Epithelial-to-Mesenchymal Transition-Related Markers in Prostate Cancer: From Bench to Bedside
Abstract
:Simple Summary
Abstract
1. Introduction
2. EMT and MET as Targets for Therapy
3. Epithelial-To-Mesenchymal Transition in Cancer Progression
3.1. EMT Initiation
3.2. EMT Transcription Factors
3.3. EMT Signaling Pathways
3.3.1. TGF-β Signaling
3.3.2. Receptor Tyrosine Kinase Signaling
3.3.3. Wnt Signaling
3.3.4. Notch Signaling
3.3.5. Hedgehog Signaling
4. Studies on EMT in Prostate Cancer
4.1. In Vitro and In Vivo Studies
4.1.1. EMT Surface Markers
4.1.2. EMT Transcription Markers
4.1.3. Tumor Microenvironment
4.1.4. TGF-β Signaling
4.1.5. RTK Signaling
4.1.6. WNT Signaling
4.1.7. Notch Signaling
4.1.8. Hedgehog Signaling
4.1.9. PI3K/AKT Signaling
4.1.10. Integration of the Different Signaling Pathways in PCa Progression
4.2. Translational and Clinical Studies
5. Potential Biomarkers of Interest for Targeted Therapy
6. Clinical Trials
7. Conclusions and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Global Burden of Disease Cancer Collaboration. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017, 3, 524–548. [Google Scholar] [CrossRef]
- Gogola, S.; Rejzer, M.; Bahmad, H.F.; Alloush, F.; Omarzai, Y.; Poppiti, R. Anti-Cancer Stem-Cell-Targeted Therapies in Prostate Cancer. Cancers 2023, 15, 1621. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
- Butler, S.S.; Muralidhar, V.; Zhao, S.G.; Sanford, N.N.; Franco, I.; Fullerton, Z.H.; Chavez, J.; D’Amico, A.V.; Feng, F.Y.; Rebbeck, T.R.; et al. Prostate cancer incidence across stage, NCCN risk groups, and age before and after USPSTF Grade D recommendations against prostate-specific antigen screening in 2012. Cancer 2020, 126, 717–724. [Google Scholar] [CrossRef]
- Ilic, D.; Djulbegovic, M.; Jung, J.H.; Hwang, E.C.; Zhou, Q.; Cleves, A.; Agoritsas, T.; Dahm, P. Prostate cancer screening with prostate-specific antigen (PSA) test: A systematic review and meta-analysis. BMJ 2018, 362, k3519. [Google Scholar] [CrossRef] [Green Version]
- Epstein, J.I. An update of the Gleason grading system. J. Urol. 2010, 183, 433–440. [Google Scholar] [CrossRef]
- 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]
- 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]
- Debes, J.D.; Tindall, D.J. Mechanisms of androgen-refractory prostate cancer. N. Engl. J. Med. 2004, 351, 1488–1490. [Google Scholar] [CrossRef]
- 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]
- Pienta, K.J.; Bradley, D. Mechanisms underlying the development of androgen-independent prostate cancer. Clin. Cancer Res. 2006, 12, 1665–1671. [Google Scholar] [CrossRef] [Green Version]
- Feldman, B.J.; Feldman, D. The development of androgen-independent prostate cancer. Nat. Rev. Cancer 2001, 1, 34–45. [Google Scholar] [CrossRef]
- Nelson, W.G.; de Marzo, A.M.; Isaacs, W.B. Prostate cancer. N. Engl. J. Med. 2003, 349, 366–381. [Google Scholar] [CrossRef]
- Shah, R.B.; Mehra, R.; Chinnaiyan, A.M.; Shen, R.; Ghosh, D.; Zhou, M.; Macvicar, G.R.; Varambally, S.; Harwood, J.; Bismar, T.A.; et al. Androgen-independent prostate cancer is a heterogeneous group of diseases: Lessons from a rapid autopsy program. Cancer Res. 2004, 64, 9209–9216. [Google Scholar] [CrossRef] [Green Version]
- Bahmad, H.F.; Demus, T.; Moubarak, M.M.; Daher, D.; Alvarez Moreno, J.C.; Polit, F.; Lopez, O.; Merhe, A.; Abou-Kheir, W.; Nieder, A.M.; et al. Overcoming Drug Resistance in Advanced Prostate Cancer by Drug Repurposing. Med. Sci. 2022, 10, 15. [Google Scholar] [CrossRef]
- Bahmad, H.F.; Cheaito, K.; Chalhoub, R.M.; Hadadeh, O.; Monzer, A.; Ballout, F.; El-Hajj, A.; Mukherji, D.; Liu, Y.N.; Daoud, G.; et al. Sphere-Formation Assay: Three-Dimensional in vitro Culturing of Prostate Cancer Stem/Progenitor Sphere-Forming Cells. Front. Oncol. 2018, 8, 347. [Google Scholar] [CrossRef] [Green Version]
- Bahmad, H.F.; Jalloul, M.; Azar, J.; Moubarak, M.M.; Samad, T.A.; Mukherji, D.; Al-Sayegh, M.; Abou-Kheir, W. Tumor Microenvironment in Prostate Cancer: Toward Identification of Novel Molecular Biomarkers for Diagnosis, Prognosis, and Therapy Development. Front. Genet. 2021, 12, 652747. [Google Scholar] [CrossRef]
- Martin-Belmonte, F.; Mostov, K. Regulation of cell polarity during epithelial morphogenesis. Curr. Opin. Cell Biol. 2008, 20, 227–234. [Google Scholar] [CrossRef]
