Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets
Abstract
:1. Introduction
1.1. Pathways Involved in the Angiogenesis of Prostate Cancer
1.1.1. Vascular Endothelial Growth Factors (VEGFs)
1.1.2. Fibroblast Growth Factors (FGFs)
1.1.3. Matrix Metalloproteinases (MMPs)
1.1.4. Transforming Growth Factor β (TGFβ)
1.1.5. Pathways of Hypoxia-Inducible Factors (HIF)
1.1.6. Cyclooxygenases (COXs)
1.1.7. Interleukins (ILs)
1.1.8. microRNAs (miRNAs)
2. Chemotherapeutic Agents with Antiangiogenic Activity
3. Anti-Angiogenic Agents in Prostate Cancer
3.1. VEGF-Directed Agents
3.1.1. Bevacizumab
3.1.2. Aflibercept
3.2. VEGFR Tyrosine Kinase Inhibitors
3.2.1. Sorafenib
3.2.2. Sunitinib
3.2.3. Cediranib
3.2.4. Vandetanib
3.2.5. Cabozantinib
3.3. PDGF-Targeted Therapy
3.4. Antiangiogenic/Immunomodulatory Drugs
3.4.1. Thalidomide
3.4.2. Lenalidomide
3.4.3. Miscellaneous Angiogenesis Inhibitors
- Tasquinimod
- ii.
- Itraconazole
- iii.
- Trebanabib
4. Mechanisms of Resistance
5. Conclusions and Future Directions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Compérat, E.; Wasinger, G.; Oszwald, A.; Kain, R.; Cancel-Tassin, G.; Cussenot, O. The Genetic Complexity of Prostate Cancer. Genes 2020, 11, 1396. [Google Scholar] [CrossRef]
- Aalinkeel, R.; Nair, M.P.; Sufrin, G.; Mahajan, S.D.; Chadha, K.C.; Chawda, R.P.; Schwartz, S.A. Gene Expression of Angiogenic Factors Correlates with Metastatic Potential of Prostate Cancer Cells. Cancer Res. 2004, 64, 5311–5321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aragon-Ching, J.B.; Madan, R.A.; Dahut, W.L. Angiogenesis Inhibition in Prostate Cancer: Current Uses and Future Promises. J. Oncol. 2010, 2010, 361836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, A.; Roobol, M.J.; Savage, C.J.; Peltola, M.; Pettersson, K.; Scardino, P.T.; Vickers, A.J.; Schröder, F.H.; Lilja, H. A four-kallikrein panel for the prediction of repeat prostate biopsy: Data from the European Randomized Study of Prostate Cancer screening in Rotterdam, Netherlands. Br. J. Cancer 2010, 103, 708–714. [Google Scholar] [CrossRef] [Green Version]
- Scattoni, V.; Lazzeri, M.; Lughezzani, G.; De Luca, S.; Passera, R.; Bollito, E.; Randone, D.; Abdollah, F.; Capitanio, U.; Larcher, A.; et al. Head-to-head comparison of prostate health index and urinary PCA3 for predicting cancer at initial or repeat biopsy. J. Urol. 2013, 190, 496–501. [Google Scholar] [CrossRef]
- Wei, J.T.; Feng, Z.; Partin, A.W.; Brown, E.; Thompson, I.; Sokoll, L.; Chan, D.W.; Lotan, Y.; Kibel, A.S.; Busby, J.E.; et al. Can urinary PCA3 supplement PSA in the early detection of prostate cancer? J. Clin. Oncol. 2014, 32, 4066–4072. [Google Scholar] [CrossRef] [Green Version]
- Weinreb, J.C.; Barentsz, J.O.; Choyke, P.L.; Cornud, F.; Haider, M.A.; Macura, K.J.; Margolis, D.; Schnall, M.D.; Shtern, F.; Tempany, C.M.; et al. PI-RADS Prostate Imaging—Reporting and Data System: 2015, Version 2. Eur. Urol. 2016, 69, 16–40. [Google Scholar] [CrossRef]
- Woo, S.; Suh, C.H.; Kim, S.Y.; Cho, J.Y.; Kim, S.H. Diagnostic Performance of Prostate Imaging Reporting and Data System Version 2 for Detection of Prostate Cancer: A Systematic Review and Diagnostic Meta-analysis. Eur. Urol. 2017, 72, 177–188. [Google Scholar] [CrossRef]
- Epstein, J.I.; Egevad, L.; Amin, M.B.; Delahunt, B.; Srigley, J.R.; Humphrey, P.A. 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]
- Mohler, J.L.; Armstrong, A.J.; Bahnson, R.R.; D’Amico, A.V.; Davis, B.J.; Eastham, J.A.; Enke, C.A.; Farrington, T.A.; Higano, C.S.; Horwitz, E.M.; et al. Prostate Cancer, Version 1.2016. J. Natl. Compr. Cancer Netw. 2016, 14, 19–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Epstein, J.I.; Zelefsky, M.J.; Sjoberg, D.D.; Nelson, J.B.; Egevad, L.; Magi-Galluzzi, C.; Vickers, A.J.; Parwani, A.V.; Reuter, V.E.; Fine, S.W.; et al. A Contemporary Prostate Cancer Grading System: A Validated Alternative to the Gleason Score. Eur. Urol. 2016, 69, 428–435. [Google Scholar] [CrossRef] [Green Version]
- Sommariva, S.; Tarricone, R.; Lazzeri, M.; Ricciardi, W.; Montorsi, F. Prognostic Value of the Cell Cycle Progression Score in Patients with Prostate Cancer: A Systematic Review and Meta-analysis. Eur. Urol. 2016, 69, 107–115. [Google Scholar] [CrossRef]
- Lindenberg, M.L.; Turkbey, B.; Mena, E.; Choyke, P.L. Imaging Locally Advanced, Recurrent, and Metastatic Prostate Cancer: A Review. JAMA Oncol. 2017, 3, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
- Filson, C.P.; Marks, L.S.; Litwin, M.S. Expectant management for men with early stage prostate cancer. CA Cancer J. Clin. 2015, 65, 265–282. [Google Scholar] [CrossRef] [Green Version]
- Ficarra, V.; Novara, G.; Rosen, R.C.; Artibani, W.; Carroll, P.R.; Costello, A.; Menon, M.; Montorsi, F.; Patel, V.R.; Stolzenburg, J.U.; et al. Systematic review and meta-analysis of studies reporting urinary continence recovery after robot-assisted radical prostatectomy. Eur. Urol. 2012, 62, 405–417. [Google Scholar] [CrossRef] [PubMed]
- Ficarra, V.; Novara, G.; Ahlering, T.E.; Costello, A.; Eastham, J.A.; Graefen, M.; Guazzoni, G.; Menon, M.; Mottrie, A.; Patel, V.R.; et al. Systematic review and meta-analysis of studies reporting potency rates after robot-assisted radical prostatectomy. Eur. Urol. 2012, 62, 418–430. [Google Scholar] [CrossRef] [PubMed]
- Wortel, R.C.; Incrocci, L.; Pos, F.J.; Lebesque, J.V.; Witte, M.G.; van der Heide, U.A.; van Herk, M.; Heemsbergen, W.D. Acute toxicity after image-guided intensity modulated radiation therapy compared to 3D conformal radiation therapy in prostate cancer patients. Int. J. Radiat. Oncol. Biol. Phys. 2015, 91, 737–744. [Google Scholar] [CrossRef] [PubMed]
- Viani, G.A.; Viana, B.S.; Martin, J.E.; Rossi, B.T.; Zuliani, G.; Stefano, E.J. Intensity-modulated radiotherapy reduces toxicity with similar biochemical control compared with 3-dimensional conformal radiotherapy for prostate cancer: A randomized clinical trial. Cancer 2016, 122, 2004–2011. [Google Scholar] [CrossRef] [Green Version]
