancer Res 33(12):5249–5254

    CAS  PubMed  Google Scholar 

  44. Tang D et al (2013) Methylation of the RARB gene increases prostate cancer risk in black Americans. J Urol 190(1):317–324

    CAS  PubMed Central  PubMed  Google Scholar 

  45. Hoque MO et al (2005) Quantitative methylation-specific polymerase chain reaction gene patterns in urine sediment distinguish prostate cancer patients from control subjects. J Clin Oncol 23(27):6569–6575

    CAS  PubMed  Google Scholar 

  46. Roupret M et al (2007) Molecular detection of localized prostate cancer using quantitative methylation-specific PCR on urinary cells obtained following prostate massage. Clin Cancer Res 13(6):1720–1725

    CAS  PubMed  Google Scholar 

  47. Vener T et al (2008) Development of a multiplexed urine assay for prostate cancer diagnosis. Clin Chem 54(5):874–882

    CAS  PubMed  Google Scholar 

  48. Rogers CG et al (2006) High concordance of gene methylation in post-digital rectal examination and post-biopsy urine samples for prostate cancer detection. J Urol 176(5):2280–2284

    CAS  PubMed  Google Scholar 

  49. Roupret M et al (2008) Promoter hypermethylation in circulating blood cells identifies prostate cancer progression. Int J Cancer 122(4):952–956

    CAS  PubMed  Google Scholar 

  50. Okegawa T, Nutahara K, Higashihara E (2010) Association of circulating tumor cells with tumor-related methylated DNA in patients with hormone-refractory prostate cancer. Int J Urol 17(5):466–475

    CAS  PubMed  Google Scholar 

  51. Henderson BE et al (1982) Endogenous hormones as a major factor in human cancer. Cancer Res 42(8):3232–3239

    CAS  PubMed  Google Scholar 

  52. Henderson BE, Ross RK, Pike MC (1991) Toward the primary prevention of cancer. Science 254(5035):1131–1138

    CAS  PubMed  Google Scholar 

  53. Ellem SJ, Risbridger GP (2010) Aromatase and regulating the estrogen:androgen ratio in the prostate gland. J Steroid Biochem Mol Biol 118(4–5):246–251

    CAS  PubMed  Google Scholar 

  54. Wang Q et al (2009) Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138(2):245–256

    CAS  PubMed Central  PubMed  Google Scholar 

  55. Eder IE et al (2000) Inhibition of LncaP prostate cancer cells by means of androgen receptor antisense oligonucleotides. Cancer Gene Ther 7(7):997–1007

    CAS  PubMed  Google Scholar 

  56. Mitchell SH, Zhu W, Young CY (1999) Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells. Cancer Res 59(23):5892–5895

    CAS  PubMed  Google Scholar 

  57. Tong Q et al (2003) Growth inhibiting effects of antisense eukaryotic expression vector of proliferating cell nuclear antigen gene on human bladder cancer cells. Chin Med J (Engl) 116(8):1203–1206

    CAS  Google Scholar 

  58. Heisler LE et al (1997) Androgen-dependent cell cycle arrest and apoptotic death in PC-3 prostatic cell cultures expressing a full-length human androgen receptor. Mol Cell Endocrinol 126(1):59–73

    CAS  PubMed  Google Scholar 

  59. Grossmann ME, Huang H, Tindall DJ (2001) Androgen receptor signaling in androgen-refractory prostate cancer. J Natl Cancer Inst 93(22):1687–1697

    CAS  PubMed  Google Scholar 

  60. Jarrard DF et al (1998) Methylation of the androgen receptor promoter CpG island is associated with loss of androgen receptor expression in prostate cancer cells. Cancer Res 58(23):5310–5314

    CAS  PubMed  Google Scholar 

  61. Yamanaka M et al (2003) Altered methylation of multiple genes in carcinogenesis of the prostate. Int J Cancer 106(3):382–387

    CAS  PubMed  Google Scholar 

  62. Kinoshita H et al (2000) Methylation of the androgen receptor minimal promoter silences transcription in human prostate cancer. Cancer Res 60(13):3623–3630

    CAS  PubMed  Google Scholar 

  63. Sasaki M et al (2002) Methylation and inactivation of estrogen, progesterone, and androgen receptors in prostate cancer. J Natl Cancer Inst 94(5):384–390

    CAS  PubMed  Google Scholar 

  64. Nakayama T et al (2000) Epigenetic regulation of androgen receptor gene expression in human prostate cancers. Lab Invest 80(12):1789–1796