- Bryant, D.M.; Mostov, K.E. From cells to organs: Building polarized tissue. Nat. Rev. Mol. Cell Biol. 2008, 9, 887–901. [Google Scholar] [CrossRef]
- Barriere, G.; Fici, P.; Gallerani, G.; Fabbri, F.; Rigaud, M. Epithelial Mesenchymal Transition: A double-edged sword. Clin. Transl. Med. 2015, 4, 14. [Google Scholar] [CrossRef] [Green Version]
- Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
- Eastham, A.M.; Spencer, H.; Soncin, F.; Ritson, S.; Merry, C.L.; Stern, P.L.; Ward, C.M. Epithelial-mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res. 2007, 67, 11254–11262. [Google Scholar] [CrossRef] [Green Version]
- Ullmann, U.; In’t Veld, P.; Gilles, C.; Sermon, K.; de Rycke, M.; van de Velde, H.; van Steirteghem, A.; Liebaers, I. Epithelial-mesenchymal transition process in human embryonic stem cells cultured in feeder-free conditions. Mol. Hum. Reprod. 2007, 13, 21–32. [Google Scholar] [CrossRef] [Green Version]
- Azar, J.; Bahmad, H.F.; Daher, D.; Moubarak, M.M.; Hadadeh, O.; Monzer, A.; Al Bitar, S.; Jamal, M.; Al-Sayegh, M.; Abou-Kheir, W. The Use of Stem Cell-Derived Organoids in Disease Modeling: An Update. Int. J. Mol. Sci. 2021, 22, 47667. [Google Scholar] [CrossRef]
- Papaccio, F.; della Corte, C.M.; Viscardi, G.; di Liello, R.; Esposito, G.; Sparano, F.; Ciardiello, F.; Morgillo, F. HGF/MET and the Immune System: Relevance for Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 113595. [Google Scholar] [CrossRef] [Green Version]
- Chaves, L.P.; Melo, C.M.; Saggioro, F.P.; Reis, R.B.D.; Squire, J.A. Epithelial-Mesenchymal Transition Signaling and Prostate Cancer Stem Cells: Emerging Biomarkers and Opportunities for Precision Therapeutics. Genes 2021, 12, 1900. [Google Scholar] [CrossRef]
- Katz, L.H.; Li, Y.; Chen, J.S.; Muñoz, N.M.; Majumdar, A.; Chen, J.; Mishra, L. Targeting TGF-β signaling in cancer. Expert Opin. Ther. Targets 2013, 17, 743–760. [Google Scholar] [CrossRef] [Green Version]
- Owusu, B.Y.; Galemmo, R.; Janetka, J.; Klampfer, L. Hepatocyte Growth Factor, a Key Tumor-Promoting Factor in the Tumor Microenvironment. Cancers 2017, 9, 40035. [Google Scholar] [CrossRef] [Green Version]
- Strutz, F.; Zeisberg, M.; Ziyadeh, F.N.; Yang, C.Q.; Kalluri, R.; Müller, G.A.; Neilson, E.G. Role of basic fibroblast growth factor-2 in epithelial-mesenchymal transformation. Kidney Int. 2002, 61, 1714–1728. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.G.; Kay, E.P. Cross-talk among Rho GTPases acting downstream of PI 3-kinase induces mesenchymal transformation of corneal endothelial cells mediated by FGF-2. Invest. Ophthalmol. Vis. Sci. 2006, 47, 2358–2368. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Kong, J.; Chang, H.; Kim, H.; Kim, A. EGF induces epithelial-mesenchymal transition through phospho-Smad2/3-Snail signaling pathway in breast cancer cells. Oncotarget 2016, 7, 85021–85032. [Google Scholar] [CrossRef] [Green Version]
- Wu, Q.; Hou, X.; Xia, J.; Qian, X.; Miele, L.; Sarkar, F.H.; Wang, Z. Emerging roles of PDGF-D in EMT progression during tumorigenesis. Cancer Treat. Rev. 2013, 39, 640–646. [Google Scholar] [CrossRef] [Green Version]
- Tam, S.Y.; Wu, V.W.C.; Law, H.K.W. Hypoxia-Induced Epithelial-Mesenchymal Transition in Cancers: HIF-1α and Beyond. Front. Oncol. 2020, 10, 486. [Google Scholar] [CrossRef]
- Nissen, N.I.; Karsdal, M.; Willumsen, N. Collagens and Cancer associated fibroblasts in the reactive stroma and its relation to Cancer biology. J. Exp. Clin. Cancer Res. 2019, 38, 115. [Google Scholar] [CrossRef] [Green Version]
- Vallés, A.M.; Boyer, B.; Tarone, G.; Thiery, J.P. Alpha 2 beta 1 integrin is required for the collagen and FGF-1 induced cell dispersion in a rat bladder carcinoma cell line. Cell Adhes. Commun. 1996, 4, 187–199. [Google Scholar] [CrossRef]
- Garamszegi, N.; Garamszegi, S.P.; Samavarchi-Tehrani, P.; Walford, E.; Schneiderbauer, M.M.; Wrana, J.L.; Scully, S.P. Extracellular matrix-induced transforming growth factor-beta receptor signaling dynamics. Oncogene 2010, 29, 2368–2380. [Google Scholar] [CrossRef] [Green Version]
- Medici, D.; Nawshad, A. Type I collagen promotes epithelial-mesenchymal transition through ILK-dependent activation of NF-kappaB and LEF-1. Matrix Biol. 2010, 29, 161–165. [Google Scholar] [CrossRef] [Green Version]
