292
Views
0
CrossRef citations to date
0
Altmetric
Review

Role of bromodomain and extraterminal (BET) proteins in prostate cancer

ORCID Icon, ORCID Icon, & ORCID Icon
Pages 213-228 | Received 08 Dec 2022, Accepted 28 Feb 2023, Published online: 09 Mar 2023

References

  • Kumaraswamy A, Welker Leng KR, Westbrook TC, et al. Recent advances in epigenetic biomarkers and epigenetic targeting in prostate cancer. Eur Urol. 2021;80(1):71–81.
  • Thompson D, Choo N, Bolton DM, et al. New approaches to targeting epigenetic regulation in prostate cancer. Curr Opin Urol. 2022;32(5):472–480
  • Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer. 2021;124:1478–1490.
  • Sarnik J, Popławski T, Tokarz P. BET proteins as attractive targets for cancer therapeutics. Int J Mol Sci [Internet]. 2021;22(20):11102. Available from
  • Gaucher J, Boussouar F, Montellier E, et al. Bromodomain-dependent stage-specific male genome programming by Brdt. EMBO J. 2012;31:3809–3820.
  • Wang N, Wu R, Tang D, et al. The BET family in immunity and disease. Signal Transduct Target Ther. 2021;6:1–22.
  • Filippakopoulos P, Picaud S, Mangos M, et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell. 2012;149:214–231.
  • Filippakopoulos P, Knapp S. The bromodomain interaction module. FEBS Lett. 2012;586:2692–2704.
  • Rahman S, Sowa ME, Ottinger M, et al. The Brd4 extraterminal domain confers transcription activation independent of pTEFb by recruiting multiple proteins, including NSD3. Mol Cell Biol. 2011;31:2641–2652.
  • Itzen F, Greifenberg AK, Bösken CA, et al. Brd4 activates P-TEFb for RNA polymerase II CTD phosphorylation. Nucleic Acids Res. 2014;42:7577–7590.
  • Yang L, Zhang Y, Shan W, et al. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Sci Transl Med [Internet]. 2017;9. Available from http://dx.doi.org/10.1126/scitranslmed.aal1645
  • Nerlakanti N, Yao J, Nguyen DT, et al. Targeting the BRD4-HOXB13 coregulated transcriptional networks with bromodomain-kinase inhibitors to suppress metastatic castration-resistant prostate cancer. Mol Cancer Ther. 2018;17:2796–2810.
  • Devaiah BN, Lewis BA, Cherman N, et al. BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase II carboxy-terminal domain. Proc Natl Acad Sci U S A. 2012;109:6927–6932.
  • Devaiah BN, Mu J, Akman B, et al. MYC protein stability is negatively regulated by BRD4. Proc Natl Acad Sci U S A. 2020;117:13457–13467.
  • Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54:728–736.
  • Asangani IA, Dommeti VL, Wang X, et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature. 2014;510:278–282.
  • Urbanucci A, Mills IG. Bromodomain-containing proteins in prostate cancer. Mol Cell Endocrinol. 2018;462:31–40.
  • Welti J, Sharp A, Yuan W, et al. Targeting bromodomain and extra-terminal (BET) family proteins in Castration-Resistant Prostate Cancer (CRPC). Clin Cancer Res. 2018;24:3149–3162.
  • Shen G, Jiang M, Pu J. Dual inhibition of BRD4 and PI3K by SF2523 suppresses human prostate cancer cell growth in vitro and in vivo. Biochem Biophys Res Commun. 2018;495:567–573.
  • Shafran JS, Jafari N, Casey AN, et al. BRD4 regulates key transcription factors that drive epithelial-mesenchymal transition in castration-resistant prostate cancer. Prostate Cancer Prostatic Dis. 2021;24:268–277.
  • Antonarakis ES, Lu C, Wang H, et al. AR-V7 and resistance to enzalutamide and Abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–1038.
  • Wyce A, Degenhardt Y, Bai Y, et al. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer. Oncotarget. 2013;4:2419–2429.
  • Asangani IA, Wilder-Romans K, Dommeti VL, et al. BET bromodomain inhibitors enhance efficacy and disrupt resistance to ar antagonists in the treatment of prostate cancer [Internet]. Mol Cancer Res. 2016;324–331. doi: 10.1158/1541-7786.mcr-15-0472
  • Urbanucci A, Barfeld SJ, Kytölä V, et al. Androgen receptor deregulation drives bromodomain-mediated chromatin alterations in prostate cancer. Cell Rep. 2017;19:2045–2059.
  • Dehm SM, Schmidt LJ, Heemers H. vessella RL and Tindall DJ: splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res. 2008;68:5469–5477.
  • Chan SC, Selth LA, Li Y, et al. Targeting chromatin binding regulation of constitutively active AR variants to overcome prostate cancer resistance to endocrine-based therapies. Nucleic Acids Res. 2015;43:5880–5897.
  • Watson PA, Chen YF, Balbas MD, et al. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proc Nat Acad Sci. 2010;107:16759–16765.
  • Cai L, Tsai Y-H, Wang P, et al. ZFX mediates non-canonical oncogenic functions of the androgen receptor splice variant 7 in castrate-resistant prostate cancer. Mol Cell. 2018;72:341–354.e6.
  • Nadiminty N, Tummala R, Liu C, et al. NF-κB2/p52:c-Myc:hnRNPA1 pathway regulates expression of androgen receptor splice variants and enzalutamide sensitivity in prostate cancer. Mol Cancer Ther. 2015;14:1884–1895.
  • Mori JO, Shafran JS, Stojanova M, et al. Novel forms of prostate cancer chemoresistance to successful androgen deprivation therapy demand new approaches: rationale for targeting BET proteins. Prostate. 2022;82:1005–1015.
  • Gao L, Schwartzman J, Gibbs A, et al. Androgen receptor promotes ligand-independent prostate cancer progression through c-Myc upregulation. PLoS One. 2013;8:e63563.
  • Kotekar A, Singh AK, Devaiah BN. BRD4 and MYC: power couple in transcription and disease. FEBS J [Internet]. 2022. doi: 10.1111/febs.16580
  • Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146:904–917.
  • Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A. 2011;108:16669–16674.
  • Llombart V, Mansour MR. Therapeutic targeting of “undruggable” MYC. eBioMedicine [Internet] Available from. 2022;75. https://doi.org/10.1016/j.ebiom.2021.103756
  • Whitfield JR, Soucek L. The long journey to bring a Myc inhibitor to the clinic. J Cell Biol [Internet] Available from. 2021;220. http://dx.doi.org/10.1083/jcb.202103090
  • Tan Y, Wang L, Du Y, et al. Inhibition of BRD4 suppresses tumor growth in prostate cancer via the enhancement of FOXO1 expression. Int J Oncol. 2018;53:2503–2517.
  • Li X, Baek G, Ramanand SG, et al. BRD4 promotes DNA repair and mediates the formation of TMPRSS2-ERG gene rearrangements in prostate cancer. Cell Rep. 2018;22:796–808.
  • Li X, Baek G, Carreira S, et al. Targeting radioresistance and replication fork stability in prostate cancer. JCI Insight [Internet]. 2022;7. Available from http://dx.doi.org/10.1172/jci.insight.152955
  • Tomlins SA, Bjartell A, Chinnaiyan AM, et al. ETS gene fusions in prostate cancer: from discovery to daily clinical practice. Eur Urol. 2009;56:275–286.
  • Tomlins SA, Rhodes DR, Perner S, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–648.
  • Adamo P, Ladomery MR. The oncogene ERG: a key factor in prostate cancer. Oncogene. 2016;35:403–414.
  • Demichelis F, Rubin MA. TMPRSS2-ETS fusion prostate cancer: biological and clinical implications. J Clin Pathol. 2007;60:1185–1186.
  • Coleman DJ, Gao L, King CJ, et al. BET bromodomain inhibition blocks the function of a critical AR-independent master regulator network in lethal prostate cancer. Oncogene. 2019;38:5658–5669.
  • Storck WK, May AM, Westbrook TC, et al. The role of epigenetic change in therapy-induced neuroendocrine prostate cancer lineage plasticity. Front Endocrinol. 2022;13:926585.
  • Kim D-H, Sun D, Storck WK, et al. BET bromodomain inhibition blocks an AR-repressed, E2F1-activated treatment-emergent neuroendocrine prostate cancer lineage plasticity program. Clin Cancer Res. 2021;27:4923–4936.
  • Schwalm MP, Knapp S. BET bromodomain inhibitors. Curr Opin Chem Biol. 2022;68:102148.
  • Piha-Paul SA, Hann CL, French CA, et al. Phase 1 Study of molibresib (GSK525762), a bromodomain and extra-terminal domain protein inhibitor, in NUT carcinoma and other solid tumors. JNCI Cancer Spectr. 2020;4:kz093
  • Gilan O, Rioja I, Knezevic K, et al. Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science. 2020;368:387–394.
  • Faivre EJ, McDaniel KF, Albert DH, et al. Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer. Nature. 2020;578:306–310.
