We are conducting translational research to develop new agents and/or therapeutic maneuvers that appear to have antitumor activity in prostate cancer, and to develop molecular profiles of patients with prostate cancer to tailor an individualized treatment plan. To achieve this goal, we are extensively involved in the efforts to understand the biology of prostate cancer. Currently, we are attempting to correlate biological variables associated with prostate cancer and response to therapy. One early achievement by the Molecular Pharmacology Section was to report the first confirmation of the therapeutic efficacy of flutamide withdrawal, as well as the enhanced activity of simultaneous adrenal suppression. It has been hypothesized that the clinical improvement associated with flutamide is a result of the presence of a mutation within the ligand-binding domain of the androgen receptor. We are interested in analyzing candidate genes at the genomic level for genetic variations that may predispose individuals to increased risk of prostate cancer. Biomarker discovery for prostate cancer is an ongoing effort and our laboratory a project that focuses on the identification of single nucleotide polymorphisms (SNP) involved in prostate cancer progression. We are currently using a candidate gene approach which uses a panel of 96 SNPs (identified from prior studies and the published literature) from over thirty genes and using DNA samples from subjects with metastatic disease or without biochemical recurrence for over 5 years after treatment. Constitutional DNA from men who were biochemical recurrence free after treatment of their prostate cancer identified from the San Antonio center for Biomarkers Of risk for prostate cancer (SABOR) are being provided by our research collaborator, Dr. Robin Leach, and these samples are being compared to the constitutional DNA from men with documented metastatic prostate cancer identified from patients treated at the National Cancer Institute. Preliminary data analysis has shown that 5 SNPs show a statistically significant difference with a p value of <0.05, without adjusting for multiple testing, between men with metastatic disease and those without. Two SNPs, rs1544410 and rs2254210, found in the vitamin D receptor gene locus appeared to be protective against metastatic disease. Three SNPs found in genes for MSR1, osteoprotegerin, and ERG (rs3789015, rs2073617, rs2836582;respectively) appeared to increase the risk of metastatic disease. Future experiments will add more samples to the groups, identify more important SNPs and give focus for biochemical and molecular biological testing of identified important genes. The organic anion transporter OATP1B3, encoded by SLCO1B3, is involved in the transport of steroid hormones. Our recent study showed that prostate cancer overexpresses OATP1B3 compared to normal or benign hyperplastic tissue, and the common SLCO1B3 GG/AA haplotype is associated with impaired testosterone transport and improved survival in patients with prostate cancer. Based on this evidence, we conducted a study to determine whether patients with advanced prostate cancer carrying a polymorphism that codes for a more active testosterone transporter have less durable responses to ADT than patients not carrying this polymorphism. We examined the association between this SLCO1B3 polymorphism in Caucasian patients with advanced prostate cancer who were treated with ADT with metastatic disease (D2) or biochemical failure with no metastatic disease (D0). We found that a polymorphism in a transporter that increases testosterone import is associated with a shorter time to androgen independence in patients with prostate cancer who are treated with ADT. We recently completed a study on 321 primary tissue samples from 21 normal and cancerous patients that examined the expression of 3 family members implicated in cancer: OATP1B3, OATP1B1 and OATP2B1. The results showed that OATP2B1 is more ubiquitously expressed in all tissues than OATP1B1 and OATP1B3. OATP1B3 was also expressed in fewer normal tissue types;however, it was expressed in 50% of cancerous samples and there was a trend of increasing OATP1B3 expression with a higher Gleason score. This supports previous data suggesting a role for OATP1B3 in advancing prostate cancer. Further experiments from quantitative PCR and Western blot suggest that hypoxia elements in the OATP1B3 promoter are activated under tumor conditions and explain the increased expression in tumor cells. In addition, transport studies in Xenopus oocytes showed that optimal transport occurs at low androgen levels such as those seen in patients after androgen deprivation therapy. In summary, upon examining the expression of OATP1B3, OATP1B1 and OATP2B1 in primary tissue samples, there appear to be several cancers for which these may be progression or cancer biomarkers, especially in prostate cancer progression, and warrant further study. The recently completed Prostate Cancer Prevention Trial (PCPT) investigated the prevention of prostate cancer using the steroid 5 alpha-reductase inhibitor finasteride over a 7-year treatment period. Through a longstanding collaboration, we have access to the tissue samples of 18,800 men enrolled in this study. The overall goals of this project are: a) to better understand associations between important androgen regulatory gene polymorphisms and prostate cancer risk;and b) to evaluate the effects of these polymorphisms and serum hormone concentrations on the use of finasteride as a chemopreventive agent for prostate cancer. We are focusing on hormone-related factors that are associated with cancer risk, which may help explain the findings of the PCPT (i.e., decreased overall occurrence of adenocarcinoma, but increased prevalence of high-grade disease in the finasteride treatment arm). We hypothesize that men with polymorphisms within genes that positively impact androgen levels will have a higher risk of developing prostate cancer and high-grade disease than those with the wild-type alleles. In addition, long-term exposure to finasteride may select for somatic alterations and increase serum levels of testosterone and potentially harmful testosterone breakdown products. Evaluation of whether the polymorphic variations in the AR, SRD5A2 and HSD3B2 genes are associated with the risk of biopsy-detected prostate cancer in the PCPT is underway. We are identifying, by laser-capture microdissection and direct nucleotide sequencing, somatic alterations in AR and HSD3B2 that may have been selected for by long-term exposure to finasteride. Furthermore, we are determining whether prostate cancer somatic mutations of these genes differ with regard to their prevalence between the placebo and finasteride arms, and among PIA, HGPIN, prostate cancer and normal epithelium. These findings will help define a pharmacogenomic profile to identify men that are most likely to benefit from treatment with 5 alpha-reductase inhibitors. We investigated the association between the length of the polymorphic trinucleotide CAG microsatellite repeats in exon 1 of the AR gene and the risk of prostate cancer and found no association of AR CAG repeat length with prostate cancer risk. We have recently shown that the estrogen receptor alpha and aromatase polymorphisms affect risk, prognosis, and therapeutic outcome in men with CRPC treated with docetaxel-based therapy. We also demonstrated that a SNP in CYP2C8 is not associated with the development of bisphosphonate-related osteonecrosis of the jaw in men with CRPC.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC010453-10
Application #
8349012
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
10
Fiscal Year
2011
Total Cost
$595,171
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Tang, Li; Platek, Mary E; Yao, Song et al. (2018) Associations between polymorphisms in genes related to estrogen metabolism and function and prostate cancer risk: results from the Prostate Cancer Prevention Trial. Carcinogenesis 39:125-133
Chau, Cindy H; Figg, William D (2018) Revisiting 5?-reductase inhibitors and the risk of prostate cancer. Nat Rev Urol :
Huang, Phoebe A; Price, Douglas K; Figg, William D (2018) Molecular drivers of metastatic castrate-resistant prostate cancer: New roads to resistance. Cancer Biol Ther :1-2
Hauke Jr, Ralph J; Sissung, Tristan M; Figg, William D (2017) Discussing the predictive, prognostic, and therapeutic value of germline DNA-repair gene mutations in metastatic prostate cancer patients. Cancer Biol Ther :1-2
Tuerff, Daniel; Sissung, Tristan; Figg, William D (2017) Cellular identity crisis: antiandrogen resistance by lineage plasticity. Cancer Biol Ther :0
Strope, Jonathan D; Price, Douglas K; Figg, William D (2016) Building a hit list for the fight against metastatic castration resistant prostate cancer. Cancer Biol Ther 17:231-2
Goey, Andrew Kl; Sissung, Tristan M; Peer, Cody J et al. (2016) Pharmacogenomics and histone deacetylase inhibitors. Pharmacogenomics 17:1807-1815
Price, Douglas K; Chau, Cindy H; Till, Cathee et al. (2016) Association of androgen metabolism gene polymorphisms with prostate cancer risk and androgen concentrations: Results from the Prostate Cancer Prevention Trial. Cancer 122:2332-40
Sissung, Tristan M; Deeken, John; Leibrand, Crystal R et al. (2016) Identification of novel SNPs associated with risk and prognosis in patients with castration-resistant prostate cancer. Pharmacogenomics 17:1979-1986
McCrea, Edel; Sissung, Tristan M; Price, Douglas K et al. (2016) Androgen receptor variation affects prostate cancer progression and drug resistance. Pharmacol Res 114:152-162

Showing the most recent 10 out of 52 publications