- Ryan, C.J.; Smith, M.R.; de Bono, J.S.; Molina, A.; Logothetis, C.J.; de Souza, P.; Fizazi, K.; Mainwaring, P.; Piulats, J.M.; Ng, S.; et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 2013, 368, 138–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Bono, J.S.; Oudard, S.; Ozguroglu, M.; Hansen, S.; Machiels, J.P.; Kocak, I.; Gravis, G.; Bodrogi, I.; Mackenzie, M.J.; Shen, L.; et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: A randomised open-label trial. Lancet 2010, 376, 1147–1154. [Google Scholar] [CrossRef]
- Beer, T.M.; Armstrong, A.J.; Rathkopf, D.E.; Loriot, Y.; Sternberg, C.N.; Higano, C.S.; Iversen, P.; Bhattacharya, S.; Carles, J.; Chowdhury, S.; et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 2014, 371, 424–433. [Google Scholar] [CrossRef] [Green Version]
- Kantoff, P.W.; Higano, C.S.; Shore, N.D.; Berger, E.R.; Small, E.J.; Penson, D.F.; Redfern, C.H.; Ferrari, A.C.; Dreicer, R.; Sims, R.B.; et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N. Engl. J. Med. 2010, 363, 411–422. [Google Scholar] [CrossRef] [Green Version]
- Fizazi, K.; Tran, N.; Fein, L.; Matsubara, N.; Rodriguez-Antolin, A.; Alekseev, B.Y.; Özgüroğlu, M.; Ye, D.; Feyerabend, S.; Protheroe, A.; et al. Abiraterone plus Prednisone in Metastatic, Castration-Sensitive Prostate Cancer. N. Engl. J. Med. 2017, 377, 352–360. [Google Scholar] [CrossRef]
- James, N.D.; de Bono, J.S.; Spears, M.R.; Clarke, N.W.; Mason, M.D.; Dearnaley, D.P.; Ritchie, A.W.S.; Amos, C.L.; Gilson, C.; Jones, R.J.; et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N. Engl. J. Med. 2017, 377, 338–351. [Google Scholar] [CrossRef] [PubMed]
- Chi, K.N.; Agarwal, N.; Bjartell, A.; Chung, B.H.; Pereira de Santana Gomes, A.J.; Given, R.; Juárez Soto, Á.; Merseburger, A.S.; Özgüroğlu, M.; Uemura, H.; et al. Apalutamide for Metastatic, Castration-Sensitive Prostate Cancer. N. Engl. J. Med. 2019, 381, 13–24. [Google Scholar] [CrossRef] [PubMed]
- Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021. Epub ahead of Print. [Google Scholar] [CrossRef]
- Dahut, W.L.; Gulley, J.L.; Arlen, P.M.; Liu, Y.; Fedenko, K.M.; Steinberg, S.M.; Wright, J.J.; Parnes, H.; Chen, C.C.; Jones, E.; et al. Randomized phase II trial of docetaxel plus thalidomide in androgen-independent prostate cancer. J. Clin. Oncol. 2004, 22, 2532–2539. [Google Scholar] [CrossRef] [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, 329–333. [Google Scholar] [CrossRef]
- Melegh, Z.; Oltean, S. Targeting Angiogenesis in Prostate Cancer. Int. J. Mol. Sci. 2019, 20, 2676. [Google Scholar] [CrossRef] [Green Version]
- Escaff, S.; Fernández, J.M.; González, L.O.; Suárez, A.; González-Reyes, S.; González, J.M.; Vizoso, F.J. Study of matrix metalloproteinases and their inhibitors in prostate cancer. Br. J. Cancer 2010, 102, 922–929. [Google Scholar] [CrossRef] [Green Version]
- Botelho, F.; Pina, F.; Lunet, N. VEGF and prostatic cancer: A systematic review. Eur. J. Cancer Prev. 2010, 19, 385–392. [Google Scholar] [CrossRef]
- Wikström, P.; Damber, J.; Bergh, A. Role of transforming growth factor-beta1 in prostate cancer. Microsc. Res. Tech. 2001, 52, 411–419. [Google Scholar] [CrossRef]
- Hussain, T.; Gupta, S.; Mukhtar, H. Cyclooxygenase-2 and prostate carcinogenesis. Cancer Lett. 2003, 191, 125–135. [Google Scholar] [CrossRef]
- Chung, L.W.; Baseman, A.; Assikis, V.; Zhau, H.E. Molecular insights into prostate cancer progression: The missing link of tumor microenvironment. J. Urol. 2005, 173, 10–20. [Google Scholar] [CrossRef]
- Bono, A.V.; Celato, N.; Cova, V.; Salvadore, M.; Chinetti, S.; Novario, R. Microvessel density in prostate carcinoma. Prostate Cancer Prostatic Dis. 2002, 5, 123–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Olsson, A.K.; Dimberg, A.; Kreuger, J.; Claesson-Welsh, L. VEGF receptor signalling—in control of vascular function. Nat. Rev. Mol. Cell Biol. 2006, 7, 359–371. [Google Scholar] [CrossRef]
- Roberts, E.; Cossigny, D.A.; Quan, G.M. The role of vascular endothelial growth factor in metastatic prostate cancer to the skeleton. Prostate Cancer 2013, 2013, 418340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delongchamps, N.B.; Peyromaure, M. The role of vascular endothelial growth factor in kidney and prostate cancer. Can. J. Urol. 2007, 14, 3669–3677. [Google Scholar]
- Li, R.; Younes, M.; Wheeler, T.M.; Scardino, P.; Ohori, M.; Frolov, A.; Ayala, G. Expression of vascular endothelial growth factor receptor-3 (VEGFR-3) in human prostate. Prostate 2004, 58, 193–199. [Google Scholar] [CrossRef]
- Duque, J.L.; Loughlin, K.R.; Adam, R.M.; Kantoff, P.W.; Zurakowski, D.; Freeman, M.R. Plasma levels of vascular endothelial growth factor are increased in patients with metastatic prostate cancer. Urology 1999, 54, 523–527. [Google Scholar] [CrossRef]
- Jennbacken, K.; Vallbo, C.; Wang, W.; Damber, J.E. Expression of vascular endothelial growth factor C (VEGF-C) and VEGF receptor-3 in human prostate cancer is associated with regional lymph node metastasis. Prostate 2005, 65, 110–116. [Google Scholar] [CrossRef] [PubMed]
- West, A.F.; O’Donnell, M.; Charlton, R.G.; Neal, D.E.; Leung, H.Y. Correlation of vascular endothelial growth factor expression with fibroblast growth factor-8 expression and clinico-pathologic parameters in human prostate cancer. Br. J. Cancer 2001, 85, 576–583. [Google Scholar] [CrossRef] [Green Version]
- Cao, S.; Durrani, F.A.; Toth, K.; Rustum, Y.M.; Seshadri, M. Bevacizumab enhances the therapeutic efficacy of Irinotecan against human head and neck squamous cell carcinoma xenografts. Oral Oncol. 2011, 47, 459–466. [Google Scholar] [CrossRef] [Green Version]
- Drugs@FDA: FDA-Approved Drugs. Available online: https://www.accessdata.fda.gov/scripts/cder/daf/ (accessed on 14 July 2021).
