Next Article in Journal
Dose Reduction to Motor Structures in Adjuvant Fractionated Stereotactic Radiotherapy of Brain Metastases: nTMS-Derived DTI-Based Motor Fiber Tracking in Treatment Planning
Next Article in Special Issue
Interactions of SNPs in Folate Metabolism Related Genes on Prostate Cancer Aggressiveness in European Americans and African Americans
Previous Article in Journal
Differentiating Ductal Adenocarcinoma of the Pancreas from Benign Conditions Using Routine Health Records: A Prospective Case-Control Study
Previous Article in Special Issue
Liver Microenvironment Response to Prostate Cancer Metastasis and Hormonal Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Prostate Cancer: Pathophysiology, Pathology and Therapy

by
Vasiliki Tzelepi
1,2
1
Department of Pathology, University of Patras, 26504 Patras, Greece
2
Department of Histopathology and Cytology, University Hospital of Patras, 26504 Patras, Greece
Cancers 2023, 15(1), 281; https://doi.org/10.3390/cancers15010281
Submission received: 26 December 2022 / Accepted: 29 December 2022 / Published: 31 December 2022
(This article belongs to the Special Issue Prostate Cancer: Pathophysiology, Pathology and Therapy)
Prostate cancer (PCa) is a major health care challenge in the developed world, being the most common type of cancer in men in the USA [1] and most European countries [2] and the second most common worldwide [3]. PCa shows remarkable heterogeneity in its clinical course. Some patients have indolent cancer that will never progress, whereas others have a remarkably aggressive disease with rapid progression to metastases and resistance to therapy [4,5], making PCa the fifth most common cause of cancer-related death worldwide [3]. In between are patients with an initially localized disease that will progress to a metastatic, incurable disease after a variable time period [4]. In this Special Issue, recent advances in precision medicine approaches for PCa in regard to clinical, pathologic, molecular and therapeutic parameters are presented.
In a review paper, Cimadarone et al. [6] summarize the novel prognostic and predictive tissue-based biomarkers for PCa developed in 2021. They discuss the contemporary PCa grading, the advantages offered by computational pathology and artificial intelligence, the morphologic and immunohistochemical features of aggressive variants of PCa, and the molecular markers that can be used in the clinic to predict an aggressive disease or a response to specific therapies.
The clinical heterogeneity of prostate cancer is a reflection of its molecular heterogeneity and the emergence of lineage plasticity, which is the ability of neoplastic cells to switch between distinct lineages and phenotypic cell states by adapting to their environment [7]. The epithelial-to-mesenchymal transition is an example of lineage plasticity and represents an important mechanism of tumor progression and therapy resistance. In a review paper in this issue, Papanikolaou et al. [8] discuss the molecular pathology of the epithelial-to-mesenchymal transition in PCa, the pathways involved in its emergence, and its effect on PCa aggressiveness and therapy resistance, as well as potential opportunities for therapeutic targeting.
Currently, prostate-specific antigen (PSA) levels, clinical stages and the biopsy Gleason Score are the main parameters used to stratify patients into risk categories at initial presentation, albeit with modest specificity and sensitivity, and are used to decide on the appropriate therapy [9]. The Gleason Score, originally described by Donald Gleason in 1974 [10], has been continuously refined over the years [11,12] in an effort to enhance its prognostic value. Reproducibility in PCa grading by pathologists is of paramount importance for accurate risk classification and therapy selection. However, in a nationwide survey among 41 laboratories and >38,000 needle biopsy reports, which was completed by Flach et al. and presented in this Special Issue [13], a significant inter- and intra-laboratory variation in the daily practice of PCa grading was noted, irrespective of the PCa case volume diagnosed by the pathologist and corrected by PSA levels atdiagnosis, year of diagnosis, age, number of biopsies taken, and number of positive biopsies. These findings highlight the need for better standardization of PCa grading, for example, through training and the use of artificial intelligence.
