Next Article in Journal
Biomarkers for Diagnosis, Prognosis and Response to Immunotherapy in Melanoma
Next Article in Special Issue
Circulating Tumour Cell Numbers Correlate with Platelet Count and Circulating Lymphocyte Subsets in Men with Advanced Prostate Cancer: Data from the ExPeCT Clinical Trial (CTRIAL-IE 15-21)
Previous Article in Journal
Loss of Paid Employment up to 4 Years after Colorectal Cancer Diagnosis—A Nationwide Register-Based Study with a Population-Based Reference Group
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Inflammation Pathways as Therapeutic Targets to Reduce Lethal Prostate Cancer in African American Men

Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(12), 2874; https://doi.org/10.3390/cancers13122874
Submission received: 7 May 2021 / Revised: 5 June 2021 / Accepted: 7 June 2021 / Published: 9 June 2021
(This article belongs to the Special Issue Role of Inflammation in Prostate Cancer)

Abstract

:

Simple Summary

Men of African descent are twice as likely to die of prostate cancer than other men. While equal access to care is the key target to improve cancer survival, it is now known that there are differences in disease biology and risk factor exposure across population groups. These differences could be causatively linked to the existing prostate cancer health disparities. In this review, we will discuss the candidate role of inflammation and the immune response as contributing factors to the excessive burden of lethal prostate cancer among men of African ancestry. Furthermore, we will introduce the concept that these immunogenic vulnerabilities could be exploited to address the adverse outcomes experienced by these men. Lastly, we will summarize how these immunogenic and inflammatory differences could be targeted using current treatments to improve survival for men of African descent.

Abstract

Despite substantial improvements in cancer survival, not all population groups have benefitted equally from this progress. For prostate cancer, men of African descent in the United States and England continue to have about double the rate of fatal disease compared to other men. Studies suggest that when there is equal access to care, survival disparities are greatly diminished. However, notable differences exist in prostate tumor biology across population groups. Ancestral factors and disparate exposures can lead to altered tumor biology, resulting in a distinct disease etiology by population group. While equal care remains the key target to improve survival, additional efforts should be made to gain comprehensive knowledge of the tumor biology in prostate cancer patients of African descent. Such an approach may identify novel intervention strategies in the era of precision medicine. A growing body of evidence shows that inflammation and the immune response may play a distinct role in prostate cancer disparities. Low-grade chronic inflammation and an inflammatory tumor microenvironment are more prevalent in African American patients and have been associated with adverse outcomes. Thus, differences in activation of immune–inflammatory pathways between African American and European American men with prostate cancer may exist. These differences may influence the response to immune therapy which is consistent with recent observations. This review will discuss mechanisms by which inflammation may contribute to the disparate outcomes experienced by African American men with prostate cancer and how these immunogenic and inflammatory vulnerabilities could be exploited to improve their survival.

Although cancer death rates have declined in the United States and other countries [1], disparities in cancer risk and outcomes persist, disproportionately affecting the systematically underserved and race/ethnic minoritized populations [2,3]. Prostate cancer is a key example of this with men of African descent in the United States and England continuing to have 2–3 times higher rates of fatal disease than other men [2]. Studies suggest that when there is equal access to care, survival disparities in prostate cancer are greatly diminished [4,5]. However, these investigations do not explain the notable differences in prostate cancer incidence, nor did they consider the now well-known differences in tumor biology across population groups. As shown recently, ancestral factors and disparate exposures may lead to distinct tumor biology in prostate cancer patients, resulting in a population-specific disease etiology [6,7,8,9]. While equal care remains the key target to improve survival, additional efforts should be made to gain comprehensive knowledge of the tumor biology in prostate cancer patients of African descent. Such an approach may identify novel intervention strategies for high risk groups in the era of precision medicine.
A growing body of evidence supports the hypothesis that inflammation plays a fundamental role in prostate cancer disparities. Key differences in activation of immune–inflammatory pathways between African American and European American men with prostate cancer are emerging and these biological processes may influence how African American men respond to therapy, as suggested by recent findings from clinical trials with the cancer vaccine, Sipuleucel T [10]. It is the aim of this review to discuss the candidate role of inflammation and the immune response as contributing factors to the excessive burden of lethal prostate cancer among men of African ancestry (Figure 1). Furthermore, we will introduce the concept that these immunogenic vulnerabilities could be exploited to address the adverse outcomes experienced by the high-risk African American population.

1. The Mutational and Immune–Oncologic Landscape of Prostate Tumors Differs between Populations

Prostate cancer displays large geographical differences in occurrence, with low incidence rates in East Asia and high rates in Western countries. Recognized risk factors for the disease include age, family history of the disease, race/ethnicity, and germline genetics [11,12,13,14]. It has been assumed that modifiable risk factors such as diet and lifestyle account for the majority of prostate cancers globally [15]. There is strong evidence from migration studies that the environment modulates prostate cancer risk [16,17]. Yet, there are few environmental factors that have consistently been linked to prostate cancer [11,18]. Notably, while prostate cancer is the leading cause of cancer death among men in many countries globally, sub-Saharan Africa and the Caribbean have more than double the age-standardized rates of mortality compared to other regions of the world, including North America and Europe [3,19]. This observation led to the hypothesis that ancestral factors may predispose men of sub-Saharan African ancestry to prostate cancer and a more aggressive disease [3,19]. Recent observations revealing the association of genetic ancestral factors with prostate cancer risk support this hypothesis [20,21,22,23,24]. Moreover, men of African ancestry are at an increased risk of developing fatal prostate cancer in the United States and England [2] and present with more aggressive disease in the Caribbean and sub-Saharan Africa [3,25]. The causes of the observed global prostate cancer health disparities are still being investigated but certainly include delayed diagnosis and lack of access to health care, ancestral, lifestyle, and environmental risk factors, and likely tumor biological differences [21,26,27].
Prostate cancer is a heterogeneous disease, in which inherited factors may account for about 40 to 50% of the cases [28]. Several familial susceptibility genes have been described, including RNASEL, BRCA1, BRCA2, and HOXB13 [28,29,30,31,32]. RNASEL, or ribonuclease L, encodes a component of the interferon-regulated 2–5A system that functions in the antiviral roles of interferons [33], suggesting the importance of immune function in prostate cancer susceptibility. Most of the inherited risk for prostate cancer arises from common genetic variants [14]. More than 200 disease susceptibility loci are now known [24], but not all of them confer risk in men of African ancestry [34]. Numerous studies have examined the possibility of low penetrance genes contributing to the excessive burden of prostate cancer in African American men. To date, the best characterized risk locus for prostate cancer is located at 8q24. Multiple common variants within this locus increase the risk of prostate cancer in many populations [13,35,36,37,38]. As shown by several studies, this locus confers an even higher risk for prostate cancer in men of West African ancestry, when compared with men of European and East Asian ancestry, partly explained by variants that were only found in men of African ancestry [13,20,34,35,39,40]. Thus, the 8q24 region accounts for some of the excessive disease risk among men of African ancestry.
Prostate cancer can be classified into genomic subtypes, such as those with ETS-fusion gene arrangements and other subtypes that are negative for ETS-fusion gene arrangements, and either overexpress the SPINK1 oncogene or carry SPOP, FOXA1, or IDH1 mutations, or represent a triple-negative subtype (negative for ERG- and other ETS-fusions and SPINK1-negative) [6,41]. Early-stage prostate cancer contains few recurrent mutations in cancer-related genes (e.g., ETS gene fusions) [42,43]. Instead, prostate tumors are characterized by allelic gains of the MYC gene and deletions of the NKX3–1, PTEN, Rb, and TP53 tumor suppressors [44]. Yet, there is strong evidence of prominent population differences in the acquisition of genetic alterations for prostate cancer. Reports showed that prostate tumors from patients of either European, African, or Asian descent exhibit notable differences in acquired chromosomal aberrations (e.g., ERG fusion and PTEN loss) and subtype distribution [6,7,8,9], indicating disparities in disease etiology and mutational events among these population groups. Comparing African American with European American patients [6,45,46,47], significant differences were observed in the frequency of TMPRSS2–ERG fusions (about 25% African American vs. 40–45% European American), SPOP mutations (about 20% African American vs. 10% European American), and PTEN deletions (about 10–15% African American vs. 30% European American). Chinese prostate cancer patients acquire mutations in FOXA1 at a high frequency (about 40%), as shown by a recent report [9]. This gene is infrequently mutated in European-ancestry populations (<10%).
Chronic inflammation has been described as a prostate cancer risk factor that is associated with aggressive disease [48,49]. We found that aspirin use significantly reduces the risk of advanced prostate cancer in African American men [50]. Yet, no study has assessed whether these men commonly develop a systemic inflammatory process that increases the risk of prostate cancer progression and mortality. While environmental exposures, such as infections, promote systemic inflammation, ancestral factors may also influence inflammatory processes and the response to infections [51,52].

