Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Mar 25.
Published in final edited form as: Can J Urol. 2008 Feb;15(1):3872–3882.

Genetic susceptibility to prostate cancer in men of African descent: implications for global disparities in incidence and outcomes

Charnita M Zeigler-Johnson 1, Elaine Spangler 1, Mohamed Jalloh 2, Serigne M Gueye 2, Hanna Rennert 3, Timothy R Rebbeck 1,4
PMCID: PMC3064717  NIHMSID: NIHMS280352  PMID: 18304397

Abstract

Introduction

Disparities in prostate cancer incidence and outcomes are a hallmark of the global pattern of prostate cancer, with men of African descent suffering disproportionately from this disease. The causes of these disparities are poorly understood.

Methods

A review of the literature was undertaken to evaluate the role that genetic susceptibility may play in prostate cancer etiology and outcomes, with a particular emphasis on disparties.

Results

The genetic contribution to prostate cancer is well established, and a number of candidate prostate cancer genes have been identified. Significant differences in the frequency of risk alleles in these genes have been identified across the major races. These allele frequency differences may in part explain an increased susceptibility to prostate cancer in some populations. In addition, non-genetic factors contribute significantly to prostate cancer disparities, and the cumulative contribution of both genetic and non-genetic factors to poor-prognosis prostate cancer may explain the poorer outcomes experienced by men of African descent.

Conclusions

Prostate cancer disparities are a function of genetic susceptibility as well as environment, behavior, and health care factors acting in the context of this genetic susceptibility. Elimination of global prostate cancer disparities requires a full understanding of the effects of all of these factors on prostate cancer etiology and outcomes.

Keywords: prostate cancer, African descent, genetics, disparities

Introduction

Prostate cancer disproportionately affects men of African descent in terms of incidence, morbidity, and mortality. There are significant gaps in our knowledge about the factors that predict disparities in prostate cancer incidence and outcomes between men of African and European descent. This is true both in the United States as well as other geographic locations, including Africa and the Caribbean. In order to fully understand the source and solution to these disparities, it is imperative to integrate diverse scientific disciplines, methods, and analytic approaches to understand the multiple contributions to disparities in prostate cancer, including factors associated with the social environment (e.g., economic status, access to health care, social isolation), the physical environment (e.g., location or type of residence or medical care setting), behavior (e.g., attitudes, beliefs, and practices associated with prostate cancer screening), and biology (e.g., inherited genotypes that may affect the development of prostate cancer or predict the aggressiveness of a prostate tumor).

Evidence of global prostate cancer disparities

Prostate cancer has one of the highest incidences and prevalences of any cancer in the world, accounting for 6.9% of all cancers diagnosed. Prostate cancer accounts for 9.7% of all cancers in men, including 15% of cancers in developed countries and 4% of cancers in less developed areas of the world. It is also responsible for almost 6% of cancer deaths in men worldwide. About 600000 new cases of prostate cancer are diagnosed each year, and approximately 200000 deaths are attributed to prostate cancer. Three-fourths of prostate cancer cases occur in men over the age of 64 years.1

Prostate cancer rates vary significantly by geographical region, Table 1. The incidence of prostate cancer is highest in countries where prostate-specific antigen (PSA) screening for prostate cancer is common.1 These rates are influenced greatly by the detection of latent or asymptomatic prostate cancers through the use of screening modalities. Screening with PSA has not been applied in all regions, including Africa, to the same degree as in the United States.

TABLE 1.

Sample prostate cancer incidence and mortality rates (per 100000)

Region Subgrouping Age-specific
incidence
Age-specific
mortality
Source
World 25.3 8.2
Developed
countries
56.2 13.5 IARC105
Developing
countries
9.4 5.2
Africa Eastern Africa 13.8 11.8
Middle Africa 24.5 21.1
Northern Africa 5.8 4.9 IARC105
Southern Africa 40.5 22.4
Western Africa 19.3 16.0
Americas The Caribbean 52.4 28.0 IARC105
Brazil 53.2 15.8
United States: all races 168.0 27.9
African American 255.5 62.3
European American 161.4 25.6 SEER, 2004
American Indian/
Alaskan Native
68.2 21.5
Asian or Pacific Islander 96.5 11.3
Hispanic 140.8 21.2
Asia Eastern Asia 3.8 1.9
Europe Northern Europe 57.5 19.7 IARC105
Western Europe 61.6 17.5
Oceania New Zealand 79.9 18.1

While rates of prostate cancer are high in Europe and North America, the incidence of prostate cancer is highest among men of African descent in North America and the Caribbean. African American men are at particularly high risk for prostate cancer. Recent SEER data (2006) indicate that the incidence of prostate cancer in African American men is higher than in any other group, with an age-adjusted incidence of 255.5 per 100000 versus 161.4 per 100000 in European Americans. African American men also present with more advanced disease at initial diagnosis,2 and have a worse prognosis than white men.3 However, the reason for these differences is not completely understood. It has been hypothesized that genetics and environmental exposures may play a role in determining these high rates.4,5

Like incidence, prostate cancer mortality rates are highest in populations of African descent. A study by Thompson et al6 found that after controlling for prognostic variables, African American men were more likely than European American men to have inferior outcomes after receiving hormonal therapy for prostate cancer. This ethnic disparity suggests that there may be a biological difference in prostate cancer as it manifests in African Americans versus European Americans.

Reported prostate cancer rates are low in native Africans, perhaps due to underreporting of the disease. Although not reflected in many international reports, prostate cancer appears to be one of the most prevalent urological malignancies in native Africans.7 Rates in East and West African populations have been reported to be higher than those in North Africa and other developing countries. However, the validity of these rates in West African populations is not known, and it is possible that rates are substantially higher than those reported in the existing registries.8

Prostate cancer may pose a significant public health burden in some African countries, as the majority of newly diagnosed cases present with advanced disease, including poor differentiation of the tumor, metastasis, and neurological symptoms.9-12 Diagnosis at this later stage of disease often is associated with poor prognosis, which has devastating effects in countries with limited medical resources.9,12 Because of differences in screening practices which increase the number of low grade or asymptomatic cancers detected, prostate cancer survival is estimated at 80% in the United States compared to 40% in developing nations.1 A combination of low socioeconomic status, late disease presentation, and limited health care access for prostate cancer treatment in many African countries may contribute to the poor prognosis that African men face after being diagnosed with prostate cancer.13

Because African and African American men have common ancestry, knowledge of prostate cancer in African men, and comparisons of prostate cancer in African American men and men of the African Diaspora may provide valuable clues about the causes, prevention and treatment of prostate cancer. International comparisons of prostate cancer rates are complicated by differences in prostate cancer screening, diagnosis and reporting systems.11,14

Research on prostate cancer disparities by ethnicity has revealed that there are ethnic differences across populations in terms of diagnostic characteristics and prognostic characteristics, with African Americans having an earlier age at diagnosis and higher PSA levels compared with European Americans.10,15,16 However, these ethnic disparities are not explained entirely by inequities in socioeconomic status or access to an use of health care.17

Despite this evidence, the reasons for disparity in prostate cancer etiology and outcomes in men of African descent are not well understood. Previous research has largely identified individual factors that may be associated with prostate cancer risk or disparities, including family history, age, race, and possibly exposures. However, this research has not generally considered the interaction of multiple factors and the neighborhood context in which they act in determining the causes of disparities. A transdisciplinary approach that considers multiple causative agents may be required to fully understand disparities in prostate cancer and the translation of this information into meaningful disparities reduction approaches. Here, we focus on the possible role of genetic susceptibility in affecting prostate cancer disparities.

