Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Molecular events in neuroendocrine prostate cancer development

Abstract

Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer. NEPC arises de novo only rarely; the disease predominantly develops from adenocarcinoma in response to drug-induced androgen receptor signalling inhibition, although the mechanisms behind this transdifferentiation are a subject of debate. The survival of patients with NEPC is poor, and few effective treatment options are available. To improve clinical outcomes, understanding of the biology and molecular mechanisms regulating NEPC development is crucial. Various NEPC molecular drivers make temporal contributions during NEPC development, and despite the limited treatment options available, several novel targeted therapeutics are currently under research.

Key points

  • Neuroendocrine prostate cancer (NEPC) is an aggressive variant form that is characterized by low or absent androgen receptor (AR) expression, gain of the neuroendocrine phenotype and is not responsive to therapies targeting AR signalling.

  • De novo NEPC accounts for less than 2% of all prostate cancers, but treatment-induced NEPC occurs in 10–17% of patients with castration-resistant prostate cancer by evolving from adenocarcinoma, probably as a result of a transdifferentiation process.

  • Molecular mechanisms underlying NEPC development include genomic alterations, abnormal regulation of epigenetic regulators, transcription factors and other molecular pathways. The temporal contribution and co-operation of NEPC drivers during adenocarcinoma to NEPC transdifferentiation is largely unknown; thus, longitudinal study of serial patient samples and preclinical models that recapitulate the entire disease progression is warranted.

  • Longitudinal analyses of the only clinically relevant patient-derived xenograft model with serial genomic and transcriptomic data available throughout the adenocarcinoma-to-NEPC transdifferentiation process (LTL331/331 R) could group NEPC-driving molecular alterations into early and terminal events, suggesting their roles during different phases of NEPC development.

  • Platinum-based chemotherapy is the only treatment currently available for NEPC. Advances in NEPC research have led to new potential therapies that are undergoing investigation in clinical trials or in preclinical development.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Hypotheses of the origin of neuroendocrine prostate cancer.
Fig. 2: Timing of the emergence of molecular events in the LTL331/LTL331R patient-derived xenograft model of neuroendocrine prostate cancer53,98.

Similar content being viewed by others

References

  1. Sung, H. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  2. Humphrey, P. A. Histological variants of prostatic carcinoma and their significance. Histopathology 60, 59–74 (2012).

    Article  PubMed  Google Scholar 

  3. Huggins, C. & Hodges, C. V. Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate. CA Cancer J. Clin. 22, 232–240 (1972).

    Article  CAS  PubMed  Google Scholar 

  4. Gillessen, S. et al. Management of patients with advanced prostate cancer: recommendations of the St Gallen Advanced Prostate Cancer Consensus Conference (APCCC) 2015. Ann. Oncol. 26, 1589–604 (2016).

    Article  Google Scholar 

  5. Watson, P. A., Arora, V. K. & Sawyers, C. L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer 15, 701–711 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chen, C. D. et al. Molecular determinants of resistance to antiandrogen therapy. Nat. Med. 10, 33–39 (2004).

    Article  PubMed  CAS  Google Scholar 

  7. Beer, T. M. et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 371, 424–433 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Scher, H. I. et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 367, 1187–1197 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. de Bono, J. S. et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 364, 1995–2005 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ryan, C. J. et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 368, 138–148 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Fizazi, K. et al. Nonmetastatic, castration-resistant prostate cancer and survival with darolutamide. N. Engl. J. Med. 383, 1040–1049 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Beltran, H. et al. Aggressive variants of castration-resistant prostate cancer. Clin. Cancer Res. 20, 2846–2850 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bluemn, E. G. et al. Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling. Cancer Cell 32, 474–489.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, H. T. et al. Neuroendocrine Prostate Cancer (NEPC) progressing from conventional prostatic adenocarcinoma: factors associated with time to development of NEPC and survival from NEPC diagnosis — a systematic review and pooled analysis. J. Clin. Oncol. 32, 3383–3390 (2014).

    Article  PubMed  Google Scholar 

  15. Zaffuto, E. et al. Contemporary incidence and cancer control outcomes of primary neuroendocrine prostate cancer: a SEER database analysis. Clin. Genitourin. Cancer 15, e793–e800 (2017).