2. Inflammation as a Possible Driver of Aggressive Prostate Cancer in African American Men

The immune–inflammation signature that was initially described by Wallace et al. to be prevalent in prostate tumors of African American patients is central to the hypothesis that inflammation is a candidate driver of prostate cancer disparities [53]. Subsequently observed by others [54] and validated in TCGA [55], this signature includes upregulation of genes in the interferon (IFN) signaling pathway and contains elements of a viral mimicry signature. Further investigations of this signature in prostate tumors from African American men describe a signature which corresponds to a previously described “interferon-related DNA damage resistance signature”, also termed IRDS [56,57]. Detection of IRDS is a marker of decreased disease-free survival in prostate cancer and has been linked to acquired resistance to radiation and chemotherapy in breast cancer. Thus, upregulation of this signature in African American tumors indicates a mechanism by which either inflammatory ancestral factors or a yet unknown infectious agent may contribute adversely to prostate cancer outcomes. Even though the presence of IRDS in a tumor may indicate an adverse outcome, this signature may also constitute a vulnerability. Tumors with an interferon-stimulated gene signature were reported to be highly susceptible to inhibition of adenosine deaminase acting on RNA (ADAR1) [58,59].
Despite the fact that we know it occurs about twice as often in African American prostate tumors when compared to European American tumors [56], the precise origin of this immune inflammation signature remains unknown. However, presence of the signature is associated with an interferon-λ4 ΔG genotype [56]. This genotype is responsible for production of the interferon lambda 4 protein (IFNL4) and is most common in people of West African ancestry and influences host viral response [56,60]. In this context, the signature may have origins in either infection history [61], pro-inflammatory diets [62], changes to the epigenome [63], or reactivation of endogenous retroviral sequences which have been reported in African American prostate cancer patients [64].
Multiple studies reported upregulation of inflammatory mediators in the tumor microenvironment (TME) of African American prostate cancer patients, many of which have implications for disease prognosis [65,66,67,68,69]. Gillard et al. investigated the role of the stroma in prostate cancer disparities by isolating prostate fibroblasts from the TME of African American and European American men and culturing prostate cancer cell lines in conditioned fibroblast media [65]. They found enhanced expression of proinflammatory mediators including TrKB, BDNF, VEGF, and IL6 by tumor cells when the conditioned media was obtained from fibroblasts of African American origin as compared to European origin. This implicates the stromal environment in African American men as a potential driver of prostate cancer progression through elevation of inflammatory mediators. Weiner et al. report higher immune content in the TME of prostate tumors from African American men compared to European American men with the proportion of plasma cells contributing the greatest difference in quantity across three independent cohorts [68]. These high intra tumoral counts of plasma cells were further associated with increased metastasis-free survival in both a Johns Hopkins Medical Institute and the TCGA cohort, implicating plasma cells as candidate regulators of the immune responsiveness in African American men with prostate cancer. High plasma cell levels correlated with increased IgG expression and IFN signaling, and B cell and natural killer (NK) cell activity in tumors of these patients, showing a possible link between high plasma cells and increased immune activity. High IgG expression and NK cell activity also showed clinical significance as they were associated with increased metastasis-free survival. Our group previously detected a B cell signature in prostate tumors from current smokers, but smoking is thought to increase the risk of metastasis [70]. Collectively, these findings suggest a regulatory network between intratumor immune cells, inflammatory cytokines, and cells in the TME. Such a network, if clearly defined, may have potential as a biomarker of responsiveness to immunotherapy and targets to improve outcomes among African American patients.
In support of these findings, Awasthi and colleagues reported distinct changes in immune pathways including overall higher immune cell content, enrichment of immune oncological pathways, and lower DNA damage repair in prostate tumors of African American men compared to European American men [66]. After exploring discovery and validation cohorts of immune-related genes, the authors focused on 38 genes that were differentially expressed between the two population groups. Of these genes, 26 with the most robust gene expression differences were identified as being consistently associated with major immune biological pathways, including IFN signaling and cytokine signaling based on discovery and validation with two separate pathway analysis tools. As a stand-out, the proinflammatory gene IFITM3 (IFN inducible transmembrane protein 3) was the only gene overexpressed in African American prostate tumors that predicted increased risk of biochemical recurrence only for African American men with prostate cancer, but not European American men.
The cause of this elevated immune–inflammation response is still under investigation. Numerous studies have shown that population differences in genetic ancestry can contribute to population differences in cancer susceptibility through processes that may involve inflammation. Genetic ancestry and natural selection are known to contribute to population differences in immune response to pathogens [52,71]. Furthermore, relationships of ancestry with expression levels of inflammatory cytokines are well documented in human populations [72,73]. As a modifiable risk factor, a pro-inflammatory diet that associates with high-grade prostate cancer is more commonly consumed by African American than European American men [62] and may lead to systemic inflammation. Other inducers of systemic inflammation may include stress exposures. Stress signaling transduces its biological effects through hypersecretion of the corticotrophin-releasing hormone and activation of the peripheral autonomic and sympathetic nervous system, which has direct effects on tumor biology and immune response, promoting inflammation, angiogenesis, mesenchymal differentiation, and metastasis [74]. As a final example, co-morbidities including chronic infections and diabetes can be excessively high in African American men [75,76,77]. They are frequently associated with increased inflammatory processes which could contribute to cancer development. This suggests that there could be a role for both biological and environmental factors in the elevated immune–inflammation pathways that are reported in the prostate tumors from men of African descent, as previously discussed [78].

3. African American Men May Have a Differential Response to Certain Therapies for Metastatic Prostate Cancer

The peer-reviewed literature now provides some evidence that men of African descent may respond differently across the gamut of both standardized and emerging options of care for prostate cancer, including radiation, hormone therapy, chemotherapy, and immunotherapy. Differences in immune response may play a key role in many of these observations. Metastatic castration-resistant prostate cancer (mCRPC) is a main cause of lethal prostate cancer and therefore remains a key focus for research. Despite patients with mCRPC having multiple treatment options targeting a variety of mechanisms (Figure 2), median overall survival is still only around 3 years [79]. This further highlights the need for inclusion of diverse biospecimens in scientific studies and historically understudied populations in clinical trials to determine who is benefitting optimally from these currently approved treatments.

4. Radiation

Radium-223 is an approved therapeutic option for mCRPC patients with symptomatic bone metastases. Zhao et al. examined the response to radium-223 treatment in men from a Veteran Affairs cohort with mCRPC [80]. With equal access to care across the cohort, this group found that African American men may have a better response to this treatment compared to European American men, resulting in a 25% decreased risk of mortality in this equal access to care study. African American men in this study were more likely to have received docetaxel beforehand and the improved response to therapy was despite the African American cases being more likely to not start radium treatment until further along in the disease course. Patients harboring DNA damage repair mutations have prolonged overall survival after radium-223 treatment compared to patients who do not have these alterations [81,82,83]. This is also the subject of another clinical trial currently in the recruitment stage (NCT04489719). With Awasthi et al. reporting decreased DNA damage repair capacity in prostate tumors of African American men, it can be speculated that inactivating mutations that decrease the DNA damage repair capacity in tumors from African American men may contribute to the positive outcomes post treatment with radium-223 [66].
A recent, small, phase II trial (NCT02463799) found combining radium-223 treatment with Sipuleucel-T increased progression-free survival and overall survival in men with mCRPC [84]. Now that studies have shown better responses from African American men treated with radium-223 and Sipuleucel-T separately [10,80], a planned larger trial may inform on whether African American men may benefit synergistically from this combination approach.

5. Immunotherapy

Immunotherapy has not been as successful in treating prostate cancer as with other hematologic or solid cancers and clinical trials show a modest [85] to no effect [79,86,87] on survival. This has been attributed to prostate cancer not being as immunogenic as other cancers. However, recent studies indicate a potential role for immunotherapy in certain patient groups with prostate cancer. Precision medicine strategies targeting immunotherapy to those men with the best response is the preferred goal. Evidence is currently being built to support the hypothesis that African American men may have a differential and perhaps superior response to certain treatments due to changes in immune cell response and a differing tumor biology.
Tumors from men of African descent may have a heightened response to immunotherapies, and specifically to cancer vaccines, as assumed from the presence of an interferon signature in their tumors and increased immune content in the TME [56,66]. Studies have shown that young people who self-report as African American mounted an increased immune response to vaccination [88,89]. Sartor et al. recently reported that African American men with mCRPC who were treated with the cancer vaccine, Sipuleucel-T, in the PROCEED trial/registry, had significantly better survival than the European American patients [10]. Median overall survival was 35.3 months for African American men compared to 25.8 months for European American men, in a PSA-matched set. This difference became even greater when measured in patients with a baseline PSA below the median, with median overall survival of 54.3 months in African American men versus 33.4 months in European American men. Increased activation of dendritic cells is a proposed mechanism of action of the vaccine and in agreement with this, activated dendritic cells in localized tumors have subsequently been associated with improved distant metastasis free survival [90]. Mechanistically, evidence points towards a complex interplay of immune cells with tumor biology which may predict prognosis and response to therapy. However, the lack of tumor specimens from African American men means that more work must be done to capitalize on the differences in the immune landscape which may improve response to treatment in this population.
Generally, poor immunogenicity has resulted in little success for PD-L1 blockade in treatment of prostate cancer [87,91]. This has been attributed in part to relatively low PD-L1 expression from tumor cells [92,93]. However, this is not consistent across the literature, with studies also reporting increased PD-L1 expression and association with biochemical recurrence [94] and shorter metastasis free survival [95]. Petitprez et al. provide preliminary evidence that a composite assessment of both PD-L1 and CD8 expression in localized prostate cancer may be a good strategy for predicting outcomes in mCRPC [95]. A group in Norway reported high PD-L1 expression in post-prostatectomy, hormone-naïve tumor epithelial cells with a non-significant trend towards an inverse association between PD-L1 expression and biochemical failure-free survival [96]. However, clinical trials investigating the effect of PD-L1 inhibition reported no significant clinical benefit. Yet, they have typically not included men of African descent [87].
Recent work has focused on PD-L1 expression on tumor-infiltrating immune cells. Bishop et al. reported enzalutamide-resistant prostate cancer patients showing increased PD-L1 expression on dendritic cells and high PD-L1 T cells when compared to enzalutamide-sensitive or treatment-naïve patients [97]. African American ethnicity and an aggressive cancer phenotype have been associated with prediction of tumor PD-L1 positivity in hormone-naïve tumors [98], suggesting a potential benefit for immunotherapy in African Americans at high risk of aggressive disease, but this has not been replicated yet [66]. When tumors are enzalutamide-sensitive, McNamara et al. preliminarily reported increased overall survival for African American, chemotherapy-naïve men with mCRPC treated with abiraterone or enzalutamide compared to European American men [99]. Overall survival was 918 days for African Americans compared to 781 days for European Americans. This study in a Veteran Affairs population was retrospective in design, again pointing to the value of equal access to care across populations. Thus, additional work is warranted, including measurement of PD-L1 in tumor samples from African American men post various treatment regimens to account for increased immunogenic response to therapy.

