Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2187 2023-10-30 10:17:36 |
2 only format change Meta information modification 2187 2023-10-31 02:41:18 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Le, T.K.; Duong, Q.H.; Baylot, V.; Fargette, C.; Baboudjian, M.; Colleaux, L.; Taïeb, D.; Rocchi, P. Mechanisms Underlying Castration-Resistant Prostate Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/50924 (accessed on 04 September 2024).
Le TK, Duong QH, Baylot V, Fargette C, Baboudjian M, Colleaux L, et al. Mechanisms Underlying Castration-Resistant Prostate Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/50924. Accessed September 04, 2024.
Le, Thi Khanh, Quang Hieu Duong, Virginie Baylot, Christelle Fargette, Michael Baboudjian, Laurence Colleaux, David Taïeb, Palma Rocchi. "Mechanisms Underlying Castration-Resistant Prostate Cancer" Encyclopedia, https://encyclopedia.pub/entry/50924 (accessed September 04, 2024).
Le, T.K., Duong, Q.H., Baylot, V., Fargette, C., Baboudjian, M., Colleaux, L., Taïeb, D., & Rocchi, P. (2023, October 30). Mechanisms Underlying Castration-Resistant Prostate Cancer. In Encyclopedia. https://encyclopedia.pub/entry/50924
Le, Thi Khanh, et al. "Mechanisms Underlying Castration-Resistant Prostate Cancer." Encyclopedia. Web. 30 October, 2023.
Mechanisms Underlying Castration-Resistant Prostate Cancer
Edit

Prostate cancer (PC) is the second most common cancer in men worldwide. Despite recent advances in diagnosis and treatment, castration-resistant prostate cancer (CRPC) remains a significant medical challenge. Prostate cancer cells can develop mechanisms to resist androgen deprivation therapy, such as AR overexpression, AR mutations, alterations in AR coregulators, increased steroidogenic signaling pathways, outlaw pathways, and bypass pathways. Various treatment options for CRPC exist, including androgen deprivation therapy, chemotherapy, immunotherapy, localized or systemic therapeutic radiation, and PARP inhibitors. However, more research is needed to combat CRPC effectively. Further investigation into the underlying mechanisms of the disease and the development of new therapeutic strategies will be crucial in improving patient outcomes. 

castration-resistant prostate cancer androgen

1. Introduction

Prostate cancer (PC) is the second most prevalent cancer in men worldwide, representing 15% of all cancers diagnosed in males [1]. In the US, it is the most frequently diagnosed cancer and the second leading cause of cancer deaths in men. An estimated 288,300 new PC cases and 34,700 deaths from the disease are predicted to occur in the US in 2023 [2]. Patients who have advanced PC initially respond very well to androgen deprivation therapy. However, the treatment finally selects cancer cells that relapse into androgen deprivation, leading to the emergence of castration-resistant prostate cancer (CRPC). Patients with metastatic CRPC (mCRPC) have reduced life expectancy, with a median overall survival rate of below 2 years. The current definition CRPC, according to the European Association of Urology (EAU), is based on biochemical and/or clinical parameters demonstrating cancer progression in a castration environment (testosterone level < 50 ng/dL or 1.7 nmol/L) and standards proposed by Prostate Cancer Working Group 3 (PCWG3) [3] and/or the Response Evaluation Criteria in Solid Tumors (RECIST 1.1) [4]. Accordingly, biological progression refers to a condition where there are three consecutive increases in PSA (Prostate Specific Antigen), each at least one week apart, resulting in two increases of at least 50% over the lowest point, and a PSA level greater than 2 ng/mL. Radiographic progression is defined by the appearance of at least two new lesions on a bone scan or the progression of a measurable lesion according to the RECIST [5].
Over the past decade, there has been significant improvement in the treatment options for CRPC. Several drugs that increase overall patient survival, including androgen-receptor (AR) signaling inhibitors (such as abiraterone acetate, enzalutamide, apalutamide, and darolutamide), and radiopharmaceutical therapies (like radium-223 and 177Lu-PSMA-617) have been approved [6]. Advancements in precision medicine have led to the discovery of distinct subtypes of prostate cancer and the identification of genetic changes that predict the effectiveness of specific treatments [7][8].

2. Mechanisms Underlying Castration-Resistant Prostate Cancer

There are currently many treatment options available for CRPC patients; however, the responses to these therapies are still limited. Knowledge of the underlying mechanisms associated with the CRPC phenotype can help to identify new promising therapeutics.