Apart from the Gleason Score, specific morphologic patterns of PCa, i.e., cribriform and intraductal carcinoma (IDC), as well as tertiary patterns, seem to have additional prognostic significance. However, confusing guidelines exist in regard to how this information is conveyed to clinicians [14,15,16]. In a paper in this Special Issue [17], Tzelepi et al. show that, depending on the grading criteria used (handling of IDC and tertiary patterns), the final grade would be different in a number of cases, with a three-point difference in a minority of them. This may be a source of confusion amongst both pathologists and urologists. They also provide support for IDC incorporation into the final grade, even though they recognize the need for additional studies with survival as the end point.
Radiotherapy (RT) is a therapeutic option for localized prostate cancer, with fractionation and dose depending on the clinical scenario [9]. Previous studies have shown that external beam radiotherapy with moderate hypofractionation is effective in low-risk, intermediate-risk and high-risk localized prostate cancer. In their study, di Muzio et al. [18] present their experience with pelvic lymph nodal irradiation through a moderately hypofractionated, high-dose, simultaneously integrated boost to the prostate and seminal vesicles and long-term androgen-deprivation therapy in unfavorable intermediate-, high- and very high-risk localized prostate cancer patients. They confirmed the good clinical outcomes and acceptable toxicity of this approach, in terms of ten-year biochemical-relapse-free survival and disease-free survival, with a longer follow-up (median of 107.6 months (IQR: 78.35; 136.10)) than that used in previous studies, and they identified a Gleason Score ≥8 as an independent predictor of biochemical relapse.
With conventional RT approaches, a prescribed dose is delivered to the prostate. With biologically targeted approaches, the specific characteristics of the tumor are considered in order to maximize tumor control while limiting the toxicity in adjacent normal tissues. The inability to spatially map tumor characteristics has limited the wide application of this approach. In their study, Hen et al. [19] compare in silico radiotherapy with a tumor and hypoxic sub-volume boost by using mpMRI-derived cell density and hypoxia maps, to conventional approaches. Their results showed that biological optimization improved rectal and bladder sparing, supporting further investigations of biologically targeted approaches by taking advantage of the knowledge on the spatial distribution of tumor heterogeneity.
Androgen deprivation therapy (ADT) using LHRH agonists/antagonists that cause pharmacologic castration, with or without androgen receptor (AR) antagonists, represents the main course of therapy for patients that present with metastatic disease and for patients that develop biochemical recurrence after their initial therapy [20]. ADT is not without side effects, with cardiovascular events, including myocardial infarction and cardiovascular-related mortality, being the most important [21]. Dementia has also been associated with ADT, albeit the reports are controversial. In this issue, Liu et al. [22] describe a population-based cohort study of 129,126 men with PCa, with data obtained from the National Health Insurance Database of Taiwan and The Health Improvement Network Database of the United Kingdom, and show that there is no difference in the incidence rates of dementia between patients receiving ADT and those that are ADT naïve, nor is there a difference in any cumulative dose effect between ADT and dementia.
The majority of patients will respond well to androgen deprivation treatment, but the tumor will, eventually, recur, leading to castrate-resistant prostate cancer (CRPC) [23]. Second generation antiandrogens and various other therapies are available for these patients, which result in a significant prolongation of patients’ survival [20]. Predictive markers as to the best sequential order of these therapies are largely lacking. In their study, Hsieh et al. [24] provide evidence that metastatic CRPC (mCRPC) patients receiving enzalutamide with a high tumor burden, defined as either appendicular bony or visceral metastasis, showed fewer good PSA response rates, fewer rates of partial radiological response and stable disease, and a shorter progression-free survival duration compared to low-tumor-burden patients. Although further studies are needed to identify a model that will inform therapeutic decisions, this study, as well as others [25,26], highlight tumor burden as a clinical factor to be considered when deciding upon an optimal therapeutic strategy.
Similarly, in an effort to identify markers of disease aggressiveness to guide therapeutic decisions, Delanoy et al. [27] present a post hoc analysis of PROSELICA, a large phase III randomized study (NCT01308580) that evaluated two doses of cabazitaxel (the standard dose of 25 mg/m2 every 3 weeks and a lower dose of 20 mg/m2 every 3 weeks) in mCRPC patients previously treated with docetaxel [28]. The authors assessed the prognostic value of the type of disease progression at cabazitaxel initiation in a post-docetaxel setting and confirmed that pain progression at cabazitaxel initiation was associated with clinical and biological features of aggressive disease and worse outcomes, andwas a better predictor of disease aggressiveness than PSA progression.
CRPC emergence involves the development of AR mutations, with the majority being located at the ligand-binding domain of the molecule [29], and the expression of AR splice variants, most being constitutively active despite lacking the ligand-binding domain [30]. Both mechanisms render most first- and second-generation AR-targeting approaches (that directly or indirectly target the ligand-binding domain) ineffective. Darolutamide, a second-generation hormone therapy, is an AR antagonist that is structurally distinct compared to other AR antagonists, including enzalutabite and apalutamide, and has been proposed to inhibit AR mutant proteins that are resistant to other antagonists. In their paper, Lallous et al. [31] examine the effect of darolutamide, as well as the commonly used antiandrogens bicalutamide and enzalutamide and the major endogenous steroids DHT, estradiol, progesterone and hydrocortisone, on 44 AR mutants (including data presented in their previous work [32]) and identified only one mutant that was not inhibited by darolutamide, highlighting this compound as a potential therapeutic option for CRPC patients with AR mutations.
Since the ligand-binding domain of AR is the most affected under the pressure of therapy and since most of the AR transcriptional activity is mediated via its N-terminal domain (NTD), targeting the NTD domain represents a promising therapeutic strategy for CRPC patients. In their paper, Ban et al. [33] characterize novel AR NTD-targeting molecules. Based on a molecule identified in their previous work, VPC-2055, they synthesized and tested 110 novel compounds. Among them, eight were found to be active in physiologic concentrations and one, VPC-220010, was three and five times more potent than VPC-2055 and EPI-001, a known NTD inhibitor, respectively, and was further extensively characterized in regard to the inhibition of AR activation and PCa cell-line viability, providing preclinical evidence for its potential clinical utility.
When progressing, PCa largely metastasizes to the bones, and cancer cell osteomimicry has been implicated in this process. MINDIN, an extracellular matrix protein, has been shown to induce osteomimicry and prostate cancer progression [34]; however, its mechanisms of action have not been elucidated. In their paper, Alvarez-Carrion et al. [35] provide evidence that MINDIN downregulates the expression of NHERF-1, a scaffold protein that interacts with various proteins including receptors, transporters and cytoplasmic signaling proteins, thereby enhancing a variety of cellular processes including cell proliferation and migration.
Another mechanism by which AR is activated in the CRPC setting is via crosstalk with growth factor receptor pathways, including the PI3K/AKT pathway, through PTEN deletion [36]. Apalutamide is a second-generation antiandrogen that binds to the ligand-binding domain of AR and inhibits its nuclear translocation. It their paper, De Velasco et al. [37] characterize the antitumor activity of apalutamide in genetically engineered, Pten-deficient PCa mouse models. They showed that apalutamide demonstrated antitumor activity in models of castration-naïve PCa (Pten-deficient) and CRPC (Pten/Trp53 double knockout). Upregulated aberrant AR and a phosphorylated S6 and proline-rich Akt substrate of 40 kDa (PRAS40) were seen in the surviving cancer cells in CRPC. Strong synergy was observed between the pan-AKT inhibitor GSK690693 and apalutamide in vivo in castration-naïve PCa, but not in CRPC. Further studies of apalutamide combinations with AKT and the inhibition of other pathways are needed.
AR-independent mechanisms have also been implicated in CRPC development. In addition, a transformation from adenocarcinoma, the prototype of PCa, to neuroendocrine carcinoma (NECa), an aggressive tumor type characterized by lytic bone metastasis, splanchnic metastasis and low PSA for disease burden, is seen in some, but not all, of the CRPC cases under the pressure of therapy [38]. Elucidating the mechanisms of AR-independent progression is of paramount importance for the discovery of therapeutic targets for this devastating type of the disease. In this Special Issue, Xu et al. [39] show that the expression of ELOVL5, the key enzyme of long-chain polyunsaturated fatty acid elongation, was elevated in NECa cell lines, and its knock down diminished their neuroendocrine phenotype and enzalutamide resistance, whereas its overexpression enhanced enzalutamide resistance in PCa cell lines in vitro and in vivo. These findings highlight ELOVL5 as a potential therapeutic target for PCa patients with the NECa phenotype.
This Special Issue includes some important developments in PCa research for the implementation of personalized medicine in the treatment of PCa patients and for using clinical, morphologic and molecular markers to inform therapeutic decisions.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2022. CA A Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Dyba, T.; Randi, G.; Bray, F.; Martos, C.; Giusti, F.; Nicholson, N.; Gavin, A.; Flego, M.; Neamtiu, L.; Dimitrova, N.; et al. The European Cancer Burden in 2020: Incidence and Mortality Estimates for 40 Countries and 25 Major Cancers. Eur. J. Cancer 2021, 157, 308–347. [Google Scholar] [CrossRef] [PubMed]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  4. Johansson, J.; Andrén, O.; Andersson, S.; Dickman, P.W.; Holmberg, L.; Magnuson, A.; Adami, H.-O. Natural History of Early, Localized Prostate Cancer. JAMA 2004, 291, 2713–2719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Aparicio, A.; Tzelepi, V. Neuroendocrine (Small-Cell) Carcinomas: Why They Teach Us Essential Lessons about Prostate Cancer. Oncology 2014, 28, 831–838. [Google Scholar]
  6. Cimadamore, A.; Mazzucchelli, R.; Lopez-Beltran, A.; Massari, F.; Santoni, M.; Scarpelli, M.; Cheng, L.; Montironi, R. Prostate Cancer in 2021: Novelties in Prognostic and Therapeutic Biomarker Evaluation. Cancers 2021, 13, 3471. [Google Scholar] [CrossRef]
  7. Thankamony, A.P.; Subbalakshmi, A.R.; Jolly, M.K.; Nair, R. Lineage Plasticity in Cancer: The Tale of a Skin-Walker. Cancers 2021, 13, 3602. [Google Scholar] [CrossRef]
  8. 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 2021, 13, 2795. [Google Scholar] [CrossRef]
  9. Mottet, N.; van den Bergh, R.C.N.; Briers, E.; Van den Broeck, T.; Cumberbatch, M.G.; De Santis, M.; Fanti, S.; Fossati, N.; Gandaglia, G.; Gillessen, S.; et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer—2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent. Eur. Urol. 2021, 79, 243–262. [Google Scholar] [CrossRef]
  10. Gleason, D.F.; Mellinger, G.T. Prediction of Prognosis for Prostatic Adenocarcinoma by Combined Histological Grading and Clinical Staging. J. Urol. 1974, 111, 58–64. [Google Scholar] [CrossRef]