- Koenig, A.; Mueller, C.; Hasel, C.; Adler, G.; Menke, A. Collagen type I induces disruption of E-cadherin-mediated cell-cell contacts and promotes proliferation of pancreatic carcinoma cells. Cancer Res. 2006, 66, 4662–4671. [Google Scholar] [CrossRef] [Green Version]
- Oyanagi, J.; Ogawa, T.; Sato, H.; Higashi, S.; Miyazaki, K. Epithelial-mesenchymal transition stimulates human cancer cells to extend microtubule-based invasive protrusions and suppresses cell growth in collagen gel. PLoS ONE 2012, 7, e53209. [Google Scholar] [CrossRef] [Green Version]
- Nisticò, P.; Bissell, M.J.; Radisky, D.C. Epithelial-mesenchymal transition: General principles and pathological relevance with special emphasis on the role of matrix metalloproteinases. Cold Spring Harb. Perspect. Biol. 2012, 4, 11908. [Google Scholar] [CrossRef]
- Stallings-Mann, M.L.; Waldmann, J.; Zhang, Y.; Miller, E.; Gauthier, M.L.; Visscher, D.W.; Downey, G.P.; Radisky, E.S.; Fields, A.P.; Radisky, D.C. Matrix metalloproteinase induction of Rac1b, a key effector of lung cancer progression. Sci. Transl. Med. 2012, 4, 142ra195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stemmler, M.P.; Eccles, R.L.; Brabletz, S.; Brabletz, T. Non-redundant functions of EMT transcription factors. Nat. Cell Biol. 2019, 21, 102–112. [Google Scholar] [CrossRef] [PubMed]
- Gras, B.; Jacqueroud, L.; Wierinckx, A.; Lamblot, C.; Fauvet, F.; Lachuer, J.; Puisieux, A.; Ansieau, S. Snail family members unequally trigger EMT and thereby differ in their ability to promote the neoplastic transformation of mammary epithelial cells. PLoS ONE 2014, 9, e92254. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Shi, J.; Chai, K.; Ying, X.; Zhou, B.P. The Role of Snail in EMT and Tumorigenesis. Curr. Cancer Drug Targets 2013, 13, 963–972. [Google Scholar] [CrossRef] [Green Version]
- Batlle, E.; Sancho, E.; Francí, C.; Domínguez, D.; Monfar, M.; Baulida, J.; García de Herreros, A. The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat. Cell Biol. 2000, 2, 84–89. [Google Scholar] [CrossRef]
- Olmeda, D.; Moreno-Bueno, G.; Flores, J.M.; Fabra, A.; Portillo, F.; Cano, A. SNAI1 is required for tumor growth and lymph node metastasis of human breast carcinoma MDA-MB-231 cells. Cancer Res. 2007, 67, 11721–11731. [Google Scholar] [CrossRef] [Green Version]
- Jin, H.; Yu, Y.; Zhang, T.; Zhou, X.; Zhou, J.; Jia, L.; Wu, Y.; Zhou, B.P.; Feng, Y. Snail is critical for tumor growth and metastasis of ovarian carcinoma. Int. J. Cancer 2010, 126, 2102–2111. [Google Scholar] [CrossRef]
- Min, A.L.; Choi, J.Y.; Woo, H.Y.; Kim, J.D.; Kwon, J.H.; Bae, S.H.; Yoon, S.K.; Shin, S.H.; Chung, Y.J.; Jung, C.K. High expression of Snail mRNA in blood from hepatocellular carcinoma patients with extra-hepatic metastasis. Clin. Exp. Metastasis 2009, 26, 759–767. [Google Scholar] [CrossRef]
- Kudo-Saito, C.; Shirako, H.; Takeuchi, T.; Kawakami, Y. Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 2009, 15, 195–206. [Google Scholar] [CrossRef] [Green Version]
- Vesuna, F.; van Diest, P.; Chen, J.H.; Raman, V. Twist is a transcriptional repressor of E-cadherin gene expression in breast cancer. Biochem. Biophys. Res. Commun. 2008, 367, 235–241. [Google Scholar] [CrossRef] [Green Version]
- Alexander, N.R.; Tran, N.L.; Rekapally, H.; Summers, C.E.; Glackin, C.; Heimark, R.L. N-cadherin gene expression in prostate carcinoma is modulated by integrin-dependent nuclear translocation of Twist1. Cancer Res. 2006, 66, 3365–3369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Casas, E.; Kim, J.; Bendesky, A.; Ohno-Machado, L.; Wolfe, C.J.; Yang, J. Snail2 is an essential mediator of Twist1-induced epithelial mesenchymal transition and metastasis. Cancer Res. 2011, 71, 245–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, Z.; Rahman, M.A.; Chen, Z.G.; Shin, D.M. Multiple biological functions of Twist1 in various cancers. Oncotarget 2017, 8, 20380–20393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eger, A.; Aigner, K.; Sonderegger, S.; Dampier, B.; Oehler, S.; Schreiber, M.; Berx, G.; Cano, A.; Beug, H.; Foisner, R. DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene 2005, 24, 2375–2385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aigner, K.; Dampier, B.; Descovich, L.; Mikula, M.; Sultan, A.; Schreiber, M.; Mikulits, W.; Brabletz, T.; Strand, D.; Obrist, P.; et al. The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene 2007, 26, 6979–6988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perez-Oquendo, M.; Gibbons, D.L. Regulation of ZEB1 Function and Molecular Associations in Tumor Progression and Metastasis. Cancers 2022, 14, 1864. [Google Scholar] [CrossRef] [PubMed]
- Pang, M.F.; Georgoudaki, A.M.; Lambut, L.; Johansson, J.; Tabor, V.; Hagikura, K.; Jin, Y.; Jansson, M.; Alexander, J.S.; Nelson, C.M.; et al. TGF-β1-induced EMT promotes targeted migration of breast cancer cells through the lymphatic system by the activation of CCR7/CCL21-mediated chemotaxis. Oncogene 2016, 35, 748–760. [Google Scholar] [CrossRef]
- Kang, M.H.; Kang, H.N.; Kim, J.L.; Kim, J.S.; Oh, S.C.; Yoo, Y.A. Inhibition of PI3 kinase/Akt pathway is required for BMP2-induced EMT and invasion. Oncol. Rep. 2009, 22, 525–534. [Google Scholar] [CrossRef] [Green Version]
- Deng, G.; Chen, Y.; Guo, C.; Yin, L.; Han, Y.; Li, Y.; Fu, Y.; Cai, C.; Shen, H.; Zeng, S. BMP4 promotes the metastasis of gastric cancer by inducing epithelial-mesenchymal transition via ID1. J. Cell Sci. 2020, 133, jcs237222. [Google Scholar] [CrossRef]
- Huang, F.; Chen, Y.G. Regulation of TGF-β receptor activity. Cell Biosci. 2012, 2, 9. [Google Scholar] [CrossRef] [Green Version]
- Garg, M. Epithelial-mesenchymal transition—Activating transcription factors—Multifunctional regulators in cancer. World J. Stem Cells 2013, 5, 188–195. [Google Scholar] [CrossRef] [PubMed]
- Irie, H.Y.; Pearline, R.V.; Grueneberg, D.; Hsia, M.; Ravichandran, P.; Kothari, N.; Natesan, S.; Brugge, J.S. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J. Cell Biol. 2005, 171, 1023–1034. [Google Scholar] [CrossRef] [PubMed]
- Chaudhury, A.; Hussey, G.S.; Ray, P.S.; Jin, G.; Fox, P.L.; Howe, P.H. TGF-beta-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI. Nat. Cell Biol. 2010, 12, 286–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, B.P.; Deng, J.; Xia, W.; Xu, J.; Li, Y.M.; Gunduz, M.; Hung, M.C. Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat. Cell Biol. 2004, 6, 931–940. [Google Scholar] [CrossRef]
- Xie, L.; Law, B.K.; Chytil, A.M.; Brown, K.A.; Aakre, M.E.; Moses, H.L. Activation of the Erk pathway is required for TGF-beta1-induced EMT in vitro. Neoplasia 2004, 6, 603–610. [Google Scholar] [CrossRef] [Green Version]
- Du, Z.; Lovly, C.M. Mechanisms of receptor tyrosine kinase activation in cancer. Mol. Cancer 2018, 17, 58. [Google Scholar] [CrossRef]
- Guo, X.; Wang, X.F. Signaling cross-talk between TGF-beta/BMP and other pathways. Cell Res. 2009, 19, 71–88. [Google Scholar] [CrossRef]
- Graham, T.R.; Zhau, H.E.; Odero-Marah, V.A.; Osunkoya, A.O.; Kimbro, K.S.; Tighiouart, M.; Liu, T.; Simons, J.W.; O’Regan, R.M. Insulin-like growth factor-I-dependent up-regulation of ZEB1 drives epithelial-to-mesenchymal transition in human prostate cancer cells. Cancer Res. 2008, 68, 2479–2488. [Google Scholar] [CrossRef] [Green Version]
- Grotegut, S.; von Schweinitz, D.; Christofori, G.; Lehembre, F. Hepatocyte growth factor induces cell scattering through MAPK/Egr-1-mediated upregulation of Snail. Embo. J. 2006, 25, 3534–3545. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Chrisman, H.; Weijer, C.J. PDGF signalling controls the migration of mesoderm cells during chick gastrulation by regulating N-cadherin expression. Development 2008, 135, 3521–3530. [Google Scholar] [CrossRef] [Green Version]
- Chuang, H.H.; Zhen, Y.Y.; Tsai, Y.C.; Chuang, C.H.; Hsiao, M.; Huang, M.S.; Yang, C.J. FAK in Cancer: From Mechanisms to Therapeutic Strategies. Int. J. Mol. Sci. 2022, 23, 31726. [Google Scholar] [CrossRef] [PubMed]
- Drake, J.M.; Graham, N.A.; Stoyanova, T.; Sedghi, A.; Goldstein, A.S.; Cai, H.; Smith, D.A.; Zhang, H.; Komisopoulou, E.; Huang, J.; et al. Oncogene-specific activation of tyrosine kinase networks during prostate cancer progression. Proc. Natl. Acad. Sci. USA 2012, 109, 1643–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tatarov, O.; Mitchell, T.J.; Seywright, M.; Leung, H.Y.; Brunton, V.G.; Edwards, J. SRC family kinase activity is up-regulated in hormone-refractory prostate cancer. Clin. Cancer Res. 2009, 15, 3540–3549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Feng, R. Inhibition of epithelial to mesenchymal transition in metastatic breast carcinoma cells by c-Src suppression. Acta Biochim. Biophys. Sin. 2010, 42, 496–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagathihalli, N.S.; Merchant, N.B. Src-mediated regulation of E-cadherin and EMT in pancreatic cancer. Front. Biosci. 2012, 17, 2059–2069. [Google Scholar] [CrossRef] [Green Version]
- Patel, S.; Alam, A.; Pant, R.; Chattopadhyay, S. Wnt Signaling and Its Significance Within the Tumor Microenvironment: Novel Therapeutic Insights. Front. Immunol. 2019, 10, 2872. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.; Lu, Z.; Hay, E.D. Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol. Int. 2002, 26, 463–476. [Google Scholar] [CrossRef]
- Zucchini-Pascal, N.; Peyre, L.; Rahmani, R. Crosstalk between beta-catenin and snail in the induction of epithelial to mesenchymal transition in hepatocarcinoma: Role of the ERK1/2 pathway. Int. J. Mol. Sci. 2013, 14, 20768–20792. [Google Scholar] [CrossRef] [Green Version]
- Wu, Z.Q.; Li, X.Y.; Hu, C.Y.; Ford, M.; Kleer, C.G.; Weiss, S.J. Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc. Natl. Acad. Sci. USA 2012, 109, 16654–16659. [Google Scholar] [CrossRef] [Green Version]
- Howe, L.R.; Watanabe, O.; Leonard, J.; Brown, A.M. Twist is up-regulated in response to Wnt1 and inhibits mouse mammary cell differentiation. Cancer Res. 2003, 63, 1906–1913. [Google Scholar]
- Bray, S.J. Notch signalling: A simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 2006, 7, 678–689. [Google Scholar] [CrossRef] [PubMed]
- Guo, D.; Ye, J.; Dai, J.; Li, L.; Chen, F.; Ma, D.; Ji, C. Notch-1 regulates Akt signaling pathway and the expression of cell cycle regulatory proteins cyclin D1, CDK2 and p21 in T-ALL cell lines. Leuk. Res. 2009, 33, 678–685. [Google Scholar] [CrossRef] [PubMed]
- Maniati, E.; Bossard, M.; Cook, N.; Candido, J.B.; Emami-Shahri, N.; Nedospasov, S.A.; Balkwill, F.R.; Tuveson, D.A.; Hagemann, T. Crosstalk between the canonical NF-κB and Notch signaling pathways inhibits Pparγ expression and promotes pancreatic cancer progression in mice. J. Clin. Invest. 2011, 121, 4685–4699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leong, K.G.; Niessen, K.; Kulic, I.; Raouf, A.; Eaves, C.; Pollet, I.; Karsan, A. Jagged1-mediated Notch activation induces epithelial-to-mesenchymal transition through Slug-induced repression of E-cadherin. J. Exp. Med. 2007, 204, 2935–2948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sahlgren, C.; Gustafsson, M.V.; Jin, S.; Poellinger, L.; Lendahl, U. Notch signaling mediates hypoxia-induced tumor cell migration and invasion. Proc. Natl. Acad. Sci. USA 2008, 105, 6392–6397. [Google Scholar] [CrossRef] [Green Version]
- Xie, M.; Zhang, L.; He, C.S.; Xu, F.; Liu, J.L.; Hu, Z.H.; Zhao, L.P.; Tian, Y. Activation of Notch-1 enhances epithelial-mesenchymal transition in gefitinib-acquired resistant lung cancer cells. J. Cell. Biochem. 2012, 113, 1501–1513. [Google Scholar] [CrossRef]
- Sari, I.N.; Phi, L.T.H.; Jun, N.; Wijaya, Y.T.; Lee, S.; Kwon, H.Y. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells 2018, 7, 110208. [Google Scholar] [CrossRef] [Green Version]
- Fendrich, V.; Waldmann, J.; Esni, F.; Ramaswamy, A.; Mullendore, M.; Buchholz, M.; Maitra, A.; Feldmann, G. Snail and Sonic Hedgehog activation in neuroendocrine tumors of the ileum. Endocr. Relat. Cancer 2007, 14, 865–874. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Deng, W.; Nail, C.D.; Bailey, S.K.; Kraus, M.H.; Ruppert, J.M.; Lobo-Ruppert, S.M. Snail induction is an early response to Gli1 that determines the efficiency of epithelial transformation. Oncogene 2006, 25, 609–621. [Google Scholar] [CrossRef] [Green Version]
- Yoo, Y.A.; Kang, M.H.; Kim, J.S.; Oh, S.C. Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Carcinogenesis 2008, 29, 480–490. [Google Scholar] [CrossRef] [Green Version]