6. Other Treatment Opportunities

Historically, participation of African American men in clinical trials has been low. Reasons for this are multifactorial but include historical mistrust of the medical profession as a result of systemic racism and major ethical breaches in the past [100]. A higher prevalence of comorbidities and a lack of access to academic medical centers involved in trials may also prevent access to trials [100,101,102]. This prevents generalizability across population groups when reporting clinical trial data. A recent example highlights the need to include diverse population groups and possibly stratify clinical trial participants by race to get a fuller picture of treatment response. Halabi et al. completed a meta-analysis of survival outcomes for African American versus European American men in phase III clinical trials treating mCRPC with docetaxel [103]. With just 6% of African American participants, they reported that while overall median survival was similar, a pooled hazard ratio of 0.81 (95% CI, 0.72 to 0.91) post adjustment for baseline prognostic factors estimated that African American men may have a significantly decreased risk of death compared to European American men. This was despite African American men having baseline characteristics known to be prognostic of overall survival including statistically significantly worse performance status, higher testosterone levels, higher PSA levels, and lower hemoglobin levels.
It is assumed by many that the prostate cancer biology in men of African ancestry is intrinsically more aggressive—at least for a subset of patients—leading to a survival health disparity in the population [104]. Yet, this does not mean that African American men would not respond as well as European American men to most standard therapies. In fact, it appears that the treatment responses of African American and European American men are mostly similar. Yet, tumors in African American men could still respond better to certain therapies compared to the average response among European American men. Some of the treatments or combination treatments discussed in this review were the subject of small clinical trials and so these therapeutic options might not be widely offered yet in the clinic. Therefore, the findings require further evaluation in larger studies but do suggest that there is a potential role for these treatments in reducing the survival disparities observed in prostate cancer. African American men are less likely to be recruited into clinical trials and may not have the opportunities to avail of these new therapeutic options.

7. Germline and Somatic Mutations in DNA Repair Pathways

A proposed feature of prostate tumors in African American men that may play a prominent role in differential response to treatment is a deficiency in DNA damage repair capacity. Both germline and somatic alterations to DNA damage repair pathways have now been found in prostate tumors across multiple studies [66,105,106,107]. Tumors from African American men were reported to have a significantly lower level of DNA repair capacity when compared to those from European American men. Notably, these tumors seemed to have an increased radiosensitivity [66].
Germline mutations in DNA repair genes have a higher occurrence in metastatic prostate cancer when compared to localized prostate cancer [105,106]. BRCA1/2 pathogenic variants have been associated with more aggressive prostate cancer and adverse survival outcomes [108,109]. DNA repair gene mutations may contribute to aggressive disease in African American men. Acquired somatic mutations may differ among patient groups, with Yadav et al. reporting that prostate tumors from African American men were twice as likely to have at least one mutation in nucleotide excision repair pathway genes compared to European American (89% vs. > 40%) [107]. Petrovics and colleagues reported that germline variants in DNA repair genes of unknown significance had an increased frequency in African American men (4.6%) compared to European American men (1.6%) [110]. As the significance of these is undetermined, there is an opportunity to investigate whether they play a pathogenic role in prostate cancer. The same authors also reported that just 0.7% of men with localized prostate cancer carried pathogenic variants of BRCA1/2 mutations, but this increased over 4-fold to 3.1% in patients with metastatic and advanced disease, indicating that the presence of known BRCA1/2 pathogenic variants is linked to disease status [110]. Because the FDA-approved PARP inhibitors, olaparib and rucaparib, have shown success in prolonging overall survival in mCRPC patients with mutations in these DNA damage response genes [111,112,113], they should be made available to all African American men with prostate cancer who carry these mutations.
Altered DNA damage repair pathways may sensitize tumors to immunotherapeutic approaches. Several clinical trials across many cancer sites including metastatic prostate cancer are currently underway, targeting DNA damage repair-deficient tumors with checkpoint inhibitors (extensively reviewed by Bever et al.) [114]. Mechanistically, in prostate cancer, the stimulator of the IFN genes (STING) pathway has been linked to the recruitment and activation of interferon-related genes in vitro, increasing sensitivity to the immune checkpoint inhibitor PD-L1 in DNA repair-deficient tumors [115,116,117,118]. As a low DNA repair capacity may increase tumor genomic instability and tumor mutational burden, this again might constitute a vulnerability to immunotherapeutic strategies [119]. It has been suggested that enhancement of this genomic instability through use of radiation, chemotherapy, or PARP inhibitors could augment the immunotherapeutic response [114,120,121].

8. Anti-Inflammatory Drug Aspirin for Prevention of Adverse Outcomes in African American Men with Prostate Cancer

While the research community plays catch-up with ensuring proper representation of population groups in clinical trials and precision medicine studies, strategies for the prevention of lethal prostate cancer and reduction in adverse outcomes are of paramount importance to men of African descent who continue to experience a disproportionally high mortality from prostate cancer. An extensive body of evidence including both preclinical and clinical studies led the United States Preventative Services Task Force to recommend the anti-inflammatory drug aspirin for prevention of colorectal cancer for an albeit narrow category of adults [122,123,124]. In keeping with the hypothesis that inflammation is one of the drivers of the prostate cancer disparities, our group explored the link between regular use of aspirin and prostate cancer in African American men. We found that regular aspirin use significantly reduces the risk of both advanced prostate cancer and disease recurrence in these men [50]. The finding is consistent with a similar observation in a previous study [125]. Inhibition of the pro-inflammatory cyclooxygenase/thromboxane A2 pathway has been identified as a potential mechanism of action for aspirin in the prevention of metastatic cancer [126]. Using a retrospective cohort, we found a distinct association between high urinary 11-dehydrothromboxane B2 (the stable metabolite of thromboxane A2) and aggressive prostate cancer as well as adverse survival outcomes for African American men. Importantly, our ongoing research showed high 11-dehydrothromboxane B2 was inversely correlated with aspirin use, indicating a potential benefit of aspirin in preventing lethal prostate cancer through inhibition of TXA2 synthesis.
Lastly, data prospectively obtained in the Southern Community Cohort Study suggested that aspirin use is tentatively associated with a reduced prostate cancer mortality in African American men [127]. Hurwitz et al. also observed this inverse relationship between aspirin use and prostate cancer mortality in both African American and European American men using the ARIC cohort [128], again pointing to the potential benefit of aspirin use for men at high risk of fatal prostate cancer.

9. Conclusions

Elevated inflammatory processes in African American men with prostate cancer are a candidate biological driver of disparate disease risks. There is a need for more clinical trials specifically focused on the treatment response of African American men with metastatic prostate cancer [129]. Improved inclusion of minority populations in trials is essential to further enhance our knowledge of how inflammation and the immune response and alterations to molecular pathways may govern the response to emerging therapies across all patient groups. With evidence now building that suggests increased clinical benefit with certain therapies among African American men when compared to European American men, targeting inflammatory processes and the immune system could be an important strategy to reduce lethal disease in high-risk populations such as men of African ancestry.

Author Contributions

Conceptualization, M.K. and S.A.; writing—original draft preparation, M.K. and S.A.; writing—review and editing, M.K. and S.A.; funding acquisition, M.K. and S.A. Both authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Intramural Research Program of the NIH, National Cancer Institute (NCI), Center for Cancer Research and the DoD award W81XWH1810588 (to S.A.); Maeve Kiely is supported by the NCI Cancer Prevention Fellowship program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study is a literature review and did not analyze any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  2. Butler, E.N.; Kelly, S.P.; Coupland, V.H.; Rosenberg, P.S.; Cook, M.B. Fatal prostate cancer incidence trends in the United States and England by race, stage, and treatment. Br. J. Cancer 2020, 123, 1–8. [Google Scholar] [CrossRef]
  3. Rebbeck, T.R.; Devesa, S.S.; Chang, B.L.; Bunker, C.H.; Cheng, I.; Cooney, K.; Eeles, R.; Fernandez, P.; Giri, V.N.; Gueye, S.M.; et al. Global patterns of prostate cancer incidence, aggressiveness, and mortality in men of african descent. Prostate Cancer 2013, 2013, 560857. [Google Scholar] [CrossRef] [Green Version]
  4. Tewari, A.; Horninger, W.; Pelzer, A.E.; Demers, R.; Crawford, E.D.; Gamito, E.J.; Divine, G.; Johnson, C.C.; Bartsch, G.; Menon, M. Factors contributing to the racial differences in prostate cancer mortality. BJU Int. 2005, 96, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  5. Dess, R.T.; Hartman, H.E.; Mahal, B.A.; Soni, P.D.; Jackson, W.C.; Cooperberg, M.R.; Amling, C.L.; Aronson, W.J.; Kane, C.J.; Terris, M.K. Association of black race with prostate cancer–specific and other-cause mortality. JAMA Oncol. 2019, 5, 975–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Faisal, F.A.; Sundi, D.; Tosoian, J.J.; Choeurng, V.; Alshalalfa, M.; Ross, A.E.; Klein, E.; Den, R.; Dicker, A.; Erho, N.; et al. Racial Variations in Prostate Cancer Molecular Subtypes and Androgen Receptor Signaling Reflect Anatomic Tumor Location. Eur. Urol. 2016, 70, 14–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Magi-Galluzzi, C.; Tsusuki, T.; Elson, P.; Simmerman, K.; Lafargue, C.; Esgueva, R.; Klein, E.; Rubin, M.A.; Zhou, M. TMPRSS2-ERG gene fusion prevalence and class are significantly different in prostate cancer of Caucasian, African-American and Japanese patients. Prostate 2011, 71, 489–497. [Google Scholar] [CrossRef]
  8. Rosen, P.; Sesterhenn, I.A.; Brassell, S.A.; McLeod, D.G.; Srivastava, S.; Dobi, A. Clinical potential of the ERG oncoprotein in prostate cancer. Nat. Rev. Urol. 2012, 9, 131–137. [Google Scholar] [CrossRef] [PubMed]
  9. Li, J.; Xu, C.; Lee, H.J.; Ren, S.; Zi, X.; Zhang, Z.; Wang, H.; Yu, Y.; Yang, C.; Gao, X.; et al. A genomic and epigenomic atlas of prostate cancer in Asian populations. Nature 2020, 580, 93–99. [Google Scholar] [CrossRef] [PubMed]
  10. Sartor, O.; Armstrong, A.J.; Ahaghotu, C.; McLeod, D.G.; Cooperberg, M.R.; Penson, D.F.; Kantoff, P.W.; Vogelzang, N.J.; Hussain, A.; Pieczonka, C.M.; et al. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: Outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis. 2020, 23, 517–526. [Google Scholar] [CrossRef]
  11. Hsing, A.W.; Chokkalingam, A.P. Prostate cancer epidemiology. Front. Biosci. 2006, 11, 1388–1413. [Google Scholar] [CrossRef] [PubMed]
  12. Giovannucci, E.; Liu, Y.; Platz, E.A.; Stampfer, M.J.; Willett, W.C. Risk factors for prostate cancer incidence and progression in the health professionals follow-up study. Int. J. Cancer 2007, 121, 1571–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Amundadottir, L.T.; Sulem, P.; Gudmundsson, J.; Helgason, A.; Baker, A.; Agnarsson, B.A.; Sigurdsson, A.; Benediktsdottir, K.R.; Cazier, J.B.; Sainz, J.; et al. A common variant associated with prostate cancer in European and African populations. Nat.Genet. 2006, 38, 652–658. [Google Scholar] [CrossRef]
  14. Zheng, S.L.; Sun, J.; Wiklund, F.; Smith, S.; Stattin, P.; Li, G.; Adami, H.O.; Hsu, F.C.; Zhu, Y.; Balter, K.; et al. Cumulative association of five genetic variants with prostate cancer. N. Engl. J. Med. 2008, 358, 910–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Hsing, A.W.; Tsao, L.; Devesa, S.S. International trends and patterns of prostate cancer incidence and mortality. Int. J. Cancer 2000, 85, 60–67. [Google Scholar] [CrossRef]
  16. Shimizu, H.; Ross, R.K.; Bernstein, L.; Yatani, R.; Henderson, B.E.; Mack, T.M. Cancers of the prostate and breast among Japanese and white immigrants in Los Angeles County. Br. J. Cancer 1991, 63, 963–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Maringe, C.; Mangtani, P.; Rachet, B.; Leon, D.A.; Coleman, M.P.; dos Santos Silva, I. Cancer incidence in South Asian migrants to England, 1986–2004: Unraveling ethnic from socioeconomic differentials. Int. J. Cancer 2013, 132, 1886–1894. [Google Scholar] [CrossRef]
  18. Bylsma, L.C.; Alexander, D.D. A review and meta-analysis of prospective studies of red and processed meat, meat cooking methods, heme iron, heterocyclic amines and prostate cancer. Nutr. J. 2015, 14, 1–18. [Google Scholar] [CrossRef] [Green Version]
  19. Warner, W.A.; Lee, T.Y.; Fang, F.; Llanos, A.A.M.; Bajracharya, S.; Sundaram, V.; Badal, K.; Sookdeo, V.D.; Roach, V.; Lamont-Greene, M.; et al. The burden of prostate cancer in Trinidad and Tobago: One of the highest mortality rates in the world. Cancer Causes Control. 2018, 29, 685–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Freedman, M.L.; Haiman, C.A.; Patterson, N.; McDonald, G.J.; Tandon, A.; Waliszewska, A.; Penney, K.; Steen, R.G.; Ardlie, K.; John, E.M.; et al. Admixture mapping identifies 8q24 as a prostate cancer risk locus in African-American men. Proc. Natl. Acad. Sci. USA 2006, 103, 14068–14073. [Google Scholar] [CrossRef] [Green Version]
  21. Lachance, J.; Berens, A.J.; Hansen, M.E.B.; Teng, A.K.; Tishkoff, S.A.; Rebbeck, T.R. Genetic Hitchhiking and Population Bottlenecks Contribute to Prostate Cancer Disparities in Men of African Descent. Cancer Res. 2018, 78, 2432–2443. [Google Scholar] [CrossRef] [Green Version]
  22. Maruthappu, M.; Barnes, I.; Sayeed, S.; Ali, R. Incidence of prostate and urological cancers in England by ethnic group, 2001-2007: A descriptive study. BMC Cancer 2015, 15, 753. [Google Scholar] [CrossRef] [PubMed]
  23. Petersen, D.C.; Jaratlerdsiri, W.; van Wyk, A.; Chan, E.K.F.; Fernandez, P.; Lyons, R.J.; Mutambirw, S.B.A.; van der Merwe, A.; Venter, P.A.; Bates, W.; et al. African KhoeSan ancestry linked to high-risk prostate cancer. BMC Med. Genom. 2019, 12, 82. [Google Scholar] [CrossRef] [PubMed]
  24. Conti, D.V.; Darst, B.F.; Moss, L.C.; Saunders, E.J.; Sheng, X.; Chou, A.; Schumacher, F.R.; Olama, A.A.A.; Benlloch, S.; Dadaev, T.; et al. Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction. Nat. Genet. 2021, 53, 65–75. [Google Scholar] [CrossRef]
  25. Heyns, C.F.; Fisher, M.; Lecuona, A.; van der Merwe, A. Prostate cancer among different racial groups in the Western Cape: Presenting features and management. S. Afr. Med. J. 2011, 101, 267–270. [Google Scholar] [CrossRef] [Green Version]
  26. Wallace, T.A.; Martin, D.N.; Ambs, S. Interactions among genes, tumor biology and the environment in cancer health disparities: Examining the evidence on a national and global scale. Carcinogenesis 2011, 32, 1107–1121. [Google Scholar] [CrossRef] [Green Version]
  27. Rebbeck, T.R. Prostate Cancer Disparities by Race and Ethnicity: From Nucleotide to Neighborhood. Cold Spring Harb. Perspect Med. 2018, 8. [Google Scholar] [CrossRef]
  28. Giri, V.N.; Beebe-Dimmer, J.L. Familial prostate cancer. Semin. Oncol. 2016, 43, 560–565. [Google Scholar] [CrossRef]
  29. Carpten, J.; Nupponen, N.; Isaacs, S.; Sood, R.; Robbins, C.; Xu, J.; Faruque, M.; Moses, T.; Ewing, C.; Gillanders, E.; et al. Germline mutations in the ribonuclease L gene in families showing linkage with HPC1. Nat. Genet. 2002, 30, 181–184. [Google Scholar] [CrossRef]
  30. Rennert, H.; Zeigler-Johnson, C.M.; Addya, K.; Finley, M.J.; Walker, A.H.; Spangler, E.; Leonard, D.G.; Wein, A.; Malkowicz, S.B.; Rebbeck, T.R. Association of susceptibility alleles in ELAC2/HPC2, RNASEL/HPC1, and MSR1 with prostate cancer severity in European American and African American men. Cancer Epidemiol. Biomark. Prev. 2005, 14, 949–957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Ewing, C.M.; Ray, A.M.; Lange, E.M.; Zuhlke, K.A.; Robbins, C.M.; Tembe, W.D.; Wiley, K.E.; Isaacs, S.D.; Johng, D.; Wang, Y.; et al. Germline mutations in HOXB13 and prostate-cancer risk. N. Engl. J. Med. 2012, 366, 141–149. [Google Scholar] [CrossRef] [Green Version]
  32. Raymond, V.M.; Mukherjee, B.; Wang, F.; Huang, S.C.; Stoffel, E.M.; Kastrinos, F.; Syngal, S.; Cooney, K.A.; Gruber, S.B. Elevated risk of prostate cancer among men with Lynch syndrome. J. Clin. Oncol. 2013, 31, 1713–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Silverman, R.H. Implications for RNase L in prostate cancer biology. Biochemistry 2003, 42, 1805–1812. [Google Scholar] [CrossRef]
  34. Haiman, C.A.; Chen, G.K.; Blot, W.J.; Strom, S.S.; Berndt, S.I.; Kittles, R.A.; Rybicki, B.A.; Isaacs, W.B.; Ingles, S.A.; Stanford, J.L.; et al. Characterizing genetic risk at known prostate cancer susceptibility loci in African Americans. PLoS Genet. 2011, 7, e1001387. [Google Scholar] [CrossRef]
  35. Haiman, C.A.; Patterson, N.; Freedman, M.L.; Myers, S.R.; Pike, M.C.; Waliszewska, A.; Neubauer, J.; Tandon, A.; Schirmer, C.; McDonald, G.J.; et al. Multiple regions within 8q24 independently affect risk for prostate cancer. Nat. Genet. 2007, 39, 638–644. [Google Scholar] [CrossRef] [Green Version]
  36. Robbins, C.; Torres, J.B.; Hooker, S.; Bonilla, C.; Hernandez, W.; Candreva, A.; Ahaghotu, C.; Kittles, R.; Carpten, J. Confirmation study of prostate cancer risk variants at 8q24 in African Americans identifies a novel risk locus. Genome Res. 2007, 17, 1717–1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Xu, J.; Kibel, A.S.; Hu, J.J.; Turner, A.R.; Pruett, K.; Zheng, S.L.; Sun, J.; Isaacs, S.D.; Wiley, K.E.; Kim, S.T.; et al. Prostate cancer risk associated loci in African Americans. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2145–2149. [Google Scholar] [CrossRef] [Green Version]
  38. Cropp, C.D.; Robbins, C.M.; Sheng, X.; Hennis, A.J.; Carpten, J.D.; Waterman, L.; Worrell, R.; Schwantes-An, T.H.; Trent, J.M.; Haiman, C.A.; et al. 8q24 risk alleles and prostate cancer in African-Barbadian men. Prostate 2014, 74, 1579–1588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Gudmundsson, J.; Sulem, P.; Manolescu, A.; Amundadottir, L.T.; Gudbjartsson, D.; Helgason, A.; Rafnar, T.; Bergthorsson, J.T.; Agnarsson, B.A.; Baker, A.; et al. Genome-wide association study identifies a second prostate cancer susceptibility variant at 8q24. Nat. Genet. 2007, 39, 631–637. [Google Scholar] [CrossRef] [PubMed]
  40. Han, Y.; Rand, K.A.; Hazelett, D.J.; Ingles, S.A.; Kittles, R.A.; Strom, S.S.; Rybicki, B.A.; Nemesure, B.; Isaacs, W.B.; Stanford, J.L.; et al. Prostate Cancer Susceptibility in Men of African Ancestry at 8q24. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef] [PubMed]
  41. Attard, G.; Parker, C.; Eeles, R.A.; Schroder, F.; Tomlins, S.A.; Tannock, I.; Drake, C.G.; de Bono, J.S. Prostate cancer. Lancet 2016, 387, 70–82. [Google Scholar] [CrossRef]
  42. Berger, M.F.; Lawrence, M.S.; Demichelis, F.; Drier, Y.; Cibulskis, K.; Sivachenko, A.Y.; Sboner, A.; Esgueva, R.; Pflueger, D.; Sougnez, C.; et al. The genomic complexity of primary human prostate cancer. Nature 2011, 470, 214–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Fraser, M.; Sabelnykova, V.Y.; Yamaguchi, T.N.; Heisler, L.E.; Livingstone, J.; Huang, V.; Shiah, Y.J.; Yousif, F.; Lin, X.; Masella, A.P.; et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature 2017, 541, 359–364. [Google Scholar] [CrossRef]
  44. Abate-Shen, C.; Shen, M.M. Molecular genetics of prostate cancer. Genes Dev. 2000, 14, 2410–2434. [Google Scholar] [CrossRef] [Green Version]
  45. Khani, F.; Mosquera, J.M.; Park, K.; Blattner, M.; O’Reilly, C.; MacDonald, T.Y.; Chen, Z.; Srivastava, A.; Tewari, A.K.; Barbieri, C.E.; et al. Evidence for molecular differences in prostate cancer between African American and Caucasian men. Clin. Cancer Res. 2014, 20, 4925–4934. [Google Scholar] [CrossRef] [Green Version]
  46. Blackburn, J.; Vecchiarelli, S.; Heyer, E.E.; Patrick, S.M.; Lyons, R.J.; Jaratlerdsiri, W.; van Zyl, S.; Bornman, M.S.R.; Mercer, T.R.; Hayes, V.M. TMPRSS2-ERG fusions linked to prostate cancer racial health disparities: A focus on Africa. Prostate 2019, 79, 1191–1196. [Google Scholar] [CrossRef] [Green Version]
  47. Koga, Y.; Song, H.; Chalmers, Z.R.; Newberg, J.; Kim, E.; Carrot-Zhang, J.; Piou, D.; Polak, P.; Abdulkadir, S.A.; Ziv, E.; et al. Genomic Profiling of Prostate Cancers from Men with African and European Ancestry. Clin. Cancer Res. 2020, 26, 4651–4660. [Google Scholar] [CrossRef] [PubMed]
  48. Gurel, B.; Lucia, M.S.; Thompson, I.M., Jr.; Goodman, P.J.; Tangen, C.M.; Kristal, A.R.; Parnes, H.L.; Hoque, A.; Lippman, S.M.; Sutcliffe, S.; et al. Chronic inflammation in benign prostate tissue is associated with high-grade prostate cancer in the placebo arm of the prostate cancer prevention trial. Cancer Epidemiol. Biomark. Prev. 2014, 23, 847–856. [Google Scholar] [CrossRef] [Green Version]
  49. Klink, J.C.; Banez, L.L.; Gerber, L.; Lark, A.; Vollmer, R.T.; Freedland, S.J. Intratumoral inflammation is associated with more aggressive prostate cancer. World J. Urol. 2013, 31, 1497–1503. [Google Scholar] [CrossRef] [PubMed]
  50. Smith, C.J.; Dorsey, T.H.; Tang, W.; Jordan, S.V.; Loffredo, C.A.; Ambs, S. Aspirin Use Reduces the Risk of Aggressive Prostate Cancer and Disease Recurrence in African-American Men. Cancer Epidemiol. Biomark. Prev. 2017, 26, 845–853. [Google Scholar] [CrossRef] [Green Version]
  51. Van Dyke, A.L.; Cote, M.L.; Wenzlaff, A.S.; Land, S.; Schwartz, A.G. Cytokine SNPs: Comparison of allele frequencies by race and implications for future studies. Cytokine 2009, 46, 236–244. [Google Scholar] [CrossRef] [Green Version]
  52. Nedelec, Y.; Sanz, J.; Baharian, G.; Szpiech, Z.A.; Pacis, A.; Dumaine, A.; Grenier, J.C.; Freiman, A.; Sams, A.J.; Hebert, S.; et al. Genetic Ancestry and Natural Selection Drive Population Differences in Immune Responses to Pathogens. Cell 2016, 167, 657–669 e621. [Google Scholar] [CrossRef] [Green Version]
  53. Wallace, T.A.; Prueitt, R.L.; Yi, M.; Howe, T.M.; Gillespie, J.W.; Yfantis, H.G.; Stephens, R.M.; Caporaso, N.E.; Loffredo, C.A.; Ambs, S. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res. 2008, 68, 927–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Reams, R.R.; Agrawal, D.; Davis, M.B.; Yoder, S.; Odedina, F.T.; Kumar, N.; Higginbotham, J.M.; Akinremi, T.; Suther, S.; Soliman, K.F. Microarray comparison of prostate tumor gene expression in African-American and Caucasian American males: A pilot project study. Infect. Agent. Cancer 2009, 4 (Suppl. 1), S3. [Google Scholar] [CrossRef] [Green Version]
  55. Yuan, J.; Kensler, K.H.; Hu, Z.; Zhang, Y.; Zhang, T.; Jiang, J.; Xu, M.; Pan, Y.; Long, M.; Montone, K.T.; et al. Integrative comparison of the genomic and transcriptomic landscape between prostate cancer patients of predominantly African or European genetic ancestry. PLoS Genet. 2020, 16, e1008641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Tang, W.; Wallace, T.A.; Yi, M.; Magi-Galluzzi, C.; Dorsey, T.H.; Onabajo, O.O.; Obajemu, A.; Jordan, S.V.; Loffredo, C.A.; Stephens, R.M.; et al. IFNL4-DeltaG Allele Is Associated with an Interferon Signature in Tumors and Survival of African-American Men with Prostate Cancer. Clin. Cancer Res. 2018, 24, 5471–5481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Weichselbaum, R.R.; Ishwaran, H.; Yoon, T.; Nuyten, D.S.; Baker, S.W.; Khodarev, N.; Su, A.W.; Shaikh, A.Y.; Roach, P.; Kreike, B. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 18490–18495. [Google Scholar] [CrossRef] [Green Version]
  58. Gannon, H.S.; Zou, T.; Kiessling, M.K.; Gao, G.F.; Cai, D.; Choi, P.S.; Ivan, A.P.; Buchumenski, I.; Berger, A.C.; Goldstein, J.T. Identification of ADAR1 adenosine deaminase dependency in a subset of cancer cells. Nat. Commun. 2018, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
  59. Liu, H.; Golji, J.; Brodeur, L.K.; Chung, F.S.; Chen, J.T.; deBeaumont, R.S.; Bullock, C.P.; Jones, M.D.; Kerr, G.; Li, L. Tumor-derived IFN triggers chronic pathway agonism and sensitivity to ADAR loss. Nat. Med. 2019, 25, 95–102. [Google Scholar] [CrossRef]
  60. Prokunina-Olsson, L.; Muchmore, B.; Tang, W.; Pfeiffer, R.M.; Park, H.; Dickensheets, H.; Hergott, D.; Porter-Gill, P.; Mumy, A.; Kohaar, I. A variant upstream of IFNL3 (IL28B) creating a new interferon gene IFNL4 is associated with impaired clearance of hepatitis C virus. Nat. Genet. 2013, 45, 164. [Google Scholar] [CrossRef]
  61. Minas, T.Z.; Tang, W.; Smith, C.J.; Onabajo, O.O.; Obajemu, A.; Dorsey, T.H.; Jordan, S.V.; Obadi, O.M.; Ryan, B.M.; Prokunina-Olsson, L.; et al. IFNL4-ΔG is associated with prostate cancer among men at increased risk of sexually transmitted infections. Commun. Biol. 2018, 1, 191. [Google Scholar] [CrossRef] [Green Version]
  62. Vidal, A.C.; Oyekunle, T.; Howard, L.E.; Shivappa, N.; De Hoedt, A.; Figueiredo, J.C.; Taioli, E.; Fowke, J.H.; Lin, P.H.; Hebert, J.R.; et al. Dietary inflammatory index (DII) and risk of prostate cancer in a case-control study among Black and White US Veteran men. Prostate Cancer Prostatic Dis. 2019, 22, 580–587. [Google Scholar] [CrossRef]
  63. Chiappinelli, K.B.; Strissel, P.L.; Desrichard, A.; Li, H.; Henke, C.; Akman, B.; Hein, A.; Rote, N.S.; Cope, L.M.; Snyder, A. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 2015, 162, 974–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wallace, T.A.; Downey, R.F.; Seufert, C.J.; Schetter, A.; Dorsey, T.H.; Johnson, C.A.; Goldman, R.; Loffredo, C.A.; Yan, P.; Sullivan, F.J.; et al. Elevated HERV-K mRNA expression in PBMC is associated with a prostate cancer diagnosis particularly in older men and smokers. Carcinogenesis 2014, 35, 2074–2083. [Google Scholar] [CrossRef] [PubMed]
  65. Gillard, M.; Javier, R.; Ji, Y.; Zheng, S.L.; Xu, J.; Brendler, C.B.; Crawford, S.E.; Pierce, B.L.; Vander Griend, D.J.; Franco, O.E. Elevation of stromal-derived mediators of inflammation promote prostate cancer progression in African-American men. Cancer Res. 2018, 78, 6134–6145. [Google Scholar] [CrossRef] [Green Version]
  66. Awasthi, S.; Berglund, A.; Abraham-Miranda, J.; Rounbehler, R.J.; Kensler, K.; Serna, A.; Vidal, A.; You, S.; Freeman, M.R.; Davicioni, E. Comparative genomics reveals distinct immune-oncologic pathways in African American men with prostate cancer. Clin. Cancer Res. 2021, 27, 320–329. [Google Scholar] [CrossRef]
  67. Nonomura, N.; Takayama, H.; Nakayama, M.; Nakai, Y.; Kawashima, A.; Mukai, M.; Nagahara, A.; Aozasa, K.; Tsujimura, A. Infiltration of tumour-associated macrophages in prostate biopsy specimens is predictive of disease progression after hormonal therapy for prostate cancer. BJU Int. 2011, 107, 1918–1922. [Google Scholar] [CrossRef]
  68. Weiner, A.B.; Vidotto, T.; Liu, Y.; Mendes, A.A.; Salles, D.C.; Faisal, F.A.; Murali, S.; McFarlane, M.; Imada, E.L.; Zhao, X. Plasma cells are enriched in localized prostate cancer in Black men and are associated with improved outcomes. Nat. Commun. 2021, 12, 1–10. [Google Scholar] [CrossRef] [PubMed]
  69. Kinseth, M.A.; Jia, Z.; Rahmatpanah, F.; Sawyers, A.; Sutton, M.; Wang-Rodriguez, J.; Mercola, D.; McGuire, K.L. Expression differences between African American and Caucasian prostate cancer tissue reveals that stroma is the site of aggressive changes. Int. J. Cancer 2014, 134, 81–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Prueitt, R.L.; Wallace, T.A.; Glynn, S.A.; Yi, M.; Tang, W.; Luo, J.; Dorsey, T.H.; Stagliano, K.E.; Gillespie, J.W.; Hudson, R.S.; et al. An Immune-Inflammation Gene Expression Signature in Prostate Tumors of Smokers. Cancer Res. 2016, 76, 1055–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Barreiro, L.B.; Quintana-Murci, L. Evolutionary and population (epi)genetics of immunity to infection. Hum. Genet. 2020, 139, 723–732. [Google Scholar] [CrossRef]
  72. Coe, C.L.; Love, G.D.; Karasawa, M.; Kawakami, N.; Kitayama, S.; Markus, H.R.; Tracy, R.P.; Ryff, C.D. Population differences in proinflammatory biology: Japanese have healthier profiles than Americans. Brain Behav. Immun. 2011, 25, 494–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yao, S.; Hong, C.C.; Ruiz-Narvaez, E.A.; Evans, S.S.; Zhu, Q.; Schaefer, B.A.; Yan, L.; Coignet, M.V.; Lunetta, K.L.; Sucheston-Campbell, L.E.; et al. Genetic ancestry and population differences in levels of inflammatory cytokines in women: Role for evolutionary selection and environmental factors. PLoS Genet. 2018, 14, e1007368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Cole, S.W.; Nagaraja, A.S.; Lutgendorf, S.K.; Green, P.A.; Sood, A.K. Sympathetic nervous system regulation of the tumour microenvironment. Nat. Rev. Cancer 2015, 15, 563–572. [Google Scholar] [CrossRef] [Green Version]
  75. Daw, J. Contribution of Four Comorbid Conditions to Racial/Ethnic Disparities in Mortality Risk. Am. J. Prev. Med. 2017, 52, S95–S102. [Google Scholar] [CrossRef]
  76. Geiss, L.S.; Wang, J.; Cheng, Y.J.; Thompson, T.J.; Barker, L.; Li, Y.; Albright, A.L.; Gregg, E.W. Prevalence and incidence trends for diagnosed diabetes among adults aged 20 to 79 years, United States, 1980-2012. JAMA 2014, 312, 1218–1226. [Google Scholar] [CrossRef] [Green Version]
  77. Panigrahi, G.; Ambs, S. How Comorbidities Shape Cancer Biology and Survival. Trends Cancer 2021, 7, 488–495. [Google Scholar] [CrossRef] [PubMed]
  78. Minas, T.Z.; Kiely, M.; Ajao, A.; Ambs, S. An overview of cancer health disparities: New approaches and insights and why they matter. Carcinogenesis 2021, 42, 2–13. [Google Scholar] [CrossRef]
  79. Beer, T.M.; Kwon, E.D.; Drake, C.G.; Fizazi, K.; Logothetis, C.; Gravis, G.; Ganju, V.; Polikoff, J.; Saad, F.; Humanski, P.; et al. Randomized, Double-Blind, Phase III Trial of Ipilimumab Versus Placebo in Asymptomatic or Minimally Symptomatic Patients With Metastatic Chemotherapy-Naive Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2017, 35, 40–47. [Google Scholar] [CrossRef] [PubMed]
  80. Zhao, H.; Howard, L.E.; De Hoedt, A.; Terris, M.K.; Amling, C.L.; Kane, C.J.; Cooperberg, M.R.; Aronson, W.J.; Klaassen, Z.; Polascik, T.J. Racial Discrepancies in Overall Survival among Men Treated with 223Radium. J. Urol. 2020, 203, 331–337. [Google Scholar] [CrossRef]
  81. Van der Doelen, M.J.; Velho, P.I.; Slootbeek, P.H.; Naga, S.P.; Bormann, M.; van Helvert, S.; Kroeze, L.I.; van Oort, I.M.; Gerritsen, W.R.; Antonarakis, E.S. Impact of DNA damage repair defects on response to radium-223 and overall survival in metastatic castration-resistant prostate cancer. Eur. J. Cancer 2020, 136, 16–24. [Google Scholar] [CrossRef]
  82. Ramos, J.D.; Mostaghel, E.A.; Pritchard, C.C.; Evan, Y.Y. DNA repair pathway alterations in metastatic castration-resistant prostate cancer responders to radium-223. Clin. Genitourin. Cancer 2018, 16, 106–110. [Google Scholar] [CrossRef] [PubMed]
  83. Steinberger, A.E.; Cotogno, P.; Ledet, E.M.; Lewis, B.; Sartor, O. Exceptional duration of radium-223 in prostate cancer with a BRCA2 mutation. Clin. Genitourin. Cancer 2017, 15, e69–e71. [Google Scholar] [CrossRef]
  84. Marshall, C.H.; Fu, W.; Wang, H.; Park, J.C.; DeWeese, T.L.; Tran, P.T.; Song, D.Y.; King, S.; Afful, M.; Hurrelbrink, J.; et al. Randomized Phase II Trial of Sipuleucel-T with or without Radium-223 in Men with Bone-metastatic Castration-resistant Prostate Cancer. Clin. Cancer Res. 2021, 27, 1623–1630. [Google Scholar] [CrossRef]
  85. Antonarakis, E.S.; Piulats, J.M.; Gross-Goupil, M.; Goh, J.; Ojamaa, K.; Hoimes, C.J.; Vaishampayan, U.; Berger, R.; Sezer, A.; Alanko, T. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: Multicohort, open-label phase II KEYNOTE-199 study. J. Clin. Oncol. 2020, 38, 395. [Google Scholar] [CrossRef]
  86. Kwon, E.D.; Drake, C.G.; Scher, H.I.; Fizazi, K.; Bossi, A.; Van den Eertwegh, A.J.; Krainer, M.; Houede, N.; Santos, R.; Mahammedi, H. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy (CA184-043): A multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. 2014, 15, 700–712. [Google Scholar] [CrossRef] [Green Version]
  87. Brahmer, J.R.; Drake, C.G.; Wollner, I.; Powderly, J.D.; Picus, J.; Sharfman, W.H.; Stankevich, E.; Pons, A.; Salay, T.M.; McMiller, T.L. Phase I study of single-agent anti–programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J. Clin. Oncol. 2010, 28, 3167. [Google Scholar] [CrossRef]
  88. Haralambieva, I.H.; Salk, H.M.; Lambert, N.D.; Ovsyannikova, I.G.; Kennedy, R.B.; Warner, N.D.; Pankratz, V.S.; Poland, G.A. Associations between race, sex and immune response variations to rubella vaccination in two independent cohorts. Vaccine 2014, 32, 1946–1953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Kurupati, R.; Kossenkov, A.; Haut, L.; Kannan, S.; Xiang, Z.; Li, Y.; Doyle, S.; Liu, Q.; Schmader, K.; Showe, L. Race-related differences in antibody responses to the inactivated influenza vaccine are linked to distinct pre-vaccination gene expression profiles in blood. Oncotarget 2016, 7, 62898. [Google Scholar] [CrossRef]
  90. Zhao, S.G.; Lehrer, J.; Chang, S.L.; Das, R.; Erho, N.; Liu, Y.; Sjöström, M.; Den, R.B.; Freedland, S.J.; Klein, E.A. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. JNCI J. Natl. Cancer Inst. 2019, 111, 301–310. [Google Scholar] [CrossRef] [PubMed]
  91. Topalian, S.L.; Sznol, M.; Brahmer, J.R.; McDermott, D.F.; Smith, D.C.; Gettinger, S.N.; Taube, J.M.; Drake, C.G.; Pardoll, D.M.; Powderly, J.D. Nivolumab (anti-PD-1; BMS-936558; ONO-4538) in Patients with Advanced Solid Tumors: Survival and Long-Term Safety in a Phase I Trial; American Society of Clinical Oncology: Alexandria, VA, USA, 2013; p. 3002. [Google Scholar]
  92. Haffner, M.C.; Guner, G.; Taheri, D.; Netto, G.J.; Palsgrove, D.N.; Zheng, Q.; Guedes, L.B.; Kim, K.; Tsai, H.; Esopi, D.M. Comprehensive evaluation of programmed death-ligand 1 expression in primary and metastatic prostate cancer. Am. J. Pathol. 2018, 188, 1478–1485. [Google Scholar] [CrossRef] [Green Version]
  93. Fankhauser, C.D.; Schüffler, P.J.; Gillessen, S.; Omlin, A.; Rupp, N.J.; Rueschoff, J.H.; Hermanns, T.; Poyet, C.; Sulser, T.; Moch, H. Comprehensive immunohistochemical analysis of PD-L1 shows scarce expression in castration-resistant prostate cancer. Oncotarget 2018, 9, 10284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Li, H.; Wang, Z.; Zhang, Y.; Sun, G.; Ding, B.; Yan, L.; Liu, H.; Guan, W.; Hu, Z.; Wang, S. The immune checkpoint regulator PDL1 is an independent prognostic biomarker for biochemical recurrence in prostate cancer patients following adjuvant hormonal therapy. J. Cancer 2019, 10, 3102. [Google Scholar] [CrossRef]
  95. Petitprez, F.; Fossati, N.; Vano, Y.; Freschi, M.; Becht, E.; Lucianò, R.; Calderaro, J.; Guédet, T.; Lacroix, L.; Rancoita, P.M. PD-L1 expression and CD8+ T-cell infiltrate are associated with clinical progression in patients with node-positive prostate cancer. Eur. Urol. Focus 2019, 5, 192–196. [Google Scholar] [CrossRef] [PubMed]
  96. Ness, N.; Andersen, S.; Khanehkenari, M.R.; Nordbakken, C.V.; Valkov, A.; Paulsen, E.E.; Nordby, Y.; Bremnes, R.M.; Donnem, T.; Busund, L.T.; et al. The prognostic role of immune checkpoint markers programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) in a large, multicenter prostate cancer cohort. Oncotarget 2017, 8, 26789–26801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Bishop, J.L.; Sio, A.; Angeles, A.; Roberts, M.E.; Azad, A.A.; Chi, K.N.; Zoubeidi, A. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget 2015, 6, 234. [Google Scholar] [CrossRef] [Green Version]
  98. Calagua, C.; Russo, J.; Sun, Y.; Schaefer, R.; Lis, R.; Zhang, Z.; Mahoney, K.; Bubley, G.J.; Loda, M.; Taplin, M.E.; et al. Expression of PD-L1 in Hormone-naïve and Treated Prostate Cancer Patients Receiving Neoadjuvant Abiraterone Acetate plus Prednisone and Leuprolide. Clin. Cancer Res. 2017, 23, 6812–6822. [Google Scholar] [CrossRef] [Green Version]
  99. McNamara, M.A.; George, D.J.; Ramaswamy, K.; Lechpammer, S.; Mardekian, J.; Schultz, N.M.; Wang, L.; Baser, O.; Huang, A.; Freedland, S.J. Overall survival by race in chemotherapy-naïve metastatic castration-resistant prostate cancer (mCRPC) patients treated with abiraterone acetate or enzalutamide. J. Clin. Oncol. 2019, 37, 212. [Google Scholar] [CrossRef]
  100. Brandon, D.T.; Isaac, L.A.; LaVeist, T.A. The legacy of Tuskegee and trust in medical care: Is Tuskegee responsible for race differences in mistrust of medical care? J. Natl. Med. Assoc. 2005, 97, 951. [Google Scholar]
  101. Duma, N.; Vera Aguilera, J.; Paludo, J.; Haddox, C.L.; Gonzalez Velez, M.; Wang, Y.; Leventakos, K.; Hubbard, J.M.; Mansfield, A.S.; Go, R.S. Representation of minorities and women in oncology clinical trials: Review of the past 14 years. J. Oncol. Pract. 2018, 14, e1–e10. [Google Scholar] [CrossRef]
  102. Loree, J.M.; Anand, S.; Dasari, A.; Unger, J.M.; Gothwal, A.; Ellis, L.M.; Varadhachary, G.; Kopetz, S.; Overman, M.J.; Raghav, K. Disparity of race reporting and representation in clinical trials leading to cancer drug approvals from 2008 to 2018. JAMA Oncol. 2019, 5, e191870. [Google Scholar] [CrossRef] [PubMed]
  103. Halabi, S.; Dutta, S.; Tangen, C.M.; Rosenthal, M.; Petrylak, D.P.; Thompson, I.M., Jr.; Chi, K.N.; Araujo, J.C.; Logothetis, C.; Quinn, D.I.; et al. Overall Survival of Black and White Men With Metastatic Castration-Resistant Prostate Cancer Treated With Docetaxel. J. Clin. Oncol. 2019, 37, 403–410. [Google Scholar] [CrossRef]
  104. Powell, I.J.; Bock, C.H.; Ruterbusch, J.J.; Sakr, W. Evidence supports a faster growth rate and/or earlier transformation to clinically significant prostate cancer in black than in white American men, and influences racial progression and mortality disparity. J. Urol. 2010, 183, 1792–1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Fantus, R.J.; Helfand, B.T. Germline Genetics of Prostate Cancer: Time to Incorporate Genetics into Early Detection Tools. Clin. Chem. 2019, 65, 74–79. [Google Scholar] [CrossRef] [Green Version]
  106. Pritchard, C.C.; Mateo, J.; Walsh, M.F.; De Sarkar, N.; Abida, W.; Beltran, H.; Garofalo, A.; Gulati, R.; Carreira, S.; Eeles, R. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N. Engl. J. Med. 2016, 375, 443–453. [Google Scholar] [CrossRef]
  107. Yadav, S.; Anbalagan, M.; Baddoo, M.; Chellamuthu, V.K.; Mukhopadhyay, S.; Woods, C.; Jiang, W.; Moroz, K.; Flemington, E.K.; Makridakis, N. Somatic mutations in the DNA repairome in prostate cancers in African Americans and Caucasians. Oncogene 2020, 39, 4299–4311. [Google Scholar] [CrossRef] [Green Version]
  108. Taylor, R.A.; Fraser, M.; Livingstone, J.; Espiritu, S.M.G.; Thorne, H.; Huang, V.; Lo, W.; Shiah, Y.-J.; Yamaguchi, T.N.; Sliwinski, A. Germline BRCA2 mutations drive prostate cancers with distinct evolutionary trajectories. Nat. Commun. 2017, 8, 1–10. [Google Scholar] [CrossRef]
  109. Narod, S.; Neuhausen, S.; Vichodez, G.; Armel, S.; Lynch, H.; Ghadirian, P.; Cummings, S.; Olopade, O.; Stoppa-Lyonnet, D.; Couch, F. Rapid progression of prostate cancer in men with a BRCA2 mutation. Br. J. Cancer 2008, 99, 371–374. [Google Scholar] [CrossRef] [PubMed]
  110. Petrovics, G.; Price, D.K.; Lou, H.; Chen, Y.; Garland, L.; Bass, S.; Jones, K.; Kohaar, I.; Ali, A.; Ravindranath, L. Increased frequency of germline BRCA2 mutations associates with prostate cancer metastasis in a racially diverse patient population. Prostate Cancer Prostatic Dis. 2019, 22, 406–410. [Google Scholar] [CrossRef]
  111. Mateo, J.; Porta, N.; Bianchini, D.; McGovern, U.; Elliott, T.; Jones, R.; Syndikus, I.; Ralph, C.; Jain, S.; Varughese, M. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): A multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 162–174. [Google Scholar] [CrossRef]
  112. Mateo, J.; McKay, R.; Abida, W.; Aggarwal, R.; Alumkal, J.; Alva, A.; Feng, F.; Gao, X.; Graff, J.; Hussain, M. Accelerating precision medicine in metastatic prostate cancer. Nat. Cancer 2020, 1, 1041–1053. [Google Scholar] [CrossRef]
  113. Ratta, R.; Guida, A.; Scotté, F.; Neuzillet, Y.; Teillet, A.B.; Lebret, T.; Beuzeboc, P. PARP inhibitors as a new therapeutic option in metastatic prostate cancer: A systematic review. Prostate Cancer Prostatic Dis. 2020, 23, 549–560. [Google Scholar] [CrossRef]
  114. Bever, K.M.; Le, D.T. DNA repair defects and implications for immunotherapy. J. Clin. Investig. 2018, 128, 4236–4242. [Google Scholar] [CrossRef]
  115. Wang, H.; Hu, S.; Chen, X.; Shi, H.; Chen, C.; Sun, L.; Chen, Z.J. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc. Natl. Acad. Sci. USA 2017, 114, 1637–1642. [Google Scholar] [CrossRef] [Green Version]
  116. Härtlova, A.; Erttmann, S.F.; Raffi, F.A.; Schmalz, A.M.; Resch, U.; Anugula, S.; Lienenklaus, S.; Nilsson, L.M.; Kröger, A.; Nilsson, J.A. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 2015, 42, 332–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Morel, K.L.; Sheahan, A.V.; Burkhart, D.L.; Baca, S.C.; Boufaied, N.; Liu, Y.; Qiu, X.; Cañadas, I.; Roehle, K.; Heckler, M. EZH2 inhibition activates a dsRNA–STING–interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer. Nat. Cancer 2021, 2, 1–13. [Google Scholar] [CrossRef] [PubMed]
  118. Esteves, A.M.; Papaevangelou, E.; Dasgupta, P.; Galustian, C. Combination of Interleukin-15 with a STING agonist, ADU-S100 analog: A potential immunotherapy for prostate cancer. Front. Oncol. 2021, 11, 621550. [Google Scholar] [CrossRef]
  119. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 1–14. [Google Scholar] [CrossRef]
  120. Gupta, P.D.; Chaudagar, K.; Sharma-Saha, S.; Bynoe, K.; Maillat, L.; Heiss, B.; Stadler, W.M.; Patnaik, A. PARP and PI3K inhibitor combination therapy eradicates c-MYC-driven murine prostate cancers via cGAS/STING pathway activation within tumor-associated macrophages. bioRxiv 2020. [Google Scholar] [CrossRef]
  121. Chabanon, R.M.; Muirhead, G.; Krastev, D.B.; Adam, J.; Morel, D.; Garrido, M.; Lamb, A.; Hénon, C.; Dorvault, N.; Rouanne, M. PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer. J. Clin. Investig. 2019, 129, 1211–1228. [Google Scholar] [CrossRef]
  122. Chubak, J.; Whitlock, E.P.; Williams, S.B.; Kamineni, A.; Burda, B.U.; Buist, D.S.; Anderson, M.L. Aspirin for the prevention of cancer incidence and mortality: Systematic evidence reviews for the US Preventive Services Task Force. Ann. Intern. Med. 2016, 164, 814–825. [Google Scholar] [CrossRef] [PubMed]
  123. Bibbins-Domingo, K. Aspirin use for the primary prevention of cardiovascular disease and colorectal cancer: US Preventive Services Task Force recommendation statement. Ann. Intern. Med. 2016, 164, 836–845. [Google Scholar] [CrossRef] [Green Version]
  124. Dehmer, S.P.; Maciosek, M.V.; Flottemesch, T.J.; LaFrance, A.B.; Whitlock, E.P. Aspirin for the primary prevention of cardiovascular disease and colorectal cancer: A decision analysis for the US Preventive Services Task Force. Ann. Intern. Med. 2016, 164, 777–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Osborn, V.W.; Chen, S.C.; Weiner, J.; Schwartz, D.; Schreiber, D. Impact of aspirin on clinical outcomes for African American men with prostate cancer undergoing radiation. Tumori 2016, 102, 65–70. [Google Scholar] [CrossRef]
  126. Lucotti, S.; Cerutti, C.; Soyer, M.; Gil-Bernabé, A.M.; Gomes, A.L.; Allen, P.D.; Smart, S.; Markelc, B.; Watson, K.; Armstrong, P.C. Aspirin blocks formation of metastatic intravascular niches by inhibiting platelet-derived COX-1/thromboxane A 2. J. Clin. Investig. 2019, 129, 1845–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Tang, W.; Fowke, J.H.; Hurwitz, L.M.; Steinwandel, M.; Blot, W.J.; Ambs, S. Aspirin Use and Prostate Cancer among African-American Men in the Southern Community Cohort Study. Cancer Epidemiol. Prev. Biomark. 2021, 30, 539–544. [Google Scholar] [CrossRef]
  128. Hurwitz, L.M.; Joshu, C.E.; Barber, J.R.; Prizment, A.E.; Vitolins, M.Z.; Jones, M.R.; Folsom, A.R.; Han, M.; Platz, E.A. Aspirin and non-aspirin NSAID use and prostate cancer incidence, mortality, and case fatality in the atherosclerosis risk in communities study. Cancer Epidemiol. Prev. Biomark. 2019, 28, 563–569. [Google Scholar] [CrossRef] [Green Version]
  129. Bitting, R.L.; Goodman, M.; George, D.J. Racial Disparity in Response to Prostate Cancer Systemic Therapies. Curr. Oncol. Rep. 2020, 22, 1–5. [Google Scholar] [CrossRef]
Figure 1. Key differences in the tumor immune–inflammation environment and the mutational spectrum between African American and European American men with prostate cancer. These differences open up potential vulnerabilities which preliminary studies have indicated could be exploited with treatment options, with some of them having demonstrated favorable responses in African American men.
Figure 1. Key differences in the tumor immune–inflammation environment and the mutational spectrum between African American and European American men with prostate cancer. These differences open up potential vulnerabilities which preliminary studies have indicated could be exploited with treatment options, with some of them having demonstrated favorable responses in African American men.
Cancers 13 02874 g001
Figure 2. Preliminary evidence for certain treatment response differences between African American (AA) and European American (EA) patients with metastatic castration-resistant prostate cancer (mCRPC). Current approved treatment modalities for mCRPC, how clinical responses may differ between the two patient groups, and where additional studies are warranted. For PARP inhibitor use, the pathologic role of germline variants of unknown significance in DNA repair genes that commonly occur in AA men needs to be investigated.
Figure 2. Preliminary evidence for certain treatment response differences between African American (AA) and European American (EA) patients with metastatic castration-resistant prostate cancer (mCRPC). Current approved treatment modalities for mCRPC, how clinical responses may differ between the two patient groups, and where additional studies are warranted. For PARP inhibitor use, the pathologic role of germline variants of unknown significance in DNA repair genes that commonly occur in AA men needs to be investigated.
Cancers 13 02874 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kiely, M.; Ambs, S. Immune Inflammation Pathways as Therapeutic Targets to Reduce Lethal Prostate Cancer in African American Men. Cancers 2021, 13, 2874. https://doi.org/10.3390/cancers13122874

AMA Style

Kiely M, Ambs S. Immune Inflammation Pathways as Therapeutic Targets to Reduce Lethal Prostate Cancer in African American Men. Cancers. 2021; 13(12):2874. https://doi.org/10.3390/cancers13122874

Chicago/Turabian Style

Kiely, Maeve, and Stefan Ambs. 2021. "Immune Inflammation Pathways as Therapeutic Targets to Reduce Lethal Prostate Cancer in African American Men" Cancers 13, no. 12: 2874. https://doi.org/10.3390/cancers13122874

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop