Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma
Abstract
:Simple Summary
Abstract
1. Introduction
2. The Hippo Signaling Pathway: Critical Components in Mammals and Basic Biology
3. Deregulation of the Hippo Signaling Pathway in Bone and Soft Tissue Sarcoma
3.1. Osteosarcomas
3.1.1. YAP
3.1.2. NF2
3.1.3. LATS1/2
3.1.4. RASSF
3.1.5. TAZ
3.2. Ewing Sarcoma
3.3. Epithelial Hemangioendothelioma
3.4. Myxoid Liposarcoma
3.5. Sclerosing Epithelioid Fibrosarcoma and Low-Grade Fibromyxoid Sarcoma
3.6. Rhabdomyosarcoma
3.6.1. Alveolar RMS
3.6.2. Spindle Cell/Sclerosing RMS
3.7. Synovial Sarcoma
3.8. Osteoblastoma
3.9. Undifferentiated Pleomorphic Sarcoma
3.10. Chondrosarcoma
3.11. Ossifying Fibromyxoid Tumor
4. Targeting the Hippo Pathway as a Therapeutic Approach for Sarcomas
4.1. Inhibition of YAP-TEAD Interaction: Verteporfin
4.2. YAP/TAZ Cytoplasmic Retention: Dasatinib, Statins, Pazopanib, and Metformin
4.3. Inhibition of TEAD-Transcription Activity
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Pan, D. The Hippo Signaling Pathway in Development and Cancer. Dev. Cell 2010, 19, 491–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, F.-X.; Zhao, B.; Guan, K.-L. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Justice, R.W.; Zilian, O.; Woods, D.F.; Noll, M.; Bryant, P.J. The Drosophila tumor suppressor gene warts encodes a homolog of human myotonic dystrophy kinase and is required for the control of cell shape and proliferation. Genes Dev. 1995, 9, 534–546. [Google Scholar] [CrossRef] [Green Version]
- Xu, T.; Wang, W.; Zhang, S.; Stewart, R.A.; Yu, W. Identifying tumor suppressors in genetic mosaics: The Drosophila lats gene encodes a putative protein kinase. Development 1995, 121, 1053–1063. [Google Scholar] [CrossRef] [PubMed]
- Udan, R.S.; Kango-Singh, M.; Nolo, R.; Tao, C.; Halder, G. Hippo promotes proliferation arrest and apoptosis in the Salvador/Warts pathway. Nat. Cell Biol. 2003, 5, 914–920. [Google Scholar] [CrossRef]
- Wu, S.; Huang, J.; Dong, J.; Pan, D. hippo encodes a Ste-20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts. Cell 2003, 114, 445–456. [Google Scholar] [CrossRef] [Green Version]
- Zhou, D.; Conrad, C.; Xia, F.; Park, J.S.; Payer, B.; Yin, Y.; Lauwers, G.Y.; Thasler, W.; Lee, J.T.; Avruch, J.; et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 2009, 16, 425–438. [Google Scholar] [CrossRef] [Green Version]
- Camargo, F.D.; Gokhale, S.; Johnnidis, J.B.; Fu, D.; Bell, G.W.; Jaenisch, R.; Brummelkamp, T.R. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. CB 2007, 17, 2054–2060. [Google Scholar] [CrossRef] [Green Version]
- Dong, J.; Feldmann, G.; Huang, J.; Wu, S.; Zhang, N.; Comerford, S.A.; Gayyed, M.F.; Anders, R.A.; Maitra, A.; Pan, D. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 2007, 130, 1120–1133. [Google Scholar] [CrossRef] [Green Version]
- Cordenonsi, M.; Zanconato, F.; Azzolin, L.; Forcato, M.; Rosato, A.; Frasson, C.; Inui, M.; Montagner, M.; Parenti, A.R.; Poletti, A.; et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 2011, 147, 759–772. [Google Scholar] [CrossRef]
- Harvey, K.F.; Zhang, X.; Thomas, D.M. The Hippo pathway and human cancer. Nat. Rev. Cancer 2013, 13, 246–257. [Google Scholar] [CrossRef] [PubMed]
- Jiao, S.; Wang, H.; Shi, Z.; Dong, A.; Zhang, W.; Song, X.; He, F.; Wang, Y.; Zhang, Z.; Wang, W.; et al. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 2014, 25, 166–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lau, A.N.; Curtis, S.J.; Fillmore, C.M.; Rowbotham, S.P.; Mohseni, M.; Wagner, D.E.; Beede, A.M.; Montoro, D.T.; Sinkevicius, K.W.; Walton, Z.E.; et al. Tumor-propagating cells and Yap/Taz activity contribute to lung tumor progression and metastasis. EMBO J. 2014, 33, 468–481. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, L.T.; Tretiakova, M.S.; Silvis, M.R.; Lucas, J.; Klezovitch, O.; Coleman, I.; Bolouri, H.; Kutyavin, V.I.; Morrissey, C.; True, L.D.; et al. ERG Activates the YAP1 Transcriptional Program and Induces the Development of Age-Related Prostate Tumors. Cancer Cell 2015, 27, 797–808. [Google Scholar] [CrossRef] [Green Version]
- Yimlamai, D.; Christodoulou, C.; Galli, G.G.; Yanger, K.; Pepe-Mooney, B.; Gurung, B.; Shrestha, K.; Cahan, P.; Stanger, B.Z.; Camargo, F.D. Hippo pathway activity influences liver cell fate. Cell 2014, 157, 1324–1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Xu, X.; Maglic, D.; Dill, M.T.; Mojumdar, K.; Ng, P.K.; Jeong, K.J.; Tsang, Y.H.; Moreno, D.; Bhavana, V.H.; et al. Comprehensive Molecular Characterization of the Hippo Signaling Pathway in Cancer. Cell Rep. 2018, 25, 1304–1317.e1305. [Google Scholar] [CrossRef] [Green Version]
- Poma, A.M.; Torregrossa, L.; Bruno, R.; Basolo, F.; Fontanini, G. Hippo pathway affects survival of cancer patients: Extensive analysis of TCGA data and review of literature. Sci. Rep. 2018, 8, 10623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harvey, K.; Tapon, N. The Salvador-Warts-Hippo pathway—An emerging tumour-suppressor network. Nat. Rev. Cancer 2007, 7, 182–191. [Google Scholar] [CrossRef] [PubMed]
- Kim, N.G.; Koh, E.; Chen, X.; Gumbiner, B.M. E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components. Proc. Natl. Acad. Sci. USA 2011, 108, 11930–11935. [Google Scholar] [CrossRef] [Green Version]
- Ma, L.; Cui, J.; Xi, H.; Bian, S.; Wei, B.; Chen, L. Fat4 suppression induces Yap translocation accounting for the promoted proliferation and migration of gastric cancer cells. Cancer Biol. Ther. 2016, 17, 36–47. [Google Scholar] [CrossRef]
- Kim, M.; Jho, E.H. Cross-talk between Wnt/β-catenin and Hippo signaling pathways: A brief review. BMB Rep. 2014, 47, 540–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azzolin, L.; Zanconato, F.; Bresolin, S.; Forcato, M.; Basso, G.; Bicciato, S.; Cordenonsi, M.; Piccolo, S. Role of TAZ as mediator of Wnt signaling. Cell 2012, 151, 1443–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azzolin, L.; Panciera, T.; Soligo, S.; Enzo, E.; Bicciato, S.; Dupont, S.; Bresolin, S.; Frasson, C.; Basso, G.; Guzzardo, V.; et al. YAP/TAZ incorporation in the β-catenin destruction complex orchestrates the Wnt response. Cell 2014, 158, 157–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varelas, X.; Samavarchi-Tehrani, P.; Narimatsu, M.; Weiss, A.; Cockburn, K.; Larsen, B.G.; Rossant, J.; Wrana, J.L. The Crumbs complex couples cell density sensing to Hippo-dependent control of the TGF-β-SMAD pathway. Dev. Cell 2010, 19, 831–844. [Google Scholar] [CrossRef] [PubMed]
- Das Thakur, M.; Feng, Y.; Jagannathan, R.; Seppa, M.J.; Skeath, J.B.; Longmore, G.D. Ajuba LIM proteins are negative regulators of the Hippo signaling pathway. Curr. Biol. CB 2010, 20, 657–662. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Xia, H.; Ge, X.; Chen, Q.; Yuan, D.; Chen, Q.; Leng, W.; Chen, L.; Tang, Q.; Bi, F. CD44 acts through RhoA to regulate YAP signaling. Cell. Signal. 2014, 26, 2504–2513. [Google Scholar] [CrossRef]
- Yu, F.X.; Zhao, B.; Panupinthu, N.; Jewell, J.L.; Lian, I.; Wang, L.H.; Zhao, J.; Yuan, H.; Tumaneng, K.; Li, H.; et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 2012, 150, 780–791. [Google Scholar] [CrossRef] [Green Version]
- Yin, F.; Yu, J.; Zheng, Y.; Chen, Q.; Zhang, N.; Pan, D. Spatial organization of Hippo signaling at the plasma membrane mediated by the tumor suppressor Merlin/NF2. Cell 2013, 154, 1342–1355. [Google Scholar] [CrossRef] [Green Version]
- Wilson, K.E.; Li, Y.W.; Yang, N.; Shen, H.; Orillion, A.R.; Zhang, J. PTPN14 forms a complex with Kibra and LATS1 proteins and negatively regulates the YAP oncogenic function. J. Biol. Chem. 2014, 289, 23693–23700. [Google Scholar] [CrossRef] [Green Version]
- Sorrentino, G.; Ruggeri, N.; Specchia, V.; Cordenonsi, M.; Mano, M.; Dupont, S.; Manfrin, A.; Ingallina, E.; Sommaggio, R.; Piazza, S.; et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 2014, 16, 357–366. [Google Scholar] [CrossRef]
- Fang, C.Y.; Lai, T.C.; Hsiao, M.; Chang, Y.C. The Diverse Roles of TAO Kinases in Health and Diseases. Int. J. Mol. Sci. 2020, 21, 7463. [Google Scholar] [CrossRef] [PubMed]
- Zhao, B.; Li, L.; Lu, Q.; Wang, L.H.; Liu, C.Y.; Lei, Q.; Guan, K.L. Angiomotin is a novel Hippo pathway component that inhibits YAP oncoprotein. Genes Dev. 2011, 25, 51–63. [Google Scholar] [CrossRef] [Green Version]
- Boggiano, J.C.; Vanderzalm, P.J.; Fehon, R.G. Tao-1 Phosphorylates Hippo/MST Kinases to Regulate the Hippo-Salvador-Warts Tumor Suppressor Pathway. Dev. Cell 2011, 21, 888–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Praskova, M.; Khoklatchev, A.; Ortiz-Vega, S.; Avruch, J. Regulation of the MST1 kinase by autophosphorylation, by the growth inhibitory proteins, RASSF1 and NORE1, and by Ras. Biochem. J. 2004, 381, 453–462. [Google Scholar] [CrossRef] [PubMed]
- Mo, J.S.; Park, H.W.; Guan, K.L. The Hippo signaling pathway in stem cell biology and cancer. EMBO Rep. 2014, 15, 642–656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Plouffe, S.W.; Meng, Z.; Lin, K.C.; Lin, B.; Hong, A.W.; Chun, J.V.; Guan, K.L. Characterization of Hippo Pathway Components by Gene Inactivation. Mol. Cell 2016, 64, 993–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, G.; Irvine, K.D. Ajuba family proteins link JNK to Hippo signaling. Sci. Signal. 2013, 6, ra81. [Google Scholar] [CrossRef] [Green Version]
- Hergovich, A. MOB control: Reviewing a conserved family of kinase regulators. Cell. Signal. 2011, 23, 1433–1440. [Google Scholar] [CrossRef] [Green Version]
- Morice, S.; Danieau, G.; Rédini, F.; Brounais-Le-Royer, B.; Verrecchia, F. Hippo/YAP Signaling Pathway: A Promising Therapeutic Target in Bone Paediatric Cancers? Cancers 2020, 12, 645. [Google Scholar] [CrossRef] [Green Version]
- Muslin, A.J.; Xing, H. 14-3-3 proteins: Regulation of subcellular localization by molecular interference. Cell. Signal. 2000, 12, 703–709. [Google Scholar] [CrossRef]