- Ribatti, D.; Vacca, A. New Insights in Anti-Angiogenesis in Multiple Myeloma. Int. J. Mol. Sci. 2018, 19, 2031. [Google Scholar] [CrossRef] [Green Version]
- Yun, Y.R.; Won, J.E.; Jeon, E.; Lee, S.; Kang, W.; Jo, H.; Jang, J.H.; Shin, U.S.; Kim, H.W. Fibroblast Growth Factors: Biology, Function, and Application for Tissue Regeneration. J. Tissue Eng. 2010, 2010, 218142. [Google Scholar] [CrossRef] [PubMed]
- Fujisato, T.; Sajiki, T.; Liu, Q.; Ikada, Y. Effect of basic fibroblast growth factor on cartilage regeneration in chondrocyte-seeded collagen sponge scaffold. Biomaterials 1996, 17, 155–162. [Google Scholar] [CrossRef]
- Wang, J.S. Basic fibroblast growth factor for stimulation of bone formation in osteoinductive or conductive implants. Acta Orthop. Scand. 1996, 269, 1–33. [Google Scholar] [CrossRef]
- Matsusaki, M.; Ochi, M.; Uchio, Y.; Shu, N.; Kurioka, H.; Kawasaki, K.; Adachi, N. Effects of Basic Fibroblast Growth Factor on Proliferation and Phenotype Expression of Chondrocytes Embedded in Collagen Gel. Gen. Pharmacol. 1998, 31, 759–764. [Google Scholar] [CrossRef]
- Harper, M.E.; Glynne-Jones, E.; Goddard, L.; Thurston, V.J.; Griffiths, K. Vascular endothelial growth factor (VEGF) expression in prostatic tumours and its relationship to neuroendocrine cells. Br. J. Cancer 1996, 74, 910–916. [Google Scholar] [CrossRef] [Green Version]
- Liang, W.C.; Wu, X.; Peale, F.V.; Lee, C.V.; Meng, Y.G.; Gutierrez, J.; Fu, L.; Malik, A.K.; Gerber, H.P.; Ferrara, N.; et al. Cross-species vascular endothelial growth factor (VEGF)-blocking antibodies completely inhibit the growth of human tumor xenografts and measure the contribution of stromal VEGF. J. Biol. Chem. 2006, 281, 951–961. [Google Scholar] [CrossRef] [Green Version]
- Deep, G.; Jain, A.; Kumar, A.; Agarwal, C.; Kim, S.; Leevy, W.M.; Agarwal, R. Exosomes secreted by prostate cancer cells under hypoxia promote matrix metalloproteinases activity at pre-metastatic niches. Mol. Carcinog. 2020, 59, 323–332. [Google Scholar] [CrossRef]
- Katiyar, S.K. Matrix Metalloproteinases in Cancer Metastasis: Molecular Targets for Prostate Cancer Prevention by Green Tea Polyphenols and Grape Seed Proanthocyanidins. Endocr. Metab. Immune Disord. Drug Targets 2006, 6, 17–24. [Google Scholar] [CrossRef]
- Franko, A.; Berti, L.; Hennenlotter, J.; Rausch, S.; Scharpf, M.O.; Angelis, M.H.; Stenzl, A.; Peter, A.; Birkenfeld, A.L.; Lutz, S.Z.; et al. Increased Expressions of Matrix Metalloproteinases (MMPs) in Prostate Cancer Tissues of Men with Type 2 Diabetes. Biomedicines 2020, 8, 507. [Google Scholar] [CrossRef]
- Alaseem, A.; Alhazzani, K.; Dondapati, P.; Alobid, S.; Bishayee, A.; Rathinavelu, A. Matrix Metalloproteinases: A challenging paradigm of cancer management. Semin. Cancer Biol. 2019, 56, 100–115. [Google Scholar] [CrossRef] [PubMed]
- Andrae, J.; Gallini, R.; Betsholtz, C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008, 22, 1276–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahel, J.; Hudorović, N.; Vičić-Hudorović, V.; Nikles, H. TGF-Beta in the Natural History of Prostate Cancer. Acta Clin. Croat. 2019, 58, 128–138. [Google Scholar] [CrossRef] [Green Version]
- Barrett, C.S.; Millena, A.C.; Khan, S.A. TGF-β Effects on Prostate Cancer Cell Migration and Invasion Require FosB. Prostate 2017, 77, 72–81. [Google Scholar] [CrossRef] [Green Version]
- Fournier, P.G.; Juárez, P.; Jiang, G.; Clines, G.A.; Niewolna, M.; Kim, H.S.; Walton, H.W.; Peng, X.H.; Liu, Y.; Mohammad, K.S.; et al. The TGF-β Signaling Regulator PMEPA1 Suppresses Prostate Cancer Metastases to Bone. Cancer Cell 2015, 27, 809–821. [Google Scholar] [CrossRef] [Green Version]
- Kloss, C.C.; Lee, J.; Zhang, A.; Chen, F.; Melenhorst, J.J.; Lacey, S.F.; Maus, M.V.; Fraietta, J.A.; Zhao, Y.; June, C.H. Dominant-Negative TGF-β Receptor Enhances PSMA-Targeted Human CAR T Cell Proliferation and Augments Prostate Cancer Eradication. Mol. Ther. 2018, 26, 1855–1866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mu, H.Q.; He, Y.H.; Wang, S.B.; Yang, S.; Wang, Y.J.; Nan, C.J.; Bao, Y.F.; Xie, Q.P.; Chen, Y.H. MiR-130b/TNF-α/NF-κB/VEGFA loop inhibits prostate cancer angiogenesis. Clin. Transl. Oncol. 2020, 22, 111–121. [Google Scholar] [CrossRef]
- Teixeira, A.F.; Ten Dijke, P.; Zhu, H.J. On-Target Anti-TGF-β Therapies Are Not Succeeding in Clinical Cancer Treatments: What Are Remaining Challenges? Front. Cell Dev. Biol. 2020, 8, 605. [Google Scholar] [CrossRef]
- Semenza, G.L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2010, 29, 625–634. [Google Scholar] [CrossRef] [Green Version]
- Janssen, H.L.; Haustermans, K.M.; Sprong, D.; Blommestijn, G.; Hofland, I.; Hoebers, F.J.; Blijweert, E.; Raleigh, J.A.; Semenza, G.L.; Varia, M.A.; et al. HIF-1A, pimonidazole, and iododeoxyuridine to estimate hypoxia and perfusion in human head-and-neck tumors. Int. J. Radiat. Oncol. Biol. Phys. 2002, 54, 1537–1549. [Google Scholar] [CrossRef]
- Boddy, J.L.; Fox, S.B.; Han, C.; Campo, L.; Turley, H.; Kanga, S.; Malone, P.R.; Harris, A.L. The Androgen Receptor Is Significantly Associated with Vascular Endothelial Growth Factor and Hypoxia Sensing via Hypoxia-Inducible Factors HIF-1a, HIF-2a, and the Prolyl Hydroxylases in Human Prostate Cancer. Clin. Cancer Res. 2005, 11, 7658–7663. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Cheng, Y.; Zhang, Z.; Bai, Z.; Jin, H.; Guo, X.; Huang, X.; Li, M.; Wang, M.; Shu, X.S.; et al. CDCA2 Inhibits Apoptosis and Promotes Cell Proliferation in Prostate Cancer and Is Directly Regulated by HIF-1α Pathway. Front. Oncol. 2020, 10, 725. [Google Scholar] [CrossRef] [PubMed]
- Pavlakis, D.; Kampantais, S.; Gkagkalidis, K.; Gourvas, V.; Memmos, D.; Tsionga, A.; Dimitriadis, G.; Vakalopoulos, I. Hypoxia-Inducible Factor 2a Expression Is Positively Correlated with Gleason Score in Prostate Cancer. Technol. Cancer Res. Treat. 2021, 20, 1533033821990010. [Google Scholar] [CrossRef]
- Zha, S.; Yegnasubramanian, V.; Nelson, W.G.; Isaacs, W.B.; De Marzo, A.M. Cyclooxygenases in cancer: Progress and perspective. Cancer Lett. 2004, 215, 1–20. [Google Scholar] [CrossRef]
- Zheng, Y.; Comaills, V.; Burr, R.; Boulay, G.; Miyamoto, D.T.; Wittner, B.S.; Emmons, E.; Sil, S.; Koulopoulos, M.W.; Broderick, K.T.; et al. COX-2 mediates tumor-stromal prolactin signaling to initiate tumorigenesis. Proc. Natl. Acad. Sci. USA 2019, 116, 5223–5232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doat, S.; Cénée, S.; Trétarre, B.; Rebillard, X.; Lamy, P.J.; Bringer, J.P.; Iborra, F.; Murez, T.; Sanchez, M.; Menegaux, F. Nonsteroidal anti-inflammatory drugs (NSAIDs) and prostate cancer risk: Results from the EPICAP study. Cancer Med. 2017, 6, 2461–2470. [Google Scholar] [CrossRef] [PubMed]
- Garg, R.; Blando, J.M.; Perez, C.J.; Lal, P.; Feldman, M.D.; Smyth, E.M.; Ricciotti, E.; Grosser, T.; Benavides, F.; Kazanietz, M.G. COX-2 mediates pro-tumorigenic effects of PKCε in prostate cancer. Oncogene 2018, 37, 4735–4749. [Google Scholar] [CrossRef]
- Tian, J.; Guo, F.; Chen, Y.; Li, Y.; Yu, B.; Li, Y. Nanoliposomal formulation encapsulating celecoxib and genistein inhibiting COX-2 pathway and Glut-1 receptors to prevent prostate cancer cell proliferation. Cancer Lett. 2019, 448, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Liu, S.; Parajuli, K.R.; Zhang, W.; Zhang, K.; Mo, Z.; Liu, J.; Chen, Z.; Yang, S.; Wang, A.R.; et al. Interleukin-17 promotes prostate cancer via MMP7-induced epithelial-to-mesenchymal transition. Oncogene 2017, 36, 687–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.H.; Ho, C.H.; Hu, S.W.; Tzou, K.Y.; Wang, Y.H.; Wu, C.C. Association between interleukin-8 rs4073 polymorphism and prostate cancer: A meta-analysis. J. Formos. Med. Assoc. 2020, 119, 1201–1210. [Google Scholar] [CrossRef]
- Culig, Z.; Puhr, M. Interleukin-6 and prostate cancer: Current developments and unsolved questions. Mol. Cell. Endocrinol. 2018, 462, 25–30. [Google Scholar] [CrossRef]
- Nguyen, D.P.; Li, J.; Tewari, A.K. Inflammation and prostate cancer: The role of interleukin 6 (IL-6): Role of IL-6 in prostate cancer. BJU Int. 2014, 113, 986–992. [Google Scholar] [CrossRef]
- Sakellariou, C.; Elhage, O.; Papaevangelou, E.; Giustarini, G.; Esteves, A.M.; Smolarek, D.; Smith, R.A.; Dasgupta, P.; Galustian, C. Prostate cancer cells enhance interleukin-15-mediated expansion of NK cells. BJU Int. 2020, 125, 89–102. [Google Scholar] [CrossRef] [Green Version]
- Bergers, G.; Benjamin, L.E. Tumorigenesis and the angiogenic switch. Nat. Rev. Cancer 2003, 3, 401–410. [Google Scholar] [CrossRef]
- Boussios, S.; Ozturk, M.A.; Moschetta, M.; Karathanasi, A.; Zakynthinakis-Kyriakou, N.; Katsanos, K.H.; Christodoulou, D.K.; Pavlidis, N. The Developing Story of Predictive Biomarkers in Colorectal Cancer. J. Pers. Med. 2019, 9, 12. [Google Scholar] [CrossRef] [Green Version]
- Zarkavelis, G.; Boussios, S.; Papadaki, A.; Katsanos, K.H.; Christodoulou, D.K.; Pentheroudakis, G. Current and future biomarkers in colorectal cancer. Ann. Gastroenterol. 2017, 30, 613–621. [Google Scholar] [CrossRef] [PubMed]
- Boussios, S.; Mikropoulos, C.; Samartzis, E.; Karihtala, P.; Moschetta, M.; Sheriff, M.; Karathanasi, A.; Sadauskaite, A.; Rassy, E.; Pavlidis, N. Wise Management of Ovarian Cancer: On the Cutting Edge. J. Pers. Med. 2020, 10, 41. [Google Scholar] [CrossRef]
- Boussios, S.; Moschetta, M.; Karathanasi, A.; Tsiouris, A.K.; Kanellos, F.S.; Tatsi, K.; Katsanos, K.H.; Christodoulou, D.K. Malignant peritoneal mesothelioma: Clinical aspects, and therapeutic perspectives. Ann. Gastroenterol. 2018, 31, 659–669. [Google Scholar] [CrossRef] [PubMed]
- Revythis, A.; Shah, S.; Kutka, M.; Moschetta, M.; Ozturk, M.A.; Pappas-Gogos, G.; Ioannidou, E.; Sheriff, M.; Rassy, E.; Boussios, S. Unraveling the Wide Spectrum of Melanoma Biomarkers. Diagnostics 2021, 11, 1341. [Google Scholar] [CrossRef]
- Fabris, L.; Ceder, Y.; Chinnaiyan, A.M.; Jenster, G.W.; Sorensen, K.D.; Tomlins, S.; Visakorpi, T.; Calin, G.A. The Potential of MicroRNAs as Prostate Cancer Biomarkers. Eur. Urol. 2016, 70, 312–322. [Google Scholar] [CrossRef] [Green Version]
- Saxby, H.; Mikropoulos, C.; Boussios, S. An Update on the Prognostic and Predictive Serum Biomarkers in Metastatic Prostate Cancer. Diagnostics 2020, 10, 549. [Google Scholar] [CrossRef] [PubMed]
- Balacescu, O.; Dumitrescu, R.G.; Marian, C. MicroRNAs Role in Prostate Cancer. Methods Mol. Biol. 2018, 1856, 103–117. [Google Scholar]