  11. Epstein, J.I.; Allsbrook, W.C.; Amin, M.B.; Egevad, L.L. The 2005 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma. Am. J. Surg. Pathol. 2005, 29, 1228–1242. [Google Scholar] [CrossRef] [PubMed]
  12. Epstein, J.I.; Egevad, L.; Amin, M.B.; Delahunt, B.; Srigley, J.R.; Humphrey, P.A.; The Grading Committee. The 2014 International Society of Urological Pathology (ISUP) Consensus Conference on Gleason Grading of Prostatic Carcinoma: Definition of Grading Patterns and Proposal for a New Grading System. Am. J. Surg. Pathol. 2016, 40, 244–252. [Google Scholar] [CrossRef] [PubMed]
  13. Flach, R.N.; Willemse, P.-P.M.; Suelmann, B.B.M.; Deckers, I.A.G.; Jonges, T.N.; van Dooijeweert, C.; van Diest, P.J.; Meijer, R.P. Significant Inter- and Intralaboratory Variation in Gleason Grading of Prostate Cancer: A Nationwide Study of 35,258 Patients in The Netherlands. Cancers 2021, 13, 5378. [Google Scholar] [CrossRef] [PubMed]
  14. van Leenders, G.J.L.H.; van der Kwast, T.H.; Grignon, D.J.; Evans, A.J.; Kristiansen, G.; Kweldam, C.F.; Litjens, G.; McKenney, J.K.; Melamed, J.; Mottet, N.; et al. The 2019 International Society of Urological Pathology (ISUP) Consensus Conference on Grading of Prostatic Carcinoma. Am. J. Surg. Pathol. 2020, 44, e87–e99. [Google Scholar] [CrossRef] [PubMed]
  15. Epstein, J.I.; Amin, M.B.; Fine, S.W.; Algaba, F.; Aron, M.; Baydar, D.E.; Beltran, A.L.; Brimo, F.; Cheville, J.C.; Colecchia, M.; et al. The 2019 Genitourinary Pathology Society (GUPS) White Paper on Contemporary Grading of Prostate Cancer. Arch. Pathol. Lab. Med. 2021, 145, 461–493. [Google Scholar] [CrossRef] [PubMed]
  16. Smith, S.C.; Gandhi, J.S.; Moch, H.; Aron, M.; Compérat, E.; Paner, G.P.; McKenney, J.K.; Amin, M.B. Similarities and Differences in the 2019 ISUP and GUPS Recommendations on Prostate Cancer Grading: A Guide for Practicing Pathologists. Adv. Anat. Pathol. 2021, 28, 1–7. [Google Scholar] [CrossRef]
  17. Tzelepi, V.; Grypari, I.M.; Logotheti, S.; Kontogiannis, S.; Kallidonis, P.; Melachrinou, M.; Zolota, V. Contemporary Grading of Prostate Cancer: The Impact of Grading Criteria and the Significance of the Amount of Intraductal Carcinoma. Cancers 2021, 13, 5454. [Google Scholar] [CrossRef]
  18. Di Muzio, N.G.; Deantoni, C.L.; Brombin, C.; Fiorino, C.; Cozzarini, C.; Zerbetto, F.; Mangili, P.; Tummineri, R.; Dell’Oca, I.; Broggi, S.; et al. Ten Year Results of Extensive Nodal Radiotherapy and Moderately Hypofractionated Simultaneous Integrated Boost in Unfavorable Intermediate-, High-, and Very High-Risk Prostate Cancer. Cancers 2021, 13, 4970. [Google Scholar] [CrossRef]
  19. Her, E.J.; Haworth, A.; Sun, Y.; Williams, S.; Reynolds, H.M.; Kennedy, A.; Ebert, M.A. Biologically Targeted Radiation Therapy: Incorporating Patient-Specific Hypoxia Data Derived from Quantitative Magnetic Resonance Imaging. Cancers 2021, 13, 4897. [Google Scholar] [CrossRef]
  20. Cornford, P.; van den Bergh, R.C.N.; Briers, E.; Van den Broeck, T.; Cumberbatch, M.G.; De Santis, M.; Fanti, S.; Fossati, N.; Gandaglia, G.; Gillessen, S.; et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer. Part II—2020 Update: Treatment of Relapsing and Metastatic Prostate Cancer. Eur. Urol. 2021, 79, 263–282. [Google Scholar] [CrossRef]
  21. Levine, G.N.; D’Amico, A.V.; Berger, P.; Clark, P.E.; Eckel, R.H.; Keating, N.L.; Milani, R.V.; Sagalowsky, A.I.; Smith, M.R.; Zakai, N. Androgen-Deprivation Therapy in Prostate Cancer and Cardiovascular Risk. Circulation 2010, 121, 833–840. [Google Scholar] [CrossRef] [PubMed]
  22. Liu, J.-M.; Shen, C.-Y.; Lau, W.C.Y.; Shao, S.-C.; Man, K.K.C.; Hsu, R.-J.; Wu, C.-T.; Lai, E.C.-C. Association between Androgen Deprivation Therapy and Risk of Dementia in Men with Prostate Cancer. Cancers 2021, 13, 3861. [Google Scholar] [CrossRef] [PubMed]
  23. Patrikidou, A.; Loriot, Y.; Eymard, J.-C.; Albiges, L.; Massard, C.; Ileana, E.; Di Palma, M.; Escudier, B.; Fizazi, K. Who Dies from Prostate Cancer? Prostate Cancer Prostatic Dis. 2014, 17, 348–352. [Google Scholar] [CrossRef] [PubMed]
  24. Hsieh, Y.-T.; Chiang, B.-J.; Wu, C.-C.; Liao, C.-H.; Lin, C.-D.; Chen, C.-H. High Tumor Burden Predicts Poor Response to Enzalutamide in Metastatic Castration-Resistant Prostate Cancer Patients. Cancers 2021, 13, 3966. [Google Scholar] [CrossRef] [PubMed]
  25. Sweeney, C.J.; Chen, Y.-H.; Carducci, M.; Liu, G.; Jarrard, D.F.; Eisenberger, M.; Wong, Y.-N.; Hahn, N.; Kohli, M.; Cooney, M.M.; et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N. Engl. J. Med. 2015, 373, 737–746. [Google Scholar] [CrossRef] [PubMed]
  26. Gravis, G.; Boher, J.-M.; Chen, Y.-H.; Liu, G.; Fizazi, K.; Carducci, M.A.; Oudard, S.; Joly, F.; Jarrard, D.M.; Soulie, M.; et al. Burden of Metastatic Castrate Naive Prostate Cancer Patients, to Identify Men More Likely to Benefit from Early Docetaxel: Further Analyses of CHAARTED and GETUG-AFU15 Studies. Eur. Urol. 2018, 73, 847–855. [Google Scholar] [CrossRef] [PubMed]
  27. Delanoy, N.; Robbrecht, D.; Eisenberger, M.; Sartor, O.; de Wit, R.; Mercier, F.; Geffriaud-Ricouard, C.; de Bono, J.; Oudard, S. Pain Progression at Initiation of Cabazitaxel in Metastatic Castration-Resistant Prostate Cancer (MCRPC): A Post Hoc Analysis of the PROSELICA Study. Cancers 2021, 13, 1284. [Google Scholar] [CrossRef] [PubMed]
  28. Eisenberger, M.; Hardy-Bessard, A.-C.; Kim, C.S.; Géczi, L.; Ford, D.; Mourey, L.; Carles, J.; Parente, P.; Font, A.; Kacso, G.; et al. Phase III Study Comparing a Reduced Dose of Cabazitaxel (20 Mg/m2) and the Currently Approved Dose (25 Mg/m2) in Postdocetaxel Patients With Metastatic Castration-Resistant Prostate Cancer—PROSELICA. J. Clin. Oncol. 2017, 35, 3198–3206. [Google Scholar] [CrossRef]
  29. Shiota, M.; Akamatsu, S.; Tsukahara, S.; Nagakawa, S.; Matsumoto, T.; Eto, M. Androgen Receptor Mutations for Precision Medicine in Prostate Cancer. Endocr.—Relat. Cancer 2022, 29, R143–R155. [Google Scholar] [CrossRef]
  30. Wadosky, K.M.; Koochekpour, S. Androgen Receptor Splice Variants and Prostate Cancer: From Bench to Bedside. Oncotarget 2017, 8, 18550–18576. [Google Scholar] [CrossRef] [Green Version]
