Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2079 word(s) 2079 2021-06-17 08:52:05 |
2 format correct -55 word(s) 2024 2021-07-14 05:29:52 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Bailey-Whyte, M. Targeting Inflammation in Prostate Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/11837 (accessed on 23 April 2024).
Bailey-Whyte M. Targeting Inflammation in Prostate Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/11837. Accessed April 23, 2024.
Bailey-Whyte, Maeve. "Targeting Inflammation in Prostate Cancer" Encyclopedia, https://encyclopedia.pub/entry/11837 (accessed April 23, 2024).
Bailey-Whyte, M. (2021, July 08). Targeting Inflammation in Prostate Cancer. In Encyclopedia. https://encyclopedia.pub/entry/11837
Bailey-Whyte, Maeve. "Targeting Inflammation in Prostate Cancer." Encyclopedia. Web. 08 July, 2021.
Targeting Inflammation in Prostate Cancer
Edit

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.

prostate cancer African American inflammation health disparity

1. Introduction

Despite substantial improvements in cancer survival overall, men of African descent continue to have about double the rate of fatal prostate cancer compared to other men[1][2]. Prostate cancer disparities are largely explained by health care disparities, lifestyle factors and disparate exposures to carcinogens but even when these are accounted for - some of the cancer disparities persist. Ancestral factors and disparate exposures can lead to altered tumor biology, resulting in a distinct disease etiology by population group.

 A growing body of evidence indicates a distinct role for inflammation and the immune response in prostate cancer disparities. Low-grade chronic inflammation is more prevalent in African American patients and has been associated with adverse outcomes[3][4]. An immune-inflammation signature more prevalent in prostate tumors of African American patients is central to the hypothesis that inflammation is a candidate driver of prostate cancer disparities[5]. This signature includes up-regulation of genes in the interferon signaling pathway corresponding to a previously described “interferon-related DNA damage resistance signature”, which has been linked to acquired resistance to radiation and chemotherapy in breast cancer[6][7]. 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. Multiple studies reported upregulation of inflammatory mediators in the tumor microenvironment of African American prostate cancer patients, many of which have implications for disease prognosis[8][9][10][11][12]. This review aims to introduce the concept that this inflammation and immune response present immunogenic vulnerabilities that could be exploited to address the adverse outcomes experienced by the African American population with prostate cancer.

2. African American men may have a differential response to certain therapies for metastatic prostate cancer.

There is now some evidence that men of African descent may respond differently to 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, median overall survival is still only around 3 years[13]. 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.

3. Radiation

Radium-223 is an approved therapeutic option for mCRPC patients with symptomatic bone metastases. Zhao et al examined response to radium-223 treatment in men from a Veteran Affairs cohort with mCRPC[14]. 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[15][16][17]. 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, it can be speculated that inactivating mutations that decrease the DNA damage repair capacity in tumors from African American may contribute to the positive outcomes post treatment with radium-223[9].

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[18]. Now that studies have shown better responses from African American men treated with radium-223 and Sipuleucel-T separately[14][19], a planned larger trial may inform on whether African American men may benefit synergistically from this combination approach.

4. Immunotherapy

Immunotherapy has not been as successful in treating prostate cancer as with other hematologic or solid cancers and clinical trials show modest[20] to no effect[13][21][22] 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[6][9]. Studies have shown that young people who self-report as African American mounted an increased immune response to vaccination[23][24]. 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[19]. 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[25]. Mechanistically, evidence points towards a complex interplay of immune cells with tumor biology which may predict prognosis and response to therapy. But 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[22][26]. This has been attributed in part to relatively low PD-L1 expression from tumor cells[27][28]. However, this is not consistent across literature with studies also reporting increased PD-L1 expression and association with  biochemical recurrence[29] and shorter metastasis free survival[30]. 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[30]. 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[31]. However, clinical trials investigating the effect of PD-1 inhibition reported no significant clinical benefit. Yet, they have typically not included men of African descent[22].

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[32]. African American ethnicity and an aggressive cancer phenotype has been associated with prediction of tumor PD-L1 positivity in hormone naïve tumors[33], suggesting a potential benefit for immunotherapy in African Americans at high risk of aggressive disease but this has not been replicated yet[9]. 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[34]. 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.