- Basu, S.; Totty, N.F.; Irwin, M.S.; Sudol, M.; Downward, J. Akt Phosphorylates the Yes-Associated Protein, YAP, to Induce Interaction with 14-3-3 and Attenuation of p73-Mediated Apoptosis. Mol. Cell 2003, 11, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.K.; Jang, J.W.; Bae, S.C. DNA binding partners of YAP/TAZ. BMB Rep. 2018, 51, 126–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vassilev, A.; Kaneko, K.J.; Shu, H.; Zhao, Y.; DePamphilis, M.L. TEAD/TEF transcription factors utilize the activation domain of YAP65, a Src/Yes-associated protein localized in the cytoplasm. Genes Dev. 2001, 15, 1229–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, B.; Ye, X.; Yu, J.; Li, L.; Li, W.; Li, S.; Yu, J.; Lin, J.D.; Wang, C.Y.; Chinnaiyan, A.M.; et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev. 2008, 22, 1962–1971. [Google Scholar] [CrossRef] [Green Version]
- Ferrigno, O.; Lallemand, F.; Verrecchia, F.; L’Hoste, S.; Camonis, J.; Atfi, A.; Mauviel, A. Yes-associated protein (YAP65) interacts with Smad7 and potentiates its inhibitory activity against TGF-beta/Smad signaling. Oncogene 2002, 21, 4879–4884. [Google Scholar] [CrossRef] [Green Version]
- Varelas, X.; Sakuma, R.; Samavarchi-Tehrani, P.; Peerani, R.; Rao, B.M.; Dembowy, J.; Yaffe, M.B.; Zandstra, P.W.; Wrana, J.L. TAZ controls Smad nucleocytoplasmic shuttling and regulates human embryonic stem-cell self-renewal. Nat. Cell Biol. 2008, 10, 837–848. [Google Scholar] [CrossRef]
- Komuro, A.; Nagai, M.; Navin, N.E.; Sudol, M. WW domain-containing protein YAP associates with ErbB-4 and acts as a co-transcriptional activator for the carboxyl-terminal fragment of ErbB-4 that translocates to the nucleus. J. Biol. Chem. 2003, 278, 33334–33341. [Google Scholar] [CrossRef] [Green Version]
- Rosenbluh, J.; Nijhawan, D.; Cox, A.G.; Li, X.; Neal, J.T.; Schafer, E.J.; Zack, T.I.; Wang, X.; Tsherniak, A.; Schinzel, A.C.; et al. β-Catenin-Driven Cancers Require a YAP1 Transcriptional Complex for Survival and Tumorigenesis. Cell 2012, 151, 1457–1473. [Google Scholar] [CrossRef] [Green Version]
- Murakami, M.; Nakagawa, M.; Olson, E.N.; Nakagawa, O. A WW domain protein TAZ is a critical coactivator for TBX5, a transcription factor implicated in Holt-Oram syndrome. Proc. Natl. Acad. Sci. USA 2005, 102, 18034–18039. [Google Scholar] [CrossRef] [Green Version]
- Qiao, Y.; Lin, S.J.; Chen, Y.; Voon, D.C.; Zhu, F.; Chuang, L.S.; Wang, T.; Tan, P.; Lee, S.C.; Yeoh, K.G.; et al. RUNX3 is a novel negative regulator of oncogenic TEAD-YAP complex in gastric cancer. Oncogene 2016, 35, 2664–2674. [Google Scholar] [CrossRef]
- Chuang, L.S.H.; Ito, Y. The Multiple Interactions of RUNX with the Hippo-YAP Pathway. Cells 2021, 10, 2925. [Google Scholar] [CrossRef] [PubMed]
- Zagurovskaya, M.; Shareef, M.M.; Das, A.; Reeves, A.; Gupta, S.; Sudol, M.; Bedford, M.T.; Prichard, J.; Mohiuddin, M.; Ahmed, M.M. EGR-1 forms a complex with YAP-1 and upregulates Bax expression in irradiated prostate carcinoma cells. Oncogene 2009, 28, 1121–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Li, Y.; Ma, Y.; Yang, L.; Wang, T.; Meng, X.; Zong, Z.; Sun, X.; Hua, X.; Li, H. Yes-associated protein (YAP) binds to HIF-1α and sustains HIF-1α protein stability to promote hepatocellular carcinoma cell glycolysis under hypoxic stress. J. Exp. Clin. Cancer Res. CR 2018, 37, 216. [Google Scholar] [CrossRef] [PubMed]
- Yagi, R.; Chen, L.F.; Shigesada, K.; Murakami, Y.; Ito, Y. A WW domain-containing yes-associated protein (YAP) is a novel transcriptional co-activator. EMBO J. 1999, 18, 2551–2562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strano, S.; Munarriz, E.; Rossi, M.; Castagnoli, L.; Shaul, Y.; Sacchi, A.; Oren, M.; Sudol, M.; Cesareni, G.; Blandino, G. Physical interaction with Yes-associated protein enhances p73 transcriptional activity. J. Biol. Chem. 2001, 276, 15164–15173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Du, S.; Lei, T.; Wang, H.; He, X.; Tong, R.; Wang, Y. Multifaceted regulation and functions of YAP/TAZ in tumors (Review). Oncol. Rep. 2018, 40, 16–28. [Google Scholar] [CrossRef]
- Zanconato, F.; Battilana, G.; Cordenonsi, M.; Piccolo, S. YAP/TAZ as therapeutic targets in cancer. Curr. Opin. Pharmacol. 2016, 29, 26–33. [Google Scholar] [CrossRef]
- Panciera, T.; Azzolin, L.; Cordenonsi, M.; Piccolo, S. Mechanobiology of YAP and TAZ in physiology and disease. Nat. Rev. Mol. Cell Biol. 2017, 18, 758–770. [Google Scholar] [CrossRef]
- Lamar, J.M.; Xiao, Y.; Norton, E.; Jiang, Z.G.; Gerhard, G.M.; Kooner, S.; Warren, J.S.A.; Hynes, R.O. SRC tyrosine kinase activates the YAP/TAZ axis and thereby drives tumor growth and metastasis. J. Biol. Chem. 2019, 294, 2302–2317. [Google Scholar] [CrossRef] [Green Version]
- Warren, J.S.A.; Xiao, Y.; Lamar, J.M. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers 2018, 10, 115. [Google Scholar] [CrossRef]
- WHO Classification of Tumours Editorial Board. Soft Tissue and Bone Tumours, 5th ed.; WHO Classification of Tumours Series; International Agency for Research on Cancer: Lyon, France, 2020; Volume 3, Available online: https://publications.iarc.fr/588 (accessed on 14 November 2022).
- Szulzewsky, F.; Holland, E.C.; Vasioukhin, V. YAP1 and its fusion proteins in cancer initiation, progression and therapeutic resistance. Dev. Biol. 2021, 475, 205–221. [Google Scholar] [CrossRef] [PubMed]
- Errani, C.; Zhang, L.; Sung, Y.S.; Hajdu, M.; Singer, S.; Maki, R.G.; Healey, J.H.; Antonescu, C.R. A novel WWTR1-CAMTA1 gene fusion is a consistent abnormality in epithelioid hemangioendothelioma of different anatomic sites. Genes Chromosom. Cancer 2011, 50, 644–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonescu, C.R.; Le Loarer, F.; Mosquera, J.M.; Sboner, A.; Zhang, L.; Chen, C.L.; Chen, H.W.; Pathan, N.; Krausz, T.; Dickson, B.C.; et al. Novel YAP1-TFE3 fusion defines a distinct subset of epithelioid hemangioendothelioma. Genes Chromosom. Cancer 2013, 52, 775–784. [Google Scholar] [CrossRef] [Green Version]
- Tremblay, A.M.; Missiaglia, E.; Galli, G.G.; Hettmer, S.; Urcia, R.; Carrara, M.; Judson, R.N.; Thway, K.; Nadal, G.; Selfe, J.L.; et al. The Hippo Transducer YAP1 Transforms Activated Satellite Cells and Is a Potent Effector of Embryonal Rhabdomyosarcoma Formation. Cancer Cell 2014, 26, 273–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seidel, C.; Schagdarsurengin, U.; Blümke, K.; Würl, P.; Pfeifer, G.P.; Hauptmann, S.; Taubert, H.; Dammann, R. Frequent hypermethylation of MST1 and MST2 in soft tissue sarcoma. Mol. Carcinog. 2007, 46, 865–871. [Google Scholar] [CrossRef] [PubMed]
- Fullenkamp, C.A.; Hall, S.L.; Jaber, O.I.; Pakalniskis, B.L.; Savage, E.C.; Savage, J.M.; Ofori-Amanfo, G.K.; Lambertz, A.M.; Ivins, S.D.; Stipp, C.S.; et al. TAZ and YAP are frequently activated oncoproteins in sarcomas. Oncotarget 2016, 7, 30094–30108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isfort, I.; Elges, S.; Cyra, M.; Berthold, R.; Renner, M.; Mechtersheimer, G.; Aman, P.; Larsson, O.; Ratner, N.; Hafner, S.; et al. Prevalence of the Hippo Effectors YAP1/TAZ in Tumors of Soft Tissue and Bone. Sci. Rep. 2019, 9, 19704. [Google Scholar] [CrossRef] [Green Version]
- Rodríguez-Núñez, P.; Romero-Pérez, L.; Amaral, A.T.; Puerto-Camacho, P.; Jordán, C.; Marcilla, D.; Grünewald, T.G.; Alonso, J.; de Alava, E.; Díaz-Martín, J. Hippo pathway effectors YAP1/TAZ induce an EWS-FLI1-opposing gene signature and associate with disease progression in Ewing sarcoma. J. Pathol. 2020, 250, 374–386. [Google Scholar] [CrossRef] [Green Version]
- Bouvier, C.; Macagno, N.; Nguyen, Q.; Loundou, A.; Jiguet-Jiglaire, C.; Gentet, J.C.; Jouve, J.L.; Rochwerger, A.; Mattei, J.C.; Bouvard, D.; et al. Prognostic value of the Hippo pathway transcriptional coactivators YAP/TAZ and β1-integrin in conventional osteosarcoma. Oncotarget 2016, 7, 64702–64710. [Google Scholar] [CrossRef] [Green Version]
- Desai, C.; Thomason, J.; Kohlmeyer, J.L.; Reisetter, A.C.; Ahirwar, P.; Jahanseir, K.; Leidinger, M.; Ofori-Amanfo, G.; Fritchie, K.; Velu, S.E.; et al. Prognostic and therapeutic value of the Hippo pathway, RABL6A, and p53-MDM2 axes in sarcomas. Oncotarget 2021, 12, 740–755. [Google Scholar] [CrossRef]
- Chan, L.H.; Wang, W.; Yeung, W.; Deng, Y.; Yuan, P.; Mak, K.K. Hedgehog signaling induces osteosarcoma development through Yap1 and H19 overexpression. Oncogene 2014, 33, 4857–4866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verma, N.K.; Gadi, A.; Maurizi, G.; Roy, U.B.; Mansukhani, A.; Basilico, C. Myeloid Zinc Finger 1 and GA Binding Protein Co-Operate with Sox2 in Regulating the Expression of Yes-Associated Protein 1 in Cancer Cells. Stem Cells 2017, 35, 2340–2350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.; Chen, C.; Xu, R.; Li, Y.; Hu, R.; Li, Z.; Zhu, X. Knockdown of HuR represses osteosarcoma cells migration, invasion and stemness through inhibition of YAP activation and increases susceptibility to chemotherapeutic agents. Biomed. Pharmacother. 2018, 102, 587–593. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Wang, Y.; Hu, R.; Xu, R.; Xu, W. LncRNA B4GALT1-AS1 recruits HuR to promote osteosarcoma cells stemness and migration via enhancing YAP transcriptional activity. Cell Prolif. 2018, 51, e12504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, G.; Huang, K.; Jie, Z.; Wu, Y.; Chen, J.; Chen, Z.; Fang, X.; Shen, S. CircFAT1 sponges miR-375 to promote the expression of Yes-associated protein 1 in osteosarcoma cells. Mol. Cancer 2018, 17, 170. [Google Scholar] [CrossRef]
- Luo, Z.; Wu, G.; Zhang, D.; Liu, J.; Ran, R. microRNA-625 targets Yes-associated protein 1 to suppress cell proliferation and invasion of osteosarcoma. Mol. Med. Rep. 2018, 17, 2005–2011. [Google Scholar] [CrossRef] [Green Version]
- Cheng, L.; Wu, B.; Zhang, L.; Bian, E.; An, R.; Yu, S.; Liu, W.; Xiong, Z. Gankyrin promotes osteosarcoma tumorigenesis by forming a positive feedback loop with YAP. Cell. Signal. 2020, 65, 109460. [Google Scholar] [CrossRef]
- Yi, X.; Deng, X.; Zhao, Y.; Deng, B.; Deng, J.; Fan, H.; Du, Y.; Hao, L. Ubiquitin-like protein FAT10 promotes osteosarcoma growth by modifying the ubiquitination and degradation of YAP1. Exp. Cell Res. 2020, 387, 111804. [Google Scholar] [CrossRef]
- Zucchini, C.; Manara, M.C.; Cristalli, C.; Carrabotta, M.; Greco, S.; Pinca, R.S.; Ferrari, C.; Landuzzi, L.; Pasello, M.; Lollini, P.L.; et al. ROCK2 deprivation leads to the inhibition of tumor growth and metastatic potential in osteosarcoma cells through the modulation of YAP activity. J. Exp. Clin. Cancer Res. CR 2019, 38, 503. [Google Scholar] [CrossRef] [Green Version]
- Deng, X.; Yi, X.; Deng, J.; Zou, Y.; Wang, S.; Shan, W.; Liu, P.; Zhang, Z.; Chen, L.; Hao, L. ROCK2 promotes osteosarcoma growth and metastasis by modifying PFKFB3 ubiquitination and degradation. Exp. Cell Res. 2019, 385, 111689. [Google Scholar] [CrossRef]
- Deng, X.; Yi, X.; Huang, D.; Liu, P.; Chen, L.; Du, Y.; Hao, L. ROCK2 mediates osteosarcoma progression and TRAIL resistance by modulating O-GlcNAc transferase degradation. Am. J. Cancer Res. 2020, 10, 781–798. [Google Scholar] [PubMed]
- McClatchey, A.I.; Saotome, I.; Mercer, K.; Crowley, D.; Gusella, J.F.; Bronson, R.T.; Jacks, T. Mice heterozygous for a mutation at the Nf2 tumor suppressor locus develop a range of highly metastatic tumors. Genes Dev. 1998, 12, 1121–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giovannini, M.; Robanus-Maandag, E.; van der Valk, M.; Niwa-Kawakita, M.; Abramowski, V.; Goutebroze, L.; Woodruff, J.M.; Berns, A.; Thomas, G. Conditional biallelic Nf2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2. Genes Dev. 2000, 14, 1617–1630. [Google Scholar] [CrossRef] [PubMed]
- Morrison, H.; Sherman, L.S.; Legg, J.; Banine, F.; Isacke, C.; Haipek, C.A.; Gutmann, D.H.; Ponta, H.; Herrlich, P. The NF2 tumor suppressor gene product, merlin, mediates contact inhibition of growth through interactions with CD44. Genes Dev. 2001, 15, 968–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, S.; Cai, X.; Wu, C.; Wu, L.; Wang, Y.; Liu, Y.; Yu, Z.; Qin, S.; Ma, F.; Thiery, J.P.; et al. Adhesion glycoprotein CD44 functions as an upstream regulator of a network connecting ERK, AKT and Hippo-YAP pathways in cancer progression. Oncotarget 2015, 6, 2951–2965. [Google Scholar] [CrossRef] [Green Version]
- Gvozdenovic, A.; Arlt, M.J.; Campanile, C.; Brennecke, P.; Husmann, K.; Born, W.; Muff, R.; Fuchs, B. Silencing of CD44 gene expression in human 143-B osteosarcoma cells promotes metastasis of intratibial tumors in SCID mice. PLoS ONE 2013, 8, e60329. [Google Scholar] [CrossRef] [Green Version]
- Ma, J.; Klemm, J.; Gerardo-Ramírez, M.; Frappart, L.; Castven, D.; Becker, D.; Zoch, A.; Parent, R.; Bartosch, B.; Minnich, K.; et al. Cluster of differentiation 44 promotes osteosarcoma progression in mice lacking the tumor suppressor Merlin. Int. J. Cancer 2020, 147, 2564–2577. [Google Scholar] [CrossRef]
- Basu-Roy, U.; Bayin, N.S.; Rattanakorn, K.; Han, E.; Placantonakis, D.G.; Mansukhani, A.; Basilico, C. Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells. Nat. Commun. 2015, 6, 6411. [Google Scholar] [CrossRef] [Green Version]
- Maurizi, G.; Verma, N.; Gadi, A.; Mansukhani, A.; Basilico, C. Sox2 is required for tumor development and cancer cell proliferation in osteosarcoma. Oncogene 2018, 37, 4626–4632. [Google Scholar] [CrossRef]
- Zhou, Y.; Jin, Q.; Xiao, W.; Sun, C. Tankyrase1 antisense oligodeoxynucleotides suppress the proliferation, migration and invasion through Hippo/YAP pathway in human osteosarcoma cells. Pathol. Res. Pract. 2019, 215, 152381. [Google Scholar] [CrossRef]
- Su, X.; Teng, J.; Jin, G.; Li, J.; Zhao, Z.; Cao, X.; Guo, Y.; Guo, M.; Li, X.; Wu, J.; et al. ELK1-induced upregulation of long non-coding RNA MIR100HG predicts poor prognosis and promotes the progression of osteosarcoma by epigenetically silencing LATS1 and LATS2. Biomed. Pharmacother. 2019, 109, 788–797. [Google Scholar] [CrossRef] [PubMed]
- Gong, M.; Liu, H.; Sun, N.; Xie, Y.; Yan, F.; Cai, L. Polyethylenimine-dextran-coated magnetic nanoparticles loaded with miR-302b suppress osteosarcoma in vitro and in vivo. Nanomedicine 2020, 15, 711–723. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.; Xiang, H.; Cong, W.; Yang, H.; Zhang, G.; Wang, Y.; Guo, Z.; Shen, Y.; Chen, B. PLOD1, a target of miR-34c, contributes to cell growth and metastasis via repressing LATS1 phosphorylation and inactivating Hippo pathway in osteosarcoma. Biochem. Biophys. Res. Commun. 2020, 527, 29–36. [Google Scholar] [CrossRef]
- Zhang, M.; Wang, D.; Zhu, T.; Yin, R. RASSF4 Overexpression Inhibits the Proliferation, Invasion, EMT, and Wnt Signaling Pathway in Osteosarcoma Cells. Oncol. Res. 2017, 25, 83–91. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.H.; Yang, C.Q.; Zhang, C.L.; Gao, Y.; Yuan, H.B.; Wang, C. RASSF5 inhibits growth and invasion and induces apoptosis in osteosarcoma cells through activation of MST1/LATS1 signaling. Oncol. Rep. 2014, 32, 1505–1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Katschnig, A.M.; Kauer, M.O.; Schwentner, R.; Tomazou, E.M.; Mutz, C.N.; Linder, M.; Sibilia, M.; Alonso, J.; Aryee, D.N.T.; Kovar, H. EWS-FLI1 perturbs MRTFB/YAP-1/TEAD target gene regulation inhibiting cytoskeletal autoregulatory feedback in Ewing sarcoma. Oncogene 2017, 36, 5995–6005. [Google Scholar] [CrossRef] [Green Version]
- Bierbaumer, L.; Katschnig, A.M.; Radic-Sarikas, B.; Kauer, M.O.; Petro, J.A.; Högler, S.; Gurnhofer, E.; Pedot, G.; Schäfer, B.W.; Schwentner, R.; et al. YAP/TAZ inhibition reduces metastatic potential of Ewing sarcoma cells. Oncogenesis 2021, 10, 2. [Google Scholar] [CrossRef]
- Gharanei, S.; Brini, A.T.; Vaiyapuri, S.; Alholle, A.; Dallol, A.; Arrigoni, E.; Kishida, T.; Hiruma, T.; Avigad, S.; Grimer, R.; et al. RASSF2 methylation is a strong prognostic marker in younger age patients with Ewing sarcoma. Epigenetics 2013, 8, 893–898. [Google Scholar] [CrossRef] [Green Version]
- Avigad, S.; Shukla, S.; Naumov, I.; Cohen, I.J.; Ash, S.; Meller, I.; Kollender, Y.; Issakov, J.; Yaniv, I. Aberrant methylation and reduced expression of RASSF1A in Ewing sarcoma. Pediatr. Blood Cancer 2009, 53, 1023–1028. [Google Scholar] [CrossRef]
- Lamar, J.M.; Motilal Nehru, V.; Weinberg, G. Epithelioid Hemangioendothelioma as a Model of YAP/TAZ-Driven Cancer: Insights from a Rare Fusion Sarcoma. Cancers 2018, 10, 229. [Google Scholar] [CrossRef]
- Rosenbaum, E.; Jadeja, B.; Xu, B.; Zhang, L.; Agaram, N.P.; Travis, W.; Singer, S.; Tap, W.D.; Antonescu, C.R. Prognostic stratification of clinical and molecular epithelioid hemangioendothelioma subsets. Mod. Pathol. 2020, 33, 591–602. [Google Scholar] [CrossRef] [PubMed]
- Trautmann, M.; Cheng, Y.Y.; Jensen, P.; Azoitei, N.; Brunner, I.; Hullein, J.; Slabicki, M.; Isfort, I.; Cyra, M.; Berthold, R.; et al. Requirement for YAP1 signaling in myxoid liposarcoma. EMBO Mol. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
- Berthold, R.; Isfort, I.; Erkut, C.; Heinst, L.; Grünewald, I.; Wardelmann, E.; Kindler, T.; Åman, P.; Grünewald, T.G.P.; Cidre-Aranaz, F.; et al. Fusion protein-driven IGF-IR/PI3K/AKT signals deregulate Hippo pathway promoting oncogenic cooperation of YAP1 and FUS-DDIT3 in myxoid liposarcoma. Oncogenesis 2022, 11, 20. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, A.; Arai, Y.; Tanzawa, Y.; Wakai, S.; Hama, N.; Kawai, A.; Shibata, T. KMT2A (MLL) fusions in aggressive sarcomas in young adults. Histopathology 2019, 75, 508–516. [Google Scholar] [CrossRef] [PubMed]
- Puls, F.; Agaimy, A.; Flucke, U.; Mentzel, T.; Sumathi, V.P.; Ploegmakers, M.; Stoehr, R.; Kindblom, L.G.; Hansson, M.; Sydow, S.; et al. Recurrent Fusions Between YAP1 and KMT2A in Morphologically Distinct Neoplasms Within the Spectrum of Low-grade Fibromyxoid Sarcoma and Sclerosing Epithelioid Fibrosarcoma. Am. J. Surg. Pathol. 2020, 44, 594–606. [Google Scholar] [CrossRef]
- Kao, Y.C.; Lee, J.C.; Zhang, L.; Sung, Y.S.; Swanson, D.; Hsieh, T.H.; Liu, Y.R.; Agaram, N.P.; Huang, H.Y.; Dickson, B.C.; et al. Recurrent YAP1 and KMT2A Gene Rearrangements in a Subset of MUC4-negative Sclerosing Epithelioid Fibrosarcoma. Am. J. Surg. Pathol. 2020, 44, 368–377. [Google Scholar] [CrossRef]
- Massoth, L.R.; Hung, Y.P.; Nardi, V.; Nielsen, G.P.; Hasserjian, R.P.; Louissaint, A., Jr.; Fisch, A.S.; Deshpande, V.; Zukerberg, L.R.; Lennerz, J.K.; et al. Pan-sarcoma genomic analysis of KMT2A rearrangements reveals distinct subtypes defined by YAP1-KMT2A-YAP1 and VIM-KMT2A fusions. Mod. Pathol. 2020, 33, 2307–2317. [Google Scholar] [CrossRef]
- Watson, S.; Perrin, V.; Guillemot, D.; Reynaud, S.; Coindre, J.M.; Karanian, M.; Guinebretiere, J.M.; Freneaux, P.; Le Loarer, F.; Bouvet, M.; et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J. Pathol. 2018, 245, 29–40. [Google Scholar] [CrossRef] [Green Version]
- Muntean, A.G.; Tan, J.; Sitwala, K.; Huang, Y.; Bronstein, J.; Connelly, J.A.; Basrur, V.; Elenitoba-Johnson, K.S.; Hess, J.L. The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis. Cancer Cell 2010, 17, 609–621. [Google Scholar] [CrossRef] [Green Version]
- Harada, K.; Toyooka, S.; Maitra, A.; Maruyama, R.; Toyooka, K.O.; Timmons, C.F.; Tomlinson, G.E.; Mastrangelo, D.; Hay, R.J.; Minna, J.D.; et al. Aberrant promoter methylation and silencing of the RASSF1A gene in pediatric tumors and cell lines. Oncogene 2002, 21, 4345–4349. [Google Scholar] [CrossRef]
- Slemmons, K.K.; Yeung, C.; Baumgart, J.T.; Juarez, J.O.M.; McCalla, A.; Helman, L.J. Targeting Hippo-Dependent and Hippo-Independent YAP1 Signaling for the Treatment of Childhood Rhabdomyosarcoma. Cancer Res. 2020, 80, 3046–3056. [Google Scholar] [CrossRef] [PubMed]
- Alaggio, R.; Zhang, L.; Sung, Y.S.; Huang, S.C.; Chen, C.L.; Bisogno, G.; Zin, A.; Agaram, N.P.; LaQuaglia, M.P.; Wexler, L.H.; et al. A Molecular Study of Pediatric Spindle and Sclerosing Rhabdomyosarcoma: Identification of Novel and Recurrent VGLL2-related Fusions in Infantile Cases. Am. J. Surg. Pathol. 2016, 40, 224–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mosquera, J.M.; Sboner, A.; Zhang, L.; Kitabayashi, N.; Chen, C.L.; Sung, Y.S.; Wexler, L.H.; LaQuaglia, M.P.; Edelman, M.; Sreekantaiah, C.; et al. Recurrent NCOA2 gene rearrangements in congenital/infantile spindle cell rhabdomyosarcoma. Genes Chromosom. Cancer 2013, 52, 538–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whittle, S.; Venkatramani, R.; Schönstein, A.; Pack, S.D.; Alaggio, R.; Vokuhl, C.; Rudzinski, E.R.; Wulf, A.L.; Zin, A.; Gruver, J.R.; et al. Congenital spindle cell rhabdomyosarcoma: An international cooperative analysis. Eur. J. Cancer 2022, 168, 56–64. [Google Scholar] [CrossRef] [PubMed]
- Tan, G.Z.L.; Saminathan, S.N.; Chang, K.T.E.; Odoño, E.G.; Kuick, C.H.; Chen, H.; Lee, V.K.M. A rare case of congenital spindle cell rhabdomyosarcoma with TEAD1-NCOA2 fusion: A subset of spindle cell rhabdomyosarcoma with indolent behavior. Pathol. Int. 2020, 70, 234–236. [Google Scholar] [CrossRef] [PubMed]
- Lin, K.C.; Park, H.W.; Guan, K.L. Regulation of the Hippo Pathway Transcription Factor TEAD. Trends Biochem. Sci. 2017, 42, 862–872. [Google Scholar] [CrossRef]
- Mesrouze, Y.; Aguilar, G.; Bokhovchuk, F.; Martin, T.; Delaunay, C.; Villard, F.; Meyerhofer, M.; Zimmermann, C.; Fontana, P.; Wille, R.; et al. A new perspective on the interaction between the Vg/VGLL1-3 proteins and the TEAD transcription factors. Sci. Rep. 2020, 10, 17442. [Google Scholar] [CrossRef]
- Yamaguchi, N. Multiple Roles of Vestigial-Like Family Members in Tumor Development. Front. Oncol. 2020, 10, 1266. [Google Scholar] [CrossRef]
- Isfort, I.; Cyra, M.; Elges, S.; Kailayangiri, S.; Altvater, B.; Rossig, C.; Steinestel, K.; Grünewald, I.; Huss, S.; Eßeling, E.; et al. SS18-SSX-Dependent YAP/TAZ Signaling in Synovial Sarcoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 3718–3731. [Google Scholar] [CrossRef] [Green Version]
- Saba, K.H.; Cornmark, L.; Hofvander, J.; Magnusson, L.; Nilsson, J.; van den Bos, H.; Spierings, D.C.; Foijer, F.; Staaf, J.; Brosjo, O.; et al. Loss of NF2 defines a genetic subgroup of non-FOS-rearranged osteoblastoma. J. Pathol. Clin. Res. 2020, 6, 231–237. [Google Scholar] [CrossRef]
- Merritt, N.M.; Fullenkamp, C.A.; Hall, S.L.; Qian, Q.; Desai, C.; Thomason, J.; Lambertz, A.M.; Dupuy, A.J.; Darbro, B.; Tanas, M.R. A comprehensive evaluation of Hippo pathway silencing in sarcomas. Oncotarget 2018, 9, 31620–31636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, S.; Liu, Y.; Fuller, A.M.; Katti, R.; Ciotti, G.E.; Chor, S.; Alam, M.Z.; Devalaraja, S.; Lorent, K.; Weber, K.; et al. TGFβ and Hippo Pathways Cooperate to Enhance Sarcomagenesis and Metastasis through the Hyaluronan-Mediated Motility Receptor (HMMR). Mol. Cancer Res. MCR 2020, 18, 560–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, S.; Lawlor, M.A.; Rivera-Reyes, A.; Egolf, S.; Chor, S.; Pak, K.; Ciotti, G.E.; Lee, A.C.; Marino, G.E.; Shah, J.; et al. YAP1-Mediated Suppression of USP31 Enhances NFκB Activity to Promote Sarcomagenesis. Cancer Res. 2018, 78, 2705–2720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, C.; Zhou, H.; Zhang, X.; Liu, Z.; Ma, X. PRMT1 potentiates chondrosarcoma development through activation of YAP activity. Mol. Carcinog. 2019, 58, 2193–2206. [Google Scholar] [CrossRef] [PubMed]
- Kao, Y.C.; Sung, Y.S.; Zhang, L.; Chen, C.L.; Huang, S.C.; Antonescu, C.R. Expanding the molecular signature of ossifying fibromyxoid tumors with two novel gene fusions: CREBBP-BCORL1 and KDM2A-WWTR1. Genes Chromosom. Cancer 2017, 56, 42–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lamplot, J.D.; Denduluri, S.; Qin, J.; Li, R.; Liu, X.; Zhang, H.; Chen, X.; Wang, N.; Pratt, A.; Shui, W.; et al. The Current and Future Therapies for Human Osteosarcoma. Curr. Cancer Ther. Rev. 2013, 9, 55–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mortus, J.R.; Zhang, Y.; Hughes, D.P.M. Developmental Pathways Hijacked by Osteosarcoma. In Current Advances in Osteosarcoma; Kleinerman, M.D.E.S., Ed.; Springer International Publishing: Cham, Switzerland, 2014; pp. 93–118. [Google Scholar]
- Martin, J.W.; Squire, J.A.; Zielenska, M. The genetics of osteosarcoma. Sarcoma 2012, 2012, 627254. [Google Scholar] [CrossRef] [Green Version]
- Sadikovic, B.; Yoshimoto, M.; Chilton-MacNeill, S.; Thorner, P.; Squire, J.A.; Zielenska, M. Identification of interactive networks of gene expression associated with osteosarcoma oncogenesis by integrated molecular profiling. Hum. Mol. Genet. 2009, 18, 1962–1975. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.H.; Li, B.; Shen, L.; Shen, Y.; Chen, X.D. The role and clinical significance of YES-associated protein 1 in human osteosarcoma. Int. J. Immunopathol. Pharmacol. 2013, 26, 157–167. [Google Scholar] [CrossRef] [Green Version]
- Chai, J.; Xu, S.; Guo, F. TEAD1 mediates the oncogenic activities of Hippo-YAP1 signaling in osteosarcoma. Biochem. Biophys. Res. Commun. 2017, 488, 297–302. [Google Scholar] [CrossRef]
- Luu, A.K.; Schott, C.R.; Jones, R.; Poon, A.C.; Golding, B.; Hamed, R.; Deheshi, B.; Mutsaers, A.; Wood, G.A.; Viloria-Petit, A.M. An evaluation of TAZ and YAP crosstalk with TGFβ signalling in canine osteosarcoma suggests involvement of hippo signalling in disease progression. BMC Vet. Res. 2018, 14, 365. [Google Scholar] [CrossRef] [PubMed]
- Luo, Y.; Liu, W.; Tang, P.; Jiang, D.; Gu, C.; Huang, Y.; Gong, F.; Rong, Y.; Qian, D.; Chen, J.; et al. miR-624-5p promoted tumorigenesis and metastasis by suppressing hippo signaling through targeting PTPRB in osteosarcoma cells. J. Exp. Clin. Cancer Res. CR 2019, 38, 488. [Google Scholar] [CrossRef] [Green Version]
- Basu-Roy, U.; Han, E.; Rattanakorn, K.; Gadi, A.; Verma, N.; Maurizi, G.; Gunaratne, P.H.; Coarfa, C.; Kennedy, O.D.; Garabedian, M.J.; et al. PPARγ agonists promote differentiation of cancer stem cells by restraining YAP transcriptional activity. Oncotarget 2016, 7, 60954–60970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, J.; Huang, K.; Ma, Y.; Zhou, M.; Fan, S. The TAZ-miR-224-SMAD4 axis promotes tumorigenesis in osteosarcoma. Cell Death Dis. 2017, 8, e2539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, S.; Huang, K.; Wu, Y.; Ma, Y.; Wang, J.; Qin, F.; Ma, J. A miR-135b-TAZ positive feedback loop promotes epithelial-mesenchymal transition (EMT) and tumorigenesis in osteosarcoma. Cancer Lett. 2017, 407, 32–44. [Google Scholar] [CrossRef] [PubMed]
- Grunewald, T.G.P.; Cidre-Aranaz, F.; Surdez, D.; Tomazou, E.M.; de Alava, E.; Kovar, H.; Sorensen, P.H.; Delattre, O.; Dirksen, U. Ewing sarcoma. Nat. Rev. Dis. Prim. 2018, 4, 5. [Google Scholar] [CrossRef]
- Ahmed, A.A.; Abedalthagafi, M.; Anwar, A.E.; Bui, M.M. Akt and Hippo Pathways in Ewing’s Sarcoma Tumors and Their Prognostic Significance. J. Cancer 2015, 6, 1005–1010. [Google Scholar] [CrossRef] [Green Version]
- Hsu, J.H.; Lawlor, E.R. BMI-1 suppresses contact inhibition and stabilizes YAP in Ewing sarcoma. Oncogene 2011, 30, 2077–2085. [Google Scholar] [CrossRef] [Green Version]
- Franzetti, G.A.; Laud-Duval, K.; van der Ent, W.; Brisac, A.; Irondelle, M.; Aubert, S.; Dirksen, U.; Bouvier, C.; de Pinieux, G.; Snaar-Jagalska, E.; et al. Cell-to-cell heterogeneity of EWSR1-FLI1 activity determines proliferation/migration choices in Ewing sarcoma cells. Oncogene 2017, 36, 3505–3514. [Google Scholar] [CrossRef] [Green Version]
- Marques Howarth, M.; Simpson, D.; Ngok, S.P.; Nieves, B.; Chen, R.; Siprashvili, Z.; Vaka, D.; Breese, M.R.; Crompton, B.D.; Alexe, G.; et al. Long noncoding RNA EWSAT1-mediated gene repression facilitates Ewing sarcoma oncogenesis. J. Clin. Investig. 2014, 124, 5275–5290. [Google Scholar] [CrossRef]
- Vlahov, N.; Scrace, S.; Soto, M.S.; Grawenda, A.M.; Bradley, L.; Pankova, D.; Papaspyropoulos, A.; Yee, K.S.; Buffa, F.; Goding, C.R.; et al. Alternate RASSF1 Transcripts Control SRC Activity, E-Cadherin Contacts, and YAP-Mediated Invasion. Curr. Biol. 2015, 25, 3019–3034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, S.; Huang, Q.; Hu, J.; Li, L.; Xiao, Y.; Yu, H.; Han, Z.; Wang, T.; Zhou, W.; Wei, H.; et al. EWS-FLI1-mediated tenascin-C expression promotes tumour progression by targeting MALAT1 through integrin alpha5beta1-mediated YAP activation in Ewing sarcoma. Br. J. Cancer 2019, 121, 922–933. [Google Scholar] [CrossRef] [PubMed]
- Hawkins, A.G.; Julian, C.M.; Konzen, S.; Treichel, S.; Lawlor, E.R.; Bailey, K.M. Microenvironmental Factors Drive Tenascin C and Src Cooperation to Promote Invadopodia Formation in Ewing Sarcoma. Neoplasia 2019, 21, 1063–1072. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, A.; Agulnik, M. Epithelioid Hemangioendothelioma: Update on Diagnosis and Treatment. Curr. Treat. Options Oncol. 2018, 19, 19. [Google Scholar] [CrossRef]
- Mendlick, M.R.; Nelson, M.; Pickering, D.; Johansson, S.L.; Seemayer, T.A.; Neff, J.R.; Vergara, G.; Rosenthal, H.; Bridge, J.A. Translocation t(1;3)(p36.3;q25) is a nonrandom aberration in epithelioid hemangioendothelioma. Am. J. Surg. Pathol. 2001, 25, 684–687. [Google Scholar] [CrossRef]
- Tanas, M.R.; Sboner, A.; Oliveira, A.M.; Erickson-Johnson, M.R.; Hespelt, J.; Hanwright, P.J.; Flanagan, J.; Luo, Y.; Fenwick, K.; Natrajan, R.; et al. Identification of a disease-defining gene fusion in epithelioid hemangioendothelioma. Sci. Transl. Med. 2011, 3, 98ra82. [Google Scholar] [CrossRef]
- Patel, N.R.; Salim, A.A.; Sayeed, H.; Sarabia, S.F.; Hollingsworth, F.; Warren, M.; Jakacky, J.; Tanas, M.; Oliveira, A.M.; Rubin, B.P.; et al. Molecular characterization of epithelioid haemangioendotheliomas identifies novel WWTR1-CAMTA1 fusion variants. Histopathology 2015, 67, 699–708. [Google Scholar] [CrossRef]
- Anderson, T.; Zhang, L.; Hameed, M.; Rusch, V.; Travis, W.D.; Antonescu, C.R. Thoracic epithelioid malignant vascular tumors: A clinicopathologic study of 52 cases with emphasis on pathologic grading and molecular studies of WWTR1-CAMTA1 fusions. Am. J. Surg. Pathol. 2015, 39, 132–139. [Google Scholar] [CrossRef] [Green Version]
- Tanas, M.R.; Ma, S.; Jadaan, F.O.; Ng, C.K.; Weigelt, B.; Reis-Filho, J.S.; Rubin, B.P. Mechanism of action of a WWTR1(TAZ)-CAMTA1 fusion oncoprotein. Oncogene 2016, 35, 929–938. [Google Scholar] [CrossRef] [Green Version]
- Dei Tos, A.P. Liposarcomas: Diagnostic pitfalls and new insights. Histopathology 2014, 64, 38–52. [Google Scholar] [CrossRef]
- Perez-Losada, J.; Pintado, B.; Gutierrez-Adan, A.; Flores, T.; Banares-Gonzalez, B.; del Campo, J.C.; Martin-Martin, J.F.; Battaner, E.; Sanchez-Garcia, I. The chimeric FUS/TLS-CHOP fusion protein specifically induces liposarcomas in transgenic mice. Oncogene 2000, 19, 2413–2422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riggi, N.; Cironi, L.; Provero, P.; Suva, M.L.; Stehle, J.C.; Baumer, K.; Guillou, L.; Stamenkovic, I. Expression of the FUS-CHOP fusion protein in primary mesenchymal progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res. 2006, 66, 7016–7023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Antonescu, C.R.; Rosenblum, M.K.; Pereira, P.; Nascimento, A.G.; Woodruff, J.M. Sclerosing Epithelioid Fibrosarcoma: A Study of 16 Cases and Confirmation of a Clinicopathologically Distinct Tumor. Am. J. Surg. Pathol. 2001, 25, 699–709. [Google Scholar] [CrossRef] [PubMed]
- Mohamed, M.; Fisher, C.; Thway, K. Low-grade fibromyxoid sarcoma: Clinical, morphologic and genetic features. Ann. Diagn. Pathol. 2017, 28, 60–67. [Google Scholar] [CrossRef] [PubMed]
- Arbajian, E.; Puls, F.; Magnusson, L.; Thway, K.; Fisher, C.; Sumathi, V.P.; Tayebwa, J.; Nord, K.H.; Kindblom, L.G.; Mertens, F. Recurrent EWSR1-CREB3L1 gene fusions in sclerosing epithelioid fibrosarcoma. Am. J. Surg. Pathol. 2014, 38, 801–808. [Google Scholar] [CrossRef]
- Prieto-Granada, C.; Zhang, L.; Chen, H.W.; Sung, Y.S.; Agaram, N.P.; Jungbluth, A.A.; Antonescu, C.R. A genetic dichotomy between pure sclerosing epithelioid fibrosarcoma (SEF) and hybrid SEF/low-grade fibromyxoid sarcoma: A pathologic and molecular study of 18 cases. Genes Chromosom. Cancer 2015, 54, 28–38. [Google Scholar] [CrossRef] [Green Version]
- Crose, L.E.; Galindo, K.A.; Kephart, J.G.; Chen, C.; Fitamant, J.; Bardeesy, N.; Bentley, R.C.; Galindo, R.L.; Chi, J.T.; Linardic, C.M. Alveolar rhabdomyosarcoma-associated PAX3-FOXO1 promotes tumorigenesis via Hippo pathway suppression. J. Clin. Investig. 2014, 124, 285–296. [Google Scholar] [CrossRef] [Green Version]
- Pipes, G.C.; Creemers, E.E.; Olson, E.N. The myocardin family of transcriptional coactivators: Versatile regulators of cell growth, migration, and myogenesis. Genes Dev. 2006, 20, 1545–1556. [Google Scholar] [CrossRef] [Green Version]
- Honda, M.; Hidaka, K.; Fukada, S.I.; Sugawa, R.; Shirai, M.; Ikawa, M.; Morisaki, T. Vestigial-like 2 contributes to normal muscle fiber type distribution in mice. Sci. Rep. 2017, 7, 7168. [Google Scholar] [CrossRef] [Green Version]
- Stacchiotti, S.; Van Tine, B.A. Synovial Sarcoma: Current Concepts and Future Perspectives. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 180–187. [Google Scholar] [CrossRef] [PubMed]
- Ladanyi, M. Fusions of the SYT and SSX genes in synovial sarcoma. Oncogene 2001, 20, 5755–5762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lam, S.W.; Cleven, A.H.G.; Kroon, H.M.; Briaire-de Bruijn, I.H.; Szuhai, K.; Bovee, J. Utility of FOS as diagnostic marker for osteoid osteoma and osteoblastoma. Virchows Arch. 2020, 476, 455–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gutierrez, J.C.; Perez, E.A.; Franceschi, D.; Moffat, F.L., Jr.; Livingstone, A.S.; Koniaris, L.G. Outcomes for soft-tissue sarcoma in 8249 cases from a large state cancer registry. J. Surg Res. 2007, 141, 105–114. [Google Scholar] [CrossRef] [PubMed]
- Thway, K. Sarcomas. In Brenner’s Encyclopedia of Genetics, 2nd ed.; Maloy, S., Hughes, K., Eds.; Academic Press: San Diego, CA, USA, 2013; pp. 327–331. [Google Scholar]
- Ballinger, M.L.; Goode, D.L.; Ray-Coquard, I.; James, P.A.; Mitchell, G.; Niedermayr, E.; Puri, A.; Schiffman, J.D.; Dite, G.S.; Cipponi, A.; et al. Monogenic and polygenic determinants of sarcoma risk: An international genetic study. Lancet Oncol. 2016, 17, 1261–1271. [Google Scholar] [CrossRef] [PubMed]
- Eisinger-Mathason, T.S.K.; Mucaj, V.; Biju, K.M.; Nakazawa, M.S.; Gohil, M.; Cash, T.P.; Yoon, S.S.; Skuli, N.; Park, K.M.; Gerecht, S.; et al. Deregulation of the Hippo pathway in soft-tissue sarcoma promotes FOXM1 expression and tumorigenesis. Proc. Natl. Acad. Sci. USA 2015, 112, E3402–E3411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mito, J.K.; Riedel, R.F.; Dodd, L.; Lahat, G.; Lazar, A.J.; Dodd, R.D.; Stangenberg, L.; Eward, W.C.; Hornicek, F.J.; Yoon, S.S.; et al. Cross Species Genomic Analysis Identifies a Mouse Model as Undifferentiated Pleomorphic Sarcoma/Malignant Fibrous Histiocytoma. PLoS ONE 2009, 4, e8075. [Google Scholar] [CrossRef]
- Kelleher, F.C.; O’Sullivan, H. FOXM1 in sarcoma: Role in cell cycle, pluripotency genes and stem cell pathways. Oncotarget 2016, 7, 42792–42804. [Google Scholar] [CrossRef] [Green Version]
- Bakkar, N.; Wang, J.; Ladner, K.J.; Wang, H.; Dahlman, J.M.; Carathers, M.; Acharyya, S.; Rudnicki, M.A.; Hollenbach, A.D.; Guttridge, D.C. IKK/NF-κB regulates skeletal myogenesis via a signaling switch to inhibit differentiation and promote mitochondrial biogenesis. J. Cell Biol. 2008, 180, 787–802. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Garzon, R.; Sun, H.; Ladner, K.J.; Singh, R.; Dahlman, J.; Cheng, A.; Hall, B.M.; Qualman, S.J.; Chandler, D.S.; et al. NF-kappaB-YY1-miR-29 regulatory circuitry in skeletal myogenesis and rhabdomyosarcoma. Cancer Cell 2008, 14, 369–381. [Google Scholar] [CrossRef] [Green Version]
- Rivera-Reyes, A.; Ye, S.; Marino, G.E.; Egolf, S.; Ciotti, G.E.; Chor, S.; Liu, Y.; Posimo, J.M.; Park, P.M.C.; Pak, K.; et al. YAP1 enhances NF-κB-dependent and independent effects on clock-mediated unfolded protein responses and autophagy in sarcoma. Cell Death Dis. 2018, 9, 1108. [Google Scholar] [CrossRef]
- Nazeri, E.; Gouran Savadkoohi, M.; Majidzadeh, A.K.; Esmaeili, R. Chondrosarcoma: An overview of clinical behavior, molecular mechanisms mediated drug resistance and potential therapeutic targets. Crit. Rev. Oncol. Hematol. 2018, 131, 102–109. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Bedford, M.T. Protein arginine methyltransferases and cancer. Nat. Rev. Cancer 2013, 13, 37–50. [Google Scholar] [CrossRef] [PubMed]
- Kao, Y.C.; Lee, J.C.; Huang, H.Y. What is new about the molecular genetics in matrix-producing soft tissue tumors? -The contributions to pathogenetic understanding and diagnostic classification. Virchows. Arch. 2020, 476, 121–134. [Google Scholar] [CrossRef] [PubMed]
- Schneider, N.; Fisher, C.; Thway, K. Ossifying fibromyxoid tumor: Morphology, genetics, and differential diagnosis. Ann. Diagn. Pathol. 2016, 20, 52–58. [Google Scholar] [CrossRef]
- Lin, L.; Sabnis, A.J.; Chan, E.; Olivas, V.; Cade, L.; Pazarentzos, E.; Asthana, S.; Neel, D.; Yan, J.J.; Lu, X.; et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat. Genet. 2015, 47, 250–256. [Google Scholar] [CrossRef]
- Li, Z.; Razavi, P.; Li, Q.; Toy, W.; Liu, B.; Ping, C.; Hsieh, W.; Sanchez-Vega, F.; Brown, D.N.; Da Cruz Paula, A.F.; et al. Loss of the FAT1 Tumor Suppressor Promotes Resistance to CDK4/6 Inhibitors via the Hippo Pathway. Cancer Cell 2018, 34, 893–905.e898. [Google Scholar] [CrossRef] [Green Version]
- Guenther, L.M.; Dharia, N.V.; Ross, L.; Conway, A.; Robichaud, A.L.; Catlett, J.L., 2nd; Wechsler, C.S.; Frank, E.S.; Goodale, A.; Church, A.J.; et al. A Combination CDK4/6 and IGF1R Inhibitor Strategy for Ewing Sarcoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 1343–1357. [Google Scholar] [CrossRef] [Green Version]
- Bressler, N.M.; Bressler, S.B. Photodynamic therapy with verteporfin (Visudyne): Impact on ophthalmology and visual sciences. Investig. Ophthalmol. Vis. Sci. 2000, 41, 624–628. [Google Scholar]
- Liu-Chittenden, Y.; Huang, B.; Shim, J.S.; Chen, Q.; Lee, S.-J.; Anders, R.A.; Liu, J.O.; Pan, D. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012, 26, 1300–1305. [Google Scholar] [CrossRef] [Green Version]
- Wang, C.; Zhu, X.; Feng, W.; Yu, Y.; Jeong, K.; Guo, W.; Lu, Y.; Mills, G.B. Verteporfin inhibits YAP function through up-regulating 14-3-3σ sequestering YAP in the cytoplasm. Am. J. Cancer Res. 2016, 6, 27–37. [Google Scholar]
- Wang, B.; Shao, W.; Shi, Y.; Liao, J.; Chen, X.; Wang, C. Verteporfin induced SUMOylation of YAP1 in endometrial cancer. Am. J. Cancer Res. 2020, 10, 1207–1217. [Google Scholar] [PubMed]
- Wei, H.; Wang, F.; Wang, Y.; Li, T.; Xiu, P.; Zhong, J.; Sun, X.; Li, J. Verteporfin suppresses cell survival, angiogenesis and vasculogenic mimicry of pancreatic ductal adenocarcinoma via disrupting the YAP-TEAD complex. Cancer Sci. 2017, 108, 478–487. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Gou, J.; Jia, J.; Yi, T.; Cui, T.; Li, Z. Verteporfin, a suppressor of YAP-TEAD complex, presents promising antitumor properties on ovarian cancer. OncoTargets Ther. 2016, 9, 5371–5381. [Google Scholar] [CrossRef] [Green Version]
- Giraud, J.; Molina-Castro, S.; Seeneevassen, L.; Sifré, E.; Izotte, J.; Tiffon, C.; Staedel, C.; Boeuf, H.; Fernandez, S.; Barthelemy, P.; et al. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int. J. Cancer 2020, 146, 2255–2267. [Google Scholar] [CrossRef] [PubMed]
- Dong, L.; Lin, F.; Wu, W.; Liu, Y.; Huang, W. Verteporfin inhibits YAP-induced bladder cancer cell growth and invasion via Hippo signaling pathway. Int. J. Med. Sci. 2018, 15, 645–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, C.; Li, X. Verteporfin inhibits cell proliferation and induces apoptosis in different subtypes of breast cancer cell lines without light activation. BMC Cancer 2020, 20, 1042. [Google Scholar] [CrossRef]
- Lui, J.W.; Xiao, S.; Ogomori, K.; Hammarstedt, J.E.; Little, E.C.; Lang, D. The Efficiency of Verteporfin as a Therapeutic Option in Pre-Clinical Models of Melanoma. J. Cancer 2019, 10, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Isakoff, S.J.; Rogers, G.S.; Hill, S.; McMullan, P.; Habin, K.R.; Park, H.; Bartenstein, D.W.; Chen, S.T.; Barry, W.T.; Overmoyer, B. An open label, phase II trial of continuous low-irradiance photodynamic therapy (CLIPT) using verteporfin for the treatment of cutaneous breast cancer metastases. J. Clin. Oncol. 2017, 35, TPS1121. [Google Scholar] [CrossRef]
- Oku, Y.; Nishiya, N.; Shito, T.; Yamamoto, R.; Yamamoto, Y.; Oyama, C.; Uehara, Y. Small molecules inhibiting the nuclear localization of YAP/TAZ for chemotherapeutics and chemosensitizers against breast cancers. FEBS Open Bio 2015, 5, 542–549. [Google Scholar] [CrossRef] [Green Version]
- Shor, A.C.; Keschman, E.A.; Lee, F.Y.; Muro-Cacho, C.; Letson, G.D.; Trent, J.C.; Pledger, W.J.; Jove, R. Dasatinib inhibits migration and invasion in diverse human sarcoma cell lines and induces apoptosis in bone sarcoma cells dependent on SRC kinase for survival. Cancer Res. 2007, 67, 2800–2808. [Google Scholar] [CrossRef] [Green Version]
- Mukaihara, K.; Tanabe, Y.; Kubota, D.; Akaike, K.; Hayashi, T.; Mogushi, K.; Hosoya, M.; Sato, S.; Kobayashi, E.; Okubo, T.; et al. Cabozantinib and dastinib exert anti-tumor activity in alveolar soft part sarcoma. PLoS ONE 2017, 12, e0185321. [Google Scholar] [CrossRef] [Green Version]
- Lopez-Acevedo, M.; Grace, L.; Teoh, D.; Whitaker, R.; Adams, D.J.; Jia, J.; Nixon, A.B.; Secord, A.A. Dasatinib (BMS-35482) potentiates the activity of gemcitabine and docetaxel in uterine leiomyosarcoma cell lines. Gynecol. Oncol. Res. Pract. 2014, 1, 2. [Google Scholar] [CrossRef]
- Timeus, F.; Crescenzio, N.; Fandi, A.; Doria, A.; Foglia, L.; Cordero di Montezemolo, L. In vitro antiproliferative and antimigratory activity of dasatinib in neuroblastoma and Ewing sarcoma cell lines. Oncol. Rep. 2008, 19, 353–359. [Google Scholar] [CrossRef] [Green Version]
- Kawakita, T.; Masato, N.; Takiguchi, E.; Abe, A.; Irahara, M. Cytotoxic effects of 15-deoxy-Δ12,14-prostaglandin J2 alone and in combination with dasatinib against uterine sarcoma in vitro. Exp. Ther. Med. 2017, 13, 2939–2945. [Google Scholar] [CrossRef] [Green Version]
- Abaza, Y.; Kantarjian, H.; Alwash, Y.; Borthakur, G.; Champlin, R.; Kadia, T.; Garcia-Manero, G.; Daver, N.; Ravandi, F.; Verstovsek, S.; et al. Phase I/II study of dasatinib in combination with decitabine in patients with accelerated or blast phase chronic myeloid leukemia. Am. J. Hematol. 2020, 95, 1288–1295. [Google Scholar] [CrossRef]
- Kimura, S.; Imagawa, J.; Murai, K.; Hino, M.; Kitawaki, T.; Okada, M.; Tanaka, H.; Shindo, M.; Kumagai, T.; Ikezoe, T.; et al. Treatment-free remission after first-line dasatinib discontinuation in patients with chronic myeloid leukaemia (first-line DADI trial): A single-arm, multicentre, phase 2 trial. Lancet. Haematol. 2020, 7, e218–e225. [Google Scholar] [CrossRef]
- Cortes, J.E.; Jiang, Q.; Wang, J.; Weng, J.; Zhu, H.; Liu, X.; Hochhaus, A.; Kim, D.W.; Radich, J.; Savona, M.; et al. Dasatinib vs. imatinib in patients with chronic myeloid leukemia in chronic phase (CML-CP) who have not achieved an optimal response to 3 months of imatinib therapy: The DASCERN randomized study. Leukemia 2020, 34, 2064–2073. [Google Scholar] [CrossRef] [Green Version]
- Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al. Dasatinib-Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623. [Google Scholar] [CrossRef]
- Morris, P.G.; Rota, S.; Cadoo, K.; Zamora, S.; Patil, S.; D’Andrea, G.; Gilewski, T.; Bromberg, J.; Dang, C.; Dickler, M.; et al. Phase II Study of Paclitaxel and Dasatinib in Metastatic Breast Cancer. Clin. Breast Cancer 2018, 18, 387–394. [Google Scholar] [CrossRef]
- Creelan, B.C.; Gray, J.E.; Tanvetyanon, T.; Chiappori, A.A.; Yoshida, T.; Schell, M.J.; Antonia, S.J.; Haura, E.B. Phase 1 trial of dasatinib combined with afatinib for epidermal growth factor receptor- (EGFR-) mutated lung cancer with acquired tyrosine kinase inhibitor (TKI) resistance. Br. J. Cancer 2019, 120, 791–796. [Google Scholar] [CrossRef] [Green Version]
- Kelley, M.J.; Jha, G.; Shoemaker, D.; Herndon, J.E., 2nd; Gu, L.; Barry, W.T.; Crawford, J.; Ready, N. Phase II Study of Dasatinib in Previously Treated Patients with Advanced Non-Small Cell Lung Cancer. Cancer Investig. 2017, 35, 32–35. [Google Scholar] [CrossRef] [PubMed]
- Schuetze, S.M.; Wathen, J.K.; Lucas, D.R.; Choy, E.; Samuels, B.L.; Staddon, A.P.; Ganjoo, K.N.; von Mehren, M.; Chow, W.A.; Loeb, D.M.; et al. SARC009: Phase 2 study of dasatinib in patients with previously treated, high-grade, advanced sarcoma. Cancer 2016, 122, 868–874. [Google Scholar] [CrossRef] [PubMed]
- Schuetze, S.M.; Bolejack, V.; Choy, E.; Ganjoo, K.N.; Staddon, A.P.; Chow, W.A.; Tawbi, H.A.; Samuels, B.L.; Patel, S.R.; von Mehren, M.; et al. Phase 2 study of dasatinib in patients with alveolar soft part sarcoma, chondrosarcoma, chordoma, epithelioid sarcoma, or solitary fibrous tumor. Cancer 2017, 123, 90–97. [Google Scholar] [CrossRef] [PubMed]
- Montemurro, M.; Cioffi, A.; Dômont, J.; Rutkowski, P.; Roth, A.D.; von Moos, R.; Inauen, R.; Toulmonde, M.; Burkhard, R.O.; Knuesli, C.; et al. Long-term outcome of dasatinib first-line treatment in gastrointestinal stromal tumor: A multicenter, 2-stage phase 2 trial (Swiss Group for Clinical Cancer Research 56/07). Cancer 2018, 124, 1449–1454. [Google Scholar] [CrossRef]
- D’Angelo, S.P.; Shoushtari, A.N.; Keohan, M.L.; Dickson, M.A.; Gounder, M.M.; Chi, P.; Loo, J.K.; Gaffney, L.; Schneider, L.; Patel, Z.; et al. Combined KIT and CTLA-4 Blockade in Patients with Refractory GIST and Other Advanced Sarcomas: A Phase Ib Study of Dasatinib plus Ipilimumab. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 2972–2980. [Google Scholar] [CrossRef] [Green Version]
- Kato, S.; Jardim, D.L.; Johnson, F.M.; Subbiah, V.; Piha-Paul, S.; Tsimberidou, A.M.; Falchook, G.S.; Karp, D.; Zinner, R.; Wheler, J.; et al. Phase I study of the combination of crizotinib (as a MET inhibitor) and dasatinib (as a c-SRC inhibitor) in patients with advanced cancer. Investig. New Drugs 2018, 36, 416–423. [Google Scholar] [CrossRef]
- Demierre, M.F.; Higgins, P.D.; Gruber, S.B.; Hawk, E.; Lippman, S.M. Statins and cancer prevention. Nat. Rev. Cancer 2005, 5, 930–942. [Google Scholar] [CrossRef]
- Matusewicz, L.; Czogalla, A.; Sikorski, A.F. Attempts to use statins in cancer therapy: An update. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2020, 42, 1010428320941760. [Google Scholar] [CrossRef]
- Yang, J.; Li, C.; Shen, Y.; Zhou, H.; Shao, Y.; Zhu, W.; Chen, Y. Impact of statin use on cancer-specific mortality and recurrence: A meta-analysis of 60 observational studies. Medicine 2020, 99, e19596. [Google Scholar] [CrossRef]
- Gachpazan, M.; Kashani, H.; Khazaei, M.; Hassanian, S.M.; Rezayi, M.; Asgharzadeh, F.; Ghayour-Mobarhan, M.; Ferns, G.A.; Avan, A. The Impact of Statin Therapy on the Survival of Patients with Gastrointestinal Cancer. Curr. Drug Targets 2019, 20, 738–747. [Google Scholar] [CrossRef]
- Borgquist, S.; Broberg, P.; Tojjar, J.; Olsson, H. Statin use and breast cancer survival—A Swedish nationwide study. BMC Cancer 2019, 19, 54. [Google Scholar] [CrossRef] [Green Version]
- Nilsson, S.; Huelsenbeck, J.; Fritz, G. Mevalonate pathway inhibitors affect anticancer drug-induced cell death and DNA damage response of human sarcoma cells. Cancer Lett. 2011, 304, 60–69. [Google Scholar] [CrossRef]
- Yang, J.; Zhang, W. New molecular insights into osteosarcoma targeted therapy. Curr. Opin. Oncol. 2013, 25, 398–406. [Google Scholar] [CrossRef]
- Majidi, A.; Na, R.; Jordan, S.J.; De Fazio, A.; Webb, P.M. Statin use and survival following a diagnosis of ovarian cancer: A prospective observational study. Int. J. Cancer 2020, 148, 1608–1615. [Google Scholar] [CrossRef]
- Brånvall, E.; Ekberg, S.; Eloranta, S.; Wästerlid, T.; Birmann, B.M.; Smedby, K.E. Statin use is associated with improved survival in multiple myeloma: A Swedish population-based study of 4315 patients. Am. J. Hematol. 2020, 95, 652–661. [Google Scholar] [CrossRef]
- Allott, E.H.; Ebot, E.M.; Stopsack, K.H.; Gonzalez-Feliciano, A.G.; Markt, S.C.; Wilson, K.M.; Ahearn, T.U.; Gerke, T.A.; Downer, M.K.; Rider, J.R.; et al. Statin Use Is Associated with Lower Risk of PTEN-Null and Lethal Prostate Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 1086–1093. [Google Scholar] [CrossRef]
- Pourlotfi, A.; Ahl, R.; Sjolin, G.; Forssten, M.P.; Bass, G.A.; Cao, Y.; Matthiessen, P.; Mohseni, S. Statin therapy and postoperative short-term mortality after rectal cancer surgery. Color. Dis. Off. J. Assoc. Coloproctology Great Br. Irel. 2020, 23, 875–881. [Google Scholar] [CrossRef]
- Raymakers, A.; Sin, D.D.; Sadatsafavi, M.; FitzGerald, J.M.; Marra, C.A.; Lynd, L.D. Statin use and lung cancer risk in chronic obstructive pulmonary disease patients: A population-based cohort study. Respir. Res. 2020, 21, 118. [Google Scholar] [CrossRef]
- Alexandre, L.; Clark, A.B.; Walton, S.; Lewis, M.P.; Kumar, B.; Cheong, E.C.; Warren, H.; Kadirkamanathan, S.S.; Parsons, S.L.; Dresner, S.M.; et al. Adjuvant statin therapy for oesophageal adenocarcinoma: The STAT-ROC feasibility study. BJS Open 2020, 4, 59–70. [Google Scholar] [CrossRef] [Green Version]
- Jameson, M.B.; Gormly, K.; Espinoza, D.; Hague, W.; Asghari, G.; Jeffery, G.M.; Price, T.J.; Karapetis, C.S.; Arendse, M.; Armstrong, J.; et al. SPAR—A randomised, placebo-controlled phase II trial of simvastatin in addition to standard chemotherapy and radiation in preoperative treatment for rectal cancer: An AGITG clinical trial. BMC Cancer 2019, 19, 1229. [Google Scholar] [CrossRef]
- Davidson, B.A.; Secord, A.A. Profile of pazopanib and its potential in the treatment of epithelial ovarian cancer. Int. J. Women’s Health 2014, 6, 289–300. [Google Scholar] [CrossRef] [Green Version]
- Bao, Y.; Nakagawa, K.; Yang, Z.; Ikeda, M.; Withanage, K.; Ishigami-Yuasa, M.; Okuno, Y.; Hata, S.; Nishina, H.; Hata, Y. A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription. J. Biochem. 2011, 150, 199–208. [Google Scholar] [CrossRef] [PubMed]
- Hosaka, S.; Horiuchi, K.; Yoda, M.; Nakayama, R.; Tohmonda, T.; Susa, M.; Nakamura, M.; Chiba, K.; Toyama, Y.; Morioka, H. A novel multi-kinase inhibitor pazopanib suppresses growth of synovial sarcoma cells through inhibition of the PI3K-AKT pathway. J. Orthop. Res. Off. Publ. Orthop. Res. Soc. 2012, 30, 1493–1498. [Google Scholar] [CrossRef]
- Kim, S.T.; Jang, H.L.; Lee, S.J.; Lee, J.; Choi, Y.L.; Kim, K.M.; Cho, J.; Park, S.H.; Park, Y.S.; Lim, H.Y.; et al. Pazopanib, a novel multitargeted kinase inhibitor, shows potent in vitro antitumor activity in gastric cancer cell lines with FGFR2 amplification. Mol. Cancer Ther. 2014, 13, 2527–2536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sloan, B.; Scheinfeld, N.S. Pazopanib, a VEGF receptor tyrosine kinase inhibitor for cancer therapy. Curr. Opin. Investig. Drugs 2008, 9, 1324–1335. [Google Scholar] [PubMed]
- Zhu, G.; Zhao, M.; Han, Q.; Tan, Y.; Sun, Y.U.; Bouvet, M.; Singh, S.R.; Ye, J.; Hoffman, R.M. Pazopanib Inhibits Tumor Growth, Lymph-node Metastasis and Lymphangiogenesis of an Orthotopic Mouse of Colorectal Cancer. Cancer Genom. Proteom. 2020, 17, 131–139. [Google Scholar] [CrossRef] [PubMed]
- Sun, J.M.; Lee, K.H.; Kim, B.S.; Kim, H.G.; Min, Y.J.; Yi, S.Y.; Yun, H.J.; Jung, S.H.; Lee, S.H.; Ahn, J.S.; et al. Pazopanib maintenance after first-line etoposide and platinum chemotherapy in patients with extensive disease small-cell lung cancer: A multicentre, randomised, placebo-controlled Phase II study (KCSG-LU12-07). Br. J. Cancer 2018, 118, 648–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dinkic, C.; Eichbaum, M.; Schmidt, M.; Grischke, E.M.; Gebauer, G.; Fricke, H.C.; Lenz, F.; Wallwiener, M.; Marme, F.; Schneeweiss, A.; et al. Pazopanib (GW786034) and cyclophosphamide in patients with platinum-resistant, recurrent, pre-treated ovarian cancer—Results of the PACOVAR-trial. Gynecol. Oncol. 2017, 146, 279–284. [Google Scholar] [CrossRef] [PubMed]
- Morgan, R.D.; Banerjee, S.; Hall, M.; Clamp, A.R.; Zhou, C.; Hasan, J.; Orbegoso, C.; Taylor, S.; Tugwood, J.; Lyon, A.R.; et al. Pazopanib and Fosbretabulin in recurrent ovarian cancer (PAZOFOS): A multi-centre, phase 1b and open-label, randomised phase 2 trial. Gynecol. Oncol. 2020, 156, 545–551. [Google Scholar] [CrossRef]
- Richardson, D.L.; Sill, M.W.; Coleman, R.L.; Sood, A.K.; Pearl, M.L.; Kehoe, S.M.; Carney, M.E.; Hanjani, P.; Van Le, L.; Zhou, X.C.; et al. Paclitaxel with and Without Pazopanib for Persistent or Recurrent Ovarian Cancer: A Randomized Clinical Trial. JAMA Oncol. 2018, 4, 196–202. [Google Scholar] [CrossRef]
- Maughan, B.L.; Pal, S.K.; Gill, D.; Boucher, K.; Martin, C.; Salgia, M.; Nussenzveig, R.; Liu, T.; Hawks, J.L.; Batten, J.; et al. Modulation of Premetastatic Niche by the Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor Pazopanib in Localized High-Risk Prostate Cancer Followed by Radical Prostatectomy: A Phase II Randomized Trial. Oncology 2018, 23, 1413-e1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Motzer, R.J.; Haas, N.B.; Donskov, F.; Gross-Goupil, M.; Varlamov, S.; Kopyltsov, E.; Lee, J.L.; Melichar, B.; Rini, B.I.; Choueiri, T.K.; et al. Randomized Phase III Trial of Adjuvant Pazopanib Versus Placebo After Nephrectomy in Patients with Localized or Locally Advanced Renal Cell Carcinoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 3916–3923. [Google Scholar] [CrossRef] [PubMed]
- Jones, R.J.; Hussain, S.A.; Protheroe, A.S.; Birtle, A.; Chakraborti, P.; Huddart, R.A.; Jagdev, S.; Bahl, A.; Stockdale, A.; Sundar, S.; et al. Randomized Phase II Study Investigating Pazopanib Versus Weekly Paclitaxel in Relapsed or Progressive Urothelial Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 1770–1777. [Google Scholar] [CrossRef] [Green Version]
- Chow, W.; Frankel, P.; Ruel, C.; Araujo, D.M.; Milhem, M.; Okuno, S.; Hartner, L.; Undevia, S.; Staddon, A. Results of a prospective phase 2 study of pazopanib in patients with surgically unresectable or metastatic chondrosarcoma. Cancer 2020, 126, 105–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grünwald, V.; Karch, A.; Schuler, M.; Schöffski, P.; Kopp, H.G.; Bauer, S.; Kasper, B.; Lindner, L.H.; Chemnitz, J.M.; Crysandt, M.; et al. Randomized Comparison of Pazopanib and Doxorubicin as First-Line Treatment in Patients with Metastatic Soft Tissue Sarcoma Age 60 Years or Older: Results of a German Intergroup Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 3555–3564. [Google Scholar] [CrossRef]
- Hirbe, A.C.; Eulo, V.; Moon, C.I.; Luo, J.; Myles, S.; Seetharam, M.; Toeniskoetter, J.; Kershner, T.; Haarberg, S.; Agulnik, M.; et al. A phase II study of pazopanib as front-line therapy in patients with non-resectable or metastatic soft-tissue sarcomas who are not candidates for chemotherapy. Eur. J. Cancer 2020, 137, 1–9. [Google Scholar] [CrossRef]
- Kim, M.; Kim, T.M.; Keam, B.; Kim, Y.J.; Paeng, J.C.; Moon, K.C.; Kim, D.W.; Heo, D.S. A Phase II Trial of Pazopanib in Patients with Metastatic Alveolar Soft Part Sarcoma. Oncology 2019, 24, 20-e29. [Google Scholar] [CrossRef] [Green Version]
- Martin-Broto, J.; Cruz, J.; Penel, N.; Le Cesne, A.; Hindi, N.; Luna, P.; Moura, D.S.; Bernabeu, D.; de Alava, E.; Lopez-Guerrero, J.A.; et al. Pazopanib for treatment of typical solitary fibrous tumours: A multicentre, single-arm, phase 2 trial. Lancet. Oncol. 2020, 21, 456–466. [Google Scholar] [CrossRef]
- Martin-Broto, J.; Stacchiotti, S.; Lopez-Pousa, A.; Redondo, A.; Bernabeu, D.; de Alava, E.; Casali, P.G.; Italiano, A.; Gutierrez, A.; Moura, D.S.; et al. Pazopanib for treatment of advanced malignant and dedifferentiated solitary fibrous tumour: A multicentre, single-arm, phase 2 trial. Lancet Oncol 2019, 20, 134–144. [Google Scholar] [CrossRef]
- Mehta, C.R.; Liu, L.; Theuer, C. An adaptive population enrichment phase III trial of TRC105 and pazopanib versus pazopanib alone in patients with advanced angiosarcoma (TAPPAS trial). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 103–108. [Google Scholar] [CrossRef]
- Pautier, P.; Penel, N.; Ray-Coquard, I.; Italiano, A.; Bompas, E.; Delcambre, C.; Bay, J.O.; Bertucci, F.; Delaye, J.; Chevreau, C.; et al. A phase II of gemcitabine combined with pazopanib followed by pazopanib maintenance, as second-line treatment in patients with advanced leiomyosarcomas: A unicancer French Sarcoma Group study (LMS03 study). Eur. J. Cancer 2020, 125, 31–37. [Google Scholar] [CrossRef] [PubMed]
- Samuels, B.L.; Chawla, S.P.; Somaiah, N.; Staddon, A.P.; Skubitz, K.M.; Milhem, M.M.; Kaiser, P.E.; Portnoy, D.C.; Priebat, D.A.; Walker, M.S.; et al. Results of a prospective phase 2 study of pazopanib in patients with advanced intermediate-grade or high-grade liposarcoma. Cancer 2017, 123, 4640–4647. [Google Scholar] [CrossRef]
- Stacchiotti, S.; Ferrari, S.; Redondo, A.; Hindi, N.; Palmerini, E.; Vaz Salgado, M.A.; Frezza, A.M.; Casali, P.G.; Gutierrez, A.; Lopez-Pousa, A.; et al. Pazopanib for treatment of advanced extraskeletal myxoid chondrosarcoma: A multicentre, single-arm, phase 2 trial. Lancet. Oncol. 2019, 20, 1252–1262. [Google Scholar] [CrossRef] [PubMed]
- Urakawa, H.; Kawai, A.; Goto, T.; Hiraga, H.; Ozaki, T.; Tsuchiya, H.; Nakayama, R.; Naka, N.; Matsumoto, Y.; Kobayashi, E.; et al. Phase II trial of pazopanib in patients with metastatic or unresectable chemoresistant sarcomas: A Japanese Musculoskeletal Oncology Group study. Cancer Sci. 2020, 111, 3303–3312. [Google Scholar] [CrossRef]
- Weiss, A.R.; Chen, Y.L.; Scharschmidt, T.J.; Chi, Y.Y.; Tian, J.; Black, J.O.; Davis, J.L.; Fanburg-Smith, J.C.; Zambrano, E.; Anderson, J.; et al. Pathological response in children and adults with large unresected intermediate-grade or high-grade soft tissue sarcoma receiving preoperative chemoradiotherapy with or without pazopanib (ARST1321): A multicentre, randomised, open-label, phase 2 trial. Lancet. Oncol. 2020, 21, 1110–1122. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Zheng, Q.; Li, Y.; Wang, G.; Gao, S.; Zhang, X.; Yan, X.; Zhang, X.; Xie, J.; Wang, Y.; et al. Metformin targets a YAP1-TEAD4 complex via AMPKα to regulate CCNE1/2 in bladder cancer cells. J. Exp. Clin. Cancer Res. CR 2019, 38, 376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, D.; Guo, J.; Wu, Y.; Chen, W.; Du, J.; Yang, L.; Wang, X.; Gong, K.; Dai, J.; Miao, S.; et al. Metformin-repressed miR-381-YAP-snail axis activity disrupts NSCLC growth and metastasis. J. Exp. Clin. Cancer Res. CR 2020, 39, 6. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Li, J.; Chen, H.; Wang, R.; Li, P.; Miao, Y.; Liu, P. Metformin suppresses proliferation and invasion of drug-resistant breast cancer cells by activation of the Hippo pathway. J. Cell. Mol. Med. 2020, 24, 5786–5796. [Google Scholar] [CrossRef] [Green Version]
- Hajimoradi Javarsiani, M.; Sajedianfard, J.; Haghjooy Javanmard, S. The effects of metformin on the hippo pathway in the proliferation of melanoma cancer cells: A preclinical study. Arch. Physiol. Biochem. 2020, 128, 1150–1155. [Google Scholar] [CrossRef]
- Gandini, S.; Guerrieri-Gonzaga, A.; Puntoni, M.; Decensi, A. Metformin and breast cancer risk. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 973–974. [Google Scholar] [CrossRef]
- De, A.; Kuppusamy, G. Metformin in breast cancer: Preclinical and clinical evidence. Curr. Probl. Cancer 2020, 44, 100488. [Google Scholar] [CrossRef] [PubMed]
- Mu, N.; Xu, T.; Gao, M.; Dong, M.; Tang, Q.; Hao, L.; Wang, G.; Li, Z.; Wang, W.; Yang, Y.; et al. Therapeutic effect of metformin in the treatment of endometrial cancer. Oncol. Lett. 2020, 20, 156. [Google Scholar] [CrossRef]
- Zhao, B.; Luo, J.; Wang, Y.; Zhou, L.; Che, J.; Wang, F.; Peng, S.; Zhang, G.; Shang, P. Metformin Suppresses Self-Renewal Ability and Tumorigenicity of Osteosarcoma Stem Cells via Reactive Oxygen Species-Mediated Apoptosis and Autophagy. Oxidative Med. Cell. Longev. 2019, 2019, 9290728. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Zhou, P.; Xu, K.; Chen, T.; Jiao, J.; Wei, H.; Yang, X.; Xu, W.; Wan, W.; Xiao, J. Metformin induces cell cycle arrest, apoptosis and autophagy through ROS/JNK signaling pathway in human osteosarcoma. Int. J. Biol. Sci. 2020, 16, 74–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uehara, T.; Eikawa, S.; Nishida, M.; Kunisada, Y.; Yoshida, A.; Fujiwara, T.; Kunisada, T.; Ozaki, T.; Udono, H. Metformin induces CD11b+-cell-mediated growth inhibition of an osteosarcoma: Implications for metabolic reprogramming of myeloid cells and anti-tumor effects. Int. Immunol. 2019, 31, 187–198. [Google Scholar] [CrossRef] [Green Version]
- Garofalo, C.; Capristo, M.; Manara, M.C.; Mancarella, C.; Landuzzi, L.; Belfiore, A.; Lollini, P.L.; Picci, P.; Scotlandi, K. Metformin as an adjuvant drug against pediatric sarcomas: Hypoxia limits therapeutic effects of the drug. PLoS ONE 2013, 8, e83832. [Google Scholar] [CrossRef]
- Chen, X.; Hu, C.; Zhang, W.; Shen, Y.; Wang, J.; Hu, F.; Yu, P. Metformin inhibits the proliferation, metastasis, and cancer stem-like sphere formation in osteosarcoma MG63 cells in vitro. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2015, 36, 9873–9883. [Google Scholar] [CrossRef]
- Nan, X.; Wang, J.; Cheng, H.; Yin, Z.; Sheng, J.; Qiu, B.; Lau, C.C.; Yustein, J.T.; Zhao, H.; Wong, S.T.C. Imatinib revives the therapeutic potential of metformin on ewing sarcoma by attenuating tumor hypoxic response and inhibiting convergent signaling pathways. Cancer Lett. 2020, 469, 195–206. [Google Scholar] [CrossRef]
- Duan, C.; Evison, A.; Taylor, L.; Onur, S.; Morten, K.; Townley, H. The common diabetes drug metformin can diminish the action of citral against Rhabdomyosarcoma cells in vitro. Phytother. Res. PTR 2020, 35, 1378–1388. [Google Scholar] [CrossRef]
- Ezewuiro, O.; Grushko, T.A.; Kocherginsky, M.; Habis, M.; Hurteau, J.A.; Mills, K.A.; Hunn, J.; Olopade, O.I.; Fleming, G.F.; Romero, I.L. Association of Metformin Use with Outcomes in Advanced Endometrial Cancer Treated with Chemotherapy. PLoS ONE 2016, 11, e0147145. [Google Scholar] [CrossRef] [Green Version]
- Bragagnoli, A.C.; Araujo, R.L.C.; Ferraz, M.W.; Dos Santos, L.V.; Abdalla, K.C.; Comar, F.; Santos, F.A.; Oliveira, M.A.; Carvalheira, J.B.C.; Cárcano, F.M.; et al. Metformin plus lrinotecan in patients with refractory colorectal cancer: A phase 2 clinical trial. Br. J. Cancer 2021, 124, 1072–1078. [Google Scholar] [CrossRef] [PubMed]
- Petchsila, K.; Prueksaritanond, N.; Insin, P.; Yanaranop, M.; Chotikawichean, N. Effect of Metformin for Decreasing Proliferative Marker in Women with Endometrial Cancer: A Randomized Double-blind Placebo-Controlled Trial. Asian Pac. J. Cancer Prev. APJCP 2020, 21, 733–741. [Google Scholar] [CrossRef] [PubMed]
- Brown, J.R.; Chan, D.K.; Shank, J.J.; Griffith, K.A.; Fan, H.; Szulawski, R.; Yang, K.; Reynolds, R.K.; Johnston, C.; McLean, K.; et al. Phase II clinical trial of metformin as a cancer stem cell-targeting agent in ovarian cancer. JCI Insight 2020, 5, e133247. [Google Scholar] [CrossRef]
- Wang, S.; Lin, Y.; Xiong, X.; Wang, L.; Guo, Y.; Chen, Y.; Chen, S.; Wang, G.; Lin, P.; Chen, H.; et al. Low-Dose Metformin Reprograms the Tumor Immune Microenvironment in Human Esophageal Cancer: Results of a Phase II Clinical Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4921–4932. [Google Scholar] [CrossRef]
- Molenaar, R.J.; Coelen, R.J.S.; Khurshed, M.; Roos, E.; Caan, M.W.A.; van Linde, M.E.; Kouwenhoven, M.; Bramer, J.A.M.; Bovée, J.; Mathôt, R.A.; et al. Study protocol of a phase IB/II clinical trial of metformin and chloroquine in patients with IDH1-mutated or IDH2-mutated solid tumours. BMJ Open 2017, 7, e014961. [Google Scholar] [CrossRef] [Green Version]
- Holden, J.K.; Cunningham, C.N. Targeting the Hippo Pathway and Cancer through the TEAD Family of Transcription Factors. Cancers 2018, 10, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Noland, C.L.; Gierke, S.; Schnier, P.D.; Murray, J.; Sandoval, W.N.; Sagolla, M.; Dey, A.; Hannoush, R.N.; Fairbrother, W.J.; Cunningham, C.N. Palmitoylation of TEAD Transcription Factors Is Required for Their Stability and Function in Hippo Pathway Signaling. Structure 2016, 24, 179–186. [Google Scholar] [CrossRef] [Green Version]
- Holden, J.K.; Crawford, J.J.; Noland, C.L.; Schmidt, S.; Zbieg, J.R.; Lacap, J.A.; Zang, R.; Miller, G.M.; Zhang, Y.; Beroza, P.; et al. Small Molecule Dysregulation of TEAD Lipidation Induces a Dominant-Negative Inhibition of Hippo Pathway Signaling. Cell Rep. 2020, 31, 107809. [Google Scholar] [CrossRef]
- Sun, Y.; Hu, L.; Tao, Z.; Jarugumilli, G.K.; Erb, H.; Singh, A.; Li, Q.; Cotton, J.L.; Greninger, P.; Egan, R.K.; et al. Pharmacological blockade of TEAD–YAP reveals its therapeutic limitation in cancer cells. Nat. Commun. 2022, 13, 6744. [Google Scholar] [CrossRef]
- Tolcher, A.W.; Lakhani, N.J.; McKean, M.; Lingaraj, T.; Victor, L.; Sanchez-Martin, M.; Kacena, K.; Malek, K.S.; Santillana, S. A phase 1, first-in-human study of IK-930, an oral TEAD inhibitor targeting the Hippo pathway in subjects with advanced solid tumors. J. Clin. Oncol. 2022, 40, TPS3168. [Google Scholar] [CrossRef]
Sarcoma Subtype | T-Sarcoma/ Non T-Sarcoma | Hippo Member | Deregulation Mechanism | Deregulating Factors or Genetic Aberration | References |
---|---|---|---|---|---|
OS | Non T-Sarcoma | YAP | Transcriptional | H19 lncRNA/ Hedgehog signalling | [72] |
SOX2 | [73] | ||||
Post-transcriptional | B4GALT1-AS1 lncRNA/HuR | [74,75] | |||
Epigenetic | circFAT1/miR-375 | [76] | |||
miR-625 | [77] | ||||
Gankyrin/ miR-200a | [78] | ||||
Post-translational | FAT10 | [79] | |||
ROCK2 | [80,81,82] | ||||
NF2 | Mutation | NF2 | [83,84] | ||
Post-translational | CD44 | [85,86,87,88] | |||
Transcriptional | SOX2 | [89,90] | |||
LATS1/2 | Protein Upregulation | Tankyrase 1 | [91] | ||
Epigenetic | miR-100HG/EZH2 | [92] | |||
Post-translational | miR-302b/YOD1 | [93] | |||
miR-34c/PLOD1 | [94] | ||||
RASSF 4/5/10 | Epigenetic | Promoter hypermethylation | [95,96] | ||
EwS | YAP | Transcriptional Interference | EWSR1::FLI1 | [69,97] | |
T-Sarcoma | TAZ | Transcriptional Repression | EWSR1::FLI1 | [69,98] | |
RASSF1/2 | Epigenetic | Promoter hypermethylation | [69,99,100] | ||
EHE | T-Sarcoma | TAZ | Chromosomal Rearrangement | WWTR1::CAMTA1 | [63,99,100,101] |
YAP | Chromosomal Rearrangement | YAP1::TFE3 | [64,102] | ||
MLS | T-Sarcoma | YAP | Transcriptional induction and nuclear localization | FUS::DDIT3 | [103,104] |
SEF and LGMFS (MUC4-) | T-Sarcoma | YAP | Chromosomal Rearrangement | YAP1::KMT2A | [105,106,107,108,109,110] |
ARMS | T-Sarcoma | MST1 | Protein inhibition by indirect fusion-dependent Mechanism | PAX3::FOXO1-dependent upregulation of RASSF4 | [14] |
RASSF1/5 | Epigenetic | Promoter hypermethylation | [66,111,112] | ||
SRMS | T-Sarcoma | TEAD | Chromosomal Rearrangement | TEAD1::NCOA2 | [113,114,115,116] |
VGLL2 | Chromosomal Rearrangement | VGLL2::NCOA2, VGLL2::CITED | [113,117,118,119] | ||
SS | T-Sarcoma | MST1, MOB1 | Protein inhibition by indirect fusion-dependent mechanism | SS18::SSX-dependent IGF-II/IGF-IR signaling loop | [120] |
non-FOS-rearranged OB | Non T-Sarcoma | NF2 | CNA | NF2 homozygous deletion | [121] |
UPS | Non T-Sarcoma | MST1/2 and LATS1/2 | Post-translational and epigenetic | Proteasomal degradation, deacetylated histones and hypermethylated promoters | [122,123,124] |
AMOT | Epigenetic | Histone deacetylation | [124] | ||
CS | Non T-Sarcoma | LATS1 and other kinases | Post-translational | PMRT1 | [125] |
OFMT | T-Sarcoma | TAZ | Chromosomal Rearrangement | KDM2A::WWTR1 | [126] |
Small Molecule | Sarcoma | Phase | ClinicalTrials.gov Identifier | Status |
---|---|---|---|---|
Dasanitib | GIST Stage III/IV Soft Tissue Sarcoma | I | NCT01643278 | Completed |
RMS, Malignant PNST, CS, EwS, ASPS, Chordoma, Epithelioid Sarcoma, GSCB, HPC, GIST | II | NCT00464620 | Completed with results | |
Sarcoma and other tumors | II | NCT00788125 | Completed with results | |
RMS, ARMS, ERMS | I/II | NCT03041701 | Completed with results | |
GIST | II | NCT00568750 | Completed | |
Statins (Simvastatin) | CCS, EwS, OS, RMS and other tumors | I | NCT02390843 | Completed |
Pazopanib | Advanced Soft Tissue Sarcoma | I/II | NCT01975519 | Completed with results |
Soft Tissue Sarcoma | II | NCT02300545 | Completed with results | |
Sarcoma | II | NCT01593748 | Completed with results | |
Soft Tissue Sarcoma | III | NCT00753688 | Completed with results | |
Soft Tissue Sarcoma | II | NCT00297258 | Completed with results | |
Stage IIA/III/IV Adult Soft Tissue Sarcoma | NA | NCT01446809 | Completed with results | |
Adult/Recurrent LPS Recurrent/Metastatic OS Recurrent/Stage IV Adult Soft Tissue Sarcoma | II | NCT02357810 | Completed with results | |
Adult ASPS, Angiosarcoma, DSRCT, EHE, Epithelioid Sarcoma, EMSC, Extraskeletal OS, Adult FS, LMS, LPS, Malignant PNST, RMS, SS, UPS, Malignant HPC, Recurrent/Stage III/ IV Adult Soft Tissue Sarcoma | II | NCT01532687 | Completed with results | |
Adult Angiosarcoma, Recurrent / Stage III/IV Adult Soft Tissue Sarcoma | II | NCT01462630 | Completed with results | |
Recurrent Uterine Corpus Sarcoma and other tumors | II | NCT01247571 | Completed with results | |
Advanced Angiosarcoma | III | NCT02979899 | Completed with results | |
Surgically and metastatic LPS | II | NCT01506596 | Completed with results | |
Advanced/ Metastatic LPS | II | NCT01692496 | Completed with results | |
CS, Metastatic CS | II | NCT01330966 | Completed with results | |
Solid Tumors | II | NCT01956669 | Completed with results | |
Solid Tumor | I | NCT01468922 | Completed with results | |
Metformin | OS, EwS | II | NCT04758000 | Recruiting |
CS and other tumors | I/II | NCT02496741 | Completed | |
Angiosarcoma and other tumors | II | NCT01042379 | Recruiting | |
IK-930 | Adult Solid Tumor, EHE, Solid Tumors With YAP1/TAZ Fusion Genes, NF2 Deficiency or YAP1 or TAZ Gene Fusions, and other tumors | I | NCT05228015 | Recruiting |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Salguero-Aranda, C.; Olmedo-Pelayo, J.; de Álava, E.; Amaral, A.T.; Díaz-Martín, J. Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma. Cancers 2022, 14, 6211. https://doi.org/10.3390/cancers14246211
Salguero-Aranda C, Olmedo-Pelayo J, de Álava E, Amaral AT, Díaz-Martín J. Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma. Cancers. 2022; 14(24):6211. https://doi.org/10.3390/cancers14246211
Chicago/Turabian StyleSalguero-Aranda, Carmen, Joaquín Olmedo-Pelayo, Enrique de Álava, Ana Teresa Amaral, and Juan Díaz-Martín. 2022. "Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma" Cancers 14, no. 24: 6211. https://doi.org/10.3390/cancers14246211
APA StyleSalguero-Aranda, C., Olmedo-Pelayo, J., de Álava, E., Amaral, A. T., & Díaz-Martín, J. (2022). Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma. Cancers, 14(24), 6211. https://doi.org/10.3390/cancers14246211