Factors Involved in Prostate Cancer Disparity in African Americans: from Health System to Molecular Mechanisms
Article Information
Marina G. Ferrari1, Jimenez-Uribe AP3, Adrian P. Mansini*, 1,2
1Department of Urology, Rush University Medical Center, Chicago, IL, USA.
2Department of Dermatology, Rush University Medical Center, Chicago, IL, USA
3Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
*Corresponding author: Adrian P. Mansini, Department of Urology and Department of Dermatology, Rush University Medical Center, Chicago, IL, USA.
Received: 04 June 2024; Accepted: 11 June 2024; Published: 04 December 2024
Citation: Marina G. Ferrari, Jimenez-Uribe AP, Adrian P. Mansini. Factors Involved in Prostate Cancer Disparity in African Americans: from Health System to Molecular Mechanisms. Fortune Journal of Health Sciences. 7 (2024): 690-700.
View / Download Pdf Share at FacebookAbstract
The high incident and mortality rate in African American patients reflects the racial disparity in prostate cancer (PCa). African American men are affected by several non-biological and biological factors that increase their susceptibility to develop aggressive PCa compared with Caucasian men. Here, we provide a general view of some factors that impact the outcome, focusing on socio-economic factors and with more detail on a molecular perspective covering the mutations, polymorphisms, or epigenetic changes that influence cell proliferation/death balance, androgen pathway and immune response involved in PCa racial disparity. Moreover, we provide an overview of how non-biological and biological factors are interconnected in properly managing diseases.
Keywords
Prostate Cancer, Cancer Disparity, Androgen Receptor
Article Details
Introduction
Prostate cancer (PCa) is the most common cancer diagnosed in men and the fifth leading cause of death worldwide [1]. The incidence among African American (AA) men in the US is 60% higher compared with their Caucasian (CA) counterparts, and the mortality rate is 2.5-fold increased [2, 3]. AA men present at the diagnosis with more aggressive disease, worst prognosis, and worst therapeutic response with a higher risk of recurrence [4-6]. In addition, AA patients exhibit more severe side effects of conventional therapies than their CA counterparts [7]. Although socio-cultural disparities are often associated with PCa outcomes in AA patients, several other biological factors are also involved.
Among the non-biological factors that impact PCa treatment and outcome are socioeconomic status, lack of access to adequate health, physician-patient barrier communication, lifestyle, and environment. Biological factors, including germline polymorphisms, family history, hormonal levels, and molecular alterations, contribute to race-specific prostate cancer development, incidence, and clinical outcome.
Socio-Cultural Factors in Racial Disparities of Prostate Cancer
One of the first barriers involved in the correct prevention, diagnosis, and treatment of PCa, in general, is the lack of knowledge related to genitourinary health and the risk factors involved in the development of PCa, including age, family history of PCa, sexually transmitted disease history, smoking, diet, among others according to American Cancer Society (cancer.org). In addition to this, other sociocultural factors involved in the PCa disparity in AA men include poor interpersonal communication between physicians and patients, including physicians' failure to provide necessary information and racism, patients' distrust in the health care system, and lack of adherence to therapy. Even if the barriers mentioned above are overcome, the socioeconomic status of the patients impact directly in the affordability of the treatment, and consequent outcome; indeed, socioeconomic status in AA patients diagnosed with PCa is directly associated with increased mortality and morbidity [8, 9]. Furthermore, fear of diagnosis and treatment strategies and poor health consciousness contribute to the late diagnosis. These and other factors were reviewed in-depth elsewhere [10]. Socio-cultural and biological factors influence dietary habits; however, the exact molecular mechanisms of how these act remain unclear. In particular, vitamin D deficiency is associated with increased susceptibility to developing prostate cancer in AA men [11]. On the other hand, high calcium intake is associated with aggressiveness of PCa [12].
Molecular Basis of Racial Disparities in Prostate Cancer
Molecular factors such as genetic modifications, including gene polymorphism and mutations, epigenetic alterations, dysregulation of miRNAs, and over-activation of several signaling pathways, are involved in PCa disparities [13].
Genetic Variants
Since PCa is one of the most recognizable inherited malignancies associated with hereditary breast and ovarian cancer (HBOC) and Lynch syndromes (LS), some germline polymorphisms and mutations are involved in the disparity susceptibility to develop PCa in AA men as described below:
Chromosome 8. The deletion of the short arm of chromosome 8 is frequently found in prostatic intraepithelial neoplasia and adenocarcinomas, which participate in prostate cancer carcinogenesis [14, 15]. Although the exact mechanisms by which alterations of 8q24 participate in PCa development are unclear, some polymorphisms may influence the expression of neighbor genes, such as c-MYC [16]. The allelic variant of 8q24 is reported to be associated with the risk of PCa in young AA men [17], and alteration of 8q24 is associated with hereditary PCa in AA men.
MSR1. Macrophage scavenger receptor 1 (MSR1), also known as CD204, is a multifunctional receptor that binds modified self- and pathogen-associated antigens. MSR1 plays an important role in maintaining immunological tolerance [18]. MSR1 gene is associated with germ-line alterations in 8p and prostate carcinogenesis. Several variants of MSR1 are found to be associated with PCa susceptibility in AA men [19]. A specific mutation in the MSR1 gene that results in 520G>T is associated with PCa, and AA men exhibit a high frequency of this mutation [20]. However, there is limited support for an association in AA men.
HPC1. Hereditary prostate cancer 1 is localized at 1q24-31; several studies suggest that HPC1 increases the risk of hereditary PCa development in AA men [21, 22]. Furthermore, in the loci of HPC have been identified several candidates that contribute to susceptibility, such as HPCX at Xq27-28 and RNASEL at 1q25 [23]. While HPCX variants are associated with an increased risk of PCa, variants in RNASEL contribute to susceptibility to the early onset of the hereditary form of PCa [23].