5. 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 and a higher prevalence of comorbidities preventing access to trials[35][36][37]. 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[38]. With just 6% of African American participants, they report 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 significant worse performance status, higher testosterone levels, higher PSA levels, and lower hemoglobin levels.

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[9][39][40][41]. 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[9].

Altered DNA damage repair pathways may also 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)[42]. Mechanistically, in prostate cancer, the stimulator of IFN genes (STING) pathway has been linked to recruitment and activation of interferon-related genes in vitro, increasing sensitivity to the immune checkpoint inhibitor PD-L1 in DNA repair deficient tumors[43][44][45][46]. As a low DNA repair capacity may increase tumor genomic instability and tumor mutational burden, this again, might constitute as a vulnerability to immunotherapeutic strategies[47]. It has been suggested that enhancement of this genomic instability through use of radiation, chemotherapy or PARP inhibitors could augment the immunotherapeutic response[42][48][49].

6. Anti-inflammatory drug aspirin for prevention of adverse outcomes in African American men with prostate cancer

The United States Preventative Services Task Force now recommend the anti-inflammatory drug aspirin for prevention of colorectal cancer for an albeit narrow category of adults[50][51][52]. 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[53]. The finding is consistent with a similar observation in a previous study[54]. 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[55]. 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, 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 (article under review). 

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[56]. 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[57], again pointing to the potential benefit of aspirin use for men at high risk of fatal prostate cancer.

7. Conclusion

Elevated inflammatory processes in African American men with prostate cancer are a candidate biological drivers of disparate disease risks. 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 like men of African ancestry.

References

  1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer Statistics, 2021. CA Cancer J Clin 2021;71(1):7-33.
  2. Butler EN, Kelly SP, Coupland VH, et al. Fatal prostate cancer incidence trends in the United States and England by race, stage, and treatment. British Journal of Cancer 2020:1-8.
  3. Gurel B, Lucia MS, Thompson IM, Jr., 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 Biomarkers Prev 2014;23(5):847-56.
  4. Klink JC, Banez LL, Gerber L, et al. Intratumoral inflammation is associated with more aggressive prostate cancer. World J Urol 2013;31(6):1497-503.
  5. Wallace TA, Prueitt RL, Yi M, et al. Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res. 2008;68(3):927-936.
  6. Tang W, Wallace TA, Yi 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(21):5471-5481.
  7. Weichselbaum RR, Ishwaran H, Yoon T, et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proceedings of the National Academy of Sciences 2008;105(47):18490-18495.
  8. Gillard M, Javier R, Ji Y, et al. Elevation of stromal-derived mediators of inflammation promote prostate cancer progression in African-American men. Cancer research 2018;78(21):6134-6145.
  9. Awasthi S, Berglund A, Abraham-Miranda J, et al. Comparative genomics reveals distinct immune-oncologic pathways in African American men with prostate cancer. Clinical Cancer Research 2021;27(1):320-329.
  10. Nonomura N, Takayama H, Nakayama M, et al. Infiltration of tumour‐associated macrophages in prostate biopsy specimens is predictive of disease progression after hormonal therapy for prostate cancer. BJU international 2011;107(12):1918-1922.
  11. Weiner AB, Vidotto T, Liu Y, et al. Plasma cells are enriched in localized prostate cancer in Black men and are associated with improved outcomes. Nature communications 2021;12(1):1-10.
  12. Kinseth MA, Jia Z, Rahmatpanah F, et al. Expression differences between African American and Caucasian prostate cancer tissue reveals that stroma is the site of aggressive changes. International journal of cancer 2014;134(1):81-91.
  13. Beer TM, Kwon ED, Drake CG, 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(1):40-47.
  14. Zhao H, Howard LE, De Hoedt A, et al. Racial Discrepancies in Overall Survival among Men Treated with 223Radium. The Journal of urology 2020;203(2):331-337.
  15. van der Doelen MJ, Velho PI, Slootbeek PH, et al. Impact of DNA damage repair defects on response to radium-223 and overall survival in metastatic castration-resistant prostate cancer. European Journal of Cancer 2020;136:16-24.
  16. Ramos JD, Mostaghel EA, Pritchard CC, et al. DNA repair pathway alterations in metastatic castration-resistant prostate cancer responders to radium-223. Clinical genitourinary cancer 2018;16(2):106-110.
  17. Steinberger AE, Cotogno P, Ledet EM, et al. Exceptional duration of radium-223 in prostate cancer with a BRCA2 mutation. Clinical genitourinary cancer 2017;15(1):e69-e71.
  18. Marshall CH, Fu W, Wang H, et al. Randomized Phase II Trial of Sipuleucel-T with or without Radium-223 in Men with Bone-metastatic Castration-resistant Prostate Cancer. Clinical Cancer Research 2021;27(6):1623-1630.
  19. Sartor O, Armstrong AJ, Ahaghotu C, et al. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis 2020;23(3):517-526.
  20. Antonarakis ES, Piulats JM, Gross-Goupil M, et al. Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. Journal of Clinical Oncology 2020;38(5):395.
  21. Kwon ED, Drake CG, Scher HI, et al. 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. The lancet oncology 2014;15(7):700-712.
  22. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti–programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. Journal of clinical oncology 2010;28(19):3167.
  23. Haralambieva IH, Salk HM, Lambert ND, et al. Associations between race, sex and immune response variations to rubella vaccination in two independent cohorts. Vaccine 2014;32(17):1946-1953.
  24. Kurupati R, Kossenkov A, Haut L, et al. 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(39):62898.
  25. Zhao SG, Lehrer J, Chang SL, et al. The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. JNCI: Journal of the National Cancer Institute 2019;111(3):301-310.
  26. Topalian SL, Sznol M, Brahmer JR, et al. Nivolumab (anti-PD-1; BMS-936558; ONO-4538) in patients with advanced solid tumors: Survival and long-term safety in a phase I trial. In: American Society of Clinical Oncology; 2013, 3002-3002.
  27. Haffner MC, Guner G, Taheri D, et al. Comprehensive evaluation of programmed death-ligand 1 expression in primary and metastatic prostate cancer. The American journal of pathology 2018;188(6):1478-1485.
  28. Fankhauser CD, Schüffler PJ, Gillessen S, et al. Comprehensive immunohistochemical analysis of PD-L1 shows scarce expression in castration-resistant prostate cancer. Oncotarget 2018;9(12):10284.
  29. Li H, Wang Z, Zhang Y, et al. The immune checkpoint regulator PDL1 is an independent prognostic biomarker for biochemical recurrence in prostate cancer patients following adjuvant hormonal therapy. Journal of Cancer 2019;10(14):3102.
  30. Petitprez F, Fossati N, Vano Y, et al. PD-L1 expression and CD8+ T-cell infiltrate are associated with clinical progression in patients with node-positive prostate cancer. European urology focus 2019;5(2):192-196.
  31. Ness N, Andersen S, Khanehkenari MR, 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(16):26789-26801.
  32. Bishop JL, Sio A, Angeles A, et al. PD-L1 is highly expressed in Enzalutamide resistant prostate cancer. Oncotarget 2015;6(1):234.
  33. Calagua C, Russo J, Sun Y, 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(22):6812-6822.
  34. McNamara MA, George DJ, Ramaswamy K, et al. Overall survival by race in chemotherapy-naïve metastatic castration-resistant prostate cancer (mCRPC) patients treated with abiraterone acetate or enzalutamide. Journal of Clinical Oncology 2019;37(7_suppl):212-212.
  35. Duma N, Vera Aguilera J, Paludo J, et al. Representation of minorities and women in oncology clinical trials: review of the past 14 years. Journal of oncology practice 2018;14(1):e1-e10.
  36. Loree JM, Anand S, Dasari A, et al. Disparity of race reporting and representation in clinical trials leading to cancer drug approvals from 2008 to 2018. Jama Oncology 2019;5(10):e191870-e191870.
  37. Brandon DT, Isaac LA, LaVeist TA. The legacy of Tuskegee and trust in medical care: is Tuskegee responsible for race differences in mistrust of medical care? Journal of the National Medical Association 2005;97(7):951.
  38. Halabi S, Dutta S, Tangen CM, et al. Overall Survival of Black and White Men With Metastatic Castration-Resistant Prostate Cancer Treated With Docetaxel. J Clin Oncol 2019;37(5):403-410.
  39. Fantus RJ, Helfand BT. Germline Genetics of Prostate Cancer: Time to Incorporate Genetics into Early Detection Tools. Clinical Chemistry 2019;65(1):74-79.
  40. Pritchard CC, Mateo J, Walsh MF, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med 2016;375:443-453.
  41. Yadav S, Anbalagan M, Baddoo M, et al. Somatic mutations in the DNA repairome in prostate cancers in African Americans and Caucasians. Oncogene 2020;39(21):4299-4311.
  42. Bever KM, Le DT. DNA repair defects and implications for immunotherapy. The Journal of clinical investigation 2018;128(10):4236-4242.
  43. Wang H, Hu S, Chen X, et al. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proceedings of the National Academy of Sciences 2017;114(7):1637-1642.
  44. Härtlova A, Erttmann SF, Raffi FA, et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 2015;42(2):332-343.
  45. Morel KL, Sheahan AV, Burkhart DL, et al. EZH2 inhibition activates a dsRNA–STING–interferon stress axis that potentiates response to PD-1 checkpoint blockade in prostate cancer. Nature Cancer 2021:1-13.
  46. Esteves AM, Papaevangelou E, Dasgupta P, et al. Combination of Interleukin-15 with a STING agonist, ADU-S100 analog: a potential immunotherapy for prostate cancer. Frontiers in Oncology 2021;11.
  47. Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome medicine 2017;9(1):1-14.
  48. Gupta PD, Chaudagar K, Sharma-Saha S, et al. PARP and PI3K inhibitor combination therapy eradicates c-MYC-driven murine prostate cancers via cGAS/STING pathway activation within tumor-associated macrophages. bioRxiv 2020.
  49. Chabanon RM, Muirhead G, Krastev DB, et al. PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer. The Journal of clinical investigation 2019;129(3):1211-1228.
  50. Chubak J, Whitlock EP, Williams SB, et al. Aspirin for the prevention of cancer incidence and mortality: systematic evidence reviews for the US Preventive Services Task Force. Annals of internal medicine 2016;164(12):814-825.
  51. Bibbins-Domingo K. Aspirin use for the primary prevention of cardiovascular disease and colorectal cancer: US Preventive Services Task Force recommendation statement. Annals of internal medicine 2016;164(12):836-845.
  52. Dehmer SP, Maciosek MV, Flottemesch TJ, et al. Aspirin for the primary prevention of cardiovascular disease and colorectal cancer: a decision analysis for the US Preventive Services Task Force. Annals of internal medicine 2016;164(12):777-786.
  53. Smith CJ, Dorsey TH, Tang W, et al. Aspirin Use Reduces the Risk of Aggressive Prostate Cancer and Disease Recurrence in African-American Men. Cancer Epidemiol Biomarkers Prev 2017;26(6):845-853.
  54. Osborn VW, Chen SC, Weiner J, et al. Impact of aspirin on clinical outcomes for African American men with prostate cancer undergoing radiation. Tumori 2016;102(1):65-70.
  55. Lucotti S, Cerutti C, Soyer M, et al. Aspirin blocks formation of metastatic intravascular niches by inhibiting platelet-derived COX-1/thromboxane A 2. The Journal of clinical investigation 2019;129(5):1845-1862.
  56. Tang W, Fowke JH, Hurwitz LM, et al. Aspirin Use and Prostate Cancer among African-American Men in the Southern Community Cohort Study. Cancer Epidemiology and Prevention Biomarkers 2021;30(3):539-544.
  57. Hurwitz LM, Joshu CE, Barber JR, et al. Aspirin and non-aspirin NSAID use and prostate cancer incidence, mortality, and case fatality in the atherosclerosis risk in communities study. Cancer Epidemiology and Prevention Biomarkers 2019;28(3):563-569.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 304
Revisions: 2 times (View History)
Update Date: 14 Jul 2021
1000/1000