Finasteride concentrations and prostate cancer risk: results from the Prostate Cancer Prevention Trial

Cindy H Chau, Douglas K Price, Cathee Till, Phyllis J Goodman, Xiaohong Chen, Robin J Leach, Teresa L Johnson-Pais, Ann W Hsing, Ashraful Hoque, Catherine M Tangen, Lisa Chu, Howard L Parnes, Jeannette M Schenk, Juergen K V Reichardt, Ian M Thompson, William D Figg, Cindy H Chau, Douglas K Price, Cathee Till, Phyllis J Goodman, Xiaohong Chen, Robin J Leach, Teresa L Johnson-Pais, Ann W Hsing, Ashraful Hoque, Catherine M Tangen, Lisa Chu, Howard L Parnes, Jeannette M Schenk, Juergen K V Reichardt, Ian M Thompson, William D Figg

Abstract

Objective: In the Prostate Cancer Prevention Trial (PCPT), finasteride reduced the risk of prostate cancer by 25%, even though high-grade prostate cancer was more common in the finasteride group. However, it remains to be determined whether finasteride concentrations may affect prostate cancer risk. In this study, we examined the association between serum finasteride concentrations and the risk of prostate cancer in the treatment arm of the PCPT and determined factors involved in modifying drug concentrations.

Methods: Data for this nested case-control study are from the PCPT. Cases were drawn from men with biopsy-proven prostate cancer and matched controls. Finasteride concentrations were measured using a liquid chromatography-mass spectrometry validated assay. The association of serum finasteride concentrations with prostate cancer risk was determined by logistic regression. We also examine whether polymorphisms in the enzyme target and metabolism genes of finasteride are related to drug concentrations using linear regression.

Results and conclusions: Among men with detectable finasteride concentrations, there was no association between finasteride concentrations and prostate cancer risk, low-grade or high-grade, when finasteride concentration was analyzed as a continuous variable or categorized by cutoff points. Since there was no concentration-dependent effect on prostate cancer, any exposure to finasteride intake may reduce prostate cancer risk. Of the twenty-seven SNPs assessed in the enzyme target and metabolism pathway, five SNPs in two genes, CYP3A4 (rs2242480; rs4646437; rs4986910), and CYP3A5 (rs15524; rs776746) were significantly associated with modifying finasteride concentrations. These results suggest that finasteride exposure may reduce prostate cancer risk and finasteride concentrations are affected by genetic variations in genes responsible for altering its metabolism pathway.

Trial registration: ClinicalTrials.gov NCT00288106.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Linkage disequilibrium (LD) pattern and…
Fig 1. Linkage disequilibrium (LD) pattern and haplotype architecture for (A) CYP3A4 & CYP3A5; (B) SRD5A2; and (C) SRD5A2L/SRD5A3 genes.
The haplotype block structure, as exhibited by Haploview is shown. LD was measured using data from all white subjects in the present study. The haplotype blocks were determined using the criteria described by Gabriel et al. The physical position of each SNP is presented in the upper diagram. Each diamond contains the level of LD measured by Hedrick's multiallelic D′ between pairs of single nucleotide polymorphisms. Shading shows the magnitude and significance of pairwise LD, with darker shades representing stronger LD; the diamond without a number corresponds to D′ = 1. Haplotypes for the variations and their population frequency (light gray color) are shown below each haplotype block of the corresponding genes. The SNP numbers across the top of the haplotypes correspond to those in the Haploview plot. D′ indicates the level of recombination between two blocks and is shown in the crossing area. The connection from one block to the next block is displayed through frequency corresponding to the thickness of the line.

