REFERENCES

1. American Cancer Society. Cancer Facts & Figures 2018. Available from: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2018.html. [Last accessed on 14 May 2019].

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018;68:7-30.

3. Antonarakis ES, Blackford AL, Garrett-Mayer E, Eisenberger MA. Survival in men with nonmetastatic prostate cancer treated with hormone therapy: a quantitative systematic review. J Clin Oncol 2007;25:4998-5008.

4. Armstrong AJ, Garrett-Mayer E, de Wit R, Tannock I, Eisenberger M. Prediction of survival following first-line chemotherapy in men with castration-resistant metastatic prostate cancer. Clin Cancer Res 2010;16:203-11.

5. Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med 2004;10:33-9.

6. Loriot Y, Fizazi K, de Bono JS, Forer D, Hirmand M, et al. Enzalutamide in castration-resistant prostate cancer patients with visceral disease in the liver and/or lung: outcomes from the randomized controlled phase 3 AFFIRM trial. Cancer 2017;123:253-62.

7. Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, et al. Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin Cancer Res 2015;21:2315-24.

8. Aparicio A, Logothetis CJ, Maity SN. Understanding the lethal variant of prostate cancer: power of examining extremes. Cancer Discov 2011;1:466-8.

9. Alanee S, Moore A, Nutt M, Holland B, Dynda D, et al. Contemporary incidence and mortality rates of neuroendocrine prostate cancer. Anticancer Res 2015;35:4145-50.

10. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015;161:1215-28.

11. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nature medicine 2016;22:298-305.

12. Liu C, Lou W, Zhu Y, Yang JC, Nadiminty N, et al. Intracrine androgens and AKR1C3 activation confer resistance to enzalutamide in prostate cancer. Cancer Res 2015;75:1413-22.

13. Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239-43.

14. Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, et al. Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol 2013;63:920-6.

15. Hara T, Miyazaki J, Araki H, Yamaoka M, Kanzaki N, et al. Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome. Cancer Res 2003;63:149-53.

16. Veldscholte J, Berrevoets CA, Ris-Stalpers C, Kuiper GG, Jenster G, et al. The androgen receptor in LNCaP cells contains a mutation in the ligand binding domain which affects steroid binding characteristics and response to antiandrogens. J Steroid Biochem Mol Biol 1992;41:665-9.

17. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, et al. A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Discov 2013;3:1020-9.

18. Balbas MD, Evans MJ, Hosfield DJ, Wongvipat J, Arora VK, et al. Overcoming mutation-based resistance to antiandrogens with rational drug design. Elife 2013;2:e00499.

19. Rathkopf DE, Morris MJ, Fox JJ, Danila DC, Slovin SF, et al. Phase I study of ARN-509, a novel antiandrogen, in the treatment of castration-resistant prostate cancer. J Clin Oncol 2013;31:3525-30.

20. Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res 2009;69:16-22.

21. Guo Z, Yang X, Sun F, Jiang R, Linn DE, et al. A novel androgen receptor splice variant is up-regulated during prostate cancer progression and promotes androgen depletion-resistant growth. Cancer Res 2009;69:2305-13.

22. Sun S, Sprenger CC, Vessella RL, Haugk K, Soriano K, et al. Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant. J Clinical Invest 2010;120:2715-30.

23. Ahrens-Fath I, Politz O, Geserick C, Haendler B. Androgen receptor function is modulated by the tissue-specific AR45 variant. FEBS J 2005;272:74-84.

24. Kohli M, Ho Y, Hillman DW, Van Etten JL, Henzler C, et al. Androgen receptor variant AR-V9 is coexpressed with AR-V7 in prostate cancer metastases and predicts abiraterone resistance. Clin Cancer Res 2017;23:4704-15.

25. Antonarakis ES, Lu C, Wang H, Luber B, Nakazawa M, et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 2014;371:1028-38.

26. Zhang X, Morrissey C, Sun S, Ketchandji M, Nelson PS, et al. Androgen receptor variants occur frequently in castration resistant prostate cancer metastases. PLoS One 2011;6:e27970.

27. Hornberg E, Ylitalo EB, Crnalic S, Antti H, Stattin P, et al. Expression of androgen receptor splice variants in prostate cancer bone metastases is associated with castration-resistance and short survival. PLoS One 2011;6:e19059.

28. Dehm SM, Schmidt LJ, Heemers HV, Vessella RL, Tindall DJ. Splicing of a novel androgen receptor exon generates a constitutively active androgen receptor that mediates prostate cancer therapy resistance. Cancer Res 2008;68:5469-77.

29. Li Y, Alsagabi M, Fan D, Bova GS, Tewfik AH, et al. Intragenic rearrangement and altered RNA splicing of the androgen receptor in a cell-based model of prostate cancer progression. Cancer Res 2011;71:2108-17.

30. Mostaghel EA, Marck BT, Plymate SR, Vessella RL, Balk S, et al. Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: induction of steroidogenesis and androgen receptor splice variants. Clin Cancer Res 2011;17:5913-25.

31. Li Y, Chan SC, Brand LJ, Hwang TH, Silverstein KA, et al. Androgen receptor splice variants mediate enzalutamide resistance in castration-resistant prostate cancer cell lines. Cancer Res 2013;73:483-9.

32. Watson PA, Chen YF, Balbas MD, Wongvipat J, Socci ND, et al. Constitutively active androgen receptor splice variants expressed in castration-resistant prostate cancer require full-length androgen receptor. Proc Natl Acad Sci U S A 2010;107:16759-65.

33. Hu R, Lu C, Mostaghel EA, Yegnasubramanian S, Gurel M, et al. Distinct transcriptional programs mediated by the ligand-dependent full-length androgen receptor and its splice variants in castration-resistant prostate cancer. Cancer Res 2012;72:3457-62.

34. Hu R, Isaacs WB, Luo J. A snapshot of the expression signature of androgen receptor splicing variants and their distinctive transcriptional activities. Prostate 2011;71:1656-67.

35. Cronauer MV, Hittmair A, Eder IE, Hobisch A, Culig Z, et al. Basic fibroblast growth factor levels in cancer cells and in sera of patients suffering from proliferative disorders of the prostate. Prostate 1997;31:223-33.

36. Luo J, Attard G, Balk SP, Bevan C, Burnstein K, et al. Role of androgen receptor variants in prostate cancer: report from the 2017 Mission Androgen Receptor Variants Meeting. Eur Urol 2018;73:715-23.

37. Chan SC, Li Y, Dehm SM. Androgen receptor splice variants activate androgen receptor target genes and support aberrant prostate cancer cell growth independent of canonical androgen receptor nuclear localization signal. J Biol Chem 2012;287:19736-49.

38. Liu C, Lou W, Zhu Y, Nadiminty N, Schwartz CT, et al. Niclosamide inhibits androgen receptor variants expression and overcomes enzalutamide resistance in castration-resistant prostate cancer. Clin Cancer Res 2014;20:3198-210.

39. Nadiminty N, Chun JY, Lou W, Lin X, Gao AC. NF-kappaB2/p52 enhances androgen-independent growth of human LNCaP cells via protection from apoptotic cell death and cell cycle arrest induced by androgen-deprivation. Prostate 2008;68:1725-33.

40. Nadiminty N, Dutt S, Tepper C, Gao AC. Microarray analysis reveals potential target genes of NF-kappaB2/p52 in LNCaP prostate cancer cells. Prostate 2010;70:276-87.

41. Nadiminty N, Tummala R, Liu C, Yang J, Lou W, et al. NF-kappaB2/p52 induces resistance to enzalutamide in prostate cancer: role of androgen receptor and its variants. Mol Cancer Ther 2013;12:1629-37.

42. Moigne RL, Zhou HJ, Obst JK, Banuelos CA, Jian K, et al. Lessons learned from the metastatic castration-resistant prostate cancer phase I trial of EPI-506, a first-generation androgen receptor N-terminal domain inhibitor. J Clin Oncol 2019;37:257.

43. Fronsdal K, Engedal N, Slagsvold T, Saatcioglu F. CREB binding protein is a coactivator for the androgen receptor and mediates cross-talk with AP-1. J Biol Chem 1998;273:31853-9.

44. Bevan CL, Hoare S, Claessens F, Heery DM, Parker MG. The AF1 and AF2 domains of the androgen receptor interact with distinct regions of SRC1. Mol Cell Biol 1999;19:8383-92.

45. Ueda T, Mawji NR, Bruchovsky N, Sadar MD. Ligand-independent activation of the androgen receptor by interleukin-6 and the role of steroid receptor coactivator-1 in prostate cancer cells. J Biol Chem 2002;277:38087-94.

46. Reid J, Murray I, Watt K, Betney R, McEwan IJ. The androgen receptor interacts with multiple regions of the large subunit of general transcription factor TFIIF. J Biol Chem 2002;277:41247-53.

47. Gao N, Zhang J, Rao MA, Case TC, Mirosevich J, et al. The role of hepatocyte nuclear factor-3 alpha (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Mol Endocrinol 2003;17:1484-507.

48. Hsieh CL, Botta G, Gao S, Li T, Van Allen EM, et al. PLZF, a tumor suppressor genetically lost in metastatic castration-resistant prostate cancer, is a mediator of resistance to androgen deprivation therapy. Cancer Res 2015;75:1944-8.

49. Agoulnik IU, Vaid A, Bingman WE 3rd, Erdeme H, Frolov A, et al. Role of SRC-1 in the promotion of prostate cancer cell growth and tumor progression. Cancer Res 2005;65:7959-67.

50. Agoulnik IU, Vaid A, Nakka M, Alvarado M, Bingman WE 3rd, et al. Androgens modulate expression of transcription intermediary factor 2, an androgen receptor coactivator whose expression level correlates with early biochemical recurrence in prostate cancer. Cancer Res 2006;66:10594-602.

51. Comuzzi B, Nemes C, Schmidt S, Jasarevic Z, Lodde M, et al. The androgen receptor co-activator CBP is up-regulated following androgen withdrawal and is highly expressed in advanced prostate cancer. J Pathol 2004;204:159-66.

52. Debes JD, Sebo TJ, Lohse CM, Murphy LM, Haugen DA, et al. p300 in prostate cancer proliferation and progression. Cancer Res 2003;63:7638-40.

53. Zhou HJ, Yan J, Luo W, Ayala G, Lin SH, et al. SRC-3 is required for prostate cancer cell proliferation and survival. Cancer Res 2005;65:7976-83.

54. Tran C, Ouk S, Clegg NJ, Chen Y, Watson PA, Arora V, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 2009;324:787-90.

55. Yang YC, Banuelos CA, Mawji NR, Wang J, Kato M, et al. Targeting androgen receptor activation function-1 with EPI to overcome resistance mechanisms in castration-resistant prostate cancer. Clin Cancer Res 2016;22:4466-77.

56. Luisi BF, Xu WX, Otwinowski Z, Freedman LP, Yamamoto KR, et al. Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature 1991;352:497-505.

57. Savory JG, Prefontaine GG, Lamprecht C, Liao M, Walther RF, et al. Glucocorticoid receptor homodimers and glucocorticoid-mineralocorticoid receptor heterodimers form in the cytoplasm through alternative dimerization interfaces. Mol Cell Biol 2001;21:781-93.

58. Galon J, Franchimont D, Hiroi N, Frey G, Boettner A, et al. Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J 2002;16:61-71.

59. Lu NZ, Collins JB, Grissom SF, Cidlowski JA. Selective regulation of bone cell apoptosis by translational isoforms of the glucocorticoid receptor. Mol Cell Biol 2007;27:7143-60.

60. Ren R, Oakley RH, Cruz-Topete D, Cidlowski JA. Dual role for glucocorticoids in cardiomyocyte hypertrophy and apoptosis. Endocrinology 2012;153:5346-60.

61. Arora VK, Schenkein E, Murali R, Subudhi SK, Wongvipat J, et al. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 2013;155:1309-22.

62. Kiemer AK, Takeuchi K, Quinlan MP. Identification of genes involved in epithelial-mesenchymal transition and tumor progression. Oncogene 2001;20:6679-88.

63. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest 2002;110:341-50.

64. Janda E, Lehmann K, Killisch I, Jechlinger M, Herzig M, et al. Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis: dissection of Ras signaling pathways. J Cell Biol 2002;156:299-313.

65. Xue C, Plieth D, Venkov C, Xu C, Neilson EG. The gatekeeper effect of epithelial-mesenchymal transition regulates the frequency of breast cancer metastasis. Cancer Res 2003;63:3386-94.

66. Liu Q, Tong D, Liu G, Xu J, Do K, et al. Metformin reverses prostate cancer resistance to enzalutamide by targeting TGF-beta1/STAT3 axis-regulated EMT. Cell Death Dis 2017;8:e3007.

67. Miao L, Yang L, Li R, Rodrigues DN, Crespo M, et al. Disrupting androgen receptor signaling induces Snail-mediated epithelial-mesenchymal plasticity in prostate cancer. Cancer Res 2017;77:3101-12.

68. Martin SK, Pu H, Penticuff JC, Cao Z, Horbinski C, et al. Multinucleation and mesenchymal-to-epithelial transition alleviate resistance to combined cabazitaxel and antiandrogen therapy in advanced prostate cancer. Cancer Res 2016;76:912-26.

69. Ware KE, Somarelli JA, Schaeffer D, Li J, Zhang T, et al. Snail promotes resistance to enzalutamide through regulation of androgen receptor activity in prostate cancer. Oncotarget 2016;7:50507-21.

70. Kong D, Sethi S, Li Y, Chen W, Sakr WA, et al. Androgen receptor splice variants contribute to prostate cancer aggressiveness through induction of EMT and expression of stem cell marker genes. Prostate 2015;75:161-74.

71. Cai C, Chen S, Ng P, Bubley GJ, Nelson PS, et al. Intratumoral de novo steroid synthesis activates androgen receptor in castration-resistant prostate cancer and is upregulated by treatment with CYP17A1 inhibitors. Cancer Res 2011;71:6503-13.

72. Ishizaki F, Nishiyama T, Kawasaki T, Miyashiro Y, Hara N, et al. Androgen deprivation promotes intratumoral synthesis of dihydrotestosterone from androgen metabolites in prostate cancer. Sci Rep 2013;3:1528.

73. Locke JA, Guns ES, Lubik AA, Adomat HH, Hendy SC, et al. Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res 2008;68:6407-15.

74. Mohler JL, Titus MA, Bai S, Kennerley BJ, Lih FB, et al. Activation of the androgen receptor by intratumoral bioconversion of androstanediol to dihydrotestosterone in prostate cancer. Cancer Res 2011;71:1486-96.

75. Fankhauser M, Tan Y, Macintyre G, Haviv I, Hong MK, et al. Canonical androstenedione reduction is the predominant source of signaling androgens in hormone-refractory prostate cancer. Clin Cancer Res 2014;20:5547-57.

76. Labrie F, Luu-The V, Lin SX, Labrie C, Simard J, et al. The key role of 17 beta-hydroxysteroid dehydrogenases in sex steroid biology. Steroids 1997;62:148-58.

77. Bauman DR, Steckelbroeck S, Williams MV, Peehl DM, Penning TM. Identification of the major oxidative 3alpha-hydroxysteroid dehydrogenase in human prostate that converts 5alpha-androstane-3alpha, 17beta-diol to 5alpha-dihydrotestosterone: a potential therapeutic target for androgen-dependent disease. Mol endocrinol 2006;20:444-58.

78. Geng H, Xue C, Mendonca J, Sun XX, Liu Q, et al. Interplay between hypoxia and androgen controls a metabolic switch conferring resistance to androgen/AR-targeted therapy. Nat commun 2018;9:4972.

79. Massie CE, Lynch A, Ramos-Montoya A, Boren J, Stark R, et al. The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO J 2011;30:2719-33.

80. Tsouko E, Khan AS, White MA, Han JJ, Shi Y, et al. Regulation of the pentose phosphate pathway by an androgen receptor-mTOR-mediated mechanism and its role in prostate cancer cell growth. Oncogenesis 2014;3:e103.

81. Reina-Campos M, Linares JF, Duran A, Cordes T, L’Hermitte A, et al. Increased serine and one-carbon pathway metabolism by PKClambda/iota deficiency promotes neuroendocrine prostate cancer. Cancer Cell 2019;35:385-400.e9.

82. Ku SY, Rosario S, Wang Y, Mu P, Seshadri M, et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 2017;355:78-83.

83. Mu P, Zhang Z, Benelli M, Karthaus WR, Hoover E, et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 2017;355:84-8.

84. Bishop JL, Thaper D, Vahid S, Davies A, Ketola K, et al. The master neural transcription factor BRN2 is an androgen receptor-suppressed driver of neuroendocrine differentiation in prostate cancer. Cancer Discov 2017;7:54-71.

85. Beltran H, Rickman DS, Park K, Chae SS, Sboner A, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov 2011;1:487-95.

86. Dardenne E, Beltran H, Benelli M, Gayvert K, Berger A, et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell 2016;30:563-77.

87. Mosquera JM, Beltran H, Park K, MacDonald TY, Robinson BD, et al. Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia 2013;15:1-10.

88. Yin Y, Xu L, Chang Y, Zeng T, Chen X, et al. N-Myc promotes therapeutic resistance development of neuroendocrine prostate cancer by differentially regulating miR-421/ATM pathway. Mol Cancer 2019;18:11.

Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/