- Bryzgunova, O.E.; Konoshenko, M.Y.; Laktionov, P.P. MicroRNA-guided gene expression in prostate cancer: Literature and database overview. J. Gene Med. 2018, 20, e3016. [Google Scholar] [CrossRef]
- Kanwal, R.; Plaga, A.R.; Liu, X.; Shukla, G.C.; Gupta, S. MicroRNAs in prostate cancer: Functional role as biomarkers. Cancer Lett. 2017, 407, 9–20. [Google Scholar] [CrossRef]
- Kasomva, K.; Sen, A.; Paulraj, M.G.; Sailo, S.; Raphael, V.; Puro, K.U.; Assumi, S.R.; Ignacimuthu, S. Roles of microRNA in prostate cancer cell metabolism. Int. J. Biochem. Cell Biol. 2018, 102, 109–116. [Google Scholar] [CrossRef]
- Khanmi, K.; Ignacimuthu, S.; Paulraj, M.G. MicroRNA in prostate cancer. Clin. Chim. Acta 2015, 451, 154–160. [Google Scholar] [CrossRef] [PubMed]
- Sharma, N.; Baruah, M.M. The microRNA signatures: Aberrantly expressed miRNAs in prostate cancer. Clin. Transl. Oncol. 2019, 21, 126–144. [Google Scholar] [CrossRef] [PubMed]
- Cassinello, J.; Carballido Rodríguez, J.; Antón Aparicio, L. Role of taxanes in advanced prostate cancer. Clin. Transl. Oncol. 2016, 18, 972–980. [Google Scholar] [CrossRef]
- Thienger, P.; Rubin, M.A. Prostate cancer hijacks the microenvironment. Nat. Cell Biol. 2021, 23, 3–5. [Google Scholar] [CrossRef]
- Luz Flores, M.; Sáez, C. Protocols for the Study of Taxanes Chemosensitivity in Prostate Cancer. Methods Mol. Biol. 2018, 1786, 153–173. [Google Scholar]
- Petrylak, D.P. Docetaxel (Taxotere) in hormone-refractory prostate cancer. Semin. Oncol. 2000, 27, 24–29. [Google Scholar]
- Stein, C.A. Mechanisms of action of taxanes in prostate cancer. Semin. Oncol. 1999, 26, 3–7. [Google Scholar]
- Small, E.J.; Srinivas, S.; Egan, B.; McMillan, A.; Rearden, T.P. Doxorubicin and dose-escalated cyclophosphamide with granulocyte colony-stimulating factor for the treatment of hormone-resistant prostate cancer. J. Clin. Oncol. 1996, 14, 1617–1625. [Google Scholar] [CrossRef]
- Ceci, C.; Atzori, M.G.; Lacal, P.M.; Graziani, G. Role of VEGFs/VEGFR-1 Signaling and its Inhibition in Modulating Tumor Invasion: Experimental Evidence in Different Metastatic Cancer Models. Int. J. Mol. Sci. 2020, 21, 1388. [Google Scholar] [CrossRef] [Green Version]
- Reese, D.M.; Fratesi, P.; Corry, M.; Novotny, W.; Holmgren, E.; Small, E.J. A Phase II Trial of Humanized Anti-Vascular Endothelial Growth Factor Antibody for the Treatment of Androgen-Independent Prostate Cancer. Prostate J. 2001, 3, 65–70. [Google Scholar] [CrossRef]
- Iacobelli, S. Hormone-refractory prostate cancer responding to bevacizumab. Int. J. Urol. 2008, 15, 754. [Google Scholar] [CrossRef] [PubMed]
- Picus, J.; Halabi, S.; Kelly, W.K.; Vogelzang, N.J.; Whang, Y.E.; Kaplan, E.B.; Stadler, W.M.; Small, E.J.; Cancer and Leukemia Group B. A phase 2 study of estramustine, docetaxel, and bevacizumab in men with castrate-resistant prostate cancer: Results from Cancer and Leukemia Group B Study 90006. Cancer 2011, 117, 526–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Lorenzo, G.; Figg, W.D.; Fossa, S.D.; Mirone, V.; Autorino, R.; Longo, N.; Imbimbo, C.; Perdonà, S.; Giordano, A.; Giuliano, M.; et al. Combination of bevacizumab and docetaxel in docetaxel-pretreated hormone-refractory prostate cancer: A phase 2 study. Eur. Urol. 2008, 54, 1089–1094. [Google Scholar] [CrossRef]
- Kelly, W.K.; Halabi, S.; Carducci, M.; George, D.; Mahoney, J.F.; Stadler, W.M.; Morris, M.; Kantoff, P.; Monk, J.P.; Kaplan, E.; et al. Randomized, double-blind, placebo-controlled phase III trial comparing docetaxel and prednisone with or without bevacizumab in men with metastatic castration-resistant prostate cancer: CALGB 90401. J. Clin. Oncol. 2012, 30, 1534–1540. [Google Scholar] [CrossRef] [PubMed]
- Holash, J.; Davis, S.; Papadopoulos, N.; Croll, S.D.; Ho, L.; Russell, M.; Boland, P.; Leidich, R.; Hylton, D.; Burova, E.; et al. VEGF-Trap: A VEGF blocker with potent antitumor effects. Proc. Natl. Acad. Sci. USA 2002, 99, 11393–11398. [Google Scholar] [CrossRef] [Green Version]
- Tannock, I.F.; Fizazi, K.; Ivanov, S.; Karlsson, C.T.; Fléchon, A.; Skoneczna, I.; Orlandi, F.; Gravis, G.; Matveev, V.; Bavbek, S.; et al. Aflibercept versus placebo in combination with docetaxel and prednisone for treatment of men with metastatic castration-resistant prostate cancer (VENICE): A phase 3, double-blind randomised trial. Lancet Oncol. 2013, 14, 760–768. [Google Scholar] [CrossRef]
- Chi, K.N.; Ellard, S.L.; Hotte, S.J.; Czaykowski, P.; Moore, M.; Ruether, J.D.; Schell, A.J.; Taylor, S.; Hansen, C.; Gauthier, I.; et al. A phase II study of sorafenib in patients with chemo-naive castration-resistant prostate cancer. Ann. Oncol. 2008, 19, 746–751. [Google Scholar] [CrossRef] [PubMed]
- Dahut, W.L.; Scripture, C.; Posadas, E.; Jain, L.; Gulley, J.L.; Arlen, P.M.; Wright, J.J.; Yu, Y.; Cao, L.; Steinberg, S.M.; et al. A phase II clinical trial of sorafenib in androgen-independent prostate cancer. Clin. Cancer Res. 2008, 14, 209–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steinbild, S.; Mross, K.; Frost, A.; Morant, R.; Gillessen, S.; Dittrich, C.; Strumberg, D.; Hochhaus, A.; Hanauske, A.R.; Edler, L.; et al. A clinical phase II study with sorafenib in patients with progressive hormone-refractory prostate cancer: A study of the CESAR Central European Society for Anticancer Drug Research-EWIV. Br. J. Cancer 2007, 97, 1480–1485. [Google Scholar] [CrossRef]
- Rini, B.I.; Garcia, J.A.; Cooney, M.M.; Elson, P.; Tyler, A.; Beatty, K.; Bokar, J.; Mekhail, T.; Bukowski, R.M.; Budd, G.T.; et al. A phase I study of sunitinib plus bevacizumab in advanced solid tumors. Clin. Cancer Res. 2009, 15, 6277–6283. [Google Scholar] [CrossRef] [Green Version]
- Dror Michaelson, M.; Regan, M.M.; Oh, W.K.; Kaufman, D.S.; Olivier, K.; Michaelson, S.Z.; Spicer, B.; Gurski, C.; Kantoff, P.W.; Smith, M.R. Phase II study of sunitinib in men with advanced prostate cancer. Ann. Oncol. 2009, 20, 913–920. [Google Scholar] [CrossRef]
- Payton, S. Targeting prostate tumour angiogenesis with cediranib. Nat. Rev. Urol. 2013, 10, 187. [Google Scholar] [CrossRef]
- Karakunnel, J.J.; Gulley, J.L.; Arlen, P.M.; Mulquin, M.; Wright, J.J.; Turkbey, I.B.; Choyke, P.; Ahlers, C.M.; Figg, W.D.; Dahut, W.L. Phase II trial of cediranib (AZD2171) in docetaxel-resistant, castrate-resistant prostate cancer (CRPC). J. Clin. Oncol. 2008, 26, 283s. [Google Scholar] [CrossRef]
- Guérin, O.; Etienne-Grimaldi, M.C.; Monteverde, M.; Sudaka, A.; Brunstein, M.C.; Formento, P.; Lattanzio, L.; Maffi, M.; Tonissi, F.; Ortholan, C.; et al. Contrasted effects of the multitarget TKi vandetanib on docetaxel-sensitive and docetaxel-resistant prostate cancer cell lines. Urol. Oncol. 2013, 31, 1567–1575. [Google Scholar] [CrossRef]
- Horti, J.; Widmark, A.; Stenzl, A.; Federico, M.H.; Abratt, R.P.; Sanders, N.; Pover, G.M.; Bodrogi, I. A Randomized, Double-Blind, Placebo-Controlled Phase II Study of Vandetanib Plus Docetaxel/Prednisolone in Patients with Hormone-Refractory Prostate Cancer. Cancer Biother. Radiopharm. 2009, 24, 175–180. [Google Scholar] [CrossRef]
- Azad, A.A.; Beardsley, E.K.; Hotte, S.J.; Ellard, S.L.; Klotz, L.; Chin, J.; Kollmannsberger, C.; Mukherjee, S.D.; Chi, K.N. A randomized phase II efficacy and safety study of vandetanib (ZD6474) in combination with bicalutamide versus bicalutamide alone in patients with chemotherapy naïve castration-resistant prostate cancer. Investig. New Drugs 2014, 32, 746–752. [Google Scholar] [CrossRef]
- Cochin, V.; Gross-Goupil, M.; Ravaud, A.; Godbert, Y.; Le Moulec, S. Cabozantinib: Mechanism of action, efficacy and indications. Bull. Cancer 2017, 104, 393–401. [Google Scholar] [CrossRef] [PubMed]
- Choueiri, T.K.; Hessel, C.; Halabi, S.; Sanford, B.; Michaelson, M.D.; Hahn, O.; Walsh, M.; Olencki, T.; Picus, J.; Small, E.J.; et al. Cabozantinib versus sunitinib as initial therapy for metastatic renal cell carcinoma of intermediate or poor risk (Alliance A031203 CABOSUN randomised trial): Progression-free survival by independent review and overall survival update. Eur. J. Cancer 2018, 94, 115–125. [Google Scholar] [CrossRef] [Green Version]
- Choueiri, T.K.; Escudier, B.; Powles, T.; Tannir, N.M.; Mainwaring, P.N.; Rini, B.I.; Hammers, H.J.; Donskov, F.; Roth, B.J.; Peltola, K.; et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): Final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016, 17, 917–927. [Google Scholar] [CrossRef] [Green Version]
- Brose, M.S.; Robinson, B.; Sherman, S.I.; Krajewska, J.; Lin, C.C.; Vaisman, F.; Hoff, A.O.; Hitre, E.; Bowles, D.W.; Hernando, J.; et al. Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 1126–1138. [Google Scholar] [CrossRef]
- Chott, A.; Sun, Z.; Morganstern, D.; Pan, J.; Li, T.; Susani, M.; Mosberger, I.; Upton, M.P.; Bubley, G.J.; Balk, S.P. Tyrosine kinases expressed in vivo by human prostate cancer bone marrow metastases and loss of the type 1 insulin-like growth factor receptor. Am. J. Pathol. 1999, 155, 1271–1279. [Google Scholar] [CrossRef] [Green Version]
- Patnaik, A.; Swanson, K.D.; Csizmadia, E.; Solanki, A.; Landon-Brace, N.; Gehring, M.P.; Helenius, K.; Olson, B.M.; Pyzer, A.R.; Wang, L.C.; et al. Cabozantinib Eradicates Advanced Murine Prostate Cancer by Activating Antitumor Innate Immunity. Cancer Discov. 2017, 7, 750–765. [Google Scholar] [CrossRef] [Green Version]
- Sidaway, P. Cabozantinib activates innate immunity. Nat. Rev. Urol. 2017, 14, 327. [Google Scholar] [CrossRef]
- Basch, E.M.; Scholz, M.; de Bono, J.S.; Vogelzang, N.; de Souza, P.; Marx, G.; Vaishampayan, U.; George, S.; Schwarz, J.K.; Antonarakis, E.S.; et al. Cabozantinib Versus Mitoxantrone-prednisone in Symptomatic Metastatic Castration-resistant Prostate Cancer: A Randomized Phase 3 Trial with a Primary Pain Endpoint. Eur. Urol. 2019, 75, 929–937. [Google Scholar] [CrossRef] [Green Version]
- Agarwal, N.; Loriot, Y.; McGregor, B.A.; Dreicer, R.; Dorff, T.B.; Maughan, B.L.; Kelly, W.K.; Pagliaro, L.C.; Srinivas, S.; Squillante, C.M.; et al. Cabozantinib in combination with atezolizumab in patients with metastatic castration-resistant prostate cancer: Results of cohort 6 of the COSMIC-021 study. J. Clin. Oncol. 2020, 38, 5564. [Google Scholar] [CrossRef]
- Lin, A.M.; Rini, B.I.; Weinberg, V.; Fong, K.; Ryan, C.J.; Rosenberg, J.E.; Fong, L.; Small, E.J. A phase II trial of imatinib mesylate in patients with biochemical relapse of prostate cancer after definitive local therapy. BJU Int. 2006, 98, 763–769. [Google Scholar] [CrossRef]
- Rao, K.; Goodin, S.; Levitt, M.J.; Dave, N.; Shih, W.J.; Lin, Y.; Capanna, T.; Doyle-Lindrud, S.; Juvidian, P.; DiPaola, R.S. A phase II trial of imatinib mesylate in patients with prostate specific antigen progression after local therapy for prostate cancer. Prostate 2005, 62, 115–122. [Google Scholar] [CrossRef]
- Bajaj, G.K.; Zhang, Z.; Garrett-Mayer, E.; Drew, R.; Sinibaldi, V.; Pili, R.; Denmeade, S.R.; Carducci, M.A.; Eisenberger, M.A.; DeWeese, T.L. Phase II study of imatinib mesylate in patients with prostate cancer with evidence of biochemical relapse after definitive radical retropubic prostatectomy or radiotherapy. Urology 2007, 69, 526–531. [Google Scholar] [CrossRef]
- Hail, N., Jr.; Chen, P.; Bushman, L.R. Teriflunomide (Leflunomide) Promotes Cytostatic, Antioxidant, and Apoptotic Effects in Transformed Prostate Epithelial Cells: Evidence Supporting a Role for Teriflunomide in Prostate Cancer Chemoprevention. Neoplasia 2010, 12, 464–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Figg, W.D.; Li, H.; Sissung, T.; Retter, A.; Wu, S.; Gulley, J.L.; Arlen, P.; Wright, J.J.; Parnes, H.; Fedenko, K.; et al. Pre-clinical and clinical evaluation of estramustine, docetaxel and thalidomide combination in androgen-independent prostate cancer. BJU Int. 2007, 99, 1047–1055. [Google Scholar] [CrossRef] [PubMed]
- Pytel, D.; Sliwinski, T.; Poplawski, T.; Ferriola, D.; Majsterek, I. Tyrosine Kinase Blockers: New Hope for Successful Cancer Therapy. Anticancer Agents. Med. Chem. 2009, 9, 66–76. [Google Scholar] [CrossRef] [PubMed]
- D’Amato, R.J.; Loughnan, M.S.; Flynn, E.; Folkman, J. Thalidomide is an inhibitor of angiogenesis. Proc. Natl. Acad. Sci. USA 1994, 91, 4082–4085. [Google Scholar] [CrossRef] [Green Version]
- Dixon, S.C.; Kruger, E.A.; Bauer, K.S.; Figg, W.D. Thalidomide up-regulates prostate-specific antigen secretion from LNCaP cells. Cancer Chemother. Pharmacol. 1999, 43, S78–S84. [Google Scholar] [CrossRef]
- Figg, W.D.; Dahut, W.; Duray, P.; Hamilton, M.; Tompkins, A.; Steinberg, S.M.; Jones, E.; Premkumar, A.; Linehan, W.M.; Floeter, M.K.; et al. A randomized phase II trial of thalidomide, an angiogenesis inhibitor, in patients with androgen-independent prostate cancer. Clin. Cancer Res. 2001, 7, 1888–1893. [Google Scholar]
- Figg, W.D.; Arlen, P.; Gulley, J.; Fernandez, P.; Noone, M.; Fedenko, K.; Hamilton, M.; Parker, C.; Kruger, E.A.; Pluda, J.; et al. A randomized phase II trial of docetaxel (taxotere) plus thalidomide in androgen-independent prostate cancer. Semin. Oncol. 2001, 28, 62–66. [Google Scholar] [CrossRef]
- Ning, Y.M.; Gulley, J.L.; Arlen, P.M.; Woo, S.; Steinberg, S.M.; Wright, J.J.; Parnes, H.L.; Trepel, J.B.; Lee, M.J.; Kim, Y.S.; et al. Phase II trial of bevacizumab, thalidomide, docetaxel, and prednisone in patients with metastatic castration-resistant prostate cancer. J. Clin. Oncol. 2010, 28, 2070–2076. [Google Scholar] [CrossRef] [Green Version]
- Xing, D.L.; Song, D.K.; Zhang, L.R. Lenalidomide in Treating Patients with Castration-Resistant Prostate Cancer. Asian Pac. J. Cancer Prev. 2015, 16, 3969–3972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sinibaldi, V.J.; Carducci, M.A.; Moore-Cooper, S.; George, B.; Denmeade, S.; Drake, C.G.; Walczak, J.; Pili, R.; Zahurak, M.L.; Eisenberger, M.A. A randomized double blind phase I-II study to determine the tolerability/efficacy of two different doses of lenalidomide (L), CC- 5013, in biochemically relapsed (BR) prostate cancer (PC) patients (pts) (M0) after local treatment (LT). J. Clin. Oncol. 2009, 27, 5130. [Google Scholar] [CrossRef]
- Petrylak, D.P.; Vogelzang, N.J.; Budnik, N.; Wiechno, P.J.; Sternberg, C.N.; Doner, K.; Bellmunt, J.; Burke, J.M.; de Olza, M.O.; Choudhury, A.; et al. Docetaxel and prednisone with or without lenalidomide in chemotherapy-naive patients with metastatic castration-resistant prostate cancer (MAINSAIL): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2015, 16, 417–425. [Google Scholar] [CrossRef]
- Dalrymple, S.L.; Becker, R.E.; Isaacs, J.T. The quinoline-3-carboxamide anti-angiogenic agent, tasquinimod, enhances the anti-prostate cancer efficacy of androgen ablation and taxotere without effecting serum PSA directly in human xenografts. Prostate 2007, 67, 790–797. [Google Scholar] [CrossRef] [PubMed]
- Bratt, O.; Häggman, M.; Ahlgren, G.; Nordle, O.; Björk, A.; Damber, J.E. Open-label, clinical phase I studies of tasquinimod in patients with castration-resistant prostate cancer. Br. J. Cancer 2009, 101, 1233–1240. [Google Scholar] [CrossRef] [Green Version]
- Pili, R.; Häggman, M.; Stadler, W.M.; Gingrich, J.R.; Assikis, V.J.; Björk, A.; Nordle, O.; Forsberg, G.; Carducci, M.A.; Armstrong, A.J. Phase II randomized, double-blind, placebo-controlled study of tasquinimod in men with minimally symptomatic metastatic castrate-resistant prostate cancer. J. Clin. Oncol. 2011, 29, 4022–4028. [Google Scholar] [CrossRef]