  31. Lallous, N.; Snow, O.; Sanchez, C.; Parra Nuñez, A.K.; Sun, B.; Hussain, A.; Lee, J.; Morin, H.; Leblanc, E.; Gleave, M.E.; et al. Evaluation of Darolutamide (ODM201) Efficiency on Androgen Receptor Mutants Reported to Date in Prostate Cancer Patients. Cancers 2021, 13, 2939. [Google Scholar] [CrossRef] [PubMed]
  32. Borgmann, H.; Lallous, N.; Ozistanbullu, D.; Beraldi, E.; Paul, N.; Dalal, K.; Fazli, L.; Haferkamp, A.; Lejeune, P.; Cherkasov, A.; et al. Moving Towards Precision Urologic Oncology: Targeting Enzalutamide-Resistant Prostate Cancer and Mutated Forms of the Androgen Receptor Using the Novel Inhibitor Darolutamide (ODM-201). Eur. Urol. 2018, 73, 4–8. [Google Scholar] [CrossRef] [PubMed]
  33. Ban, F.; Leblanc, E.; Cavga, A.D.; Huang, C.-C.F.; Flory, M.R.; Zhang, F.; Chang, M.E.K.; Morin, H.; Lallous, N.; Singh, K.; et al. Development of an Androgen Receptor Inhibitor Targeting the N-Terminal Domain of Androgen Receptor for Treatment of Castration Resistant Prostate Cancer. Cancers 2021, 13, 3488. [Google Scholar] [CrossRef] [PubMed]
  34. Ardura, J.A.; Gutiérrez-Rojas, I.; Álvarez-Carrión, L.; Rodríguez-Ramos, M.R.; Pozuelo, J.M.; Alonso, V. The Secreted Matrix Protein Mindin Increases Prostate Tumor Progression and Tumor-Bone Crosstalk via ERK 1/2 Regulation. Carcinogenesis 2019, 40, 828–839. [Google Scholar] [CrossRef]
  35. Álvarez-Carrión, L.; Gutiérrez-Rojas, I.; Rodríguez-Ramos, M.R.; Ardura, J.A.; Alonso, V. MINDIN Exerts Protumorigenic Actions on Primary Prostate Tumors via Downregulation of the Scaffold Protein NHERF-1. Cancers 2021, 13, 436. [Google Scholar] [CrossRef]
  36. Sircar, K.; Yoshimoto, M.; Monzon, F.A.; Koumakpayi, I.H.; Katz, R.L.; Khanna, A.; Alvarez, K.; Chen, G.; Darnel, A.D.; Aprikian, A.G.; et al. PTEN Genomic Deletion Is Associated with P-Akt and AR Signalling in Poorer Outcome, Hormone Refractory Prostate Cancer. J. Pathol. 2009, 218, 505–513. [Google Scholar] [CrossRef]
  37. De Velasco, M.A.; Kura, Y.; Ando, N.; Sako, N.; Banno, E.; Fujita, K.; Nozawa, M.; Yoshimura, K.; Sakai, K.; Yoshikawa, K.; et al. Context-Specific Efficacy of Apalutamide Therapy in Preclinical Models of Pten-Deficient Prostate Cancer. Cancers 2021, 13, 3975. [Google Scholar] [CrossRef]
  38. Aparicio, A.; Logothetis, C.J.; Maity, S.N. Understanding the Lethal Variant of Prostate Cancer: Power of Examining Extremes. Cancer Discov. 2011, 1, 466–468. [Google Scholar] [CrossRef] [Green Version]
  39. Xu, H.; Li, S.; Sun, Y.; Xu, L.; Hong, X.; Wang, Z.; Hu, H. ELOVL5-Mediated Long Chain Fatty Acid Elongation Contributes to Enzalutamide Resistance of Prostate Cancer. Cancers 2021, 13, 3957. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tzelepi, V. Prostate Cancer: Pathophysiology, Pathology and Therapy. Cancers 2023, 15, 281. https://doi.org/10.3390/cancers15010281

AMA Style

Tzelepi V. Prostate Cancer: Pathophysiology, Pathology and Therapy. Cancers. 2023; 15(1):281. https://doi.org/10.3390/cancers15010281

Chicago/Turabian Style

Tzelepi, Vasiliki. 2023. "Prostate Cancer: Pathophysiology, Pathology and Therapy" Cancers 15, no. 1: 281. https://doi.org/10.3390/cancers15010281

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop