Includes irregular control of molecules involved in cell survival and death.Upregulation of GATA2 increases IGF2 resulting in docetaxel resistance.Overexpression of certain cytokines including interleukin (IL)-6, IL-8, TGF-β1, CCL2, and MIC-1 promotes the growth and prevent cell death.AR-V7 has shown sensitivity to the microtubule stabilization caused by taxanes thus contributing resistance.
Genes associated in resistance are: HSP, STAT1, NF-κB, STAT3 and clusterin.Decreased p53 activity is associated with a lack of response to docetaxel.Higher expression of class IV β-tubulin and mutations in class 1 β-tubulin, has lead to impaired polymerization in cells resistant to docetaxel.
Chemotherapeutic.Overexpression of ETS related gene (ERG) in prostate cells causes resistance to cabazitaxel.Tumors lacking retinoblastoma exhibit a more favourable response to cabazitaxel.
Re-activation of androgen synthesis in PC is accompanying with resistance to abiraterone in patients.Steroidogenesis: upregulation of steroidal hormones by IL-6.
Enzymes involved in steroidogenesis are AKR1C3, HSD3B2, and AKR1C3.Glucocorticoids minimizes the adverse effects linked to abiraterone treatment and can stimulate AR and trigger androgen independent cell growth.
Resistance in cells is linked to changes in steroidogenesis, glucose metabolism, and autophagy.Genes related to steroid biosynthesis show increased expression, with AKR1C3.Overexpression of p52 leads to enzalutamide resistance, caused by alterations in glucose metabolism and the expression of AR splice variants.
Exhibits an increase in the levels of androgen and its precursors, such as cholesterol, DHEA, and progesterone.AR mutations in the ligand binding domain affecting around 10–30% of CRPC patients.Overexpression of IL-6 leads to constitutive STAT3 activation, as a result enzalutamide becomes ineffective.
Lower likelihood of cross-resistance between cabazitaxel and AR-targeted therapies compared to docetaxel.
Prior treatment with abiraterone or docetaxel may experience a decreased response to subsequent enzalutamide treatment, resulting in diminished PSA decline and progression-free survival.
Authors are thankful to Graphic Era Deemed to be University for providing the environment in which we conducted this review. During the preparation of this work, author(s) used the BioRender.com for preparation of Figures 1 and 2. After using the tool/service, author(s) reviewed and edited the content as needed and take(s) full responsibility for the content of the publication.
Wade CA, Kyprianou N. Profiling Prostate Cancer Therapeutic Resistance.Int J Mol Sci. 2018;19:904. [DOI] [PubMed] [PMC]
Diaz KA, Amaya SL, García-Perdomo HA. Perspectives on prostate cancer: advances and pending challenges for a multidisciplinary oncological approach in South America.Int Urol Nephrol. 2024;56:1–7. [DOI] [PubMed] [PMC]
Leslie SW, Soon-Sutton TL, R I A, Sajjad H, Skelton WP. Prostate Cancer. Treasure Island (FL): StatPearls Publishing; 2023.
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018.CA Cancer J Clin. 2018;68:7–30. [DOI] [PubMed]
Snowden FM. Epidemics and society: From the black death to the present. Yale University Press; 2019.
Park JH, Pyun WY, Park HW. Cancer Metabolism: Phenotype, Signaling and Therapeutic Targets.Cells. 2020;9:2308. [DOI] [PubMed] [PMC]
Le TK, Duong QH, Baylot V, Fargette C, Baboudjian M, Colleaux L, et al. Castration-Resistant Prostate Cancer: From Uncovered Resistance Mechanisms to Current Treatments.Cancers (Basel). 2023;15:5047. [DOI] [PubMed] [PMC]
McCaffery K, Nickel B, Pickles K, Moynihan R, Kramer B, Barratt A, et al. Resisting recommended treatment for prostate cancer: a qualitative analysis of the lived experience of possible overdiagnosis.BMJ Open. 2019;9:e026960. [DOI] [PubMed] [PMC]