References

    1. Bruchovsky N, Wilson JD (1968) The conversion of testosterone to 5-alpha-androstan-17-beta-ol-3-one by rat prostate in vivo and in vitro. J Biol Chem 243: 2012–2021.
    1. Imperato-McGinley J, Guerrero L, Gautier T, Peterson RE (1974) Steroid 5alpha-reductase deficiency in man: an inherited form of male pseudohermaphroditism. Science 186: 1213–1215.
    1. Andersson S, Russell DW (1990) Structural and biochemical properties of cloned and expressed human and rat steroid 5 alpha-reductases. Proc Natl Acad Sci U S A 87: 3640–3644.
    1. Tamura K, Furihata M, Tsunoda T, Ashida S, Takata R, Obara W, et al. (2007) Molecular features of hormone-refractory prostate cancer cells by genome-wide gene expression profiles. Cancer Res 67: 5117–5125.
    1. Uemura M, Tamura K, Chung S, Honma S, Okuyama A, Nakamura Y, et al. (2008) Novel 5 alpha-steroid reductase (SRD5A3, type-3) is overexpressed in hormone-refractory prostate cancer. Cancer Sci 99: 81–86.
    1. Chaudhary UB, Turner JS (2010) Finasteride. Expert Opin Drug Metab Toxicol 6: 873–881. 10.1517/17425255.2010.495944
    1. Gormley GJ, Stoner E, Bruskewitz RC, Imperato-McGinley J, Walsh PC, McConnell JD, et al. (1992) The Effect of Finasteride in Men with Benign Prostatic Hyperplasia. N Engl J Med 327: 1185–1191.
    1. Stoner E (1992) The clinical effects of a 5 alpha-reductase inhibitor, finasteride, on benign prostatic hyperplasia. The Finasteride Study Group. J Urol 147: 1298–1302.
    1. Bull HG, Garcia-Calvo M, Andersson S, Baginsky WF, Chan HK, Ellsworth DE, et al. (1996) Mechanism-Based Inhibition of Human Steroid 5α-Reductase by Finasteride: Enzyme-Catalyzed Formation of NADP—Dihydrofinasteride, a Potent Bisubstrate Analog Inhibitor. J Am Chem Soc 118: 2359–2365.
    1. Yamana K, Labrie F, Luu-The V (2010) Human type 3 5α-reductase is expressed in peripheral tissues at higher levels than types 1 and 2 and its activity is potently inhibited by finasteride and dutasteride. Horm Mol Biol Clin Invest 2: 293–299.
    1. Makridakis NM, di Salle E, Reichardt JK (2000) Biochemical and pharmacogenetic dissection of human steroid 5 alpha-reductase type II. Pharmacogenetics 10: 407–413.
    1. Makridakis N, Akalu A, Reichardt JK (2004) Identification and characterization of somatic steroid 5alpha-reductase (SRD5A2) mutations in human prostate cancer tissue. Oncogene 23: 7399–7405.
    1. Huskey SW, Dean DC, Miller RR, Rasmusson GH, Chiu SH (1995) Identification of human cytochrome P450 isozymes responsible for the in vitro oxidative metabolism of finasteride. Drug Metab Dispos 23: 1126–1135.
    1. Kuehl P, Zhang J, Lin Y, Lamba J, Assem M, Schuetz J, et al. (2001) Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 27: 383–391.
    1. Thompson IM, Goodman PJ, Tangen CM, Lucia MS, Miller GJ, Ford LG, et al. (2003) The influence of finasteride on the development of prostate cancer. N Engl J Med 349: 215–224.
    1. Feigl P, Blumenstein B, Thompson I, Crowley J, Wolf M, Kramer BS, et al. (1995) Design of the Prostate Cancer Prevention Trial (PCPT). Control Clin Trials 16: 150–163.
    1. Chen X, Gardner ER, Price DK, Figg WD (2008) Development and validation of an LC-MS assay for finasteride and its application to prostate cancer prevention trial sample analysis. J Chromatogr Sci 46: 356–361.
    1. Barrett JC, Fry B, Maller J, Daly MJ (2005) Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics 21: 263–265.
    1. Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, et al. (2002) The structure of haplotype blocks in the human genome. Science 296: 2225–2229.
    1. Gong Z, Kristal AR, Schenk JM, Tangen CM, Goodman PJ, Thompson IM (2009) Alcohol consumption, finasteride, and prostate cancer risk: results from the Prostate Cancer Prevention Trial. Cancer 115: 3661–3669. 10.1002/cncr.24423
    1. He BX, Shi L, Qiu J, Tao L, Li R, Yang L, et al. (2011) A functional polymorphism in the CYP3A4 gene is associated with increased risk of coronary heart disease in the Chinese Han population. Basic Clin Pharmacol Toxicol 108: 208–213. 10.1111/j.1742-7843.2010.00657.x
    1. Gao Y, Zhang LR, Fu Q (2008) CYP3A4*1G polymorphism is associated with lipid-lowering efficacy of atorvastatin but not of simvastatin. Eur J Clin Pharmacol 64: 877–882. 10.1007/s00228-008-0502-x
    1. Miura M, Satoh S, Kagaya H, Saito M, Numakura K, Tsuchiya N, et al. (2011) Impact of the CYP3A4*1G polymorphism and its combination with CYP3A5 genotypes on tacrolimus pharmacokinetics in renal transplant patients. Pharmacogenomics 12: 977–984. 10.2217/pgs.11.33
    1. Schirmer M, Rosenberger A, Klein K, Kulle B, Toliat MR, Nurnberg P, et al. (2007) Sex-dependent genetic markers of CYP3A4 expression and activity in human liver microsomes. Pharmacogenomics 8: 443–453.
    1. van Schaik RH, de Wildt SN, Brosens R, van Fessem M, van den Anker JN, Lindemans J (2001) The CYP3A4*3 allele: is it really rare? Clin Chem 47: 1104–1106.
    1. Kim DH, Sriharsha L, Xu W, Kamel-Reid S, Liu X, Siminovitch K, et al. (2009) Clinical relevance of a pharmacogenetic approach using multiple candidate genes to predict response and resistance to imatinib therapy in chronic myeloid leukemia. Clin Cancer Res 15: 4750–4758. 10.1158/1078-0432.CCR-09-0145
    1. Garcia-Donas J, Esteban E, Leandro-Garcia LJ, Castellano DE, del Alba AG, Climent MA, et al. (2011) Single nucleotide polymorphism associations with response and toxic effects in patients with advanced renal-cell carcinoma treated with first-line sunitinib: a multicentre, observational, prospective study. Lancet Oncol 12: 1143–1150. 10.1016/S1470-2045(11)70266-2
    1. Onizuka M, Kunii N, Toyosaki M, Machida S, Ohgiya D, Ogawa Y, et al. (2011) Cytochrome P450 genetic polymorphisms influence the serum concentration of calcineurin inhibitors in allogeneic hematopoietic SCT recipients. Bone Marrow Transplant 46: 1113–1117. 10.1038/bmt.2010.273

Source: PubMed

Подписаться