Apoptotic genes: Cell cycle regulation is disrupted in cancer by the imbalance between cell growth and cell death; many genes related to apoptotic functions are altered in PCa as described as follows:
Anti-apoptotic BCL2. BCL2 protein plays a crucial role in cancer development and progression and exhibits an anti-apoptotic effect in cancer cells. BCL2 expression in PCa is associated with increased resistance to therapies. AA men present higher expression of BCL2 compared with their CA counterparts. Thus, the overexpression of BCL2 in AA men may participate in prostate tumor growth and aggressiveness [24]. It was reported that a functional single nucleotide polymorphism (c.-938C>A, rs2279115) in the inhibitory P2 promoter of BCL2 results in altered transcription factor binding and expression [25]. While the BCL2 -938C allele was significantly associated with increased P2 promoter activity, which results in decreased BCL2 transcriptional activity and protein expression [26, 27], the BCL2-938 CC genotype (CA allele +AA allele) is associated with an increased risk of biochemical recurrence [28]. In 2014, Renner et al., showed a strong association between the BCL2-938 CC genotype and reduced survival in PCa patients [29]. However, the study by Bachmann et al., reported an association of the BCL2-938 AA allele with a worse outcome in PCa patients [26]. The C allele is the most common in AA populations. The cause of this discrepancy is unclear.
MDM2. This ubiquitin ligase protein is involved in several functions, such as promoting the degradation of the tumor suppressor p53, DNA repair, and apoptosis [30, 31]. MDM2 is significantly highly expressed in CA patients compared with AA, and the overexpression in PCa is associated with cancer progression in a mechanism dependent on p53 degradation [32]. However, the expression in AA patients is associated with poor prognosis. Bond et al., reported a single nucleotide polymorphism (SNP309), which enhances transcriptional activation of MDM2 downregulating p53 pathway [33]. In addition, Wang et al., reported no difference in the expression level of SNP309 between AA and CA patients [34]. Still, interestingly, in a meta-analysis performed by Yang et al., they showed that the MDM2 309G variant was markedly associated with a low risk of developing PCa and slower clinical progression in CA men [35]. Thus, some controversies need to be studied more deeply.
Caveolin-1. This structural protein is involved in tumor progression and metastasis and was reported to participate actively in radio-resistance acquisition [36]. Since caveolin-1 is significantly upregulated in PCa cells and mediates downstream signaling mechanisms related to the development of aggressive PCa, caveolin-1 was proposed as a prognostic biomarker to monitor tumor radioresistance [36]. Furthermore, caveolin-1 is involved in the suppression of apoptosis by suppressing c-MYC. In addition, Caveolin-1 was found overexpressed in prostate adenocarcinoma cells and is implicated in the progression of androgen-dependent PCa to androgen-independent [37]. It is known that a high level of caveolin-1 is associated with poor treatment outcomes [36]. AA men diagnosed with PCa exhibit a significant increment in the level of caveolin-1 compared with their CA counterparts [38]. Although caveolin-1 polymorphism rs7804372 is associated with the risk of several types of cancer (10.4236/ym.2020.43020), there is no current information on AA risk to PCa with this polymorphism.
Growth factors and receptors: As a counterpart to balance cell death, cell survival, and proliferation are driven by several growth factors. Epidermal growth factor and receptor (EGFR) and EPH receptor (EPHB2) are the most common receptors involved in the racial disparity of PCa. AA men exhibit a higher level of IGF-1 and lower levels of IGFB-3 that may participate in tumor growth and progression. IGF and ligands are effectors of AKT signaling, activating PCa development, metastasis, and anti-apoptosis [39].
EGFR. The EGFR signaling pathway is one of the most critical pathways to PCa development. This pathway is implicated in the progression of PCa from androgen-dependent to androgen-independent and is associated with metastasis [40]. Several studies have identified racial differences in dinucleotide (CA)n repeat polymorphisms in intron 1 of the gene [41]. The number of CA repeats is related to transcriptional activity. Thus, the longer allele is associated with reduced protein expression. In contrast, shorter alleles, which are more frequent in AA men, are associated with overexpression and PCa development [42].
EPHB2. The gene encodes a tyrosine kinase receptor and is a tumor suppressor gene. Somatic inactivating mutations occur in approximately 10% of sporadic tumors. The nonsense mutation K1019X (3055A-T) is more frequently found in AA men and is associated with an increased risk of PCa development. [43].
Androgens pathway. Several studies have shown that sex steroid hormone levels are also implicated in the racial disparities in PCa. These studies have shown that AA men exhibit approximately 11-15% higher levels of testosterone, 13% higher free testosterone, and higher activity of 5-alpha reductase than CA men [44-46]. Although there is no established relationship between the level of circulating androgens and PCa, the level of androgens is considered a risk factor. Furthermore, mutation or alteration in several genes involved in androgen biosynthesis may contribute to the racial disparity.
Androgen receptors. Androgens bind androgen receptor (AR), inducing several androgen-regulated genes required for prostate cell growth and maintenance. AR is a transcription factor, a gene mapped in Xq11-12 and composed of 8 exons. Exon 1 encodes the N-terminal domain, which is a transactivation domain. This domain controls the transcriptional activation of the receptor and exhibits several highly repetitive DNA sequences. Polymorphic trinucleotide repeats (CAG) are associated with PCa disparity. One study performed in 587 cases of PCa compared with 588 controls, which involved 95% of CA patients, showed that shorter CAG repeat sequence was associated with metastasis and high grade of the disease [47]. In addition, Do et al., analyzed CAG repeat length in 109 cases of PCa and found that the median CAG repeat length was 25 in patients with early status of PCa, while in advanced status, the CAG repeat length was present between 22-23. Further, they found a significant correlation between these CAG repeats and the age at onset of the disease, suggesting that these repeats may be associated with an increased risk of developing PCa [48]. On the other hand, Sartor et al., analyzed the presence of these repeats in 130 CA and 65 AA average men, finding that AA men exhibit significantly shorter repeats than their CA counterparts [49]. Thus, shorter CAG repeats may explain the occurrence at a younger age and the rapid progression of PCa in AA men.