Nakazawa M, Paller C, Kyprianou N. Mechanisms of Therapeutic Resistance in Prostate Cancer.Curr Oncol Rep. 2017;19:13. [DOI] [PubMed] [PMC]
Attard G, Murphy L, Clarke NW, Sachdeva A, Jones C, Hoyle A, et al.; STAMPEDE investigators. Abiraterone acetate plus prednisolone with or without enzalutamide for patients with metastatic prostate cancer starting androgen deprivation therapy: final results from two randomised phase 3 trials of the STAMPEDE platform protocol.Lancet Oncol. 2023;24:443–56. [DOI] [PubMed]
Giridharan M, Rupani V, Banerjee S. Signaling Pathways and Targeted Therapies for Stem Cells in Prostate Cancer.ACS Pharmacol Transl Sci. 2022;5:193–206. [DOI] [PubMed] [PMC]
He Y, Xu W, Xiao YT, Huang H, Gu D, Ren S. Targeting signaling pathways in prostate cancer: mechanisms and clinical trials.Signal Transduct Target Ther. 2022;7:198. [DOI] [PubMed] [PMC]
Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012.Int J Cancer. 2015;136:E359–86. [DOI] [PubMed]
Shi T, Ma Y, Cao L, Zhan S, Xu Y, Fu F, et al. B7-H3 promotes aerobic glycolysis and chemoresistance in colorectal cancer cells by regulating HK2.Cell Death Dis. 2019;10:308. [DOI] [PubMed] [PMC]
Kim SK, Cho SW. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment.Front Pharmacol. 2022;13:868695. [DOI] [PubMed] [PMC]
Congregado Ruiz B, Rivero Belenchón I, Lendínez Cano G, Medina López RA. Strategies to Re-Sensitize Castration-Resistant Prostate Cancer to Antiandrogen Therapy.Biomedicines. 2023;11:1105. [DOI] [PubMed] [PMC]
Sejda A, Sigorski D, Gulczyński J, Wesołowski W, Kitlińska J, Iżycka-Świeszewska E. Complexity of Neural Component of Tumor Microenvironment in Prostate Cancer.Pathobiology. 2020;87:87–99. [DOI] [PubMed]
Bahmad HF, Jalloul M, Azar J, Moubarak MM, Samad TA, Mukherji D, et al. Tumor Microenvironment in Prostate Cancer: Toward Identification of Novel Molecular Biomarkers for Diagnosis, Prognosis, and Therapy Development.Front Genet. 2021;12:652747. [DOI] [PubMed] [PMC]
Nepali PR, Kyprianou N. Anoikis in phenotypic reprogramming of the prostate tumor microenvironment.Front Endocrinol (Lausanne). 2023;14:1160267. [DOI] [PubMed] [PMC]
Maitland NJ. Resistance to Antiandrogens in Prostate Cancer: Is It Inevitable, Intrinsic or Induced?Cancers (Basel). 2021;13:327. [DOI] [PubMed] [PMC]
Archer M, Dogra N, Kyprianou N. Inflammation as a Driver of Prostate Cancer Metastasis and Therapeutic Resistance.Cancers (Basel). 2020;12:2984. [DOI] [PubMed] [PMC]
Kallenbach J, Atri Roozbahani G, Heidari Horestani M, Baniahmad A. Distinct mechanisms mediating therapy-induced cellular senescence in prostate cancer.Cell Biosci. 2022;12:200. [DOI] [PubMed] [PMC]
Mijatović S, Maksimović-Ivanić D. Basic Insights into Tumor Microenvironment in PC. In: Kocic G, Hadzi-Djokic J, Simic T, editors. Prostate Cancer: Advancements in the Pathogenesis, Diagnosis and Personalized Therapy. Cham: Springer; 2024. pp. 43–71. [DOI]
Lang JM. Understanding dynamic interactions in the prostate tumor microenvironment.Urol Oncol. 2019;37:532–4. [DOI]
Hill BS, Sarnella A, D’Avino G, Zannetti A. Recruitment of stromal cells into tumor microenvironment promote the metastatic spread of breast cancer.Semin Cancer Biol. 2020;60:202–13. [DOI]
Ioannidou E, Moschetta M, Shah S, Parker JS, Ozturk MA, Pappas-Gogos G, et al. Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets.Int J Mol Sci. 2021;22:9926. [DOI] [PubMed] [PMC]
Bedeschi M, Marino N, Cavassi E, Piccinini F, Tesei A. Cancer-Associated Fibroblast: Role in Prostate Cancer Progression to Metastatic Disease and Therapeutic Resistance.Cells. 2023;12:802. [DOI] [PubMed] [PMC]
Bonollo F, Thalmann GN, Kruithof-de Julio M, Karkampouna S. The Role of Cancer-Associated Fibroblasts in Prostate Cancer Tumorigenesis.Cancers (Basel). 2020;12:1887. [DOI] [PubMed] [PMC]
Bu L, Baba H, Yoshida N, Miyake K, Yasuda T, Uchihara T, et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment.Oncogene. 2019;38:4887–901. [DOI] [PubMed]
Crowley F, Sterpi M, Buckley C, Margetich L, Handa S, Dovey Z. A Review of the Pathophysiological Mechanisms Underlying Castration-resistant Prostate Cancer.Res Rep Urol. 2021;13:457–72. [DOI] [PubMed] [PMC]
Papanikolaou S, Vourda A, Syggelos S, Gyftopoulos K. Cell Plasticity and Prostate Cancer: The Role of Epithelial-Mesenchymal Transition in Tumor Progression, Invasion, Metastasis and Cancer Therapy Resistance.Cancers (Basel). 2021;13:2795. [DOI] [PubMed] [PMC]
Dicken H, Hensley PJ, Kyprianou N. Prostate tumor neuroendocrine differentiation via EMT: The road less traveled.Asian J Urol. 2019;6:82–90. [DOI] [PubMed] [PMC]
Moris L, Cumberbatch MG, Van den Broeck T, Gandaglia G, Fossati N, Kelly B, et al. Benefits and Risks of Primary Treatments for High-risk Localized and Locally Advanced Prostate Cancer: An International Multidisciplinary Systematic Review.Eur Urol. 2020;77:614–27. [DOI] [PubMed]
Testa U, Castelli G, Pelosi E. Cellular and Molecular Mechanisms Underlying Prostate Cancer Development: Therapeutic Implications.Medicines (Basel). 2019;6:82. [DOI] [PubMed] [PMC]
Pederzoli F, Raffo M, Pakula H, Ravera F, Nuzzo PV, Loda M. “Stromal cells in prostate cancer pathobiology: friends or foes?”.Br J Cancer. 2023;128:930–9. [DOI] [PubMed] [PMC]
Bahmad HF. Therapy Resistance in Prostate Cancer: Mechanisms and Insights. Academic Press; 2023. [DOI]
Pearson GW. Control of Invasion by Epithelial-to-Mesenchymal Transition Programs during Metastasis.J Clin Med. 2019;8:646. [DOI] [PubMed] [PMC]
Nallasamy P, Nimmakayala RK, Parte S, Are AC, Batra SK, Ponnusamy MP. Tumor microenvironment enriches the stemness features: the architectural event of therapy resistance and metastasis.Mol Cancer. 2022;21:225. [DOI] [PubMed] [PMC]
Zheng X, Yu C, Xu M. Linking Tumor Microenvironment to Plasticity of Cancer Stem Cells: Mechanisms and Application in Cancer Therapy.Front Oncol. 2021;11:678333. [DOI] [PubMed] [PMC]
Scott LJ. Abiraterone Acetate: A Review in Metastatic Castration-Resistant Prostrate Cancer.Drugs. 2017;77:1565–76. [DOI] [PubMed]
James ND, Spears MR, Sydes MR. Abiraterone in Metastatic Prostate Cancer.N Engl J Med. 2017;377:1696–7. [DOI] [PubMed]
Fizazi K, Tran N, Fein L, Matsubara N, Rodriguez-Antolin A, Alekseev BY, et al.; LATITUDE Investigators. Abiraterone plus Prednisone in Metastatic, Castration-Sensitive Prostate Cancer.N Engl J Med. 2017;377:352–60. [DOI] [PubMed]
Ciccarese C, Nobili E, Grilli D, Casolari L, Rihawi K, Gelsomino F, et al. The safety and efficacy of enzalutamide in the treatment of advanced prostate cancer.Expert Rev Anticancer Ther. 2016;16:681–96. [DOI] [PubMed]
Moreira RB, Debiasi M, Francini E, Nuzzo PV, Velasco G, Maluf FC, et al. Differential side effects profile in patients with mCRPC treated with abiraterone or enzalutamide: a meta-analysis of randomized controlled trials.Oncotarget. 2017;8:84572–8. [DOI] [PubMed] [PMC]
Zhu J, Liao R, Su C, Liang D, Wu J, Qiu K, et al. Toxicity profile characteristics of novel androgen-deprivation therapy agents in patients with prostate cancer: a meta-analysis.Expert Rev Anticancer Ther. 2018;18:193–8. [DOI] [PubMed]
Sumanasuriya S, De Bono J. Treatment of Advanced Prostate Cancer-A Review of Current Therapies and Future Promise.Cold Spring Harb Perspect Med. 2018;8:a030635. [DOI] [PubMed] [PMC]
Rathkopf DE, Antonarakis ES, Shore ND, Tutrone RF, Alumkal JJ, Ryan CJ, et al. Safety and Antitumor Activity of Apalutamide (ARN-509) in Metastatic Castration-Resistant Prostate Cancer with and without Prior Abiraterone Acetate and Prednisone.Clin Cancer Res. 2017;23:3544–51. [DOI] [PubMed] [PMC]
Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, et al.; SPARTAN Investigators. Apalutamide Treatment and Metastasis-free Survival in Prostate Cancer.N Engl J Med. 2018;378:1408–18. [DOI] [PubMed]
Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, et al. ARN-509: a novel antiandrogen for prostate cancer treatment.Cancer Res. 2012;72:1494–503. [DOI] [PubMed] [PMC]
Baydoun A, Sun Y, Sandler HM, Bolla M, Nabid A, Denham JW, et al. Efficacy of Bicalutamide Monotherapy in PC: A Network Meta-Analysis of 10 Randomized Trials.Int J Radiat Oncol Biol Phys. 2022;114:e211–2. [DOI]
Nightingale G, Ryu J. Cabazitaxel (jevtana): a novel agent for metastatic castration-resistant prostate cancer.P T. 2012;37:440–8. [PubMed] [PMC]
Fizazi K, Albiges L, Loriot Y, Massard C. ODM-201: a new-generation androgen receptor inhibitor in castration-resistant prostate cancer.Expert Rev Anticancer Ther. 2015;15:1007–17. [DOI] [PubMed] [PMC]
Fizazi K, Smith MR, Tombal B. Clinical Development of Darolutamide: A Novel Androgen Receptor Antagonist for the Treatment of Prostate Cancer.Clin Genitourin Cancer. 2018;16:332–40. [DOI] [PubMed]
Steinberg M. Degarelix: a gonadotropin-releasing hormone antagonist for the management of prostate cancer.Clin Ther. 2009;31:2312–31. [DOI] [PubMed]
Persson BE, Kold Olesen T, Jensen JK. Degarelix: a new approach for the treatment of prostate cancer.Neuroendocrinology. 2009;90:235–44. [DOI] [PubMed]
Bissery MC, Nohynek G, Sanderink GJ, Lavelle F. Docetaxel (Taxotere): a review of preclinical and clinical experience. Part I: Preclinical experience.Anticancer Drugs. 1995;6:339–55. [DOI] [PubMed]
Pienta KJ. Preclinical mechanisms of action of docetaxel and docetaxel combinations in prostate cancer.Semin Oncol. 2001;28:3–7. [DOI] [PubMed]
Heinlein CA, Chang C. Androgen receptor in prostate cancer.Endocr Rev. 2004;25:276–308. [DOI] [PubMed]
Shore ND, Saltzstein D, Sieber P, Mehlhaff B, Gervasi L, Phillips J, et al. Results of a Real-world Study of Enzalutamide and Abiraterone Acetate With Prednisone Tolerability (REAAcT).Clin Genitourin Cancer. 2019;17:457–63.e6. [DOI] [PubMed]
Nedeljković M, Damjanović A. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer-How We Can Rise to the Challenge.Cells. 2019;8:957. [DOI] [PubMed] [PMC]
Hoter A, Rizk S, Naim HY. The Multiple Roles and Therapeutic Potential of Molecular Chaperones in Prostate Cancer.Cancers (Basel). 2019;11:1194. [DOI] [PubMed] [PMC]
Kalampounias G, Zafeiropoulou K, Androutsopoulou T, Alexis S, Symeonidis A, Katsoris P. The ERK1/2-Elk1, JNK-cJun, and JAK-STAT Transcriptional Axes as Potential Bortezomib Resistance Mediators in Prostate Cancer.bioRxiv [Preprint]. 2024 [cited 2024 May 17]. Available from: https://www.biorxiv.org/content/10.1101/2024.04.15.589569v1
Ehsani M, David FO, Baniahmad A. Androgen Receptor-Dependent Mechanisms Mediating Drug Resistance in Prostate Cancer.Cancers (Basel). 2021;13:1534. [DOI] [PubMed] [PMC]
Kumari S, Sharma V, Tiwari R, Maurya JP, Subudhi BB, Senapati D. Therapeutic potential of p53 reactivation in prostate cancer: Strategies and opportunities.Eur J Pharmacol. 2022;919:174807. [DOI] [PubMed]
Cai M, Song XL, Li XA, Chen M, Guo J, Yang DH, et al. Current therapy and drug resistance in metastatic castration-resistant prostate cancer.Drug Resist Updat. 2023;68:100962. [DOI] [PubMed]
Balaguer FA, Mühlethaler T, Estévez-Gallego J, Calvo E, Giménez-Abián JF, Risinger AL, et al. Crystal Structure of the Cyclostreptin-Tubulin Adduct: Implications for Tubulin Activation by Taxane-Site Ligands.Int J Mol Sci. 2019;20:1392. [DOI] [PubMed] [PMC]
