Register      Login
Australian Journal of Chemistry Australian Journal of Chemistry Society
An international journal for chemical science
REVIEW (Open Access)

Hippo signaling in cancer: regulatory mechanisms and therapeutic strategies

Zhao Huang https://orcid.org/0000-0001-9994-2070 A , Yunhan Tan A B , Wei Zhang C D E , Xiangdong Tang F , Edouard C. Nice G * and Canhua Huang https://orcid.org/0000-0003-2247-7750 A *
+ Author Affiliations
- Author Affiliations

A Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.

B West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.

C Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.

D West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.

E Medical Big Data Center, Sichuan University, Chengdu, 610041, China.

F Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.

G Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia.




Zhao Huang is currently postdoctor in Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University. His research focuses on the tumor biology including the growth, metastasis and drug resistance of cancers.



Yunhan Tan is currently an undergraduate student in West China School of Stomatology, Sichuan University. Her research focuses on the application of nanotechnology and immune therapy for head and neck squamous cell carcinomas.



Prof. Wei Zhang graduated from the Clinical School of Medicine of West China University of Medical Sciences in 1985 and obtained a master’s degree in medicine in 1992. He has been working in West China Hospital since then. He is currently deputy secretary of the Party Committee of Sichuan University, the chief scientist of the Medical Big Data Center of Sichuan University and the director of the Biomedical Big Data Center of West China Hospital. He has successively undertaken over 20 projects including National 863 Program, National 973 Program, the 10th Five-Year Plan, the 11th Five-Year Plan, the 12th Five-Year Plan, the 13th Five-Year Plan, and National Natural Science Foundation projects. He has been awarded the First Prize of Natural Science by the Ministry of Education and the First Prize of Scientific and Technological Progress by Sichuan Province, among other awards. Under his leadership, the team has established a comprehensive biological database of depression and anxiety disorders in the Chinese population. His team is interested in providing a basis for early prevention, early diagnosis, and early personalized treatment of mental disorders.



Prof. Xiangdong Tang is the chief physician of Mental Health Center, West China Hospital, Sichuan University. He has been engaged in the basic and clinical research of sleep disorders for a long time, and is good at the diagnosis and treatment of common sleep problems such as insomnia, dreaminess and rhythm disorder, as well as sleep disorders related to breathing. He is the academic and technical leader in Sichuan Province, vice chairman of the Sleep Medicine Expert Committee of the Chinese Medical Doctor Association, deputy director of the Sleep and Mental Health Professional Committee of the Chinese Sleep Research Association, and deputy director of the Sleep Disorders Professional Committee. He has been responsible for and participated in 7 scientific research projects, and has published over 100 SCI papers.



Prof. Ed Nice obtained his Licentiate from the Royal Institute of Chemistry in London in Advanced Analytical Chemistry in 1972 and Fellowship of the Chemical Society London in 1973. He is currently Adjunct Professor at Monash University where he is Head of the Clinical Biomarker Discovery and Validation (Department of Biochemistry and Molecular Biology) and a scientific advisor to the Monash Antibody Technologies Facility (MATF), of which he was Director from 2009–2013. He holds a Visiting Professorship at Sichuan University s West China Hospital and an Adjunct position at Macquarie University. Ed s long-term research interests have been in biomarker discovery and validation, high throughput monoclonal antibody production and validation and clinical biomarker assay development, with a strong translational focus on colorectal cancer.



Prof. Canhua Huang returned to China in 2005 and was appointed as a Professor of the State Key Lab of Biotherapy, West China Hospital, Sichuan University. In 2012, he won the National Science Fund for Distinguished Young Scholars of China, and took up the post of the Chief Scientist for National 973 Program entitled “Proteomics Profiling of the Redoxomes Associated with Virus-induced Carcinogenesis” from 2013 to 2017. In 2014, he was hired as the Changjiang Scholars Program Endowed Professor. In 2018, he served as the director of an Innovative Research Group of the National Nature Science Foundation of China, entitled “Redox Signaling Regulation and Carcinogenesis”. In 2020, he was a member of the 8th Discipline Evaluation Group of Academic Degrees Committee of The State Council (Basic Medicine Group). He was selected by Elsevier 2020 and 2021 China Highly Cited Scholar and won the First Prize of Natural Science Award of the Ministry of Education in 2020.

# These authors contributed equally to this paper

Handling Editor: Mibel Aguilar

Australian Journal of Chemistry 76(8) 399-412 https://doi.org/10.1071/CH22241
Submitted: 21 November 2022  Accepted: 1 June 2023   Published: 7 July 2023

© 2023 The Author(s) (or their employer(s)). Published by CSIRO Publishing. This is an open access article distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND)

Abstract

As an evolutionarily conserved pathway, Hippo signaling not only plays a key role in embryonic development, but also regulates the initiation and progression of cancer. The upstream factors regulating the Hippo pathway are complex, including cell–cell contact, cell–extracellular matrix contact, membrane receptor–ligand binding, and cytoskeletal tension. In response to these mechanical or soluble cues, the Hippo core kinases are activated or inactivated, regulating the activity of key transcription co-factor YAP/TAZ thus yielding biological consequences. In the context of neoplasm, dysregulation of Hippo signaling contributes to cancer hallmarks such as sustained proliferation, stem-like properties, and metastasis. Importantly, targeting Hippo signaling by chemicals is emerging as a promising anticancer strategy. This article briefly introduces the discovery process of the Hippo pathway, summarizes the upstream signals regulating the Hippo pathway, discusses the relationship between Hippo inactivation and cancer development, and highlights the potential use of chemicals targeting Hippo signaling in cancer treatment.

Keywords: cancer hallmarks, chemical inhibitors, embryonic development, Hippo signaling, oncogene, targeted therapy, tumor suppressor, YAP/TAZ.


References

[1]  B Mintz, Gene expression in neoplasia and differentiation. Harvey Lect 1978, 71, 193.

[2]  LM Postovit, NV Margaryan, EA Seftor, DA Kirschmann, A Lipavsky, WW Wheaton, et al. Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells. Proc Natl Acad Sci U S A 2008, 105, 4329.
         | Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells.Crossref | GoogleScholarGoogle Scholar |

[3]  A Klaus, W Birchmeier, Wnt signalling and its impact on development and cancer. Nat Rev Cancer 2008, 8, 387.
         | Wnt signalling and its impact on development and cancer.Crossref | GoogleScholarGoogle Scholar |

[4]  J Briscoe, PP Thérond, The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 2013, 14, 416.
         | The mechanisms of Hedgehog signalling and its roles in development and disease.Crossref | GoogleScholarGoogle Scholar |

[5]  V Bolo’s, J Grego-Bessa, JL de la Pompa, Notch signaling in development and cancer. Endocr Rev 2007, 28, 339.
         | Notch signaling in development and cancer.Crossref | GoogleScholarGoogle Scholar |

[6]  CJ David, J Massagué, Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol 2018, 19, 419.
         | Contextual determinants of TGFβ action in development, immunity and cancer.Crossref | GoogleScholarGoogle Scholar |

[7]  FX Yu, B Zhao, KL Guan, Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 2015, 163, 811.
         | Hippo pathway in organ size control, tissue homeostasis, and cancer.Crossref | GoogleScholarGoogle Scholar |

[8]  A Sebio, HJ Lenz, Molecular pathways: Hippo signaling, a critical tumor suppressor. Clin Cancer Res 2015, 21, 5002.
         | Molecular pathways: Hippo signaling, a critical tumor suppressor.Crossref | GoogleScholarGoogle Scholar |

