MYC drives aggressive prostate cancer by disrupting transcriptional pause release at androgen receptor targets

Xintao Qiu, Nadia Boufaied, Tarek Hallal, Avery Feit, Anna de Polo, Adrienne M Luoma, Walaa Alahmadi, Janie Larocque, Giorgia Zadra, Yingtian Xie, Shengqing Gu, Qin Tang, Yi Zhang, Sudeepa Syamala, Ji-Heui Seo, Connor Bell, Edward O'Connor, Yang Liu, Edward M Schaeffer, R Jeffrey Karnes, Sheila Weinmann, Elai Davicioni, Colm Morrissey, Paloma Cejas, Leigh Ellis, Massimo Loda, Kai W Wucherpfennig, Mark M Pomerantz, Daniel E Spratt, Eva Corey, Matthew L Freedman, X Shirley Liu, Myles Brown, Henry W Long, David P Labbé, Xintao Qiu, Nadia Boufaied, Tarek Hallal, Avery Feit, Anna de Polo, Adrienne M Luoma, Walaa Alahmadi, Janie Larocque, Giorgia Zadra, Yingtian Xie, Shengqing Gu, Qin Tang, Yi Zhang, Sudeepa Syamala, Ji-Heui Seo, Connor Bell, Edward O'Connor, Yang Liu, Edward M Schaeffer, R Jeffrey Karnes, Sheila Weinmann, Elai Davicioni, Colm Morrissey, Paloma Cejas, Leigh Ellis, Massimo Loda, Kai W Wucherpfennig, Mark M Pomerantz, Daniel E Spratt, Eva Corey, Matthew L Freedman, X Shirley Liu, Myles Brown, Henry W Long, David P Labbé

Abstract

c-MYC (MYC) is a major driver of prostate cancer tumorigenesis and progression. Although MYC is overexpressed in both early and metastatic disease and associated with poor survival, its impact on prostate transcriptional reprogramming remains elusive. We demonstrate that MYC overexpression significantly diminishes the androgen receptor (AR) transcriptional program (the set of genes directly targeted by the AR protein) in luminal prostate cells without altering AR expression. Analyses of clinical specimens reveal that concurrent low AR and high MYC transcriptional programs accelerate prostate cancer progression toward a metastatic, castration-resistant disease. Data integration of single-cell transcriptomics together with ChIP-seq uncover an increase in RNA polymerase II (Pol II) promoter-proximal pausing at AR-dependent genes following MYC overexpression without an accompanying deactivation of AR-bound enhancers. Altogether, our findings suggest that MYC overexpression antagonizes the canonical AR transcriptional program and contributes to prostate tumor initiation and progression by disrupting transcriptional pause release at AR-regulated genes.

Conflict of interest statement

R.J.K. receive royalties from GenomeDx (now Veracyte) for Decipher testing. S.W. receives research funding from PreludeDX. K.W.W. serves on the scientific advisory board of T-Scan Therapeutics, SQZ Biotech, Nextechinvest and receives sponsored research funding from Novartis. He is a co-founder of Immunitas, a biotech company. These activities are not related to the research reported in this publication. D.E.S. receives personal fees from Janssen, AstraZeneca, and Blue Earth and funding from Janssen. E.C. received research funding under institutional SRA from Janssen Research and Development, Bayer Pharmaceuticals, KronosBio, Forma Pharmaceutics, Foghorn, Gilead, Sanofi, AbbVie, MacroGenics, and GSK. M.L.F. reports other support from Nuscan Diagnostics outside the submitted work. X.S.L. conducted the work while being a faculty at the Dana-Farber Cancer Institute and is currently a board member and CEO of GV20. M.B. and H.W.L. receives sponsored research support from Novartis. M.B. is a consultant to Aleta Biotherapeutics and H3 Biomedicine and serves on the SAB of Kronos Bio. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. MYC induces a profound transcriptional…
Fig. 1. MYC induces a profound transcriptional reprogramming in murine prostate lobes.
a Graphical summary of the experimental design. b, c Transcriptional profiling of WT and MYC-transformed VP reveal high concordance for the total number of genes detected (b) and their expression levels (c) between bulk and single-cell RNA-seq (VP; matched bulk and single-cell RNA-seq; n = 1 per genotype). d, e Sample-sample correlation (d) and principal component analysis (e) between bulk and matched single-cell transcriptome identifies distinct transcriptional profiles across murine prostate lobes (AP, DLP, VP; matched bulk and single-cell RNA-seq; n = 1 per genotype). f Single-cell census of WT and MYC-transformed AP, DLP and VP. tSNE of scRNA-seq profiles colored using known markers identified nine major subpopulations across prostate lobes (AP, DLP, VP; n = 1 per genotype). gi The human MYC transgene (hg19MYC) expression is largely restricted to the luminal compartment (g AP, DLP, VP; n = 1 per genotype) and predominantly expressed in the VP (h Source data are provided as a Source Data file), in accordance with the penetrance of prostatic intraepithelial neoplasia (i PIN; n = biologically independent animals; mean ± SD; Source data are provided as a Source Data file). WT: wild-type; VP: ventral prostate; DLP: dorsolateral prostate; AP: anterior prostate.
Fig. 2. Single-cell transcriptome reveals distinct luminal…
Fig. 2. Single-cell transcriptome reveals distinct luminal cell subpopulations.
a, b Single cell census of the WT and MYC-transformed VP (a) followed by unsupervised clustering revealed four luminal subsets (b VP; n = 1 per genotype). c, d Human MYC transcript (hg19MYC) is only observed in MYC-transformed VP and mostly restricted to the luminal subsets while murine Myc transcript (mm10Myc) is expressed across cellular populations and genotypes (c VP; n = 1 per genotype) and is not correlated with hg19MYC expression in luminal cells (d VP; n = 1 per genotype; edgeR: two-sided quasi-likelihood F-test).
Fig. 3. MYC-driven luminal cells transformation dampens…
Fig. 3. MYC-driven luminal cells transformation dampens the AR transcriptional program.
a Gene Set Enrichment Analysis (GSEA, Hallmark, P < 0.05 and FDR < 0.1) revealed that the bulk RNA-seq transcriptional program associated with MYC overexpression is mostly driven by the luminal subset (VP; matched bulk and single-cell RNA-seq; n = 1 per genotype; Source data are provided as a Source Data file). b, c MYC overexpression is associated with an enriched MYC transcriptional program (bP < 0.001 and FDR < 0.001) and a depleted AR response (cP < 0.016 and FDR < 0.040) in the luminal subset (GSEA; VP; n = 1 per genotype). d, e MYC overexpression does not alter AR transcript expression in the luminal compartment (d VP; n = 1 per genotype; edgeR: two-sided quasi-likelihood F-test) and protein levels in the VP (e VP; n = 3 per genotype; numbers at the bottom represent AR levels relative to β-Actin; Source data are provided as a Source Data file). f Schematic representation of covariance analysis to determine co-expression (i.e. positive covariance) or mutually exclusive expression (i.e. negative covariance) between two genes at a single cell level. g Covariance analysis in the luminal subset reveals a shift from canonical AR target genes in the transcripts co-expressed with Ar upon MYC overexpression (VP; n = 1 per genotype). NES: normalized enrichment score; ES: enrichment score.
Fig. 4. MYC overexpression alters the AR…
Fig. 4. MYC overexpression alters the AR cistrome.
a AR ChIP-seq identifies an androgen response element (ARE) as the top AR binding motif in WT and MYC-transformed VP (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). b Unsupervised pairwise correlation of the murine AR cistrome from all specimens (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). c MYC overexpression expands the AR cistrome as demonstrated by the heatmaps indicating AR binding intensity across 4 kb intervals (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). d Motif analysis of MYC-associated AR gained sites reveal forkhead response element (FHRE) and androgen response element (ARE; VP; n = 2 pools of biological replicates (n = 8–13) per genotype). e, f AR gained sites are characterized by increased FOXA1 binding (e) and H3K27ac mark (f) in MYC-transformed VP (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). g Integration of the 1695 AR bindings sites gained in MYC tumors with luminal single cell transcriptome grouped by k-means clustering (n = 3 clusters). h GSEA analysis (Hallmark) revealed an enforced MYC transcriptional program (Cluster 1) and a diminished androgen response (Cluster 2) associated to MYC-dependent AR gained binding sites (Source data are provided as a Source Data file).
