poster slide final

1
A Review of the Expression of Genes Involved in Sex Steroid Hormone Metabolism and Activity in Prostate Tissue: A Need for Epigenetic Information Jamie Ritchey 1 , MPH; Wilfried Karmaus 1 , MD, Dr.med, MPH; Tara Sabo-Attwood 1,2 , PhD; Susan E. Steck 1,3 , PhD, MPH, RD; Hongmei Zhang 1 , PhD; 1 Department of Epidemiology and Biostatistics, 2 Department of Environmental Health Sciences, 3 Cancer Prevention and Control Program, University of South Carolina, 800 Sumter Street, Columbia, SC 29208. INTRODUCTION A pathway-oriented approach guided the literature searches, conducted in Pub Med, limited to English language and human prostate only OBJECTIVE To review the literature for gene expression of enzymes and receptors involved in sex steroid hormone metabolism and activity in normal, benign, and cancerous prostate tissues To determine if there is an association with gene expression and age, body mass index, estrogen and testosterone levels, and cancer treatments METHODS RESULTS REFERENCES RATIONALE Age, race, and family history are the only confirmed risk factors for prostate cancer [1] Observational, clinical, and laboratory evidence indicate that sex steroid hormones are important to the development and progression of prostate cancer [2-14] Epidemiology focusing on sex steroid hormone risk and prostate cancer is inconclusive [2-14] Most epidemiologic studies have focused on gene single nucleotide polymorphisms (SNPs) of hormone metabolic enzymes and serum hormone levels [2-3] Gene expression may provide etiologic clues regarding how sex steroid metabolism is associated with prostate cancer not detected at the DNA sequence or serum level [130] The expression of genes involved in sex steroid metabolism in prostate tissues may differ by different levels of age, body mass index, race, estrogen and testosterone levels, and/or cancer treatments administered STRENGTHS LIMITATIONS To guide our searches, we constructed a figure of a plausible gene network for E and T expression [3,11, 19-20]. Past studies have only included portions of pathways, or the T or E pathways alone, and often do not include preliminary pathways (the dashed line from androstenedione to DHT indicates that this reaction is preliminary) [3,11,19-20]. Used a pathway approach to guide searches Omitted cell studies since results may not be consistent with tissue studies for methodological reasons Reviewed references cited in publications found during searches to improve completeness Information for many genes was scant Only publications in English language were considered Only includes citations from PubMed SUMMARY GSTP1 was consistently down regulated or not expressed in prostate cancer, which coincides with previous research indicating that GSTP1 is methylated in prostate cancer tissue [16,95,99-109,111-118,130,136] For all other genes, studies were either scant or inconsistent, and conflicting expression results may be due to limitations, including: tissue collection, laboratory methods, or gene functionality Few studies were found examining the association of gene expression with age, race, body mass index, estrogen, testosterone, and cancer treatments Future studies should focus not only on gene expression, but also epigenetic mechanisms such as methylation to assess prostate cancer risk Table 1. Literature search summary Gene & Chromosomal location Total articles retrieved Laboratory methods† Androgen Metabolism AR Ch Xq12 26 IHC, Microarrays, IS, ISV, real-time PCR HSD17B2 Ch 16q24.1-q24.2 4 IHC, ISH, Microarray, real-time PCR HSD17B3 Ch 9q22 4 IHC, microarray, real-time PCR SRD5A1 Ch 5p15.31 6 IHC, ISH, Microarray SRD5A2 Ch 2p23.1 10 IHC, ISH, Microarray, real-time PCR CYP3A4 Ch 7q21.1-22 5 IHC, real-time PCR CYP3A5 Ch 7q21.1-22 6 IHC, real-time PCR, meta-analysis CYP3A7 Ch 7q21.1-22 3 IHC, real-time PCR CYP3A43 Ch 7q21.1 1 ICH, real-time PCR AKR1C3 Ch 10p15-14 6 Array, real-time PCR, Northern blot, review AKR1C2 Ch 10p15-14 5 Array, real-time PCR, review HSD3B1 Ch 1p13-11 2 Array, real-time PCR HSD3B2 Ch 1p13.1 3 Array, review UGT2B15 Ch 4q13 3 Array, microarray UGT2B17 Ch 4q13 3 IHC, ISH, real-time PCR Estrogen Metabolism ESR1 Ch 6q24-27 9 IHC, ISH, real-time PCR ESR2 Ch 14q31-22 10 IHC, ISH, microarray, real-time PCR CYP19A1 Ch 15q21 9 Avidin-biotin, IHC, ISH, microarray, real-time PCR HSD17B1 Ch 17q11-21 2 IHC, real-time PCR HSD17B4 Ch 5q2 5 IHC, ISH, microarray, real-time PCR, meta-analysis HSD17B7 Ch 1q23 1 IHC SULT1A1 Ch 16p21.1 3 IHC, Western blot SULT1A3 Ch 16p11.2 2 IHC, Western blot SULT2B1a Ch 19 q13.3 3 Northern blot, immunoblot, IHC SULT2Bb Ch 19 q13.3 3 Northern blot, immunoblot, IHC HSD17B12 Ch 11p11 1 IHC, ISH CYP1A1 Ch 15q24.1 4 IHC, real-time PCR CYP1A2 Ch 15q24.1 4 IHC, real-time PCR CYP1B1 Ch 2p22.2 6 IHC, ISH, real-time PCR COMT Ch 22q11.21 2 IHC, real-time PCR GSTT1 Ch 22q11.23 1 Microarray, real-time PCR GSTM1 Ch 1p13.1 3 IHC, microarray, real-time PCR GSTP1 Ch 11q13.2 10 IHC, IS, microarray, real-time PCR †Laboratory method abbreviations: Immunohistochemistry (IHC), Real-time polymerase chain reaction (real-time PCR), Immunostaining (IS), In situ hybridization (ISH), Immunostaining with optimized IHC criteria and video image analysis (ISV) A total of 85 studies were identified Most studies focused on the expression of one or a few genes, rather than the complete pathway, ignoring compensatory or alternate response Studies comparing normal, benign, and cancerous prostate tissue showed limited consistency, with one exception, the down-regulation of GSTP1 in cancerous tissue 1. American Cancer Society. Cancer Facts & Figures 2008. Atlanta: American Cancer Society, 2008. 2. Chu, L.W., J.K. Reichardt, and A.W. Hsing, Androgens and the molecular epidemiology of prostate cancer. Curr Opin Endocrinol Diabetes Obes, 2008. 15(3): p. 261-70. 3. Chokkalingam, A.P., et al., Molecular epidemiology of prostate cancer: hormone-related genetic loci. Front Biosci, 2007. 12: p. 3436-60. 4. Pollard, M., P.H. Luckert, and M.A. Schmidt, Induction of prostate adenocarcinomas in Lobund Wistar rats by testosterone. Prostate, 1982. 3(6): p. 563-8. 5. Cavalieri, E.L., et al., Catechol estrogen metabolites and conjugates in different regions of the prostate of Noble rats treated with 4-hydroxyestradiol: implications for estrogen-induced initiation of prostate cancer. Carcinogenesis, 2002. 23(2): p. 329-33. 6. Morgentaler, A., Guilt by association: a historial perspective on Huggins, testosterone therapy, and prostate cancer. J Sex Med, 2008. 5: p. 1834-1840. 7. Sharifi, N., J.L. Gulley, and W.L. Dahut, Androgen deprivation therapy for prostate cancer. Jama, 2005. 294(2): p. 238-44. 8. Hsing, A.W., J.K. Reichardt, and F.Z. Stanczyk, Hormones and prostate cancer: current perspectives and future directions. Prostate., 2002. 52(3): p. 213-35. 9. Huggins, C. and R. Hodges, Studies on prostate cancer: 1. The effects of castration, of estrogen, and androgen injection on serum phosphatases in metastatic carcinoma of the prostate Cancer Research, 1941. 1: p. 203. 10. Cunningham, J.M., et al., Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomarkers Prev, 2007. 16(5): p. 969-78. 11. Hsing, A.W., L.W. Chu, and F.Z. Stanczyk, Androgen and prostate cancer: is the hypothesis dead? Cancer Epidemiol Biomarkers Prev, 2008. 17(10): p. 2525-30. 12. Weiss, J.M., et al., Endogenous sex hormones and the risk of prostate cancer: a prospective study. Int J Cancer, 2008. 122(10): p. 2345-50. 13. Rohrmann, S., et al., Serum estrogen, but not testosterone, levels differ between black and white men in a nationally representative sample of Americans. J Clin Endocrinol Metab, 2007. 92(7): p. 2519-25. 14. Roddam, A.W., et al., Endogenous sex hormones and prostate cancer: a collaborative analysis of 18 prospective studies. J Natl Cancer Inst, 2008. 100(3): p. 170-83. 15. Li, J., et al., Steroid 5-{alpha}-Reductase Type 2 (SRD5a2) Gene Polymorphisms and Risk of Prostate Cancer: A HuGE Review. Am J Epidemiol, 2009. 16. Mo, Z., et al., An updating meta-analysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: a HuGE review. Prostate, 2009. 69(6): p. 662-88. 17. Keshava, C., E.C. McCanlies, and A. Weston, CYP3A4 polymorphisms--potential risk factors for breast and prostate cancer: a HuGE review. Am J Epidemiol, 2004. 160(9): p. 825-41. 18. Takase, Y., et al., Expression of enzymes involved in estrogen metabolism in human prostate. J Histochem Cytochem, 2006. 54(8): p. 911-21. 19. Guillemette, C., A. Belanger, and J. Lepine, Metabolic inactivation of estrogens in breast tissue by UDP-glucuronosyltransferase enzymes: an overview. Breast Cancer Res, 2004. 6(6): p. 246-54. 20. Maglott, D., et al., Entrez Gene: gene-centered information at NCBI. Nucleic Acids Res, 2007. 35(Database issue): p. D26-31. 21. HGNC Database, HUGO Gene Nomenclature Committee (HGNC), EMBL Outstation - Hinxton, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK. (http://www.genenames.org) . June 2010. 22. Lu, N.Z., et al., International Union of Pharmacology. LXV. The pharmacology and classification of the nuclear receptor superfamily: glucocorticoid, mineralocorticoid, progesterone, and androgen receptors. Pharmacol Rev, 2006. 58(4): p. 782-97. 23. van der Kwast, T.H. and B. Tetu, Androgen receptors in untreated and treated prostatic intraepithelial neoplasia. Eur Urol, 1996. 30(2): p. 265-8. 24. Sweat, S.D., et al., Androgen receptor expression in prostate cancer lymph node metastases is predictive of outcome after surgery. J Urol, 1999. 161(4): p. 1233-7. 25. Sweat, S.D., et al., Androgen receptor expression in prostatic intraepithelial neoplasia and cancer. J Urol, 1999. 161(4): p. 1229-32. 26. Olapade-Olaopa, E.O., et al., Androgen receptor protein expression in prostatic tissues in Black and Caucasian men. Prostate, 2004. 59(4): p. 460-8. 27. Mohler, J.L., et al., The androgen axis in recurrent prostate cancer. Clin Cancer Res, 2004. 10(2): p. 440-8. 28. Kim, D., et al., Immunohistochemical quantitation of androgen receptor expression using color video image analysis. Cytometry, 1999. 35(1): p. 2-10. 29. Elo, J.P., et al., Characterization of 17beta-hydroxysteroid dehydrogenase isoenzyme expression in benign and malignant human prostate. Int J Cancer, 1996. 66(1): p. 37-41. 30. Brolin, J., L. Skoog, and P. Ekman, Immunohistochemistry and biochemistry in detection of androgen, progesterone, and estrogen receptors in benign and malignant human prostatic tissue. Prostate, 1992. 20(4): p. 281-95. 31. Gaston, K.E., et al., Racial differences in androgen receptor protein expression in men with clinically localized prostate cancer. J Urol, 2003. 170(3): p. 990-3. 32. Mohler, J.L., et al., Androgen and glucocorticoid receptors in the stroma and epithelium of prostatic hyperplasia and carcinoma. Clin Cancer Res, 1996. 2(5): p. 889-95. 33. Olapade-Olaopa, E.O., et al., Malignant transformation of human prostatic epithelium is associated with the loss of androgen receptor immunoreactivity in the surrounding stroma. Clin Cancer Res, 1999. 5(3): p. 569-76. 34. Rosner, I.L., et al., Higher tumor to benign ratio of the androgen receptor mRNA expression associates with prostate cancer progression after radical prostatectomy. Urology, 2007. 70(6): p. 1225-9. 35. Latil, A., et al., Evaluation of androgen, estrogen (ER alpha and ER beta), and progesterone receptor expression in human prostate cancer by real-time quantitative reverse transcription-polymerase chain reaction assays. Cancer Res, 2001. 61(5): p. 1919-26. 36. Leav, I., et al., Comparative studies of the estrogen receptors beta and alpha and the androgen receptor in normal human prostate glands, dysplasia, and in primary and metastatic carcinoma. Am J Pathol, 2001. 159(1): p. 79-92. 37. Qiu, Y.Q., I. Leuschner, and P.M. Braun, Androgen receptor expression in clinically localized prostate cancer: immunohistochemistry study and literature review. Asian J Androl, 2008. 10(6): p. 855-63. 38. Stanbrough, M., et al., Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res, 2006. 66(5): p. 2815-25. 39. Mohler, J.L., A role for the androgen-receptor in clinically localized and advanced prostate cancer. Best Pract Res Clin Endocrinol Metab, 2008. 22(2): p. 357-72. 40. Pertschuk, L.P., et al., Immunostaining for prostate cancer androgen receptor in paraffin identifies a subset of men with a poor prognosis. Lab Invest, 1995. 73(2): p. 302-5. 41. Koh, E., et al., Differential expression of 17beta-hydroxysteroid dehydrogenase isozyme genes in prostate cancer and noncancer tissues. Prostate, 2002. 53(2): p. 154-9. 42. Febbo, P.G., et al., Neoadjuvant docetaxel before radical prostatectomy in patients with high-risk localized prostate cancer. Clin Cancer Res, 2005. 11(14): p. 5233-40. 43. Montgomery, R.B., et al., Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res, 2008. 68(11): p. 4447-54. 44. Lunacek, A., et al., Fetal distribution of 5alpha-reductase 1 and 5alpha-reductase 2, and their input on human prostate development. J Urol, 2007. 178(2): p. 716-21. 45. Xu, Y., et al., Pharmacologic basis for the enhanced efficacy of dutasteride against prostatic cancers. Clin Cancer Res, 2006. 12(13): p. 4072-9. 46. Bonkhoff, H., et al., Differential expression of 5 alpha-reductase isoenzymes in the human prostate and prostatic carcinomas. Prostate, 1996. 29(4): p. 261-7. 47. Luo, J., et al., Decreased gene expression of steroid 5 alpha-reductase 2 in human prostate cancer: implications for finasteride therapy of prostate carcinoma. Prostate, 2003. 57(2): p. 134-9. 48. Ji, Q., et al., Selective reduction of AKR1C2 in prostate cancer and its role in DHT metabolism. Prostate, 2003. 54(4): p. 275-89. 49. Iehle, C., et al., Differences in steroid 5alpha-reductase iso-enzymes expression between normal and pathological human prostate tissue. J Steroid Biochem Mol Biol, 1999. 68(5-6): p. 189-95. 50. Bjelfman, C., et al., Differential gene expression of steroid 5 alpha-reductase 2 in core needle biopsies from malignant and benign prostatic tissue. J Clin Endocrinol Metab, 1997. 82(7): p. 2210-4. 51. Habib, F.K., et al., The loss of 5alpha-reductase type I and type II mRNA expression in metastatic prostate cancer to bone and lymph node metastasis. Clin Cancer Res, 2003. 9(5): p. 1815-9. 52. Daly, A.K., Significance of the minor cytochrome P450 3A isoforms. Clin Pharmacokinet, 2006. 45(1): p. 13-31. 53. Koch, I., et al., Interindividual variability and tissue-specificity in the expression of cytochrome P450 3A mRNA. Drug Metab Dispos, 2002. 30(10): p. 1108-14. 54. Leskela, S., et al., Cytochrome P450 3A5 is highly expressed in normal prostate cells but absent in prostate cancer. Endocr Relat Cancer, 2007. 14(3): p. 645-54. 55. Yokose, T., et al., Immunohistochemical study of cytochrome P450 2C and 3A in human non-neoplastic and neoplastic tissues. Virchows Arch, 1999. 434(5): p. 401-11. 56. Di Paolo, O.A., et al., Expression of cytochromes P450 and glutathione S-transferases in human prostate, and the potential for activation of heterocyclic amine carcinogens via acetyl-coA-, PAPS- and ATP-dependent pathways. Int J Cancer, 2005. 117(1): p. 8-13. 