[9]  Z Meng, T Moroishi, KL Guan, Mechanisms of Hippo pathway regulation. Genes Dev 2016, 30, 1.
         | Mechanisms of Hippo pathway regulation.Crossref | GoogleScholarGoogle Scholar |

[10]  T Moroishi, CG Hansen, KL Guan, The emerging roles of YAP and TAZ in cancer. Nat Rev Cancer 2015, 15, 73.
         | The emerging roles of YAP and TAZ in cancer.Crossref | GoogleScholarGoogle Scholar |

[11]  BH Sohn, JJ Shim, SB Kim, KY Jang, SM Kim, JH Kim, et al. Inactivation of Hippo pathway is significantly associated with poor prognosis in hepatocellular carcinoma. Clin Cancer Res 2016, 22, 1256.
         | Inactivation of Hippo pathway is significantly associated with poor prognosis in hepatocellular carcinoma.Crossref | GoogleScholarGoogle Scholar |

[12]  D Zhou, C Conrad, F Xia, JS Park, B Payer, Y Yin, et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 2009, 16, 425.
         | Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene.Crossref | GoogleScholarGoogle Scholar |

[13]  W Kim, SK Khan, Y Liu, R Xu, O Park, Y He, et al. Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma. Gut 2018, 67, 1692.
         | Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma.Crossref | GoogleScholarGoogle Scholar |

[14]  Y Wang, X Xu, D Maglic, MT Dill, K Mojumdar, PK-S Ng, et al. Comprehensive molecular characterization of the Hippo signaling pathway in cancer. Cell Rep 2018, 25, 1304.
         | Comprehensive molecular characterization of the Hippo signaling pathway in cancer.Crossref | GoogleScholarGoogle Scholar |

[15]  B Zhao, X Wei, W Li, RS Udan, Q Yang, J Kim, et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev 2007, 21, 2747.
         | Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control.Crossref | GoogleScholarGoogle Scholar |

[16]  B Zhao, L Li, L Wang, CY Wang, J Yu, KL Guan, Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis. Genes Dev 2012, 26, 54.
         | Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikis.Crossref | GoogleScholarGoogle Scholar |

[17]  FX Yu, B Zhao, N Panupinthu, JL Jewell, I Lian, LH Wang, et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 2012, 150, 780.
         | Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling.Crossref | GoogleScholarGoogle Scholar |

[18]  C Rauskolb, S Sun, G Sun, Y Pan, KD Irvine, Cytoskeletal tension inhibits Hippo signaling through an Ajuba-Warts complex. Cell 2014, 158, 143.
         | Cytoskeletal tension inhibits Hippo signaling through an Ajuba-Warts complex.Crossref | GoogleScholarGoogle Scholar |

[19]  Z Meng, Y Qiu, KC Lin, A Kumar, JK Placone, C Fang, et al. RAP2 mediates mechanoresponses of the Hippo pathway. Nature 2018, 560, 655.
         | RAP2 mediates mechanoresponses of the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[20]  X Zheng, H Han, GP Liu, YX Ma, RL Pan, LJ Sang, et al. LncRNA wires up Hippo and Hedgehog signaling to reprogramme glucose metabolism. EMBO J 2017, 36, 3325.
         | LncRNA wires up Hippo and Hedgehog signaling to reprogramme glucose metabolism.Crossref | GoogleScholarGoogle Scholar |

[21]  U Ehmer, J Sage, Control of proliferation and cancer growth by the Hippo signaling pathway. Mol Cancer Res 2016, 14, 127.
         | Control of proliferation and cancer growth by the Hippo signaling pathway.Crossref | GoogleScholarGoogle Scholar |

[22]  HJ Janse van Rensburg, X Yang, The roles of the Hippo pathway in cancer metastasis. Cell Signal 2016, 28, 1761.
         | The roles of the Hippo pathway in cancer metastasis.Crossref | GoogleScholarGoogle Scholar |

[23]  JH Park, JE Shin, HW Park, The role of Hippo pathway in cancer stem cell biology. Mol Cells 2018, 41, 83.
         | The role of Hippo pathway in cancer stem cell biology.Crossref | GoogleScholarGoogle Scholar |

[24]  A Britschgi, S Duss, S Kim, JP Couto, H Brinkhaus, S Koren, et al. The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα. Nature 2017, 541, 541.
         | The Hippo kinases LATS1 and 2 control human breast cell fate via crosstalk with ERα.Crossref | GoogleScholarGoogle Scholar |

[25]  S-H Jeong, H-B Kim, M-C Kim, J-m Lee, JH Lee, J-H Kim, et al. Hippo-mediated suppression of IRS2/AKT signaling prevents hepatic steatosis and liver cancer. J Clin Invest 2018, 128, 1010.
         | Hippo-mediated suppression of IRS2/AKT signaling prevents hepatic steatosis and liver cancer.Crossref | GoogleScholarGoogle Scholar |

[26]  K Tumaneng, K Schlegelmilch, RC Russell, D Yimlamai, H Basnet, N Mahadevan, et al. YAP mediates crosstalk between the Hippo and PI(3)K-TOR pathways by suppressing PTEN via miR-29. Nat Cell Biol 2012, 14, 1322.
         | YAP mediates crosstalk between the Hippo and PI(3)K-TOR pathways by suppressing PTEN via miR-29.Crossref | GoogleScholarGoogle Scholar |

[27]  K Brodowska, A Al-Moujahed, A Marmalidou, M Meyer Zu Horste, J Cichy, JW Miller, et al. The clinically used photosensitizer Verteporfin (VP) inhibits YAP-TEAD and human retinoblastoma cell growth in vitro without light activation. Exp Eye Res 2014, 124, 67.
         | The clinically used photosensitizer Verteporfin (VP) inhibits YAP-TEAD and human retinoblastoma cell growth in vitro without light activation.Crossref | GoogleScholarGoogle Scholar |

[28]  GK Michalopoulos, MC DeFrances, Liver regeneration. Science 1997, 276, 60.
         | Liver regeneration.Crossref | GoogleScholarGoogle Scholar |

[29]  RW Justice, O Zilian, DF Woods, M Noll, PJ Bryant, 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.
         | 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.Crossref | GoogleScholarGoogle Scholar |

[30]  MAR St John, W Tao, X Fei, R Fukumoto, ML Carcangiu, DG Brownstein, et al. Mice deficient of Lats1 develop soft-tissue sarcomas, ovarian tumours and pituitary dysfunction. Nat Genet 1999, 21, 182.
         | Mice deficient of Lats1 develop soft-tissue sarcomas, ovarian tumours and pituitary dysfunction.Crossref | GoogleScholarGoogle Scholar |

[31]  S Visser, X Yang, LATS tumor suppressor: a new governor of cellular homeostasis. Cell Cycle 2010, 9, 3892.
         | LATS tumor suppressor: a new governor of cellular homeostasis.Crossref | GoogleScholarGoogle Scholar |

[32]  KF Harvey, CM Pfleger, IK Hariharan, The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell 2003, 114, 457.
         | The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis.Crossref | GoogleScholarGoogle Scholar |

[33]  J Jia, W Zhang, B Wang, R Trinko, J Jiang, The Drosophila Ste20 family kinase dMST functions as a tumor suppressor by restricting cell proliferation and promoting apoptosis. Genes Dev 2003, 17, 2514.
         | The Drosophila Ste20 family kinase dMST functions as a tumor suppressor by restricting cell proliferation and promoting apoptosis.Crossref | GoogleScholarGoogle Scholar |