Genes involved in androgen biosynthesis. Polymorphism in genes involved in androgen biosynthesis and metabolism may modify the expression of androgen levels, contributing to racial disparity. Some of these polymorphism genes are the following:
CYP17. The CYP17 gene is located on chromosome 10 and encodes the cytochrome P450c17a enzyme, which participates in the steroid biosynthesis pathway [50]. The polymorphic T-to-C substitution in the 5’ promoter region results in A1(T) and A2(C) alleles [51]. One study conducted by Wadelius et al., evaluated the association between the polymorphism in the CYP17 gene and prostate cancer in 178 CA patients diagnosed with PCa using as control 160 age-matched control individuals. After evaluating the presence of the polymorphism in blood samples, they reported the frequency of the CYP17 A1 allele was significantly higher in prostate cancer patients [52]. Supporting the results as mentioned earlier, Habuchi et al., published a study performed in a Japanese population that included 252 prostate cancer patients, 202 benign prostatic hyperplasia (BPH) patients, and 131 male controls, which found that the A1 allele is associated with an increased risk of both prostate cancer and BPH, but not influence in the status of the disease [53]. Despite several studies reporting that the A1 allele is associated with an increased risk of PCa in CA and Japanese patients [52, 53], other studies show the opposite. Lunn et al., performed a study in 108 CA prostate cancer cases where they found that the A2 allele (genotype A1/A2 and A2/A2) was the most frequent in CA PCa patients compared with control urology patients [54]. Contrary to the report in CA, Ntais et al., by performing a meta-analysis of 10 studies that included 2404 patients with prostate cancer and 2755 controls, found that while the allele A2 seems not to affect the predisposition to develop PCa in CA people, in AA men the presence of A2 allele is associated with PCa development [55]. In addition, and supporting this finding, Kittles et al., by analyzing the presence of the A2 allele (genotype A1/A2 and A2/A2) in Nigerian (n=56), CA (n=74), and A-A (n=111) healthy male as a control found that AA men homozygous for the allele A2 present a higher risk to develop PCa [56]. Thus, the association is not conclusive.
CYP3A4. A germinal variant (A to G) in the 5’ regulatory region of the gene determines the variant CYP3A4-V. One study conducted by Rebbeck et al., on 230 Caucasians shows that CYP3A4-V is associated with the development and aggressiveness of PCa in CA but not in AA men [57]. In AA men, the variant CYP3A4 G is more frequent and is associated with poor prognosis [58].
CYP19A1. The CYP19A1 gene is mapped in 15q21.1 and encodes the enzyme aromatase. At least 30 SNPs have been identified and associated with a high estradiol level in men's serum. The polymorphisms rs2470152, rs12439137, rs3751592, and rs2470164 are associated with a high risk of PCa in both AA and CA. [59]. However, CA men bearing rs2470164 polymorphism present a higher risk.
SRD5A2. This gene encodes steroid 5-α reductase type 2, which converts testosterone to DHT and is exclusively expressed in the prostate [60]. SRD5A is highly polymorphic in AA men, and SRD5A2 TA repeats are exclusively present in high-risk AA men. Other variants, such as V89L and A49T, are involved in converting DHT from testosterone; the first variant decreases the production, while the second increases the production in AA men [61].
The HSD3B family, HSD3B1 and HSD3B2, encode 3β-hydroxysteroid dehydrogenase type 1 and 2, respectively. A notable variation that frequently occurs in prostate cancer is the N367T (rs10473003) polymorphism in HSD3B1. While this variation is more prevalent in CA than in AA, it does not significantly modify its activity compared with the wild type, underscoring its importance in prostate cancer genetics [62]. The presence of (TG)n(TA)n(CA)n dinucleotide repeats in the intron 3 of HSD3B2 gene presence variation between racial populations, the longer sequences are associated with faster degradation of DHT. In contrast, the shorter alleles are associated with PCa risk and aggressiveness in CA men [63]. Two SNPs were reported in HSD3B1 (rs1819689 and rs1538989), which are more frequent in AA men and are associated with PCa in that racial population [59].
miRNAs. MicroRNAs are endogenous non-coding RNA that regulate gene expression. The correct regulation of the expression at transcriptional and post-transcriptional levels by miRNAs is required for the basic cellular process. These small sequences of oligonucleotides could function as a tumor suppressor or oncogenes according to their expression level [64, 65]. Since the miRNAs are specific to the type of cancer, tumor grade, and level of metastasis, miRNAs have the potential to be used as a stage-specific biomarker of cancer [66]. Some miRNAs such as (miR-21, miR-17-5P, miR-191, miR-29-b2, miR-223, miR-199-a1, miR-146, miR-181-b1, miR-20a, miR-32, miR-92-2) are up-regulated in PCa but also are up-regulated in other solid tumor. Thus, these miRNAs are not specific but belong to the signatures of some solid cancers [67]. Recently, Sharma et al., published a review identifying a panel of miRNAs differentially expressed in PCa. These miRNAs include miR-141, miR-375, miR-221, and miR-21 and are the most common dysregulated miRNAs in prostate cancer independent of the racial disparity [66]. The role of these miRNAs in PCa is shown in Table 1. Calin et al., reported at least five miRNAs that are differentially expressed between AA and CA patients diagnosed with PCa ( miR-26a, miR-30c-1, miR-1b-1, miR-219, and miR-301) [68]. In addition, Ren et al., working with a cohort of 27 cases of radical prostatectomy PCa samples, reported that the expression of miR-30c and miR-219 were downregulated in PCa, but miR-21 and miR-30c were significantly downregulated in PCa in AA cases compared with CA cases. They also found the downregulation of let-7c in PCa stroma cells was significantly associated with metastasis [69]. However, it is necessary to note the limited number of cases analyzed in the study. Another group using a combined platform intergraded by cancer cell lines, transgenic mice, and human tissue samples has shown that loss of miR-34b expression occurs in AA patients more frequently than in CA, and the loss of the expression is associated with PCa progression in a SOX2 dependent mechanism [70]. Working with the AA and CA cancer cell lines model, Theodore found that mir-26a is significantly expressed in AA compared with CA [71].
Table 1: Common dysregulated miRNAs in prostate cancer
Epigenetic Changes
Epigenetic processes regulate gene expression, a reversible process that, contrary to mutation, consists of DNA methylation, modification of histones, chemical modification, and chromatin remodeling that result in changes in gene expression without modification of the DNA sequences—epigenetic process blockage of the access of transcriptional factors to the target genes promoter. Thus, genes are not able to be transcribed. Since the epigenetic changes are reversible, they allow the development of new opportunities for therapies to recover the initial condition. Several genes exhibit aberrant methylation in PCa. Among these genes are found GSTP1, MGMT, CDH1, CD44, CDKN2A, APC, RARβ, RARRES1, and RASSF1. GSTP1 gene is localized in chromosome 11q13 and encodes glutathione S-transferase. Hypermethylation of GSTP1 seems to be exclusive of PCa cells and is present at all stages of PCa. MGMT is localized in chromosome 10q23 and encodes DNA methyltransferase enzyme. Its methylation was reported in PCa patients and cell lines and is associated with carcinogenesis [72]. Since both enzymes are involved in cell detoxification and DNA repair, they may be involved in the genomic instability of the cells. CDH1 gene is another gene hypermethylated in PCa, which encodes E-cadherin protein that participates in cell-cell adhesion. Hypermethylation of CDH1 results in loss of E-cadherin expression, which is associated with metastasis development [73, 74]. CD44 hypermethylation is found in 78% of patients diagnosed with PCa, a characteristic feature of epithelial-mesenchymal transition [74, 75]. CCND2 promoter hypermethylation is found in 32% of PCa, and the high methylation level of the gene is correlated with tumor aggressiveness [75, 76]. APC protein encoded by the APC gene participates in cell cycling regulation, migration, and differentiation. The silencing of the APC protein product of promoter methylation is associated with high-grade and advanced stages of PCa [77]. Other genes were reported to be hypermethylated in PCa, such as the retinoic acid receptor (RARβ), RARRES1, and RASSF1, in which proteins are found in low levels and are involved in PCa development [78-80]. On the other hand, hypomethylation is associated with cancer metastasis and is frequently found in repetitive sequences of LINE-1 in PCa metastasis [81, 82]. Higher expression of urokinase plasminogen activator, heparanase, cytochrome p450s, WNT5A, S100P, and cysteine-rich intestinal protein 1 due to promoter hypomethylation was reported in PCa [83, 84]. Furthermore, aberrant histone methylation is found in PCa patients. Chervona et al., found reduced levels of H3K4me3 and H3K18Ac in PCa, which were associated with relapse and negative prognosis [85].
Fusion Gene in Racial Prostate Cancer Disparity
Gene rearrangement involving the androgen-regulated gene transmembrane protease serine 2 (TMPRSS2) and erythroblastosis virus E26 oncogene homolog (ERG) is the most common fusion gene in PCa. Its presence is found in approximately 50% of the patients diagnosed with PCa. However, this aberration is more frequently found in CA than in AA men, whose frequency is 31% [86]. Although ERG alteration is more commonly found in CA patients, AA patients with no ERG alteration exhibit higher-grade index tumors [87]. TMPRSS2-ERG fusion gene is associated with an aggressive tumor and is seen to be generated by gamma-irradiation-induced DNA double-strand breaks [88]. In addition, aberrant androgen receptor signaling may induce chromosome aberration.
Immune System in Prostate Cancer Disparity
The tumor microenvironment is constituted by various soluble factors such as cytokines, interleukins, proteins, several immune cells such as natural killer (NK), lymphocytes, macrophages, and non-immune cells such as fibroblast and endothelial cells. The fact that AA men develop more frequently PCa, and more aggressive diseases compared with CA men suggests that an inappropriate tumor microenvironment may contribute to the increased disparity. A study conducted by Eastham et al., found that AA specimens exhibit higher inflammation compared with CA specimens in prostate biopsy specimens obtained from patients diagnosed with PCa [89]. In another study, Wallace et al., using microarray technology, evaluated the gene expression profile in PCa primary tumors obtained from a cohort of 69 patients (AA=33 and CA=36). They found that autocrine mobility factor receptor (AMFR), chemokine receptor 4 (CXCR4), and matrix metalloprotease 9 (MMP9) were the most differentially expressed genes [90]. In 2014, Kinseth et al., in a study integrated by 17 pairs of arrays for AA and CA, found a significant difference in the gene expression between both populations. Interestingly, most genes differentially expressed were associated with tumor-adjacent stroma, not tumor tissue [91]. Among the genes differentially expressed and associated with tumor tissue, genes involved in immune-related pathways were overexpressed [91], while from the genes differentially expressed in PCa stroma tissue, approximately 20% of them were involved in immune response, including cytokines such as TGF-β and IL-10 [91]. Interestingly, these cytokines are involved in immune suppression and PCa progression.
Another protein that seems to be involved in immune racial disparity is the well-known transmembrane protein MHC class I polypeptide-related sequence A (MICA). This protein participates in the surveillance and antitumor immunity by interacting with NK cells, cytotoxic T, and other T cell subsets expressing NKG2D receptor [92, 93]. Even though the expression of MICA allows immune cells to recognize tumors, some tumor cells exhibit an evasion mechanism by which MICA is cleaved from the membrane of the cancer. This soluble form (sMICA) binds the NKGD2 receptor, which is internalized. Thus, sMICA impairs the activation of immune cells [94, 95]. In 2004, Wu et al., reported a positive correlation between the level of sMICA and deficiency in NK function in patients with advanced PCa. Moreover, the group proposes sMICA as a novel biomarker for prostate cancer [96]. Recently, Sakiyama et al., in a cohort composed of a total of 52 patients diagnosed with PCa, including AA and CA men, found that prostate tumor tissues express a higher level of MICA compared with normal tissues. Furthermore, the MICA expression at molecular and protein levels was lower in AA patients compared with CA, and low levels of MICA were associated with poor prognosis [97]. Interestingly, sMICA was found in prostate cancer cell lines representative of CA but not in cell lines representative of AA. Thus, MICA expression participates in racial disparity. Another entity suggested to participate in immune race disparity in PCa is benign ethnic neutropenia, which is very frequent in AA [98, 99]. In 2012, Sadeghi et al., reported that neutropenia is an independent risk factor for developing poorly differentiated PCa among AA men [100]. However, more studies need to be performed to confirm the strength of this association.
Conclusion
Several factors are involved in the racial disparities observed between AA and CA men. Some increase the susceptibility to developing PCa, and others are related to aggressiveness and capacity to induce metastasis, resulting in the worst outcome. Among these factors, we can find mutations and gene variations such as polymorphisms, alteration of miRNA expression, aberrant activation of signaling pathways, epigenetic changes, and altered protein expression. Although for therapy use or the development of new ones, it is essential to know the molecular mechanisms implicated in the pathophysiology of PCa, some fundamental limitations still exist in the diagnosis and successful treatment. Due to the molecular complexity of PCa and the difficulties of therapies in targeting genetic factors such as mutations or polymorphisms, primary therapies are focused on surgical removal of the prostate combined with radiotherapy or chemotherapy to limit cancer cell proliferation. However, one of the best approaches that lead to successful therapies for cancer in general is preventive medicine since this allows for early diagnosis.
Preventive medicine encompasses educational strategies to draw attention to risk factors such as ancestry-related predisposition and environment. Recently, Garraway IP et al., [101] published a guideline for early screening to detect PCa in AA men, containing key points such as screening for blood PSA in the early 40s and family history. Furthermore, other strategies in preventive medicine include education related to dietary habits, smoking, and exposure to environmental pollutants. In conclusion, the combination of managing non-biological and biological factors involved in PCa is essential to eliminate racial disparities in diagnosis and treatment in AA men.
No Conflict Statement
All authors have participated in writing and have approved this version. The authors declare no potential conflicts of interest.
Financial Declaration
Swim Across America funding, awarded to Dr. Mansini, supported this study.
References
- Rawla P, Epidemiology of Prostate Cancer. World J Oncol 10 (2019): 63-89.
- Powell IJ, Epidemiology and pathophysiology of prostate cancer in African-American men. J Urol 177 (2007): 444-9.
- Bedolla RG, et al, Receptor tyrosine kinase recepteur d'origine nantais as predictive marker for aggressive prostate cancer in African Americans. Mol Carcinog 58 (2019): 854-861.
- McGinley, K.F, K.J. Tay, and J.W. Moul, Prostate cancer in men of African origin. Nat Rev Urol 13 (2016): 99-107.
- Thorpe RJ Jr, et al, Health behaviors and all-cause mortality in African American men. Am J Mens Health 7 (2013): 8s-18s.
- Sundi D, et al, African American men with very low-risk prostate cancer exhibit adverse oncologic outcomes after radical prostatectomy: should active surveillance still be an option for them? J Clin Oncol 31 (2013): 2991-7.
- Kerns SL, et al, Genome-wide association study to identify single nucleotide polymorphisms (SNPs) associated with the development of erectile dysfunction in African-American men after radiotherapy for prostate cancer. Int J Radiat Oncol Biol Phys 78 (2010): 1292-300.
- Chhatre, S, et al, Understanding the Racial and Ethnic Differences in Cost and Mortality Among Advanced Stage Prostate Cancer Patients (STROBE). Medicine (Baltimore) 94 (2015): e1353.
- Hoffman RM, et al, Racial and ethnic differences in advanced-stage prostate cancer: the Prostate Cancer Outcomes Study. J Natl Cancer Inst 93 (2001): 388-95.
- Lillard JW Jr, et al, Racial disparities in Black men with prostate cancer: A literature review. Cancer 128 (2022): 3787-3795.
- Batai K, et al, Vitamin D and Immune Response: Implications for Prostate Cancer in African Americans. Front Immunol 7 (2016): 53.
- Batai K, et al, Race and BMI modify associations of calcium and vitamin D intake with prostate cancer. BMC Cancer 17 (2017): 64.
- Bhardwaj A, et al, Racial disparities in prostate cancer: a molecular perspective. Front Biosci (Landmark Ed) 22 (2017): 772-782.
- Barry KH, et al, Prospective study of DNA methylation at chromosome 8q24 in peripheral blood and prostate cancer risk. Br J Cancer 116 (2017): 1470-1479.
- Trock BJ, et al. PTEN loss and chromosome 8 alterations in Gleason grade 3 prostate cancer cores predicts the presence of un-sampled grade 4 tumor: implications for active surveillance. Mod Pathol 29 (2016): 764-71.
- Ahmadiyeh N, et al. 8q24 prostate, breast, and colon cancer risk loci show tissue-specific long-range interaction with MYC. Proc Natl Acad Sci U S A 107 (2010): 9742-6.
- Freedman ML, et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc Natl Acad Sci U S A 103 (2006): 14068-73.
- Haasken S, et al. Macrophage scavenger receptor 1 (Msr1, SR-A) influences B cell autoimmunity by regulating soluble autoantigen concentration. J Immunol 191 (2013): 1055-62.
- Miller DC, et al. Germ-line mutations of the macrophage scavenger receptor 1 gene: association with prostate cancer risk in African-American men. Cancer Res 63 (2003): 3486-9.
- Singh SK, JW, Lillard Jr, and R Singh, Molecular basis for prostate cancer racial disparities. Front Biosci (Landmark Ed) 22 (2017): 428-450.
- Smith JR, et al. Major susceptibility locus for prostate cancer on chromosome 1 suggested by a genome-wide search. Science 274 (1996): 1371-4.
- Gronberg H, et al. Early age at diagnosis in families providing evidence of linkage to the hereditary prostate cancer locus (HPC1) on chromosome 1. Cancer Res 57 (1997): 4707-9.
- Agalliu I, et al. Contribution of HPC1 (RNASEL) and HPCX variants to prostate cancer in a founder population. Prostate 70 (2010): 1716-27.
- deVere White RW, et al Racial differences in clinically localized prostate cancers of black and white men. J Urol 159 (1998): 1979-82.
- El Hindy N, et al Association of the CC genotype of the regulatory BCL2 promoter polymorphism (-938C>A) with better 2-year survival in patients with glioblastoma multiforme. J Neurosurg 114 (2011): 1631-9.
- Bachmann HS, et al. Regulatory BCL2 promoter polymorphism (-938C>A) is associated with adverse outcome in patients with prostate carcinoma. Int J Cancer 129 (2011): 2390-9.
- Nuckel H, et al. Association of a novel regulatory polymorphism (-938C>A) in the BCL2 gene promoter with disease progression and survival in chronic lymphocytic leukemia. Blood 109 (2007): 290-7.
- Hirata H, et al. Bcl2 -938C/A polymorphism carries increased risk of biochemical recurrence after radical prostatectomy. J Urol 181 (2009): 1907-12.
- Renner W, et al. BCL2 genotypes and prostate cancer survival. Strahlenther Onkol 193 (2017): 466-471.
- Shi D and W Gu. Dual Roles of MDM2 in the Regulation of p53: Ubiquitination Dependent and Ubiquitination Independent Mechanisms of MDM2 Repression of p53 Activity. Genes Cancer 3 (2012): 240-8.
- Zhao Y, H Yu and W Hu. The regulation of MDM2 oncogene and its impact on human cancers. Acta Biochim Biophys Sin (Shanghai) 46 (2014): 180-9.
- Osman I, et al. Inactivation of the p53 pathway in prostate cancer: impact on tumor progression. Clin Cancer Res 5 (1999): 2082-8.
- Bond GL, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 119 (2004): 591-602.
- Wang G, et al. MDM2 expression and regulation in prostate cancer racial disparity. Int J Clin Exp Pathol 2 (2009): 353-60.
- Yang J, et al, The risks, degree of malignancy and clinical progression of prostate cancer associated with the MDM2 T309G polymorphism: a meta-analysis. Asian J Androl 14 (2012): 726-31.
- Mahmood, J, et al, Caveolin-1: a novel prognostic biomarker of radioresistance in cancer. Int J Radiat Biol 92 (2016): 747-753.
- Freeman MR, W Yang and D Di Vizio. Caveolin-1 and prostate cancer progression. Adv Exp Med Biol 729 (2012): 95-110.
- Yang G, et al. Elevated caveolin-1 levels in African-American versus white-American prostate cancer. Clin Cancer Res 6 (2000): 3430-3.
- da Silva HB et al, Dissecting Major Signaling Pathways throughout the Development of Prostate Cancer. Prostate Cancer (2013): 920612.
- Munoz-Moreno L, et al. Growth hormone-releasing hormone (GHRH) promotes metastatic phenotypes through EGFR/HER2 transactivation in prostate cancer cells. Mol Cell Endocrinol 446 (2017): 59-69.
- Liu W, et al. Interethnic difference in the allelic distribution of human epidermal growth factor receptor intron 1 polymorphism. Clin Cancer Res 9 (2003): 1009-12.
- Shuch B, et al, Racial disparity of epidermal growth factor receptor expression in prostate cancer. J Clin Oncol 22 (2004): 4725-9.
- Kittles RA, et al. A common nonsense mutation in EphB2 is associated with prostate cancer risk in African American men with a positive family history. J Med Genet 43 (2006): 507-11.
- Ross, R, et al, Serum testosterone levels in healthy young black and white men. J Natl Cancer Inst 76 (1986): 45-8.
- Kheirandish P and F Chinegwundoh, Ethnic differences in prostate cancer. Br J Cancer 105 (2011): 481-5.
- Ross, R.K, et al, 5-alpha-reductase activity and risk of prostate cancer among Japanese and US white and black males. Lancet 339 (1992): 887-9.
- Giovannucci, E, et al, The CAG repeat within the androgen receptor gene and its relationship to prostate cancer. Proc Natl Acad Sci U S A 94 (1997): 3320-3.
- Hardy DO, et al, Androgen receptor CAG repeat lengths in prostate cancer: correlation with age of onset. J Clin Endocrinol Metab 81 (1996): 4400-5.
- Sartor, O, Q. Zheng, and J.A. Eastham, Androgen receptor gene CAG repeat length varies in a race-specific fashion in men without prostate cancer. Urology 53 (1999): 378-80.
- Picado-Leonard, J. and W.L. Miller, Cloning and sequence of the human gene for P450c17 (steroid 17 alpha-hydroxylase/17,20 lyase): similarity with the gene for P450c21. DNA 6 (1987): 439-48.
- Carey, A.H, et al, Polycystic ovaries and premature male pattern baldness are associated with one allele of the steroid metabolism gene CYP17. Hum Mol Genet 3 (1994): 1873-6.
- Wadelius M, et al. Prostate cancer associated with CYP17 genotype. Pharmacogenetics 9 (1999): 635-9.
- Habuchi, T, et al, Increased risk of prostate cancer and benign prostatic hyperplasia associated with a CYP17 gene polymorphism with a gene dosage effect. Cancer Res 60 (2000).
- Lunn RM, et al. Prostate cancer risk and polymorphism in 17 hydroxylase (CYP17) and steroid reductase (SRD5A2). Carcinogenesis 20 (1999): 1727-31.
- Ntais CA, Polycarpou and JP. Ioannidis, Association of the CYP17 gene polymorphism with the risk of prostate cancer: a meta-analysis. Cancer Epidemiol Biomarkers Prev 12 (2003): 120-6.
- Kittles, R.A, et al. Cyp17 promoter variant associated with prostate cancer aggressiveness in African Americans. Cancer Epidemiol Biomarkers Prev 10 (2001): 943-7.
- Rebbeck TR, et al. Modification of clinical presentation of prostate tumors by a novel genetic variant in CYP3A4. J Natl Cancer Inst 90 (1998): 1225-9.
- Powell IJ, et al. CYP3A4 genetic variant and disease-free survival among white and black men after radical prostatectomy. J Urol 172 (2004): 1848-52.
- Huang YC et al. CYP19 TCT tri-nucleotide Del/Del genotype is a susceptibility marker for prostate cancer in a Taiwanese population. Urology 69 (2007): 996-1000.
- Thigpen AE, et al. Tissue distribution and ontogeny of steroid 5 alpha-reductase isozyme expression. J Clin Invest 92 (1993): 903-10.
- Makridakis NM, et al. Association of mis-sense substitution in SRD5A2 gene with prostate cancer in African-American and Hispanic men in Los Angeles, USA. Lancet 354 (1999): 975-8.
- Wang L, et al. Human 3beta-hydroxysteroid dehydrogenase types 1 and 2: Gene sequence variation and functional genomics. J Steroid Biochem Mol Biol 107 (2007): 88-99.
- Neslund-Dudas C, et al. SRD5A2 and HSD3B2 polymorphisms are associated with prostate cancer risk and aggressiveness. Prostate 67 (2007): 1654-63.
- Ozen M, et al. Overexpression of miR-145-5p inhibits proliferation of prostate cancer cells and reduces SOX2 expression. Cancer Invest 33 (2015): 251-8.
- Karatas, O.F, et al, miR-33a is a tumor suppressor microRNA that is decreased in prostate cancer. Oncotarget 8 (2017): 60243-60256.
- Sharma N. and M.M. Baruah, The microRNA signatures: aberrantly expressed miRNAs in prostate cancer. Clin Transl Oncol 21 (2019): 126-144.
- Volinia S, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 103 (2006) 2257-61.
- Calin GA and CM Croce. MicroRNA signatures in human cancers. Nat Rev Cancer 6 (2006): 857-66.
- Ren Q, et al. Epithelial and stromal expression of miRNAs during prostate cancer progression. Am J Transl Res, 2014. 6(4): p. 329-39.
- Forno, I, et al, Deregulation of MiR-34b/Sox2 Predicts Prostate Cancer Progression. PLoS One 10 (2015): e0130060.
- Theodore, S.C, et al, MiRNA 26a expression in a novel panel of African American prostate cancer cell lines. Ethn Dis 21 (2010): S1-96-100.
- Nowacka-Zawisza M and E Wisnik. DNA methylation and histone modifications as epigenetic regulation in prostate cancer (Review). Oncol Rep 38 (2017): 2587-2596.
- Keil KP, et al. DNA methylation of E-cadherin is a priming mechanism for prostate development. Dev Biol, 387 (2014): 142-53.
- Kito H, et al Hypermethylation of the CD44 gene is associated with progression and metastasis of human prostate cancer. Prostate 49 (2001): 110-5.
- Woodson, K, et al, Hypermethylation of GSTP1, CD44, and E-cadherin genes in prostate cancer among US Blacks and Whites. Prostate 55 (2003): 199-205.
- et al, Hypermethylation of Cyclin D2 is associated with loss of mRNA expression and tumor development in prostate cancer. J Mol Med (Berl) 84 (2006): 911-8.
- Delgado-Cruzata, L, et al, DNA methylation changes correlate with Gleason score and tumor stage in prostate cancer. DNA Cell Biol 31 (2012): 187-92.
- Jeronimo, C, et al, A quantitative promoter methylation profile of prostate cancer. Clin Cancer Res 10 (2004): 8472-8.
- Youssef EM, et al. Hypermethylation and silencing of the putative tumor suppressor Tazarotene-induced gene 1 in human cancers. Cancer Res 64 (2004): 2411-7.
- Zhang JL. Liu and GP. Pfeifer, Methylation of the retinoid response gene TIG1 in prostate cancer correlates with methylation of the retinoic acid receptor beta gene. Oncogene 23 (2004): 2241-9.
- Florl AR, et al. Coordinate hypermethylation at specific genes in prostate carcinoma precedes LINE-1 hypomethylation. Br J Cancer 91 (2004): 985-94.
- Schulz WA, et al. Genomewide DNA hypomethylation is associated with alterations on chromosome 8 in prostate carcinoma. Genes Chromosomes Cancer 35 (2002): 58-65.
- Ogishima T, et al. Increased heparanase expression is caused by promoter hypomethylation and up-regulation of transcriptional factor early growth response-1 in human prostate cancer. Clin Cancer Res 11 (2005): 1028-36.
- Tokizane, T, et al, Cytochrome P450 1B1 is overexpressed and regulated by hypomethylation in prostate cancer. Clin Cancer Res, 2005. 11(16): p. 5793-801.
- Chervona, Y. and M. Costa, Histone modifications and cancer: biomarkers of prognosis? Am J Cancer Res, 2012. 2(5): p. 589-97.
- Magi-Galluzzi C, et al. TMPRSS2-ERG gene fusion prevalence and class are significantly different in prostate cancer of Caucasian, African-American and Japanese patients. Prostate 71 (2011): 489-97.
- Rosen P, et al. Differences in frequency of ERG oncoprotein expression between index tumors of Caucasian and African American patients with prostate cancer. Urology 80 (2012): 749-53.
- Mani RS, et al. Induced chromosomal proximity and gene fusions in prostate cancer. Science 326 (2009): 1230.
- Eastham JA, et al. Clinical characteristics and biopsy specimen features in African-American and white men without prostate cancer. J Natl Cancer Inst 90 (1998): 756-60.
- Wallace TA, et al. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res 68 (2008): 927-36.
- Kinseth MA, et al. Expression differences between African American and Caucasian prostate cancer tissue reveals that stroma is the site of aggressive changes. Int J Cancer 134 (2014): 81-91.
- Lopez-Soto A, et al. NKG2D signaling in cancer immunosurveillance. Int J Cancer 136 (2015): 1741-50.
- Blery M and E Vivier. NKG2D-MICA Interaction: A Paradigm Shift in Innate Recognition. J Immunol 200 (2018): 2229-2230.
- Chitadze G, et al. Shedding of endogenous MHC class I-related chain molecules A and B from different human tumor entities: heterogeneous involvement of the "a disintegrin and metalloproteases" 10 and 17. Int J Cancer 133 (2013): 1557-66.
- Maurer S, et al. Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. Oncoimmunology 7 (2018): e1364827.
- Wu JD, et al. Prevalent expression of the immunostimulatory MHC class I chain-related molecule is counteracted by shedding in prostate cancer. J Clin Invest 114 (2004): 560-8.
- Sakiyama MJ, et al. Race-associated expression of MHC class I polypeptide-related sequence A (MICA) in prostate cancer. Exp Mol Pathol 108 (2019): 173-182.
- Shoenfeld Y, et al. Benign familial leukopenia and neutropenia in different ethnic groups. Eur J Haematol 41 (1988): 273-7.
- Hsieh MM, et al. Neutrophil count in African Americans: lowering the target cutoff to initiate or resume chemotherapy? J Clin Oncol 28 (2010): 1633-7.
- Sadeghi N, et al. Does absolute neutrophil count predict high tumor grade in African-American men with prostate cancer? Prostate 72 (2012): 386-91.
- Garraway IP, et al. Prostate Cancer Foundation Screening Guidelines for Black Men in the United States. NEJM Evidence 3 (2024): EVIDoa2300289.
- Waltering KK, et al Androgen regulation of micro-RNAs in prostate cancer. Prostate 71 (2011): 604-14.
- Sun T, et al. The role of microRNA-221 and microRNA-222 in androgen-independent prostate cancer cell lines. Cancer Res 69 (2009): 3356-63.
- Li JZ, et al. MiR-141-3p promotes prostate cancer cell proliferation through inhibiting kruppel-like factor-9 expression. Biochem Biophys Res Commun 482 (2017): 1381-1386.
- Liu C, et al. MicroRNA-141 suppresses prostate cancer stem cells and metastasis by targeting a cohort of pro-metastasis genes. Nat Commun 8 (2017): 14270.
- Chu M, et al. Androgen receptor is negatively correlated with the methylation-mediated transcriptional repression of miR-375 in human prostate cancer cells. Oncol Rep 31 (2014): 34-40.
- Pickl JM, et al. Ago-RIP-Seq identifies Polycomb repressive complex I member CBX7 as a major target of miR-375 in prostate cancer progression. Oncotarget 7 (2016): 59589-59603.
- Selth LA, et al. A ZEB1-miR-375-YAP1 pathway regulates epithelial plasticity in prostate cancer. Oncogene 36 (2017): 24-34.
- Zheng CS, Yinghao and J Li. MiR-221 expression affects invasion potential of human prostate carcinoma cell lines by targeting DVL2. Med Oncol 29 (2012): 815-22.
- Hu CD, R Choo and J Huang. Neuroendocrine differentiation in prostate cancer: a mechanism of radioresistance and treatment failure. Front Oncol 5 (2015): 90.
- Yang X, et al. Down-regulation of mir-221 and mir-222 restrain prostate cancer cell proliferation and migration that is partly mediated by activation of SIRT1. PLoS One 9 (2014): e98833.
- Kneitz B, et al. Survival in patients with high-risk prostate cancer is predicted by miR-221, which regulates proliferation, apoptosis, and invasion of prostate cancer cells by inhibiting IRF2 and SOCS3. Cancer Res 74 (2014): 2591-603.
- Wang L, et al. Effects of microRNA-221/222 on cell proliferation and apoptosis in prostate cancer cells. Gene 572 (2015): 252-8.
- Ribas J and SE. Lupold, The transcriptional regulation of miR-21, its multiple transcripts, and their implication in prostate cancer. Cell Cycle 9 (2010): 923-9.
- Coppola V, et al. BTG2 loss and miR-21 upregulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial-mesenchymal transition. Oncogene 32 (2013): 1843-53.
- Yang Y, JX Guo and ZQ Shao. miR-21 targets and inhibits tumor suppressor gene PTEN to promote prostate cancer cell proliferation and invasion: An experimental study. Asian Pac J Trop Med 10 (2017): 87-91.