- Xia, R.; Xu, M.; Yang, J.; Ma, X. The role of Hedgehog and Notch signaling pathway in cancer. Mol. Biomed. 2022, 3, 44. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Ma, L.; Zhang, Z.; Liu, X.; Gao, H.; Zhuang, Y.; Yang, P.; Kornmann, M.; Tian, X.; Yang, Y. Hedgehog Signaling Regulates Epithelial-Mesenchymal Transition in Pancreatic Cancer Stem-Like Cells. J. Cancer 2016, 7, 408–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsai, Y.C.; Chen, W.Y.; Abou-Kheir, W.; Zeng, T.; Yin, J.J.; Bahmad, H.; Lee, Y.C.; Liu, Y.N. Androgen deprivation therapy-induced epithelial-mesenchymal transition of prostate cancer through downregulating SPDEF and activating CCL2. Biochim. Biophys. Acta Mol. Basis. Dis. 2018, 1864, 1717–1727. [Google Scholar] [CrossRef]
- Cheaito, K.A.; Bahmad, H.F.; Hadadeh, O.; Saleh, E.; Dagher, C.; Hammoud, M.S.; Shahait, M.; Mrad, Z.A.; Nassif, S.; Tawil, A.; et al. EMT Markers in Locally-Advanced Prostate Cancer: Predicting Recurrence? Front. Oncol. 2019, 9, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Xu, H.; Cheng, C.; Ji, Z.; Zhao, H.; Sheng, Y.; Li, X.; Wang, J.; Shu, Y.; He, Y.; et al. Identification of a Zeb1 expressing basal stem cell subpopulation in the prostate. Nat. Commun. 2020, 11, 706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nolan, K.D.; Franco, O.E.; Hance, M.W.; Hayward, S.W.; Isaacs, J.S. Tumor-secreted Hsp90 subverts polycomb function to drive prostate tumor growth and invasion. J. Biol. Chem. 2015, 290, 8271–8282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Hong, W.; Wei, X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J. Hematol. Oncol. 2022, 15, 129. [Google Scholar] [CrossRef]
- Zhou, H.; He, Q.; Li, C.; Alsharafi, B.L.M.; Deng, L.; Long, Z.; Gan, Y. Focus on the tumor microenvironment: A seedbed for neuroendocrine prostate cancer. Front. Cell Dev. Biol. 2022, 10, 955669. [Google Scholar] [CrossRef]
- Kang, J.; la Manna, F.; Bonollo, F.; Sampson, N.; Alberts, I.L.; Mingels, C.; Afshar-Oromieh, A.; Thalmann, G.N.; Karkampouna, S. Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer. Cancer Lett. 2022, 530, 156–169. [Google Scholar] [CrossRef]
- Comito, G.; Giannoni, E.; Segura, C.P.; Barcellos-de-Souza, P.; Raspollini, M.R.; Baroni, G.; Lanciotti, M.; Serni, S.; Chiarugi, P. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene 2014, 33, 2423–2431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bezzi, M.; Seitzer, N.; Ishikawa, T.; Reschke, M.; Chen, M.; Wang, G.; Mitchell, C.; Ng, C.; Katon, J.; Lunardi, A.; et al. Diverse genetic-driven immune landscapes dictate tumor progression through distinct mechanisms. Nat. Med. 2018, 24, 165–175. [Google Scholar] [CrossRef] [PubMed]
- Su, W.; Han, H.H.; Wang, Y.; Zhang, B.; Zhou, B.; Cheng, Y.; Rumandla, A.; Gurrapu, S.; Chakraborty, G.; Su, J.; et al. The Polycomb Repressor Complex 1 Drives Double-Negative Prostate Cancer Metastasis by Coordinating Stemness and Immune Suppression. Cancer Cell 2019, 36, 139–155.e110. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Gu, M.; Cai, Z.K.; Zhao, H.; Sun, S.C.; Liu, C.; Zhan, M.; Chen, Y.B.; Wang, Z. TGF-β1 promotes epithelial-to-mesenchymal transition and stemness of prostate cancer cells by inducing PCBP1 degradation and alternative splicing of CD44. Cell Mol. Life Sci. 2021, 78, 949–962. [Google Scholar] [CrossRef] [PubMed]
- Bluemn, E.G.; Coleman, I.M.; Lucas, J.M.; Coleman, R.T.; Hernandez-Lopez, S.; Tharakan, R.; Bianchi-Frias, D.; Dumpit, R.F.; Kaipainen, A.; Corella, A.N.; et al. Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. Cancer Cell 2017, 32, 474.e476–489.e476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gregory, C.W.; Whang, Y.E.; McCall, W.; Fei, X.; Liu, Y.; Ponguta, L.A.; French, F.S.; Wilson, E.M.; Earp, H.S., 3rd. Heregulin-induced activation of HER2 and HER3 increases androgen receptor transactivation and CWR-R1 human recurrent prostate cancer cell growth. Clin. Cancer Res. 2005, 11, 1704–1712. [Google Scholar] [CrossRef] [Green Version]
- Giannoni, E.; Bianchini, F.; Masieri, L.; Serni, S.; Torre, E.; Calorini, L.; Chiarugi, P. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010, 70, 6945–6956. [Google Scholar] [CrossRef] [Green Version]
- Acevedo, V.D.; Gangula, R.D.; Freeman, K.W.; Li, R.; Zhang, Y.; Wang, F.; Ayala, G.E.; Peterson, L.E.; Ittmann, M.; Spencer, D.M. Inducible FGFR-1 activation leads to irreversible prostate adenocarcinoma and an epithelial-to-mesenchymal transition. Cancer Cell 2007, 12, 559–571. [Google Scholar] [CrossRef] [Green Version]