- Pantziarka, P.; Sukhatme, V.; Bouche, G.; Meheus, L.; Sukhatme, V.P. Repurposing Drugs in Oncology (ReDO)-itraconazole as an anti-cancer agent. Ecancermedicalscience 2015, 9, 521. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.; Hong, H.; Kim, W.; Zhang, L.; Friedlander, T.W.; Fong, L.; Lin, A.M.; Small, E.J.; Wei, X.X.; Rodvelt, T.J.; et al. Itraconazole as a Noncastrating Treatment for Biochemically Recurrent Prostate Cancer: A Phase 2 Study. Clin. Genitourin. Cancer 2019, 17, e92–e96. [Google Scholar] [CrossRef]
- Antonarakis, E.S.; Heath, E.I.; Smith, D.C.; Rathkopf, D.; Blackford, A.L.; Danila, D.C.; King, S.; Frost, A.; Ajiboye, A.S.; Zhao, M.; et al. Repurposing Itraconazole as a Treatment for Advanced Prostate Cancer: A Noncomparative Randomized Phase II Trial in Men With Metastatic Castration-Resistant Prostate Cancer. Oncologist 2013, 18, 163–173. [Google Scholar] [CrossRef] [PubMed]
- Bilusic, M.; Wong, Y.N. Anti-angiogenesis in prostate cancer: Knocked down but not out. Asian J. Androl. 2014, 16, 372–377. [Google Scholar] [PubMed]
- Leary, S.E.S.; Park, J.R.; Reid, J.M.; Ralya, A.T.; Baruchel, S.; Wu, B.; Roberts, T.P.L.; Liu, X.; Minard, C.G.; Fox, E.; et al. Pediatric Phase I Trial and Pharmacokinetic Study of Trebananib in Relapsed Solid Tumors, Including Primary Tumors of the Central Nervous System ADVL1115: A Children’s Oncology Group Phase I Consortium Report. Clin. Cancer. Res. 2017, 23, 6062–6069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pan, J.; Zang, L.; Zhang, Y.; Hong, J.; Yao, Y.; Zou, C.; Zhang, L.; Chen, Y. Early changes in apparent diffusion coefficients predict radiosensitivity of human nasopharyngeal carcinoma xenografts. Laryngoscope 2012, 122, 839–843. [Google Scholar] [CrossRef]
- Gule, M.K.; Chen, Y.; Sano, D.; Frederick, M.J.; Zhou, G.; Zhao, M.; Milas, Z.L.; Galer, C.E.; Henderson, Y.C.; Jasser, S.A.; et al. Targeted therapy of VEGFR2 and EGFR significantly inhibits growth of anaplastic thyroid cancer in an orthotopic murine model. Clin. Cancer Res. 2011, 17, 2281–2291. [Google Scholar] [CrossRef] [Green Version]
- Isayeva, T.; Chanda, D.; Kallman, L.; Eltoum, I.E.; Ponnazhagan, S. Effects of sustained antiangiogenic therapy in multistage prostate cancer in TRAMP model. Cancer Res. 2007, 67, 5789–5797. [Google Scholar] [CrossRef] [Green Version]
Pathway | Factor | Clinical Impact | References |
---|---|---|---|
Vascular Endothelial Growth factors (VEGFs) | VEGFR 1, -2 and -3 | Stimulators | [36,37,38,39,40,41,42,43,44,45] |
Fibroblast growth factors (FGFs) | FGF-1 and -2, bFGF | [46,47,48,49,50,51] | |
Matrix metalloproteinases (MMPs) | MMP-2, -7 and -9 | [52,53,54,55] | |
Transforming growth factor β (TGFβ) | TGFβ1 | [56,57,58,59,60,61,62] | |
Hypoxia-inducible factors (HIF) | HIF-1a | [63,64,65,66,67] | |
Cyclooxygenases (COXs) | COX2 | [68,69,70,71,72] | |
Interleukins (ILs) | IL8 | Stimulator | [74,76,77] |
IL10 and IL27 | Inhibitors | [73,76,78] | |
microRNAs (miRNAs) | miR-296, miR-30d, miR-323, miR-21, miR-182 and miR-130b | Stimulators | [88] |
miR-195, miR-218 and miR-146a | Inhibitors | [87] |
Agent | Mechanism of Action | Phase | Primary Endpoint | Identifier |
---|---|---|---|---|
Bevacizumab | Recombinant humanized monoclonal antibody that blocks VEGF-A | II | ORR | NCT01083368 |
II | PSA rFS | NCT00776594 | ||
Aflibercept | Binds to circulating VEGF-A | III | OS | NCT00519285 |
Sunitinib | Receptor tyrosine kinase inhibitor | II | ≥30% PSA decline | NCT00879619 |
II | PFS | NCT00734851 | ||
Sorafenib | II | Overall response rate | NCT00414388 | |
II | ≥50% PSA decline | NCT00589420 | ||
Cediranib | Inhibitor of VEGFR-1, -2, and -3 | II | PFS | NCT00527124 |
II | PFS | NCT01260688 | ||
Cabozantinib | Inhibits VEGFRs, MET, and RET | II | PFS | NCT01428219 |
Thalidomide | Inhibition of VEGF, PI3K/Akt/NF-kappaB, and mTOR pathways | II | ORR | NCT00307294 |
Lenalidomide | Inhibition of VEGF-induced PI3K/Akt pathway signalling | II | OS | NCT00942578 |
Tasquinimod | S100A9 protein that inhibits VEGF | III | PFS | NCT01234311 |
Itraconazole | Inhibition of the Hedgehog pathway | II | ≥30% PSA decline | NCT01450683 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ioannidou, E.; Moschetta, M.; Shah, S.; Parker, J.S.; Ozturk, M.A.; Pappas-Gogos, G.; Sheriff, M.; Rassy, E.; Boussios, S. Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets. Int. J. Mol. Sci. 2021, 22, 9926. https://doi.org/10.3390/ijms22189926
Ioannidou E, Moschetta M, Shah S, Parker JS, Ozturk MA, Pappas-Gogos G, Sheriff M, Rassy E, Boussios S. Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets. International Journal of Molecular Sciences. 2021; 22(18):9926. https://doi.org/10.3390/ijms22189926
Chicago/Turabian StyleIoannidou, Evangelia, Michele Moschetta, Sidrah Shah, Jack Steven Parker, Mehmet Akif Ozturk, George Pappas-Gogos, Matin Sheriff, Elie Rassy, and Stergios Boussios. 2021. "Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets" International Journal of Molecular Sciences 22, no. 18: 9926. https://doi.org/10.3390/ijms22189926
APA StyleIoannidou, E., Moschetta, M., Shah, S., Parker, J. S., Ozturk, M. A., Pappas-Gogos, G., Sheriff, M., Rassy, E., & Boussios, S. (2021). Angiogenesis and Anti-Angiogenic Treatment in Prostate Cancer: Mechanisms of Action and Molecular Targets. International Journal of Molecular Sciences, 22(18), 9926. https://doi.org/10.3390/ijms22189926