  • Waring MJ, Chen H, Rabow AA, et al. Potent and selective bivalent inhibitors of BET bromodomains. Nat Chem Biol. 2016;12:1097–1104.
  • Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21:181–200.
  • Winter GE, Buckley DL, Paulk J, et al. DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science 2015;348:1376–1381.
  • Gadd MS, Testa A, Lucas X, et al. Structural basis of PROTAC cooperative recognition for selective protein degradation. Nat Chem Biol. 2017;13:514–521.
  • Lu J, Qian Y, Altieri M, et al. Hijacking the E3 ubiquitin ligase cereblon to efficiently target BRD4. Chem Biol. 2015;22:755–763.
  • Lewin J, Soria J-C, Stathis A, et al. Phase Ib trial with birabresib, a small-molecule inhibitor of bromodomain and extraterminal proteins, in patients with selected advanced solid tumors. J Clin Oncol. 2018;36:3007–3014.
  • Piha-Paul SA, Sachdev JC, Barve M, et al. First-in-human study of mivebresib (ABBV-075), an oral pan-inhibitor of bromodomain and extra terminal proteins, in patients with relapsed/refractory solid tumors. Clin Cancer Res. 2019;25:6309–6319.
  • Aggarwal RR, Schweizer MT, Nanus DM, et al. A phase Ib/IIa study of the Pan-BET inhibitor ZEN-3694 in combination with enzalutamide in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res. 2020;26:5338–5347.
  • Cousin S, Blay J-Y, Garcia IB, et al. Safety, pharmacokinetic, pharmacodynamic and clinical activity of molibresib for the treatment of nuclear protein in testis carcinoma and other cancers: results of a Phase I/II open-label, dose escalation study. Int J Cancer. 2022;150:993–1006.
  • Vaishampayan UN, Narayan V, Wise D, et al. A phase Ib open-label, dose escalation and expansion study to investigate the safety, pharmacokinetics, pharmacodynamics and clinical activity of GSK525762 in combination with Abiraterone or enzalutamide in metastatic castrate-resistant prostate cancer. J Clin Orthod. 2018;36: TPS391–TPS391.
  • Ameratunga M, Braña I, Bono P, et al. First-in-human Phase 1 open label study of the BET inhibitor ODM-207 in patients with selected solid tumours. Br J Cancer. 2020;123:1730–1736.
  • Aggarwal R, Starodub AN, Koh BD, et al. Phase 1b study of the BET inhibitor GS-5829 as monotherapy and combined with enzalutamide in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res [Internet]. 2022. doi: 10.1158/1078-0432.CCR-22-0175
  • Bechter O, Schöffski P. Make your best BET: the emerging role of BET inhibitor treatment in malignant tumors. Pharmacol Ther. 2020;208:107479.
  • Dai X, Gan W, Li X, et al. Prostate cancer–associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat Med. 2017;23:1063–1071.
  • Janouskova H, El Tekle G, Bellini E, et al. Opposing effects of cancer-type-specific SPOP mutants on BET protein degradation and sensitivity to BET inhibitors. Nat Med. 2017;23:1046–1054.
  • Zhang P, Wang D, Zhao Y, et al. Intrinsic BET inhibitor resistance in SPOP-mutated prostate cancer is mediated by BET protein stabilization and AKT–mTORC1 activation. Nat Med. 2017;23:1055–1062.
  • Barbieri I, Cannizzaro E, Dawson MA. Bromodomains as therapeutic targets in cancer. Brief Funct Genomics. 2013;12:219–230.
  • Jin X, Yan Y, Wang D, et al. DUB3 promotes BET inhibitor resistance and cancer progression by deubiquitinating BRD4. Mol Cell. 2018;71:592–605.e4.
  • Yang G-F, Zhang X, Su Y-G, et al. The role of the deubiquitinating enzyme DUB3/USP17 in cancer: a narrative review. Cancer Cell Int. 2021;21:455.
  • Sena LA, Denmeade SR, Antonarakis ES. Targeting the spectrum of immune checkpoints in prostate cancer. Expert Rev Clin Pharmacol. 2021;14:1253–1266.
  • 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. J Clin Oncol. 2020;38:395–405.
  • Flammiger A, Weisbach L, Huland H, et al. High tissue density of FOXP3+ T cells is associated with clinical outcome in prostate cancer. Eur J Cancer. 2013;49:1273–1279.
  • Blades RA, Keating PJ, McWilliam LJ, et al. Loss of HLA class I expression in prostate cancer: implications for immunotherapy. Urology. 1995;46:681–686. discussion 686-7
  • Mao W, Ghasemzadeh A, Freeman ZT, et al. Immunogenicity of prostate cancer is augmented by BET bromodomain inhibition. J Immunother Cancer. 2019;7:277.
  • Bauer K, Berghoff AS, Preusser M, et al. Degradation of BRD4 - a promising treatment approach not only for hematologic but also for solid cancer. Am J Cancer Res. 2021;11:530–545.
  • Olson B, Chaudagar K, Bao R, et al. BET inhibition sensitizes immunologically-cold Rb-deficient prostate cancer to immune checkpoint blockade [Internet]. bioRxiv. 2022;2022. Available from https://www.biorxiv.org/content/10.1101/2022.03.24.485685v1
  • Ding D, Zheng R, Tian Y, et al. Retinoblastoma protein as an intrinsic BRD4 inhibitor modulates small molecule BET inhibitor sensitivity in cancer. Nat Commun. 2022;13:6311.
  • Hajmirza A, Emadali A, Gauthier A, et al. BET family protein BRD4: an emerging actor in NFκB signaling in inflammation and cancer. Biomedicines [Internet]. 2018;6. doi: 10.3390/biomedicines6010016
  • Vázquez R, Civenni G, Kokanovic A, et al. Efficacy of novel bromodomain and extraterminal inhibitors in combination with chemotherapy for castration-resistant prostate cancer. Eur Urol Oncol. 2021;4:437–446.
  • Xu Y, Pachnikova G, Przybilla D, et al. Evaluation of JQ1 combined with docetaxel for the treatment of prostate cancer cells in 2D- and 3D-culture systems. Front Pharmacol. 2022;13:839620.
  • Wang J, Wang Y, Mei H, et al. The BET bromodomain inhibitor JQ1 radiosensitizes non-small cell lung cancer cells by upregulating p21. Cancer Lett. 2017;391:141–151.
  • Camero S, Camicia L, Marampon F, et al. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett. 2020;479:71–88.
  • Sweat SD, Pacelli A, Murphy GP, et al. Prostate-specific membrane antigen expression is greatest in prostate adenocarcinoma and lymph node metastases. Urology. 1998;52:637–640.
  • Paschalis A, Sheehan B, Riisnaes R, et al. PSMA heterogeneity and DNA repair defects in prostate cancer. J Clin Orthod. 2019;37:5002.
  • Vlachostergios PJ, Niaz MJ, Sun M, et al. Prostate-specific membrane antigen uptake and survival in metastatic castration-resistant prostate cancer. Front Oncol. 2021;11:630589.
  • Graf F, Fahrer J, Maus S, et al. DNA double strand breaks as predictor of efficacy of the alpha-particle emitter Ac-225 and the electron emitter Lu-177 for somatostatin receptor targeted radiotherapy. PLoS One. 2014;9:e88239.
  • Shigeta S, Lui GYL, Shaw R, et al. Targeting BET proteins BRD2 and BRD3 in combination with PI3K-AKT inhibition as a therapeutic strategy for ovarian clear cell carcinoma. Mol Cancer Ther. 2021;20:691–703.
  • Ilic N, Utermark T, Widlund HR, et al. PI3K-targeted therapy can be evaded by gene amplification along the MYC-eukaryotic translation initiation factor 4E (eIF4E) axis. Proc Natl Acad Sci U S A. 2011;108:E699–708.
  • Stratikopoulos EE, Dendy M, Szabolcs M, et al. Kinase and BET inhibitors together clamp inhibition of PI3K signaling and overcome resistance to therapy. Cancer Cell. 2015;27:837–851.
  • Shen G, Chen J, Zhou Y, et al. AZD5153 inhibits prostate cancer cell growth in vitro and in vivo. Cell Physiol Biochem. 2018;50:798–809.
  • Greenwell IB, Ip A, Cohen JB. PI3K inhibitors: understanding toxicity mechanisms and management. Oncology. 2017;31:821–828.
  • Hanlon A, Brander DM. Managing toxicities of phosphatidylinositol-3-kinase (PI3K) inhibitors. Hematology Am Soc Hematol Educ Program. 2020;2020:346–356.
  • Wang H, Liu G, Jin X, et al. BET inhibitor JQ1 enhances anti-tumor immunity and synergizes with PD-1 blockade in CRC. J Cancer. 2022;13:2126–2137.
  • Zhang Y, Yan H, Xu Z, et al. Molecular basis for class side effects associated with PI3K/AKT/mTOR pathway inhibitors. Expert Opin Drug Metab Toxicol. 2019;15:767–774.
  • Alghamdi S, Khan I, Beeravolu N, et al. BET protein inhibitor JQ1 inhibits growth and modulates WNT signaling in mesenchymal stem cells. Stem Cell Res Ther. 2016;7:22.
  • Iglehart JD, Dirk Iglehart J, Silver DP. Synthetic lethality — a new direction in cancer-drug development [Internet]. N Engl J Med. 2009;189–191. doi: 10.1056/nejme0903044
  • Teply BA, Antonarakis ES. Treatment strategies for DNA repair-deficient prostate cancer. Expert Rev Clin Pharmacol. 2017;10:889–898.
  • Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell. 2015;163:1011–1025.
  • Robinson D, Van Allen EM, Wu Y-M, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;162:454.
  • 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.
  • Antonarakis ES. Germline DNA repair mutations and response to hormonal therapy in advanced prostate cancer [Internet]. Eur Urol. 2017;43–44. doi: 10.1016/j.eururo.2017.03.003
  • Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373:1697–1708.
  • Floyd SR, Pacold ME, Huang Q, et al. The bromodomain protein Brd4 insulates chromatin from DNA damage signalling. Nature. 2013;498:246–250.
  • Wang L, Xie L, Ramachandran S, et al. Non-canonical bromodomain within DNA-PKcs promotes DNA damage response and radioresistance through recognizing an IR-induced acetyl-lysine on H2AX. Chem Biol. 2015;22:849–861.
  • Isaacsson Velho P, Fu W, Wang H, et al. Wnt-pathway activating mutations are associated with resistance to first-line abiraterone and enzalutamide in castration-resistant prostate cancer. Eur Urol. 2020;77:14–21.
  • Gera JF, Mellinghoff IK, Shi Y, et al. AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem. 2004;279:2737–2746.
  • Hubbard GK, Mutton LN, Khalili M, et al. Combined MYC activation and PTEN loss are sufficient to create genomic instability and lethal metastatic prostate cancer. Cancer Res. 2016;76:283–292.
  • Song L-N, Herrell R, Byers S, et al. Beta-catenin binds to the activation function 2 region of the androgen receptor and modulates the effects of the N-terminal domain and TIF2 on ligand-dependent transcription. Mol Cell Biol. 2003;23:1674–1687.
  • Truica CI, Byers S, Gelmann EP. Beta-catenin affects androgen receptor transcriptional activity and ligand specificity. Cancer Res. 2000;60:4709–4713.
  • Yang F, Li X, Sharma M, et al. Linking β-catenin to androgen-signaling pathway. J Biol Chem. 2002;277:11336–11344.
  • Asem MS, Buechler S, Wates RB, et al. Wnt5a signaling in cancer. Cancers [Internet]. 2016;8. Available from.;. http://dx.doi.org/10.3390/cancers8090079
  • Rathert P, Roth M, Neumann T, et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature. 2015;525:543–547.
  • Fong CY, Gilan O, Lam EYN, et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature. 2015;525:538–542.
  • Metzger E, Wissmann M, Yin N, et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature. 2005;437:436–439.
  • Suikki HE, Kujala PM, Tammela TLJ, et al. Genetic alterations and changes in expression of histone demethylases in prostate cancer. Prostate. 2010;70:889–898.
  • Kahl P, Gullotti L, Heukamp LC, et al. Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res. 2006;66:11341–11347.
  • Cai C, He HH, Chen S, et al. Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1. Cancer Cell. 2011;20:457–471.
  • Etani T, Naiki T, Naiki-Ito A, et al. NCL1, A highly selective lysine-specific demethylase 1 inhibitor, suppresses castration-resistant prostate cancer growth via regulation of apoptosis and autophagy. J Clin Med Res [Internet]. 2019;8. Available from.;. http://dx.doi.org/10.3390/jcm8040442
  • Wang J, Yu Q, Qiu Z, et al. The combined effect of epigenetic inhibitors for LSD1 and BRD4 alters prostate cancer growth and invasion. Aging (Albany NY). 2020;12:397–415.
  • Beltran H, Prandi D, Mosquera JM, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22:298–305.
  • Abida W, Cyrta J, Heller G, et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc Natl Acad Sci U S A. 2019;116:11428–11436.
  • Sehrawat A, Gao L, Wang Y, et al. LSD1 activates a lethal prostate cancer gene network independently of its demethylase function. Proc Natl Acad Sci U S A. 2018;115:E4179–E4188.
  • Sashida G, Wang C, Tomioka T, et al. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J Exp Med. 2016;213:1459–1477.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.