Prassanawar SS, Panda D. Tubulin heterogeneity regulates functions and dynamics of microtubules and plays a role in the development of drug resistance in cancer.Biochem J. 2019;476:1359–76. [DOI] [PubMed]
Rafi IK, Hoque M. Short notes on molecular mechanisms behind antimicrobial drug resistance.GSC Biol Pharm Sci. 2023;24:1–009. [DOI]
Haider T, Pandey V, Banjare N, Gupta PN, Soni V. Drug resistance in cancer: mechanisms and tackling strategies.Pharmacol Rep. 2020;72:1125–51. [DOI] [PubMed]
Hongo H, Kosaka T, Oya M. Analysis of cabazitaxel-resistant mechanism in human castration-resistant prostate cancer.Cancer Sci. 2018;109:2937–45. [DOI] [PubMed] [PMC]
Roshan-Moniri M. Development of ERG inhibitors as potential drugs for the treatment of metastatic prostate cancer [dissertation]. Vancouver: University of British Columbia; 2021. [DOI]
Ruiz de Porras V, Font A, Aytes A. Chemotherapy in metastatic castration-resistant prostate cancer: Current scenario and future perspectives.Cancer Lett. 2021;523:162–9. [DOI] [PubMed]
Bungaro M, Buttigliero C, Tucci M. Overcoming the mechanisms of primary and acquired resistance to new generation hormonal therapies in advanced prostate cancer: focus on androgen receptor independent pathways.Cancer Drug Resist. 2020;3:726–41. [DOI] [PubMed] [PMC]
Mostaghel EA, Marck BT, Plymate SR, Vessella RL, Balk S, Matsumoto AM, 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. [DOI] [PubMed] [PMC]
Xiao L, Wang Y, Xu K, Hu H, Xu Z, Wu D, et al. Nuclear Receptor LRH-1 Functions to Promote Castration-Resistant Growth of Prostate Cancer via Its Promotion of Intratumoral Androgen Biosynthesis.Cancer Res. 2018;78:2205–18. [DOI] [PubMed]
Verma S, Prajapati KS, Kushwaha PP, Shuaib M, Kumar Singh A, Kumar S, et al. Resistance to second generation antiandrogens in prostate cancer: pathways and mechanisms.Cancer Drug Resist. 2020;3:742–61. [DOI] [PubMed] [PMC]
Gao K, Li X, Ni J, Wu B, Guo J, Zhang R, et al. Non-coding RNAs in enzalutamide resistance of castration-resistant prostate cancer.Cancer Lett. 2023;566:216247. [DOI] [PubMed]
Hou Z, Huang S, Li Z. Androgens in prostate cancer: A tale that never ends.Cancer Lett. 2021;516:1–12. [DOI] [PubMed]
Maylin ZR, Nicolescu RC, Pandha H, Asim M. Breaking androgen receptor addiction of prostate cancer by targeting different functional domains in the treatment of advanced disease.Transl Oncol. 2021;14:101115. [DOI] [PubMed] [PMC]
Ha S, Luo G, Xiang H. A Comprehensive Overview of Small-Molecule Androgen Receptor Degraders: Recent Progress and Future Perspectives.J Med Chem. 2022;65:16128–54. [DOI] [PubMed]
Linder S, van der Poel HG, Bergman AM, Zwart W, Prekovic S. Enzalutamide therapy for advanced prostate cancer: efficacy, resistance and beyond.Endocr Relat Cancer. 2018;26:R31–52. [DOI] [PubMed] [PMC]
Wang Y, Chen J, Wu Z, Ding W, Gao S, Gao Y, et al. Mechanisms of enzalutamide resistance in castration-resistant prostate cancer and therapeutic strategies to overcome it.Br J Pharmacol. 2021;178:239–61. [DOI] [PubMed]
Chen MK, Liang ZJ, Luo DS, Xue KY, Liao DY, Li Z, et al. Abiraterone, Orteronel, Enzalutamide and Docetaxel: Sequential or Combined Therapy?Front Pharmacol. 2022;13:843110. [DOI] [PubMed] [PMC]
Cesca MG, Silveira MT, Pandolfi NC, Oliveira TB, Rinck JA, Da Costa AA, et al. Comparison of enzalutamide versus abiraterone in castration-resistant PC before docetaxel: Results of a propensity score-matched analysis.JCO. 2019;37:e16540. [DOI]
Maroto P, Pinto A, de Liaño AG, González-Billalabeitia E, Lorente D, Puente J. Resistance to the Androgen Receptor Centred Therapies: Biology and Management.SN Compr Clin Med. 2021;3:1593–609. [DOI]
Pinto F, Dibitetto F, Ragonese M, Bassi P. Mechanisms of Resistance to Second-Generation Antiandrogen Therapy for Prostate Cancer: Actual Knowledge and Perspectives.Med Sci (Basel). 2022;10:25. [DOI] [PubMed] [PMC]
Farrow JM, Yang JC, Evans CP. Autophagy as a modulator and target in prostate cancer.Nat Rev Urol. 2014;11:508–16. [DOI] [PubMed] [PMC]
Erkisa M, Aydinlik S, Cevatemre B, Aztopal N, Akar RO, Celikler S, et al. A promising therapeutic combination for metastatic prostate cancer: Chloroquine as autophagy inhibitor and palladium(II) barbiturate complex.Biochimie. 2020;175:159–72. [DOI] [PubMed]
Loizzo D, Pandolfo SD, Rogers D, Cerrato C, di Meo NA, Autorino R, et al. Novel Insights into Autophagy and Prostate Cancer: A Comprehensive Review.Int J Mol Sci. 2022;23:3826. [DOI] [PubMed] [PMC]
Grossi V, Lucarelli G, Forte G, Peserico A, Matrone A, Germani A, et al. Loss of STK11 expression is an early event in prostate carcinogenesis and predicts therapeutic response to targeted therapy against MAPK/p38.Autophagy. 2015;11:2102–13. [DOI] [PubMed] [PMC]
Ashrafizadeh M, Paskeh MDA, Mirzaei S, Gholami MH, Zarrabi A, Hashemi F, et al. Targeting autophagy in prostate cancer: preclinical and clinical evidence for therapeutic response.J Exp Clin Cancer Res. 2022;41:105. [DOI] [PubMed] [PMC]
Maughan BL, Antonarakis ES. Androgen pathway resistance in prostate cancer and therapeutic implications.Expert Opin Pharmacother. 2015;16:1521–37. [DOI] [PubMed] [PMC]
Wadosky KM, Koochekpour S. Molecular mechanisms underlying resistance to androgen deprivation therapy in prostate cancer.Oncotarget. 2016;7:64447–70. [DOI] [PubMed] [PMC]
Edwards J, Krishna NS, Grigor KM, Bartlett JM. Androgen receptor gene amplification and protein expression in hormone refractory prostate cancer.Br J Cancer. 2003;89:552–6. [DOI] [PubMed] [PMC]
Rehman Y, Rosenberg JE. Abiraterone acetate: oral androgen biosynthesis inhibitor for treatment of castration-resistant prostate cancer.Drug Des Devel Ther. 2012;6:13–8. [DOI] [PubMed] [PMC]
Jacob A, Raj R, Allison DB, Myint ZW. Androgen Receptor Signaling in Prostate Cancer and Therapeutic Strategies.Cancers (Basel). 2021;13:5417. [DOI] [PubMed] [PMC]
Leung JK, Sadar MD. Non-Genomic Actions of the Androgen Receptor in Prostate Cancer.Front Endocrinol (Lausanne). 2017;8:2. [DOI] [PubMed] [PMC]
Gerratana L, Basile D, Buono G, De Placido S, Giuliano M, Minichillo S, et al. Androgen receptor in triple negative breast cancer: A potential target for the targetless subtype.Cancer Treat Rev. 2018;68:102–10. [DOI] [PubMed]
Shorning BY, Dass MS, Smalley MJ, Pearson HB. The PI3K-AKT-mTOR Pathway and Prostate Cancer: At the Crossroads of AR, MAPK, and WNT Signaling.Int J Mol Sci. 2020;21:4507. [DOI] [PubMed] [PMC]
Hashemi M, Taheriazam A, Daneii P, Hassanpour A, Kakavand A, Rezaei S, et al. Targeting PI3K/Akt signaling in prostate cancer therapy.J Cell Commun Signal. 2023;17:423–43. [DOI] [PubMed] [PMC]
Saxby H, Mikropoulos C, Boussios S. An Update on the Prognostic and Predictive Serum Biomarkers in Metastatic Prostate Cancer.Diagnostics (Basel). 2020;10:549. [DOI] [PubMed] [PMC]
Boussios S, Rassy E, Moschetta M, Ghose A, Adeleke S, Sanchez E, et al. BRCA Mutations in Ovarian and Prostate Cancer: Bench to Bedside.Cancers (Basel). 2022;14:3888. [DOI] [PubMed] [PMC]
Teply BA, Antonarakis ES. Treatment strategies for DNA repair-deficient prostate cancer.Expert Rev Clin Pharmacol. 2017;10:889–98. [DOI] [PubMed] [PMC]
Ghose A, Moschetta M, Pappas-Gogos G, Sheriff M, Boussios S. Genetic Aberrations of DNA Repair Pathways in Prostate Cancer: Translation to the Clinic.Int J Mol Sci. 2021;22:9783. [DOI] [PubMed] [PMC]
Huggins C, Hodges CV. 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. 1972;22:232–40. [DOI] [PubMed]
