601
Views
0
CrossRef citations to date
0
Altmetric
Research paper

Neuroendocrine gene subsets are uniquely dysregulated in prostate adenocarcinoma

, , , , , , , , , , , , , & ORCID Icon show all
Article: 2364433 | Received 22 Mar 2024, Accepted 02 Jun 2024, Published online: 26 Jun 2024

References

  • Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12–9. doi:10.3322/caac.21820.
  • Haffner MC, Zwart W, Roudier MP, True LD, Nelson WG, Epstein JI, De Marzo AM, Nelson PS, Yegnasubramanian S. Genomic and phenotypic heterogeneity in prostate cancer. Nat Rev Urol. 2021;18(2):79–92. doi:10.1038/s41585-020-00400-w.
  • Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014;371(5):424–33. doi:10.1056/NEJMoa1405095.
  • Sainio M, Visakorpi T, Tolonen T, Ilvesaro J, Bova GS. Expression of neuroendocrine differentiation markers in lethal metastatic castration-resistant prostate cancer. Pathol Res Pract. 2018;214(6):848–56. doi:10.1016/j.prp.2018.04.015.
  • Moller A, Lobb RJ. The evolving translational potential of small extracellular vesicles in cancer. Nat Rev Cancer. 2020;20(12):697–709. doi:10.1038/s41568-020-00299-w.
  • Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17. doi:10.1038/s41556-018-0250-9.
  • Welsh JA, Goberdhan DCI, O’Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024;13(2):e12404. doi:10.1002/jev2.12404.
  • Jankovicova J, Secova P, Michalkova K, Antalikova JT. Tetraspanins, more than markers of extracellular vesicles in reproduction. Int J Mol Sci. 2020;21(20):7568. doi:10.3390/ijms21207568.
  • Santos MF, Rappa G, Fontana S, Karbanová J, Aalam F, Tai D, Li Z, Pucci M, Alessandro R, Morimoto C. et al. Anti-human CD9 fab fragment antibody blocks the extracellular vesicle-mediated increase in Malignancy of colon cancer cells. Cells. 2022;11(16):2474. doi:10.3390/cells11162474.
  • Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478). doi:10.1126/science.aau6977.
  • DeRita RM, Zerlanko B, Singh A, Lu H, Iozzo RV, Benovic JL, Languino LR. c-Src, insulin-like growth factor I receptor, G-Protein-coupled receptor kinases and focal adhesion kinase are enriched into prostate cancer cell exosomes. J Cell Biochem. 2017;118(1):66–73. doi:10.1002/jcb.25611.
  • Krishn SR, Salem I, Quaglia F, Naranjo NM, Agarwal E, Liu Q, Sarker S, Kopenhaver J, McCue PA, Weinreb PH. et al. The αvβ6 integrin in cancer cell-derived small extracellular vesicles enhances angiogenesis. J Extracell Vesicles. 2020;9(1):1763594. doi:10.1080/20013078.2020.1763594.
  • Osti D, Del Bene M, Rappa G, Santos M, Matafora V, Richichi C, Faletti S, Beznoussenko GV, Mironov A, Bachi A, et al. Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin Cancer Res. 2019;25(1):266–76. doi:10.1158/1078-0432.CCR-18-1941.
  • Block T, Zezulinski D, Kaplan DE, Lu J, Zanine S, Zhan T, Doria C, Sayeed A. Circulating messenger RNA variants as a potential biomarker for surveillance of hepatocellular carcinoma. Front Oncol. 2022;12:963641. doi:10.3389/fonc.2022.963641.
  • Hofsli E, Wheeler TE, Langaas M, Laegreid A, Thommesen L. Identification of novel neuroendocrine-specific tumour genes. Br J Cancer. 2008;99(8):1330–39. doi:10.1038/sj.bjc.6604565.
  • Waks AG, Winer EP. Breast cancer treatment: a review. JAMA. 2019;321(3):288–300. doi:10.1001/jama.2018.19323.
  • Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J, Ficarra V. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3(1):17009. doi:10.1038/nrdp.2017.9.
  • Haake SM, Rathmell WK. Renal cancer subtypes: should we be lumping or splitting for therapeutic decision making? Cancer. 2017;123(2):200–09. doi:10.1002/cncr.30314.
  • Mendhiratta N, Muraki P, Sisk AE Jr., Shuch B. Papillary renal cell carcinoma: review. Urol Oncol. 2021;39(6):327–37. doi:10.1016/j.urolonc.2021.04.013.
  • Alaghehbandan R, Przybycin CG, Verkarre V, Mehra R. Chromophobe renal cell carcinoma: novel molecular insights and clinicopathologic updates. Asian J Urol. 2022;9(1):1–11. doi:10.1016/j.ajur.2021.11.010.
  • Stridsberg M. The use of chromogranin, synaptophysin and islet amyloid polypeptide as markers for neuroendocrine tumours. Ups J Med Sci. 1995;100(3):169–99. doi:10.3109/03009739509178905.
  • Louthan O. Chromogranin a in physiology and oncology. Folia Biol (Praha). 2011;57(5):173–81.
  • Soh MA, Garrett SH, Somji S, Dunlevy JR, Zhou XD, Sens MA, Bathula CS, Allen C, Sens DA. Arsenic, cadmium and neuron specific enolase (ENO2, γ-enolase) expression in breast cancer. Cancer Cell Int. 2011;11(1):41. doi:10.1186/1475-2867-11-41.
  • Korse CM, Taal BG, Vincent A, van Velthuysen MLF, Baas P, Buning-Kager JCGM, Linders TC, Bonfrer JMG. Choice of tumour markers in patients with neuroendocrine tumours is dependent on the histological grade. A marker study of chromogranin A, neuron specific enolase, progastrin-releasing peptide and cytokeratin fragments. Eur J Cancer. 2012;48(5):662–71. doi:10.1016/j.ejca.2011.08.012.
  • Astarloa R, Sanchez-Franco F, Cacicedo L, Garcia-Villanueva M. Differential expression of neurofilament triplet proteins in carcinoid tumours: an immunohistochemical study. Br J Cancer. 1991;63(5):715–18. doi:10.1038/bjc.1991.161.
  • Lee JK, Phillips JW, Smith BA, Park JW, Stoyanova T, McCaffrey EF, Baertsch R, Sokolov A, Meyerowitz JG, Mathis C, et al. N-Myc drives neuroendocrine prostate cancer initiated from human prostate epithelial cells. Cancer Cell. 2016;29(4):536–47. doi:10.1016/j.ccell.2016.03.001.
  • Li Z, Xie J, Fei Y, Gao P, Xie Q, Gao W, Xu Z. GDNF family receptor alpha 2 promotes neuroblastoma cell proliferation by interacting with PTEN. Biochem Biophys Res Commun. 2019;510(3):339–44. doi:10.1016/j.bbrc.2018.12.169.
  • Cancer Genome Atlas Research N, Ahn J, Akbani R, Ally A, Amin S, Andry C, Annala M, Aprikian A, Armenia J, Arora A, et al. The molecular taxonomy of primary prostate cancer. Cell. 2015;163(4):1011–25. doi:10.1016/j.cell.2015.10.025.
  • Bowes AL, Tarabichi M, Pillay N, Van Loo P. Leveraging single-cell sequencing to unravel intratumour heterogeneity and tumour evolution in human cancers. J Pathol. 2022;257(4):466–78. doi:10.1002/path.5914.
  • Hoshino A, Kim HS, Bojmar L, Gyan KE, Cioffi M, Hernandez J, Zambirinis CP, Rodrigues G, Molina H, Heissel S, et al. Extracellular Vesicle and particle biomarkers define multiple human cancers. Cell. 2020;182(4):1044–61.e18. doi:10.1016/j.cell.2020.07.009.
  • Krishn SR, Singh A, Bowler N, Duffy AN, Friedman A, Fedele C, Kurtoglu S, Tripathi SK, Wang K, Hawkins A, et al. Prostate cancer sheds the αvβ3 integrin in vivo through exosomes. Matrix Biol. 2019;77:41–57. doi:10.1016/j.matbio.2018.08.004.
  • Matoso A, Epstein JI. Grading of prostate cancer: past, present, and future. Curr Urol Rep. 2016;17(3):25. doi:10.1007/s11934-016-0576-4.
  • Cheng L, Montironi R, Bostwick DG, Lopez-Beltran A, Berney DM. Staging of prostate cancer. Histopathology. 2012;60(1):87–117. doi:10.1111/j.1365-2559.2011.04025.x.
  • Bery F, Cancel M, Chantôme A, Guibon R, Bruyère F, Rozet F, Mahéo K, Fromont G. The calcium-sensing receptor is a marker and potential driver of neuroendocrine differentiation in prostate cancer. Cancers (Basel). 2020;12(4):860. doi:10.3390/cancers12040860.
  • Yamada Y, Beltran H. Clinical and biological features of neuroendocrine prostate cancer. Curr Oncol Rep. 2021;23(2):15. doi:10.1007/s11912-020-01003-9.
  • Bhinder B, Ferguson A, Sigouros M, Uppal M, Elsaeed AG, Bareja R, Alnajar H, Eng KW, Conteduca V, Sboner A, et al. Immunogenomic landscape of neuroendocrine prostate cancer. Clin Cancer Res. 2023;29(15):2933–43. doi:10.1158/1078-0432.CCR-22-3743.
  • Saraswat M, Joenväära S, Musante L, Peltoniemi H, Holthofer H, Renkonen R. N-linked (N-) glycoproteomics of urinary exosomes [Corrected]. Mol Cell Proteomics: mCP. 2015;14(2):263–76. doi:10.1074/mcp.M114.040345.
  • Goetzl EJ, Kapogiannis D, Schwartz JB, Lobach IV, Goetzl L, Abner EL, Jicha GA, Karydas AM, Boxer A, Miller BL, et al. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 2016;30(12):4141–48. doi:10.1096/fj.201600816R.
  • Yang J, Zhang Y, Gao X, Yuan Y, Zhao J, Zhou S, Wang H, Wang L, Xu G, Li X, et al. Plasma-derived exosomal ALIX as a novel biomarker for diagnosis and classification of pancreatic cancer. Front Oncol. 2021;11:628346. doi:10.3389/fonc.2021.628346.
  • Pietrowska M, Zebrowska A, Gawin M, Marczak L, Sharma P, Mondal S, Mika J, Polańska J, Ferrone S, Kirkwood JM, et al. Proteomic profile of melanoma cell-derived small extracellular vesicles in patients’ plasma: a potential correlate of melanoma progression. J Extracell Vesicles. 2021;10(4):e12063. doi:10.1002/jev2.12063.
  • Kim O, Hwangbo C, Tran PT, Lee JH. Syntenin-1-mediated small extracellular vesicles promotes cell growth, migration, and angiogenesis by increasing onco-miRnas secretion in lung cancer cells. Cell Death Dis. 2022;13(2):122. doi:10.1038/s41419-022-04594-2.
  • Folkmanis K, Junk E, Merdane E, Folkmane I, Folkmanis V, Ivanovs I, Eglitis J, Jakubovskis M, Laabs S, Isajevs S, et al. Clinicopathological significance of exosomal proteins CD9 and CD63 and DNA mismatch repair proteins in prostate adenocarcinoma and benign hyperplasia. Diagnostics (Basel). 2022;12(2):287. doi:10.3390/diagnostics12020287.
  • Franceschini GM, Quaini O, Mizuno K, Orlando F, Ciani Y, Ku S-Y, Sigouros M, Rothmann E, Alonso A, Benelli M, et al. Noninvasive detection of neuroendocrine prostate cancer through targeted cell-free DNA methylation. Cancer Discov. 2024;14:424–45. doi:10.1158/2159-8290.CD-23-0754.
  • Mugoni V, Ciani Y, Quaini O, Tomasini S, Notarangelo M, Vannuccini F, Marinelli A, Leonardi E, Pontalti S, Martinelli A. et al. Integrating extracellular vesicle and circulating cell-free DNA analysis using a single plasma aliquot improves the detection of HER2 positivity in breast cancer patients. J Extracell Biol. 2023;2(9):e108. doi:10.1002/jex2.108.
  • Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44(8):e71. doi:10.1093/nar/gkv1507.
  • Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. doi:10.1093/bioinformatics/btp616.
  • Benjamini Y, Hochberg Y. Controlling the false discovery rate - a practical and powerful approach to multiple testing. J Roy Stat Soc B Met. 1995;57(1):289–300. doi:10.1111/j.2517-6161.1995.tb02031.x.
  • Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, Marotz C, Giannopoulou E, Chakravarthi BVSK, Varambally S, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22(3):298–305. doi:10.1038/nm.4045.
  • Salem I, Naranjo NM, Singh A, DeRita R, Krishn SR, Sirman LS, Quaglia F, Duffy A, Bowler N, Sayeed A, et al. Methods for extracellular vesicle isolation from cancer cells. Cancer Drug Resist. 2020;3:1–14. doi:10.20517/cdr.2019.118.
  • Testa A, Quaglia F, Naranjo NM, Verrillo CE, Shields CD, Lin S, Pickles MW, Hamza DF, Von Schalscha T, Cheresh DA, et al. Targeting the αVβ3/NgR2 pathway in neuroendocrine prostate cancer. Matrix Biol. 2023;124:49–62. doi:10.1016/j.matbio.2023.11.003.
  • Naranjo NM, Salem I, Harris MA, Languino LR. IFIT3 (Interferon Induced Protein with Tetratricopeptide Repeats 3) modulates STAT1 expression in small extracellular vesicles. Biochem J. 2021;478(21):3905–21. doi:10.1042/BCJ20210580.
  • Quaglia F, Krishn SR, Sossey-Alaoui K, Rana PS, Pluskota E, Park PH, Shields CD, Lin S, McCue P, Kossenkov AV, et al. The NOGO receptor NgR2, a novel αVβ3 integrin effector, induces neuroendocrine differentiation in prostate cancer. Sci Rep. 2022;12(1):18879. doi:10.1038/s41598-022-21711-5.