2.1. Androgen Receptor Overexpression

Androgen Receptor (AR) overexpression enables the survival and proliferation of tumor cells in limited-androgen conditions during androgen suppression treatment. Elevated AR expression at both the mRNA and protein levels has been frequently observed in CRPC. Amplification of the AR has been observed in 70% of cases of CRPC and is associated with significantly increased AR mRNA expression [9]. David A. et al. reported the existence of an enhancer that is amplified in 81% of castration-resistant metastatic patients. This enhancer has the potential to increase the expression of the AR gene independently of AR locus amplification in response to first-line androgen deprivation therapy (ADT) [9]. It should be noted that AR amplification is only seen in prostate tumors that have been exposed to androgen deprivation, indicating that AR amplification is a consequence of hormone therapy [10]. Additionally, epigenetic modifications, such as DNA methylation, histone acetylation, and miRNA modulation, could lead to AR overexpression in CRPC (Figure 1) [11].
Figure 1. Mechanisms of castration-resistant prostate cancer (CRPC). AR overexpression enables the survival and proliferation of tumor cells in limited-androgen conditions during androgen suppression treatment. A high AR expression level can be due to AR gene amplification, epigenetics, and miRNA modulation (1). Point mutations in the ligand-binding domain of the AR gene lead to the increased affinity of the mutated AR (mAR) to other hormones, such as progesterone and estrogen, thereby modulating androgen-responsive gene transcription independently from androgen (2). The emergence of AR-splicing isoforms, such as AR3 (also called AR-V7), AR4, and AR5, encoding the truncated AR protein (tAR), which lacks the ligand-binding domain, results in constitutive activation of the AR, thereby promoting variant-carrying tumor cells to ignore the need for androgen (3). Overexpression of AR coactivators and the decreased expression of AR corepressors will result in an increase in AR-regulated transcription (4). Increased production of 5α-reductase can provide sufficient androgens for AR activation in cancer cells (5). CRPC can be induced by outlaw pathways in which AR signaling can be activated in a ligand-independent manner by other molecules than androgens, such as growth factors, cytokines, and kinases (6). The bypass pathways, involved in CRPC progression, increase the activity of MAPK, PI3K, and PCK cascades, leading to either the stimulation of alternative growth pathways or the enhancement of survival signaling independently from AR signaling. Alternative pathways also involve the overexpression of heat-shock protein 27 (HSP27), which mediates its cytoprotective function by protecting its interacting proteins (eIF4E, TCTP, Menin, DDX5) from their degradation by the proteasome (7) (↑ in red: increase, ↓in red: decrease).

2.2. AR Mutations

AR gene mutations have been found in around 10–20% of CRPC cases, while they are rarely observed in localized PC (0–4%) [12][13]. Most recurrent AR mutations are found in the ligand-binding domain (LBD) and/or cofactor binding regions [14]. These alterations could decrease the specificity of the AR for its main ligand, androgen; however, they allow AR specificity to extend to other hormones, such as progesterone and estrogen [12][15][16][17][18]. In addition, they could also disrupt the effectiveness of AR antagonists, causing them to act as AR agonists and resulting in resistance to treatments [19]. It has been reported that the H875Y mutation in CWR22 causes cells to exhibit altered ligand specificity. This mutant AR has shown transcriptional activity in response to testicular androgens, similarly to the wild-type AR. However, it differs from the wild type in its additional activation by adrenal androgens, dehydroepiandrosterone, and the antiandrogen hydroxyflutamide, as well as by estradiol and progesterone [20]. Along with AR coding mutations, several AR splicing mutations, leading to the abnormal expression of splice variants such as AR3 (also called AR-V7), AR4, and AR5, which lack the ligand-binding domain, are frequently observed in CRPC [21]. These variants encode a shorter receptor that lacks the ligand-binding domain and thereby constitutively activates AR pathways [22].