    Article  PubMed  Google Scholar 

  16. Vlachostergios, P. J., Puca, L. & Beltran, H. Emerging variants of castration-resistant prostate cancer. Curr. Oncol. Rep. 19, 32 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Aggarwal, R. et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J. Clin. Oncol. 36, 2492–2503 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Abida, W. et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc. Natl Acad. Sci. USA 116, 11428–11436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chedgy, E. C. et al. Biallelic tumour suppressor loss and DNA repair defects in de novo small-cell prostate carcinoma. J. Pathol. 246, 244–253 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Epstein, J. I. et al. Proposed morphologic classification of prostate cancer with neuroendocrine differentiation. Am. J. Surg. Pathol. 38, 756–767 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Aparicio, A. M. et al. Platinum-based chemotherapy for variant castrate-resistant prostate cancer. Clin. Cancer Res. 19, 3621–3630 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, W. & Epstein, J. I. Small cell carcinoma of the prostate. A morphologic and immunohistochemical study of 95 cases. Am. J. Surg. Pathol. 32, 65–71 (2008).

    Article  PubMed  Google Scholar 

  23. Beltran, H. et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 1, 487–495 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Conteduca, V. et al. Clinical features of neuroendocrine prostate cancer. Eur. J. Cancer 121, 7–18 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Beltran, H. et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin. Cancer Res. 25, 6916–6924 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Butler, W. & Huang, J. Neuroendocrine cells of the prostate: histology, biological functions, and molecular mechanisms. Precis. Clin. Med. 4, 25–34 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Huang, Y. H., Zhang, Y. Q. & Huang, J. T. Neuroendocrine cells of prostate cancer: biologic functions and molecular mechanisms. Asian J. Androl. 21, 291–295 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zelivianski, S. et al. Multipathways for transdifferentiation of human prostate cancer cells into neuroendocrine-like phenotype. Biochim. Biophys. Acta 1539, 28–43 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Bonkhoff, H. Neuroendocrine cells in benign and malignant prostate tissue: morphogenesis, proliferation, and androgen receptor status. Prostate Suppl. 8, 18–22 (1998).

    Article  CAS  PubMed  Google Scholar 

  30. Bonkhoff, H., Stein, U. & Remberger, K. Endocrine-paracrine cell types in the prostate and prostatic adenocarcinoma are postmitotic cells. Hum. Pathol. 26, 167–170 (1995).

    Article  CAS  PubMed  Google Scholar 

  31. Garabedian, E. M., Humphrey, P. A. & Gordon, J. I. A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells. Proc. Natl Acad. Sci. USA 95, 15382–15387 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Terry, S. & Beltran, H. The many faces of neuroendocrine differentiation in prostate cancer progression. Front. Oncol. 4, 60 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Han, B. et al. Characterization of ETS gene aberrations in select histologic variants of prostate carcinoma. Mod. Pathol. 22, 1176–1185 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lotan, T. L. et al. ERG gene rearrangements are common in prostatic small cell carcinomas. Mod. Pathol. 24, 820–828 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Williamson, S. R. et al. ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin. Mod. Pathol. 24, 1120–1127 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Demichelis, F. et al. TMPRSS2:ERG gene fusion associated with lethal prostate cancer in a watchful waiting cohort. Oncogene 26, 4596–4599 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Rajput, A. B. et al. Frequency of the TMPRSS2:ERG gene fusion is increased in moderate to poorly differentiated prostate cancers. J. Clin. Pathol. 60, 1238–1243 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Tan, H. L. et al. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin. Cancer Res. 20, 890–903 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Hansel, D. E. et al. Shared TP53 gene mutation in morphologically and phenotypically distinct concurrent primary small cell neuroendocrine carcinoma and adenocarcinoma of the prostate. Prostate 69, 603–609 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sauer, C. G., Roemer, A. & Grobholz, R. Genetic analysis of neuroendocrine tumor cells in prostatic carcinoma. Prostate 66, 227–234 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Beltran, H. et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat. Med. 22, 298–305 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dong, B. et al. Single-cell analysis supports a luminal-neuroendocrine transdifferentiation in human prostate cancer. Commun. Biol. 3, 778 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Horoszewicz, J. S. et al. LNCaP model of human prostatic carcinoma. Cancer Res. 43, 1809–1818 (1983).

    CAS  PubMed  Google Scholar 

  44. Burchardt, T. et al. Transdifferentiation of prostate cancer cells to a neuroendocrine cell phenotype in vitro and in vivo. J. Urol. 162, 1800–1805 (1999).

    Article  CAS  PubMed  Google Scholar 

  45. Bishop, J. L. et al. The master neural transcription factor BRN2 is an androgen receptor-suppressed driver of neuroendocrine differentiation in prostate cancer. Cancer Discov. 7, 54–71 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Greenberg, N. M. et al. The rat probasin gene promoter directs hormonally and developmentally regulated expression of a heterologous gene specifically to the prostate in transgenic mice. Mol. Endocrinol. 8, 230–239 (1994).

    CAS  PubMed  Google Scholar 

  47. Gingrich, J. R. et al. Androgen-independent prostate cancer progression in the TRAMP model. Cancer Res. 57, 4687–4691 (1997).

    CAS  PubMed  Google Scholar 

  48. Greenberg, N. M. et al. Prostate cancer in a transgenic mouse. Proc. Natl Acad. Sci. USA 92, 3439–3443 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kaplan-Lefko, P. J. et al. Pathobiology of autochthonous prostate cancer in a pre-clinical transgenic mouse model. Prostate 55, 219–237 (2003).

    Article  PubMed  Google Scholar 

  50. Chiaverotti, T. et al. Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer. Am. J. Pathol. 172, 236–246 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ku, S. Y. et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78–83 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zou, M. et al. Transdifferentiation as a mechanism of treatment resistance in a mouse model of castration-resistant prostate cancer. Cancer Discov. 7, 736–749 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lin, D. et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer Res. 74, 1272–1283 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Lee, J. K. et al. N-Myc drives neuroendocrine prostate cancer initiated from human prostate epithelial cells. Cancer Cell 29, 536–547 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ci, X. et al. Heterochromatin protein 1alpha mediates development and aggressiveness of neuroendocrine prostate cancer. Cancer Res. 78, 2691–2704 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. Aparicio, A. M. et al. Combined tumor suppressor defects characterize clinically defined aggressive variant prostate cancers. Clin. Cancer Res. 22, 1520–1530 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Zhou, Z. et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer Res. 66, 7889–7898 (2006).

    Article  CAS  PubMed  Google Scholar 

  58. Tran, C. et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787–790 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mu, P. et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 355, 84–88 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Nyquist, M. D. et al. Combined TP53 and RB1 loss promotes prostate cancer resistance to a spectrum of therapeutics and confers vulnerability to replication stress. Cell Rep. 31, 107669 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Dardenne, E. et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell 30, 563–577 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yan, M. et al. Aurora-a kinase: a potent oncogene and target for cancer therapy. Med. Res. Rev. 36, 1036–1079 (2016).

    Article  PubMed  Google Scholar 

  63. Otto, T. et al. Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma. Cancer Cell 15, 67–78 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Brockmann, M. et al. Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell 24, 75–89 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Li, E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat. Rev. Genet. 3, 662–673 (2002).

    Article  CAS  PubMed  Google Scholar 

  66. Richly, H., Aloia, L. & Di Croce, L. Roles of the Polycomb group proteins in stem cells and cancer. Cell Death Dis. 2, e204 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sauvageau, M. & Sauvageau, G. Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell 7, 299–313 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Di Croce, L. & Helin, K. Transcriptional regulation by Polycomb group proteins. Nat. Struct. Mol. Biol. 20, 1147–1155 (2013).

    Article  PubMed  CAS  Google Scholar 

  69. Clermont, P. L. et al. Polycomb-mediated silencing in neuroendocrine prostate cancer. Clin. Epigenetics 7, 40 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Kim, K. H. & Roberts, C. W. Targeting EZH2 in cancer. Nat. Med. 22, 128–134 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Kleb, B. et al. Differentially methylated genes and androgen receptor re-expression in small cell prostate carcinomas. Epigenetics 11, 184–193 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Nielsen, A. L. et al. Heterochromatin formation in mammalian cells: interaction between histones and HP1 proteins. Mol. Cell 7, 729–739 (2001).

    Article  CAS  PubMed  Google Scholar 

  73. Nielsen, A. L. et al. Interaction with members of the heterochromatin protein 1 (HP1) family and histone deacetylation are differentially involved in transcriptional silencing by members of the TIF1 family. EMBO J. 18, 6385–6395 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kraemer, D., Wozniak, R. W., Blobel, G. & Radu, A. The human CAN protein, a putative oncogene product associated with myeloid leukemogenesis, is a nuclear pore complex protein that faces the cytoplasm. Proc. Natl Acad. Sci. USA 91, 1519–1523 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Cleary, J. et al. p300/CBP-associated factor drives DEK into interchromatin granule clusters. J. Biol. Chem. 280, 31760–31767 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Hollenbach, A. D., McPherson, C. J., Mientjes, E. J., Iyengar, R. & Grosveld, G. Daxx and histone deacetylase II associate with chromatin through an interaction with core histones and the chromatin-associated protein Dek. J. Cell Sci. 115, 3319–3330 (2002).

    Article  CAS  PubMed  Google Scholar 

  77. Lin, D. et al. Identification of DEK as a potential therapeutic target for neuroendocrine prostate cancer. Oncotarget 6, 1806–1820 (2015).

    Article  PubMed  Google Scholar 

  78. Mittal, P. & Roberts, C. W. M. The SWI/SNF complex in cancer - biology, biomarkers and therapy. Nat. Rev. Clin. Oncol. 17, 435–448 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Roberts, C. W. & Orkin, S. H. The SWI/SNF complex–chromatin and cancer. Nat. Rev. Cancer 4, 133–142 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Kadoch, C. & Crabtree, G. R. Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics. Sci. Adv. 1, e1500447 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Kadoch, C. & Crabtree, G. R. Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell 153, 71–85 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Buscarlet, M. et al. Essential role of BRG, the ATPase subunit of BAF chromatin remodeling complexes, in leukemia maintenance. Blood 123, 1720–1728 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Jubierre, L. et al. BRG1/SMARCA4 is essential for neuroblastoma cell viability through modulation of cell death and survival pathways. Oncogene 35, 5179–5190 (2016).

    Article  CAS  PubMed  Google Scholar 

  84. Laurette, P. et al. Chromatin remodellers Brg1 and Bptf are required for normal gene expression and progression of oncogenic Braf-driven mouse melanoma. Cell Death Differ. 27, 29–43 (2020).

    Article  CAS  PubMed  Google Scholar 

  85. Cyrta, J. et al. Role of specialized composition of SWI/SNF complexes in prostate cancer lineage plasticity. Nat. Commun. 11, 5549 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Wapinski, O. & Chang, H. Y. Long noncoding RNAs and human disease. Trends Cell Biol. 21, 354–361 (2011).

    Article  CAS  PubMed  Google Scholar 

  87. Statello, L., Guo, C. J., Chen, L. L. & Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 22, 96–118 (2021).

    Article  CAS  PubMed  Google Scholar 

  88. Walsh, A. L., Tuzova, A. V., Bolton, E. M., Lynch, T. H. & Perry, A. S. Long noncoding RNAs and prostate carcinogenesis: the missing ‘linc’? Trends Mol. Med. 20, 428–436 (2014).

    Article  CAS  PubMed  Google Scholar 

  89. Ramnarine, V. R. et al. The long noncoding RNA landscape of neuroendocrine prostate cancer and its clinical implications. Gigascience 7, giy050 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  90. Crea, F. et al. The role of epigenetics and long noncoding RNA MIAT in neuroendocrine prostate cancer. Epigenomics 8, 721–731 (2016).

    Article  CAS  PubMed  Google Scholar 

  91. Zhang, A. et al. LncRNA HOTAIR enhances the androgen-receptor-mediated transcriptional program and drives castration-resistant prostate cancer. Cell Rep. 13, 209–221 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Mather, R. L., Wang, Y. & Crea, F. Is HOTAIR really involved in neuroendocrine prostate cancer differentiation? Epigenomics 10, 1259–1261 (2018).

    Article  CAS  PubMed  Google Scholar 

  93. Luo, J. et al. LncRNA-p21 alters the antiandrogen enzalutamide-induced prostate cancer neuroendocrine differentiation via modulating the EZH2/STAT3 signaling. Nat. Commun. 10, 2571 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

  95. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Orkin, S. H. Diversification of haematopoietic stem cells to specific lineages. Nat. Rev. Genet. 1, 57–64 (2000).

    Article  CAS  PubMed  Google Scholar 

  97. Panman, L. et al. Transcription factor-induced lineage selection of stem-cell-derived neural progenitor cells. Cell Stem Cell 8, 663–675 (2011).

    Article  CAS  PubMed  Google Scholar 

  98. Akamatsu, S. et al. The placental gene PEG10 promotes progression of neuroendocrine prostate cancer. Cell Rep. 12, 922–936 (2015).

    Article  CAS  PubMed  Google Scholar 

  99. Akamatsu, S., Inoue, T., Ogawa, O. & Gleave, M. E. Clinical and molecular features of treatment-related neuroendocrine prostate cancer. Int. J. Urol. 25, 345–351 (2018).

    Article  CAS  PubMed  Google Scholar 

  100. Lapuk, A. V. et al. From sequence to molecular pathology, and a mechanism driving the neuroendocrine phenotype in prostate cancer. J. Pathol. 227, 286–297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Svensson, C. et al. REST mediates androgen receptor actions on gene repression and predicts early recurrence of prostate cancer. Nucleic Acids Res. 42, 999–1015 (2014).

    Article  CAS  PubMed  Google Scholar 

  102. Schoenherr, C. J. & Anderson, D. J. The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes. Science 267, 1360–1363 (1995).

    Article  CAS  PubMed  Google Scholar 

  103. Ballas, N., Grunseich, C., Lu, D. D., Speh, J. C. & Mandel, G. REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell 121, 645–657 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Zhang, X. et al. SRRM4 expression and the loss of REST activity may promote the emergence of the neuroendocrine phenotype in castration-resistant prostate cancer. Clin. Cancer Res. 21, 4698–4708 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Sarkar, A. & Hochedlinger, K. The sox family of transcription factors: versatile regulators of stem and progenitor cell fate. Cell Stem Cell 12, 15–30 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kregel, S. et al. Sox2 is an androgen receptor-repressed gene that promotes castration-resistant prostate cancer. PLoS ONE 8, e53701 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Russo, M. V. et al. SOX2 boosts major tumor progression genes in prostate cancer and is a functional biomarker of lymph node metastasis. Oncotarget 7, 12372–12385 (2016).

    Article  PubMed  Google Scholar 

  108. Choi, Y. J. et al. miR-34 miRNAs provide a barrier for somatic cell reprogramming. Nat. Cell Biol. 13, 1353–1360 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kareta, M. S. et al. Inhibition of pluripotency networks by the Rb tumor suppressor restricts reprogramming and tumorigenesis. Cell Stem Cell 16, 39–50 (2015).

    Article  CAS  PubMed  Google Scholar 

  110. Dominguez, M. H., Ayoub, A. E. & Rakic, P. POU-III transcription factors (Brn1, Brn2, and Oct6) influence neurogenesis, molecular identity, and migratory destination of upper-layer cells of the cerebral cortex. Cereb Cortex 23, 2632–2643 (2013).

    Article  PubMed  Google Scholar 

  111. Cirillo, L. A. et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 (2002).

    Article  CAS  PubMed  Google Scholar 

  112. Iwafuchi-Doi, M. et al. The pioneer transcription factor foxa maintains an accessible nucleosome configuration at enhancers for tissue-specific gene activation. Mol. Cell 62, 79–91 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Carroll, J. S. et al. Genome-wide analysis of estrogen receptor binding sites. Nat. Genet. 38, 1289–1297 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Clarke, C. L. & Graham, J. D. Non-overlapping progesterone receptor cistromes contribute to cell-specific transcriptional outcomes. PLoS ONE 7, e35859 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Gao, N. et al. The role of hepatocyte nuclear factor-3 alpha (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol. Endocrinol. 17, 1484–1507 (2003).

    Article  CAS  PubMed  Google Scholar 

  116. Mirosevich, J., Gao, N. & Matusik, R. J. Expression of Foxa transcription factors in the developing and adult murine prostate. Prostate 62, 339–352 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Kim, J. et al. FOXA1 inhibits prostate cancer neuroendocrine differentiation. Oncogene 36, 4072–4080 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Adams, E. J. et al. FOXA1 mutations alter pioneering activity, differentiation and prostate cancer phenotypes. Nature 571, 408–412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Park, J. W., Lee, J. K., Witte, O. N. & Huang, J. FOXA2 is a sensitive and specific marker for small cell neuroendocrine carcinoma of the prostate. Mod. Pathol. 30, 1262–1272 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Roy, A. et al. Onecut transcription factors act upstream of Isl1 to regulate spinal motoneuron diversification. Development 139, 3109–3119 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Clotman, F. et al. Control of liver cell fate decision by a gradient of TGF beta signaling modulated by Onecut transcription factors. Genes Dev. 19, 1849–1854 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Margagliotti, S. et al. The Onecut transcription factors HNF-6/OC-1 and OC-2 regulate early liver expansion by controlling hepatoblast migration. Dev. Biol. 311, 579–589 (2007).

    Article  CAS  PubMed  Google Scholar 

  123. Espana, A. & Clotman, F. Onecut factors control development of the Locus Coeruleus and of the mesencephalic trigeminal nucleus. Mol. Cell Neurosci. 50, 93–102 (2012).

    Article  CAS  PubMed  Google Scholar 

  124. Espana, A. & Clotman, F. Onecut transcription factors are required for the second phase of development of the A13 dopaminergic nucleus in the mouse. J. Comp. Neurol. 520, 1424–1441 (2012).

    Article  CAS  PubMed  Google Scholar 

  125. Rotinen, M. et al. ONECUT2 is a targetable master regulator of lethal prostate cancer that suppresses the androgen axis. Nat. Med. 24, 1887–1898 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Guo, H. et al. ONECUT2 is a driver of neuroendocrine prostate cancer. Nat. Commun. 10, 278 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Milosevic, M. et al. Tumor hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer. Clin. Cancer Res. 18, 2108–2114 (2012).

    Article  CAS  PubMed  Google Scholar 

  128. Qi, J. et al. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell 18, 23–38 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Calarco, J. A. et al. Regulation of vertebrate nervous system alternative splicing and development by an SR-related protein. Cell 138, 898–910 (2009).

    Article  CAS  PubMed  Google Scholar 

  130. Taylor, B. S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Li, Y. et al. SRRM4 drives neuroendocrine transdifferentiation of prostate adenocarcinoma under androgen receptor pathway inhibition. Eur. Urol. 71, 68–78 (2017).

    Article  CAS  PubMed  Google Scholar 

  132. Lee, A. R. et al. Alternative RNA splicing of the MEAF6 gene facilitates neuroendocrine prostate cancer progression. Oncotarget 8, 27966–27975 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Chapman, G., Sparrow, D. B., Kremmer, E. & Dunwoodie, S. L. Notch inhibition by the ligand DELTA-LIKE 3 defines the mechanism of abnormal vertebral segmentation in spondylocostal dysostosis. Hum. Mol. Genet. 20, 905–916 (2011).

    Article  CAS  PubMed  Google Scholar 

  134. Puca, L. et al. Delta-like protein 3 expression and therapeutic targeting in neuroendocrine prostate cancer. Sci. Transl. Med. 11, eaav0891 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Saunders, L. R. et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl. Med. 7, 302ra136 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Brcic, L. et al. Comparison of four DLL3 antibodies performance in high grade neuroendocrine lung tumor samples and cell cultures. Diagn. Pathol. 14, 47 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Chen, H. et al. Pathogenesis of prostatic small cell carcinoma involves the inactivation of the P53 pathway. Endocr. Relat. Cancer 19, 321–331 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Huang, J. et al. Differential expression of interleukin-8 and its receptors in the neuroendocrine and non-neuroendocrine compartments of prostate cancer. Am. J. Pathol. 166, 1807–1815 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Li, Y. et al. Targeting cellular heterogeneity with CXCR2 blockade for the treatment of therapy-resistant prostate cancer. Sci. Transl. Med. 11, eaax0428 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Hanks, S. K. & Hunter, T. Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J. 9, 576–596 (1995).

    Article  CAS  PubMed  Google Scholar 

  141. Moscat, J. & Diaz-Meco, M. T. The atypical protein kinase Cs. Functional specificity mediated by specific protein adapters. EMBO Rep. 1, 399–403 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Reina-Campos, M. et al. Increased serine and one-carbon pathway metabolism by PKClambda/iota deficiency promotes neuroendocrine prostate cancer. Cancer Cell 35, 385–400 e389 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Tagawa, S. T. Neuroendocrine prostate cancer after hormonal therapy: knowing is half the battle. J. Clin. Oncol. 32, 3360–3364 (2014).

    Article  PubMed  Google Scholar 

  144. Tritschler, S., Erdelkamp, R., Stief, C. & Hentrich, M. Neuroendocrine prostate cancer. Urologe A 56, 1475–1484 (2017).

    Article  CAS  PubMed  Google Scholar 

  145. Amato, R. J. et al. Chemotherapy for small cell carcinoma of prostatic origin. J. Urol. 147, 935–937 (1992).

    Article  CAS  PubMed  Google Scholar 

  146. Dasari, S. & Tchounwou, P. B. Cisplatin in cancer therapy: molecular mechanisms of action. Eur. J. Pharmacol. 740, 364–378 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Montecucco, A., Zanetta, F. & Biamonti, G. Molecular mechanisms of etoposide. EXCLI J. 14, 95–108 (2015).

    PubMed  PubMed Central  Google Scholar 

  148. Chang, H. M., Moudgil, R., Scarabelli, T., Okwuosa, T. M. & Yeh, E. T. H. Cardiovascular complications of cancer therapy: best practices in diagnosis, prevention, and management: part 1. J. Am. Coll. Cardiol. 70, 2536–2551 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  149. Papandreou, C. N. et al. Results of a phase II study with doxorubicin, etoposide, and cisplatin in patients with fully characterized small-cell carcinoma of the prostate. J. Clin. Oncol. 20, 3072–3080 (2002).

    Article  CAS  PubMed  Google Scholar 

  150. Flechon, A. et al. Phase II study of carboplatin and etoposide in patients with anaplastic progressive metastatic castration-resistant prostate cancer (mCRPC) with or without neuroendocrine differentiation: results of the French Genito-Urinary Tumor Group (GETUG) P01 trial. Ann. Oncol. 22, 2476–2481 (2011).

    Article  CAS  PubMed  Google Scholar 

  151. Culine, S. et al. Docetaxel and cisplatin in patients with metastatic androgen independent prostate cancer and circulating neuroendocrine markers. J. Urol. 178, 844–848 discussion 848 (2007).

    Article  CAS  PubMed  Google Scholar 

  152. Corn, P. G. et al. Cabazitaxel plus carboplatin for the treatment of men with metastatic castration-resistant prostate cancers: a randomised, open-label, phase 1-2 trial. Lancet Oncol. 20, 1432–1443 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Beltran, H. et al. A phase II trial of the aurora kinase a inhibitor alisertib for patients with castration-resistant and neuroendocrine prostate cancer: efficacy and biomarkers. Clin. Cancer Res. 25, 43–51 (2019).

    Article  CAS  PubMed  Google Scholar 

  154. Rickman, D. S., Beltran, H., Demichelis, F. & Rubin, M. A. Biology and evolution of poorly differentiated neuroendocrine tumors. Nat. Med. 23, 1–10 (2017).

    Article  CAS  PubMed  Google Scholar 

  155. Alexandrov, L. B. et al. Mutational signatures associated with tobacco smoking in human cancer. Science 354, 618–622 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Horn, L. et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N. Engl. J. Med. 379, 2220–2229 (2018).

    Article  CAS  PubMed  Google Scholar 

  157. Vicier, C., Xie, W., Hamid, A., Evan, C. & Sweeney, C. Impact of new systemic therapies on outcomes of patients with non-metastatic castration resistant prostate cancer (nmCRPC). J. Clin. Oncol. 37, 244–244 (2019).

    Article  Google Scholar 

  158. Nappi, L. et al. Immunogenomic landscape of neuroendocrine small cell prostate cancer. J. Clin. Oncol. 37, 217–217 (2019).

    Article  Google Scholar 

  159. Landon Carter Brown, S. H. et al. Efficacy of the PD-L1 inhibitor avelumab in neuroendocrine or aggressive variant prostate cancer: results from a phase II, single-arm study. J. Clin. Oncol. 39, 1 (2021).

    Google Scholar 

  160. Morgensztern, D. et al. Efficacy and safety of rovalpituzumab tesirine in third-line and beyond patients with DLL3-expressing, relapsed/refractory small-cell lung cancer: results from the phase II trinity study. Clin. Cancer Res. 25, 6958–6966 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Blackhall, F. et al. Efficacy and safety of rovalpituzumab tesirine compared with topotecan as second-line therapy in DLL3-high SCLC: results from the phase 3 TAHOE study. J. Thorac. Oncol. (2021).

  162. Mansfield, A. S. et al. A phase I/II study of rovalpituzumab tesirine in delta-like 3-expressing, advanced solid tumors. J. Clin. Oncol. 38, 3552–3552 (2020).

    Article  Google Scholar 

  163. Isobe, Y. et al. Near infrared photoimmunotherapy targeting DLL3 for small cell lung cancer. EBioMedicine 52, 102632 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  164. Giffin, M. J. et al. AMG 757, a half-life extended, DLL3-targeted bispecific T-cell engager, shows high potency and sensitivity in preclinical models of small cell lung cancer. Clin. Cancer Res. (2020).

  165. Hipp, S. et al. A bispecific DLL3/CD3 IgG-Like T-cell engaging antibody induces antitumor responses in small cell lung cancer. Clin. Cancer Res. 26, 5258–5268 (2020).

    Article  CAS  PubMed  Google Scholar 

  166. Puca, L. et al. Patient derived organoids to model rare prostate cancer phenotypes. Nat. Commun. 9, 2404 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Navone, N. M. et al. Movember GAP1 PDX project: an international collection of serially transplantable prostate cancer patient-derived xenograft (PDX) models. Prostate 78, 1262–1282 (2018).

    Article  CAS  PubMed  Google Scholar 

  168. Ci, X. et al. Conditionally reprogrammed cells from patient-derived xenograft to model neuroendocrine prostate cancer development. Cells 9, 1398 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  169. Gleave, A. M., Ci, X., Lin, D. & Wang, Y. A synopsis of prostate organoid methodologies, applications, and limitations. Prostate 80, 518–526 (2020).

    Article  PubMed  Google Scholar 

  170. Sequist, L. V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 3, 75ra26 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  171. Mohler, J. L. et al. Prostate cancer, version 2.2019, NCCN clinical practice guidelines in oncology. J. Natl Compr. Canc Netw. 17, 479–505 (2019).

    Article  CAS  PubMed  Google Scholar 

  172. Pomerantz, M. M. et al. The association between germline BRCA2 variants and sensitivity to platinum-based chemotherapy among men with metastatic prostate cancer. Cancer 123, 3532–3539 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank all members of the Living Tumor Laboratory (www.livingtumorlab.com) for helpful discussions. This research was supported in part by the Canadian Institutes of Health Research (#141635, #144159, #153081, #173338, Yuzhuo W.), Terry Fox Research Institute (#1062, Yuzhuo W.), Mitacs Accelerate Program (#IT10125, #IT06414, #IT12387, IT14958, Yuzhuo W., NCI grant (P50CA097186) pilot project award (Yuzhuo W.), Prostate Cancer Foundation BC (X.C.), and China Scholar Council award (Yu W.).

Author information

Authors and Affiliations

Authors

Contributions

Yong W. and Yu W. researched data for the article, Yong W., Yu W., X.C. and D.L. wrote the article, Yuzhuo W., D.L., Yong W., Yu W., X.C., S.C. and F.C. made a substantial contribution to discussion of the content of the manuscript and Yuzhuo W., D.L., S.Y.C.C. and F.C. reviewed and edited the manuscript before submission.

Corresponding authors

Correspondence to Dong Lin or Yuzhuo Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Urology thanks A. Lopez-Beltran and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Living tumor lab: https://www.livingtumorlab.com

Glossary

Lineage plasticity

The ability of a cell to convert from one cell type to another, which could refer to the potential of a fully differentiated cell to de-differentiate and then re-differentiate into a different cell lineage status.

Simian virus 40 T antigens

(SV40 T- Ag). Dominant-acting oncoproteins encoded by the polyomavirus SV40, which are capable of inducing malignant transformation of a variety of cell types.

Allelic imbalance

A phenomenon in which the two alleles of a given gene are expressed at different levels in a given cell.

Genomic profiles

Identification of genomic alterations in a particular cell or tissue type.

Divergent clonal evolution

A tumour evolution model, in which different tumour clones share some genetic alterations inherited from a common ancestor cell, but also display unique alterations that are acquired during early evolutionary divergence.

Probasin promoter

The promoter of rat probasin, an androgen-regulated protein, which commonly uses prostate-specific promoters to drive targeting expression of genes of interest to the prostate epithelium.

Transcriptomic profiles

Identification and quantification of RNA transcripts expressed in a particular cell or tissue type.

AR-indifferent status

Sustained growth of prostate tumour cells that is not driven by, or dependent on, canonical AR signalling.

Notch signalling pathway

A highly conserved signalling pathway that occurs through direct interaction between Notch, the receptor, and the Jagged or Delta family of ligands to trigger proteolytic cleavage to release Notch intracellular fragment that functions to regulate transcription.

Human kinome

The complete set of protein kinases expressed in human cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Wang, Y., Ci, X. et al. Molecular events in neuroendocrine prostate cancer development. Nat Rev Urol 18, 581–596 (2021). https://doi.org/10.1038/s41585-021-00490-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-021-00490-0

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing