Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 1.
Published in final edited form as: Int J Cancer. 2014 Oct 13;136(9):2166–2171. doi: 10.1002/ijc.29241

Confirmation of Genetic Variants Associated with Lethal Prostate Cancer in a Cohort of Men from Hereditary Prostate Cancer Families

Danielle M Karyadi 1,*, Shanshan Zhao 2,*, Qianchuan He 2, Laura McIntosh 2, Jonathan L Wright 2,3, Elaine A Ostrander 1, Ziding Feng 4, Janet L Stanford 2,5
PMCID: PMC4331209  NIHMSID: NIHMS633289  PMID: 25273821

Abstract

Germline genetic variants have been suggested as prognostic biomarkers for identifying patients at high risk for lethal prostate cancer (PCa). Validation studies have confirmed the association of several single nucleotide polymorphisms (SNPs) with fatal PCa, but whether these variants affect PCa-specific mortality (PCSM) in patients with an inherited predisposition to PCa, based on familial history, is unknown. For this study, a cohort of 957 PCa patients from 270 hereditary prostate cancer (HPC) families of European ancestry was genotyped for a panel of 22 PCSM-associated SNPs. Death certificates were reviewed to confirm cause of death. Mixed-effect Cox proportional hazards models were used to assess survival according to genotypes, accounting for relatedness and clinicopathological factors. Within this cohort, 98 PCa deaths were confirmed over an average follow-up period of 12.7 years after diagnosis. Variant allele carriers for three SNPs had significantly altered risk for PCSM (rs635261 at RNASEL, HR, 0.35, 95% CI, 0.18–0.66; P = 0.002; rs915927 in XRCC1, HR, 1.91, 95% CI, 1.21–3.02; P = 0.009; and rs2494750 at AKT1, HR, 0.45, 95% CI, 0.23–0.90; P = 0.016). These results confirm the association of genetic variation in three genes with PCa lethality in a cohort of men with an inherited susceptibility to the disease and provide validation evidence that germline SNPs provide prognostic information for PCa patients. Development of a panel of germline biomarkers with clinical utility for distinguishing patients at detection who have an increased risk for fatal PCa is warranted.

Keywords: Hereditary prostate cancer, mortality, SNPs, XRCC1, AKT1

Introduction

Prostate cancer (PCa) ranks second in the United States (U.S.) and fifth internationally as a cause of cancer-related deaths in men, with approximately 30,000 men in the U.S. and over 300,000 men worldwide dying of PCa annually 1, 2. Recent survival data from the Surveillance, Epidemiology and End Results (SEER) Program indicate that approximately one in 11 (9%) patients diagnosed with PCa will die of their disease 3, 4.

Prostate cancer is a heterogeneous disorder that exhibits a range of clinical behaviors, from indolent, slow-growing to aggressive, rapidly growing tumors with lethal progression. Although a significant number of prostate tumors may not lead to morbidity or mortality, even if left untreated, most clinically localized tumors are treated with curative intent via surgery or radiation 5. Despite treatment, however, up to one-third of patients will recur during long-term follow-up 6, 7. Even within those experiencing PCa recurrence, there is heterogeneity in outcomes as only a subset will die of their disease. The standard criteria for risk stratifying clinically localized PCa, namely pre-biopsy PSA level, tumor histologic grade (Gleason score), and tumor stage, do not accurately distinguish patients who will go on to have aggressive as opposed to indolent PCa, especially when evaluating men within a risk stratum. Thus, research focused on discovery and confirmation of biomarkers associated with a high risk for progression to lethal PCa is urgently needed.

Toward this end, we previously identified a panel of genetic variants that were significantly associated with PCa-specific mortality (PCSM) in a population-based cohort from Seattle-King County, WA, and completed an initial validation effort confirming five genetic variants (one each in the LEPR, CRY1, RNASEL, IL4, and ARVCF genes) as predictive of lethal PCa in a second large independent population-based cohort of PCa patients from Sweden 8. Most recently, a confirmation study within the Physician’s Health Study cohort further validated the role of one of these SNPs (rs5993891 in the ARVCF gene) as being significantly associated with a lower risk of fatal PCa 9. A meta-analysis revealed a significant 48% reduction in the risk of lethal PCa in cases carrying the variant T allele (hazard ratio, HR = 0.52, 95% CI 0.3–0.9; P = 0.03). The above study cohorts were mainly comprised of PCa patients without a family history of the disease. However, a subset (5%–10%) of PCa patients has the familial form of the disease due to an inherited predisposition, and it is unknown whether genetic biomarkers for lethal PCa are associated with outcomes in these patients. To further evaluate the ability of the SNP panel to distinguish patients at the time of detection who are at high risk for disease lethality, we genotyped a cohort of PCa patients ascertained through a large study of hereditary prostate cancer (HPC) families.

Materials and Methods

Study cohorts

The Seattle-based PCa patient cohort in which these 22 SNP genotypes were first associated with lethal outcomes has been described elsewhere 8. In brief, 1,309 patients of European ancestry diagnosed with adenocarcinoma of the prostate in 1993–1996 or 2002–2005 were ascertained through the Seattle-Puget Sound population-based SEER cancer registry and provided a blood sample for genetic studies. Over an average follow-up period of 8.5 years, 60 patients were confirmed to have died of PCa based on SEER data and review of death certificates.

The validation patient cohort is comprised of men participating in the Prostate Cancer Genetic Research Study (PROGRESS), which was initiated in 1995 and has ascertained and followed over 2,200 members of 307 families with a pattern of familial PCa 10. Families with two or more PCa survivors who were willing to participate and provide a blood sample were ascertained from across North America by advertising a toll-free number via public media, health-related publications, and the internet as well as communications with urologists, other health-care professionals, and PCa support groups. To be eligible for inclusion, families were required to meet at least one of the following criteria: (1) have three or more first-degree relatives with PCa; (2) have three generations of relatives with PCa; or (3) have two first-degree relatives with PCa diagnosed before age 65. For this analysis, a cohort of 957 men of European ancestry who were diagnosed with histologically confirmed PCa and for whom germline DNA was available for genotyping was included. Medical records were abstracted to obtain clinical and pathological data such as Gleason score (biopsy, surgery), extent of disease (tumor stage), diagnostic level PSA, and primary therapy. Data were coded according to SEER guidelines 11, incorporating the information from surgery for patients whose primary treatment was radical prostatectomy. The ascertainment of information on secondary treatment and vital status was based on two follow-up surveys sent to patients and their relatives, followed by collection and review of death certificates for decedents to determine underlying cause of death (i.e., prostate cancer vs. another cause), which was used to classify PCSM events.

Written informed consent was obtained from all participants in each of the study cohorts. Both studies were reviewed and approved by the Fred Hutchinson Cancer Research Center institutional review board.

Genotyping

Blood samples were collected using ACD tubes and shipped to the Fred Hutchinson Cancer Research Center for processing. Genomic DNA was extracted from peripheral blood lymphocytes using standard techniques12 and stored at −80°C. For this study, one DNA aliquot per patient was shipped to Dr. Ostrander’s laboratory (National Human Genome Research Institute) for genotyping. The MassARRAY iPLEX system (Sequenom, Inc., San Diego, CA) was used to genotype 20 of the 22 SNPs, and the remaining two SNPs (rs2308327 and rs4583514) were genotyped with TaqMan SNP genotyping assays (Applied Biosystems, Foster City, CA).

Blind duplicate DNA samples from 48 patients were distributed evenly across all genotyping batches, and laboratory personnel were blinded to patient outcome status (alive, deceased). The average concordance for the 22 SNP genotypes among the 48 blind duplicates was 99.8%. All 22 SNPs were successfully genotyped in 96% of the cohort. Hardy-Weinberg Equilibrium (HWE) was assessed in the patients who did not die of PCa. For this analysis, we randomly sampled one PCa patient per family to avoid genotype correlations within the family. All 22 SNP genotypes were in HWE (all P-values > 0.05). In addition, the minor allele frequency (MAF) for each SNP was calculated based on the genotype in PROGRESS cases who did not die of PCa.

Statistical analysis

The mixed-effect Cox proportional hazards model was used for analysis as it accounts for the correlation of genotypes between family members 13. For the main models, the best-fitting genetic model (i.e., dominant, recessive, or trend) for each SNP as determined from the Seattle-based cohort that initially reported these SNP-PCSM associations was assumed (i.e., the underlying genetic model was fixed), and two sets of covariates were considered: 1) age at diagnosis only; and 2) age at diagnosis, together with Gleason score, stage, diagnostic PSA level, and primary treatment (categorical with missing indicator variables for clinicopathological covariates). The best-fitting of the two models in the PROGRESS cohort was selected based on which model had the lowest P-value. For the correlation structure, we used the pedigree() and kinship() function in the kinship2 library in R to construct the kinship matrix that represents the genetic relationships within families 14. The mixed-effect Cox model was fitted with the R function coxme() in the coxme library, incorporating the constructed kinship matrix 14. Follow-up time was calculated from the date of PCa diagnosis to the date of death from PCa, death from another cause or last follow-up.

A genetic variant was considered as validated if one of the two covariate adjusted models had a 1-sided p-value < 0.05 since for validation we required that the effect of the variant allele on PCSM in the PROGRESS cohort be in the same direction as that previously observed in the Seattle PCa cohort 8. Hazard ratios (HR) and associated 95% confidence intervals (CI) were calculated.

Results

Descriptive characteristics of the HPC patient cohort are shown in Table 1. The mean age at diagnosis was 64.5 years and the average length of follow-up was 12.7 years. A total of 98 (10%) deaths were attributed to PCa during the follow-up period. The majority of patients were diagnosed with clinically localized disease and 52.4% underwent radical prostatectomy as primary therapy. At diagnosis, 13.1% of patients had a Gleason score of 7 (4+3) or higher.

Table 1.

Descriptive Characteristics of Prostate Cancer Patients in the PROGRESS Cohort

Characteristic No. = 957 %
Age at diagnosis, years
 Mean 64.5
 Range 40.0–87.0
Follow-up time, years
 Mean 12.73
 Range 0.28–32.6
Prostate cancer-specific death
 No1 782 81.7
 Yes 98 10.2
 Unknown2 77 8.0
Age at death, years
 Mean 80.5
 Range 54.0–99.0
Stage at diagnosis
 Local 623 65.1
 Regional 240 25.1
 Distant 27 2.8
 Missing 67 7.0
Gleason score at diagnosis
 ≤ 6 561 58.6
 7 (3+4) 159 16.6
 7 (4+3) 45 4.7
 8–10 80 8.4
 Missing 112 11.7
Diagnostic PSA level, ng/mL
 < 4.0 77 8.0
 4–9.9 360 37.6
 10–19.9 157 16.4
 ≥ 20 130 13.6
 Missing 233 24.3
Primary therapy
 Radical prostatectomy 501 52.4
 Radiation therapy 256 26.8
 Androgen deprivation 75 7.8
 Active surveillance 21 2.2
 Other 18 1.9
 Missing 86 9.0
1

Includes men who died of other causes (n=258) or were alive at last follow-up (n=524).

2

Includes men who died but underlying cause of death is unknown.

The results highlighted three genetic variants for which there was validation evidence for an association with PCSM in the PROGRESS cohort (Table 2). The strongest result was for rs635261 downstream of the RNASEL gene, with a HR = 0.35 (95% CI, 0.18–0.66; P = 0.002) for carriers of the variant C allele. In addition, rs915927 in the XRCC1 gene (HR, 1.91, 95% CI, 1.21–3.02; P = 0.009) and rs2494750 at AKT1 (HR, 0.45, 95% CI, 0.23–0.90; P = 0.016) were associated with risks for lethal PCa. For comparison, the published results from the previously analyzed Seattle-based PCa cohort are also shown (Table 2). The underlying genetic model for all three of these PCSM-associated SNPs was identical to the earlier Seattle-based model, and the covariates in the model that best fit the PROGRESS data for the RNASEL variant were also the same as in the Seattle cohort. However, the covariates differed in the best-fitting models for the XRCC1 and AKT1 gene variants in the PROGRESS cohort.

Table 2.

Hazard Ratios (HR) for Prostate Cancer-Specific Mortality Associated with a Panel of 22 SNPs in Two Patient Cohorts

Genetic Variants Seattle Cohort PROGRESS Cohort
SNP Chr. Gene Alleles1 MAF2 HR 95% CI Model3 MAF2 HR 95% CI P-value4 Model
rs1137100 1p31 LEPR A/G 0.27 0.29 0.14–0.60 Dom: ACP 0.26 1.65 1.08–2.51 NC Dom: ACP
rs228697 1p36 PER3 C/G 0.11 0.25 0.10–0.60 Dom: ACP 0.10 1.39 0.84–2.30 NC Dom: ACP
rs635261 1q25 RNASEL G/C 0.36 0.22 0.070.65 Rec: ACP 0.39 0.35 0.180.66 0.002 Rec: ACP
rs627839 1q25 RNASEL G/T 0.47 3.98 1.64–9.65 Dom: ACP 0.48 0.83 0.56–1.21 NC Dom: ACP
rs4583514 2p21 MSH2 G/A 0.38 2.49 1.21–5.10 Dom: ACP 0.38 0.68 0.45–1.03 NC Dom: ACP
rs4608577 2p21 MSH2 T/G 0.17 2.04 1.36–3.07 Tre: A 0.17 1.09 0.77–1.55 0.33 Tre: ACP
rs523349 2p23 SRD5A2 C/G 0.29 0.49 0.28–0.86 Dom: A 0.29 1.08 0.71–1.67 NC Dom: ACP
rs12467911 2p23 SRD5A2 C/T 0.28 0.45 0.24–0.81 Dom: A 0.30 1.14 0.75–1.72 NC Dom: ACP
rs11710277 3p21 SEMA3F A/G 0.09 3.71 1.75–7.90 Dom: ACP 0.06 0.75 0.39–1.42 NC Dom: ACP
rs11205 5p23 HSD17B4 A/G 0.39 0.21 0.06–0.70 Rec: ACP 0.43 1.21 0.79–1.86 NC Rec: A
rs2070874 5q31 IL4 C/T 0.16 2.16 1.27–3.67 Dom: A 0.16 1.17 0.80–1.72 0.25 Dom: A
rs1799964 6p21 TNF/LTA T/C 0.21 0.39 0.20–0.77 Dom: A 0.22 1.19 0.82–1.72 NC Dom: A
rs4645959 8q24 C-MYC A/G 0.04 0 0.00–inf. Tre: ACP 0.04 1.14 0.56–2.32 NC Tre: ACP
rs1029153 10q11 CXCL12 T/C 0.31 0.22 0.07–0.75 Tre: A 0.29 1.14 0.87–1.51 NC Tre: A
rs2839685 10q11 CXCL12 C/T 0.15 28.2 7.21–110.2 Rec: ACP 0.14 0.58 0.11–3.10 NC Rec: A
rs2308327 10q26 MGMT A/G 0.13 0.32 0.13–0.78 Tre: A 0.11 0.85 0.55–1.31 0.26 Tre: ACP
rs10778534 12q23 CRY1 T/C 0.36 2.21 1.19–4.12 Dom: A 0.36 0.68 0.45–1.03 NC Dom: ACP
rs2494750 14q32 AKT1 C/G 0.07 0.22 0.070.70 Tre: ACP 0.05 0.45 0.230.90 0.016 Tre: A
rs1799814 15q24 CYP1A1 C/A 0.05 0.13 0.03–0.57 Tre: ACP 0.05 1.27 0.61–2.64 NC Tre: ACP
rs25487 19q13 XRCC1 G/A 0.36 0.49 0.31–0.77 Tre: A 0.37 0.85 0.65–1.12 0.17 Tre: A
rs915927 19q13 XRCC1 A/G 0.43 2.54 1.245.18 Dom: A 0.41 1.91 1.213.02 0.009 Dom: ACP
rs5993891 22q11 ARVCF C/T 0.05 0.21 0.07–0.61 Dom: ACP 0.05 1.53 0.91–2.57 NC Dom: A
1

Major/minor allele.

2

MAF: minor allele frequency, calculated from cases who did not die of prostate cancer.

3

Model: Dom= dominant, Rec= recessive, Tre= trend; A= age at diagnosis, ACP= age plus clinicopathological covariates (i.e., Gleason score, diagnostic PSA, stage, and primary treatment).

4

NC: not confirmed (i.e., the direction of the HR in the PROGRESS confirmation cohort was in the opposite direction compared to the HR in the Seattle discovery cohort).

In further analyses the use of secondary treatment was considered. A covariate was added to the Cox models to assess whether HRs for the three confirmed SNPs remained significant. Data on use of secondary therapy were available for 525 (61%) cases who did not die of PCa and 30 (31%) fatal cases, so we assigned a separate missing category for the remaining cases. The HRs for the three SNPs were not substantially different than those shown in Table 2 (i.e., rs635261, HR = 0.32; rs915927, HR = 1.82; rs2494750, HR = 0.49) and all were statistically significant (all P ≤ 0.035).

One SNP that was not considered confirmed in the PROGRESS cohort because the association was not in the same direction as in the Seattle-based PCa discovery cohort, however, was associated with survival. For rs1137100 in LEPR, the variant G allele was associated with a significant increase in the risk of lethal PCa (HR, 1.65, 95% CI, 1.08–2.51), but this risk estimate was in the opposite direction compared to results from both the Seattle and the Swedish population-based PCa cohorts 8.

Discussion

The results from this study provide validation for specific germline alterations that are associated with lethal PCa in men with an inherited predisposition for the disease based on family history. This is the first evidence that several SNPs in our initial panel of 22 PCSM-associated genetic polymorphisms 8 are associated with prognosis in patients from hereditary PCa (HPC) families. We confirmed the association of three genetic variants with the risk of fatal events, one downstream of RNASEL, one in XRCC1 and the third upstream of AKT1. There was one other variant (rs1137100 in LEPR) that achieved statistical significance in this HPC cohort, but it was not considered as validated because its effect on PCSM was in the opposite direction as compared to results from the Seattle-based cohort that first highlighted this panel of 22 SNPs associated with lethality 8.

The strongest confirmation evidence was for rs635261 downstream of the RNASEL gene. Interestingly, another SNP in this gene (rs627839) achieved borderline significance in the prior validation study of the 22 PCSM-associated variants in a Swedish cohort 8. RNASEL is involved in the interferon-regulated antiviral response and also functions in diverse cellular mechanisms, including cell proliferation, differentiation, apoptosis and tumorigenesis 15. The RNASEL R462Q mutation is a candidate for the hereditary PCa allele HPC1 16. In addition, there is evidence that RNASEL can interact with the androgen receptor to promote tumor progression 17. Recently RNASEL variants have also been associated with outcomes after radiation therapy 18, and with increased serum levels of C-reactive protein and interleukin-6 19, which together suggest a role for inflammation in response to radiation treatment 18. Still other studies suggest that a subset of variants within the gene have predictive value for aggressive disease 19, 20. The gene may not act alone, and at least one study suggests that interactions between variants in RNASEL and 8q24 are associated with aggressive prostate cancer 21.

The second strongest validation evidence was for a variant in the X-ray repair cross-complementing group 1 (XRCC1) gene. XRCC1 is a DNA repair gene whose protein product is involved in a number of DNA repair pathways including base excision repair 22, 23. XRCC1 is thought to act as a scaffold protein enabling the recruitment of the DNA repair enzymes involved in the insertion of new nucleotides and sealing of the repair break 22, 23. As such, reduced activity of XRCC1 could lead to an increase in somatic mutations, which in turn might promote disease progression. Multiple studies have evaluated the association between XRCC1 variants and overall risk of PCa with conflicting results and, in fact, two meta-analyses failed to find any association 24. However, none of these studies analyzed outcomes such as PCa mortality or focused on HPC families. Interestingly, the XRCC1 Arg280His variant has been associated with another PCa outcome, radiation induced late stage toxicity 25.

A third variant upstream of AKT1 was also validated in this study. AKT1 is a member of the AKT/PKB family of kinases. AKT acts as an intermediate signaling molecule in a number of important processes including cell survival, proliferation, tumor invasion, metastasis and angiogenesis 26, 27. Of particular interest for PCSM is the role of the AKT pathway in epithelial-mesenchymal transition (EMT). EMT is a normal and important developmental mechanism whereby epithelial cells obtain mesenchymal, fibroblast-like properties, including reduced intercellular adhesion and increased motility 28. Activation of AKT can induce EMT-like events, which in the course of tumor development could give cancer cells invasive and metastatic properties, thus promoting tumor progression and in the long term, PCSM 28.

The rs635261 SNP is approximately 3,700 bp downstream of RNASEL, the XRCC1 variant, rs915927, is a synonymous change at Pro206, and rs2494750 is approximately 830 bp upstream of the AKT1 gene. Given the critical role both of the latter proteins perform during development and the activity of XRCC1 in DNA repair, a major disruption to either protein’s function is unlikely. The role of these genetic variants is unknown at this time, but they could potentially alter gene expression in a tissue specific manner. Alternatively the variants may tag another mutation that directly affects RNASEL, XRCC1 or AKT1 function, or that of other critical, nearby genes. Additional studies are needed to resolve these possibilities and to understand the role of each gene in the progression pathway to lethal PCa.

Currently, available clinicopathological characteristics of PCa do not accurately predict an individual patient’s outcome. Distinguishing patients at elevated risk for fatal PCa from those with indolent tumors is critical not only to identify high-risk patients who would benefit from early aggressive therapy, but to avoid over-treatment of low-risk patients as well. Our results provide further support that germline biomarkers may enable differentiation between patients with indolent versus aggressive PCa at the time of detection and have clinical utility for identifying those at increased risk for lethal PCa. Additional studies aimed at the development of a germline biomarkers panel with improved prognostic ability over the current methods are warranted.

Acknowledgments

This work was supported by a grant from the National Cancer Institute (R01 CA080122), with additional support provided by the Fred Hutchinson Cancer Research Center, the Intramural Program of the National Human Genome Research Institute, and the Prostate Cancer Foundation. The authors gratefully acknowledge the contributions of participants in the Prostate Cancer Genetic Research Study.

Abbreviations

PCa

prostate cancer

HPC

hereditary prostate cancer

SNP

single nucleotide polymorphism

HR

hazard ratio

CI

confidence interval

Footnotes

Brief Description: This study evaluated a panel of 22 germline genetic variants previously associated with lethal PCa in a high-risk patient cohort from HPC families. Results validate the association of three SNPs with lethal PCa (rs635261 at RNASEL, HR, 0.35, 95% CI, 0.18–0.66; P = 0.002; rs915927 in XRCC1, HR, 1.91, 95% CI, 1.21–3.02; P = 0.009; and rs2494750 at AKT1, HR, 0.45, 95% CI, 0.23–0.90; P = 0.016). Germline SNPs may provide prognostic information for distinguishing patients at increased risk for progression to fatal PCa.

References

  • 1.Howlader N, Noone AM, Krapcho M, et al. SEER Cancer Statistics Review, 1975–2011. National Cancer Institute; Bethesda, MD: Available at: http://seer.cancer.gov/csr/1975_2011/ [Google Scholar]
  • 2.Ferlay J, Soerjomataram I, Ervik M, et al. GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet] Lyon, France: International Agency for Research on Cancer; 2013. Available at: http://globocan.iarc.fr/ [Google Scholar]
  • 3.Epstein MM, Edgren G, Rider JR, et al. Temporal trends in cause of death among Swedish and US men with prostate cancer. J Natl Cancer Inst. 2012;104:1335–42. doi: 10.1093/jnci/djs299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10–29. doi: 10.3322/caac.20138. [DOI] [PubMed] [Google Scholar]
  • 5.Cooperberg MR, Broering JM, Carroll PR. Time trends and local variation in primary treatment of localized prostate cancer. J Clin Oncol. 2010;28:1117–23. doi: 10.1200/JCO.2009.26.0133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Boorjian SA, Eastham JA, Graefen M, et al. A critical analysis of the long-term impact of radical prostatectomy on cancer control and function outcomes. Eur Urol. 2012;61:664–75. doi: 10.1016/j.eururo.2011.11.053. [DOI] [PubMed] [Google Scholar]
  • 7.Thompson I, Thrasher JB, Canby-Hagino ED, et al. Guideline for the management of clinically localized prostate cancer: 2007 update. American Urological Association Education and Research, Inc; 2007. pp. 1–254. [Google Scholar]
  • 8.Lin DW, FitzGerald LM, Fu R, et al. Genetic variants in the LEPR, CRY1, RNASEL, IL4, and ARVCF genes are prognostic markers of prostate cancer-specific mortality. Cancer Epidemiol Biomarkers Prev. 2011;20:1928–36. doi: 10.1158/1055-9965.EPI-11-0236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Penney KL, Shui IM, Feng Z, et al. Replication of a genetic variant for prostate cancer-specific mortality. Prostate. 2014 doi: 10.1038/pcan.2015.18. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stanford JL, FitzGerald LM, McDonnell SK, et al. Dense genome-wide SNP linkage scan in 301 hereditary prostate cancer families identifies multiple regions with suggestive evidence for linkage. Hum Mol Genet. 2009;18:1839–48. doi: 10.1093/hmg/ddp100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fritz A, Ries L. SEER Extent of Disease, 1988, Codes and Coding Instructions. (3) 1998 Available at: http://www.seer.cancer.gov/manuals/EOD10Dig.3rd.pdf.
  • 12.Sambrook J, Fritsch EF, Maniatis T. Isolation of high-molecular weight DNA from mammalian cells. In: Nolan C, editor. Molecular Cloning: A Laboratory Manual. Vol. 2. 1989. pp. 9.16–9.9. [Google Scholar]
  • 13.Pankratz VS, de Andrade M, Therneau TT. Random-effects Cox proportional hazards model: general variance components methods for time-to-event data. Genetic Epidemiol. 2005;28:97–109. doi: 10.1002/gepi.20043. [DOI] [PubMed] [Google Scholar]
  • 14.R: A language and environment for statistical computing. The R Foundation for Statistical Computing; Vienna, Austria: 2014. Available at: http://www.R-project.org/ [Google Scholar]
  • 15.Brennan-Laun SE, Ezelle HJ, Li X-L, et al. RNase-L control of cellular mRNAs: roles in biologic functions and mechanisms of substrate targeting. J Inteferon & Cytokine Res. 2014;34:275–88. doi: 10.1089/jir.2013.0147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Carpten J, Nupponen N, Isaacs S, et al. Germline mutations in the ribonuclease L gene in families showing linkage with HPC1. Nat Genet. 2002;30:181–4. doi: 10.1038/ng823. [DOI] [PubMed] [Google Scholar]
  • 17.Bettoun DJ, Scafonas A, Rutledge SJ, et al. Interaction between the androgen receptor and Rnase L mediates cross-talk between the interferon and androgen signaling pathways. J Biol Chem. 2005;280:38898–901. doi: 10.1074/jbc.C500324200. [DOI] [PubMed] [Google Scholar]
  • 18.Schoenfeld JD, Margalit DN, Kasperzyk JL, et al. A single nucleotide polymorphism in inflammatory gene RNASEL predicts outcome after radiation therapy for loclized prostate cancer. Clin Cancer Res. 2013;19:1612–19. doi: 10.1158/1078-0432.CCR-12-2718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Meyer MS, Penney KL, Stark JR, et al. Genetic variation in RNASEL associated with prostate cancer risk and progression. Carcinogenesis. 2010;31:1597–603. doi: 10.1093/carcin/bgq132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Alvarez-Cubero MJ, Entrala C, Fernandez-Rosado F, et al. Predictive value in the analysis of RNASEL genotypes in relation to prostate cancer. Prostate Cancer Prostatic Dis. 2012;15:144–9. doi: 10.1038/pcan.2011.56. [DOI] [PubMed] [Google Scholar]
  • 21.San Francisco IF, Rojas PA, Torres-Estay V, et al. Association of RNASEL and 8q24 variants with the presence and aggressiveness of hereditary and sporadic prostate cancer in a Hispanic population. J Cell Mol Med. 2014;18:125–33. doi: 10.1111/jcmm.12171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ginsberg G, Angle K, Guyton K, et al. Polymorphism in the DNA repair enzyme XRCC1: utility of current database and implications for human health risk assessment. Mutat Res. 2011;727:1–15. doi: 10.1016/j.mrrev.2011.02.001. [DOI] [PubMed] [Google Scholar]
  • 23.Iyama T, Wilson DM., 3rd DNA repair mechanisms in dividing and non-dividing cells. DNA Repair. 2013;12:620–36. doi: 10.1016/j.dnarep.2013.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wei B, Zhou Y, Xu Z, et al. XRCC1 Arg399Gln and Arg194Trp polymorphisms in prostate cancer risk: a meta-analysis. Prostate Cancer Prostatic Dis. 2011;14:225–31. doi: 10.1038/pcan.2011.26. [DOI] [PubMed] [Google Scholar]
  • 25.Langsenlehner T, Renner W, Gerger A, et al. Association between single nucleotide polymorphisms in the gene for XRCC1 and radiation-induced late toxicity in prostate cancer patients. Radiother Oncol. 2011;98:387–93. doi: 10.1016/j.radonc.2011.01.021. [DOI] [PubMed] [Google Scholar]
  • 26.Cheung M, Testa JR. Diverse mechanisms of AKT pathway activation in human malignancy. Curr Cancer Drug Targets. 2013;13:234–44. doi: 10.2174/1568009611313030002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hers I, Vincent EE, Tavare JM. Akt signalling in health and disease. Cell Signalling. 2011;23:1515–27. doi: 10.1016/j.cellsig.2011.05.004. [DOI] [PubMed] [Google Scholar]
  • 28.Larue L, Bellacosa A. Epithelial-mesenchymal transition in development and cancer: role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene. 2005;24:7443–54. doi: 10.1038/sj.onc.1209091. [DOI] [PubMed] [Google Scholar]

RESOURCES