[34]  J Huang, S Wu, J Barrera, K Matthews, D Pan, The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell 2005, 122, 421.
         | The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP.Crossref | GoogleScholarGoogle Scholar |

[35]  M Ota, H Sasaki, Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling. Development 2008, 135, 4059.
         | Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling.Crossref | GoogleScholarGoogle Scholar |

[36]  B Zhao, X Ye, J Yu, L Li, W Li, S Li, et al. TEAD mediates YAP-dependent gene induction and growth control. Genes Dev 2008, 22, 1962.
         | TEAD mediates YAP-dependent gene induction and growth control.Crossref | GoogleScholarGoogle Scholar |

[37]  S Wu, J Huang, J Dong, D Pan, 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.
         | hippo encodes a Ste-20 family protein kinase that restricts cell proliferation and promotes apoptosis in conjunction with salvador and warts.Crossref | GoogleScholarGoogle Scholar |

[38]  X Wei, T Shimizu, ZC Lai, Mob as tumor suppressor is activated by Hippo kinase for growth inhibition in Drosophila. EMBO J 2007, 26, 1772.
         | Mob as tumor suppressor is activated by Hippo kinase for growth inhibition in Drosophila.Crossref | GoogleScholarGoogle Scholar |

[39]  C Guo, X Zhang, GP Pfeifer, The tumor suppressor RASSF1A prevents dephosphorylation of the mammalian STE20-like kinases MST1 and MST2. J Biol Chem 2011, 286, 6253.
         | The tumor suppressor RASSF1A prevents dephosphorylation of the mammalian STE20-like kinases MST1 and MST2.Crossref | GoogleScholarGoogle Scholar |

[40]  JC Boggiano, PJ Vanderzalm, RG Fehon, Tao-1 phosphorylates Hippo/MST kinases to regulate the Hippo–Salvador–Warts tumor suppressor pathway. Dev Cell 2011, 21, 888.
         | Tao-1 phosphorylates Hippo/MST kinases to regulate the Hippo–Salvador–Warts tumor suppressor pathway.Crossref | GoogleScholarGoogle Scholar |

[41]  Z Meng, T Moroishi, V Mottier-Pavie, SW Plouffe, CG Hansen, AW Hong, et al. MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat Commun 2015, 6, 8357.
         | MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[42]  S Lim, N Hermance, T Mudianto, HM Mustaly, IPM Mauricio, MA Vittoria, et al. Identification of the kinase STK25 as an upstream activator of LATS signaling. Nat Commun 2019, 10, 1547.
         | Identification of the kinase STK25 as an upstream activator of LATS signaling.Crossref | GoogleScholarGoogle Scholar |

[43]  X Feng, N Arang, DC Rigiracciolo, JS Lee, H Yeerna, Z Wang, et al. A platform of synthetic lethal gene interaction networks reveals that the GNAQ uveal melanoma oncogene controls the Hippo pathway through FAK. Cancer Cell 2019, 35, 457.
         | A platform of synthetic lethal gene interaction networks reveals that the GNAQ uveal melanoma oncogene controls the Hippo pathway through FAK.Crossref | GoogleScholarGoogle Scholar |

[44]  S Qi, Y Zhu, X Liu, P Li, Y Wang, Y Zeng, et al. WWC proteins mediate LATS1/2 activation by Hippo kinases and imply a tumor suppression strategy. Mol Cell 2022, 82, 1850.
         | WWC proteins mediate LATS1/2 activation by Hippo kinases and imply a tumor suppression strategy.Crossref | GoogleScholarGoogle Scholar |

[45]  W Kim, E-h Jho, The history and regulatory mechanism of the Hippo pathway. BMB Rep 2018, 51, 106.
         | The history and regulatory mechanism of the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[46]  M Luo, Z Meng, T Moroishi, KC Lin, G Shen, F Mo, et al. Heat stress activates YAP/TAZ to induce the heat shock transcriptome. Nat Cell Biol 2020, 22, 1447.
         | Heat stress activates YAP/TAZ to induce the heat shock transcriptome.Crossref | GoogleScholarGoogle Scholar |

[47]  H Han, HJ Nakaoka, L Hofmann, JJ Zhou, C Yu, L Zeng, et al. The Hippo pathway kinases LATS1 and LATS2 attenuate cellular responses to heavy metals through phosphorylating MTF1. Nat Cell Biol 2022, 24, 74.
         | The Hippo pathway kinases LATS1 and LATS2 attenuate cellular responses to heavy metals through phosphorylating MTF1.Crossref | GoogleScholarGoogle Scholar |

[48]  Q Liu, J Li, W Zhang, C Xiao, S Zhang, C Nian, et al. Glycogen accumulation and phase separation drives liver tumor initiation. Cell 2021, 184, 5559.
         | Glycogen accumulation and phase separation drives liver tumor initiation.Crossref | GoogleScholarGoogle Scholar |

[49]  N Nishioka, K Inoue, K Adachi, H Kiyonari, M Ota, A Ralston, et al. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell 2009, 16, 398.
         | The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass.Crossref | GoogleScholarGoogle Scholar |

[50]  NG Kim, E Koh, X Chen, BM Gumbiner, E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components. Proc Natl Acad Sci U S A 2011, 108, 11930.
         | E-cadherin mediates contact inhibition of proliferation through Hippo signaling-pathway components.Crossref | GoogleScholarGoogle Scholar |

[51]  C Yi, S Troutman, D Fera, A Stemmer-Rachamimov, JL Avila, N Christian, et al. A tight junction-associated Merlin-angiomotin complex mediates Merlin’s regulation of mitogenic signaling and tumor suppressive functions. Cancer Cell 2011, 19, 527.
         | A tight junction-associated Merlin-angiomotin complex mediates Merlin’s regulation of mitogenic signaling and tumor suppressive functions.Crossref | GoogleScholarGoogle Scholar |

[52]  Y Li, H Zhou, F Li, SW Chan, Z Lin, Z Wei, et al. Angiomotin binding-induced activation of Merlin/NF2 in the Hippo pathway. Cell Res 2015, 25, 801.
         | Angiomotin binding-induced activation of Merlin/NF2 in the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[53]  P Chiarugi, E Giannoni, Anoikis: a necessary death program for anchorage-dependent cells. Biochem Pharmacol 2008, 76, 1352.
         | Anoikis: a necessary death program for anchorage-dependent cells.Crossref | GoogleScholarGoogle Scholar |

[54]  C Penaloza, L Lin, RA Lockshin, Z Zakeri, Cell death in development: shaping the embryo. Histochem Cell Biol 2006, 126, 149.
         | Cell death in development: shaping the embryo.Crossref | GoogleScholarGoogle Scholar |

[55]  S Mathew, L Fu, M Fiorentino, H Matsuda, B Das, YB Shi, Differential regulation of cell type-specific apoptosis by stromelysin-3: a potential mechanism via the cleavage of the laminin receptor during tail resorption in Xenopus laevis. J Biol Chem 2009, 284, 18545.
         | Differential regulation of cell type-specific apoptosis by stromelysin-3: a potential mechanism via the cleavage of the laminin receptor during tail resorption in Xenopus laevis.Crossref | GoogleScholarGoogle Scholar |

[56]  CL Buchheit, KJ Weigel, ZT Schafer, Cancer cell survival during detachment from the ECM: multiple barriers to tumour progression. Nat Rev Cancer 2014, 14, 632.
         | Cancer cell survival during detachment from the ECM: multiple barriers to tumour progression.Crossref | GoogleScholarGoogle Scholar |

[57]  Y Cheng, T Hou, J Ping, T Chen, B Yin, LMO3 promotes hepatocellular carcinoma invasion, metastasis and anoikis inhibition by directly interacting with LATS1 and suppressing Hippo signaling. J Exp Clin Cancer Res 2018, 37, 228.
         | LMO3 promotes hepatocellular carcinoma invasion, metastasis and anoikis inhibition by directly interacting with LATS1 and suppressing Hippo signaling.Crossref | GoogleScholarGoogle Scholar |

[58]  M Larsen, VV Artym, JA Green, KM Yamada, The matrix reorganized: extracellular matrix remodeling and integrin signaling. Curr Opin Cell Biol 2006, 18, 463.
         | The matrix reorganized: extracellular matrix remodeling and integrin signaling.Crossref | GoogleScholarGoogle Scholar |

[59]  R Fan, NG Kim, BM Gumbiner, Regulation of Hippo pathway by mitogenic growth factors via phosphoinositide 3-kinase and phosphoinositide-dependent kinase-1. Proc Natl Acad Sci U S A 2013, 110, 2569.
         | Regulation of Hippo pathway by mitogenic growth factors via phosphoinositide 3-kinase and phosphoinositide-dependent kinase-1.Crossref | GoogleScholarGoogle Scholar |

[60]  NG Kim, BM Gumbiner, Adhesion to fibronectin regulates Hippo signaling via the FAK–Src–PI3K pathway. J Cell Biol 2015, 210, 503.
         | Adhesion to fibronectin regulates Hippo signaling via the FAK–Src–PI3K pathway.Crossref | GoogleScholarGoogle Scholar |

[61]  H Sabra, M Brunner, V Mandati, B Wehrle-Haller, D Lallemand, AS Ribba, et al. β1 integrin–dependent Rac/group I PAK signaling mediates YAP activation of Yes-associated protein 1 (YAP1) via NF2/merlin. J Biol Chem 2017, 292, 19179.
         | β1 integrin–dependent Rac/group I PAK signaling mediates YAP activation of Yes-associated protein 1 (YAP1) via NF2/merlin.Crossref | GoogleScholarGoogle Scholar |

[62]  KD Irvine, Integration of intercellular signaling through the Hippo pathway. Semin Cell Dev Biol 2012, 23, 812.
         | Integration of intercellular signaling through the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[63]  Z Huang, JK Zhou, K Wang, H Chen, S Qin, J Liu, et al. PDLIM1 inhibits tumor metastasis through activating Hippo signaling in hepatocellular carcinoma. Hepatology 2020, 71, 1643.
         | PDLIM1 inhibits tumor metastasis through activating Hippo signaling in hepatocellular carcinoma.Crossref | GoogleScholarGoogle Scholar |

[64]  S Mana-Capelli, M Paramasivam, S Dutta, D McCollum, Angiomotins link F-actin architecture to Hippo pathway signaling. Mol Biol Cell 2014, 25, 1676.
         | Angiomotins link F-actin architecture to Hippo pathway signaling.Crossref | GoogleScholarGoogle Scholar |

[65]  KF Harvey, X Zhang, DM Thomas, The Hippo pathway and human cancer. Nat Rev Cancer 2013, 13, 246.
         | The Hippo pathway and human cancer.Crossref | GoogleScholarGoogle Scholar |

[66]  L Sansores-Garcia, W Bossuyt, K-I Wada, S Yonemura, C Tao, H Sasaki, et al. Modulating F-actin organization induces organ growth by affecting the Hippo pathway. EMBO J 2011, 30, 2325.
         | Modulating F-actin organization induces organ growth by affecting the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[67]  BG Fernández, P Gaspar, C Brás-Pereira, B Jezowska, SR Rebelo, F Janody, Actin-Capping Protein and the Hippo pathway regulate F-actin and tissue growth in Drosophila. Development 2011, 138, 2337.
         | Actin-Capping Protein and the Hippo pathway regulate F-actin and tissue growth in Drosophila.Crossref | GoogleScholarGoogle Scholar |

[68]  S Dupont, L Morsut, M Aragona, E Enzo, S Giulitti, M Cordenonsi, et al. Role of YAP/TAZ in mechanotransduction. Nature 2011, 474, 179.
         | Role of YAP/TAZ in mechanotransduction.Crossref | GoogleScholarGoogle Scholar |

[69]  K-I Wada, K Itoga, T Okano, S Yonemura, H Sasaki, Hippo pathway regulation by cell morphology and stress fibers. Development 2011, 138, 3907.
         | Hippo pathway regulation by cell morphology and stress fibers.Crossref | GoogleScholarGoogle Scholar |

[70]  AM Babic, C-C Chen, LF Lau, Fisp12/mouse connective tissue growth factor mediates endothelial cell adhesion and migration through integrin αvβ3, promotes endothelial cell survival, and induces angiogenesis in vivo. Mol Cell Biol 1999, 19, 2958.
         | Fisp12/mouse connective tissue growth factor mediates endothelial cell adhesion and migration through integrin αvβ3, promotes endothelial cell survival, and induces angiogenesis in vivo.Crossref | GoogleScholarGoogle Scholar |

[71]  A Jedsadayanmata, CC Chen, ML Kireeva, LF Lau, SC-T Lam, Activation-dependent adhesion of human platelets to Cyr61 and Fisp12/mouse connective tissue growth factor is mediated through integrin αIIbβ3. J Biol Chem 1999, 274, 24321.
         | Activation-dependent adhesion of human platelets to Cyr61 and Fisp12/mouse connective tissue growth factor is mediated through integrin αIIbβ3.Crossref | GoogleScholarGoogle Scholar |

[72]  JR Misra, KD Irvine, The Hippo signaling network and its biological functions. Annu Rev Genet 2018, 52, 65.
         | The Hippo signaling network and its biological functions.Crossref | GoogleScholarGoogle Scholar |

[73]  JH Koo, KL Guan, Interplay between YAP/TAZ and metabolism. Cell Metab 2018, 28, 196.
         | Interplay between YAP/TAZ and metabolism.Crossref | GoogleScholarGoogle Scholar |

[74]  S Ma, Z Meng, R Chen, KL Guan, The Hippo pathway: biology and pathophysiology. Annu Rev Biochem 2019, 88, 577.
         | The Hippo pathway: biology and pathophysiology.Crossref | GoogleScholarGoogle Scholar |

[75]  S Piccolo, S Dupont, M Cordenonsi, The biology of YAP/TAZ: Hippo signaling and beyond. Physiol Rev 2014, 94, 1287.
         | The biology of YAP/TAZ: Hippo signaling and beyond.Crossref | GoogleScholarGoogle Scholar |

[76]  S Noguchi, A Saito, M Horie, Y Mikami, HI Suzuki, Y Morishita, et al. An integrative analysis of the tumorigenic role of TAZ in human non-small cell lung cancer. Clin Cancer Res 2014, 20, 4660.
         | An integrative analysis of the tumorigenic role of TAZ in human non-small cell lung cancer.Crossref | GoogleScholarGoogle Scholar |

[77]  J Díaz-Martín, MÁ López-García, L Romero-Pérez, MR Atienza-Amores, ML Pecero, MÁ Castilla, et al. Nuclear TAZ expression associates with the triple-negative phenotype in breast cancer. Endocr Relat Cancer 2015, 22, 443.
         | Nuclear TAZ expression associates with the triple-negative phenotype in breast cancer.Crossref | GoogleScholarGoogle Scholar |

[78]  KW Lee, SS Lee, SB Kim, BH Sohn, HS Lee, HJ Jang, et al. Significant association of oncogene YAP1 with poor prognosis and cetuximab resistance in colorectal cancer patients. Clin Cancer Res 2015, 21, 357.
         | Significant association of oncogene YAP1 with poor prognosis and cetuximab resistance in colorectal cancer patients.Crossref | GoogleScholarGoogle Scholar |

[79]  S-x Han, E Bai, G-h Jin, C-c He, X-j Guo, L-j Wang, et al. Expression and clinical significance of YAP, TAZ, and AREG in hepatocellular carcinoma. J Immunol Res 2014, 2014, 261365.
         | Expression and clinical significance of YAP, TAZ, and AREG in hepatocellular carcinoma.Crossref | GoogleScholarGoogle Scholar |

[80]  M Yu, Z Peng, M Qin, Y Liu, J Wang, C Zhang, et al. Interferon-γ induces tumor resistance to anti-PD-1 immunotherapy by promoting YAP phase separation. Mol Cell 2021, 81, 1216.
         | Interferon-γ induces tumor resistance to anti-PD-1 immunotherapy by promoting YAP phase separation.Crossref | GoogleScholarGoogle Scholar |

[81]  F Zanconato, M Cordenonsi, S Piccolo, YAP/TAZ at the roots of cancer. Cancer Cell 2016, 29, 783.
         | YAP/TAZ at the roots of cancer.Crossref | GoogleScholarGoogle Scholar |

[82]  W Zhang, N Nandakumar, Y Shi, M Manzano, A Smith, G Graham, et al. Downstream of mutant KRAS, the transcription regulator YAP is essential for neoplastic progression to pancreatic ductal adenocarcinoma. Sci Signal 2014, 7, ra42.
         | Downstream of mutant KRAS, the transcription regulator YAP is essential for neoplastic progression to pancreatic ductal adenocarcinoma.Crossref | GoogleScholarGoogle Scholar |

[83]  DD Shao, W Xue, EB Krall, A Bhutkar, F Piccioni, X Wang, et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 2014, 158, 171.
         | KRAS and YAP1 converge to regulate EMT and tumor survival.Crossref | GoogleScholarGoogle Scholar |

[84]  LMR Ferreira, A Hebrant, JE Dumont, Metabolic reprogramming of the tumor. Oncogene 2012, 31, 3999.
         | Metabolic reprogramming of the tumor.Crossref | GoogleScholarGoogle Scholar |

[85]  PB Gupta, I Pastushenko, A Skibinski, C Blanpain, C Kuperwasser, Phenotypic plasticity: driver of cancer initiation, progression, and therapy resistance. Cell Stem Cell 2019, 24, 65.
         | Phenotypic plasticity: driver of cancer initiation, progression, and therapy resistance.Crossref | GoogleScholarGoogle Scholar |

[86]  SH Patel, FD Camargo, D Yimlamai, Hippo signaling in the liver regulates organ size, cell fate, and carcinogenesis. Gastroenterology 2017, 152, 533.
         | Hippo signaling in the liver regulates organ size, cell fate, and carcinogenesis.Crossref | GoogleScholarGoogle Scholar |

[87]  B Zhao, L Li, Q Lei, KL Guan, The Hippo-YAP pathway in organ size control and tumorigenesis: an updated version. Genes Dev 2010, 24, 862.
         | The Hippo-YAP pathway in organ size control and tumorigenesis: an updated version.Crossref | GoogleScholarGoogle Scholar |

[88]  P Li, Y Chen, KK Mak, CK Wong, CC Wang, P Yuan, Functional role of Mst1/Mst2 in embryonic stem cell differentiation. PLoS One 2013, 8, e79867.
         | Functional role of Mst1/Mst2 in embryonic stem cell differentiation.Crossref | GoogleScholarGoogle Scholar |

[89]  M Overholtzer, J Zhang, GA Smolen, B Muir, W Li, DC Sgroi, et al. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc Natl Acad Sci U S A 2006, 103, 12405.
         | Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon.Crossref | GoogleScholarGoogle Scholar |

[90]  M Malumbres, M Barbacid, Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer 2009, 9, 153.
         | Cell cycle, CDKs and cancer: a changing paradigm.Crossref | GoogleScholarGoogle Scholar |

[91]  DA Barron, JD Kagey, The role of the Hippo pathway in human disease and tumorigenesis. Clin Transl Med 2014, 3, e25.
         | The role of the Hippo pathway in human disease and tumorigenesis.Crossref | GoogleScholarGoogle Scholar |

[92]  Z Shen, BZ Stanger, YAP regulates S-phase entry in endothelial cells. PLoS One 2015, 10, e0117522.
         | YAP regulates S-phase entry in endothelial cells.Crossref | GoogleScholarGoogle Scholar |

[93]  A Hergovich, BA Hemmings, Hippo signalling in the G2/M cell cycle phase: lessons learned from the yeast MEN and SIN pathways. Semin Cell Dev Biol 2012, 23, 794.
         | Hippo signalling in the G2/M cell cycle phase: lessons learned from the yeast MEN and SIN pathways.Crossref | GoogleScholarGoogle Scholar |

[94]  FA Dick, SM Rubin, Molecular mechanisms underlying RB protein function. Nat Rev Mol Cell Biol 2013, 14, 297.
         | Molecular mechanisms underlying RB protein function.Crossref | GoogleScholarGoogle Scholar |

[95]  K Thalmeier, H Synovzik, R Mertz, EL Winnacker, M Lipp, Nuclear factor E2F mediates basic transcription and trans-activation by E1a of the human MYC promoter. Genes Dev 1989, 3, 527.
         | Nuclear factor E2F mediates basic transcription and trans-activation by E1a of the human MYC promoter.Crossref | GoogleScholarGoogle Scholar |

[96]  RM Neto-Silva, S de Beco, LA Johnston, Evidence for a growth-stabilizing regulatory feedback mechanism between Myc and Yorkie, the Drosophila homolog of Yap. Dev Cell 2010, 19, 507.
         | Evidence for a growth-stabilizing regulatory feedback mechanism between Myc and Yorkie, the Drosophila homolog of Yap.Crossref | GoogleScholarGoogle Scholar |

[97]  A Kapoor, W Yao, H Ying, S Hua, A Liewen, Q Wang, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014, 158, 185.
         | Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer.Crossref | GoogleScholarGoogle Scholar |

[98]  U Ehmer, AF Zmoos, RK Auerbach, D Vaka, AJ Butte, MA Kay, et al. Organ size control is dominant over Rb family inactivation to restrict proliferation in vivo. Cell Rep 2014, 8, 371.
         | Organ size control is dominant over Rb family inactivation to restrict proliferation in vivo.Crossref | GoogleScholarGoogle Scholar |

[99]  NK Lytle, AG Barber, T Reya, Stem cell fate in cancer growth, progression and therapy resistance. Nat Rev Cancer 2018, 18, 669.
         | Stem cell fate in cancer growth, progression and therapy resistance.Crossref | GoogleScholarGoogle Scholar |

[100]  T Shibue, RA Weinberg, EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol 2017, 14, 611.
         | EMT, CSCs, and drug resistance: the mechanistic link and clinical implications.Crossref | GoogleScholarGoogle Scholar |

[101]  EM Morin-Kensicki, BN Boone, M Howell, JR Stonebraker, J Teed, JG Alb, et al. Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65. Mol Cell Biol 2006, 26, 77.
         | Defects in yolk sac vasculogenesis, chorioallantoic fusion, and embryonic axis elongation in mice with targeted disruption of Yap65.Crossref | GoogleScholarGoogle Scholar |

[102]  C Tamm, N Böwer, C Annerén, Regulation of mouse embryonic stem cell self-renewal by a Yes-YAP-TEAD2 signaling pathway downstream of LIF. J Cell Sci 2011, 124, 1136.
         | Regulation of mouse embryonic stem cell self-renewal by a Yes-YAP-TEAD2 signaling pathway downstream of LIF.Crossref | GoogleScholarGoogle Scholar |

[103]  I Lian, J Kim, H Okazawa, J Zhao, B Zhao, J Yu, et al. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation. Genes Dev 2010, 24, 1106.
         | The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation.Crossref | GoogleScholarGoogle Scholar |

[104]  D Yimlamai, C Christodoulou, GG Galli, K Yanger, B Pepe-Mooney, B Gurung, et al. Hippo pathway activity influences liver cell fate. Cell 2014, 157, 1324.
         | Hippo pathway activity influences liver cell fate.Crossref | GoogleScholarGoogle Scholar |

[105]  J Hao, Y Zhang, D Jing, Y Li, J Li, Z Zhao, Role of Hippo signaling in cancer stem cells. J Cell Physiol 2014, 229, 266.
         | Role of Hippo signaling in cancer stem cells.Crossref | GoogleScholarGoogle Scholar |

[106]  JS Mo, HW Park, KL Guan, The Hippo signaling pathway in stem cell biology and cancer. EMBO Rep 2014, 15, 642.
         | The Hippo signaling pathway in stem cell biology and cancer.Crossref | GoogleScholarGoogle Scholar |

[107]  M Cordenonsi, F Zanconato, L Azzolin, M Forcato, A Rosato, C Frasson, et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 2011, 147, 759.
         | The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells.Crossref | GoogleScholarGoogle Scholar |

[108]  A Fernandez-L, PA Northcott, J Dalton, C Fraga, D Ellison, S Angers, et al. YAP1 is amplified and up-regulated in hedgehog-associated medulloblastomas and mediates Sonic hedgehog-driven neural precursor proliferation. Genes Dev 2009, 23, 2729.
         | YAP1 is amplified and up-regulated in hedgehog-associated medulloblastomas and mediates Sonic hedgehog-driven neural precursor proliferation.Crossref | GoogleScholarGoogle Scholar |

[109]  U Basu-Roy, NS Bayin, K Rattanakorn, E Han, DG Placantonakis, A Mansukhani, et al. Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells. Nat Commun 2015, 6, 6411.
         | Sox2 antagonizes the Hippo pathway to maintain stemness in cancer cells.Crossref | GoogleScholarGoogle Scholar |

[110]  P Mehlen, A Puisieux, Metastasis: a question of life or death. Nat Rev Cancer 2006, 6, 449.
         | Metastasis: a question of life or death.Crossref | GoogleScholarGoogle Scholar |

[111]  N Sethi, Y Kang, Notch signalling in cancer progression and bone metastasis. Br J Cancer 2011, 105, 1805.
         | Notch signalling in cancer progression and bone metastasis.Crossref | GoogleScholarGoogle Scholar |

[112]  K VanderVorst, CA Dreyer, SE Konopelski, H Lee, H-YH Ho, KL Carraway III, Wnt/PCP signaling contribution to carcinoma collective cell migration and metastasis. Cancer Res 2019, 79, 1719.
         | Wnt/PCP signaling contribution to carcinoma collective cell migration and metastasis.Crossref | GoogleScholarGoogle Scholar |

[113]  Z Yao, L Han, Y Chen, F He, B Sun, S Kamar, et al. Hedgehog signalling in the tumourigenesis and metastasis of osteosarcoma, and its potential value in the clinical therapy of osteosarcoma. Cell Death Dis 2018, 9, 701.
         | Hedgehog signalling in the tumourigenesis and metastasis of osteosarcoma, and its potential value in the clinical therapy of osteosarcoma.Crossref | GoogleScholarGoogle Scholar |

[114]  Y Drabsch, P ten Dijke, TGF-β signalling and its role in cancer progression and metastasis. Cancer Metastasis Rev 2012, 31, 553.
         | TGF-β signalling and its role in cancer progression and metastasis.Crossref | GoogleScholarGoogle Scholar |

[115]  JSA Warren, Y Xiao, JM Lamar, YAP/TAZ activation as a target for treating metastatic cancer. Cancers (Basel) 2018, 10, 115.
         | YAP/TAZ activation as a target for treating metastatic cancer.Crossref | GoogleScholarGoogle Scholar |

[116]  XY Lin, XP Zhang, JH Wu, XS Qiu, EH Wang, Expression of LATS1 contributes to good prognosis and can negatively regulate YAP oncoprotein in non-small-cell lung cancer. Tumour Biol 2014, 35, 6435.
         | Expression of LATS1 contributes to good prognosis and can negatively regulate YAP oncoprotein in non-small-cell lung cancer.Crossref | GoogleScholarGoogle Scholar |

[117]  Y Takahashi, Y Miyoshi, C Takahata, N Irahara, T Taguchi, Y Tamaki, et al. Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers. Clin Cancer Res 2005, 11, 1380.
         | Down-regulation of LATS1 and LATS2 mRNA expression by promoter hypermethylation and its association with biologically aggressive phenotype in human breast cancers.Crossref | GoogleScholarGoogle Scholar |

[118]  K Liang, G Zhou, Q Zhang, J Li, C Zhang, Expression of hippo pathway in colorectal cancer. Saudi J Gastroenterol 2014, 20, 188.
         | Expression of hippo pathway in colorectal cancer.Crossref | GoogleScholarGoogle Scholar |

[119]  GX Zhou, XY Li, Q Zhang, K Zhao, CP Zhang, CH Xue, et al. Effects of the hippo signaling pathway in human gastric cancer. Asian Pac J Cancer Prev 2013, 14, 5199.
         | Effects of the hippo signaling pathway in human gastric cancer.Crossref | GoogleScholarGoogle Scholar |

[120]  JM Lamar, P Stern, H Liu, JW Schindler, ZG Jiang, RO Hynes, The Hippo pathway target, YAP, promotes metastasis through its TEAD-interaction domain. Proc Natl Acad Sci U S A 2012, 109, E2441.
         | The Hippo pathway target, YAP, promotes metastasis through its TEAD-interaction domain.Crossref | GoogleScholarGoogle Scholar |

[121]  Z Zhou, JS Zhu, CP Gao, LP Li, C Zhou, H Wang, et al. siRNA targeting YAP gene inhibits gastric carcinoma growth and tumor metastasis in SCID mice. Oncol Lett 2016, 11, 2806.
         | siRNA targeting YAP gene inhibits gastric carcinoma growth and tumor metastasis in SCID mice.Crossref | GoogleScholarGoogle Scholar |

[122]  HH Ling, CC Kuo, BX Lin, YH Huang, CW Lin, Elevation of YAP promotes the epithelial-mesenchymal transition and tumor aggressiveness in colorectal cancer. Exp Cell Res 2017, 350, 218.
         | Elevation of YAP promotes the epithelial-mesenchymal transition and tumor aggressiveness in colorectal cancer.Crossref | GoogleScholarGoogle Scholar |

[123]  Y Qu, L Zhang, J Wang, P Chen, Y Jia, C Wang, et al. Yes-associated protein (YAP) predicts poor prognosis and regulates progression of esophageal squamous cell cancer through epithelial-mesenchymal transition. Exp Ther Med 2019, 18, 2993.
         | Yes-associated protein (YAP) predicts poor prognosis and regulates progression of esophageal squamous cell cancer through epithelial-mesenchymal transition.Crossref | GoogleScholarGoogle Scholar |

[124]  M Zhou, Y Zhang, H Wei, J He, D Wang, B Chen, et al. Furin inhibitor D6R suppresses epithelial-mesenchymal transition in SW1990 and PaTu8988 cells via the Hippo-YAP signaling pathway. Oncol Lett 2018, 15, 3192.
         | Furin inhibitor D6R suppresses epithelial-mesenchymal transition in SW1990 and PaTu8988 cells via the Hippo-YAP signaling pathway.Crossref | GoogleScholarGoogle Scholar |

[125]  X Tang, Y Sun, G Wan, J Sun, J Sun, C Pan, Knockdown of YAP inhibits growth in Hep-2 laryngeal cancer cells via epithelial-mesenchymal transition and the Wnt/β-catenin pathway. BMC Cancer 2019, 19, 654.
         | Knockdown of YAP inhibits growth in Hep-2 laryngeal cancer cells via epithelial-mesenchymal transition and the Wnt/β-catenin pathway.Crossref | GoogleScholarGoogle Scholar |

[126]  S Visser, X Yang, Identification of LATS transcriptional targets in HeLa cells using whole human genome oligonucleotide microarray. Gene 2010, 449, 22.
         | Identification of LATS transcriptional targets in HeLa cells using whole human genome oligonucleotide microarray.Crossref | GoogleScholarGoogle Scholar |

[127]  Z Li, Y Wang, Y Zhu, C Yuan, D Wang, W Zhang, et al. The Hippo transducer TAZ promotes epithelial to mesenchymal transition and cancer stem cell maintenance in oral cancer. Mol Oncol 2015, 9, 1091.
         | The Hippo transducer TAZ promotes epithelial to mesenchymal transition and cancer stem cell maintenance in oral cancer.Crossref | GoogleScholarGoogle Scholar |

[128]  Y Zhang, P Xie, X Wang, P Pan, Y Wang, H Zhang, et al. YAP promotes migration and invasion of human glioma cells. J Mol Neurosci 2018, 64, 262.
         | YAP promotes migration and invasion of human glioma cells.Crossref | GoogleScholarGoogle Scholar |

[129]  SW Chan, CJ Lim, K Guo, CP Ng, I Lee, W Hunziker, et al. A role for TAZ in migration, invasion, and tumorigenesis of breast cancer cells. Cancer Res 2008, 68, 2592.
         | A role for TAZ in migration, invasion, and tumorigenesis of breast cancer cells.Crossref | GoogleScholarGoogle Scholar |

[130]  A Dey, X Varelas, KL Guan, Targeting the Hippo pathway in cancer, fibrosis, wound healing and regenerative medicine. Nat Rev Drug Discov 2020, 19, 480.
         | Targeting the Hippo pathway in cancer, fibrosis, wound healing and regenerative medicine.Crossref | GoogleScholarGoogle Scholar |

[131]  Y Liu-Chittenden, B Huang, JS Shim, Q Chen, SJ Lee, RA Anders, et al. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev 2012, 26, 1300.
         | Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP.Crossref | GoogleScholarGoogle Scholar |

[132]  C Wang, X Zhu, W Feng, Y Yu, K Jeong, W Guo, et al. Verteporfin inhibits YAP function through up-regulating 14-3-3σ sequestering YAP in the cytoplasm. Am J Cancer Res 2016, 6, 27.

[133]  H Zhang, SK Ramakrishnan, D Triner, B Centofanti, D Maitra, B Győrffy, et al. Tumor-selective proteotoxicity of verteporfin inhibits colon cancer progression independently of YAP1. Sci Signal 2015, 8, ra98.
         | Tumor-selective proteotoxicity of verteporfin inhibits colon cancer progression independently of YAP1.Crossref | GoogleScholarGoogle Scholar |

[134]  JG Shamul, SR Shah, J Kim, P Schiapparelli, CA Vazquez-Ramos, BJ Lee, et al. Verteporfin-loaded anisotropic poly(beta-amino ester)-based micelles demonstrate brain cancer-selective cytotoxicity and enhanced pharmacokinetics. Int J Nanomedicine 2019, 14, 10047.
         | Verteporfin-loaded anisotropic poly(beta-amino ester)-based micelles demonstrate brain cancer-selective cytotoxicity and enhanced pharmacokinetics.Crossref | GoogleScholarGoogle Scholar |

[135]  JM Houle, A Strong, Clinical pharmacokinetics of verteporfin. J Clin Pharmacol 2002, 42, 547.
         | Clinical pharmacokinetics of verteporfin.Crossref | GoogleScholarGoogle Scholar |

[136]  MT Huggett, M Jermyn, A Gillams, R Illing, S Mosse, M Novelli, et al. Phase I/II study of verteporfin photodynamic therapy in locally advanced pancreatic cancer. Br J Cancer 2014, 110, 1698.
         | Phase I/II study of verteporfin photodynamic therapy in locally advanced pancreatic cancer.Crossref | GoogleScholarGoogle Scholar |

[137]  W Lu, J Wang, Y Li, H Tao, H Xiong, F Lian, et al. Discovery and biological evaluation of vinylsulfonamide derivatives as highly potent, covalent TEAD autopalmitoylation inhibitors. Eur J Med Chem 2019, 184, 111767.
         | Discovery and biological evaluation of vinylsulfonamide derivatives as highly potent, covalent TEAD autopalmitoylation inhibitors.Crossref | GoogleScholarGoogle Scholar |

[138]  A Kaneda, T Seike, T Danjo, T Nakajima, N Otsubo, D Yamaguchi, et al. The novel potent TEAD inhibitor, K-975, inhibits YAP1/TAZ-TEAD protein-protein interactions and exerts an anti-tumor effect on malignant pleural mesothelioma. Am J Cancer Res 2020, 10, 4399.

[139]  X Deng, L Fang, VGLL4 is a transcriptional cofactor acting as a novel tumor suppressor via interacting with TEADs. Am J Cancer Res 2018, 8, 932.

[140]  S Jiao, H Wang, Z Shi, A Dong, W Zhang, X Song, et al. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 2014, 25, 166.
         | A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer.Crossref | GoogleScholarGoogle Scholar |

[141]  S Jiao, C Li, Q Hao, H Miao, L Zhang, L Li, et al. VGLL4 targets a TCF4–TEAD4 complex to coregulate Wnt and Hippo signalling in colorectal cancer. Nat Commun 2017, 8, 14058.
         | VGLL4 targets a TCF4–TEAD4 complex to coregulate Wnt and Hippo signalling in colorectal cancer.Crossref | GoogleScholarGoogle Scholar |

[142]  W Zhang, Y Gao, P Li, Z Shi, T Guo, F Li, et al. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res 2014, 24, 331.
         | VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex.Crossref | GoogleScholarGoogle Scholar |

[143]  PC Calses, JJ Crawford, JR Lill, A Dey, Hippo pathway in cancer: aberrant regulation and therapeutic opportunities. Trends Cancer 2019, 5, 297.
         | Hippo pathway in cancer: aberrant regulation and therapeutic opportunities.Crossref | GoogleScholarGoogle Scholar |

[144]  HW Park, KL Guan, Regulation of the Hippo pathway and implications for anticancer drug development. Trends Pharmacol Sci 2013, 34, 581.
         | Regulation of the Hippo pathway and implications for anticancer drug development.Crossref | GoogleScholarGoogle Scholar |

[145]  N Yang, CD Morrison, P Liu, J Miecznikowski, W Bshara, S Han, et al. TAZ induces growth factor-independent proliferation through activation of EGFR ligand amphiregulin. Cell Cycle 2012, 11, 2922.
         | TAZ induces growth factor-independent proliferation through activation of EGFR ligand amphiregulin.Crossref | GoogleScholarGoogle Scholar |

[146]  C He, X Lv, G Hua, SM Lele, S Remmenga, J Dong, et al. YAP forms autocrine loops with the ERBB pathway to regulate ovarian cancer initiation and progression. Oncogene 2015, 34, 6040.
         | YAP forms autocrine loops with the ERBB pathway to regulate ovarian cancer initiation and progression.Crossref | GoogleScholarGoogle Scholar |

[147]  T Ando, N Arang, Z Wang, DE Costea, X Feng, Y Goto, et al. EGFR regulates the Hippo pathway by promoting the tyrosine phosphorylation of MOB1. Commun Biol 2021, 4, 1237.
         | EGFR regulates the Hippo pathway by promoting the tyrosine phosphorylation of MOB1.Crossref | GoogleScholarGoogle Scholar |

[148]  J Zhang, JY Ji, M Yu, M Overholtzer, GA Smolen, R Wang, et al. YAP-dependent induction of amphiregulin identifies a non-cell-autonomous component of the Hippo pathway. Nat Cell Biol 2009, 11, 1444.
         | YAP-dependent induction of amphiregulin identifies a non-cell-autonomous component of the Hippo pathway.Crossref | GoogleScholarGoogle Scholar |

[149]  DO Wennmann, B Vollenbröker, AK Eckart, J Bonse, F Erdmann, DA Wolters, et al. The Hippo pathway is controlled by Angiotensin II signaling and its reactivation induces apoptosis in podocytes. Cell Death Dis 2014, 5, e1519.
         | The Hippo pathway is controlled by Angiotensin II signaling and its reactivation induces apoptosis in podocytes.Crossref | GoogleScholarGoogle Scholar |

[150]  I Fujiwara, ME Zweifel, N Courtemanche, TD Pollard, Latrunculin A accelerates actin filament depolymerization in addition to sequestering actin monomers. Curr Biol 2018, 28, 3183.
         | Latrunculin A accelerates actin filament depolymerization in addition to sequestering actin monomers.Crossref | GoogleScholarGoogle Scholar |

[151]  KA Sayed, MA Khanfar, HM Shallal, A Muralidharan, B Awate, DT Youssef, et al. Latrunculin A and its C-17-O-carbamates inhibit prostate tumor cell invasion and HIF-1 activation in breast tumor cells. J Nat Prod 2008, 71, 396.
         | Latrunculin A and its C-17-O-carbamates inhibit prostate tumor cell invasion and HIF-1 activation in breast tumor cells.Crossref | GoogleScholarGoogle Scholar |

[152]  H Konishi, S Kikuchi, T Ochiai, H Ikoma, T Kubota, D Ichikawa, et al. Latrunculin A has a strong anticancer effect in a peritoneal dissemination model of human gastric cancer in mice. Anticancer Res 2009, 29, 2091.

[153]  M Kim, K Song, EJ Jin, J Sonn, Staurosporine and cytochalasin D induce chondrogenesis by regulation of actin dynamics in different way. Exp Mol Med 2012, 44, 521.
         | Staurosporine and cytochalasin D induce chondrogenesis by regulation of actin dynamics in different way.Crossref | GoogleScholarGoogle Scholar |

[154]  J Yu, A Alharbi, H Shan, Y Hao, B Snetsinger, MJ Rauh, et al. TAZ induces lung cancer stem cell properties and tumorigenesis by up-regulating ALDH1A1. Oncotarget 2017, 8, 38426.
         | TAZ induces lung cancer stem cell properties and tumorigenesis by up-regulating ALDH1A1.Crossref | GoogleScholarGoogle Scholar |

[155]  MZ Xu, SW Chan, AM Liu, KF Wong, ST Fan, J Chen, et al. AXL receptor kinase is a mediator of YAP-dependent oncogenic functions in hepatocellular carcinoma. Oncogene 2011, 30, 1229.
         | AXL receptor kinase is a mediator of YAP-dependent oncogenic functions in hepatocellular carcinoma.Crossref | GoogleScholarGoogle Scholar |

[156]  E Ghiso, C Migliore, V Ciciriello, E Morando, A Petrelli, S Corso, et al. YAP-Dependent AXL Overexpression Mediates Resistance to EGFR Inhibitors in NSCLC. Neoplasia 2017, 19, 1012.
         | YAP-Dependent AXL Overexpression Mediates Resistance to EGFR Inhibitors in NSCLC.Crossref | GoogleScholarGoogle Scholar |

[157]  S Aveic, D Corallo, E Porcù, M Pantile, D Boso, C Zanon, et al. TP-0903 inhibits neuroblastoma cell growth and enhances the sensitivity to conventional chemotherapy. Eur J Pharmacol 2018, 818, 435.
         | TP-0903 inhibits neuroblastoma cell growth and enhances the sensitivity to conventional chemotherapy.Crossref | GoogleScholarGoogle Scholar |

[158]  G Wang, X Lu, P Dey, P Deng, CC Wu, S Jiang, et al. Targeting YAP-dependent MDSC infiltration impairs tumor progression. Cancer Discov 2016, 6, 80.
         | Targeting YAP-dependent MDSC infiltration impairs tumor progression.Crossref | GoogleScholarGoogle Scholar |

[159]  N Zhu, R Yang, X Wang, L Yuan, X Li, F Wei, et al. The Hippo signaling pathway: from multiple signals to the hallmarks of cancers. Acta Biochim Biophys Sin (Shanghai) 2023, 55, 1.
         | The Hippo signaling pathway: from multiple signals to the hallmarks of cancers.Crossref | GoogleScholarGoogle Scholar |

[160]  F Gibault, M Corvaisier, F Bailly, G Huet, P Melnyk, P Cotelle, Non-photoinduced biological properties of Verteporfin. Curr Med Chem 2016, 23, 1171.
         | Non-photoinduced biological properties of Verteporfin.Crossref | GoogleScholarGoogle Scholar |

[161]  EK Konstantinou, S Notomi, C Kosmidou, K Brodowska, A Al-Moujahed, F Nicolaou, et al. Verteporfin-induced formation of protein cross-linked oligomers and high molecular weight complexes is mediated by light and leads to cell toxicity. Sci Rep 2017, 7, 46581.
         | Verteporfin-induced formation of protein cross-linked oligomers and high molecular weight complexes is mediated by light and leads to cell toxicity.Crossref | GoogleScholarGoogle Scholar |

[162]  P Karpowicz, J Perez, N Perrimon, The Hippo tumor suppressor pathway regulates intestinal stem cell regeneration. Development 2010, 137, 4135.
         | The Hippo tumor suppressor pathway regulates intestinal stem cell regeneration.Crossref | GoogleScholarGoogle Scholar |

[163]  Y Qiao, J Chen, YB Lim, ML Finch-Edmondson, VP Seshachalam, L Qin, et al. YAP regulates actin dynamics through ARHGAP29 and promotes metastasis. Cell Rep 2017, 19, 1495.
         | YAP regulates actin dynamics through ARHGAP29 and promotes metastasis.Crossref | GoogleScholarGoogle Scholar |

[164]  D Lai, X Yang, BMP4 is a novel transcriptional target and mediator of mammary cell migration downstream of the Hippo pathway component TAZ. Cell Signal 2013, 25, 1720.
         | BMP4 is a novel transcriptional target and mediator of mammary cell migration downstream of the Hippo pathway component TAZ.Crossref | GoogleScholarGoogle Scholar |