Comparative analysis of 1152 African-American and European-American men with prostate cancer identifies distinct genomic and immunological differences

Walter Rayford, Alp Tuna Beksac, Jordan Alger, Mohammed Alshalalfa, Mohsen Ahmed, Irtaza Khan, Ugo G Falagario, Yang Liu, Elai Davicioni, Daniel E Spratt, Edward M Schaeffer, Felix Y Feng, Brandon Mahal, Paul L Nguyen, Robert B Den, Mark D Greenberger, Randy Bradley, Justin M Watson, Matthew Beamer, Lambros Stamatakis, Darrell J Carmen, Shivanshu Awasthi, Jonathan Hwang, Rachel Weil, Harri Merisaari, Nihal Mohamed, Leslie A Deane, Dimple Chakravarty, Kamlesh K Yadav, Kosj Yamoah, Sujit S Nair, Ashutosh K Tewari, Walter Rayford, Alp Tuna Beksac, Jordan Alger, Mohammed Alshalalfa, Mohsen Ahmed, Irtaza Khan, Ugo G Falagario, Yang Liu, Elai Davicioni, Daniel E Spratt, Edward M Schaeffer, Felix Y Feng, Brandon Mahal, Paul L Nguyen, Robert B Den, Mark D Greenberger, Randy Bradley, Justin M Watson, Matthew Beamer, Lambros Stamatakis, Darrell J Carmen, Shivanshu Awasthi, Jonathan Hwang, Rachel Weil, Harri Merisaari, Nihal Mohamed, Leslie A Deane, Dimple Chakravarty, Kamlesh K Yadav, Kosj Yamoah, Sujit S Nair, Ashutosh K Tewari

Abstract

Racial disparities in prostate cancer have not been well characterized on a genomic level. Here we show the results of a multi-institutional retrospective analysis of 1,152 patients (596 African-American men (AAM) and 556 European-American men (EAM)) who underwent radical prostatectomy. Comparative analyses between the race groups were conducted at the clinical, genomic, pathway, molecular subtype, and prognostic levels. The EAM group had increased ERG (P < 0.001) and ETS (P = 0.02) expression, decreased SPINK1 expression (P < 0.001), and basal-like (P < 0.001) molecular subtypes. After adjusting for confounders, the AAM group was associated with higher expression of CRYBB2, GSTM3, and inflammation genes (IL33, IFNG, CCL4, CD3, ICOSLG), and lower expression of mismatch repair genes (MSH2, MSH6) (p < 0.001 for all). At the pathway level, the AAM group had higher expression of genes sets related to the immune response, apoptosis, hypoxia, and reactive oxygen species. EAM group was associated with higher levels of fatty acid metabolism, DNA repair, and WNT/beta-catenin signaling. Based on cell lines data, AAM were predicted to have higher potential response to DNA damage. In conclusion, biological characteristics of prostate tumor were substantially different in AAM when compared to EAM.

Trial registration: ClinicalTrials.gov NCT02609269.

Conflict of interest statement

The authors declare the following competing interests: Y.L. and E.D. are employees of Decipher Biosciences. R.B.D. has received research funding and has served as a consultant for Decipher Biosciences. P.L.N. has served as a consultant for Decipher Biosciences, Ferring, Bayer, Astellas Medivation, Dendreon, Blue Earth, Nanobiotix, Augmenix, and Infinity Pharmaceuticals. F.F.Y.F. is an employee of PFS genomics and has served as a consultant for Medivation/Astellas, Decipher, Celgene, Dendreon, EMD Serono, Janssen Oncology, Ferring, and Bayer. D.E.S. has served as a consultant for Dendreon. E.M.S. has served as a consultant for Decipher, OPKO Health, Abbvie. K.K.Y. is an employee of Sema4, a Mount Sinai venture company, and has received royalties from Inthera Bioscience. A.K.T. has served as a site-PI on pharma/industry-sponsored clinical trials from Kite Pharma, Lumicell Inc, Dendreon, and Oncovir Inc. He has received research funding (grants) to his institution from DOD, NIH, Axogen, Intuitive surgical, AMBFF, and other philanthropy. A.K.T. has served as an unpaid consultant to Roivant Biosciences and advisor to Promaxo. He owns equity in Promaxo.

Figures

Fig. 1. Comparison of genomic-risk signatures and…
Fig. 1. Comparison of genomic-risk signatures and their grade group associations in AAM and EAM.
a Prognostic differences in AAM vs. EAM. 20 prognostic signatures show similar trends in both groups. Overall, both Decipher (b) and average genomic-risk scores (c) positively correlate with grade groups in both races. b Decipher is higher in AAM with low grades but not different in high-grade tumors. c In contrast, average genomic risk is lower in the AAM with high-grade tumors. AAM African-American men, EAM European-American men. Error bars represent the 95% confidence interval.
Fig. 2. Genome-wide differential expression in AAM…
Fig. 2. Genome-wide differential expression in AAM and EAM.
a Top 22 genes differentially expressed between the two groups in our cohort with multivariate analysis logistic regression P value <1e−10. Mean difference in TCGA prostate was also shown for these genes to show that these genes show the same directionality. Genes were sorted based on the mean difference between AAM and EAM. b Boxplot of genes differentially expressed in TCGA prostate cohort. Abbreviations as in Fig. 1. Error bars represent the 95% confidence interval.
Fig. 3. Activation of different signaling pathways…
Fig. 3. Activation of different signaling pathways drives prostate cancer in AAM and EAM.
a The bar plot depicts the log odds of pathways that have a significant association with AAM and EAM after adjusting for clinical variables and false discovery. Many of the pathways more active in the AAM are related to the immune response. Pathways more active in the EAM include those related to DNA repair, glycolytic metabolism, and the cell cycle. b African Americans showed higher inflammation activity and higher expression of CD3, IFNG, IL33, and immune checkpoint inhibitors (PDL2, ICOSLG). Abbreviations as in Fig. 1. Error bars represent the 95% confidence interval.
Fig. 4. Differential expression of DNA repair…
Fig. 4. Differential expression of DNA repair pathways and genes.
a DNA repair activity and mismatch repair were calculated as mean of genes sets. Both showed lower activity in African Americans. b Key DNA repair genes (MSH2, RAD52, and PRKCD) were the most upregulated in patients. Error bars represent the 95% confidence interval.
Fig. 5. Tumor inflammation heterogeneity in AAM.
Fig. 5. Tumor inflammation heterogeneity in AAM.
Using a set of 124 inflammation and immune-response genes in AAM, consensus clustering revealed a cluster of patients with high expression of immune genes (CD3, CD45, GZMA, B2M, STAT1), nominating this cluster for immunotherapy treatment intervention. AAM African-American men, APF adverse pathology feature.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J. Clin. 2018;68:7–30. doi: 10.3322/caac.21442.
    1. Friedlander DF, et al. Racial disparity in delivering definitive therapy for intermediate/high-risk localized prostate cancer: the impact of facility features and socioeconomic characteristics. Eur. Urol. 2018;73:445–451. doi: 10.1016/j.eururo.2017.07.023.
    1. Palmer NR, et al. Disparities in barriers to follow-up care between African American and White breast cancer survivors. Support Care Cancer. 2015;23:3201–3209. doi: 10.1007/s00520-015-2706-9.
    1. Bhardwaj A, et al. Racial disparities in prostate cancer: a molecular perspective. Front. Biosci. 2017;22:772–782. doi: 10.2741/4515.
    1. Yuan J, 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. doi: 10.1371/journal.pgen.1008641.
    1. Karakas C, et al. Molecular mechanisms involving prostate cancer racial disparity. Am. J. Clin. Exp. Urol. 2017;5:34–48.
    1. Kumar S, Singh R, Malik S, Manne U, Mishra M. Prostate cancer health disparities: an immuno-biological perspective. Cancer Lett. 2018;414:153–165. doi: 10.1016/j.canlet.2017.11.011.
    1. Hatcher D, Daniels G, Osman I, Lee P. Molecular mechanisms involving prostate cancer racial disparity. Am. J. Transl. Res. 2009;1:235–248.
    1. Echevarria, M. I. et al. African American-specific gene panel predictive of poor prostate cancer outcome. J. Urol.10.1097/JU.0000000000000193 (2019).
    1. Yamoah K, et al. Novel biomarker signature that may predict aggressive disease in African American men with prostate cancer. J. Clin. Oncol. 2015;33:2789–2796. doi: 10.1200/jco.2014.59.8912.
    1. Ross AE, et al. Tissue-based genomics augments post-prostatectomy risk stratification in a natural history cohort of intermediate- and high-risk men. Eur. Urol. 2016;69:157–165. doi: 10.1016/j.eururo.2015.05.042.
    1. Zhao SG, et al. Associations of Luminal and basal subtyping of prostate cancer with prognosis and response to androgen deprivation therapy. JAMA Oncol. 2017;3:1663–1672. doi: 10.1001/jamaoncol.2017.0751.
    1. Field LA, et al. Identification of differentially expressed genes in breast tumors from African American compared with Caucasian women. Cancer. 2012;118:1334–1344. doi: 10.1002/cncr.26405.
    1. Jovov B, et al. Differential gene expression between African American and European American colorectal cancer patients. PLoS One. 2012;7:e30168. doi: 10.1371/journal.pone.0030168.
    1. Zhao SG, et al. Development and validation of a 24-gene predictor of response to postoperative radiotherapy in prostate cancer: a matched, retrospective analysis. Lancet Oncol. 2016;17:1612–1620. doi: 10.1016/S1470-2045(16)30491-0.
    1. Kim W, et al. Luminal and basal subtyping of metastatic castration-resistant prostate cancer (mCRPC) and its clinical implications. J. Clin. Oncol. 2018;36:197–197. doi: 10.1200/JCO.2018.36.6_suppl.197.
    1. Reinhold WC, et al. CellMiner: a web-based suite of genomic and pharmacologic tools to explore transcript and drug patterns in the NCI-60 cell line set. Cancer Res. 2012;72:3499–3511. doi: 10.1158/0008-5472.CAN-12-1370.
    1. Petrelli F, et al. Predictive factors for reclassification and relapse in prostate cancer eligible for active surveillance: a systematic review and meta-analysis. Urology. 2016;91:136–142. doi: 10.1016/j.urology.2016.01.034.
    1. Faisal FA, et al. SPINK1 expression is enriched in African American prostate cancer but is not associated with altered immune infiltration or oncologic outcomes post-prostatectomy. Prostate Cancer Prostatic Dis. 2019;22:552–559. doi: 10.1038/s41391-019-0139-0.
    1. Tomlins SA, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–648. doi: 10.1126/science.1117679.
    1. Taris M, et al. ERG expression in prostate cancer: the prognostic paradox. Prostate. 2014;74:1481–1487. doi: 10.1002/pros.22863.
    1. Johnson MH, et al. SPINK1 defines a molecular subtype of prostate cancer in men with more rapid progression in an at risk, natural history radical prostatectomy cohort. J. Urol. 2016;196:1436–1444. doi: 10.1016/j.juro.2016.05.092.
    1. Khani F, et al. Evidence for molecular differences in prostate cancer between African American and Caucasian men. Clin. Cancer Res. 2014;20:4925–4934. doi: 10.1158/1078-0432.CCR-13-2265.
    1. Cornford P, et al. EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur. Urol. 2017;71:630–642. doi: 10.1016/j.eururo.2016.08.002.
    1. Higano CS, et al. Real-world outcomes of sipuleucel-T treatment in PROCEED, a prospective registry of men with metastatic castration-resistant prostate cancer. Cancer. 2019;125:4172–4180. doi: 10.1002/cncr.32445.
    1. Spratt D, et al. Androgen receptor activity and radiotherapeutic sensitivity in African-American men with prostate cancer: a Large Scale Gene Expression Analysis and Meta-analysis of RTOG Trials. Int. J. Radiat. Oncol.• Biol.• Phys. 2018;102:S3. doi: 10.1016/j.ijrobp.2018.06.104.
    1. Spratt DE, et al. Transcriptomic heterogeneity of androgen receptor activity defines a de novo low AR-active subclass in treatment naive primary prostate cancer. Clin. Cancer Res. 2019;25:6721–6730. doi: 10.1158/1078-0432.Ccr-19-1587.
    1. Gandaglia G, et al. Adjuvant and salvage radiotherapy after radical prostatectomy in prostate cancer patients. Eur. Urol. 2017;72:689–709. doi: 10.1016/j.eururo.2017.01.039.
    1. Dalela D, et al. Genomic classifier augments the role of pathological features in identifying optimal candidates for adjuvant radiation therapy in patients with prostate cancer: development and internal validation of a multivariable prognostic model. J. Clin. Oncol. 2017;35:1982–1990. doi: 10.1200/JCO.2016.69.9918.
    1. Gray PJ, Lin CC, Cooperberg MR, Jemal A, Efstathiou JA. Temporal trends and the impact of race, insurance, and socioeconomic status in the management of localized prostate cancer. Eur. Urol. 2017;71:729–737. doi: 10.1016/j.eururo.2016.08.047.
    1. Cooperberg MR, et al. The diverse genomic landscape of clinically low-risk prostate cancer. Eur. Urol. 2018;74:444–452. doi: 10.1016/j.eururo.2018.05.014.
    1. Epstein JI, et al. The 2014 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma: definition of grading patterns and proposal for a new grading system. Am. J. Surg. Pathol. 2016;40:244–252. doi: 10.1097/PAS.0000000000000530.
    1. Cooperberg MR, Hilton JF, Carroll PR. The CAPRA-S score: a straightforward tool for improved prediction of outcomes after radical prostatectomy. Cancer. 2011;117:5039–5046. doi: 10.1002/cncr.26169.
    1. Den RB, et al. Decipher correlation patterns post prostatectomy: initial experience from 2 342 prospective patients. Prostate Cancer Prostatic Dis. 2016;19:374–379. doi: 10.1038/pcan.2016.38.
    1. Lockstone HE. Exon array data analysis using Affymetrix power tools and R statistical software. Brief. Bioinform. 2011;12:634–644. doi: 10.1093/bib/bbq086.
    1. Piccolo SR, et al. A single-sample microarray normalization method to facilitate personalized-medicine workflows. Genomics. 2012;100:337–344. doi: 10.1016/j.ygeno.2012.08.003.
    1. Nair SS, Li DQ, Kumar R. A core chromatin remodeling factor instructs global chromatin signaling through multivalent reading of nucleosome codes. Mol. Cell. 2013;49:704–718. doi: 10.1016/j.molcel.2012.12.016.
    1. Chakravarty D, et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat. Commun. 2014;5:5383. doi: 10.1038/ncomms6383.
    1. Liberzon A, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425. doi: 10.1016/j.cels.2015.12.004.
    1. Kishan AU, et al. Transcriptomic heterogeneity of Gleason grade group 5 prostate cancer. Eur. Urol. 2020;78:327–332. doi: 10.1016/j.eururo.2020.05.009.
    1. Kuleshov MV, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44:W90–W97. doi: 10.1093/nar/gkw377.

Source: PubMed

3
Subscribe