Genetic susceptibility

Genetic contribution to prostate cancer risk is well established. Men with one, two or three first-degree affected relatives have a 2-,5- and 11-fold increased risk of developing prostate cancer, respectively.18 In a study of the risk of prostate cancer among 44788 pairs of twins in the Scandinavian countries, 42% of cases were attributed to inheritance, with the remainder considered to be most likely due to environmental factors.19 These findings have been confirmed by other studies as well.20,21 Epidemiological studies also suggested that about 9% of familial prostate cancer cases diagnosed by age 85 are caused by transmission of a rare high risk allele, and that this allele accounts for approximately half of the prostate cancer cases diagnosed before age 55.18 Examples of the genes that may explain these effects are provided below.

Candidate androgen metabolism genes

Of the potential candidate gene pathways, a substantial amount of work has been focused on androgen metabolism genetics. Testosterone is a major determinant of prostate growth and differentiation. There are numerous lines of evidence that support the role of androgen metabolism in prostate cancer etiology. Circulating levels of androgens have been reported to be higher in populations at increased prostate cancer risk, including African American men,22 and lower in populations at decreased prostate cancer risk, such as Chinese men.23 Although serum levels of testosterone do not correlate well with prostate cancer risk,24, serum levels of dihydrotestosterone (DHT) and other testosterone metabolites do correlate with prostate cancer risk.22,24 Second, there is abundant clinical evidence that androgens are related to the growth and development of prostate cancers. Androgen ablation in men with hormone-sensitive prostate cancer reduces tumor size, and decreases the associated disease burden.25 This evidence suggests that the disposition of testosterone may be important in determining prostate cancer risk.

There are several enzymes that determine the activation or inactivation of testosterone, which subsequently influences the signaling capability of testosterone metabolites in androgen-sensitive cells. These genes include the 5 alpha-reductase type II (SRD5A2) and the cytochromes p450 CYP3A4, CYP3A5 and CYP3A43. In addition, the androgen receptor (AR) gene encodes a ligand-activated receptor that mediates androgen signaling response.

The SRD5A2 gene encodes the steroid 5α-reductase type II, which converts testosterone to DHT. The valine to leucine missense variant at codon 89 (V89L) and the alanine to threonine missense variant at codon 49 (A49T) are common SRD5A2 variants that have been associated with prostate cancer etiology or severity.26-31 The V89L polymorphism on the SRD5A2 is believed to decrease the conversion of testosterone to DHT32 while A49T is believed to increase the conversion of testosterone to DHT.30 However, associations of prostate cancer risk involving these variants have not been consistent in all studies.29,33-40

The CYP3A multigene family lies in a region of chromosome 7q21-q22, which includes CYP3A4, CYP3A5, CYP3A7 and CYP3A43 in addition to pseudogenes. Only CYP3A4, CYP3A5, CYP3A7, and CYP3A43 are expressed in adults. Previous reports suggested that linkage disequilibrium exists at the CYP3A locus.41-43 Linkage disequilibrium between CYP3A4 and CYP3A5 in particular, suggests that associations at one locus could be the result of causative effects at the other locus.42

The CYP3A genes are involved in the metabolic deactivation (hydroxylation) of testosterone.44,45 These genes convert testosterone to 2β-, 6β-, or 15β-hydroxytestosterone, and therefore shunt testosterone away from the more biologically active DHT. However, the function of CYP3A4*1B has been controversial. In addition to epidemiological evidence that CYP3A4*1B is associated with prostate cancer, the basic science literature has not consistently supported a functionally significant effect. A number of authors have studied the relationship of CYP3A4 expression or function of CYP3A4*1B.46-52 Most of these authors concluded that no biologically meaningful effects existed given the small magnitude of effects that were observed. However, almost all studies have reported consistent elevations in expression associated with CYP3A4*1B in the range of 20%-200% increase over the consensus CYP3A4*1A.

CYP3A5*1 is the only CYP3A5 allele to date that produces high levels of full length CYP3A5 mRNA and expresses CYP3A5.41 The more common CYP3A5 polymorphism in European Americans, CYP3A5*3, produces an aberrantly spliced mRNA with a premature stop codon. Therefore, there is ample reason to believe that the CYP3A5 alleles studied here could have a functionally meaningful effect on disease etiology. CYP3A5*1 has been inversely associated with prostate cancer.43

In addition, reports have shown a significant association of CYP3A4 and CYP3A43 with occurrence of prostate cancer.42 While CYP3A5*1 had no effect on disease occurrence alone in that study, CYP3A43 increased risk of disease in men with a family history of disease, while CYP3A4*1B had an overall protective effect. It is unclear why CYP3A43 is associated with prostate cancer when examined alone. CYP3A43 is preferentially expressed in the prostate,53 but it has rarely been studied. As a result, there is not enough basic science about these genes that can explain the associations with family history-positive prostate cancer. However, one might speculate that this variant is more commonly inherited in men who have a family history of prostate cancer and may be a candidate hereditary gene for prostate cancer.

When CYP3A4 and CYP3A43 were considered in pairwise interactions in our earlier study to determine the effect of having genotypes with at least one CYP3A4*1B and at least one CYP3A43*3 allele, there were highly significant protective effects for early onset prostate cancer.42 This is an interesting finding, as it suggests an association that may have its greatest impact in African Americans. While only 4% of European Americans carry this allelic combination, it was a more common haplotype observed in our African American sample (35%). Therefore, a subset of men who carry the CYP3A4*1B and CYP3A43*3 combination and are likely African American are significantly less likely to have been diagnosed with prostate cancer before age 60. The reasons for this association are not understood, as African Americans, in general, are at high risk to be diagnosed with prostate cancer prior to the age of 60 years.

The androgen receptor (AR), located on the X chromosome, plays a major role in the development and functioning of the prostate gland. AR is expressed in all histologic types and stages of prostate cancer and its transactivation domain is highly polymorphic.54 Several regions of repetitive polymorphic DNA sequences exist in AR, including CAG trinucleotide repeats encoding polyglutamine residues and GGN repeats encoding polyglycine residues. Several studies have demonstrated an inverse association between the number of CAG and GGN repeats and risk of prostate cancer, advanced cancer, and risk of associated mortality.55-62 Still, some studies suggest that a postivie association exists between prostate cancer and long GGN repeats in combination with short CAG repeats62 Vijayalakshmi.63 The combination of GGN/CAG repeats may be the key to understanding how the AR is functioning to promote or inhibit prostate cancer development and progression.62-64

Race-specific effects

Genotypes involved in prostate cancer etiology differ significantly across ethnicities. For example, the allele frequencies in candidate prostate cancer susceptibility genes such as CYP3A4 and SRD5A2 differ substantially by ethnicity. We have reported a 4-5-fold higher rate of this CYP3A4 vriant in African Americans relative to European Americans.65 Similarly, Ross et al66 and Jaffe et al29 have reported significant ethnic variation in SRD5A2 genotype frequencies that rack with race-specific differences in prostate cancer risk. More recently, Zeigler-Johnson et al67 reported significant differences in the frequency of V89L variant in the SRD5A2 gene by ethnicity, with an L allele frequency of 30% in European Americans, 27% in African Americans, 19% in Ghanaians, and 18% in Senegalese (p = 0.002). Differences were also observed for CYP3A4*1B, with *1B frequencies of 8% in European Americans, 59% in African Americans, 81% in Ghanaians, and 78% in Senegalese (p = 0.0001). When these data were pooled with data from previous studies, significant ethnic differences were observed for each of the polymorphisms. Overall, Asians were least likely to have SRD5A2-V89L and CYP3A4*1B while Africans, followed by African Americans, were most likely to have those alleles. These results suggest that ethnicity-specific differences in allele and genotype frequencies exist for candidate prostate cancer genes. They further suggest that prostate cancer risk across ethnicity (lowest in Asians and highest in African Americans) may be correlated with allele frequencies at candidate prostate cancer susceptibility genes. It remains unknown whether or how these inherited genotypes may explain prostate cancer risk and variability in that risk across racial or ethnic groups. However, these findings suggest that African populations may be at genotypically increased prostate cancer risk, even though the actual magnitude of risk is not well characterized. Because so little is known about the genetics of prostate cancer in Africa, research in this area will provide insight into disease etiology and ethnic disparities worldwide that are associated with prostate cancer incidence and mortality.

Non-candidate susceptibility genes identified by other methods

Family-based studies have yielded numerous prostate cancer susceptibility genes, including ELAC2/HPC2 at 17p (MIM 605367),68 2′-5′-oligoadenylate-dependent RNase L (RNASEL/HPC1) (MIM 180435)69 and macrophage scavenger receptor 1 (MSR1) (MIM 153622).70 Two of these genes, RNASEL and MSR1 have been shown to play a major role in inflammation and innate immunity.

2′-5′-oligoadenylate-dependent RNase L (RNASEL) is a constitutively expressed latent endonuclease that mediates the antiviral and proapoptotic activities of the interferon-inducible 2-5A system.71,72 Initial linkage analysis studies revealed the presence of two deleterious mutations, Met1Ile and Glu265X in African American and European American families, respectively, that segregated with the disease.69 Glu265X was also associated with hereditary prostate cancer in other patients from European origin,73 while Met1Ile was not found in other studies. Two additional deleterious frameshift mutations, 471delAAAG and 147dupCAAT, in this gene was detected in Ashkenazi Jewish and Asian Indian men, respectively.74 Several missense variants in RNASEL have also been detected.73,75 One of these variants, Arg462Gln, was implicated in up to 13% of prostate cancer cases using a family-based case-control study.76 Although there is a low frequency of deleterious mutations in RNASEL, functional studies strongly implicate this gene in the disease. Prostate cancer patients carrying the Glu265X or the 471delAAAG mutation showed loss of heterozygosity of the wild type allele in microdissected tumor DNA,69,74 while the Arg462Gln had a significantly lower RNASEL enzymatic activity compared to the normal protein.76

Macrophage scavenger receptor 1 (MSR1; Chromosome 8p22) is a member of an expanded family of membrane receptors collectively termed scavenger receptors. MSR1 can bind many chemically modified molecules ranging from bacteria to modified lipoproteins. Correspondingly, MSR1 has been associated with a wide variety of normal and pathological processes, including inflammation, innate and adaptive immunity, oxidative stress and apoptosis.77 MSR1 maps to the 8p22 chromosome region, wich is commonly deleted in prostate cancer.78 Six rare missense variants and one nonsense mutation within MSR1 were observed to co-segregate with the disease in hereditary prostate cancer families.70 Furthermore, the prevalence of MSR1 mutations in prostate cancer cases of European and African American descent was substantially higher compared to unaffected men.70 Arg293X and Ser41Tyr were the most common mutations detected among prostate cancer patients of European and African American descent, respectively.79

ELAC2/HPC2 was predicted to encode an evolutionarily conserved, metal-dependent hydrolase, which could partially explain environmental effects on human prostate epithelial cells by postulating differential interactions with environmental exposures.68 ELAC2 was also shown to encode a 3′ processing endoribonuclease, an enzyme responsible for the removal of a 3′ trailer from precursor RNA80 and to interact with γ-tubulin, a component of the mitotic apparatus,81 suggesting a possible role for ELAC2 in cell cycle control. Initial sequence analyses of the ELAC2 gene identified rare mutations and two common missense changes, Ser217Leu and Ala541Thr that were reported to be associated with prostate cancer risk.68,82,83 However, confirmation of these results has been difficult with only weak consensus among studies.84 85

Race-specific effects

Rennert et al86 observed significant differences in ELAC2, RNASEL and MSR1 allele frequencies by race. Although no significant association has been found with prostate cancer risk overall, certain effects for MSR1 IVS7delinsTTA and RNASEL Arg462Gln were observed when stratified by race, family history or disease severity in both African American and European American men. No association between the common ELAC2 Ser217Leu and Ala541Thr sequence variants and prostate cancer risk was found. Moreover, Ala541Thr was rare among African Americans compared to European Americans, and therefore was unlikely to explain the higher rate of prostate cancer in the African American racial group. RNASEL Arg462Gln gene variation is of particular interest since it was clearly associated with reduced functional activity, and although not statistically significant, it was more common among African American cases compared to controls (0.12 versus 0.16), Table 2. When stratified by prostate tumor characteristics, Arg462Gln was associated with low-grade (OR = 1.5, 95% CI 1.04-2.2) and early-stage (OR = 1.5, 95% CI 1.02-2.1) disease in family history negative European Americans, while in family history positive individuals, Arg462Gln was inversely associated with low grade (OR = 0.43 95% CI 0.21-0.88) and low stage (OR = 0.46 95% CI 0.22-0.95) disease. In African Americans however, Arg462Gln was associated with positive family history high stage disease (OR = 14.8 95% CI 1.6-135.7).86

TABLE 2.

Allele frequencies and SIFT analysis of missense polymorphisms in ELAC2, MSR1 and RNASEL genes

Gene Nucleotide
sequence
variant
Amino
acid
change
Substitution
probability
(Pi)
Allele frequency (total no. of alleles in sample)
African American European American
Controls Cases Controls Cases
ELAC2/ 650C > T Ser217Leu 0.15 0.228 (254)a 0.211 (180) 0.296 (656) 0.301 (1020)
HPC2 1621G > A Ala541Thr 0.26 0.004 (268)a 0 (190) 0.027 (698) 0.033 (1072)
MSR1 520G > T Asp174Tyr 0.13 0.015 (266)a 0.022 (186) 0 (696) 0 (1064)
876C > T Arg293X 0 0 (262) 0 (166) 0.012 (694)b 0.007 (994)
−14,742A > G None N/A 0.269 (260)ab 0.253 (186)b 0.105 (686) 0.105 (1060)
IVS5-59C > A None N/A 0.012 (246)a 0.006 (170) 0.059 (628) 0.050 (968)
IVS7delTTA None N/A 0.264 (220)a,b 0.218 (156) 0.057 (562) 0.045 (894)
RNASEL/ 793G > T Glu265X 0 0 (268) 0 (188) 0.001 (714) 0.003 (1080)
HPC1 354C > T None N/A 0.004 (266) 0 (182) 0.003 (682) 0.002 (1002)
1385G > A Arg462Gln 0.02 0.119 (252)a 0.159 (176) 0.361 (642) 0.370 (1010)
a

Frequency in control groups differs by race (two-sided Fisher’s exact tests: p < 0.05).

b

Deviations from Hardy-Weinberg proportions based on χ2. Test at p < 0.05.

These conflicting associatons among the different studies may be explained by clinical and genetic heterogeneity of prostate cancer, heterogeneity of study populatons, incomplete penetrance, or non-genetic etiologies (e.g., environmental factors). Screening practices for prostate cancer and the inclusion of patients with clinically insignificant disease probably play a role as well. Follow-up meta-analysis studies on the association of prostate cancer with ELAC2, RNASEL, or MSR1 variants found no association or very weak effects of gene variations in these genes with prostate cancer risk overall in either European Americans or African Americans.84,85,87-90 However due to power constrains, these studies did not evaluate gene variation effects by disease or prostate tumor characteristics. Taken together, these results suggest that although MRS1 and RNASEL do not seem to be associated with disease risk overall, they may still influence disease severity and that this effect possibly varies by family history of cancer and by racial background.

8q24 locus

Recently, a genome-wide linkage study in Iceland91 and an admixture study among African Americans92 independently detected markes associated with prostate cancer risk on chromosome 8q24. Of these, a signel nucleotide, rs1447295, at 8q24.21 (denoted region1) was most strongly associated with prostate cancer risk in pooled case-control studies of Caucasians from several countries.91.93-95 Weaker effects, however, were noted for this SNP among African American men.93 SNP marker rs1447295 was also reported to be associated with prostate cancer aggressiveness, but this effect varied widely by popylation and study group.96,97 Moreover, a second region approximately 350 kb upstream to the previously reported rs1447295, denoted region 2, also demonstrated strong association with prostate cancer93,94 in both Caucasians and African Americans. These effects were independent of those detected for region 1. Finally, a third region located between region 1 and region 2 approximately 70 kb centromeric to rs1447295 was also found to be strongly associated with prostate risk in alarge nested case-control study of Caucasians, suggesting the presence of at least two independent loci within 8q24 that may contribute to prostate cancer riks in this population.95

Relationship of genotypic susceptibility and other factors

Genetic susceptibility represents only one piece of the very complex nature of prostate cancer etiology. Underlying differences in susceptibility to develop prostate cancer is manifest through exposure to environmental agents, social environmental and neighborhood context, behavior, access to quality health care, and other factors. To fully understand the causes of prostate cancer diparities across groups, a comprehensive approach to the study of many factors involved in prostate cancer causation is required.

Environmental exposure

Relatively few exposures have been consistently associated with prostate cancer risk, and very little information about the role of exposures in establishing prostate cancer disparties is available. Thus, expanded definitions of “the environment” could be developed to include social environment (e.g., socioeconomic status, access to health care, social isolation, cultural beliefs and values); physical environment (e.g., location or type of residence, access to computer and internet resources, or medical care); and behavioral facrtors (e.g., attitudes, beliefs and practices associated with cancer screening). Thus “environment” could include both individual-level factors as well as neighborhood- or community-level factors using a multilevel approach. For example, neighborhood-level factors could include housing density, measures of social captical such as cultural/civic participation or neighborhood cohesiveness, neighborhood stability such as the percent of rental housing, measures of deprivation such as violent crime rate, and social conditions such as violent crime rate, and social conditions such as percent of individuals in the neighborhood who live below the poverty level or average educational attainment. Similarly, institutional factors such as health care patterns, access to care, insurance, and type and quality of health care that has been accessed could also be considered. In general, research has focused on individual-level variables, and therefore has not been able to address the larger context in which genes, biological factors, or individual environmental exposures are acting.

Health care

Access to and choice of prostate cancer treatment may have a profound effect on disparities in outcome.98 African American men are more likely than European American men to undergo watchful waiting instead of aggressive therapy for localized prostate cancer99-101 and differences in treatment and mortality persist after adjusting for individual factors such as stage, grade, and socioeconomic status.99 This remains true despite the observation that mortality differences can be explained by lower rates of aggressive treatment in African American men.102,103 A strong relationship exists between cancer outcomes and hospital and provider characteristics, prevalent distrust of research/teaching hospitals among African Americans, and links between residential segregation and racial alienation.98 However, there remain little data about the role of social environment, including factors that affect access to care, contributes to racial disparities in access to quality prostate cancer treatment.

Social environment and behavior

Social environment, including culture, is increasingly recognized as having an important impact on cancer outcomes in ethnically diverse population.98 Environmental stressors such as life stress, racism, and discrimination may have a deleterious impact on physiological (i.e., immune functioning, cardiovascular reactivity) and behavioral (i.e., coping efforts, dietary behaviours, smoking) responses to prostate cancer.104 While African ancestry is a marker for other factors that might better explain quality of life, ethnicity alone does not provide information on the causes of these disparities. Incorporating biological and other factors into the application of assessment of risk and outcomes could improve interventions that could be addressed through psycho-educational approaches designed to facilitate stress reduction and increased confidence to cope with treatment-related side effects98.

Implications for prostate cancer disparities

Our limited knowledge about the genetic and other biological events that cause prostate cancer disparities presents a major barrier to eliminating these disparities. The accumulating knowledge of the human genome provides an opportunity to apply knowledge of carcinogenic mechanisms to the problem of prostate cancer disparities. The incorporation of biomarkers in cancer disparties research provides new opportunities for clinical and public health research and practice, and has the potential to catalyze needed improvements in the prevention and management of cancer to eliminate cancer disparities.

Prostate cancer may be a major cancer burden in African men, so it will be important to understand etiological factors including inherited genotypes that confer risk of developing prostate cancer. Understanding prostate cancer in Africa may inform us about inherited predisposition, modifiable risk factors, and disease prevention in the high risk African American population. Additionally, knowledge of the interactions of prostate cancer susceptibility genes, environment, and behavior could be used to identify individuals at risk of developing prostate cancer with poor outcomes for heightened screening or prevention modalities, and to identify optimal treatment strategies for men of African descent.

Acknowledgements

This project was supported by grants to Dr. Timothy R. Rebbeck (RO1-CA85074, P50-CA105641), Dr. Charnita M. Zeigler-Johnson (K07-CA106730), and Dr. Hanna Rennert (Department of Defense W81XWH-05-1-0198).

References

  • 1.Parkin DM, Bray FI, Devesa SS. Cancer burden in the year 2000. The global picture. Eur J Cancer. 2001;37:S4. doi: 10.1016/s0959-8049(01)00267-2. [DOI] [PubMed] [Google Scholar]
  • 2.Landis SH, Murray T, Bolden S, et al. Cancer statistics, 1999. CA Cancer J Clin. 1999;49:8. doi: 10.3322/canjclin.49.1.8. [DOI] [PubMed] [Google Scholar]
  • 3.Moul JW. Prostate cancer in African American men. Prostate Cancer Prostalic Dis. 1998;1:109. doi: 10.1038/sj.pcan.4500222. [DOI] [PubMed] [Google Scholar]
  • 4.Hayes RB, Liff JM, Pottern LM, et al. Prostate cancer risk in U.S. blacks and whites with a family history of cancer. Int J Cancer. 1995;60:361. doi: 10.1002/ijc.2910600315. [DOI] [PubMed] [Google Scholar]
  • 5.Cunningham GR, Ashton CM, Annegers JF, et al. Familial aggregation of prostate cancer in African-Americans and white Americans. Prostate. 2003;56:256. doi: 10.1002/pros.10252. [DOI] [PubMed] [Google Scholar]
  • 6.Thompson I, Tangen C, Tolcher A, et al. Association of African-American ethnic background with survival in men with metastatic prostate cancer. J Natl Cancer Inst. 2001;93:219. doi: 10.1093/jnci/93.3.219. [DOI] [PubMed] [Google Scholar]
  • 7.Koulibaly M, Kabba IS, Cisse A, et al. Cancer incidence in Conakry, Guinea: first results from the Cancer Registry 1992-1995. Int J Cancer. 1997;70:39. doi: 10.1002/(sici)1097-0215(19970106)70:1<39::aid-ijc6>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
  • 8.Wabinga HR, Parkin DM, Wabwire-Mangen F, et al. Cancer in Kampala, Uganda, in 1989-91: changes in incidence in the era of AIDS. Int J Cancer. 1993;54:26. doi: 10.1002/ijc.2910540106. [DOI] [PubMed] [Google Scholar]
  • 9.Osegbe DN. Prostate cancer in Nigerians: facts and nonfacts. J Urol. 1997;157:1340. [PubMed] [Google Scholar]
  • 10.Gueye SM, Ziegler-Johnson CM, Friebel T, et al. Clinical characteristics of prostate cancer in African Americans, American whites, and Senegalese men. Urology. 2003;61:987. doi: 10.1016/s0090-4295(02)02588-8. [DOI] [PubMed] [Google Scholar]
  • 11.Ekwere PD, Egbe SN. The changing pattern of prostate cancer in Nigerians: current status in the southeastern states. J Natl Med Assoc. 2002;94:619. [PMC free article] [PubMed] [Google Scholar]
  • 12.Dawam D, Rafindadi AH, Kalayi GD. Benign prostatic hyperplasia nd prostate carcinoma in native Africans. BJU Int. 2000;85:1074. doi: 10.1046/j.1464-410x.2000.00677.x. [DOI] [PubMed] [Google Scholar]
  • 13.Kehinde EO. The geography of prostate cancer and its treatment in Africa. Cancer Surv. 1995;23:281. [PubMed] [Google Scholar]
  • 14.Oliver SE, May MT, Gunnell D. International trends in prostate-cancer mortality in the “PSA ERA”. Int J Cancer. 2001;92:893. doi: 10.1002/ijc.1260. [DOI] [PubMed] [Google Scholar]
  • 15.Fowler JE, Jr, Bigler SA. Racial differences in prostate carcinogenesis. Histologic and clinical observations. Urol Clin North Am. 2002;29:183. doi: 10.1016/s0094-0143(02)00003-4. [DOI] [PubMed] [Google Scholar]
  • 16.Bunker CH, Patrick AL, Maharaj G, et al. Prostate cancer risk is three-fold higher among men, aged 50-64, of African descent compared with men of Asian-Indian descent in Trinidad and Tobago. Ethn Dis. 2002;12:S3. [PubMed] [Google Scholar]
  • 17.Hoffman RM, Gilliland FD, Eley JW, et al. Racial and ethnic differences in advanced-stage prostate cancer: the Prostate Cancer Outcomes Study. J Natl Cancer Inst. 2001;93:388. doi: 10.1093/jnci/93.5.388. [DOI] [PubMed] [Google Scholar]
  • 18.Carter BS, Beaty TH, Steinberg GD, et al. Mendelian inheritance of familial prostate cancer. Proc Natl Acad Sci USA. 1992;89:3367. doi: 10.1073/pnas.89.8.3367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lichtenstein P, Holm NV, Verkasalo PK, et al. Environmental and heritable factors in the causation of cancer— analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 2000;343:78. doi: 10.1056/NEJM200007133430201. [DOI] [PubMed] [Google Scholar]
  • 20.Gronberg H, Damber L, Damber JE. Studies of genetic factors in prostate cancer in a twin population. J Urol. 1994;152:1484. doi: 10.1016/s0022-5347(17)32452-7. [DOI] [PubMed] [Google Scholar]
  • 21.Ghadirian P, Howe GR, Hislop TG, et al. Family history of prostate cancer: a multi-center case-control study in Canada. Int J Cancer. 1997;70:679. doi: 10.1002/(sici)1097-0215(19970317)70:6<679::aid-ijc9>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 22.Ross RK, Bernstein L, Lobo RA, et al. 5-alpha-reductase activity and risk of prostate cancer among Japanese and US white and black males. Lancet. 1992;339:887. doi: 10.1016/0140-6736(92)90927-u. [DOI] [PubMed] [Google Scholar]
  • 23.Lookingbill DP, Demers LM, Wang C, et al. Clinical and biochemical parameters of androgen action in normal healthy Caucasian versus Chinese subjects. J Clin Endocrinol Metab. 1991;72:1242. doi: 10.1210/jcem-72-6-1242. [DOI] [PubMed] [Google Scholar]
  • 24.Gann PH, Hennekens CH, Ma J, et al. Prospective study of sex hormone levels and risk of prostate cancer. J Natl Cancer Inst. 1996;88:1118. doi: 10.1093/jnci/88.16.1118. [DOI] [PubMed] [Google Scholar]
  • 25.Oesterling JE, Roy J, Agha A, et al. The Finasteride PSA Study Group Biologic variability of prostate-specific antigen and its usefulness a marker for prostate cancer: effects of finasteride. Urology. 1997;50:13. doi: 10.1016/s0090-4295(97)00156-8. [DOI] [PubMed] [Google Scholar]
  • 26.Nam RK, Toi A, Vesprini D, et al. V89L polymorphism of type-2, 5-alpha reductase enzyme gene predicts prostate cancer presence and progression. Urology. 2001;57:199. doi: 10.1016/s0090-4295(00)00928-6. [DOI] [PubMed] [Google Scholar]
  • 27.Soderstrom TW. 5alpha-reductase 2 polymorphisms as risk factors in prostate cancer. Pharmacogenetics. 2002;12:307. doi: 10.1097/00008571-200206000-00006. [DOI] [PubMed] [Google Scholar]
  • 28.Shibata A, Garcia MI, Cheng I, Stamey TA, McNeal JE, Brooks JD, Henderson S, Yemoto CE, Peehl DM. Polymorphisms in the androgen receptor and type II 5 alpha-reductase genes and prostate cancer prognosis. Prostate. 2002;52:269. doi: 10.1002/pros.10119. [DOI] [PubMed] [Google Scholar]
  • 29.Jaffe JM, Malkowicz SB, Walker AH, et al. Association of SRD5A2 genotype and pathological characteristics of prostate tumors. Cancer Res. 2000;60:1626. [PubMed] [Google Scholar]
  • 30.Makridakis NM, Ross RK, Pike MC, et al. Association of mis-sense substitution in SRD5A2 gene with prostate cancer in African-American and Hispanic men in Los Angeles, USA. Lancet. 1999;354:975. doi: 10.1016/S0140-6736(98)11282-5. [DOI] [PubMed] [Google Scholar]
  • 31.Li Z, Habuchi T, Mitsumori K, et al. Association of V89LSRD5A2 polymorphism with prostate cancer development in a Japanese population. J Urol. 2003;169:2378. doi: 10.1097/01.ju.0000056152.57018.31. [DOI] [PubMed] [Google Scholar]
  • 32.Makridakis N, Ross RK, Pike MC, et al. A prevalent missense substitution that modulates activity of prostatic steroid 5alpha-reductase. Cancer Res. 1997;57:1020. [PubMed] [Google Scholar]
  • 33.Lamharzi N, Johnson MM, Goodman G, et al. Polymorphic markers in the 5alpha-reductase type II gene and the incidence of prostate cancer. Int J Cancer. 2003;105:480. doi: 10.1002/ijc.11126. [DOI] [PubMed] [Google Scholar]
  • 34.Lunn RM, Bell DA, Mohler JL, et al. Prostate cancer risk and polymorphism in 17 hydroxylase (CYP17) and steroid reductase (SRD5A2) Carcinogenesis. 1999;20:1727. doi: 10.1093/carcin/20.9.1727. [DOI] [PubMed] [Google Scholar]
  • 35.Febbo PG, Kantoff PW, Platz EA, et al. The V89L polymorphism in the 5alpha-reductase type 2 gene and risk of prostate cancer. Cancer Res. 1999;59:5878. [PubMed] [Google Scholar]
  • 36.Hsing AW, Chen C, Chokkalingam AP, et al. Polymorphic markers in the SRD5A2 gene and prostate cancer risk: population-based case-control study. Cancer Epidemiol Biomarkers Prev. 2001;10:1077. [PubMed] [Google Scholar]
  • 37.Pearce CL, Makridakis NM, Ross RK, et al. Steroid 5-alpha reductase type II V89L substitution is not associated with risk of prostate cancer in a multiethnic population study. Cancer Epidemiol Biomarkers Prev. 2002;11:417. [PubMed] [Google Scholar]
  • 38.Mononen N, Ikonen T, Syrjakoski K, et al. A missense substitution A49T in the steroid 5-alpha-reductase gene (SRD5A2) is not associated with prostate cancer in Finland. Br J Cancer. 2001;84:1344. doi: 10.1054/bjoc.2001.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Latil AG, Azzouzi R, Cancel GS, et al. Prostate carcinoma risk and allelic variants of genes involved in androgen biosynthesis and metabolism pathways. Cancer. 2001;92:1130. doi: 10.1002/1097-0142(20010901)92:5<1130::aid-cncr1430>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 40.Giwercman YL, Abrahamsson PA, Giwercman A, et al. The 5alpha-reductase type II A49T and V89L high-activity allelic variants are more common in men with prostate cancer compared with the general population. Eur Urol. 2005;48:679. doi: 10.1016/j.eururo.2005.06.011. [DOI] [PubMed] [Google Scholar]
  • 41.Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27:383. doi: 10.1038/86882. [DOI] [PubMed] [Google Scholar]
  • 42.Zeigler-Johnson C, Friebel T, Walker AH, et al. CYP3A4, CYP3A5, and CYP3A43 genotypes and haplotypes in the etiology and severity of prostate cancer. Cancer Res. 2004;64:8461. doi: 10.1158/0008-5472.CAN-04-1651. [DOI] [PubMed] [Google Scholar]
  • 43.Plummer SJ, Conti DV, Paris PL, et al. CYP3A4 and CYP3A5 genotypes, haplotypes, and risk of prostate cancer. Cancer Epidemiol Biomarkers Prev. 2003;12:928. [PubMed] [Google Scholar]
  • 44.Waxman DJ, Attisano C, Guengerich FP, et al. Human liver microsomal steroid metabolism: identification of the major microsomal steroid hormone 6 beta-hydroxylase cytochrome P-450 enzyme. Arch Biochem Biophys. 1988;263:424. doi: 10.1016/0003-9861(88)90655-8. [DOI] [PubMed] [Google Scholar]
  • 45.Domanski TL, He YA, Khan KK, et al. Phenylalanine and tryptophan scanning mutagenesis of CYP3A4 substrate recognition site residues and effect on substrate oxidation and cooperativity. Biochemistry. 2001;40:10150. doi: 10.1021/bi010758a. [DOI] [PubMed] [Google Scholar]
  • 46.Westlind A, Lofberg L, Tindberg N, et al. Interindividual differences in hepatic expression of CYP3A4: relationship to genetic polymorphism in the 5′-upstream regulatory region. Biochem Biophys Res Commun. 1999;259:201. doi: 10.1006/bbrc.1999.0752. [DOI] [PubMed] [Google Scholar]
  • 47.Amirimani B, Ning B, Deitz AC, et al. Increased transcriptional activity of the CYP3A4*1B promoter variant. Environ Mol Mutagen. 2003;42:299. doi: 10.1002/em.10199. [DOI] [PubMed] [Google Scholar]
  • 48.Amirimani B, Walker AH, Weber BL, et al. RESPONSE: re: modification of clinical presentation of prostate tumors by a movel genetic variant in CYP3A4. J Natl Cancer Inst. 1999;91:1588. doi: 10.1093/jnci/91.18.1588. [DOI] [PubMed] [Google Scholar]
  • 49.Ando Y, Tateishi T, Sekido Y, et al. Re: Modification of clinical presentation of prostate tumors by a novel genetic variant in CYP3A4. J Natl Cancer Inst. 1999;91:1587. doi: 10.1093/jnci/91.18.1587. [DOI] [PubMed] [Google Scholar]
  • 50.Spurdle AB, Goodwin B, Hodgson E, et al. The CYP4A4*1B polymorphism has no functional significance and is not associated with risk of breast or ovarian cancer. Pharmacogenetics. 2002;12:355. doi: 10.1097/00008571-200207000-00003. [DOI] [PubMed] [Google Scholar]
  • 51.Floyd MD, Gervasini G, Masica AL, et al. Genotype-phenotype associations for common CYP3A4 and CYP43A5 variants in the basal and induced metabolism of midazolam in European- and African-American men and women. Pharmacogenetics. 2003;13:595. doi: 10.1097/00008571-200310000-00003. [DOI] [PubMed] [Google Scholar]
  • 52.Hamzeiy H, Vahdati-Mashhadian N, Edwards HJ, et al. Mutation analysis of the human CYP3A4 gene 5′ regulatory region: population screening using non-radioactive SSCP. Mutat Res. 2002;500:103. doi: 10.1016/s0027-5107(01)00305-0. [DOI] [PubMed] [Google Scholar]
  • 53.Domanski TL, Finta C, Halpert JR, et al. cDNA cloning and initial characterization of CYP3A43, a novel human cytochrome P450. Mol Pharmacol. 2001;59:386. doi: 10.1124/mol.59.2.386. [DOI] [PubMed] [Google Scholar]
  • 54.Montgomery JS, Price DK, Figg WD. The androgen receptor gene and its influence on the development and progression of prostate cancer. J Pathol. 2001;195:138. doi: 10.1002/1096-9896(200109)195:2<138::AID-PATH961>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 55.Ingles SA, Ross RK, Yu MC, et al. Association of prostate cancer risk with genetic polymorphisms in vitamin D receptor and androgen receptor. J Natl Cancer Inst. 1997;89:166. doi: 10.1093/jnci/89.2.166. [DOI] [PubMed] [Google Scholar]
  • 56.Hakimi JM, Schoenberg MP, Rondinelli RH, et al. Androgen receptor variants with short glutamine or glycine repeats may identify unique subpopulations of men with prostate cancer. Clin. Cancer Res. 1997;3:1599. [PubMed] [Google Scholar]
  • 57.Zeegers MP, Kiemeney LA, Nieder AM, et al. How strong is the association between CAG and GGN repeat length polymorphisms in the androgen receptor gene and prostate cancer risk? Cancer Epidemiol Biomarkers Prev. 2004;13:1765. [PubMed] [Google Scholar]
  • 58.Binnie MC, Alexander FE, Heald C, et al. Polymorphic forms of prostate specific antigen and their interaction with androgen receptor trinucleotide repeats in prostate cancer. Prostate. 2005;63:309. doi: 10.1002/pros.20178. [DOI] [PubMed] [Google Scholar]
  • 59.Giwercman C, Giwercman A, Pedersen HS, et al. Polymorphisms in genes regulating androgen activity among prostate cancer low-risk Inuit men and high-risk Scandinavians. Int J Androl. 2007 doi: 10.1111/j.1365-2605.2007.00750.x. [DOI] [PubMed] [Google Scholar]
  • 60.Cude KJ, Montgomery JS, Price DK, et al. The role of an androgen receptor polymorphism in the clinical outcome of patients with metastatic prostate cancer. Urol Int. 2002;68:16. doi: 10.1159/000048412. [DOI] [PubMed] [Google Scholar]
  • 61.Giovannucci E, Platz EA, Stampfer MJ, et al. The CAG repeat within the androgen receptor gene and benign prostatic hyperplasia. Urology. 1999;53:121. doi: 10.1016/s0090-4295(98)00468-3. [DOI] [PubMed] [Google Scholar]
  • 62.Hsing AW, Gao YT, Wu G, et al. Polymorphic CAG and GGN repeat lengths in the androgen receptor gene and prostate cancer risk: a population-based case-control study in China. Cancer Res. 2000;60:5111. [PubMed] [Google Scholar]
  • 63.Vijayalakshmi K, Thangaraj K, Rajender S, et al. GGN repeat length and GGN/CAG haplotype variations in the androgen receptor gene and prostate cancer risk in south Indian men. J Hum Genet. 2006;51:998. doi: 10.1007/s10038-006-0051-z. [DOI] [PubMed] [Google Scholar]
  • 64.Ruhayel Y, Lundin K, Giwercman Y, et al. Androgen receptor gene GGN and CAG polymorphisms among severely oligozoospermic and azoospermic Swedish men. Hum Reprod. 2004;19:2076. doi: 10.1093/humrep/deh349. [DOI] [PubMed] [Google Scholar]
  • 65.Walker AH, Jaffe JM, Gunasegaram S, et al. Characterization of an allelic variant in the nifedipine-specific element of CYP3A4: ethnic distribution and implications for prostate cancer risk. Hum Mutat. 1998;12:289. Mutations in brief no. 191. Online. [PubMed] [Google Scholar]
  • 66.Ross RK, Coetzee GA, Pearce CL, et al. Androgen metabolism and prostate cancer: establishing a model of genetic susceptibility. Eur Urol. 1999;35:355. doi: 10.1159/000019909. [DOI] [PubMed] [Google Scholar]
  • 67.Zeigler-Johnson CM, Walker AH, Mancke B, et al. Ethnic differences in the frequency of prostate cancer susceptibility alleles at SRD5A2 and CYP3A4. Hum Hered. 2002;54:13. doi: 10.1159/000066695. [DOI] [PubMed] [Google Scholar]
  • 68.Tavtigian SV, Simard J, Teng DH, et al. A candidate prostate cancer susceptibility gene at chromosome 17p. Nat Genet. 2001;27:172. doi: 10.1038/84808. [DOI] [PubMed] [Google Scholar]
  • 69.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. doi: 10.1038/ng823. [DOI] [PubMed] [Google Scholar]
  • 70.Xu J, Zheng SL, Komiya A, et al. Germline mutations and Sequence variants of the macrophage scavenger receptor 1 gene are associated with prostate cancer risk. Nat Genet. 2002;32:321. doi: 10.1038/ng994. [DOI] [PubMed] [Google Scholar]
  • 71.Zhou A, Hassel BA, Silverman RH. Expression cloning of 2-5A-dependent RNAase: a uniquely regulated mediator of interferon action. Cell. 1993;72:753. doi: 10.1016/0092-8674(93)90403-d. [DOI] [PubMed] [Google Scholar]
  • 72.Zhou A, Paranjape J, Brown TL, et al. Interferon action and apoptosis are defective in mice devoid of 2′,5′-oligoadenylate-dependent RNase L. Embo J. 1997;16:6355. doi: 10.1093/emboj/16.21.6355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rokman A, Ikonen T, Seppala EH, et al. Germline alterations of the RNASEL gene, a candidate HPC1 gene at 1q25, in patients and families with prostate cancer. Am J Hum Genet. 2002;70:1299. doi: 10.1086/340450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rennert H, Bercovich D, Hubert A, et al. A Novel Founder Mutation in the RNASEL Gene, 471delAAAG, Is Associated with Prostate Cancer in Ashkenazi Jews. Am J Hum Genet. 2002;71:4. doi: 10.1086/342775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rennert H, Zeigler-Johnson C, Mittal R, et al. Analysis of the RNASEL/HPC1, and Macrophage Scavenger Receptor 1 in Asian-Indian Advanced Prostate Cancer. doi: 10.1016/j.urology.2007.11.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Casey G, Neville PJ, Plummer SJ, et al. RNASEL Arg462Gln variant is implicated in up to 13% of prostate cancer cases. Nat Genet. 2002;32:581. doi: 10.1038/ng1021. [DOI] [PubMed] [Google Scholar]
  • 77.Platt N, Gordon S. Is the class A macrophage scavenger receptor (SR-A) multifunctional? - The mouse’s tale. J Clin Invest. 2001;108:649. doi: 10.1172/JCI13903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lieberfarb ME, Lin M, Lechpammer M, et al. Genome-wide loss of heterozygoslty analysis from laser capture microdissected prostate cancer using single nucleotide polymorphic allele (SNP) arrays and a novel bioinformatics platform dChipSNP. Cancer Res. 2003;63:4781. [PubMed] [Google Scholar]
  • 79.Xu J, Zheng SL, Komiya A, et al. Common sequence variants of the macrophage scavenger receptor 1 gene are associated with prostate cancer risk. Am. J Hum Genet. 2003;72:208. doi: 10.1086/345802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Takaku H, Minagawa A, Takagi M, et al. A candidate prostate cancer susceptibility gene encodes tRNA 3′ processing endoribonuclease. Nucleic Acids Res. 2003;31:2272. doi: 10.1093/nar/gkg337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Korver W, Guevara C, Chen Y, et al. The product of the candidate prostate cancer susceptibility gene ELAC2 interacts with the gamma-tubulin complex. Int J Cancer. 2003;104:283. doi: 10.1002/ijc.10945. [DOI] [PubMed] [Google Scholar]
  • 82.Rebbeck TR, Walker AH, Zeigler-Johnson C, et al. Association of HPC2/ELAC2 genotypes and prostate cancer. Am J Hum Genet. 2000;67:1014. doi: 10.1086/303096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wang L, McDonnell SK, Elkins DA, et al. Role of HPC2/ELAC2 in hereditary prostate cancer. Cancer Res. 2001;61:6494. [PubMed] [Google Scholar]
  • 84.Meitz JC, Edwards SM, Easton DF, et al. HPC2/ELAC2 polymorphisms and prostate cancer risk: analysis by age of onset of disease. Br J Cancer. 2002;87:905. doi: 10.1038/sj.bjc.6600564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Severi G, Giles GG, Southey MC, et al. ELAC2/HPC2 polymorphisms, prostate-specific antigen levels, and prostate cancer. J Natl Cancer Inst. 2003;95:818. doi: 10.1093/jnci/95.11.818. [DOI] [PubMed] [Google Scholar]
  • 86.Rennert H, Zeigler-Johnson CM, Addya K, et al. Association of susceptibility alleles in ELAC2/HPC2, RNASEL/HPC1, and MSR1 with prostate cancer severity in European American and African American men. Cancer Epidemiol Biomarkers Prev. 2005;14:949. doi: 10.1158/1055-9965.EPI-04-0637. [DOI] [PubMed] [Google Scholar]
  • 87.Camp NJ, Tavtigian SV. Meta-analysis of associations of the Ser217Leu and Ala541Thr variants in ELAC2 (HPC2) and prostate cancer. Am J Hum Genet. 2002;71:1475. doi: 10.1086/344516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vesprini D, Nam RK, Trachtenberg J, et al. HPC2 variants and screen-detected prostate cancer. Am J Hum Genet. 2001;68:912. doi: 10.1086/319502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sun J, Hsu FC, Turner AR, et al. Meta-analysis of association of rare mutations and common sequence variants in the MSR1 gene and prostate cancer risk. Prostate. 2006;66:728. doi: 10.1002/pros.20396. [DOI] [PubMed] [Google Scholar]
  • 90.Li H, Tai BC. RNASEL gene polymorphisms and the risk of prostate cancer: a meta-analysis. Clin Cancer Res. 2006;12:5713. doi: 10.1158/1078-0432.CCR-05-2799. [DOI] [PubMed] [Google Scholar]
  • 91.Amundadottir LT, Sulem P, Gudmundsson J, et al. A common variant associated with prostate cancer in European and African populations. Nat. Genet. 2006;38:652. doi: 10.1038/ng1808. [DOI] [PubMed] [Google Scholar]
  • 92.Freedman ML, Haiman CA, Patterson N, et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc Natl Acad Sci USA. 2006;103:14068. doi: 10.1073/pnas.0605832103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gudmundsson J, Sulem P, Manolescu A, et al. Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24. Nat Genet. 2007;39:631. doi: 10.1038/ng1999. [DOI] [PubMed] [Google Scholar]
  • 94.Haiman CA, Patterson N, Freedman ML, et al. Multiple regions within 8q24 independently affect risk for prostate cancer. Nat Genet. 2007;39:638. doi: 10.1038/ng2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yeager M, Orr N, Hayes RB, et al. Genome-wide association study of prostate cancer identifies a second risk locus at 8q24. Nat Genet. 2007;39:645. doi: 10.1038/ng2022. [DOI] [PubMed] [Google Scholar]
  • 96.Severi G, Hayes VM, Padilla EJ, et al. The common variant rs1447295 on chromosome 8q24 and prostate cancer risk: results from an Australian population-based case-control study. Cancer Epidemiol Biomarkers Prev. 2007;16:610. doi: 10.1158/1055-9965.EPI-06-0872. [DOI] [PubMed] [Google Scholar]
  • 97.Wang L, McDonnell SK, Slusser JP, et al. Two common chromosome 8q24 variants are associated with increased risk for prostate cancer. Cancer Res. 2007;67:2944. doi: 10.1158/0008-5472.CAN-06-3186. [DOI] [PubMed] [Google Scholar]
  • 98.Hughes Halbert C, Armstrong K, Holmes J, et al. Transdisciplinary Approaches to Ameliorating Racial Disparities in Prostate Cancer Outcomes. Journal of Health Disparities Research and Practice. 2006;1:19. [Google Scholar]
  • 99.Klabunde CN, Potosky AL, Harlan LC, et al. Trends and black/white differences in treatment for nonmetastatic prostate cancer. Med Care. 1998;36:1337. doi: 10.1097/00005650-199809000-00006. [DOI] [PubMed] [Google Scholar]
  • 100.Desch CE, Penberthy L, Newschaffer CJ, et al. Factors that determine the treatment for local and regional prostate cancer. Med Care. 1996;4:152. doi: 10.1097/00005650-199602000-00007. [DOI] [PubMed] [Google Scholar]
  • 101.Schapira MM, McAuliffe TL, Nattinger AB. Treatment of localized prostate cancer in African-American compared with Caucasian men. Less use of aggressive therapy for comparable disease. Med Care. 1995;33:1079. doi: 10.1097/00005650-199511000-00002. [DOI] [PubMed] [Google Scholar]
  • 102.Merrill RM, Stephenson RA. Trends in mortality rates in patients with prostate cancer during the era of prostate specific antigen screening. J Urol. 2000;163:503. [PubMed] [Google Scholar]
  • 103.Wong YN, Mitra N, Hudes G, et al. Survival associated with treatment vs observation of localized prostate cancer in elderly men. JAMA. 2006;296:2683. doi: 10.1001/jama.296.22.2683. [DOI] [PubMed] [Google Scholar]
  • 104.Ellison GL, Coker AL, Hebert JR, et al. Psychosocial stress and prostate cancer: a theoretical model. Ethn Dis. 2001;11:484. [PubMed] [Google Scholar]
  • 105.Ferlay J, Bray F, Pisani P, et al. GLOBOCAN 2002: Cancer Incidence, Mortality and Prevalence Worldwide. IARC Press; Lyon: 2002. [Google Scholar]

RESOURCES