    CAS  PubMed  Google Scholar 

  65. Reibenwein J et al (2007) Promoter hypermethylation of GSTP1, AR, and 14-3-3sigma in serum of prostate cancer patients and its clinical relevance. Prostate 67(4):427–432

    CAS  PubMed  Google Scholar 

  66. Schayek H et al (2010) Progression to metastatic stage in a cellular model of prostate cancer is associated with methylation of the androgen receptor gene and transcriptional suppression of the insulin-like growth factor-I receptor gene. Exp Cell Res 316(9):1479–1488

    CAS  PubMed Central  PubMed  Google Scholar 

  67. Patra SK, Bettuzzi S (2007) Epigenetic DNA-methylation regulation of genes coding for lipid raft-associated components: a role for raft proteins in cell transformation and cancer progression (review). Oncol Rep 17(6):1279–1290

    CAS  PubMed  Google Scholar 

  68. Woodson K et al (2004) Heterogeneous gene methylation patterns among pre-invasive and cancerous lesions of the prostate: a histopathologic study of whole mount prostate specimens. Prostate 60(1):25–31

    CAS  PubMed  Google Scholar 

  69. Woodson K et al (2003) Hypermethylation of GSTP1, CD44, and E-cadherin genes in prostate cancer among US Blacks and Whites. Prostate 55(3):199–205

    CAS  PubMed  Google Scholar 

  70. Gao X, Porter AT, Honn KV (1997) Involvement of the multiple tumor suppressor genes and 12-lipoxygenase in human prostate cancer. Therapeutic implications. Adv Exp Med Biol 407:41–53

    CAS  PubMed  Google Scholar 

  71. Kito H et al (2001) Hypermethylation of the CD44 gene is associated with progression and metastasis of human prostate cancer. Prostate 49(2):110–115

    CAS  PubMed  Google Scholar 

  72. Lou W et al (1999) Methylation of the CD44 metastasis suppressor gene in human prostate cancer. Cancer Res 59(10):2329–2331

    CAS  PubMed  Google Scholar 

  73. Singal R et al (2004) Methylation of multiple genes in prostate cancer and the relationship with clinicopathological features of disease. Oncol Rep 12(3):631–637

    CAS  PubMed  Google Scholar 

  74. Woodson K, Hanson J, Tangrea J (2004) A survey of gene-specific methylation in human prostate cancer among black and white men. Cancer Lett 205(2):181–188

    CAS  PubMed  Google Scholar 

  75. Bastian PJ et al (2008) CpG island hypermethylation profile in the serum of men with clinically localized and hormone refractory metastatic prostate cancer. J Urol 179(2):529–534, discussion 534-5

    CAS  PubMed Central  PubMed  Google Scholar 

  76. Graziano F, Humar B, Guilford P (2003) The role of the E-cadherin gene (CDH1) in diffuse gastric cancer susceptibility: from the laboratory to clinical practice. Ann Oncol 14(12):1705–1713

    CAS  PubMed  Google Scholar 

  77. Li LC et al (2001) Methylation of the E-cadherin gene promoter correlates with progression of prostate cancer. J Urol 166(2):705–709

    CAS  PubMed  Google Scholar 

  78. Yao Q et al (2006) Promotor hypermethylation of E-cadherin, p16 and estrogen receptor in prostate carcinoma. Zhonghua Nan Ke Xue 12(1):28–31

    CAS  PubMed  Google Scholar 

  79. Saha B et al (2008) Unmethylated E-cadherin gene expression is significantly associated with metastatic human prostate cancer cells in bone. Prostate 68(15):1681–1688

    CAS  PubMed  Google Scholar 

  80. Konishi N et al (2002) Heterogeneous methylation and deletion patterns of the INK4a/ARF locus within prostate carcinomas. Am J Pathol 160(4):1207–1214

    CAS  PubMed Central  PubMed  Google Scholar 

  81. Nguyen TT et al (2000) Analysis of cyclin-dependent kinase inhibitor expression and methylation patterns in human prostate cancers. Prostate 43(3):233–242

    CAS  PubMed  Google Scholar 

  82. Gu K et al (1998) Analysis of the p16 tumor suppressor gene in early-stage prostate cancer. Mol Carcinog 21(3):164–170

    CAS  PubMed  Google Scholar 

  83. Jarrard DF et al (1997) Deletional, mutational, and methylation analyses of CDKN2 (p16/MTS1) in primary and metastatic prostate cancer. Genes Chromosomes Cancer 19(2):90–96

    CAS  PubMed  Google Scholar 

  84. Konishi N et al (2002) DNA hypermethylation status of multiple genes in prostate adenocarcinomas. Jpn J Cancer Res 93(7):767–773

    CAS  PubMed  Google Scholar 

  85. Higuchi T et al (2008) HRK inactivation associated with promoter methylation and LOH in prostate cancer. Prostate 68(1):105–113

    CAS  PubMed  Google Scholar 

  86. Herman JG et al (1995) Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res 55(20):4525–4530

    CAS  PubMed  Google Scholar 

  87. Nakamura M et al (2001) p14ARF deletion and methylation in genetic pathways to glioblastomas. Brain Pathol 11(2):159–168

    CAS  PubMed  Google Scholar 

  88. Lin HH et al (2010) Increase sensitivity in detecting superficial, low grade bladder cancer by combination analysis of hypermethylation of E-cadherin, p16, p14, RASSF1A genes in urine. Urol Oncol 28(6):597–602

    Google Scholar 

  89. Chim CS, Chan WW, Kwong YL (2008) Epigenetic dysregulation of the DAP kinase/p14/HDM2/p53/Apaf-1 apoptosis pathway in acute leukaemias. J Clin Pathol 61(7):844–847

    CAS  PubMed  Google Scholar 

  90. Calmon MF et al (2007) Methylation profile of genes CDKN2A (p14 and p16), DAPK1, CDH1, and ADAM23 in head and neck cancer. Cancer Genet Cytogenet 173(1):31–37

    CAS  PubMed  Google Scholar 

  91. Park JY et al (1999) Association between glutathione S-transferase pi genetic polymorphisms and oral cancer risk. Pharmacogenetics 9(4):497–504

    CAS  PubMed  Google Scholar 

  92. Henrique R, Jeronimo C (2004) Molecular detection of prostate cancer: a role for GSTP1 hypermethylation. Eur Urol 46(5):660–669, discussion 669

    CAS  PubMed  Google Scholar 

  93. Nelson CP et al (2001) Protection against 2-hydroxyamino-1-methyl-6-phenylimidazo[4,5-b]pyridine cytotoxicity and DNA adduct formation in human prostate by glutathione S-transferase P1. Cancer Res 61(1):103–109

    CAS  PubMed  Google Scholar 

  94. Richiardi L et al (2013) Methylation of APC and GSTP1 in non-neoplastic tissue adjacent to prostate tumour and mortality from prostate cancer. PLoS One 8(7):e68162

    CAS  PubMed Central  PubMed  Google Scholar 

  95. Lee WH et al (1994) Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A 91(24):11733–11737

    CAS  PubMed Central  PubMed  Google Scholar 

  96. Lee WH et al (1997) CG island methylation changes near the GSTP1 gene in prostatic carcinoma cells detected using the polymerase chain reaction: a new prostate cancer biomarker. Cancer Epidemiol Biomarkers Prev 6(6):443–450

    CAS  PubMed  Google Scholar 

  97. Santourlidis S et al (1999) High frequency of alterations in DNA methylation in adenocarcinoma of the prostate. Prostate 39(3):166–174

    CAS  PubMed  Google Scholar 

  98. Jeronimo C et al (2001) Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ-confined prostate adenocarcinoma. J Natl Cancer Inst 93(22):1747–1752

    CAS  PubMed  Google Scholar 

  99. Kollermann J et al (2003) Methylation-specific PCR for DNA-based detection of occult tumor cells in lymph nodes of prostate cancer patients. Eur Urol 44(5):533–538

    PubMed  Google Scholar 

  100. Lin X et al (2001) GSTP1 CpG island hypermethylation is responsible for the absence of GSTP1 expression in human prostate cancer cells. Am J Pathol 159(5):1815–1826

    CAS  PubMed Central  PubMed  Google Scholar 

  101. Chu DC et al (2002) The use of real-time quantitative polymerase chain reaction to detect hypermethylation of the CpG islands in the promoter region flanking the GSTP1 gene to diagnose prostate carcinoma. J Urol 167(4):1854–1858

    CAS  PubMed  Google Scholar 

  102. Zhou M et al (2004) Quantitative GSTP1 methylation levels correlate with Gleason grade and tumor volume in prostate needle biopsies. J Urol 171(6 Pt 1):2195–2198

    CAS  PubMed  Google Scholar 

  103. Bernardini S et al (2004) Hypermethylation of the CpG islands in the promoter region of the GSTP1 gene in prostate cancer: a useful diagnostic and prognostic marker? Clin Chim Acta 350(1–2):181–188

    CAS  PubMed  Google Scholar 

  104. Bastian PJ et al (2005) Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy. Clin Cancer Res 11(11):4037–4043

    CAS  PubMed  Google Scholar 

  105. Yegnasubramanian S et al (2006) Combination of methylated-DNA precipitation and methylation-sensitive restriction enzymes (COMPARE-MS) for the rapid, sensitive and quantitative detection of DNA methylation. Nucleic Acids Res 34(3):e19

    PubMed Central  PubMed  Google Scholar 

  106. Vanaja DK et al (2006) PDLIM4 repression by hypermethylation as a potential biomarker for prostate cancer. Clin Cancer Res 12(4):1128–1136

    CAS  PubMed  Google Scholar 

  107. Eilers T et al (2007) Prospective diagnostic efficiency of biopsy washing DNA GSTP1 island hypermethylation for detection of adenocarcinoma of the prostate. Prostate 67(7):757–763

    CAS  PubMed  Google Scholar 

  108. Perry AS et al (2007) In silico mining identifies IGFBP3 as a novel target of methylation in prostate cancer. Br J Cancer 96(10):1587–1594

    CAS  PubMed Central  PubMed  Google Scholar 

  109. Zon G et al (2009) Formamide as a denaturant for bisulfite conversion of genomic DNA: bisulfite sequencing of the GSTPi and RARbeta2 genes of 43 formalin-fixed paraffin-embedded prostate cancer specimens. Anal Biochem 392(2):117–125

    CAS  PubMed  Google Scholar 

  110. Ahmed H et al (2009) Evidence of heavy methylation in the galectin 3 promoter in early stages of prostate adenocarcinoma: development and validation of a methylated marker for early diagnosis of prostate cancer. Transl Oncol 2(3):146–156

    PubMed Central  PubMed  Google Scholar 

  111. Devaney J et al (2011) Epigenetic deregulation across chromosome 2q14.2 differentiates normal from prostate cancer and provides a regional panel of novel DNA methylation cancer biomarkers. Cancer Epidemiol Biomarkers Prev 20(1):148–159

    CAS  PubMed  Google Scholar 

  112. Goessl C et al (2001) DNA-based detection of prostate cancer in blood, urine, and ejaculates. Ann N Y Acad Sci 945:51–58

    CAS  PubMed  Google Scholar 

  113. Jeronimo C et al (2002) Quantitative GSTP1 hypermethylation in bodily fluids of patients with prostate cancer. Urology 60(6):1131–1135

    PubMed  Google Scholar 

  114. Goessl C et al (2002) Methylation-specific PCR for detection of neoplastic DNA in biopsy washings. J Pathol 196(3):331–334

    CAS  PubMed  Google Scholar 

  115. Van Neste L et al (2012) The epigenetic promise for prostate cancer diagnosis. Prostate 72(11):1248–1261

    PubMed  Google Scholar 

  116. Cairns P et al (2001) Molecular detection of prostate cancer in urine by GSTP1 hypermethylation. Clin Cancer Res 7(9):2727–2730

    CAS  PubMed  Google Scholar 

  117. Gonzalgo ML et al (2003) Prostate cancer detection by GSTP1 methylation analysis of postbiopsy urine specimens. Clin Cancer Res 9(7):2673–2677

    CAS  PubMed  Google Scholar 

  118. Payne SR et al (2009) DNA methylation biomarkers of prostate cancer: confirmation of candidates and evidence urine is the most sensitive body fluid for non-invasive detection. Prostate 69(12):1257–1269

    Google Scholar 

  119. Ellinger J et al (2008) CpG island hypermethylation in cell-free serum DNA identifies patients with localized prostate cancer. Prostate 68(1):42–49

    CAS  PubMed  Google Scholar 

  120. Suh CI et al (2000) Comparison of telomerase activity and GSTP1 promoter methylation in ejaculate as potential screening tests for prostate cancer. Mol Cell Probes 14(4):211–217

    CAS  PubMed  Google Scholar 

  121. Friedberg EC (2001) How nucleotide excision repair protects against cancer. Nat Rev Cancer 1(1):22–33

    CAS  PubMed  Google Scholar 

  122. Mullaart E et al (1990) Spontaneous DNA breaks in the rat brain during development and aging. Mutat Res 237(1):9–15

    CAS  PubMed  Google Scholar 

  123. Wood RD et al (2001) Human DNA repair genes. Science 291(5507):1284–1289

    CAS  PubMed  Google Scholar 

  124. Wood RD, Mitchell M, Lindahl T (2005) Human DNA repair genes, 2005. Mutat Res 577(1–2):275–283

    CAS  PubMed  Google Scholar 

  125. Park JY, Huang Y, Sellers TA (2009) Single nucleotide polymorphisms in DNA repair genes and prostate cancer risk. Methods Mol Biol 471:361–385

    CAS  PubMed Central  PubMed  Google Scholar 

  126. Hayashi K et al (2001) Overexpression of retinoic acid receptor beta induces growth arrest and apoptosis in oral cancer cell lines. Jpn J Cancer Res 92(1):42–50

    CAS  PubMed  Google Scholar 

  127. Nakayama T et al (2001) The role of epigenetic modifications in retinoic acid receptor beta2 gene expression in human prostate cancers. Lab Invest 81(7):1049–1057

    CAS  PubMed  Google Scholar 

  128. Zhang J, Liu L, Pfeifer GP (2004) Methylation of the retinoid response gene TIG1 in prostate cancer correlates with methylation of the retinoic acid receptor beta gene. Oncogene 23(12):2241–2249

    CAS  PubMed  Google Scholar 

  129. Jeronimo C et al (2004) Quantitative RARbeta2 hypermethylation: a promising prostate cancer marker. Clin Cancer Res 10(12 Pt 1):4010–4014

    CAS  PubMed  Google Scholar 

  130. Kuzmin I et al (2002) The RASSF1A tumor suppressor gene is inactivated in prostate tumors and suppresses growth of prostate carcinoma cells. Cancer Res 62(12):3498–3502

    CAS  PubMed  Google Scholar 

  131. Li QL et al (2002) Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 109(1):113–124

    CAS  PubMed  Google Scholar 

  132. Liu L et al (2002) Frequent hypermethylation of the RASSF1A gene in prostate cancer. Oncogene 21(44):6835–6840

    CAS  PubMed  Google Scholar 

  133. Kawamoto K et al (2007) Epigenetic modifications of RASSF1A gene through chromatin remodeling in prostate cancer. Clin Cancer Res 13(9):2541–2548

    CAS  PubMed  Google Scholar 

  134. Aitchison A et al (2007) RASSF1A promoter methylation is frequently detected in both pre-malignant and non-malignant microdissected prostatic epithelial tissues. Prostate 67(6):638–644

    CAS  PubMed  Google Scholar 

  135. Syeed N et al (2010) Promoter methylation profile of GSTP1 and RASSF1A in benign hyperplasia and metastatic prostate cancer patients in a Kashmiri population. Mol Med Rep 3(5):883–887

    CAS  PubMed  Google Scholar 

  136. Pan J et al (2013) Association between RASSF1A promoter methylation and prostate cancer: a systematic review and meta-analysis. PLoS One 8(9):e75283

    CAS  PubMed Central  PubMed  Google Scholar 

  137. Mack GS (2006) Epigenetic cancer therapy makes headway. J Natl Cancer Inst 98(20):1443–1444

    PubMed  Google Scholar 

  138. Muller CI et al (2006) DNA hypermethylation of myeloid cells, a novel therapeutic target in MDS and AML. Curr Pharm Biotechnol 7(5):315–321

    PubMed  Google Scholar 

  139. Oki Y, Aoki E, Issa JP (2007) Decitabine–bedside to bench. Crit Rev Oncol Hematol 61(2):140–152

    PubMed  Google Scholar 

  140. Fenaux P et al (2009) Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 10(3):223–232

    CAS  PubMed Central  PubMed  Google Scholar 

  141. Silverman LR et al (2002) Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol 20(10):2429–2440

    CAS  PubMed  Google Scholar 

  142. Muller A, Florek M (2010) 5-Azacytidine/Azacitidine. Recent Results Cancer Res 184:159–170

    PubMed  Google Scholar 

  143. Kim YJ et al (2013) Use of azacitidine for myelodysplastic syndromes: controversial issues and practical recommendations. Blood Res 48(2):87–98

    CAS  PubMed Central  PubMed  Google Scholar 

  144. Harden SV et al (2003) Quantitative GSTP1 methylation and the detection of prostate adenocarcinoma in sextant biopsies. J Natl Cancer Inst 95(21):1634–1637

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jong Y. Park Ph.D., M.P.H. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2015 Springer Science+Business Media New York

About this protocol

Cite this protocol

Park, J.Y. (2015). Promoter Hypermethylation as a Biomarker in Prostate Adenocarcinoma. In: Verma, M. (eds) Cancer Epigenetics. Methods in Molecular Biology, vol 1238. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-1804-1_32

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-1804-1_32

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-1803-4

  • Online ISBN: 978-1-4939-1804-1

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics