Next Article in Journal
Oral Dysbiosis in Pancreatic Cancer and Liver Cirrhosis: A Review of the Literature
Previous Article in Journal
Differences in Anxiety Levels of Various Murine Models in Relation to the Gut Microbiota Composition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Yes-Associated Protein (YAP) in Regulating Programmed Death-Ligand 1 (PD-L1) in Thoracic Cancer

1
Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94115, USA
2
Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
*
Author to whom correspondence should be addressed.
Biomedicines 2018, 6(4), 114; https://doi.org/10.3390/biomedicines6040114
Submission received: 10 November 2018 / Revised: 29 November 2018 / Accepted: 4 December 2018 / Published: 7 December 2018

Abstract

:
The programmed death-ligand 1(PD-L1)/PD-1 pathway is an immunological checkpoint in cancer cells. The binding of PD-L1 and PD-1 promotes T-cell tolerance and helps tumor cells escape from host immunity. Immunotherapy targeting the PD-L1/PD-1 axis has been developed as an anti-cancer therapy and used in treating advanced human non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). Yes-associated protein (YAP) is a key mediator of the Hippo/YAP signaling pathway, and plays important roles in promoting cancer development, drug resistance and metastasis in human NSCLC and MPM. YAP has been suggested as a new therapeutic target in NSCLC and MPM. The role of YAP in regulating tumor immunity such as PD-L1 expression has just begun to be explored, and the correlation between YAP-induced tumorigenesis and host anti-tumor immune responses is not well known. Here, we review recent studies investigating the correlation between YAP and PD-L1 and demonstrating the mechanism by which YAP regulates PD-L1 expression in human NSCLC and MPM. Future work should focus on the interactions between Hippo/YAP signaling pathways and the immune checkpoint PD-L1/PD-1 pathway. The development of new synergistic drugs for immune checkpoint PD-L1/PD-1 blockade in NSCLC and MPM is warranted.

Graphical Abstract

1. Programmed Death-Ligand 1 in Non-Small Cell Lung Cancer and Malignant Pleural Mesothelioma

Programmed death-ligand 1 (PD-L1) (also known as B7-H1 or CD274) is a type I transmembrane surface glycoprotein that belongs to the B7 family of costimulatory molecules. PD-L1 is a ligand of programmed cell death protein 1 (PD-1; also known as CD279), which is one of the co-inhibitory receptors expressed on the surface of antigen-stimulated T cells. The PD-L1/PD-1 pathway is an immunological checkpoint, and the binding of PD-L1 and PD-1 promotes T-cell tolerance and escape from host immunity through inhibiting CD8+ T-cell survival, effector function, and inducing Fas-mediated T-cell apoptosis [1,2]. PD-L1 is expressed in hematopoietic cells including T cells, B cells, macrophages, dendritic cells, and mast cells. PD-L1 is also broadly expressed in non-hematopoietic healthy tissue cells including vascular endothelial cells, pancreatic islet cells, astrocytes, and corneal epithelial and endothelial cells [3,4,5]. PD-L1 is expressed in cancer cells, and cancers can engage the immune checkpoint PD-L1/PD-1 axis to escape antitumor immune responses. Therefore, the PD-L1/PD-1 immune checkpoint blockade has been developed as an anti-cancer therapy [6,7,8]. PD-L1 has been shown to be expressed in human non-small-cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM) [9,10,11,12,13,14,15,16]. Anti-PD-L1/PD-1 inhibitors have used clinically to treat advanced NSCLC and MPM [11,15,16,17,18].
Currently, there are 2 anti-PD-1 (pembrolizumab and nivolumab) and 2 anti-PD-L1 (atezolizumab and durvalumab) inhibitors used in treating NSCLC. The efficacy of all 4 was shown in phase III clinical trials—all 4 have shown promising results, with ~30% of NSCLC responding [18,19,20,21,22,23,24,25].
MPM is a very aggressive thoracic cancer, and unresectable MPM has a poor prognosis with a median survival of about 12 months. Treatment options for advanced unresectable MPM are very limited [26,27,28,29]. Immune checkpoint inhibitors targeting the PD-L1/PD-1 pathway have recently been used to treat advanced MPM, and the efficacy is being investigated in several clinical trials. Some patients with advanced MPM benefited from immunotherapy with anti-PD-L1/PD-1 inhibitors [14,15,16,17,30,31,32,33]. A phase II clinical trial (NCT02628067; KEYNOTE-158) to investigate the efficacy of pembrolizumab (anti-PD-1) in advanced solid tumors, including MPM, is ongoing; patients are encouraged to participate in this trial to facilitate advancement in the treatment of MPM. The rationale for immune checkpoint PD-L1/PD-1 blockade is summarized in Figure 1.

2. Yes-Associated Protein in Human NSCLC and MPM

YAP (yes-associated protein) is the main downstream effector of the Hippo (also known as the Salvador-Warts-Hippo) signaling pathway. YAP is negatively regulated by upstream components of the Hippo pathway and while that pathway is activated, YAP will be sequestered by Hippo kinase in the cytoplasm and degraded. Conversely, when the Hippo pathway is inactivated, YAP will translocate into the nucleus and activate transcription of downstream genes by forming complexes with transcriptional enhancer factors (TEF; also known as TEAD). In normal cells, Hippo/YAP plays a key role in regulating organ size [34,35]. Overexpression of YAP has been found in many cancers because of abnormal amplification, loss of Hippo signaling by mutation, and/or downregulation of the core Hippo components. YAP has shown a correlation with stem cell renewal and differentiation, a crucial step in oncogenic transformation [36], and was reported to promote cancer development in various cancers [37,38,39,40].
YAP was identified in human NSCLC, and is correlated with drug resistance, tumorigenesis, cancer progression and metastasis [41,42,43,44,45]. For instance, we previously reported crosstalk between Hippo/YAP and epidermal growth factor receptor (EGFR)-mitogen-activated protein kinase (MAPK) signaling pathways in human NSCLC and found that YAP could promote erlotinib resistance in EGFR mutant NSCLC cells [43,44]. Two prior studies showed that YAP appears to take over K-ras as a cancer driver in NSCLC cells harboring K-ras mutations [46,47]. We also recently reported a key role for YAP in promoting brain metastasis in NSCLC H2030-BrM3 (K-rasG12C mutation) cells and that inhibition of YAP can suppress brain metastasis in a murine model [48].
YAP overexpression was also found in human MPM and reported to correlate with tumorigenesis and cancer development. The mutations of Hippo kinase genes including neurofibromatosis 2 (NF2), large tumor suppressor homolog 1 (LATS1), LATS2, and mammalian sterile-20 like kinase 1 (MST1) frequently occur in human MPM [49,50]. The mutations alter the activation of Hippo kinases, which inhibit YAP and lead to increased YAP protein expression [49,50]. We previously reported that inhibition of YAP suppresses the growth, migration and invasion of human MPM cells that have strong YAP expression, and suppresses expression of its downstream genes. Thus, YAP has been suggested as a therapeutic target in advanced unresectable MPM [49,50,51]. The regulation of Hippo/YAP signaling pathway in NSCLC and MPM is summarized in Figure 2.

3. YAP Regulates PD-L1 Expression in Human NSCLC and MPM

Though tumor PD-L1 expression has been used as a predictive biomarker for anti-PD-1/PD-L1 immunotherapy, more than 50% of tumors with strong PD-L1 expression did not respond to PD-1/PD-L1 inhibitors [10,52,53]. A better understanding of the mechanism of how tumor PD-L1 expression is regulated may help identify biomarkers and develop therapeutic strategies for clinical use.
The role of YAP in cancer immunity has just begun to be studied [54]. YAP is a negative regulator of innate immunity through its interaction with interferon regulatory factor 3 [55]. Recent studies found that YAP regulates tumor-associated immune cells like myeloid-derived suppressor cells, tumor-associated macrophages and regulatory T cells, and indicated that YAP is involved in the regulation of tumor-associated immune cells and the immune checkpoint [56,57,58]. However, the role of YAP in the context of reciprocal interactions between cancer cells and host anti-immune responses remains unclear because of the complexity of tumorigenesis and immune regulation. In two recent studies, we investigated the role of YAP in regulating PD-L1 expression in human NSCLC and MPM [59,60]. First, in human NSCLC and MPM tumor samples, immunohistochemistry showed that positive nuclear YAP staining was significantly associated with positive PD-L1 expression. Second, NSCLC and MPM cell lines (H460, SKLU-1, H1299, H2052 and 211H) with increased PD-L1 protein and mRNA expression had a lower p-YAP/YAP ratio and increased GTIIC reporter activity of the Hippo pathway compared to other NSCLC and MPM cell lines with low PD-L1 protein and mRNA expression. Third, YAP knockdown by small interfering RNAs (siRNAs) decreased the protein and mRNA levels of PD-L1 in NSCLC and MPM cell lines (H460, SKLU-1, H1299, H2052 and 211H). Fourth, forced overexpression of the YAP gene increased the PD-L1 protein expression level in A549 (NSCLC cell line) and H2452 (MPM cell line) cells, which have low YAP and PD-L1 expression. In addition, chromatin immunoprecipitation (ChIP) assays using a YAP-specific monoclonal antibody showed the precipitation of the PD-L1 enhancer region encompassing two putative TEAD binding sites in NSCLC and MPM cell lines. Our findings from those two studies indicate that YAP regulates the transcription of PD-L1 in human NSCLC and MPM [59,60]. A recent study conducted by van Rensburg et al. showed that another main mediator of the Hippo pathway, TAZ, promotes immune evasion in human NSCLC and breast cancer through PD-L1, and the experimental findings in this study also suggested that YAP is involved in the regulation of PD-L1 [61,62]. Another recent study demonstrated that YAP also regulates PD-L1 expression in EGFR-TKI-resistant NSCLC [63]. Two other studies concluded that the EGFR pathway regulates PD-L1 expression in EGFR mutant NSCLC [64,65]. We reported crosstalk between YAP and EGFR/Extracellular signal-regulated kinase (ERK) signaling pathways in human EGFR mutant NSCLC cells, and demonstrated that inhibition of the EGFR/ERK signaling pathway decreased YAP expression in human NSCLC cells [43,44]. These findings suggest that EGFR may be involved in regulating PD-L1 through the activation or inhibition of YAP. The mechanism by which YAP regulates PD-L1 expression in human NSCLC and MPM according to our findings is summarized in Figure 3.

4. Future perspectives: The Interactions between YAP Signaling Pathways and Immune Checkpoints

To date, the work to investigate the interaction between the YAP signaling pathway and the PD-L1/PD-1 immune checkpoint has been mostly done in cell lines, and only a few cohort patient samples showed the relevance of this pathway [58,59,60,61,62]. Anti-PD-L1/PD1 inhibitors have just been approved, and will be widely used in treating advanced NSCLC, and are investigated in clinical trials for treating advanced MPM [15,16,17,18,19,20,21,22,23,24,25]. In the future, more prospective patient samples can be gathered to investigate the molecular relevance between YAP and PD-L1.
In addition, the issue of establishing an ideal in vivo small animal model for investigating the interaction between human tumor and immune system interaction emerges. Recently, a humanized mouse model was generated by transplanting human CD34+ hematopoietic progenitor and stem cells into immunodeficient mice, which restores human hematopoietic and immune systems in a mouse model [66,67]. A NSCLC patient-derived xenograft humanized mouse model used to test the efficacy of anti-PD-1 immunotherapy was developed recently [67]. Several drugs potentially regulate YAP activity, including verteporfin, dasatinib, cyclin-dependent kinase 1 inhibitors, and cyclin-dependent kinase 9 (CDK9) inhibitors [50,68,69,70,71]. The combination of inhibitors of YAP activity with other drugs has shown some promising anti-tumor effects in NSCLC mouse models [72]. Recent studies suggest that the YAP inhibitor, verteporfin, and CDK9 inhibitors, may synergize with anti-PD-1/PD-L1 immunotherapy in their anti-tumor effects [73,74]. Future work to investigate the efficacy of YAP inhibitors in combination with anti-PD-L1/PD-1 inhibitors in NSCLC and MPM by using humanized mouse models is feasible.

5. Conclusions

Our review indicates that YAP plays an important role in partially regulating the tumor immune checkpoint PD-L1/PD-1 pathway in human NSCLC and MPM. Future work should focus on the development of new synergistic drugs targeting YAP for immune checkpoint PD-L1/PD-1 blockade in NSCLC and MPM.

Author Contributions

Conception and design: P.-C-H., L.Y.; Acquisition of data: P.-C-H., D.M.J.; Writing, review, and/or revision of the manuscript: P.-C-H., C.-T.Y, L.Y.; Administrative, technical, or material support (organizing data): P.-C-H., D.M.J.

Funding

This study was supported by the National Institutes of Health (NIH; Grant No. R01 CA140654, to LY) and Chang-Gung Medical Research Project (CMRP No. CMRPG3G1661 to PCH).

Acknowledgments

We are grateful for support from the Kazan McClain Partner’s Foundation; the Estate of Robert Griffiths; the Jeffrey and Karen Peterson Family Foundation; Paul and Michelle Zygielbaum; the Estate of Norman Mancini; and the Barbara Isackson Lung Cancer Research Fund. We thank Pamela Derish at Department of Surgery of UCSF for editorial assistance with this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed]
  2. Okazaki, T.; Honjo, T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006, 27, 195–201. [Google Scholar] [CrossRef] [PubMed]
  3. Ishida, M.; Iwai, Y.; Tanaka, Y.; Okazaki, T.; Freeman, G.J.; Minato, N.; Honjo, T. Differential expression of PD-L1 and PD-L2, ligands for an inhibitory receptor PD-1, in the cells of lymphohematopoietic tissues. Immunol. Lett. 2002, 84, 57–62. [Google Scholar] [CrossRef]
  4. Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [PubMed]
  6. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zielinski, C.; Knapp, S.; Mascaux, C.; Hirsch, F. Rationale for targeting the immune system through checkpoint molecule blockade in the treatment of non-small-cell lung cancer. Ann. Oncol. 2013, 24, 1170–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef] [Green Version]
  9. Cooper, W.A.; Tran, T.; Vilain, R.E.; Madore, J.; Selinger, C.I.; Kohonen-Corish, M.; Yip, P.; Yu, B.; O’Toole, S.A.; McCaughan, B.C.; et al. PD-L1 expression is a favorable prognostic factor in early stage non-small cell carcinoma. Lung Cancer 2015, 89, 181–188. [Google Scholar] [CrossRef]
  10. Passiglia, F.; Bronte, G.; Bazan, V.; Natoli, C.; Rizzo, S.; Galvano, A.; Listì, A.; Cicero, G.; Rolfo, C.; Santini, D.; et al. PD-L1 expression as predictive biomarker in patients with NSCLC: A pooled analysis. Oncotarget 2016, 7, 19738–19747. [Google Scholar] [CrossRef]
  11. Ogunleyea, F.; Blankenshipa, L.; Millisorb, V.; Anderson, J.; Jaiyesimi, I. Programmed cell death-1/Programmed cell death ligand-1(PD-1/PD-L1) inhibitors, heralding a new era of immunotherapy in the management of advanced Non-Small Cell Lung Cancer (NSCLC). Cancer Treat. Res. Commun. 2017, 12, 6–13. [Google Scholar] [CrossRef]
  12. Tang, D.; Zhao, D.; Wu, Y.; Yao, R.; Zhou, L.; Lu, L.; Gao, W.; Sun, Y. The miR-3127-5p/p-STAT3 axis up-regulates PD-L1 inducing chemoresistance in non-small-cell lung cancer. J. Cell. Mol. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  13. Tunger, A.; Kießler, M.; Wehner, R.; Temme, A.; Meier, F.; Bachmann, M.; Schmitz, M. Immune Monitoring of Cancer Patients Prior to and During CTLA-4 or PD-1/PD-L1 Inhibitor Treatment. Biomedicines 2018, 6, 26. [Google Scholar] [CrossRef] [PubMed]
  14. Calabrò, L.; Ceresoli, G.L.; D’Incecco, A.; Scherpereel, A.; Aerts, J.; Maio, M. Immune checkpoint therapy of mesothelioma: Pre-clinical bases and clinical evidences. Cytokine Growth Factor Rev. 2017. [Google Scholar] [CrossRef] [PubMed]
  15. Dozier, J.; Zheng, H.; Adusumilli, P.S. Immunotherapy for malignant pleural mesothelioma: Current status and future directions. Transl. Lung Cancer Res. 2017, 6, 315–324. [Google Scholar] [CrossRef] [PubMed]
  16. Cedrés, S.; Ponce-Aix, S.; Zugazagoitia, J.; Sansano, I.; Enguita, A.; Navarro-Mendivil, A.; Martinez-Marti, A.; Martinez, P.; Felip, E. Analysis of expression of programmed cell death 1 ligand 1 (PD-L1) in malignant pleural mesothelioma (MPM). PLoS ONE 2015, 10, e0121071. [Google Scholar] [CrossRef] [PubMed]
  17. Dumoulin, D.W.; Aerts, J.G.; Cornelissen, R. Is immunotherapy a viable option in treating mesothelioma? Future Oncol. 2017, 13, 1747–1750. [Google Scholar] [CrossRef] [Green Version]
  18. Sui, H.; Ma, N.; Wang, Y.; Li, H.; Liu, X.; Su, Y.; Yang, J. Anti-PD-1/PD-L1 Therapy for Non-Small-Cell Lung Cancer: Toward Personalized Medicine and Combination Strategies. J. Immunol. Res. 2018, 2018, 6984948. [Google Scholar] [CrossRef]
  19. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
  20. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef]
  21. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
  22. Horn, L.; Spigel, D.R.; Vokes, E.E.; Holgado, E.; Ready, N.; Steins, M.; Poddubskaya, E.; Borghaei, H.; Felip, E.; Paz-Ares, L.; et al. Nivolumab Versus Docetaxel in Previously Treated Patients With Advanced Non-Small-Cell Lung Cancer: Two-Year Outcomes From Two Randomized, Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057). J. Clin. Oncol. 2017, 35, 3924–3933. [Google Scholar] [CrossRef] [PubMed]
  23. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  24. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
  25. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
  26. Flores, R.M.; Zakowski, M.; Venkatraman, E.; Krug, L.; Rosenzweig, K.; Dycoco, J.; Lee, C.; Yeoh, C.; Bains, M.; Rusch, V. Prognostic factors in the treatment of malignant pleural mesothelioma at a large tertiary referral center. J. Thorac. Oncol. 2007, 2, 957–965. [Google Scholar] [CrossRef]
  27. Gaudino, G.; Yang, H.; Carbone, M. HGF/Met Signaling Is a Key Player in Malignant Mesothelioma Carcinogenesis. Biomedicines 2014, 2, 327–344. [Google Scholar] [CrossRef] [Green Version]
  28. Hung, M.S.; Mao, J.H.; Xu, Z.; Yang, C.T.; Yu, J.S.; Harvard, C.; Lin, Y.C.; Bravo, D.T.; Jablons, D.M.; You, L. Cul4A is an oncogene in malignant pleural mesothelioma. J. Cell. Mol. Med. 2011, 15, 350–358. [Google Scholar] [CrossRef] [Green Version]
  29. Yang, Y.L.; Ni, J.; Hsu, P.C.; Mao, J.H.; Hsieh, D.; Xu, A.; Chan, G.; Au, A.; Xu, Z.; Jablons, D.M.; et al. Cul4A overexpression associated with Gli1 expression in malignant pleural mesothelioma. J. Cell. Mol. Med. 2015, 19, 2385–2396. [Google Scholar] [CrossRef] [Green Version]
  30. Alley, E.W.; Lopez, J.; Santoro, A.; Morosky, A.; Saraf, S.; Piperdi, B.; van Brummelen, E. Clinical safety and activity of pembrolizumab in patients with malignant pleural mesothelioma (KEYNOTE-028): Preliminary results from a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2017, 18, 623–630. [Google Scholar] [CrossRef]
  31. Chatwal, M.S.; Tanvetyanon, T. Malignant mesothelioma clinical trial combines immunotherapy drugs. Immunotherapy 2018, 10, 341–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Quispel-Janssen, J.; van der Noort, V.; de Vries, J.F.; Zimmerman, M.; Lalezari, F.; Thunnissen, E.; Monkhorst, K.; Schouten, R.; Schunselaar, L.; Disselhorst, M.; et al. Programmed Death 1 Blockade with Nivolumab in Patients with Recurrent Malignant Pleural Mesothelioma. J. Thorac. Oncol. 2018, 13, 1569–1576. [Google Scholar] [CrossRef] [PubMed]
  33. Metaxas, Y.; Rivalland, G.; Mauti, L.A.; Klingbiel, D.; Kao, S.; Schmid, S.; Nowak, A.K.; Gautschi, O.; Bartnick, T.; Hughes, B.G.; et al. Pembrolizumab as Palliative Immunotherapy in Malignant Pleural Mesothelioma. J. Thorac. Oncol. 2018, 13, 1784–1791. [Google Scholar] [CrossRef] [PubMed]
  34. Johnson, R.; Halder, G. The two faces of Hippo: Targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat. Rev. Drug Discov. 2014, 13, 63–79. [Google Scholar] [CrossRef] [PubMed]
  35. Guo, L.; Teng, L. YAP/TAZ for cancer therapy: Opportunities and challenges (review). Int. J. Oncol. 2015, 46, 1444–1452. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, L.; Feng, P.; Peng, A.; Qiu, X.; Zhu, X.; He, S.; Zhou, D. cAMP response element-binding protein and Yes-associated protein form a feedback loop that promotes neurite outgrowth. J. Cell. Mol. Med. 2018, 22, 374–381. [Google Scholar] [CrossRef] [PubMed]
  37. Yu, F.X.; Luo, J.; Mo, J.S.; Liu, G.; Kim, Y.C.; Meng, Z.; Zhao, B.; Peyman, G.; Ouyang, H.; Jiang, W.; et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 2014, 25, 822–830. [Google Scholar] [CrossRef]
  38. Feng, X.; Degese, M.S.; Iglesias-Bartolome, R.; Vaque, J.P.; Molinolo, A.A.; Rodrigues, M.; Zaidi, M.R.; Ksander, B.R.; Merlino, G.; Sodhi, A.; et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell 2014, 25, 831–845. [Google Scholar] [CrossRef]
  39. Zhang, Y.; Yan, S.; Chen, J.; Gan, C.; Chen, D.; Li, Y.; Wen, J.; Kremerskothen, J.; Chen, S.; Zhang, J.; et al. WWC2 is an independent prognostic factor and prevents invasion via Hippo signalling in hepatocellular carcinoma. J. Cell. Mol. Med. 2017, 21, 3718–3729. [Google Scholar] [CrossRef] [Green Version]
  40. Xia, J.; Zeng, M.; Zhu, H.; Chen, X.; Weng, Z.; Li, S. Emerging role of Hippo signalling pathway in bladder cancer. J. Cell. Mol. Med. 2018, 22, 4–15. [Google Scholar] [CrossRef]
  41. Ye, X.Y.; Luo, Q.Q.; Xu, Y.H.; Tang, N.W.; Niu, X.M.; Li, Z.M.; Shen, S.P.; Lu, S.; Chen, Z.W. 17-AAG suppresses growth and invasion of lung adenocarcinoma cells via regulation of the LATS1/YAP pathway. J. Cell. Mol. Med. 2015, 19, 651–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. 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] [PubMed] [Green Version]
  43. You, B.; Yang, Y.L.; Xu, Z.; Dai, Y.; Liu, S.; Mao, J.H.; Tetsu, O.; Li, H.; Jablons, D.M.; You, L. Inhibition of ERK1/2 down-regulates the Hippo/YAP signaling pathway in human NSCLC cells. Oncotarget 2015, 6, 4357–4368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Hsu, P.C.; You, B.; Yang, Y.L.; Zhang, W.Q.; Wang, Y.C.; Xu, Z.; Dai, Y.; Liu, S.; Yang, C.T.; Li, H.; et al. YAP promotes erlotinib resistance in human non-small cell lung cancer cells. Oncotarget 2016, 7, 51922–51933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Dai, Y.; Liu, S.; Zhang, W.Q.; Yang, Y.L.; Hang, P.; Wang, H.; Cheng, L.; Hsu, P.C.; Wang, Y.C.; Xu, Z.; et al. YAP1 regulates ABCG2 and cancer cell side population in human lung cancer cells. Oncotarget 2017, 8, 4096–4109. [Google Scholar] [CrossRef] [PubMed]
  46. Shao, D.D.; Xue, W.; Krall, E.B.; Bhutkar, A.; Piccioni, F.; Wang, X.; Schinzel, A.C.; Sood, S.; Rosenbluh, J.; Kim, J.W.; et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 2014, 158, 171–184. [Google Scholar] [CrossRef] [PubMed]
  47. Greten, F.R. YAP1 takes over when oncogenic K-Ras slumbers. Cell 2014, 158, 11–12. [Google Scholar] [CrossRef]
  48. Hsu, P.C.; Miao, J.; Huang, Z.; Yang, Y.L.; Xu, Z.; You, J.; Dai, Y.; Yeh, C.C.; Chan, G.; Liu, S.; et al. Inhibition of yes-associated protein suppresses brain metastasis of human lung adenocarcinoma in a murine model. J. Cell. Mol. Med. 2018, 22, 3073–3085. [Google Scholar] [CrossRef] [PubMed]
  49. Felley-Bosco, E.; Stahel, R. Hippo/YAP pathway for targeted therapy. Transl. Lung Cancer Res. 2014, 3, 75–83. [Google Scholar] [PubMed]
  50. Zhang, W.Q.; Dai, Y.Y.; Hsu, P.C.; Wang, H.; Cheng, L.; Yang, Y.L.; Wang, Y.C.; Xu, Z.; Liu, S.; Chan, G.; et al. Targeting YAP in malignant pleural mesothelioma. J. Cell. Mol. Med. 2017, 21, 2663–2676. [Google Scholar] [CrossRef] [PubMed]
  51. Woodard, G.A.; Yang, Y.L.; You, L.; Jablons, D.M. Drug development against the hippo pathway in mesothelioma. Transl. Lung Cancer Res. 2017, 6, 335–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Aguiar, P.N., Jr.; Santoro, I.L.; Tadokoro, H.; de Lima, L.G.; Filardi, B.A.; Oliveira, P.; Castelo-Branco, P.; Mountzios, G.; de Mello, R.A. A pooled analysis of nivolumab for the treatment of advanced non-small-cell lung cancer and the role of PD-L1 as a predictive biomarker. Immunotherapy 2016, 8, 1011–1019. [Google Scholar] [CrossRef] [PubMed]
  53. Maleki Vareki, S.; Garrigós, C.; Duran, I. Biomarkers of response to PD-1/PD-L1 inhibition. Crit. Rev. Oncol. Hematol. 2017, 116, 116–124. [Google Scholar] [CrossRef] [PubMed]
  54. Gebhardt, T.; Harvey, K.F. Hippo Wades into Cancer Immunology. Dev. Cell 2016, 39, 635–637. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, S.; Xie, F.; Chu, F.; Zhang, Z.; Yang, B.; Dai, T.; Gao, L.; Wang, L.; Ling, L.; Jia, J.; et al. YAP antagonizes innate antiviral immunity and is targeted for lysosomal degradation through IKKε-mediated phosphorylation. Nat. Immunol. 2017, 18, 733–743. [Google Scholar] [CrossRef]
  56. Wang, G.; Lu, X.; Dey, P.; Deng, P.; Wu, C.C.; Jiang, S.; Fang, Z.; Zhao, K.; Konaparthi, R.; Hua, S.; et al. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov. 2016, 6, 80–95. [Google Scholar] [CrossRef] [PubMed]
  57. Murakami, S.; Shahbazian, D.; Surana, R.; Zhang, W.; Chen, H.; Graham, G.T.; White, S.M.; Weiner, L.M.; Yi, C. Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma. Oncogene 2017, 36, 1232–1244. [Google Scholar] [CrossRef]
  58. Ni, X.; Tao, J.; Barbi, J.; Chen, Q.; Park, B.V.; Li, Z.; Zhang, N.; Lebid, A.; Ramaswamy, A.; Wei, P.; et al. YAP Is Essential for Treg-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2018, 8, 1026–1043. [Google Scholar] [CrossRef] [Green Version]
  59. Miao, J.; Hsu, P.C.; Yang, Y.L.; Xu, Z.; Dai, Y.; Wang, Y.; Chan, G.; Huang, Z.; Hu, B.; Li, H.; et al. YAP regulates PD-L1 expression in human NSCLC cells. Oncotarget 2017, 8, 114576–114587. [Google Scholar] [CrossRef] [Green Version]
  60. Hsu, P.C.; Miao, J.; Wang, Y.C.; Zhang, W.Q.; Yang, Y.L.; Wang, C.W.; Yang, C.T.; Huang, Z.; You, J.; Xu, Z.; et al. Inhibition of yes-associated protein down-regulates PD-L1 (CD274) expression in human malignant pleural mesothelioma. J. Cell. Mol. Med. 2018, 22, 3139–3148. [Google Scholar] [CrossRef] [Green Version]
  61. Janse van Rensburg, H.J.; Azad, T.; Ling, M.; Hao, Y.; Snetsinger, B.; Khanal, P.; Minassian, L.M.; Graham, C.H.; Rauh, M.J.; Yang, X. The Hippo Pathway Component TAZ Promotes Immune Evasion in Human Cancer through PD-L1. Cancer Res. 2018, 78, 1457–1470. [Google Scholar] [CrossRef]
  62. Taha, Z.; Janse van Rensburg, H.J.; Yang, X. The Hippo Pathway: Immunity and Cancer. Cancers (Basel) 2018, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, B.S.; Park, D.I.; Lee, D.H.; Lee, J.E.; Yeo, M.K.; Park, Y.H.; Lim, D.S.; Choi, W.; Lee, D.H.; Yoo, G.; et al. Hippo effector YAP directly regulates the expression of PD-L1 transcripts in EGFR-TKI-resistant lung adenocarcinoma. Biochem. Biophys. Res. Commun. 2017, 491, 493–499. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, N.; Fang, W.; Zhan, J.; Hong, S.; Tang, Y.; Kang, S.; Zhang, Y.; He, X.; Zhou, T.; Qin, T.; et al. Upregulation of PD-L1 by EGFR Activation Mediates the Immune Escape in EGFR-Driven NSCLC: Implication for Optional Immune Targeted Therapy for NSCLC Patients with EGFR Mutation. J. Thorac. Oncol. 2015, 10, 910–923. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, N.; Zeng, Y.; Du, W.; Zhu, J.; Shen, D.; Liu, Z.; Huang, J.A. The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int. J. Oncol. 2016, 49, 1360–1368. [Google Scholar] [CrossRef] [PubMed]
  66. Ashizawa, T.; Iizuka, A.; Nonomura, C.; Kondou, R.; Maeda, C.; Miyata, H.; Sugino, T.; Mitsuya, K.; Hayashi, N.; Nakasu, Y.; et al. Antitumor Effect of Programmed Death-1 (PD-1) Blockade in Humanized the NOG-MHC Double Knockout Mouse. Clin. Cancer Res. 2017, 23, 149–158. [Google Scholar] [CrossRef]
  67. Wang, M.; Yao, L.C.; Cheng, M.; Cai, D.; Martinek, J.; Pan, C.X.; Shi, W.; Ma, A.H.; De Vere White, R.W.; Airhart, S.; et al. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J. 2018, 32, 1537–1549. [Google Scholar] [CrossRef] [Green Version]
  68. Brinda, B.; Khan, I.; Parkin, B.; Konig, H. The rocky road to personalized medicine in acute myeloid leukaemia. J. Cell. Mol. Med. 2018, 22, 1411–1427. [Google Scholar] [CrossRef] [Green Version]
  69. Yang, S.; Zhang, L.; Liu, M.; Chong, R.; Ding, S.J.; Chen, Y.; Dong, J. CDK1 phosphorylation of YAP promotes mitotic defects and cell motility and is essential for neoplastic transformation. Cancer Res. 2013, 73, 6722–6733. [Google Scholar] [CrossRef]
  70. Galli, G.G.; Carrara, M.; Yuan, W.C.; Valdes-Quezada, C.; Gurung, B.; Pepe-Mooney, B.; Zhang, T.; Geeven, G.; Gray, N.S.; de Laat, W.; et al. YAP Drives Growth by Controlling Transcriptional Pause Release from Dynamic Enhancers. Mol. Cell 2015, 60, 328–337. [Google Scholar] [CrossRef]
  71. Bae, J.S.; Kim, S.M.; Lee, H. The Hippo signaling pathway provides novel anti-cancer drug targets. Oncotarget 2017, 8, 16084–16098. [Google Scholar] [CrossRef] [PubMed]
  72. Chaib, I.; Karachaliou, N.; Pilotto, S.; Codony Servat, J.; Cai, X.; Li, X.; Drozdowskyj, A.; Servat, C.C.; Yang, J.; Hu, C.; et al. Co-activation of STAT3 and YES-Associated Protein 1 (YAP1) Pathway in EGFR-Mutant NSCLC. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Shibata, M.; Ham, K.; Hoque, M.O. A time for YAP1: Tumorigenesis, immunosuppression and targeted therapy. Int. J. Cancer 2018, 143, 2133–2144. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, H.; Pandey, S.; Travers, M.; Sun, H.; Morton, G.; Madzo, J.; Chung, W.; Khowsathit, J.; Perez-Leal, O.; Barrero, C.A.; et al. Targeting CDK9 reactivates epigenetically silenced genes in cancer. Cell 2018, 175, 1244–1258.e26. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The mechanism of anti-programmed death-ligand 1(PD-L1)/PD-1 inhibitors in cancer therapy. In tumor cells, including non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM), the binding of PD-L1 and PD-1 promotes T-cell tolerance and escape from host immunity. Immunotherapy targeting immune checkpoints for either anti-PD-1 or anti-PD-L1 has been developed and used in cancer therapy. Pembrolizumab and nivolumab are anti-PD-1 inhibitors, and atezolizumab and durvalumab are anti-PD-L1 inhibitors.
Figure 1. The mechanism of anti-programmed death-ligand 1(PD-L1)/PD-1 inhibitors in cancer therapy. In tumor cells, including non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM), the binding of PD-L1 and PD-1 promotes T-cell tolerance and escape from host immunity. Immunotherapy targeting immune checkpoints for either anti-PD-1 or anti-PD-L1 has been developed and used in cancer therapy. Pembrolizumab and nivolumab are anti-PD-1 inhibitors, and atezolizumab and durvalumab are anti-PD-L1 inhibitors.
Biomedicines 06 00114 g001
Figure 2. The regulation of the Hippo/YAP signaling pathway in malignant pleural mesothelioma (MPM) and non-small cell lung cancer (NSCLC). Hippo kinase, including NF2 and LATS1/LATS2, phosphorylate yes-associated protein (YAP) in the cytoplasm and lead to the degradation of YAP. In MPM, genetic mutations with loss of function in NF2, LATS1/LATS2, SAV1 and RASSF frequently occur, which leads to the degradation of YAP in the cytoplasm. Therefore, more YAP proteins translocate into the nucleus and activate transcription of downstream genes by forming complexes with transcriptional enhancer factors (TEAD), and promote tumorigenesis of MPM. In some NSCLC cells with high potential for drug resistance and metastasis, YAP expression increases at the protein level in the cytoplasm, and more YAP proteins translocate into the nucleus, which activates downstream genes including CTGF, cyr61 or other EGF expression genes, and then form autocrine loops to activate oncogenic pathways, such as MAPK signaling, which would inhibit the Hippo kinase. Therefore, the formation of these autocrine loops enhances YAP signaling, which promotes tumor cell proliferation, drug resistance and metastasis in NSCLC. Abbreviations: NF2, neurofibromatosis 2; LATS1, large tumor suppressor homolog 1; MST1, mammalian sterile-20 like kinase 1; SAV1, protein salvador homolog 1; RASSF, Ras-association domain family.
Figure 2. The regulation of the Hippo/YAP signaling pathway in malignant pleural mesothelioma (MPM) and non-small cell lung cancer (NSCLC). Hippo kinase, including NF2 and LATS1/LATS2, phosphorylate yes-associated protein (YAP) in the cytoplasm and lead to the degradation of YAP. In MPM, genetic mutations with loss of function in NF2, LATS1/LATS2, SAV1 and RASSF frequently occur, which leads to the degradation of YAP in the cytoplasm. Therefore, more YAP proteins translocate into the nucleus and activate transcription of downstream genes by forming complexes with transcriptional enhancer factors (TEAD), and promote tumorigenesis of MPM. In some NSCLC cells with high potential for drug resistance and metastasis, YAP expression increases at the protein level in the cytoplasm, and more YAP proteins translocate into the nucleus, which activates downstream genes including CTGF, cyr61 or other EGF expression genes, and then form autocrine loops to activate oncogenic pathways, such as MAPK signaling, which would inhibit the Hippo kinase. Therefore, the formation of these autocrine loops enhances YAP signaling, which promotes tumor cell proliferation, drug resistance and metastasis in NSCLC. Abbreviations: NF2, neurofibromatosis 2; LATS1, large tumor suppressor homolog 1; MST1, mammalian sterile-20 like kinase 1; SAV1, protein salvador homolog 1; RASSF, Ras-association domain family.
Biomedicines 06 00114 g002
Figure 3. The regulation of programmed death-ligand 1 (PD-L1) expression by yes-associated protein (YAP) in human non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). We examined the PD-L1 enhancer region (−10,000 bps) upstream of the transcription starting site of PD-L1 and found 2 putative TEAD-binding sites (CATTCC), 7941 bps and 7911 bps upstream of the PD-L1 transcription start site. The results of chromatin immunoprecipitation (ChIP) assays in our previous two studies indicate that YAP regulates PD-L1 at the transcriptional level in the nucleus.
Figure 3. The regulation of programmed death-ligand 1 (PD-L1) expression by yes-associated protein (YAP) in human non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). We examined the PD-L1 enhancer region (−10,000 bps) upstream of the transcription starting site of PD-L1 and found 2 putative TEAD-binding sites (CATTCC), 7941 bps and 7911 bps upstream of the PD-L1 transcription start site. The results of chromatin immunoprecipitation (ChIP) assays in our previous two studies indicate that YAP regulates PD-L1 at the transcriptional level in the nucleus.
Biomedicines 06 00114 g003

Share and Cite

MDPI and ACS Style

Hsu, P.-C.; Yang, C.-T.; Jablons, D.M.; You, L. The Role of Yes-Associated Protein (YAP) in Regulating Programmed Death-Ligand 1 (PD-L1) in Thoracic Cancer. Biomedicines 2018, 6, 114. https://doi.org/10.3390/biomedicines6040114

AMA Style

Hsu P-C, Yang C-T, Jablons DM, You L. The Role of Yes-Associated Protein (YAP) in Regulating Programmed Death-Ligand 1 (PD-L1) in Thoracic Cancer. Biomedicines. 2018; 6(4):114. https://doi.org/10.3390/biomedicines6040114

Chicago/Turabian Style

Hsu, Ping-Chih, Cheng-Ta Yang, David M. Jablons, and Liang You. 2018. "The Role of Yes-Associated Protein (YAP) in Regulating Programmed Death-Ligand 1 (PD-L1) in Thoracic Cancer" Biomedicines 6, no. 4: 114. https://doi.org/10.3390/biomedicines6040114

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

Article Metrics

Back to TopTop