- Nath, D.; Li, X.; Mondragon, C.; Post, D.; Chen, M.; White, J.R.; Hryniewicz-Jankowska, A.; Caza, T.; Kuznetsov, V.A.; Hehnly, H.; et al. Abi1 loss drives prostate tumorigenesis through activation of EMT and non-canonical WNT signaling. Cell Commun. Signal 2019, 17, 120. [Google Scholar] [CrossRef] [Green Version]
- Qi, Y.; Liu, J.; Chao, J.; Scheuerman, M.P.; Rahimi, S.A.; Lee, L.Y.; Li, S. PTEN suppresses epithelial-mesenchymal transition and cancer stem cell activity by downregulating Abi1. Sci. Rep. 2020, 10, 12685. [Google Scholar] [CrossRef]
- Shen, Y.; Cao, J.; Liang, Z.; Lin, Q.; Wang, J.; Yang, X.; Zhang, R.; Zong, J.; Du, X.; Peng, Y.; et al. Estrogen receptor α-NOTCH1 axis enhances basal stem-like cells and epithelial-mesenchymal transition phenotypes in prostate cancer. Cell Commun. Signal 2019, 17, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishii, A.; Shigemura, K.; Kitagawa, K.; Sung, S.Y.; Chen, K.C.; Yi-Te, C.; Liu, M.C.; Fujisawa, M. Anti-tumor Effect of Hedgehog Signaling Inhibitor, Vismodegib, on Castration-resistant Prostate Cancer. Anticancer Res. 2020, 40, 5107–5114. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Ma, C.; Li, F.; Nie, G.; Zhang, H. The Role of Contactin 1 in Cancers: What We Know So Far. Front. Oncol. 2020, 10, 574208. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; Zhang, Y.; Li, C.; Xu, P.; Gao, Y.; Xu, Y. CNTN-1 promotes docetaxel resistance and epithelial-to-mesenchymal transition via the PI3K/Akt signaling pathway in prostate cancer. Arch. Med. Sci. 2021, 17, 152–165. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.I.; Hamid, A.; Adhami, V.M.; Lall, R.K.; Mukhtar, H. Role of epithelial mesenchymal transition in prostate tumorigenesis. Curr. Pharm. Des. 2015, 21, 1240–1248. [Google Scholar] [CrossRef] [Green Version]
- Xin, L.; Lawson, D.A.; Witte, O.N. The Sca-1 cell surface marker enriches for a prostate-regenerating cell subpopulation that can initiate prostate tumorigenesis. Proc. Natl. Acad. Sci. USA 2005, 102, 6942–6947. [Google Scholar] [CrossRef] [Green Version]
- Vasaikar, S.V.; Deshmukh, A.P.; den Hollander, P.; Addanki, S.; Kuburich, N.A.; Kudaravalli, S.; Joseph, R.; Chang, J.T.; Soundararajan, R.; Mani, S.A. EMTome: A resource for pan-cancer analysis of epithelial-mesenchymal transition genes and signatures. Br. J. Cancer 2021, 124, 259–269. [Google Scholar] [CrossRef]
- Stylianou, N.; Lehman, M.L.; Wang, C.; Fard, A.T.; Rockstroh, A.; Fazli, L.; Jovanovic, L.; Ward, M.; Sadowski, M.C.; Kashyap, A.S.; et al. A molecular portrait of epithelial-mesenchymal plasticity in prostate cancer associated with clinical outcome. Oncogene 2019, 38, 913–934. [Google Scholar] [CrossRef]
- Cmero, M.; Kurganovs, N.J.; Stuchbery, R.; McCoy, P.; Grima, C.; Ngyuen, A.; Chow, K.; Mangiola, S.; Macintyre, G.; Howard, N.; et al. Loss of SNAI2 in Prostate Cancer Correlates With Clinical Response to Androgen Deprivation Therapy. JCO Precis. Oncol. 2021, 5, 337. [Google Scholar] [CrossRef]
- Jędroszka, D.; Orzechowska, M.; Hamouz, R.; Górniak, K.; Bednarek, A.K. Markers of epithelial-to-mesenchymal transition reflect tumor biology according to patient age and Gleason score in prostate cancer. PLoS ONE 2017, 12, e0188842. [Google Scholar] [CrossRef] [Green Version]
- He, M.X.; Cuoco, M.S.; Crowdis, J.; Bosma-Moody, A.; Zhang, Z.; Bi, K.; Kanodia, A.; Su, M.J.; Ku, S.Y.; Garcia, M.M.; et al. Transcriptional mediators of treatment resistance in lethal prostate cancer. Nat. Med. 2021, 27, 426–433. [Google Scholar] [CrossRef] [PubMed]
Target | NCT Number | Phase | Status | Outcome of Interest |
---|---|---|---|---|
TGF-β | NCT02452008 | 2 | Recruiting | Compare the progression-free survival of those with mCRPC who are treated with enzalutamide alone vs. enzalutamide plus LY2157299, a TGF-β receptor inhibitor. |
PRC2 | NCT05413421 | 1 | Recruiting | Establish a phase 2 dose and/or MTD of ORIC-944, a potent, highly selective, allosteric, orally bioavailable, small molecule inhibitor of PRC2. |
FGF | NCT01741116 | 2 | Completed | Evaluate the efficacy and safety of Dovitinib, an RTK inhibitor with unique inhibitory effects on FGF. No results posted. |
NCT01994590 | 2 | Terminated | Evaluate the safety of adding Dovitinib to abiraterone acetate and prednisone in those with mCRPC. Terminated due to sponsor no longer supplying study drug. | |
NCT02065323 | 2 | Withdrawn | Evaluate if adding Dovitinib to ADT will prolong time to disease progression in those with mCRPC receiving ADT for the first time. Withdrawn due to budgeting considerations and time length to development. | |
EZH2 | NCT03480646 | 1, 2 | Active, not recruiting | Determine the dose-limiting toxicities in those with mCRPC receiving CPI-1205, a small molecule inhibitor of EZH2. |
NCT03460977 | 1 | Recruiting | Evaluate the safety and efficacy of PF-06821497, an EZH2 inhibitor, in those with CRPC. | |
NCT05567679 | 1 | Not yet recruiting | Evaluate if the underlying prostate cancer tumor is more sensitive to the patient’s immune system after receiving radical prostatectomy following preoperative treatment with Tazverik, an EZH2 inhibitor. | |
NCT04179864 | 1, 2 | Recruiting | Determine the safety and efficacy of combining Tazverik with either enzalutamide or abiraterone/prednisone in those with CRPC who have not received chemotherapy. | |
HSP90 | NCT01685268 | 1, 2 | Completed | Determine the safety and antitumor activity of AT13387, an HSP90 inhibitor, either alone or in combination with abiraterone. No results posted. |
NCT01270880 | 2 | Completed | Evaluate the progression-free survival of those receiving STA-9090, an HSP90 inhibitor, with mCRPC refractory to docetaxel. At 6 months, all 18 participants had disease progression. | |
CDH1 | NCT02140996 | 1 | Unknown | Assessment of safety and dose level of Ad-sig-hMUC-1/ecdCD40L vector vaccine meant to disrupt e-cadherin. Last status update in 2016 listed it as recruiting. |
LSD1 | NCT05268666 | 1, 2 | Recruiting | Assessment of the MTD and efficacy of JBI-802, an LSD1/HDAC6 inhibitor. |
IL6 | NCT00433446 | 2 | Completed | Determine PSA response to CNTO-328, an anti-IL6 chimeric monoclonal antibody, in those with mCRPC. 3.8% of participants had a reduction in PSA of at least 50%. |
NCT00385827 | 2 | Terminated | Determine the number of participants with adverse events and the progression-free survival in those receiving CNTO-328 for mCRPC. Terminated after determination of a lack of efficacy. | |
NCT00401765 | 1 | Completed | Determine the safety and efficacy of CNTO-328 in combination with docetaxel in those with mCRPC. No results posted. | |
CCL2 | NCT00992186 | 2 | Completed | Determine the safety and efficacy of Carlumab, an anti-CCL2 therapy, in those with mCRPC. Of 41 participants, 0 had a composite response as measured by: a change in skeletal lesions, extra-skeletal lesions, or PSA. |
N-Cadherin and Vimentin | NCT01990196 | 2 | Active, not recruiting | Measure vimentin and N-cadherin expression following radical prostatectomy and treatment with degarelix, enzalutamide, trametinib, or dasatinib, which are capable of SRC and/or MEK inhibition of tyrosine kinase. |
NCT02204943 | 2 | Completed | Measure changes in biomarkers of epithelial plasticity such as N-cadherin and vimentin following bone targeting radium-223 in those with mCRPC. No results posted. | |
NCT00887640 | 2 | Terminated | Measure percent change in N-cadherin expression at baseline and at 8 weeks following treatment with Temsirolimus in those with mCRPC refractory to treatment. |
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. |
© 2023 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
Gogola, S.; Rejzer, M.; Bahmad, H.F.; Abou-Kheir, W.; Omarzai, Y.; Poppiti, R. Epithelial-to-Mesenchymal Transition-Related Markers in Prostate Cancer: From Bench to Bedside. Cancers 2023, 15, 2309. https://doi.org/10.3390/cancers15082309
Gogola S, Rejzer M, Bahmad HF, Abou-Kheir W, Omarzai Y, Poppiti R. Epithelial-to-Mesenchymal Transition-Related Markers in Prostate Cancer: From Bench to Bedside. Cancers. 2023; 15(8):2309. https://doi.org/10.3390/cancers15082309
Chicago/Turabian StyleGogola, Samantha, Michael Rejzer, Hisham F. Bahmad, Wassim Abou-Kheir, Yumna Omarzai, and Robert Poppiti. 2023. "Epithelial-to-Mesenchymal Transition-Related Markers in Prostate Cancer: From Bench to Bedside" Cancers 15, no. 8: 2309. https://doi.org/10.3390/cancers15082309
APA StyleGogola, S., Rejzer, M., Bahmad, H. F., Abou-Kheir, W., Omarzai, Y., & Poppiti, R. (2023). Epithelial-to-Mesenchymal Transition-Related Markers in Prostate Cancer: From Bench to Bedside. Cancers, 15(8), 2309. https://doi.org/10.3390/cancers15082309