Boussios S, Rassy E, Shah S, Ioannidou E, Sheriff M, Pavlidis N. Aberrations of DNA repair pathways in prostate cancer: a cornerstone of precision oncology.Expert Opin Ther Targets. 2021;25:329–33. [DOI] [PubMed]
Kornberg Z, Chou J, Feng FY, Ryan CJ. Prostate cancer in the era of “Omic” medicine: recognizing the importance of DNA damage repair pathways.Ann Transl Med. 2018;6:161. [DOI] [PubMed] [PMC]
Saxby H, Boussios S, Mikropoulos C. Androgen Receptor Gene Pathway Upregulation and Radiation Resistance in Oligometastatic Prostate Cancer.Int J Mol Sci. 2022;23:4786. [DOI] [PubMed] [PMC]
Li LY, Guan YD, Chen XS, Yang JM, Cheng Y. DNA Repair Pathways in Cancer Therapy and Resistance.Front Pharmacol. 2021;11:629266. [DOI] [PubMed] [PMC]
Neboori HJ, Haffty BG, Wu H, Yang Q, Aly A, Goyal S, et al. Low p53 binding protein 1 (53BP1) expression is associated with increased local recurrence in breast cancer patients treated with breast-conserving surgery and radiotherapy.Int J Radiat Oncol Biol Phys. 2012;83:e677–83. [DOI] [PubMed]
Beltran H, Rickman DS, Park K, Chae SS, Sboner A, MacDonald TY, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets.Cancer Discov. 2011;1:487–95. [DOI] [PubMed] [PMC]
Lo UG, Chen YA, Cen J, Deng S, Luo J, Zhau H, et al. The driver role of JAK-STAT signalling in cancer stemness capabilities leading to new therapeutic strategies for therapy- and castration-resistant prostate cancer.Clin Transl Med. 2022;12:e978. [DOI] [PubMed] [PMC]
Deng S, Wang C, Wang Y, Xu Y, Li X, Johnson NA, et al. Ectopic JAK-STAT activation enables the transition to a stem-like and multilineage state conferring AR-targeted therapy resistance.Nat Cancer. 2022;3:1071–87. [DOI] [PubMed] [PMC]
Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology E-Book: Cellular and Molecular Immunology E-Book. Elsevier Health Sciences; 2014.
Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors.Clin Cancer Res. 2019;25:3753–8. [DOI] [PubMed]
de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer.N Engl J Med. 2020;382:2091–102. [DOI] [PubMed]
Kim TJ, Lee YH, Koo KC. Current Status and Future Perspectives of Androgen Receptor Inhibition Therapy for Prostate Cancer: A Comprehensive Review.Biomolecules. 2021;11:492. [DOI] [PubMed] [PMC]
Chen Y, Zhou Q, Hankey W, Fang X, Yuan F. Second generation androgen receptor antagonists and challenges in prostate cancer treatment.Cell Death Dis. 2022;13:632. [DOI] [PubMed] [PMC]
Miricescu D, Totan A, Stanescu-Spinu II, Badoiu SC, Stefani C, Greabu M. PI3K/AKT/mTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects.Int J Mol Sci. 2020;22:173. [DOI] [PubMed] [PMC]
Liu C, Lou W, Zhu Y, Nadiminty N, Schwartz CT, Evans CP, et al. Niclosamide inhibits androgen receptor variants expression and overcomes enzalutamide resistance in castration-resistant prostate cancer.Clin Cancer Res. 2014;20:3198–210. [DOI] [PubMed] [PMC]
Zhang Z, Zhou C, Li X, Barnes SD, Deng S, Hoover E, et al. Loss of CHD1 Promotes Heterogeneous Mechanisms of Resistance to AR-Targeted Therapy via Chromatin Dysregulation.Cancer Cell. 2020;37:584–98.e11. [DOI] [PubMed] [PMC]
Howard EW, Lee DT, Chiu YT, Chua CW, Wang X, Wong YC. Evidence of a novel docetaxel sensitizer, garlic-derived S-allylmercaptocysteine, as a treatment option for hormone refractory prostate cancer.Int J Cancer. 2008;122:1941–8. [DOI] [PubMed]
O’Neill AJ, Prencipe M, Dowling C, Fan Y, Mulrane L, Gallagher WM, et al. Characterisation and manipulation of docetaxel resistant prostate cancer cell lines.Mol Cancer. 2011;10:126. [DOI] [PubMed] [PMC]
Armstrong CM, Gao AC. Drug resistance in castration resistant prostate cancer: resistance mechanisms and emerging treatment strategies.Am J Clin Exp Urol. 2015;3:64–76. [PubMed] [PMC]
Sekino Y, Teishima J. Molecular mechanisms of docetaxel resistance in prostate cancer.Cancer Drug Resist. 2020;3:676–85. [DOI] [PubMed] [PMC]
Lima TS, Iglesias-Gato D, Souza LDO, Stenvang J, Lima DS, Røder MA, et al. Molecular Profiling of Docetaxel-Resistant Prostate Cancer Cells Identifies Multiple Mechanisms of Therapeutic Resistance.Cancers (Basel). 2021;13:1290. [DOI] [PubMed] [PMC]
Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic Nanoparticles and Their Targeted Delivery Applications.Molecules. 2020;25:2193. [DOI] [PubMed] [PMC]
Armstrong CM, Gao AC. Dysregulated androgen synthesis and anti-androgen resistance in advanced prostate cancer.Am J Clin Exp Urol. 2021;9:292–300. [PubMed] [PMC]
Bishop JL, Thaper D, Zoubeidi A. The Multifaceted Roles of STAT3 Signaling in the Progression of Prostate Cancer.Cancers (Basel). 2014;6:829–59. [DOI] [PubMed] [PMC]
Saranyutanon S, Srivastava SK, Pai S, Singh S, Singh AP. Therapies Targeted to Androgen Receptor Signaling Axis in Prostate Cancer: Progress, Challenges, and Hope.Cancers (Basel). 2019;12:51. [DOI] [PubMed] [PMC]
Kregel S, Wang C, Han X, Xiao L, Fernandez-Salas E, Bawa P, et al. Androgen receptor degraders overcome common resistance mechanisms developed during prostate cancer treatment.Neoplasia. 2020;22:111–9. [DOI] [PubMed] [PMC]
Liu Y, Yu C, Shao Z, Xia X, Hu T, Kong W, et al. Selective degradation of AR-V7 to overcome castration resistance of prostate cancer.Cell Death Dis. 2021;12:857. [DOI] [PubMed] [PMC]
Henry MC, Riley CM, Hunter I, Elwood JML, Lopez-Fernandez JD, Minty L, et al. Synthesis and Evaluation of Small Molecule Inhibitors of the Androgen Receptor N-Terminal Domain.ACS Med Chem Lett. 2023;14:1800–6. [DOI] [PubMed] [PMC]
Wang Y, Wang Y, Ci X, Choi SYC, Crea F, Lin D, et al. Molecular events in neuroendocrine prostate cancer development.Nat Rev Urol. 2021;18:581–96. [DOI] [PubMed] [PMC]
Patel GK, Chugh N, Tripathi M. Neuroendocrine Differentiation of Prostate Cancer-An Intriguing Example of Tumor Evolution at Play.Cancers (Basel). 2019;11:1405. [DOI] [PubMed] [PMC]
Beltran H, Hruszkewycz A, Scher HI, Hildesheim J, Isaacs J, Yu EY, et al. The Role of Lineage Plasticity in Prostate Cancer Therapy Resistance.Clin Cancer Res. 2019;25:6916–24. [DOI] [PubMed] [PMC]
Chen R, Dong X, Gleave M. Molecular model for neuroendocrine prostate cancer progression.BJU Int. 2018;122:560–70. [DOI] [PubMed]
Shui X, Xu R, Zhang C, Meng H, Zhao J, Shi C. Advances in neuroendocrine prostate cancer research: From model construction to molecular network analyses.Lab Invest. 2022;102:332–40. [DOI] [PubMed]
Mitsiades N, Kaochar S. Androgen receptor signaling inhibitors: post-chemotherapy, pre-chemotherapy and now in castration-sensitive prostate cancer.Endocr Relat Cancer. 2021;28:T19–38. [DOI] [PubMed]
Aldea M, Andre F, Marabelle A, Dogan S, Barlesi F, Soria JC. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies.Cancer Discov. 2021;11:874–99. [DOI] [PubMed]
Shore ND, Ionescu-Ittu R, Laliberté F, Yang L, Lejeune D, Yu L, et al. Beyond Frontline Therapy with Abiraterone and Enzalutamide in Metastatic Castration-Resistant Prostate Cancer: A Real-World US Study.Clin Genitourin Cancer. 2021;19:480–90. [DOI] [PubMed]
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance.Cancers (Basel). 2020;12:3323. [DOI] [PubMed] [PMC]
Luo J, Tian J, Chou F, Lin C, Xing EZ, Zuo L, et al. Targeting the androgen receptor (AR) with AR degradation enhancer ASC-J9® led to increase docetaxel sensitivity via suppressing the p21 expression.Cancer Lett. 2019;444:35–44. [DOI] [PubMed]
Ovejero-Sánchez M, Asensio-Juárez G, González M, Puebla P, Vicente-Manzanares M, Pélaez R, et al. Panobinostat Synergistically Enhances the Cytotoxicity of Microtubule Destabilizing Drugs in Ovarian Cancer Cells.Int J Mol Sci. 2022;23:13019. [DOI] [PubMed] [PMC]