57. Finnstrom, N., et al., Detection of cytochrome P450 mRNA transcripts in prostate samples by RT-PCR. Eur J Clin Invest, 2001. 31(10): p. 880-6. 58. Gorlov, I.P., et al., Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data. BMC Med Genomics, 2009. 2: p. 48. 59. Moilanen, A.M., et al., Characterization of androgen-regulated expression of CYP3A5 in human prostate. Carcinogenesis, 2007. 28(5): p. 916-21. 60. Matsuura, K., et al., Identification of a principal mRNA species for human 3alpha-hydroxysteroid dehydrogenase isoform (AKR1C3) that exhibits high prostaglandin D2 11-ketoreductase activity. J Biochem, 1998. 124(5): p. 940-6. 61. Penning, T.M., et al., Human 3alpha-hydroxysteroid dehydrogenase isoforms (AKR1C1-AKR1C4) of the aldo-keto reductase superfamily: functional plasticity and tissue distribution reveals roles in the inactivation and formation of mal e and female sex hormones. Biochem J, 2000. 351(Pt 1): p. 67-77. 62. Penning, T.M. and M.C. Byrns, Steroid hormone transforming aldo-keto reductases and cancer. Ann N Y Acad Sci, 2009. 1155: p. 33-42. 63. Penning, T.M., et al., Structure-function aspects and inhibitor design of type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3). Mol Cell Endocrinol, 2001. 171(1-2): p. 137-49. 65. Neslund-Dudas, C., et al., SRD5A2 and HSD3B2 polymorphisms are associated with prostate cancer risk and aggressiveness. Prostate, 2007. 67(15): p. 1654-63. 66. Febbo, P.G., et al., Androgen mediated regulation and functional implications of fkbp51 expression in prostate cancer. J Urol, 2005. 173(5): p. 1772-7. 67. Barbier, O., et al., Cellular localization of uridine diphosphoglucuronosyltransferase 2B enzymes in the human prostate by in situ hybridization and immunohistochemistry. J Clin Endocrinol Metab, 2000. 85(12): p. 4819-26. 68. Park, J., et al., Deletion polymorphism of UDP-glucuronosyltransferase 2B17 and risk of prostate cancer in African American and Caucasian men. Cancer Epidemiol Biomarkers Prev, 2006. 15(8): p. 1473-8. 69. Prins, G.S. and K.S. Korach, The role of estrogens and estrogen receptors in normal prostate growth and disease. Steroids, 2008. 73(3): p. 233-44. 70. O'Dowd, B.F., et al., Discovery of three novel G-protein-coupled receptor genes. Genomics, 1998. 47(2): p. 310-3. 71. Fromont, G., et al., Differential expression of genes related to androgen and estrogen metabolism in hereditary versus sporadic prostate cancer. Cancer Epidemiol Biomarkers Prev, 2008. 17(6): p. 1505-9. 72. Ito, T., et al., Expression of estrogen receptor (ER-alpha and ER-beta) mRNA in human prostate cancer. Eur Urol, 2001. 40(5): p. 557-63. 73. Singh, P.B., et al., Quantified gene expression levels for phase I/II metabolizing enzyme and estrogen receptor levels in benign prostate from cohorts designated as high-risk (UK) versus low-risk (India) for adenocarcinoma at this organ site: a preliminary study. Asian J Androl, 2009. 74. Torlakovic, E., et al., Differential expression of steroid receptors in prostate tissues before and after radiation therapy for prostatic adenocarcinoma. Int J Cancer, 2005. 117(3): p. 381-6. 75. Yang, G.S., et al., Expression of oestrogen receptor-alpha and oestrogen receptor-beta in prostate cancer. Chin Med J (Engl), 2007. 120(18): p. 1611-5. 76. Fixemer, T., K. Remberger, and H. Bonkhoff, Differential expression of the estrogen receptor beta (ERbeta) in human prostate tissue, premalignant changes, and in primary, metastatic, and recurrent prostatic adenocarcinoma. Prostate, 2003. 54(2): p. 79-87. 77. Haqq, C., et al., Ethnic and racial differences in prostate stromal estrogen receptor alpha. Prostate, 2005. 65(2): p. 101-9. 78. Ellem, S.J., et al., Local aromatase expression in human prostate is altered in malignancy. J Clin Endocrinol Metab, 2004. 89(5): p. 2434-41. 79. Matzkin, H. and M.S. Soloway, Immunohistochemical evidence of the existence and localization of aromatase in human prostatic tissues. Prostate, 1992. 21(4): p. 309-14. 80. Hiramatsu, M., et al., Aromatase in hyperplasia and carcinoma of the human prostate. Prostate, 1997. 31(2): p. 118-24. 81. Gross, M., et al., Expression of androgen and estrogen related proteins in normal weight and obese prostate cancer patients. Prostate, 2009. 69(5): p. 520-7. 82. Vihko, P., et al., 17 beta-hydroxysteroid dehydrogenases--their role in pathophysiology. Mol Cell Endocrinol, 2004. 215(1-2): p. 83-8. 83. Rasiah, K.K., et al., HSD17B4 overexpression, an independent biomarker of poor patient outcome in prostate cancer. Mol Cell Endocrinol, 2009. 301(1-2): p. 89-96. 84. Kristiansen, G., et al., Expression profiling of microdissected matched prostate cancer samples reveals CD166/MEMD and CD24 as new prognostic markers for patient survival. J Pathol, 2005. 205(3): p. 359-76. 85. Williams, J.A., et al., Metabolic activation of carcinogens and expression of various cytochromes P450 in human prostate tissue. Carcinogenesis, 2000. 21(9): p. 1683-9. 86. Ragavan, N., et al., CYP1B1 expression in prostate is higher in the peripheral than in the transition zone. Cancer Letters, 2004. 215(1): p. 69-78. 87. John, K., et al., Quantification of phase I/II metabolizing enzyme gene expression and polycyclic aromatic hydrocarbon-DNA adduct levels in human prostate. Prostate, 2009. 69(5): p. 505-19. 88. Martin, F.L., et al., Constitutive expression of bioactivating enzymes in normal human prostate suggests a capability to activate pro-carcinogens to DNA-damaging metabolites. Prostate, 2010. 70(14): p. 1586-99. 89. Singh, P.B., et al., Quantified gene expression levels for phase I/II metabolizing enzyme and estrogen receptor levels in benign prostate from cohorts designated as high-risk (UK) versus low-risk (India) for adenocarcinoma at this organ site: a preliminary study. Asian J Androl, 2010. 12(2): p. 203-14. 90. Al-Buheissi, S.Z., et al., N-Acetyltransferase and sulfotransferase activity in human prostate: potential for carcinogen activation. Pharmacogenet Genomics, 2006. 16(6): p. 391-9. 91. Geese, W.J. and R.B. Raftogianis, Biochemical characterization and tissue distribution of human SULT2B1. Biochem Biophys Res Commun, 2001. 288(1): p. 280-9. 92. He, D., et al., Different subcellular localization of sulphotransferase 2B1b in human placenta and prostate. Biochem J, 2004. 379(Pt 3): p. 533-40. 93. Meloche, C.A. and C.N. Falany, Expression and characterization of the human 3 beta-hydroxysteroid sulfotransferases (SULT2B1a and SULT2B1b). J Steroid Biochem Mol Biol, 2001. 77(4-5): p. 261-9. 94. Elek, J., K.H. Park, and R. Narayanan, Microarray-based expression profiling in prostate tumors. In Vivo, 2000. 14(1): p. 173-82. 95. Bostwick, D.G., I. Meiers, and J.H. Shanks, Glutathione S-transferase: differential expression of alpha, mu, and pi isoenzymes in benign prostate, prostatic intraepithelial neoplasia, and prostatic adenocarcinoma. Hu m Pathol, 2007. 38(9): p. 1394-401. 96. Chetcuti, A., et al., Identification of differentially expressed genes in organ-confined prostate cancer by gene expression array. Prostate, 2001. 47(2): p. 132-40. 97. Stephenson, A.J., et al., Integration of gene expression profiling and clinical variables to predict prostate carcinoma recurrence after radical prostatectomy. Cancer, 2005. 104(2): p. 290-8. 98. Liu, A.J., et al., Quantitative analysis of a panel of gene expression in prostate cancer--with emphasis on NPY expression analysis. J Zhejiang Univ Sci B, 2007. 8(12): p. 853-9. 99. Cookson, M.S., et al., Glutathione S-transferase PI (GST-pi) class expression by immunohistochemistry in benign and malignant prostate tissue. J Urol, 1997. 157(2): p. 673-6. 100. Moskaluk, C.A., et al., Immunohistochemical expression of pi-class glutathione S-transferase is down-regulated in adenocarcinoma of the prostate. Cancer, 1997. 79(8): p. 1595-9. 101. Parsons, J.K., et al., GSTA1 expression in normal, preneoplastic, and neoplastic human prostate tissue. Prostate, 2001. 49(1): p. 30-7. 102. Montironi, R., et al., Expression of pi-class glutathione S-transferase: two populations of high grade prostatic intraepithelial neoplasia with different relations to carcinoma. Mol Pathol, 2000. 53(3): p. 122-8. 103. Montironi, R., et al., Immunohistochemical expression of pi class glutathione S-transferase in the basal cell layer of benign prostate tissue following chronic treatment with finasteride. J Clin Pathol, 1999. 52(5): p. 350-4. 104. Li, M., et al., Glutathione S-transferase pi is upregulated in the stromal compartment of hormone independent prostate cancer. Prostate, 2003. 56(2): p. 98-105. 105. Nelson, W.G., A.M. De Marzo, and S. Yegnasubramanian, Epigenetic alterations in human prostate cancers. Endocrinology, 2009. 150(9): p. 3991-4002. 106. Gilbert, S.F., Ageing and cancer as diseases of epigenesis. J Biosci, 2009. 34(4): p. 601-4. 107. Petronis, A., Epigenetics and twins: three variations on the theme. Trends Genet, 2006. 22(7): p. 347-50. 108. Fraga, M.F. and M. Esteller, Epigenetics and aging: the targets and the marks. Trends Genet, 2007. 23(8): p. 413-8. 109. Liu, R., et al., Functional alterations in the glutathione S-transferase family associated with enhanced occurrence of esophageal carcinoma in China. J Toxicol Environ Health A, 2010. 73(7): p. 471-82. 110. Stephen, J.K., et al., DNA hypermethylation profiles in squamous cell carcinoma of the vulva. Int J Gynecol Pathol, 2009. 28(1): p. 63-75. 111. Yuan, Y., et al., Reduction of GSTP1 expression by DNA methylation correlates with clinicopathological features in pituitary adenomas. Mod Pathol, 2008. 21(7): p. 856-65. 112. Kim, J., et al., Silencing and CpG island methylation of GSTP1 is rare in ordinary gastric carcinomas but common in Epstein-Barr virus-associated gastric carcinomas. Anticancer Res, 2005. 25(6B): p. 4013-9. 113. Brabender, J., et al., Glutathione S-transferase-pi expression is downregulated in patients with Barrett's esophagus and esophageal adenocarcinoma. J Gastrointest Surg, 2002. 6(3): p. 359-67. 114. Laborde, E., Glutathione transferases as mediators of signaling pathways involved in cell proliferation and cell death. Cell Death Differ, 2010. 17(9): p. 1373-80. 115. Tew, K.D., TLK-286: a novel glutathione S-transferase-activated prodrug. Expert Opin Investig Drugs, 2005. 14(8): p. 1047-54. 116. Goldstein, A.S., et al., Identification of a cell of origin for human prostate cancer. Science, 2010. 329(5991): p. 568-71. 117. Bianco-Miotto, T., et al., Global Levels of Specific Histone Modifications and an Epigenetic Gene Signature Predict Prostate Cancer Progression and Development. Cancer Epidemiol Biomarkers Prev, 2010. 118. Cooper, C.S., C. Campbell, and S. Jhavar, Mechanisms of Disease: biomarkers and molecular targets from microarray gene expression studies in prostate cancer. Nat Clin Pract Urol, 2007. 4(12): p. 677-87. 119. Qian, D.Z., et al., Prostate cancer-associated gene expression alterations determined from needle biopsies. Clin Cancer Res, 2009. 15(9): p. 3135-42. 120. Bostwick, D.G., et al., Independent origin of multiple foci of prostatic intraepithelial neoplasia: comparison with matched foci of prostate carcinoma. Cancer, 1998. 83(9): p. 1995-2002. 121. Lin, D.W., et al., Influence of surgical manipulation on prostate gene expression: implications for molecular correlates of treatment effects and disease prognosis. J Clin Oncol, 2006. 24(23): p. 3763-70. 122. True, L. and Z. Feng, Immunohistochemical validation of expression microarray results. J Mol Diagn, 2005. 7(2): p. 149-51. 123. Singh, S.S., et al., Feasibility of constructing tissue microarrays from diagnostic prostate biopsies. Prostate, 2007. 67(10): p. 1011-8. 124. Camp, R.L., V. Neumeister, and D.L. Rimm, A decade of tissue microarrays: progress in the discovery and validation of cancer biomarkers. J Clin Oncol, 2008. 26(34): p. 5630-7. 125. Jhavar, S., et al., Construction of tissue microarrays from prostate needle biopsy specimens. Br J Cancer, 2005. 93(4): p. 478-82. 126. Takase, Y., et al., Expression of sulfotransferase 1E1 in human prostate as studied by in situ hybridization and immunocytochemistry. Prostate, 2007. 67(4): p. 405-9. 127. Ahmed, H., Promoter Methylation in Prostate Cancer and its Application for the Early Detection of Prostate Cancer Using Serum and Urine Samples. Biomark Cancer, 2010. 2010(2): p. 17-33. 128. Baker, M., Epigenome: mapping in motion. Nature Methods, 2010. 7(3): p. 181-185. 129. Kondo, Y., Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J, 2009. 50(4): p. 455-63. 130. Sharma, S., T.K. Kelly, and P.A. Jones, Epigenetics in cancer. Carcinogenesis, 2010. 31(1): p. 27-36. 131. Muhonen, P. and H. Holthofer, Epigenetic and microRNA-mediated regulation in diabetes. Nephrol Dial Transplant, 2009. 24(4): p. 1088-96. 132. Lim, S., et al., Epigenetic regulation of cancer growth by histone demethylases. Int J Cancer, 2010. 127(9): p. 1991-8. 133. Rauhala, H.E., et al., miR-193b is an epigenetically regulated putative tumor suppressor in prostate cancer. Int J Cancer, 2010. 127(6): p. 1363-72. 134. Zhang, H.L., et al., Serum miRNA-21: Elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate, 2010. 135. Pang, Y., C.Y. Young, and H. Yuan, MicroRNAs and prostate cancer. Acta Biochim Biophys Sin (Shanghai), 2010. 42(6): p. 363-9. 136. Manoharan, M., et al., Epigenetic targets in the diagnosis and treatment of prostate cancer. Int Braz J Urol, 2007. 33(1): p. 11-8. 137. Chandran, U.R., et al., Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer, 2007. 7: p. 64. 138. Chodak, G.W., et al., Nuclear localization of androgen receptor in heterogeneous samples of normal, hyperplastic and neoplastic human prostate. J Urol, 1992. 147(3 Pt 2): p. 798-803. 139. Diallo, J.S., et al., Co-assessment of cytoplasmic and nuclear androgen receptor location in prostate specimens: potential implications for prostate cancer development and prognosis. BJU Int, 2008. 101(10): p. 1302-9. 140. Mohler, J.L., et al., Racial differences in prostate androgen levels in men with clinically localized prostate cancer. J Urol, 2004. 171(6 Pt 1): p. 2277-80. 141. Segawa, N., et al., Expression and somatic mutation on androgen receptor gene in prostate cancer. Int J Urol, 2002. 9(10): p. 545-53. 142. Van der Kwast, T.H., et al., Androgen receptor modulation in benign human prostatic tissue and prostatic adenocarcinoma during neoadjuvant endocrine combination therapy. Prostate, 1996. 28(4): p. 227-31. 143. Zencir, S., et al., Detection of cytochrome P450-2A6, -3A5 and -4B1 with real-time polymerase chain reaction in prostate tissue. Z Naturforsch C, 2008. 63(9-10): p. 780-4. 144. Horvath, L.G., et al., Frequent loss of estrogen receptor-beta expression in prostate cancer. Cancer Res, 2001. 61(14): p. 5331-5. 145. Tsugaya, M., et al., Aromatase mRNA levels in benign prostatic hyperplasia and prostate cancer. Int J Urol, 1996. 3(4): p. 292-6. 146. Muskhelishvili, L., et al., In situ hybridization and immunohistochemical analysis of cytochrome P450 1B1 expression in human normal tissues. J Histochem Cytochem, 2001. 49(2): p. 229-36. Figure 1. A plausible gene expression network for genes and receptors involved in Estrogen (E) and Testosterone (T) metabolism in the prostate [3, 11, 19-20]

Upload: jamieritchey

Post on 08-Jul-2015

328 views

Category:

Health & Medicine


0 download

TRANSCRIPT

Page 1: Poster Slide Final

A Review of the Expression of Genes Involved in Sex Steroid Hormone Metabolism and Activity in Prostate Tissue: A Need for Epigenetic Information

Jamie Ritchey1, MPH; Wilfried Karmaus1, MD, Dr.med, MPH; Tara Sabo-Attwood1,2, PhD; Susan E. Steck1,3, PhD, MPH, RD; Hongmei Zhang1, PhD; 1Department of Epidemiology and Biostatistics, 2Department of Environmental Health Sciences, 3Cancer Prevention and Control Program, University of SouthCarolina, 800 Sumter Street, Columbia, SC 29208.

INTRODUCTION

• A pathway-oriented approach guided the literature searches, conducted in Pub Med, limited to English language and human prostate only

OBJECTIVE• To review the literature for gene expression of enzymes and receptors involved in sex steroid hormone metabolism and activity in normal, benign, and cancerous prostate tissues

• To determine if there is an association with gene expression and age, body mass index, estrogen and testosterone levels, and cancer treatments

METHODS

RESULTS

REFERENCES

RATIONALE

• Age, race, and family history are the only confirmed risk factors for prostate cancer [1]

• Observational, clinical, and laboratory evidence indicate that sex steroid hormones are important to the development and progression of prostate cancer [2-14]

• Epidemiology focusing on sex steroid hormone risk and prostate cancer is inconclusive [2-14]

• Most epidemiologic studies have focused on gene single nucleotide polymorphisms (SNPs) of hormone metabolic enzymes and serum hormone levels [2-3]

• Gene expression may provide etiologic clues regarding how sex steroid metabolism is associated with prostate cancer not detected at the DNA sequence or serum level [130]

• The expression of genes involved in sex steroid metabolism in prostate tissues may differ by different levels of age, body mass index, race, estrogen and testosterone levels, and/or cancer treatments administered

STRENGTHS

LIMITATIONS

• To guide our searches, we constructed a figure of a

plausible gene network for E and T expression [3,11, 19-20].

• Past studies have only included portions of pathways, or the T or E pathways alone, and often do not include preliminary pathways (the dashed line from androstenedione to DHT indicates that this reaction is preliminary) [3,11,19-20].

• Used a pathway approach to guide searches

• Omitted cell studies since results may not be consistent with tissue studies for methodological reasons

• Reviewed references cited in publications found during searches to improve completeness

• Information for many genes was scant

• Only publications in English language were considered

• Only includes citations from PubMed

SUMMARY• GSTP1 was consistently down regulated or not expressed in prostate cancer, which coincides with previous research indicating that GSTP1 is methylated in prostate cancer tissue [16,95,99-109,111-118,130,136]

• For all other genes, studies were either scant or inconsistent, and conflicting expression results may be due to limitations, including: tissue collection, laboratory methods, or gene functionality

• Few studies were found examining the association of gene expression with age, race, body mass index, estrogen, testosterone, and cancer treatments

• Future studies should focus not only on gene expression, but also epigenetic mechanisms such as methylation to assess prostate cancer risk

Table 1. Literature search summary

Gene &

Chromosomal location

Total articles

retrieved

Laboratory

methods†

Androgen Metabolism

AR

Ch Xq12

26 IHC, Microarrays, IS, ISV, real-time PCR

HSD17B2

Ch 16q24.1-q24.2

4 IHC, ISH, Microarray, real-time PCR

HSD17B3

Ch 9q22

4 IHC, microarray, real-time PCR

SRD5A1

Ch 5p15.31

6 IHC, ISH, Microarray

SRD5A2

Ch 2p23.1

10 IHC, ISH, Microarray, real-time PCR

CYP3A4

Ch 7q21.1-22

5 IHC, real-time PCR

CYP3A5

Ch 7q21.1-22

6 IHC, real-time PCR, meta-analysis

CYP3A7

Ch 7q21.1-22

3 IHC, real-time PCR

CYP3A43

Ch 7q21.1

1 ICH, real-time PCR

AKR1C3

Ch 10p15-14

6 Array, real-time PCR, Northern blot, review

AKR1C2

Ch 10p15-14

5 Array, real-time PCR, review

HSD3B1

Ch 1p13-11

2 Array, real-time PCR

HSD3B2

Ch 1p13.1

3 Array, review

UGT2B15

Ch 4q13

3 Array, microarray

UGT2B17

Ch 4q13

3 IHC, ISH, real-time PCR

Estrogen Metabolism

ESR1

Ch 6q24-27

9 IHC, ISH, real-time PCR

ESR2

Ch 14q31-22

10 IHC, ISH, microarray, real-time PCR

CYP19A1

Ch 15q21

9 Avidin-biotin, IHC, ISH, microarray, real-time PCR

HSD17B1

Ch 17q11-21

2 IHC, real-time PCR

HSD17B4

Ch 5q2

5 IHC, ISH, microarray, real-time PCR, meta-analysis

HSD17B7

Ch 1q23

1 IHC

SULT1A1

Ch 16p21.1

3 IHC, Western blot

SULT1A3

Ch 16p11.2

2 IHC, Western blot

SULT2B1a

Ch 19 q13.3

3 Northern blot, immunoblot, IHC

SULT2Bb

Ch 19 q13.3

3 Northern blot, immunoblot, IHC

HSD17B12

Ch 11p11

1 IHC, ISH

CYP1A1

Ch 15q24.1

4 IHC, real-time PCR

CYP1A2

Ch 15q24.1

4 IHC, real-time PCR

CYP1B1

Ch 2p22.2

6 IHC, ISH, real-time PCR

COMT

Ch 22q11.21

2 IHC, real-time PCR

GSTT1

Ch 22q11.23

1 Microarray, real-time PCR

GSTM1

Ch 1p13.1

3 IHC, microarray, real-time PCR

GSTP1

Ch 11q13.2

10 IHC, IS, microarray, real-time PCR

†Laboratory method abbreviations: Immunohistochemistry (IHC), Real-time polymerase chain reaction (real-time PCR), Immunostaining (IS), In situ

hybridization (ISH), Immunostaining with optimized IHC criteria and video image analysis (ISV)

• A total of 85 studies were identified

• Most studies focused on the expression of one or a few genes, rather than the complete pathway, ignoring compensatory or alternate response

• Studies comparing normal, benign, and cancerous prostate tissue showed limited consistency, with one exception, the down-regulation of GSTP1 in cancerous tissue

1. American Cancer Society. Cancer Facts & Figures 2008. Atlanta: American Cancer Society, 2008.2. Chu, L.W., J.K. Reichardt, and A.W. Hsing, Androgens and the molecular epidemiology of prostate cancer. Curr Opin Endocrinol Diabetes Obes, 2008. 15(3): p. 261-70.3. Chokkalingam, A.P., et al., Molecular epidemiology of prostate cancer: hormone-related genetic loci. Front Biosci, 2007. 12: p. 3436-60.4. Pollard, M., P.H. Luckert, and M.A. Schmidt, Induction of prostate adenocarcinomas in Lobund Wistar rats by testosterone. Prostate, 1982. 3(6): p. 563-8.5. Cavalieri, E.L., et al., Catechol estrogen metabolites and conjugates in different regions of the prostate of Noble rats treated with 4-hydroxyestradiol: implications for estrogen-induced initiation of prostate cancer. Carcinogenesis, 2002. 23(2): p. 329-33.6. Morgentaler, A., Guilt by association: a historial perspective on Huggins, testosterone therapy, and prostate cancer. J Sex Med, 2008. 5: p. 1834-1840.7. Sharifi, N., J.L. Gulley, and W.L. Dahut, Androgen deprivation therapy for prostate cancer. Jama, 2005. 294(2): p. 238-44.8. Hsing, A.W., J.K. Reichardt, and F.Z. Stanczyk, Hormones and prostate cancer: current perspectives and future directions. Prostate., 2002. 52(3): p. 213-35.9. Huggins, C. and R. Hodges, Studies on prostate cancer: 1. The effects of castration, of estrogen, and androgen injection on serum phosphatases in metastatic carcinoma of the prostate Cancer Research, 1941. 1: p. 203.10. Cunningham, J.M., et al., Evaluation of genetic variations in the androgen and estrogen metabolic pathways as risk factors for sporadic and familial prostate cancer. Cancer Epidemiol Biomarkers Prev, 2007. 16(5): p. 969-78.11. Hsing, A.W., L.W. Chu, and F.Z. Stanczyk, Androgen and prostate cancer: is the hypothesis dead? Cancer Epidemiol Biomarkers Prev, 2008. 17(10): p. 2525-30.12. Weiss, J.M., et al., Endogenous sex hormones and the risk of prostate cancer: a prospective study. Int J Cancer, 2008. 122(10): p. 2345-50.13. Rohrmann, S., et al., Serum estrogen, but not testosterone, levels differ between black and white men in a nationally representative sample of Americans. J Clin Endocrinol Metab, 2007. 92(7): p. 2519-25.14. Roddam, A.W., et al., Endogenous sex hormones and prostate cancer: a collaborative analysis of 18 prospective studies. J Natl Cancer Inst, 2008. 100(3): p. 170-83.15. Li, J., et al., Steroid 5-{alpha}-Reductase Type 2 (SRD5a2) Gene Polymorphisms and Risk of Prostate Cancer: A HuGE Review. Am J Epidemiol, 2009.16. Mo, Z., et al., An updating meta-analysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: a HuGE review. Prostate, 2009. 69(6): p. 662-88.17. Keshava, C., E.C. McCanlies, and A. Weston, CYP3A4 polymorphisms--potential risk factors for breast and prostate cancer: a HuGE review. Am J Epidemiol, 2004. 160(9): p. 825-41.18. Takase, Y., et al., Expression of enzymes involved in estrogen metabolism in human prostate. J Histochem Cytochem, 2006. 54(8): p. 911-21.19. Guillemette, C., A. Belanger, and J. Lepine, Metabolic inactivation of estrogens in breast tissue by UDP-glucuronosyltransferase enzymes: an overview. Breast Cancer Res, 2004. 6(6): p. 246-54.20. Maglott, D., et al., Entrez Gene: gene-centered information at NCBI. Nucleic Acids Res, 2007. 35(Database issue): p. D26-31.21. HGNC Database, HUGO Gene Nomenclature Committee (HGNC), EMBL Outstation - Hinxton, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK. (http://www.genenames.org). June 2010.22. Lu, N.Z., et al., International Union of Pharmacology. LXV. The pharmacology and classification of the nuclear receptor superfamily: glucocorticoid, mineralocorticoid, progesterone, and androgen receptors. Pharmacol Rev, 2006. 58(4): p. 782-97.23. van der Kwast, T.H. and B. Tetu, Androgen receptors in untreated and treated prostatic intraepithelial neoplasia. Eur Urol, 1996. 30(2): p. 265-8.24. Sweat, S.D., et al., Androgen receptor expression in prostate cancer lymph node metastases is predictive of outcome after surgery. J Urol, 1999. 161(4): p. 1233-7.25. Sweat, S.D., et al., Androgen receptor expression in prostatic intraepithelial neoplasia and cancer. J Urol, 1999. 161(4): p. 1229-32.26. Olapade-Olaopa, E.O., et al., Androgen receptor protein expression in prostatic tissues in Black and Caucasian men. Prostate, 2004. 59(4): p. 460-8.27. Mohler, J.L., et al., The androgen axis in recurrent prostate cancer. Clin Cancer Res, 2004. 10(2): p. 440-8.28. Kim, D., et al., Immunohistochemical quantitation of androgen receptor expression using color video image analysis. Cytometry, 1999. 35(1): p. 2-10.29. Elo, J.P., et al., Characterization of 17beta-hydroxysteroid dehydrogenase isoenzyme expression in benign and malignant human prostate. Int J Cancer, 1996. 66(1): p. 37-41.30. Brolin, J., L. Skoog, and P. Ekman, Immunohistochemistry and biochemistry in detection of androgen, progesterone, and estrogen receptors in benign and malignant human prostatic tissue. Prostate, 1992. 20(4): p. 281-95.31. Gaston, K.E., et al., Racial differences in androgen receptor protein expression in men with clinically localized prostate cancer. J Urol, 2003. 170(3): p. 990-3.32. Mohler, J.L., et al., Androgen and glucocorticoid receptors in the stroma and epithelium of prostatic hyperplasia and carcinoma. Clin Cancer Res, 1996. 2(5): p. 889-95.33. Olapade-Olaopa, E.O., et al., Malignant transformation of human prostatic epithelium is associated with the loss of androgen receptor immunoreactivity in the surrounding stroma. Clin Cancer Res, 1999. 5(3): p. 569-76.34. Rosner, I.L., et al., Higher tumor to benign ratio of the androgen receptor mRNA expression associates with prostate cancer progression after radical prostatectomy. Urology, 2007. 70(6): p. 1225-9.35. Latil, A., et al., Evaluation of androgen, estrogen (ER alpha and ER beta), and progesterone receptor expression in human prostate cancer by real-time quantitative reverse transcription-polymerase chain reaction assays. Cancer Res, 2001. 61(5): p. 1919-26.36. Leav, I., et al., Comparative studies of the estrogen receptors beta and alpha and the androgen receptor in normal human prostate glands, dysplasia, and in primary and metastatic carcinoma. Am J Pathol, 2001. 159(1): p. 79-92.37. Qiu, Y.Q., I. Leuschner, and P.M. Braun, Androgen receptor expression in clinically localized prostate cancer: immunohistochemistry study and literature review. Asian J Androl, 2008. 10(6): p. 855-63.38. Stanbrough, M., et al., Increased expression of genes converting adrenal androgens to testosterone in androgen-independent prostate cancer. Cancer Res, 2006. 66(5): p. 2815-25.39. Mohler, J.L., A role for the androgen-receptor in clinically localized and advanced prostate cancer. Best Pract Res Clin Endocrinol Metab, 2008. 22(2): p. 357-72.40. Pertschuk, L.P., et al., Immunostaining for prostate cancer androgen receptor in paraffin identifies a subset of men with a poor prognosis. Lab Invest, 1995. 73(2): p. 302-5.41. Koh, E., et al., Differential expression of 17beta-hydroxysteroid dehydrogenase isozyme genes in prostate cancer and noncancer tissues. Prostate, 2002. 53(2): p. 154-9.42. Febbo, P.G., et al., Neoadjuvant docetaxel before radical prostatectomy in patients with high-risk localized prostate cancer. Clin Cancer Res, 2005. 11(14): p. 5233-40.43. Montgomery, R.B., et al., Maintenance of intratumoral androgens in metastatic prostate cancer: a mechanism for castration-resistant tumor growth. Cancer Res, 2008. 68(11): p. 4447-54.44. Lunacek, A., et al., Fetal distribution of 5alpha-reductase 1 and 5alpha-reductase 2, and their input on human prostate development. J Urol, 2007. 178(2): p. 716-21.45. Xu, Y., et al., Pharmacologic basis for the enhanced efficacy of dutasteride against prostatic cancers. Clin Cancer Res, 2006. 12(13): p. 4072-9.46. Bonkhoff, H., et al., Differential expression of 5 alpha-reductase isoenzymes in the human prostate and prostatic carcinomas. Prostate, 1996. 29(4): p. 261-7.47. Luo, J., et al., Decreased gene expression of steroid 5 alpha-reductase 2 in human prostate cancer: implications for finasteride therapy of prostate carcinoma. Prostate, 2003. 57(2): p. 134-9.48. Ji, Q., et al., Selective reduction of AKR1C2 in prostate cancer and its role in DHT metabolism. Prostate, 2003. 54(4): p. 275-89.49. Iehle, C., et al., Differences in steroid 5alpha-reductase iso-enzymes expression between normal and pathological human prostate tissue. J Steroid Biochem Mol Biol, 1999. 68(5-6): p. 189-95.50. Bjelfman, C., et al., Differential gene expression of steroid 5 alpha-reductase 2 in core needle biopsies from malignant and benign prostatic tissue. J Clin Endocrinol Metab, 1997. 82(7): p. 2210-4.51. Habib, F.K., et al., The loss of 5alpha-reductase type I and type II mRNA expression in metastatic prostate cancer to bone and lymph node metastasis. Clin Cancer Res, 2003. 9(5): p. 1815-9.52. Daly, A.K., Significance of the minor cytochrome P450 3A isoforms. Clin Pharmacokinet, 2006. 45(1): p. 13-31.53. Koch, I., et al., Interindividual variability and tissue-specificity in the expression of cytochrome P450 3A mRNA. Drug Metab Dispos, 2002. 30(10): p. 1108-14.54. Leskela, S., et al., Cytochrome P450 3A5 is highly expressed in normal prostate cells but absent in prostate cancer. Endocr Relat Cancer, 2007. 14(3): p. 645-54.55. Yokose, T., et al., Immunohistochemical study of cytochrome P450 2C and 3A in human non-neoplastic and neoplastic tissues. Virchows Arch, 1999. 434(5): p. 401-11.56. Di Paolo, O.A., et al., Expression of cytochromes P450 and glutathione S-transferases in human prostate, and the potential for activation of heterocyclic amine carcinogens via acetyl-coA-, PAPS- and ATP-dependent pathways. Int J Cancer, 2005. 117(1): p. 8-13.57. Finnstrom, N., et al., Detection of cytochrome P450 mRNA transcripts in prostate samples by RT-PCR. Eur J Clin Invest, 2001. 31(10): p. 880-6.58. Gorlov, I.P., et al., Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data. BMC Med Genomics, 2009. 2: p. 48.59. Moilanen, A.M., et al., Characterization of androgen-regulated expression of CYP3A5 in human prostate. Carcinogenesis, 2007. 28(5): p. 916-21.60. Matsuura, K., et al., Identification of a principal mRNA species for human 3alpha-hydroxysteroid dehydrogenase isoform (AKR1C3) that exhibits high prostaglandin D2 11-ketoreductase activity. J Biochem, 1998. 124(5): p. 940-6.61. Penning, T.M., et al., Human 3alpha-hydroxysteroid dehydrogenase isoforms (AKR1C1-AKR1C4) of the aldo-keto reductase superfamily: functional plasticity and tissue distribution reveals roles in the inactivation and formation of male and female sex hormones. Biochem J, 2000. 351(Pt 1): p. 67-77.62. Penning, T.M. and M.C. Byrns, Steroid hormone transforming aldo-keto reductases and cancer. Ann N Y Acad Sci, 2009. 1155: p. 33-42.63. Penning, T.M., et al., Structure-function aspects and inhibitor design of type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3). Mol Cell Endocrinol, 2001. 171(1-2): p. 137-49.

65. Neslund-Dudas, C., et al., SRD5A2 and HSD3B2 polymorphisms are associated with prostate cancer risk and aggressiveness. Prostate, 2007. 67(15): p. 1654-63.66. Febbo, P.G., et al., Androgen mediated regulation and functional implications of fkbp51 expression in prostate cancer. J Urol, 2005. 173(5): p. 1772-7.67. Barbier, O., et al., Cellular localization of uridine diphosphoglucuronosyltransferase 2B enzymes in the human prostate by in situ hybridization and immunohistochemistry. J Clin Endocrinol Metab, 2000. 85(12): p. 4819-26.68. Park, J., et al., Deletion polymorphism of UDP-glucuronosyltransferase 2B17 and risk of prostate cancer in African American and Caucasian men. Cancer Epidemiol Biomarkers Prev, 2006. 15(8): p. 1473-8.69. Prins, G.S. and K.S. Korach, The role of estrogens and estrogen receptors in normal prostate growth and disease. Steroids, 2008. 73(3): p. 233-44.70. O'Dowd, B.F., et al., Discovery of three novel G-protein-coupled receptor genes. Genomics, 1998. 47(2): p. 310-3.71. Fromont, G., et al., Differential expression of genes related to androgen and estrogen metabolism in hereditary versus sporadic prostate cancer. Cancer Epidemiol Biomarkers Prev, 2008. 17(6): p. 1505-9.72. Ito, T., et al., Expression of estrogen receptor (ER-alpha and ER-beta) mRNA in human prostate cancer. Eur Urol, 2001. 40(5): p. 557-63.73. Singh, P.B., et al., Quantified gene expression levels for phase I/II metabolizing enzyme and estrogen receptor levels in benign prostate from cohorts designated as high-risk (UK) versus low-risk (India) for adenocarcinoma at thisorgan site: a preliminary study. Asian J Androl, 2009.74. Torlakovic, E., et al., Differential expression of steroid receptors in prostate tissues before and after radiation therapy for prostatic adenocarcinoma. Int J Cancer, 2005. 117(3): p. 381-6.75. Yang, G.S., et al., Expression of oestrogen receptor-alpha and oestrogen receptor-beta in prostate cancer. Chin Med J (Engl), 2007. 120(18): p. 1611-5.76. Fixemer, T., K. Remberger, and H. Bonkhoff, Differential expression of the estrogen receptor beta (ERbeta) in human prostate tissue, premalignant changes, and in primary, metastatic, and recurrent prostatic adenocarcinoma.Prostate, 2003. 54(2): p. 79-87.77. Haqq, C., et al., Ethnic and racial differences in prostate stromal estrogen receptor alpha. Prostate, 2005. 65(2): p. 101-9.78. Ellem, S.J., et al., Local aromatase expression in human prostate is altered in malignancy. J Clin Endocrinol Metab, 2004. 89(5): p. 2434-41.79. Matzkin, H. and M.S. Soloway, Immunohistochemical evidence of the existence and localization of aromatase in human prostatic tissues. Prostate, 1992. 21(4): p. 309-14.80. Hiramatsu, M., et al., Aromatase in hyperplasia and carcinoma of the human prostate. Prostate, 1997. 31(2): p. 118-24.81. Gross, M., et al., Expression of androgen and estrogen related proteins in normal weight and obese prostate cancer patients. Prostate, 2009. 69(5): p. 520-7.82. Vihko, P., et al., 17 beta-hydroxysteroid dehydrogenases--their role in pathophysiology. Mol Cell Endocrinol, 2004. 215(1-2): p. 83-8.83. Rasiah, K.K., et al., HSD17B4 overexpression, an independent biomarker of poor patient outcome in prostate cancer. Mol Cell Endocrinol, 2009. 301(1-2): p. 89-96.84. Kristiansen, G., et al., Expression profiling of microdissected matched prostate cancer samples reveals CD166/MEMD and CD24 as new prognostic markers for patient survival. J Pathol, 2005. 205(3): p. 359-76.85. Williams, J.A., et al., Metabolic activation of carcinogens and expression of various cytochromes P450 in human prostate tissue. Carcinogenesis, 2000. 21(9): p. 1683-9.86. Ragavan, N., et al., CYP1B1 expression in prostate is higher in the peripheral than in the transition zone. Cancer Letters, 2004. 215(1): p. 69-78.87. John, K., et al., Quantification of phase I/II metabolizing enzyme gene expression and polycyclic aromatic hydrocarbon-DNA adduct levels in human prostate. Prostate, 2009. 69(5): p. 505-19.88. Martin, F.L., et al., Constitutive expression of bioactivating enzymes in normal human prostate suggests a capability to activate pro-carcinogens to DNA-damaging metabolites. Prostate, 2010. 70(14): p. 1586-99.89. Singh, P.B., et al., Quantified gene expression levels for phase I/II metabolizing enzyme and estrogen receptor levels in benign prostate from cohorts designated as high-risk (UK) versus low-risk (India) for adenocarcinoma atthis organ site: a preliminary study. Asian J Androl, 2010. 12(2): p. 203-14.90. Al-Buheissi, S.Z., et al., N-Acetyltransferase and sulfotransferase activity in human prostate: potential for carcinogen activation. Pharmacogenet Genomics, 2006. 16(6): p. 391-9.91. Geese, W.J. and R.B. Raftogianis, Biochemical characterization and tissue distribution of human SULT2B1. Biochem Biophys Res Commun, 2001. 288(1): p. 280-9.92. He, D., et al., Different subcellular localization of sulphotransferase 2B1b in human placenta and prostate. Biochem J, 2004. 379(Pt 3): p. 533-40.93. Meloche, C.A. and C.N. Falany, Expression and characterization of the human 3 beta-hydroxysteroid sulfotransferases (SULT2B1a and SULT2B1b). J Steroid Biochem Mol Biol, 2001. 77(4-5): p. 261-9.94. Elek, J., K.H. Park, and R. Narayanan, Microarray-based expression profiling in prostate tumors. In Vivo, 2000. 14(1): p. 173-82.95. Bostwick, D.G., I. Meiers, and J.H. Shanks, Glutathione S-transferase: differential expression of alpha, mu, and pi isoenzymes in benign prostate, prostatic intraepithelial neoplasia, and prostatic adenocarcinoma. Hum Pathol, 2007. 38(9): p. 1394-401.96. Chetcuti, A., et al., Identification of differentially expressed genes in organ-confined prostate cancer by gene expression array. Prostate, 2001. 47(2): p. 132-40.97. Stephenson, A.J., et al., Integration of gene expression profiling and clinical variables to predict prostate carcinoma recurrence after radical prostatectomy. Cancer, 2005. 104(2): p. 290-8.98. Liu, A.J., et al., Quantitative analysis of a panel of gene expression in prostate cancer--with emphasis on NPY expression analysis. J Zhejiang Univ Sci B, 2007. 8(12): p. 853-9.99. Cookson, M.S., et al., Glutathione S-transferase PI (GST-pi) class expression by immunohistochemistry in benign and malignant prostate tissue. J Urol, 1997. 157(2): p. 673-6.100. Moskaluk, C.A., et al., Immunohistochemical expression of pi-class glutathione S-transferase is down-regulated in adenocarcinoma of the prostate. Cancer, 1997. 79(8): p. 1595-9.101. Parsons, J.K., et al., GSTA1 expression in normal, preneoplastic, and neoplastic human prostate tissue. Prostate, 2001. 49(1): p. 30-7.102. Montironi, R., et al., Expression of pi-class glutathione S-transferase: two populations of high grade prostatic intraepithelial neoplasia with different relations to carcinoma. Mol Pathol, 2000. 53(3): p. 122-8.103. Montironi, R., et al., Immunohistochemical expression of pi class glutathione S-transferase in the basal cell layer of benign prostate tissue following chronic treatment with finasteride. J Clin Pathol, 1999. 52(5): p. 350-4.104. Li, M., et al., Glutathione S-transferase pi is upregulated in the stromal compartment of hormone independent prostate cancer. Prostate, 2003. 56(2): p. 98-105.105. Nelson, W.G., A.M. De Marzo, and S. Yegnasubramanian, Epigenetic alterations in human prostate cancers. Endocrinology, 2009. 150(9): p. 3991-4002.106. Gilbert, S.F., Ageing and cancer as diseases of epigenesis. J Biosci, 2009. 34(4): p. 601-4.107. Petronis, A., Epigenetics and twins: three variations on the theme. Trends Genet, 2006. 22(7): p. 347-50.108. Fraga, M.F. and M. Esteller, Epigenetics and aging: the targets and the marks. Trends Genet, 2007. 23(8): p. 413-8.109. Liu, R., et al., Functional alterations in the glutathione S-transferase family associated with enhanced occurrence of esophageal carcinoma in China. J Toxicol Environ Health A, 2010. 73(7): p. 471-82.110. Stephen, J.K., et al., DNA hypermethylation profiles in squamous cell carcinoma of the vulva. Int J Gynecol Pathol, 2009. 28(1): p. 63-75.111. Yuan, Y., et al., Reduction of GSTP1 expression by DNA methylation correlates with clinicopathological features in pituitary adenomas. Mod Pathol, 2008. 21(7): p. 856-65.112. Kim, J., et al., Silencing and CpG island methylation of GSTP1 is rare in ordinary gastric carcinomas but common in Epstein-Barr virus-associated gastric carcinomas. Anticancer Res, 2005. 25(6B): p. 4013-9.113. Brabender, J., et al., Glutathione S-transferase-pi expression is downregulated in patients with Barrett's esophagus and esophageal adenocarcinoma. J Gastrointest Surg, 2002. 6(3): p. 359-67.114. Laborde, E., Glutathione transferases as mediators of signaling pathways involved in cell proliferation and cell death. Cell Death Differ, 2010. 17(9): p. 1373-80.115. Tew, K.D., TLK-286: a novel glutathione S-transferase-activated prodrug. Expert Opin Investig Drugs, 2005. 14(8): p. 1047-54.116. Goldstein, A.S., et al., Identification of a cell of origin for human prostate cancer. Science, 2010. 329(5991): p. 568-71.117. Bianco-Miotto, T., et al., Global Levels of Specific Histone Modifications and an Epigenetic Gene Signature Predict Prostate Cancer Progression and Development. Cancer Epidemiol Biomarkers Prev, 2010.118. Cooper, C.S., C. Campbell, and S. Jhavar, Mechanisms of Disease: biomarkers and molecular targets from microarray gene expression studies in prostate cancer. Nat Clin Pract Urol, 2007. 4(12): p. 677-87.119. Qian, D.Z., et al., Prostate cancer-associated gene expression alterations determined from needle biopsies. Clin Cancer Res, 2009. 15(9): p. 3135-42.120. Bostwick, D.G., et al., Independent origin of multiple foci of prostatic intraepithelial neoplasia: comparison with matched foci of prostate carcinoma. Cancer, 1998. 83(9): p. 1995-2002.121. Lin, D.W., et al., Influence of surgical manipulation on prostate gene expression: implications for molecular correlates of treatment effects and disease prognosis. J Clin Oncol, 2006. 24(23): p. 3763-70.122. True, L. and Z. Feng, Immunohistochemical validation of expression microarray results. J Mol Diagn, 2005. 7(2): p. 149-51.123. Singh, S.S., et al., Feasibility of constructing tissue microarrays from diagnostic prostate biopsies. Prostate, 2007. 67(10): p. 1011-8.124. Camp, R.L., V. Neumeister, and D.L. Rimm, A decade of tissue microarrays: progress in the discovery and validation of cancer biomarkers. J Clin Oncol, 2008. 26(34): p. 5630-7.125. Jhavar, S., et al., Construction of tissue microarrays from prostate needle biopsy specimens. Br J Cancer, 2005. 93(4): p. 478-82.126. Takase, Y., et al., Expression of sulfotransferase 1E1 in human prostate as studied by in situ hybridization and immunocytochemistry. Prostate, 2007. 67(4): p. 405-9.127. Ahmed, H., Promoter Methylation in Prostate Cancer and its Application for the Early Detection of Prostate Cancer Using Serum and Urine Samples. Biomark Cancer, 2010. 2010(2): p. 17-33.128. Baker, M., Epigenome: mapping in motion. Nature Methods, 2010. 7(3): p. 181-185.129. Kondo, Y., Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J, 2009. 50(4): p. 455-63.

130. Sharma, S., T.K. Kelly, and P.A. Jones, Epigenetics in cancer. Carcinogenesis, 2010. 31(1): p. 27-36.131. Muhonen, P. and H. Holthofer, Epigenetic and microRNA-mediated regulation in diabetes. Nephrol Dial Transplant, 2009. 24(4): p. 1088-96.132. Lim, S., et al., Epigenetic regulation of cancer growth by histone demethylases. Int J Cancer, 2010. 127(9): p. 1991-8.133. Rauhala, H.E., et al., miR-193b is an epigenetically regulated putative tumor suppressor in prostate cancer. Int J Cancer, 2010. 127(6): p. 1363-72.134. Zhang, H.L., et al., Serum miRNA-21: Elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate, 2010.135. Pang, Y., C.Y. Young, and H. Yuan, MicroRNAs and prostate cancer. Acta Biochim Biophys Sin (Shanghai), 2010. 42(6): p. 363-9.136. Manoharan, M., et al., Epigenetic targets in the diagnosis and treatment of prostate cancer. Int Braz J Urol, 2007. 33(1): p. 11-8.137. Chandran, U.R., et al., Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process. BMC Cancer, 2007. 7: p. 64.138. Chodak, G.W., et al., Nuclear localization of androgen receptor in heterogeneous samples of normal, hyperplastic and neoplastic human prostate. J Urol, 1992. 147(3 Pt 2): p. 798-803.139. Diallo, J.S., et al., Co-assessment of cytoplasmic and nuclear androgen receptor location in prostate specimens: potential implications for prostate cancer development and prognosis. BJU Int, 2008. 101(10): p. 1302-9.140. Mohler, J.L., et al., Racial differences in prostate androgen levels in men with clinically localized prostate cancer. J Urol, 2004. 171(6 Pt 1): p. 2277-80.141. Segawa, N., et al., Expression and somatic mutation on androgen receptor gene in prostate cancer. Int J Urol, 2002. 9(10): p. 545-53.142. Van der Kwast, T.H., et al., Androgen receptor modulation in benign human prostatic tissue and prostatic adenocarcinoma during neoadjuvant endocrine combination therapy. Prostate, 1996. 28(4): p. 227-31.143. Zencir, S., et al., Detection of cytochrome P450-2A6, -3A5 and -4B1 with real-time polymerase chain reaction in prostate tissue. Z Naturforsch C, 2008. 63(9-10): p. 780-4.144. Horvath, L.G., et al., Frequent loss of estrogen receptor-beta expression in prostate cancer. Cancer Res, 2001. 61(14): p. 5331-5.145. Tsugaya, M., et al., Aromatase mRNA levels in benign prostatic hyperplasia and prostate cancer. Int J Urol, 1996. 3(4): p. 292-6.146. Muskhelishvili, L., et al., In situ hybridization and immunohistochemical analysis of cytochrome P450 1B1 expression in human normal tissues. J Histochem Cytochem, 2001. 49(2): p. 229-36.

Figure 1. A plausible gene expression network for genes and receptors involved in Estrogen (E) and Testosterone (T) metabolism in the prostate [3, 11, 19-20]