2.3. Alteration in AR CoRegulators

As a transcription factor (TF), the AR cooperates with a variety of regulatory proteins to form a productive transcription complex. These coregulators can either enhance (coactivators) or depress (cosuppressors) the transcriptional activity of the AR, thereby modulating the expression of androgen-regulated genes. As a consequence, alteration in the expression of AR coregulators can give a survival advantage to the cancer cells during castration therapy, promoting CRPC progression [23]. Several well-documented AR coactivators, including SRC1, SRC2, SRC3, ARA70, PIAS1, and Tip60, interact with the AR and enhance its transcriptional activity [24]. Levels of TIF2 and SRC1 expression have been observed to rise alongside the expression of the AR during the progression of prostate cancer cells following androgen deprivation. A previous study has also shown that these proteins are overexpressed in the majority of CRPC samples. The overexpression of TIF2 enhances the transcriptional activation of the AR in response to weaker steroids, such as adrenal androgens (DHEA and androstenedione), in two cell lines carrying AR mutations: LNCaP (T877A) and CWR22 (H874Y) [25]. The steroid receptor coactivator-3 (SRC3), also called amplified-in-breast cancer-1 (AIB1), has been identified as a potent coactivator of the hormone-activated AR [26] and has been demonstrated to promote CRPC progression [27]. Conversely, two well-known corepressors of the AR, NCoR (nuclear receptor corepressor) and SMRT (silencing mediator for retinoid and thyroid hormone receptors), have been shown to interact with the AR and decrease its transcriptional activity triggered by dihydrotestosterone [28][29][30].

2.4. Increased Steroidogenic Signaling Pathways

Accumulating evidence has reported that PC cells could survive after castration therapy by regulating intracrine androgen synthesis within the prostate. A typical example is the 5α-reductase enzyme, which catalyzes the conversion of testosterone (the most abundant free androgen) to the higher-affinity ligand 5α-dihydrotestosterone (DHT). Upon ADT, intraprostatic testosterone and DHT levels do not decline as markedly as do serum levels after ADT, buffering tumor cells from the loss of testicular androgen [31]. Increased 5α-reductase enzyme levels result from a polymorphism where there is a substitution of a valine with a leucine at the codon 89 [32]. Intraprostatic androgens can also be synthesized from cholesterol or other molecular precursors, such as DHEA (dehydroepiandrosterone). Indeed, DHEA could be converted into androstenedione, a substrate for conversion to testosterone. Another report has shown that CRPC samples have higher levels of expression of a variety of enzymes involved in de novo steroid synthesis, such as FASN, CYP17A1, CYP19A1, and UGT2B17, compared to primary PC [33].

2.5. Outlaw Pathways

In addition of being activated primarily by endogenous androgen ligands, the AR protein can also be stimulated through ligand-independent mechanisms, which are known as outlaw pathways. CRPC can thus be induced by the interactions between the cytosolic AR and many molecules such as different growth factors, cytokines, and kinases. Several growth factors, such as insulin-like growth factor-I (IGF-I), keratinocyte growth factor (KGF) and the epidermal growth factor (EGF), enable the stimulation of AR transcription activity at limited androgen levels or even in the absence of androgens [17]. Particularly, IGF1 has been reported to interact with the AR, facilitate AR translocation, and modulate the transcription of androgen-responsive genes. Moreover, it has been demonstrated to upregulate the expression of the AR coactivator TIF2 [34]. Along with growth factors, many cytokines, such as IL-6 and IL-8, have proven their ability to activate AR signaling in a ligand-independent manner [35]. Additionally, receptor tyrosine kinases (RTK) such as HER2/ERBB2 have been shown to be overexpressed in CRPC and restore AR signaling in low-androgen-concentration conditions [25].

2.6. Non-AR-Related Pathways: Bypass Pathways

While the mechanisms described above rely on AR transactivation to promote CRPC progression, alternative survival and growth pathways that are independent of AR activation can also drive androgen deprivation resistance. Of note, many outlaw pathways can also drive CRPC progression in an AR-signaling independent manner. High levels of serum insulin-like growth factor-I (IGF-I) and its receptor (IGF-1R) have been increasingly recognized to play a key role in prostate cancer progression. Binding of IGF-I with IGF-IR enables the stimulation of the transduction of a signal cascade, which, in turn, activates the expression of genes responsible for cellular growth and proliferation [10][23]. The upregulation of Akt pathway has been reported in a variety of human malignancies, including prostate cancer [36]. Indeed, AKT plays a central role in antiapoptotic pathways by phosphorylating and depressing Bcl2-associated agonist of cell death (BAD), a proapoptotic member of the BCL-2 protein family, and pro-caspase 9 [37]. AKT also promotes cellular proliferation by decreasing the expression of p27, a cell cycle inhibitor [38]. Several reports have shown that loss of PTEN, a tumor suppressor that inhibits PI3K/AKT signaling, frequently occurs in CRPC samples [39].
In addition, the heat-shock protein Hsp27 has previously been demonstrated to drive therapy resistance in PC, and its inhibitors, such as Hsp27 ASO (OGX-427) and small interference RNA (siRNA), have shown the ability to enhance chemotherapy. More recently, people have shown that Hsp27 plays a role in promoting the development of CRPC through its chaperone activity by selectively protecting some partner proteins involved in castration resistance, such as eIF4E, TCTP, Menin, and DDX5, from their ubiquitin–proteasome degradation [40][41][42][43]. CRPC has the potential to develop resistance to the antiandrogen enzalutamide by bypassing the AR blockade, which occurs due to increased activity of the glucocorticoid receptor (GR) [44]. Recent studies have indicated that heightened activity of the GR has been linked to the emergence of resistance to antiandrogen therapies in different clinical and preclinical models [44][45]. These findings could possibly be explained by the fact that GR expression is downregulated by AR signaling [46]. Additionally, the GR interacts with and mediates a subset of AR targets [44].

2.7. Evolution of t-NEPC from CRPC

During the initial diagnosis of PC, neuroendocrine prostate cancer (NEPC) is rare and represents only 0.5–2% of all PC cases. However, a larger proportion of treatment-induced neuroendocrine prostate cancer (t-NEPC) has been reported. Recent studies have indicated that approximately 17–30% of CRPC patients, following ADT and other treatments, may experience progression to t-NEPC [47][48]. The majority of evidence suggests that the origin of tNEPC is the transdifferentiation of adenocarcinoma cells into NEPC cells in response to different therapies, including ADT [47][48][49]. The transition to the NEPC phenotype following ADT treatments has been demonstrated in various preclinical models, including cell lines, genetically engineered mouse (GEM), and patient-derived xenografts [50][51][52]. In addition to ADT, other therapies, such as radiation and chemotherapy, have been reported to stimulate the NEPC phenotype [53][54][55]. Moreover, genetic and epigenetic alterations and tumor microenvironments (TMEs) containing a variety of cells and factors could support NEPC transition in CRPC patients [56][57][58][59]. Due to the absence of AR and PSA expression (AR-/PSA-), t-NEPC patients have shown limited responses to existing treatment options. To date, several therapeutics have been proposed for patients with t-NEPC, including cisplatin or carboplatin combined with etoposide or docetaxel.

References

  1. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer Incidence and Mortality Worldwide: Sources, Methods and Major Patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386.
  2. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA. Cancer J. Clin. 2023, 73, 17–48.
  3. Scher, H.I.; Morris, M.J.; Stadler, W.M.; Higano, C.S.; Halabi, S.; Smith, M.R.; Basch, E.M.; Fizazi, K.; Ryan, C.J.; Antonarakis, E.S.; et al. The Prostate Cancer Working Group 3 (PCWG3) Consensus for Trials in Castration-Resistant Prostate Cancer (CRPC). J. Clin. Oncol. 2015, 33, 5000.
  4. Castration-Resistant Prostate Cancer: Mechanisms, Targets and Treatment. SpringerLink. Available online: https://link.springer.com/chapter/10.1007/978-3-319-99286-0_7 (accessed on 24 May 2023).
  5. Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New Response Evaluation Criteria in Solid Tumours: Revised RECIST Guideline (Version 1.1). Eur. J. Cancer 2009, 45, 228–247.
  6. Iannantuono, G.M.; Chandran, E.; Floudas, C.S.; Choo-Wosoba, H.; Butera, G.; Roselli, M.; Gulley, J.L.; Karzai, F. Efficacy and Safety of PARP Inhibitors in Metastatic Castration-Resistant Prostate Cancer: A Systematic Review and Meta-Analysis of Clinical Trials. Cancer Treat. Rev. 2023, 120, 102623.
  7. Abida, W.; Armenia, J.; Gopalan, A.; Brennan, R.; Walsh, M.; Barron, D.; Danila, D.; Rathkopf, D.; Morris, M.; Slovin, S.; et al. Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making. JCO Precis. Oncol. 2017, 1, 1–16.
  8. Van Dessel, L.F.; van Riet, J.; Smits, M.; Zhu, Y.; Hamberg, P.; van der Heijden, M.S.; Bergman, A.M.; van Oort, I.M.; de Wit, R.; Voest, E.E.; et al. The Genomic Landscape of Metastatic Castration-Resistant Prostate Cancers Reveals Multiple Distinct Genotypes with Potential Clinical Impact. Nat. Commun. 2019, 10, 5251.
  9. Quigley, D.A.; Dang, H.X.; Zhao, S.G.; Lloyd, P.; Aggarwal, R.; Alumkal, J.J.; Foye, A.; Kothari, V.; Perry, M.D.; Bailey, A.M.; et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018, 175, 889.
  10. Saraon, P.; Jarvi, K.; Diamandis, E.P. Molecular Alterations during Progression of Prostate Cancer to Androgen Independence. Clin. Chem. 2011, 57, 1366–1375.
  11. Powell, S.M.; Christiaens, V.; Voulgaraki, D.; Waxman, J.; Claessens, F.; Bevan, C.L. Mechanisms of Androgen Receptor Signalling via Steroid Receptor Coactivator-1 in Prostate. Endocr. Relat. Cancer 2004, 11, 117–130.
  12. Taplin, M.-E.; Bubley, G.J.; Shuster, T.D.; Frantz, M.E.; Spooner, A.E.; Ogata, G.K.; Keer, H.N.; Balk, S.P. Mutation of the Androgen-Receptor Gene in Metastatic Androgen-Independent Prostate Cancer. N. Engl. J. Med. 1995, 332, 1393–1398.
  13. Newmark, J.R.; Hardy, D.O.; Tonb, D.C.; Carter, B.S.; Epstein, J.I.; Isaacs, W.B.; Brown, T.R.; Barrack, E.R. Androgen Receptor Gene Mutations in Human Prostate Cancer. Proc. Natl. Acad. Sci. USA 1992, 89, 6319–6323.
  14. Wyatt, A.W.; Gleave, M.E. Targeting the Adaptive Molecular Landscape of Castration-Resistant Prostate Cancer. EMBO Mol. Med. 2015, 7, 878–894.
  15. Wilding, G.; Chen, M.; Gelmann, E.P. Aberrant Response in Vitro of Hormone-Responsive Prostate Cancer Cells to Antiandrogens. Prostate 1989, 14, 103–115.
  16. Taplin, M.-E.; Bubley, G.J.; Ko, Y.-J.; Small, E.J.; Upton, M.; Rajeshkumar, B.; Balk, S.P. Selection for Androgen Receptor Mutations in Prostate Cancers Treated with Androgen Antagonist1. Cancer Res. 1999, 59, 2511–2515.
  17. Culig, Z.; Hobisch, A.; Cronauer, M.V.; Radmayr, C.; Trapman, J.; Hittmair, A.; Bartsch, G.; Klocker, H. Androgen Receptor Activation in Prostatic Tumor Cell Lines by Insulin-Like Growth Factor-1,Keratinocyte Growth Factor and Epidermal Growth Factor. Eur. Urol. 2017, 27, 45–47.
  18. Hara, T.; Miyazaki, J.; Araki, H.; Yamaoka, M.; Kanzaki, N.; Kusaka, M.; Miyamoto, M. Novel Mutations of Androgen Receptor: A Possible Mechanism of Bicalutamide Withdrawal Syndrome. Cancer Res. 2003, 63, 149–153.
  19. Watson, P.A.; Arora, V.K.; Sawyers, C.L. Emerging Mechanisms of Resistance to Androgen Receptor Inhibitors in Prostate Cancer. Nat. Rev. Cancer 2015, 15, 701–711.
  20. Tan, J.; Sharief, Y.; Hamil, K.G.; Gregory, C.W.; Zang, D.Y.; Sar, M.; Gumerlock, P.H.; White, R.W.D.; Pretlow, T.G.; Harris, S.E.; et al. Dehydroepiandrosterone Activates Mutant Androgen Receptors Expressed in the Androgen-Dependent Human Prostate Cancer Xenograft CWR22 and LNCaP Cells. Mol. Endocrinol. 1997, 11, 450–459.
  21. Guo, Z.; Yang, X.; Sun, F.; Jiang, R.; Linn, D.E.; Chen, H.; Chen, H.; Kong, X.; Melamed, J.; Tepper, C.G.; 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, 2305–2313.
  22. Zhang, T.; Karsh, L.I.; Nissenblatt, M.J.; Canfield, S.E. Androgen Receptor Splice Variant, AR-V7, as a Biomarker of Resistance to Androgen Axis-Targeted Therapies in Advanced Prostate Cancer. Clin. Genitourin. Cancer 2020, 18, 1–10.
  23. Saraon, P.; Drabovich, A.P.; Jarvi, K.A.; Diamandis, E.P. Mechanisms of Androgen-Independent Prostate Cancer. EJIFCC 2014, 25, 42–54.
  24. Heinlein, C.A.; Chang, C. Androgen Receptor (AR) Coregulators: An Overview. Endocr. Rev. 2002, 23, 175–200.
  25. Gregory, C.W.; He, B.; Johnson, R.T.; Ford, O.H.; Mohler, J.L.; French, F.S.; Wilson, E.M. A Mechanism for Androgen Receptor-Mediated Prostate Cancer Recurrence after Androgen Deprivation Therapy. Cancer Res. 2001, 61, 4315–4319.
  26. Zhou, X.E.; Suino-Powell, K.M.; Li, J.; He, Y.; Mackeigan, J.P.; Melcher, K.; Yong, E.-L.; Xu, H.E. Identification of SRC3/AIB1 as a Preferred Coactivator for Hormone-Activated Androgen Receptor. J. Biol. Chem. 2010, 285, 9161–9171.
  27. Tien, J.C.-Y.; Liu, Z.; Liao, L.; Wang, F.; Xu, Y.; Wu, Y.-L.; Zhou, N.; Ittmann, M.; Xu, J. The Steroid Receptor Coactivator-3 Is Required for the Development of Castration-Resistant Prostate Cancer. Cancer Res. 2013, 73, 3997–4008.
  28. Cheng, S.; Brzostek, S.; Lee, S.R.; Hollenberg, A.N.; Balk, S.P. Inhibition of the Dihydrotestosterone-Activated Androgen Receptor by Nuclear Receptor Corepressor. Mol. Endocrinol. 2002, 16, 1492–1501.
  29. Godoy, A.S.; Sotomayor, P.C.; Villagran, M.; Yacoub, R.; Montecinos, V.P.; McNerney, E.M.; Moser, M.; Foster, B.A.; Onate, S.A. Altered Corepressor SMRT Expression and Recruitment to Target Genes as a Mechanism That Change the Response to Androgens in Prostate Cancer Progression. Biochem. Biophys. Res. Commun. 2012, 423, 564–570.
  30. Liao, G.; Chen, L.-Y.; Zhang, A.; Godavarthy, A.; Xia, F.; Ghosh, J.C.; Li, H.; Chen, J.D. Regulation of Androgen Receptor Activity by the Nuclear Receptor Corepressor SMRT*. J. Biol. Chem. 2003, 278, 5052–5061.
  31. Nacusi, L.P.; Tindall, D.J. Targeting 5α-Reductase for Prostate Cancer Prevention and Treatment. Nat. Rev. Urol. 2011, 8, 378–384.
  32. Dutt, S.S.; Gao, A.C. Molecular Mechanisms of Castration-Resistant Prostate Cancer Progression. Future Oncol. 2009, 5, 1403–1413.
  33. Montgomery, R.B.; Mostaghel, E.A.; Vessella, R.; Hess, D.L.; Kalhorn, T.F.; Higano, C.S.; True, L.D.; Nelson, P.S. Maintenance of Intratumoral Androgens in Metastatic Prostate Cancer: A Mechanism for Castration-Resistant Tumor Growth. Cancer Res. 2008, 68, 4447–4454.
  34. Wu, J.D.; Haugk, K.; Woodke, L.; Nelson, P.; Coleman, I.; Plymate, S.R. Interaction of IGF Signaling and the Androgen Receptor in Prostate Cancer Progression. J. Cell. Biochem. 2006, 99, 392–401.
  35. Malinowska, K.; Neuwirt, H.; Cavarretta, I.T.; Bektic, J.; Steiner, H.; Dietrich, H.; Moser, P.L.; Fuchs, D.; Hobisch, A.; Culig, Z. Interleukin-6 Stimulation of Growth of Prostate Cancer in Vitro and in Vivo through Activation of the Androgen Receptor. Endocr. Relat. Cancer 2009, 16, 155–169.
  36. Sarker, D.; Reid, A.H.M.; Yap, T.A.; de Bono, J.S. Targeting the PI3K/AKT Pathway for the Treatment of Prostate Cancer. Clin. Cancer Res. 2009, 15, 4799–4805.
  37. Wen, Y.; Hu, M.C.; Makino, K.; Spohn, B.; Bartholomeusz, G.; Yan, D.H.; Hung, M.C. HER-2/Neu Promotes Androgen-Independent Survival and Growth of Prostate Cancer Cells through the Akt Pathway. Cancer Res. 2000, 60, 6841–6845.
  38. Graff, J.R.; Konicek, B.W.; McNulty, A.M.; Wang, Z.; Houck, K.; Allen, S.; Paul, J.D.; Hbaiu, A.; Goode, R.G.; Sandusky, G.E.; et al. Increased AKT Activity Contributes to Prostate Cancer Progression by Dramatically Accelerating Prostate Tumor Growth and Diminishing p27Kip1 Expression. J. Biol. Chem. 2000, 275, 24500–24505.
  39. Jamaspishvili, T.; Berman, D.M.; Ross, A.E.; Scher, H.I.; De Marzo, A.M.; Squire, J.A.; Lotan, T.L. Clinical Implications of PTEN Loss in Prostate Cancer. Nat. Rev. Urol. 2018, 15, 222–234.
  40. Andrieu, C.; Taieb, D.; Baylot, V.; Ettinger, S.; Soubeyran, P.; De-Thonel, A.; Nelson, C.; Garrido, C.; So, A.; Fazli, L.; et al. Heat Shock Protein 27 Confers Resistance to Androgen Ablation and Chemotherapy in Prostate Cancer Cells through eIF4E. Oncogene 2010, 29, 1883–1896.
  41. Baylot, V.; Katsogiannou, M.; Andrieu, C.; Taieb, D.; Acunzo, J.; Giusiano, S.; Fazli, L.; Gleave, M.; Garrido, C.; Rocchi, P. Targeting TCTP as a New Therapeutic Strategy in Castration-Resistant Prostate Cancer. Mol. Ther. J. Am. Soc. Gene Ther. 2012, 20, 2244–2256.
  42. Cherif, C.; Nguyen, D.T.; Paris, C.; Le, T.K.; Sefiane, T.; Carbuccia, N.; Finetti, P.; Chaffanet, M.; Kaoutari, A.E.; Vernerey, J.; et al. Menin Inhibition Suppresses Castration-Resistant Prostate Cancer and Enhances Chemosensitivity. Oncogene 2022, 41, 125–137.
  43. Le, T.K.; Cherif, C.; Omabe, K.; Paris, C.; Lannes, F.; Audebert, S.; Baudelet, E.; Hamimed, M.; Barbolosi, D.; Finetti, P.; et al. DDX5 mRNA-Targeting Antisense Oligonucleotide as a New Promising Therapeutic in Combating Castration-Resistant Prostate Cancer. Mol. Ther. 2022, 31, 471–486.
  44. Arora, V.K.; Schenkein, E.; Murali, R.; Subudhi, S.K.; Wongvipat, J.; Balbas, M.D.; Shah, N.; Cai, L.; Efstathiou, E.; Logothetis, C.; et al. Glucocorticoid Receptor Confers Resistance to Antiandrogens by Bypassing Androgen Receptor Blockade. Cell 2013, 155, 1309–1322.
  45. Puhr, M.; Hoefer, J.; Eigentler, A.; Ploner, C.; Handle, F.; Schaefer, G.; Kroon, J.; Leo, A.; Heidegger, I.; Eder, I.; et al. The Glucocorticoid Receptor Is a Key Player for Prostate Cancer Cell Survival and a Target for Improved Antiandrogen Therapy. Clin. Cancer Res. 2018, 24, 927–938.
  46. Xie, N.; Cheng, H.; Lin, D.; Liu, L.; Yang, O.; Jia, L.; Fazli, L.; Gleave, M.E.; Wang, Y.; Rennie, P.; et al. The Expression of Glucocorticoid Receptor Is Negatively Regulated by Active Androgen Receptor Signaling in Prostate Tumors. Int. J. Cancer 2015, 136, E27–E38.
  47. Patel, G.K.; Chugh, N.; Tripathi, M. Neuroendocrine Differentiation of Prostate Cancer—An Intriguing Example of Tumor Evolution at Play. Cancers 2019, 11, 1405.
  48. Beltran, H.; Tomlins, S.; Aparicio, A.; Arora, V.; Rickman, D.; Ayala, G.; Huang, J.; True, L.; Gleave, M.E.; Soule, H.; et al. Aggressive Variants of Castration Resistant Prostate Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 2846–2850.
  49. Beltran, H.; Rickman, D.S.; Park, K.; Chae, S.S.; Sboner, A.; MacDonald, T.Y.; Wang, Y.; Sheikh, K.L.; Terry, S.; Tagawa, S.T.; et al. Molecular Characterization of Neuroendocrine Prostate Cancer and Identification of New Drug Targets. Cancer Discov. 2011, 1, 487–495.
  50. Lin, D.; Wyatt, A.W.; Xue, H.; Wang, Y.; Dong, X.; Haegert, A.; Wu, R.; Brahmbhatt, S.; Mo, F.; Jong, L.; et al. High Fidelity Patient-Derived Xenografts for Accelerating Prostate Cancer Discovery and Drug Development. Cancer Res. 2014, 74, 1272–1283.
  51. Ku, S.Y.; Rosario, S.; Wang, Y.; Mu, P.; Seshadri, M.; Goodrich, Z.W.; Goodrich, M.M.; Labbé, D.P.; Gomez, E.C.; Wang, J.; et al. Rb1 and Trp53 Cooperate to Suppress Prostate Cancer Lineage Plasticity, Metastasis, and Antiandrogen Resistance. Science 2017, 355, 78–83.
  52. Zou, M.; Toivanen, R.; Mitrofanova, A.; Floch, N.; Hayati, S.; Sun, Y.; Le Magnen, C.; Chester, D.; Mostaghel, E.A.; Califano, A.; et al. Transdifferentiation as a Mechanism of Treatment Resistance in a Mouse Model of Castration-Resistant Prostate Cancer. Cancer Discov. 2017, 7, 736–749.
  53. Deng, X.; Liu, H.; Huang, J.; Cheng, L.; Keller, E.T.; Parsons, S.J.; Hu, C.-D. Ionizing Radiation Induces Prostate Cancer Neuroendocrine Differentiation through Interplay of CREB and ATF2: Implications for Disease Progression. Cancer Res. 2008, 68, 9663–9670.
  54. Bonkhoff, H. Factors Implicated in Radiation Therapy Failure and Radiosensitization of Prostate Cancer. Prostate Cancer 2012, 2012, 593241.
  55. Berruti, A.; Dogliotti, L.; Mosca, A.; Bellina, M.; Mari, M.; Torta, M.; Tarabuzzi, R.; Bollito, E.; Fontana, D.; Angeli, A. Circulating Neuroendocrine Markers in Patients with Prostate Carcinoma. Cancer 2000, 88, 2590–2597.
  56. Davies, A.; Zoubeidi, A.; Selth, L.A. The Epigenetic and Transcriptional Landscape of Neuroendocrine Prostate Cancer. Endocr. Relat. Cancer 2020, 27, R35–R50.
  57. Baca, S.C.; Takeda, D.Y.; Seo, J.-H.; Hwang, J.; Ku, S.Y.; Arafeh, R.; Arnoff, T.; Agarwal, S.; Bell, C.; O’Connor, E.; et al. Reprogramming of the FOXA1 Cistrome in Treatment-Emergent Neuroendocrine Prostate Cancer. Nat. Commun. 2021, 12, 1979.
  58. Dang, Q.; Li, L.; Xie, H.; He, D.; Chen, J.; Song, W.; Chang, L.S.; Chang, H.-C.; Yeh, S.; Chang, C. Anti-Androgen Enzalutamide Enhances Prostate Cancer Neuroendocrine (NE) Differentiation via Altering the Infiltrated Mast Cells→Androgen Receptor (AR)→miRNA32 Signals. Mol. Oncol. 2015, 9, 1241–1251.
  59. Mishra, R.; Haldar, S.; Placencio, V.; Madhav, A.; Rohena-Rivera, K.; Agarwal, P.; Duong, F.; Angara, B.; Tripathi, M.; Liu, Z.; et al. Stromal Epigenetic Alterations Drive Metabolic and Neuroendocrine Prostate Cancer Reprogramming. J. Clin. Investig. 2018, 128, 4472–4484.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 216
Revisions: 2 times (View History)
Update Date: 31 Oct 2023
1000/1000
ScholarVision Creations