Fig. 5. Divergent MYC and AR transcriptional…
Fig. 5. Divergent MYC and AR transcriptional programs dictate disease progression.
a, b Kaplan–Meier curves (a) and log-rank tests (b) reveal that patients bearing a primary tumor characterized by low AR-activity (AR-A) and concurrent high MYC transcriptional signature (Hallmark) have a shorter time to biochemical recurrence (BCR) within the discovery cohort (TCGA). c, d Kaplan–Meier curves (c), univariable and multivariable analysis (d Cox proportional hazards model) confirms that tumors with concurrent low AR-A and high MYC transcriptional signatures develop BCR after radical prostatectomy more rapidly than low AR-A tumors without an active MYC transcriptional program in the validation cohort (Spratt et al., 2017; n = 855; HR ± 95% CI). e, f Kaplan–Meier curves (e), univariable and multivariable analyses (f Cox proportional hazards model) reveal that tumors with concurrent low AR-A and high MYC transcriptional signatures are more likely to develop a metastatic disease (n = 855; HR ± 95% CI). PSA: prostate-specific antigen; HR: hazard ratio; CI: confidence interval; GS: Gleason score; ECE: extracapsular extension; SVI: seminal vesicles invasion; LNI: lymph node involvement.
Fig. 6. High MYC expression is associated…
Fig. 6. High MYC expression is associated with a dampened AR transcriptional program and resistance to AR signaling inhibitors in castration-resistant tumors.
a, b AR activity is inversely correlated with MYC expression in CRPC clinical samples (a Pearson correlation coefficient (ρ) and P value; linear regression ± 95% CI; Source data are provided as a Source Data file) and significantly lower in MYC-high tumors (b Two-way ANNOVA followed by a Tukey–Kramer test; median; box boundaries: 25th and 75th percentiles; whiskers: ± lowest/smallest value no further than 1.5 interquartile range; n = 59; Source data are provided as a Source Data file). c Gene Set Enrichment Analysis (GSEA, Hallmark, P < 0.05 and FDR < 0.1) revealed an enriched MYC transcriptional program (P < 0.001 and FDR < 0.001) and a depleted AR response (P < 0.001 and FDR < 0.001) in MYC-high CRPC (Source data are provided as a Source Data file). d, eMYC-high mCRPC LuCaP patient-derived xenografts (PDXs) have similar levels of AR (d Wilcoxon rank-sum test; n = 8 biologically independent animals; median, whiskers ± min to max; Source data are provided as a Source Data file) but are associated with an expanded AR cistrome as demonstrated by the increased binding intensity across 4 kb intervals at AR gained sites (e). f Motif analysis of MYC-associated AR gained sites reveal ARE and FHRE. g, h AR gained sites are characterized by increased FOXA1 binding (g) and H3K27ac mark (h) in MYC-high mCRPC LuCaP. i AR cistrome in MYC-high mCRPC LuCaP PDXs is associated with a diminished androgen response (GSEA; P < 0.001 and FDR < 0.001). j, k Kaplan-Meier curves (j) and univariable analysis (k Cox proportional hazards model) revealed that patients with mCRPC tumors harboring an AR_low/MYC_high signature are more likely to resist ARSI treatment and die of their disease (n = 75; HR ± 95% CI). HR: hazard ratio; CI: confidence interval; NES: normalized enrichment score; ES: enrichment score.
Fig. 7. MYC overexpression disrupts the AR…
Fig. 7. MYC overexpression disrupts the AR transcriptional program by pausing AR regulated genes.
a BETA analysis revealed that AR binding sites are associated with gene downregulation following MYC overexpression. b, c Despite a dampened AR transcriptional program, higher levels of the AR binding (b) and H3K27ac mark (c) are observed nearby AR response genes (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). d, e AR, FOXA1 and H3K27ac tracks at Pbsn locus, an AR-dependent gene, reveal unchanged or heightened AR and FOXA1 binding (d VP; n = 2 pools of biological replicates (n = 8–13) per genotype) albeit decreased transcript level (e VP; n = 1 per genotype; edgeR: two-sided quasi-likelihood F-test) following MYC overexpression. f, g Unchanged AR and FOXA1 binding and H3K27ac mark at Mmsb locus (f VP; n = 2 pools of biological replicates (n = 8–13) per genotype), an AR-dependent gene downregulated by MYC overexpression (g VP; n = 1 per genotype; edgeR: two-sided quasi-likelihood F-test). h RNA Pol II traveling ratio differences following MYC overexpression in murine VP (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). i, j Pause release genes following MYC overexpression are characterized by greater RNA Pol II occupancy at gene body (i VP; n = 2 pools of biological replicates (n = 8–13) per genotype) and are enriched for MYC transcriptional signatures (j GSEA, Hallmark, P < 0.05 and FDR < 0.1; Source data are provided as a Source Data file). k, l Pause genes following MYC overexpression are characterized by greater promoter-proximal RNA Pol II occupancy (k VP; n = 2 pools of biological replicates (n = 8–13) per genotype) and are enriched for AR transcriptional signature (l GSEA, Hallmark, P < 0.05 and FDR < 0.1; Source data are provided as a Source Data file). m, n Increased RNA Pol II occupancy at the promoter of Pbsn (m) and decreased occupancy at the gene body of Msmb (n) following MYC overexpression (VP; n = 2 pools of biological replicates (n = 8–13) per genotype). TSS: transcription start site; TES: transcription end site.
Fig. 8. MYC disrupts transcriptional pause release…
Fig. 8. MYC disrupts transcriptional pause release at androgen receptor targets.
a RNA Pol II traveling ratio reveals greater promoter-proximal pausing at Androgen_response genes (two-tailed t-test). b Graphical summary. TSS: transcription start site; TES: transcription end site; BCR: biochemical recurrence; CRPC: castration resistant prostate cancer; FHRE: forkhead response elements; ARE: androgen response elements.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J. Clin. 2019;69:7–34.
    1. White JW., II The present position of the surgery of the hypertrophied prostate. Ann. Surg. 1893;18:152–188. doi: 10.1097/00000658-189307000-00020.
    1. White JWI. The results of double castration in hypertrophy of the prostate. Ann. Surg. 1895;22:1–80. doi: 10.1097/00000658-189507000-00001.
    1. Huggins C, Hodges CV. Studies on prostatic cancer - I The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. Cancer Res. 1941;1:293–297.
    1. Huggins C, Stevens RE, Hodges CV. Studies on prostate cancer II The effects of castration on advanced carcinoma of the prostate gland. Arch. Surg.-Chic. 1941;43:209–223. doi: 10.1001/archsurg.1941.01210140043004.
    1. Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer. 2015;15:701–711. doi: 10.1038/nrc4016.
    1. Labbé, D. P. & Brown, M. Transcriptional regulation in prostate cancer. Cold Spring Harb Perspect Med8, a030437 (2018).
    1. Baca SC, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–677. doi: 10.1016/j.cell.2013.03.021.
    1. Cancer Genome Atlas Research, N. The molecular taxonomy of primary prostate cancer. Cell. 2015;163:1011–1025. doi: 10.1016/j.cell.2015.10.025.
    1. Gurel B, et al. Nuclear MYC protein overexpression is an early alteration in human prostate carcinogenesis. Mod. Pathol. 2008;21:1156–1167. doi: 10.1038/modpathol.2008.111.
    1. Matejcic M, et al. Germline variation at 8q24 and prostate cancer risk in men of European ancestry. Nat. Commun. 2018;9:4616. doi: 10.1038/s41467-018-06863-1.
    1. Pomerantz MM, et al. The 8q24 cancer risk variant rs6983267 shows long-range interaction with MYC in colorectal cancer. Nat. Genet. 2009;41:882–884. doi: 10.1038/ng.403.
    1. Wasserman NF, Aneas I, Nobrega MA. An 8q24 gene desert variant associated with prostate cancer risk confers differential in vivo activity to a MYC enhancer. Genome Res. 2010;20:1191–1197. doi: 10.1101/gr.105361.110.
    1. Ellwood-Yen K, et al. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer Cell. 2003;4:223–238. doi: 10.1016/S1535-6108(03)00197-1.
    1. Barfeld SJ, et al. c-Myc antagonises the transcriptional activity of the androgen receptor in prostate cancer affecting key gene networks. EBioMedicine. 2017;18:83–93. doi: 10.1016/j.ebiom.2017.04.006.
    1. Bostwick DG, Liu L, Brawer MK, Qian J. High-grade prostatic intraepithelial neoplasia. Rev. Urol. 2004;6:171–179.
    1. Labbé DP, et al. High-fat diet fuels prostate cancer progression by rewiring the metabolome and amplifying the MYC program. Nat. Commun. 2019;10:4358. doi: 10.1038/s41467-019-12298-z.
    1. Crowell PD, et al. Expansion of luminal progenitor cells in the aging mouse and human prostate. Cell Rep. 2019;28:1499–1510. doi: 10.1016/j.celrep.2019.07.007.
    1. Dahlman A, et al. Effect of androgen deprivation therapy on the expression of prostate cancer biomarkers MSMB and MSMB-binding protein CRISP3. Prostate Cancer Prostatic Dis. 2010;13:369–375. doi: 10.1038/pcan.2010.25.
    1. Zhang J, Thomas TZ, Kasper S, Matusik RJ. A small composite probasin promoter confers high levels of prostate-specific gene expression through regulation by androgens and glucocorticoids in vitro and in vivo. Endocrinology. 2000;141:4698–4710. doi: 10.1210/endo.141.12.7837.
    1. Dang, C. V. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb Perspect Med3, a014217 (2013).
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Azizi E, et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell. 2018;174:1293–1308. doi: 10.1016/j.cell.2018.05.060.
    1. Mezzetti G, Loor R, Liao S. Androgen-sensitive spermine-binding protein of rat ventral prostate. Purification of the protein and characterization of the hormonal effect. Biochem J. 1979;184:431–440. doi: 10.1042/bj1840431.
    1. Pihlajamaa P, et al. Tissue-specific pioneer factors associate with androgen receptor cistromes and transcription programs. EMBO J. 2014;33:312–326.
    1. Gross M, et al. Beta-2-microglobulin is an androgen-regulated secreted protein elevated in serum of patients with advanced prostate cancer. Clin. Cancer Res. 2007;13:1979–1986. doi: 10.1158/1078-0432.CCR-06-1156.
    1. Stopkova R, Klempt P, Kuntova B, Stopka P. On the tear proteome of the house mouse (Mus musculus musculus) in relation to chemical signalling. PeerJ. 2017;5:e3541. doi: 10.7717/peerj.3541.
    1. Wubah JA, et al. Ventral prostate predominant l, a novel mouse gene expressed exclusively in the prostate. Prostate. 2002;51:21–29. doi: 10.1002/pros.10060.
    1. Rasanen K, Itkonen O, Koistinen H, Stenman UH. Emerging roles of SPINK1 in cancer. Clin. Chem. 2016;62:449–457. doi: 10.1373/clinchem.2015.241513.
    1. Wang R, et al. Preclinical study using Malat1 small interfering RNA or androgen receptor splicing variant 7 degradation enhancer ASC-J9((R)) to suppress enzalutamide-resistant prostate cancer progression. Eur. Urol. 2017;72:835–844. doi: 10.1016/j.eururo.2017.04.005.
    1. van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat. Rev. Cancer. 2010;10:301–309. doi: 10.1038/nrc2819.
    1. Pomerantz MM, et al. The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis. Nat. Genet. 2015;47:1346–1351. doi: 10.1038/ng.3419.
    1. Ernst J, et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature. 2011;473:43–49. doi: 10.1038/nature09906.
    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. Spratt DE, et al. Individual patient-level meta-analysis of the performance of the decipher genomic classifier in high-risk men after prostatectomy to predict development of metastatic disease. J. Clin. Oncol. 2017;35:1991–1998. doi: 10.1200/JCO.2016.70.2811.
    1. Taylor BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. doi: 10.1016/j.ccr.2010.05.026.
    1. Labrecque MP, et al. Molecular profiling stratifies diverse phenotypes of treatment-refractory metastatic castration-resistant prostate cancer. J. Clin. Invest. 2019;129:4492–4505. doi: 10.1172/JCI128212.
    1. Nguyen HM, et al. LuCaP prostate cancer patient-derived xenografts reflect the molecular heterogeneity of advanced disease an-d serve as models for evaluating cancer therapeutics. Prostate. 2017;77:654–671. doi: 10.1002/pros.23313.
    1. Abida W, et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc. Natl Acad. Sci. USA. 2019;116:11428–11436. doi: 10.1073/pnas.1902651116.
    1. Wang S, et al. Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat. Protoc. 2013;8:2502–2515. doi: 10.1038/nprot.2013.150.
    1. Pomerantz MM, et al. Analysis of the 10q11 cancer risk locus implicates MSMB and NCOA4 in human prostate tumorigenesis. PLoS Genet. 2010;6:e1001204. doi: 10.1371/journal.pgen.1001204.
    1. Rahl PB, et al. c-Myc regulates transcriptional pause release. Cell. 2010;141:432–445. doi: 10.1016/j.cell.2010.03.030.
    1. Chen FX, Smith ER, Shilatifard A. Born to run: control of transcription elongation by RNA polymerase II. Nat. Rev. Mol. Cell Biol. 2018;19:464–478. doi: 10.1038/s41580-018-0010-5.
    1. Ni M, et al. Amplitude modulation of androgen signaling by c-MYC. Genes Dev. 2013;27:734–748. doi: 10.1101/gad.209569.112.
    1. Karthaus WR, et al. Regenerative potential of prostate luminal cells revealed by single-cell analysis. Science. 2020;368:497–505. doi: 10.1126/science.aay0267.
    1. Joseph, D. B. et al. Urethral luminal epithelia are castration-insensitive cells of the proximal prostate. Prostate80, 872–884 (2020).
    1. Zhang J, et al. Characterization of cis elements of the probasin promoter necessary for prostate-specific gene expression. Prostate. 2010;70:934–951. doi: 10.1002/pros.21128.
    1. Harris AW, et al. The E mu-myc transgenic mouse. A model for high-incidence spontaneous lymphoma and leukemia of early B cells. J. Exp. Med. 1988;167:353–371. doi: 10.1084/jem.167.2.353.
    1. Kumar A, et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 2016;22:369–378. doi: 10.1038/nm.4053.
    1. Antonarakis ES, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014;371:1028–1038. doi: 10.1056/NEJMoa1315815.
    1. Joseph JD, et al. A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Disco. 2013;3:1020–1029. doi: 10.1158/-13-0226.
    1. Arriaga JM, et al. A MYC and RAS co-activation signature in localized prostate cancer drives bone metastasis and castration resistance. Nat. Cancer. 2020;1:1082–1096. doi: 10.1038/s43018-020-00125-0.
    1. Bai S, et al. A positive role of c-Myc in regulating androgen receptor and its splice variants in prostate cancer. Oncogene. 2019;38:4977–4989. doi: 10.1038/s41388-019-0768-8.
    1. Dardenne E, et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell. 2016;30:563–577. doi: 10.1016/j.ccell.2016.09.005.
    1. Malynn BA, et al. N-myc can functionally replace c-myc in murine development, cellular growth, and differentiation. Genes Dev. 2000;14:1390–1399. doi: 10.1101/gad.14.11.1390.
    1. Schmidt SF, Larsen BD, Loft A, Mandrup S. Cofactor squelching: artifact or fact? Bioessays. 2016;38:618–626. doi: 10.1002/bies.201600034.
    1. Han H, et al. Small-molecule MYC inhibitors suppress tumor growth and enhance immunotherapy. Cancer Cell. 2019;36:483–497. doi: 10.1016/j.ccell.2019.10.001.
    1. Cornwell M, et al. VIPER: Visualization Pipeline for RNA-seq, a Snakemake workflow for efficient and complete RNA-seq analysis. BMC Bioinformatics. 2018;19:135. doi: 10.1186/s12859-018-2139-9.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Trapnell C, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 2010;28:511–515. doi: 10.1038/nbt.1621.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
    1. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018;36:411–420. doi: 10.1038/nbt.4096.
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32:2847–2849. doi: 10.1093/bioinformatics/btw313.
    1. Pomerantz MM, et al. Prostate cancer reactivates developmental epigenomic programs during metastatic progression. Nat. Genet. 2020;52:790–799. doi: 10.1038/s41588-020-0664-8.
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25. doi: 10.1186/gb-2009-10-3-r25.
    1. Zhang Y, et al. Model-based analysis of ChIP-Seq (MACS) Genome Biol. 2008;9:R137. doi: 10.1186/gb-2008-9-9-r137.
    1. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell38, 576–589 (2010).
    1. Qiu, X. et al. CoBRA: containerized bioinformatics workflow for reproducible ChIP/ATAC-seq analysis. Genomics Proteomics Bioinformatics19, 652–661 (2021).
    1. Qin Q, et al. ChiLin: a comprehensive ChIP-seq and DNase-seq quality control and analysis pipeline. BMC Bioinforma. 2016;17:404. doi: 10.1186/s12859-016-1274-4.
    1. Gel B, Serra E. karyoploteR: an R/Bioconductor package to plot customizable genomes displaying arbitrary data. Bioinformatics. 2017;33:3088–3090. doi: 10.1093/bioinformatics/btx346.
    1. Colaprico A, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44:e71. doi: 10.1093/nar/gkv1507.
    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. Foroutan M, et al. Single sample scoring of molecular phenotypes. BMC Bioinformatics. 2018;19:404. doi: 10.1186/s12859-018-2435-4.
    1. Hothorn, T. maxstat: maximally selected rank statistics. R Package Version 07-25. Available online: (2017).
    1. Therneau, T. A., Lumley, T., Elizabeth, A. & Cynthia, C. Survival: survival analysis. R Package Version 3.2-3. Available online: (2020).
    1. Kassambara, A., Kosinski, M., Biecek, P. & Fabian, S. survminer: drawing survival curves using ‘ggplot2’. R package version 0.4.8. Available online: (2020).
    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.

Source: PubMed

3
Subscribe