簡易檢索 / 詳目顯示

研究生: 張簡安
Chang, Chien-An
論文名稱: 藉由大數據挖掘和全基因組識別建構基因與表觀遺傳網路並比較肺腺癌和肺鱗狀細胞癌之間的進展分子機制
Investigating the Genome-wide Genetic and Epigenetic Networks for Comparing Progression Molecular Mechanisms between Lung Adenocarcinoma and Lung Squamous Cell Carcinoma via Big Data Mining and Genome-wide Identification
指導教授: 陳博現
Chen, Bor-Sen
口試委員: 王慧菁
Wang, Hui-Ching
詹鴻霖
Chan, Hong-Lin
蘇士哲
Sue, Shih-Che
王禹超
Wang, Yu-Chao
學位類別: 碩士
Master
系所名稱: 電機資訊學院 - 電機工程學系
Department of Electrical Engineering
論文出版年: 2017
畢業學年度: 105
語文別: 英文
論文頁數: 102
中文關鍵詞: 肺腺癌肺鱗狀細胞癌非小細胞肺癌基因與表觀遺傳網路主成份網路投影方法多分子藥物
外文關鍵詞: lung adenocarcinoma, lung squamous cell carcinoma, NSCLC, genetic and epigenetic network, principal network projection (PNP), multi-molecule drug
相關次數: 點閱:3下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報
  • 肺癌是最常見的惡性腫瘤,是導致全世界癌症相關死亡人數最多的疾病。只有15.9%的肺癌患者存活5年以上。作為非小細胞肺癌(NSCLC)亞型的肺腺癌(LADC)和肺鱗狀細胞癌(LSCC)在全球中是肺癌的主要形式。由於其各自獨特的分子特徵,不同細胞的起源與不同的臨床反應,使它們被認為是不同的疾病。透過使用全基因組候選基因與表觀遺傳網路(GENs)、正常細胞與LADC和LSCC患者癌細胞的次世代定序(NGS)數據和DNA甲基化譜,並藉由系統識別方法,系統階層測定方法,以及利用蛋白質與蛋白質相互作用網絡(PPIN)與基因/ miRNA / lncRNA調控網絡(GRN)所建立的系統模型,和表觀遺傳網路,我們分別建立了LADC和LSCC在各自正常,早期,中期,以及晚期階段的基因與表觀遺傳網路。透過應用主成份網路投影方法(PNP),我們分別得到LADC和LSCC各自正常,前期,中期,以及晚期階段的核心GENs。為了比較LADC和LSCC之間基因和表觀遺傳機制,我們藉由比較LADC和LSCC各自的兩相鄰疾病階段間的核心GENs來萃取出差異的核心信號途徑。根據我們的結果,我們發現造成LADC和LSCC各自從正常階段到早期階段,早期階段到中期,中期到晚期階段的分子進展原因分別是由細菌感染(LPS),EGFR突變,miR-130b,miR-100HG和miR-1292的失調,E2F1泛素化,SFTPA2去泛素化和乙醯化,MYC和RB1的DNA甲基化; 缺氧環境,miR-30c2和miR-27b的失調,VIM,MYH9和ETS1的乙醯化,MDM4泛素化;過氧化氫導致的氧化壓力,miR-143HG和miR-19a的失調,ZEB1乙醯化,RHOB去乙醯化,PCK1的DNA甲基化; 暴露於尼古丁,miR-24-2和miR-1247的失調,以及lncRNA TUG1,KLF12泛素化和乙醯化,ERCC1泛素化和SHOX2的DNA甲基化;缺氧環境,HIF1A,IGF1和ETS1的乙醯化,ITGB1,TNS1,miR-21的DNA甲基化; NNK導致的氧化壓力,miR-9-2的失調,GATA3泛素化和去乙醯化,FOXL1去乙醯化,CDH3去泛素化,PDPN的DNA甲基化。最後,我們提出了六個基因和表觀遺傳多分子藥物,分別在LADC和LSCC各自的進展階段中標靶重要的生物標誌。


    Lung cancer, the most common malignancy, results in the largest number of cancer-related deaths worldwide. Only 15.9% of lung cancer patients survive more than 5 years. Lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC), which are subtype of Non-small-cell lung cancer (NSCLC), are the predominant form of lung cancer in worldwide and they are thought to be distinct diseases due to their unique molecular characteristics, distinct cells of origin, and different clinical response. By using genome-wide candidate genetic and epigenetic networks (GENs), next-generation sequencing (NGS) data, and DNA methylation profiles of normal cells and cancer cells in LADC and LSCC patients, we respectively constructed GENs in normal , early , middle , and advanced stage of LADC and LSCC based on system identification method, system order detection scheme, and system modeling via protein-protein interaction networks (PPINs), gene/miRNA/lncRNA regulatory networks (GRNs), and epigenetic networks. Through applying principle network projection (PNP), we then identified the core GENs of LADC and LSCC in each stage from normal stage to advanced stage. To investigate and compare the progression genetic and epigenetic mechanisms between LADC and LSCC, we then extracted differential core signaling pathways by comparing core GENs between two connective stages of LADC and LSCC, respectively. Based on our results, we found that the progression from normal stage to early stage, early stage to middle stage, and middle stage to advanced stage LADC and LSCC are respectively caused by stimulation of bacteria infection (LPS), EGFR mutation, dysfunction of miR-130b, miR-100HG, and miR-1292, E2F1 ubiquitination, SFTPA2 deubiquitination and acetylation, DNA methylation of MYC and RB1; hypoxia, dysfunction of miR-30c2 and miR-27b, acetylation of VIM, MYH9, and ETS1, MDM4 ubiquitination; hydrogen peroxide induced oxidative stress, dysfunction of miR-143HG and miR-19a, ZEB1 acetylation, RHOB deacetylation, DNA methylation of PCK1; nicotine exposure, dysfunction of miR-24-2 and miR-1247, and lncRNA TUG1, KLF12 ubiquitination and acetylation, ERCC1 ubiquitination, and DNA methylation of SHOX2; hypoxia, acetylation of HIF1A, IGF1, and ETS1, DNA methylation of ITGB1, TNS1, miR-21; NNK induced oxidative stress, dysfunction of miR-9-2, GATA3 ubiquitination and deacetylation, FOXL1 deacetylation, CDH3 deubiquitination, DNA methylation of PDPN. Finally, we proposed six genetic and epigenetic multiple-molecule drugs to target significant biomarker in each progression stage LADC and LSCC, respectively.

    Contents 誌謝 I 摘要 II Abstract III Contents IV List of Tables VIII List of Figures IX Chapter 1. Introduction 1 Chapter 2. Materials and Methods 6 2.1 Overview of the construction for genome-wide GENS, core GENs, and core pathways of each progression stage in LADC and LSCC 6 2.2 Big data mining and data preprocessing of NGS data and methylation data for information of constructing GENs of human 7 2.3 Constructing genome-wide candidate GENs 8 2.4 Constructing stochastic regression models of candidate PPIN and candidate GRN of candidate GENs for LADC and LSCC 8 2.5 Parameter estimation of the stochastic regression models of candidate GENs via system identification method and system order detection 12 2.6 Extracting core GENs from the real GENs by using the PNP method 17 Chapter 3. Results 22 3.1 Analysis of core pathways to investigate different progression genetic and epigenetic mechanisms of LADC and LSCC from normal stage to early stage 23 3.2 Analysis of core pathways to investigate different progression genetic and epigenetic mechanisms of LADC and LSCC from early stage to middle stage 25 3.3 Analysis of core pathways to investigate different progression genetic and epigenetic mechanisms of LADC and LSCC from middle stage to advanced stage 28 Chapter 4. Discussion 33 4.1 Microenvironment change, dysregulation of miRNA/lncRNA regulation, DNA methylation, and epigenetic modification contribute to different progression genetic and epigenetic mechanisms from normal stage to progress to early stage of LADC and LSCC 33 4.1.1 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from normal stage to early stage LADC 34 4.1.1.1 The inflammatory microenvironment caused by infection of bacteria can lead to dysregulation of EGFR and TLR4 signaling pathways to progress from normal stage to early stage LADC 34 4.1.1.2 Dysregulations of miR-130b, miR-100HG, and miR-1292, epigenetic modifications of EGFL7, SFTPA2, and E2F1, and DNA methylation of MYC and RB1, mutation of EGFR contribute to the progression from normal stage to early stage LADC 35 4.1.2 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from normal stage to early stage LADC 37 4.1.2.1 The inflammatory microenvironment caused by exposure to nicotine can lead to the dysregulation of FGFR1, DDR1, CHRNA5 (α5 nAChR subunit) signaling pathways from normal stage to early stage of LSCC 37 4.1.2.2 Dysregulations of miR-24-2, miR-1247, epigenetic modifications of KLF12 and ERCC1, and DNA methylation of SHOX2 contribute to the progression from normal stage to early stage LSCC 39 4.2 Microenvironment change, dysregulation of miRNA/lncRNA regulation, DNA methylation, and epigenetic modification contribute to different progression genetic and epigenetic mechanisms from early stage to progress to middle stage of LADC and LSCC 40 4.2.1 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from early stage to middle stage LADC 41 4.2.1.1 The hypoxic tumor microenvironment can lead to dysregulation of NOTCH1 and CD49d signaling pathways from early stage to middle stage LADC 41 4.2.1.2 Dysregulations of miR-30c-2 and miR-27b, epigenetic modifications of VIM, MYH9, MDM4, and ETS1 contribute to the progression from early stage to middle stage LADC 43 4.2.2 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from early stage to middle stage LSCC 45 4.2.2.1 The hypoxic tumor microenvironment exposed to nicotine can lead to dysregulation of IGF-1R, TNS1, and ITGB1 (CD29) signaling pathways from early stage to middle stage LSCC 45 4.2.2.2 Dysregulation of miR106b, epigenetic modifications of HIF1α, IGF-1, and ETS1, and DNA methylation of ITGB1, TNS1, and miR21 contribute to the progression from early stage to middle stage LSCC 47 4.3 Microenvironment change, dysregulation of miRNA/lncRNA regulation, DNA methylation, and epigenetic modification contribute to different progression genetic and epigenetic mechanisms from middle stage to progress to advanced stage of LADC and LSCC 49 4.3.1 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from early stage to middle stage LADC 50 4.3.1.1 The hydrogen peroxide secreted by cancer cells leads to the alteration of tumor microenvironment, causing dysregulation of EGFR, EPOR and ITGB1 (CD29) signaling pathways from middle stage to advanced stage of LADC 50 4.3.1.2 Dysregulations of miR-143HG and miR-19a, epigenetic modifications of ZEB1 and RHOB, and DNA methylation of PCK1 contribute to the progression from middle stage to advanced stage LADC 52 4.3.2 Dysfunctions of miRNA regulation, DNA methylation, epigenetic modification and microenvironment alteration contribute to the progression from middle stage to advanced stage LSCC 53 4.3.2.1 The oxidative stress induced by the stimulation of nicotine derived nitrosaminoketone (NNK) can potentially lead to the alteration of microenvironment, causing dysregulation of ERBB4, PDPN, and EMR2 signaling pathways from middle stage to advanced stage LSCC 53 4.3.2.2 Dysregulation of miR-9-2, epigenetic modifications of GATA3, FOXL1, and CDH3, and DNA methylation of PDPN contribute to the progression from middle stage to advanced stage LSCC 55 4.4 Discocery of genetic and epigenetic multiple drugs for treating early stage, middle stage, and advanced stage LADC and LSCC 57 Chapter 5. Conclusion 63 Reference 91 List of Tables Table 1. The number of nodes and edges in candidate GENs 64 Table 2. The number of identified nodes and edges of real GENs in each stage of LADC 65 Table 3. The number of identified nodes and edges of real GENs in each stage of LSCC 66 Table 4. Design of genetic and epigenetic multiple drug for the therapeutic treatment of early stage LADC 67 Table 5. Design of genetic and epigenetic multiple drug for the therapeutic treatment of middle stage LADC 68 Table 6. Design of genetic and epigenetic multiple drug for the therapeutic treatment of advanced stage LADC 69 Table 7. Design of genetic and epigenetic multiple drug for the therapeutic treatment of early stage LSCC 70 Table 8. Design of genetic and epigenetic multiple drug for the therapeutic treatment of middle stage LSCC 71 Table 9. Design of genetic and epigenetic multiple drugs for the therapeutic treatment of advanced stage LSCC 72 List of Figures Figure 1. Flowchart of the construction for genome-wide GENS, core GENs, and core signaling pathways of each progression stage of LADC and LSCC and the discovery of potential genetic and epigenetic multiple drugs. 73 Figure 2. The genetic and epigenetic networks (GENs) of normal stage, early stage, middle stage, and advanced stage LADC. 75 Figure 3. The genetic and epigenetic networks (GENs) of normal stage, early stage, middle stage, and advanced stage LSCC. 76 Figure 4. The core genetic and epigenetic network (GEN) of normal stage of lung cells near LADC cancer cells. 77 Figure 5. The core genetic and epigenetic network (GEN) of early stage LADC. 78 Figure 6. The core genetic and epigenetic network (GEN) of middle stage LADC. 79 Figure 7. The core genetic and epigenetic network (GEN) of advanced stage LADC. 80 Figure 8. The core genetic and epigenetic network (GEN) of normal stage of lung cells near LSCC cancer cells. 81 Figure 9. The core genetic and epigenetic network (GEN) of early stage LSCC. 82 Figure 10. The core genetic and epigenetic network (GEN) of middle stage LSCC. 83 Figure 11. The core genetic and epigenetic network (GEN) of advanced stage LSCC. 84 Figure 12. Core signaling pathways extracted from comparing genetic and epigenetic networks (GENs) between normal lung cells and early stage LADC and LSCC. 85 Figure 13. Core signaling pathways extracted from comparing genetic and epigenetic networks (GENs) between early stage and middle stage LADC and LSCC. 86 Figure 14. Core signaling pathways extracted from comparing genetic and epigenetic networks (GENs) between middle stage and advanced stage LADC and LSCC. 87 Figure 15. The specific core signaling pathways extracted from Figures 12-14 for investigating the differential progression molecular mechanisms between LADC and LSCC. 89 Figure 16. Summarizing the differential progression genetic and epigenetic mechanisms from normal stage to early stage, early stage to middle stage, and middle stage to advanced stage LADC and LSCC. 90

    1. Jemal, A., et al., Global cancer statistics. CA Cancer J Clin, 2011. 61(2): p. 69-90.
    2. Siegel, R., D. Naishadham, and A. Jemal, Cancer statistics, 2013. CA Cancer J Clin, 2013. 63(1): p. 11-30.
    3. Ettinger, D.S., et al., Non-small cell lung cancer, version 2.2013. J Natl Compr Canc Netw, 2013. 11(6): p. 645-53; quiz 653.
    4. Balgkouranidou, I., T. Liloglou, and E.S. Lianidou, Lung cancer epigenetics: emerging biomarkers. Biomark Med, 2013. 7(1): p. 49-58.
    5. Davidson, M.R., A.F. Gazdar, and B.E. Clarke, The pivotal role of pathology in the management of lung cancer. J Thorac Dis, 2013. 5 Suppl 5: p. S463-78.
    6. Shukla, S., et al., Genetics and Epigenetics of Lung Cancer: Mechanisms and Future Perspectives. Current Cancer Therapy Reviews, 2013. 9(2): p. 97-110.
    7. Chen, Z., et al., Non-small-cell lung cancers: a heterogeneous set of diseases. Nat Rev Cancer, 2014. 14(8): p. 535-46.
    8. Langer, C.J., et al., The evolving role of histology in the management of advanced non-small-cell lung cancer. J Clin Oncol, 2010. 28(36): p. 5311-20.
    9. Minna, J.D., J.A. Roth, and A.F. Gazdar, Focus on lung cancer. Cancer Cell, 2002. 1(1): p. 49-52.
    10. Langevin, S.M., R.A. Kratzke, and K.T. Kelsey, Epigenetics of lung cancer. Transl Res, 2015. 165(1): p. 74-90.
    11. Massion, P.P. and D.P. Carbone, The molecular basis of lung cancer: molecular abnormalities and therapeutic implications. Respir Res, 2003. 4: p. 12.
    12. Khuder, S.A., Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis. Lung Cancer, 2001. 31(2-3): p. 139-48.
    13. Brzezianska, E., A. Dutkowska, and A. Antczak, The significance of epigenetic alterations in lung carcinogenesis. Mol Biol Rep, 2013. 40(1): p. 309-25.
    14. Chen, Q.W., et al., Epigenetic regulation and cancer (review). Oncol Rep, 2014. 31(2): p. 523-32.
    15. Castro, M., et al., Multiplexed methylation profiles of tumor suppressor genes and clinical outcome in lung cancer. J Transl Med, 2010. 8: p. 86.
    16. Hawes, S.E., et al., DNA hypermethylation of tumors from non-small cell lung cancer (NSCLC) patients is associated with gender and histologic type. Lung Cancer, 2010. 69(2): p. 172-9.
    17. Kwon, Y.J., et al., Genome-wide analysis of DNA methylation and the gene expression change in lung cancer. J Thorac Oncol, 2012. 7(1): p. 20-33.
    18. Rouhi, A., et al., MiRNAs, epigenetics, and cancer. Mamm Genome, 2008. 19(7-8): p. 517-25.
    19. Calin, G.A., et al., Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A, 2004. 101(9): p. 2999-3004.
    20. Inamura, K. and Y. Ishikawa, MicroRNA In Lung Cancer: Novel Biomarkers and Potential Tools for Treatment. J Clin Med, 2016. 5(3).
    21. Lin, P.Y., S.L. Yu, and P.C. Yang, MicroRNA in lung cancer. Br J Cancer, 2010. 103(8): p. 1144-8.
    22. Shi, X., et al., Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett, 2013. 339(2): p. 159-66.
    23. Wei, M.M. and G.B. Zhou, Long Non-coding RNAs and Their Roles in Non-small-cell Lung Cancer. Genomics Proteomics Bioinformatics, 2016. 14(5): p. 280-288.
    24. Zhang, H., et al., Long non-coding RNA: a new player in cancer. J Hematol Oncol, 2013. 6: p. 37.
    25. Kumegawa, K., et al., A genomic screen for long noncoding RNA genes epigenetically silenced by aberrant DNA methylation in colorectal cancer. Sci Rep, 2016. 6: p. 26699.
    26. Weber, M., et al., Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet, 2007. 39(4): p. 457-66.
    27. Li, C.W., P.Y. Chang, and B.S. Chen, Investigating the mechanism of hepatocellular carcinoma progression by constructing genetic and epigenetic networks using NGS data identification and big database mining method. Oncotarget, 2016. 7(48): p. 79453-79473.
    28. Li, C.W. and B.S. Chen, Network Biomarkers of Bladder Cancer Based on a Genome-Wide Genetic and Epigenetic Network Derived from Next-Generation Sequencing Data. Dis Markers, 2016. 2016: p. 4149608.
    29. Wang, Y.C. and B.S. Chen, A network-based biomarker approach for molecular investigation and diagnosis of lung cancer. BMC Med Genomics, 2011. 4: p. 2.
    30. Tong, A.H., et al., Global mapping of the yeast genetic interaction network. Science, 2004. 303(5659): p. 808-13.
    31. Wasserman, S. and K. Faust, Social Network Analysis: Methods and Applications. 1994: Cambridge University Press.
    32. Salwinski, L., et al., The Database of Interacting Proteins: 2004 update. Nucleic Acids Res, 2004. 32(Database issue): p. D449-51.
    33. Bader, G.D., D. Betel, and C.W. Hogue, BIND: the Biomolecular Interaction Network Database. Nucleic Acids Res, 2003. 31(1): p. 248-50.
    34. Chatr-Aryamontri, A., et al., The BioGRID interaction database: 2015 update. Nucleic Acids Res, 2015. 43(Database issue): p. D470-8.
    35. Orchard, S., et al., The MIntAct project--IntAct as a common curation platform for 11 molecular interaction databases. Nucleic Acids Res, 2014. 42(Database issue): p. D358-63.
    36. Licata, L., et al., MINT, the molecular interaction database: 2012 update. Nucleic Acids Res, 2012. 40(Database issue): p. D857-61.
    37. Zheng, G., et al., ITFP: an integrated platform of mammalian transcription factors. Bioinformatics, 2008. 24(20): p. 2416-7.
    38. Bovolenta, L.A., M.L. Acencio, and N. Lemke, HTRIdb: an open-access database for experimentally verified human transcriptional regulation interactions. BMC Genomics, 2012. 13: p. 405.
    39. Wingender, E., The TRANSFAC project as an example of framework technology that supports the analysis of genomic regulation. Brief Bioinform, 2008. 9(4): p. 326-32.
    40. Agarwal, V., et al., Predicting effective microRNA target sites in mammalian mRNAs. Elife, 2015. 4.
    41. Friard, O., et al., CircuitsDB: a database of mixed microRNA/transcription factor feed-forward regulatory circuits in human and mouse. BMC Bioinformatics, 2010. 11: p. 435.
    42. Li, J.H., et al., starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res, 2014. 42(Database issue): p. D92-7.
    43. Medvedeva, Y.A., et al., Effects of cytosine methylation on transcription factor binding sites. BMC Genomics, 2014. 15: p. 119.
    44. Bandres, E., et al., Epigenetic regulation of microRNA expression in colorectal cancer. Int J Cancer, 2009. 125(11): p. 2737-43.
    45. Coleman, T.F. and Y. Li, A Reflective Newton Method for Minimizing a Quadratic Function Subject to Bounds on Some of the Variables. SIAM Journal on Optimization, 1996. 6(4): p. 1040-1058.
    46. Lu, P., et al., Absolute protein expression profiling estimates the relative contributions of transcriptional and translational regulation. Nat Biotechnol, 2007. 25(1): p. 117-24.
    47. Johansson, R., System Modeling and Identification. 1993: Prentice Hall.
    48. Chen, L.Z., S.K. Nguang, and X.D. Chen, On-line identification and optimization of feed rate profiles for high productivity fed-batch culture of hybridoma cells using genetic algorithms. ISA Trans, 2002. 41(4): p. 409-19.
    49. Shannon, P., et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 2003. 13(11): p. 2498-504.
    50. Han, X., et al., Transdifferentiation of lung adenocarcinoma in mice with Lkb1 deficiency to squamous cell carcinoma. Nat Commun, 2014. 5: p. 3261.
    51. Coussens, L.M. and Z. Werb, Inflammation and cancer. Nature, 2002. 420(6917): p. 860-7.
    52. Lin, W.W. and M. Karin, A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest, 2007. 117(5): p. 1175-83.
    53. Ribeiro, R., et al., Immunoinflammatory mechanisms in lung cancer development: is leptin a mediator? J Thorac Oncol, 2007. 2(2): p. 105-8.
    54. Lighthart, B., Mini-review of the concentration variations found inthe alfresco atmospheric bacterial populations. Aerobiologia, 2000. 16(1): p. 7-16.
    55. Fischer, J.R., et al., Constitutive secretion of bioactive transforming growth factor beta 1 by small cell lung cancer cell lines. Eur J Cancer, 1994. 30A(14): p. 2125-9.
    56. Domagala-Kulawik, J., et al., Elevated TGF-beta1 concentration in bronchoalveolar lavage fluid from patients with primary lung cancer. Arch Immunol Ther Exp (Warsz), 2006. 54(2): p. 143-7.
    57. Bennett, W.P., et al., p21waf1/cip1 and transforming growth factor beta 1 protein expression correlate with survival in non-small cell lung cancer. Clin Cancer Res, 1998. 4(6): p. 1499-506.
    58. Flavell, R.A., et al., The polarization of immune cells in the tumour environment by TGFbeta. Nat Rev Immunol, 2010. 10(8): p. 554-67.
    59. Donin, N., et al., Complement resistance of human carcinoma cells depends on membrane regulatory proteins, protein kinases and sialic acid. Clin Exp Immunol, 2003. 131(2): p. 254-63.
    60. Jurianz, K., et al., Complement resistance of tumor cells: basal and induced mechanisms. Mol Immunol, 1999. 36(13-14): p. 929-39.
    61. Verstak, B., et al., MyD88 adapter-like (Mal)/TIRAP interaction with TRAF6 is critical for TLR2- and TLR4-mediated NF-kappaB proinflammatory responses. J Biol Chem, 2009. 284(36): p. 24192-203.
    62. He, W., et al., TLR4 signaling promotes immune escape of human lung cancer cells by inducing immunosuppressive cytokines and apoptosis resistance. Mol Immunol, 2007. 44(11): p. 2850-9.
    63. Borron, P., et al., Surfactant-associated protein A inhibits LPS-induced cytokine and nitric oxide production in vivo. Am J Physiol Lung Cell Mol Physiol, 2000. 278(4): p. L840-7.
    64. Beckman, G., et al., Haptoglobin groups and lung cancer. Hum Hered, 1986. 36(4): p. 258-60.
    65. Chang, Y.K., et al., Haptoglobin is a serological biomarker for adenocarcinoma lung cancer by using the ProteomeLab PF2D combined with mass spectrometry. Am J Cancer Res, 2016. 6(8): p. 1828-36.
    66. Delfortrie, S., et al., Egfl7 promotes tumor escape from immunity by repressing endothelial cell activation. Cancer Res, 2011. 71(23): p. 7176-86.
    67. Fitch, M.J., et al., Egfl7, a novel epidermal growth factor-domain gene expressed in endothelial cells. Dev Dyn, 2004. 230(2): p. 316-24.
    68. Tang, J., et al., Erlotinib resistance in lung cancer: current progress and future perspectives. Front Pharmacol, 2013. 4: p. 15.
    69. Hara, H., et al., Clinicopathologic significance of telomerase activity and hTERT mRNA expression in non-small cell lung cancer. Lung Cancer, 2001. 34(2): p. 219-26.
    70. Tan, Q., et al., Identification of FGF19 as a prognostic marker and potential driver gene of lung squamous cell carcinomas in Chinese smoking patients. Oncotarget, 2016. 7(14): p. 18394-402.
    71. Grando, S.A., Connections of nicotine to cancer. Nat Rev Cancer, 2014. 14(6): p. 419-29.
    72. Sopori, M., Effects of cigarette smoke on the immune system. Nat Rev Immunol, 2002. 2(5): p. 372-7.
    73. Nouri-Shirazi, M. and E. Guinet, Evidence for the immunosuppressive role of nicotine on human dendritic cell functions. Immunology, 2003. 109(3): p. 365-73.
    74. Hanahan, D. and R.A. Weinberg, Hallmarks of cancer: the next generation. Cell, 2011. 144(5): p. 646-74.
    75. Su, B., et al., Let-7d suppresses growth, metastasis, and tumor macrophage infiltration in renal cell carcinoma by targeting COL3A1 and CCL7. Mol Cancer, 2014. 13: p. 206.
    76. Berger, W., et al., Evidence for a role of FGF-2 and FGF receptors in the proliferation of non-small cell lung cancer cells. Int J Cancer, 1999. 83(3): p. 415-23.
    77. Godin-Heymann, N., et al., Tumour-suppression function of KLF12 through regulation of anoikis. Oncogene, 2016. 35(25): p. 3324-34.
    78. Lomberk, G. and R. Urrutia, The family feud: turning off Sp1 by Sp1-like KLF proteins. Biochem J, 2005. 392(Pt 1): p. 1-11.
    79. Hsu, T.I., et al., Sp1 expression regulates lung tumor progression. Oncogene, 2012. 31(35): p. 3973-88.
    80. Lin, P.C., et al., Long noncoding RNA TUG1 is downregulated in non-small cell lung cancer and can regulate CELF1 on binding to PRC2. BMC Cancer, 2016. 16: p. 583.
    81. Zhang, E., et al., Increased expression of long noncoding RNA TUG1 predicts a poor prognosis of gastric cancer and regulates cell proliferation by epigenetically silencing of p57. Cell Death Dis, 2016. 7: p. e2109.
    82. Zhang, M., et al., Downregulation of the long noncoding RNA TUG1 inhibits the proliferation, migration, invasion and promotes apoptosis of renal cell carcinoma. J Mol Histol, 2016. 47(4): p. 421-8.
    83. Prudkin, L., et al., Epithelial-to-mesenchymal transition in the development and progression of adenocarcinoma and squamous cell carcinoma of the lung. Mod Pathol, 2009. 22(5): p. 668-78.
    84. Baumgart, A., et al., ADAM17 regulates epidermal growth factor receptor expression through the activation of Notch1 in non-small cell lung cancer. Cancer Res, 2010. 70(13): p. 5368-78.
    85. Donnem, T., et al., Prognostic impact of Notch ligands and receptors in nonsmall cell lung cancer: coexpression of Notch-1 and vascular endothelial growth factor-A predicts poor survival. Cancer, 2010. 116(24): p. 5676-85.
    86. Hsu, Y.L., et al., Galectin-1 promotes lung cancer tumor metastasis by potentiating integrin alpha6beta4 and Notch1/Jagged2 signaling pathway. Carcinogenesis, 2013. 34(6): p. 1370-81.
    87. Jiang, X., et al., [Expression and significance of Notch1, Jagged1 and VEGF in human non-small cell lung cancer]. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 2007. 32(6): p. 1031-6.
    88. Yang, Y., et al., The Notch ligand Jagged2 promotes lung adenocarcinoma metastasis through a miR-200-dependent pathway in mice. J Clin Invest, 2011. 121(4): p. 1373-85.
    89. Yuan, X., et al., Meta-analysis reveals the correlation of Notch signaling with non-small cell lung cancer progression and prognosis. Sci Rep, 2015. 5: p. 10338.
    90. Zhou, L., et al., [Expression of CD133 and Notch1 in non-small cell lung cancer and the clinicopathological significance]. Nan Fang Yi Ke Da Xue Xue Bao, 2015. 35(2): p. 196-201.
    91. Chen, Y., et al., Oxygen concentration determines the biological effects of NOTCH-1 signaling in adenocarcinoma of the lung. Cancer Res, 2007. 67(17): p. 7954-9.
    92. Oh, M.K., et al., Hypoxia-inducible factor-1alpha enhances haptoglobin gene expression by improving binding of STAT3 to the promoter. J Biol Chem, 2011. 286(11): p. 8857-65.
    93. Ding, F., et al., USP22 promotes NSCLC tumorigenesis via MDMX up-regulation and subsequent p53 inhibition. Int J Mol Sci, 2014. 16(1): p. 307-20.
    94. Wang, X., p53 regulation: teamwork between RING domains of Mdm2 and MdmX. Cell Cycle, 2011. 10(24): p. 4225-9.
    95. Jin, Y., et al., MDMX promotes proteasomal turnover of p21 at G1 and early S phases independently of, but in cooperation with, MDM2. Mol Cell Biol, 2008. 28(4): p. 1218-29.
    96. Pan, Y. and J. Chen, MDM2 promotes ubiquitination and degradation of MDMX. Mol Cell Biol, 2003. 23(15): p. 5113-21.
    97. Slabakova, E., et al., Opposite regulation of MDM2 and MDMX expression in acquisition of mesenchymal phenotype in benign and cancer cells. Oncotarget, 2015. 6(34): p. 36156-71.
    98. Takeyama, Y., et al., Knockdown of ZEB1, a master epithelial-to-mesenchymal transition (EMT) gene, suppresses anchorage-independent cell growth of lung cancer cells. Cancer Lett, 2010. 296(2): p. 216-24.
    99. Larsen, J.E., et al., ZEB1 drives epithelial-to-mesenchymal transition in lung cancer. J Clin Invest, 2016. 126(9): p. 3219-35.
    100. Pietras, A., et al., JAG2 induction in hypoxic tumor cells alters Notch signaling and enhances endothelial cell tube formation. Mol Cancer Res, 2011. 9(5): p. 626-36.
    101. Harris, A.L., Hypoxia--a key regulatory factor in tumour growth. Nat Rev Cancer, 2002. 2(1): p. 38-47.
    102. Pouyssegur, J., F. Dayan, and N.M. Mazure, Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature, 2006. 441(7092): p. 437-43.
    103. Semenza, G.L., Hypoxia-inducible factors in physiology and medicine. Cell, 2012. 148(3): p. 399-408.
    104. Giatromanolaki, A., et al., Relation of hypoxia inducible factor 1 alpha and 2 alpha in operable non-small cell lung cancer to angiogenic/molecular profile of tumours and survival. Br J Cancer, 2001. 85(6): p. 881-90.
    105. Lee, C.H., et al., Differential expression of hypoxia inducible factor-1 alpha and tumor cell proliferation between squamous cell carcinomas and adenocarcinomas among operable non-small cell lung carcinomas. J Korean Med Sci, 2003. 18(2): p. 196-203.
    106. Zhang, H., et al., The expression of hypoxia inducible factor 1-alpha in lung cancer and its correlation with P53 and VEGF. J Huazhong Univ Sci Technolog Med Sci, 2004. 24(2): p. 124-7.
    107. Karetsi, E., et al., Differential expression of hypoxia-inducible factor 1alpha in non-small cell lung cancer and small cell lung cancer. Clinics (Sao Paulo), 2012. 67(12): p. 1373-8.
    108. Erler, J.T., et al., Lysyl oxidase is essential for hypoxia-induced metastasis. Nature, 2006. 440(7088): p. 1222-6.
    109. Ping, W., et al., Expression and significance of hypoxia inducible factor-1alpha and lysyl oxidase in non-small cell lung cancer. Asian Pac J Cancer Prev, 2013. 14(6): p. 3613-8.
    110. Wilgus, M.L., et al., Lysyl oxidase: a lung adenocarcinoma biomarker of invasion and survival. Cancer, 2011. 117(10): p. 2186-91.
    111. Mendez, M.G., S. Kojima, and R.D. Goldman, Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J, 2010. 24(6): p. 1838-51.
    112. Thiery, J.P., Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer, 2002. 2(6): p. 442-54.
    113. Jiang, J., et al., MicroRNA-27b suppresses growth and invasion of NSCLC cells by targeting Sp1. Tumour Biol, 2014. 35(10): p. 10019-23.
    114. Wan, L., et al., MiR-27b targets LIMK1 to inhibit growth and invasion of NSCLC cells. Mol Cell Biochem, 2014. 390(1-2): p. 85-91.
    115. Goto, Y., et al., The microRNA-23b/27b/24-1 cluster is a disease progression marker and tumor suppressor in prostate cancer. Oncotarget, 2014. 5(17): p. 7748-59.
    116. Veliceasa, D., et al., Therapeutic manipulation of angiogenesis with miR-27b. Vasc Cell, 2015. 7: p. 6.
    117. Betapudi, V., L.S. Licate, and T.T. Egelhoff, Distinct roles of nonmuscle myosin II isoforms in the regulation of MDA-MB-231 breast cancer cell spreading and migration. Cancer Res, 2006. 66(9): p. 4725-33.
    118. Derycke, L., et al., The role of non-muscle myosin IIA in aggregation and invasion of human MCF-7 breast cancer cells. Int J Dev Biol, 2011. 55(7-9): p. 835-40.
    119. Katono, K., et al., Prognostic significance of MYH9 expression in resected non-small cell lung cancer. PLoS One, 2015. 10(3): p. e0121460.
    120. Ramaswamy, S., et al., A molecular signature of metastasis in primary solid tumors. Nat Genet, 2003. 33(1): p. 49-54.
    121. Oleksiewicz, U., et al., COL1A1, PRPF40A, and UCP2 correlate with hypoxia markers in non-small cell lung cancer. J Cancer Res Clin Oncol, 2017.
    122. Nilsson, K. and G. Landberg, Subcellular localization, modification and protein complex formation of the cdk-inhibitor p16 in Rb-functional and Rb-inactivated tumor cells. Int J Cancer, 2006. 118(5): p. 1120-5.
    123. Souza-Rodrigues, E., et al., Proteomic analysis of p16ink4a-binding proteins. Proteomics, 2007. 7(22): p. 4102-11.
    124. Zhang, Q., et al., Nicotine induces hypoxia-inducible factor-1alpha expression in human lung cancer cells via nicotinic acetylcholine receptor-mediated signaling pathways. Clin Cancer Res, 2007. 13(16): p. 4686-94.
    125. Acosta, J.C., et al., Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell, 2008. 133(6): p. 1006-18.
    126. Behrens, P., et al., The Ets-1 transcription factor is up-regulated together with MMP 1 and MMP 9 in the stroma of pre-invasive breast cancer. J Pathol, 2001. 194(1): p. 43-50.
    127. Saeki, H., et al., Expression of ets-1 transcription factor is correlated with penetrating tumor progression in patients with squamous cell carcinoma of the esophagus. Cancer, 2000. 89(8): p. 1670-6.
    128. Logan, S.K., et al., Synergistic transcriptional activation of the tissue inhibitor of metalloproteinases-1 promoter via functional interaction of AP-1 and Ets-1 transcription factors. J Biol Chem, 1996. 271(2): p. 774-82.
    129. Pollak, M.N., E.S. Schernhammer, and S.E. Hankinson, Insulin-like growth factors and neoplasia. Nat Rev Cancer, 2004. 4(7): p. 505-18.
    130. Chen, H.X. and E. Sharon, IGF-1R as an anti-cancer target - trials and tribulations. 2013.
    131. Sachdev, D. and D. Yee, Disrupting insulin-like growth factor signaling as a potential cancer therapy. Mol Cancer Ther, 2007. 6(1): p. 1-12.
    132. Nurwidya, F., et al., Treatment with insulin-like growth factor 1 receptor inhibitor reverses hypoxia-induced epithelial-mesenchymal transition in non-small cell lung cancer. Biochem Biophys Res Commun, 2014. 455(3-4): p. 332-8.
    133. Bjorndahl, M., et al., Insulin-like growth factors 1 and 2 induce lymphangiogenesis in vivo. Proc Natl Acad Sci U S A, 2005. 102(43): p. 15593-8.
    134. Zhang, Q., et al., CCL19/CCR7 upregulates heparanase via specificity protein-1 (Sp1) to promote invasion of cell in lung cancer. Tumour Biol, 2013. 34(5): p. 2703-8.
    135. Tutunea-Fatan, E., et al., The role of CCL21/CCR7 chemokine axis in breast cancer-induced lymphangiogenesis. Mol Cancer, 2015. 14: p. 35.
    136. Lee, H.W., et al., Alpha-smooth muscle actin (ACTA2) is required for metastatic potential of human lung adenocarcinoma. Clin Cancer Res, 2013. 19(21): p. 5879-89.
    137. Rhodes, D.R., et al., Large-scale meta-analysis of cancer microarray data identifies common transcriptional profiles of neoplastic transformation and progression. Proc Natl Acad Sci U S A, 2004. 101(25): p. 9309-14.
    138. Wang, X.M., et al., Integrative analyses identify osteopontin, LAMB3 and ITGB1 as critical pro-metastatic genes for lung cancer. PLoS One, 2013. 8(2): p. e55714.
    139. Zheng, W., C. Jiang, and R. Li, Integrin and gene network analysis reveals that ITGA5 and ITGB1 are prognostic in non-small-cell lung cancer. Onco Targets Ther, 2016. 9: p. 2317-27.
    140. Birsoy, K., et al., Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides. Nature, 2014. 508(7494): p. 108-12.
    141. Popper, H.H., Progression and metastasis of lung cancer. Cancer Metastasis Rev, 2016. 35(1): p. 75-91.
    142. Kasapovic, J., et al., Antioxidant status and lipid peroxidation in the blood of breast cancer patients of different ages after chemotherapy with 5-fluorouracil, doxorubicin and cyclophosphamide. Clin Biochem, 2010. 43(16-17): p. 1287-93.
    143. Martinez-Outschoorn, U.E., et al., Cancer cells metabolically "fertilize" the tumor microenvironment with hydrogen peroxide, driving the Warburg effect: implications for PET imaging of human tumors. Cell Cycle, 2011. 10(15): p. 2504-20.
    144. Martinez-Outschoorn, U.E., et al., Cytokine production and inflammation drive autophagy in the tumor microenvironment: role of stromal caveolin-1 as a key regulator. Cell Cycle, 2011. 10(11): p. 1784-93.
    145. Whitaker-Menezes, D., et al., Evidence for a stromal-epithelial "lactate shuttle" in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fibroblasts. Cell Cycle, 2011. 10(11): p. 1772-83.
    146. Witkiewicz, A.K., et al., Molecular profiling of a lethal tumor microenvironment, as defined by stromal caveolin-1 status in breast cancers. Cell Cycle, 2011. 10(11): p. 1794-809.
    147. Lisanti, M.P., et al., Hydrogen peroxide fuels aging, inflammation, cancer metabolism and metastasis: the seed and soil also needs "fertilizer". Cell Cycle, 2011. 10(15): p. 2440-9.
    148. Ndele, J.K., K. Yoshioka, and J.W. Fisher, Hydrogen peroxide in the regulation of erythropoietin (Epo) gene expression in hepatocellular carcinoma cells. East Afr Med J, 1996. 73(2): p. 143-6.
    149. Tovari, J., et al., Erythropoietin in cancer: an update. Curr Mol Med, 2008. 8(6): p. 481-91.
    150. Kinsella, M.G., S.L. Bressler, and T.N. Wight, The regulated synthesis of versican, decorin, and biglycan: extracellular matrix proteoglycans that influence cellular phenotype. Crit Rev Eukaryot Gene Expr, 2004. 14(3): p. 203-34.
    151. Maishi, N., et al., Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan. Sci Rep, 2016. 6: p. 28039.
    152. Wadhwa, S., et al., Regulation, regulatory activities, and function of biglycan. Crit Rev Eukaryot Gene Expr, 2004. 14(4): p. 301-15.
    153. Yamamoto, K., et al., Biglycan is a specific marker and an autocrine angiogenic factor of tumour endothelial cells. Br J Cancer, 2012. 106(6): p. 1214-23.
    154. Wang, X. and Z. Chen, MicroRNA-19a functions as an oncogenic microRNA in non-small cell lung cancer by targeting the suppressor of cytokine signaling 1 and mediating STAT3 activation. Int J Mol Med, 2015. 35(3): p. 839-46.
    155. Labernadie, A., et al., A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol, 2017. 19(3): p. 224-237.
    156. Yamauchi, M., et al., N-cadherin expression is a potential survival mechanism of gefitinib-resistant lung cancer cells. Am J Cancer Res, 2011. 1(7): p. 823-33.
    157. Zhang, X., et al., N-cadherin expression is associated with acquisition of EMT phenotype and with enhanced invasion in erlotinib-resistant lung cancer cell lines. PLoS One, 2013. 8(3): p. e57692.
    158. Leithner, K., PEPCK in cancer cell starvation. Oncoscience, 2015. 2(10): p. 805-6.
    159. Ridley, A.J., Rho proteins and cancer. Breast Cancer Res Treat, 2004. 84(1): p. 13-9.
    160. Sahai, E. and C.J. Marshall, RHO-GTPases and cancer. Nat Rev Cancer, 2002. 2(2): p. 133-42.
    161. Mazieres, J., et al., Loss of RhoB expression in human lung cancer progression. Clin Cancer Res, 2004. 10(8): p. 2742-50.
    162. Sato, N., et al., RhoB is frequently downregulated in non-small-cell lung cancer and resides in the 2p24 homozygous deletion region of a lung cancer cell line. Int J Cancer, 2007. 120(3): p. 543-51.
    163. Wang, S., et al., Histone deacetylase 1 represses the small GTPase RhoB expression in human nonsmall lung carcinoma cell line. Oncogene, 2003. 22(40): p. 6204-13.
    164. Wang, Z., et al., DDX5 promotes proliferation and tumorigenesis of non-small-cell lung cancer cells by activating beta-catenin signaling pathway. Cancer Sci, 2015. 106(10): p. 1303-12.
    165. Lai, A.Y. and P.A. Wade, Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat Rev Cancer, 2011. 11(8): p. 588-96.
    166. King, E.R., et al., The anterior gradient homolog 3 (AGR3) gene is associated with differentiation and survival in ovarian cancer. Am J Surg Pathol, 2011. 35(6): p. 904-12.
    167. Obacz, J., et al., The role of AGR2 and AGR3 in cancer: similar but not identical. Eur J Cell Biol, 2015. 94(3-4): p. 139-47.
    168. Emerling, B.M., et al., PTEN regulates p300-dependent hypoxia-inducible factor 1 transcriptional activity through Forkhead transcription factor 3a (FOXO3a). Proc Natl Acad Sci U S A, 2008. 105(7): p. 2622-7.
    169. Furukawa-Hibi, Y., et al., FOXO forkhead transcription factors induce G(2)-M checkpoint in response to oxidative stress. J Biol Chem, 2002. 277(30): p. 26729-32.
    170. Kops, G.J., et al., Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature, 2002. 419(6904): p. 316-21.
    171. Tran, H., et al., DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science, 2002. 296(5567): p. 530-4.
    172. Blake, D.C., Jr., et al., FOXO3a elicits a pro-apoptotic transcription program and cellular response to human lung carcinogen nicotine-derived nitrosaminoketone (NNK). Lung Cancer, 2010. 67(1): p. 37-47.
    173. Huang, H. and D.J. Tindall, Dynamic FoxO transcription factors. J Cell Sci, 2007. 120(Pt 15): p. 2479-87.
    174. Nakamura, N., et al., Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN. Mol Cell Biol, 2000. 20(23): p. 8969-82.
    175. Skovseth, D.K., et al., Endostatin dramatically inhibits endothelial cell migration, vascular morphogenesis, and perivascular cell recruitment in vivo. Blood, 2005. 105(3): p. 1044-51.
    176. Zhang, Y., et al., Prognostic values of VEGF and endostatin with malignant pleural effusions in patients with lung cancer. Asian Pac J Cancer Prev, 2014. 15(19): p. 8435-40.
    177. Wang, Q., et al., Overexpression of serine/threonine-protein kinase-1 in pancreatic cancer tissue: Serine/threonine-protein kinase-1 knockdown increases the chemosensitivity of pancreatic cancer cells. Mol Med Rep, 2015. 12(1): p. 475-81.
    178. Xu, G.F., et al., The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell, 1990. 62(3): p. 599-608.
    179. Cawthon, R.M., et al., A major segment of the neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and point mutations. Cell, 1990. 62(1): p. 193-201.
    180. Martin, G.A., et al., The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21. Cell, 1990. 63(4): p. 843-9.
    181. Le, L.Q. and L.F. Parada, Tumor microenvironment and neurofibromatosis type I: connecting the GAPs. Oncogene, 2007. 26(32): p. 4609-16.
    182. Ozawa, T., et al., The neurofibromatosis type 1 gene product neurofibromin enhances cell motility by regulating actin filament dynamics via the Rho-ROCK-LIMK2-cofilin pathway. J Biol Chem, 2005. 280(47): p. 39524-33.
    183. Kato, Y., et al., Enhanced expression of Aggrus (T1alpha/podoplanin), a platelet-aggregation-inducing factor in lung squamous cell carcinoma. Tumour Biol, 2005. 26(4): p. 195-200.
    184. Wicki, A., et al., Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell, 2006. 9(4): p. 261-72.
    185. Cueni, L.N., et al., Tumor lymphangiogenesis and metastasis to lymph nodes induced by cancer cell expression of podoplanin. Am J Pathol, 2010. 177(2): p. 1004-16.
    186. Arenas, M.I., et al., E-, N- and P-cadherin, and alpha-, beta- and gamma-catenin protein expression in normal, hyperplastic and carcinomatous human prostate. Histochem J, 2000. 32(11): p. 659-67.
    187. Kumper, S. and A.J. Ridley, p120ctn and P-cadherin but not E-cadherin regulate cell motility and invasion of DU145 prostate cancer cells. PLoS One, 2010. 5(7): p. e11801.
    188. Paredes, J., et al., P-cadherin overexpression is an indicator of clinical outcome in invasive breast carcinomas and is associated with CDH3 promoter hypomethylation. Clin Cancer Res, 2005. 11(16): p. 5869-77.
    189. Sun, L., et al., P-cadherin promotes liver metastasis and is associated with poor prognosis in colon cancer. Am J Pathol, 2011. 179(1): p. 380-90.
    190. Yi, S., et al., N-cadherin and P-cadherin are biomarkers for invasion, metastasis, and poor prognosis of gallbladder carcinomas. Pathol Res Pract, 2014. 210(6): p. 363-8.
    191. Ng, M.R., et al., Substrate stiffness regulates cadherin-dependent collective migration through myosin-II contractility. J Cell Biol, 2012. 199(3): p. 545-63.
    192. Nguyen-Ngoc, K.V., et al., ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc Natl Acad Sci U S A, 2012. 109(39): p. E2595-604.
    193. Plutoni, C., et al., P-cadherin promotes collective cell migration via a Cdc42-mediated increase in mechanical forces. J Cell Biol, 2016. 212(2): p. 199-217.
    194. Davies, J.Q., et al., Leukocyte adhesion-GPCR EMR2 is aberrantly expressed in human breast carcinomas and is associated with patient survival. Oncol Rep, 2011. 25(3): p. 619-27.
    195. Safaee, M., et al., The role of epidermal growth factor-like module containing mucin-like hormone receptor 2 in human cancers. Oncol Rev, 2014. 8(1): p. 242.
    196. Lamb, J., et al., The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science, 2006. 313(5795): p. 1929-35.
    197. Bauman, J., et al., A Phase I Protocol of Hydralazine and Valproic Acid in Advanced, Previously Treated Solid Cancers. Transl Oncol, 2014.
    198. Trachtenberg, J., Ketoconazole therapy in advanced prostatic cancer. J Urol, 1987. 137(5): p. 959.
    199. Vanuytsel, L., et al., Ketoconazole therapy for advanced prostatic cancer: feasibility and treatment results. J Urol, 1987. 137(5): p. 905-8.
    200. Jones, G.R., Successful cancer therapy with promethazine: the rationale. Med Hypotheses, 1996. 46(1): p. 25-9.
    201. Drag-Zalesinska, M., et al., Anticancer properties of ester derivatives of betulin in human metastatic melanoma cells (Me-45). Cancer Cell Int, 2017. 17: p. 4.
    202. Seufferlein, T., et al., Mechanisms of nordihydroguaiaretic acid-induced growth inhibition and apoptosis in human cancer cells. Br J Cancer, 2002. 86(7): p. 1188-96.
    203. Li, X., et al., Nordihydroguaiaretic acid impairs prostate cancer cell migration and tumor metastasis by suppressing neuropilin 1. Oncotarget, 2016. 7(52): p. 86225-86238.
    204. Jendzelovsky, R., et al., Inhibition of GSK-3beta reverses the pro-apoptotic effect of proadifen (SKF-525A) in HT-29 colon adenocarcinoma cells. Toxicol In Vitro, 2012. 26(6): p. 775-82.
    205. Tennis, M.A., et al., Prostacyclin inhibits non-small cell lung cancer growth by a frizzled 9-dependent pathway that is blocked by secreted frizzled-related protein 1. Neoplasia, 2010. 12(3): p. 244-53.
    206. Chen, Y.H., et al., Methotrexate conjugated to gold nanoparticles inhibits tumor growth in a syngeneic lung tumor model. Mol Pharm, 2007. 4(5): p. 713-22.
    207. Djerassi, I., et al., Phase I study of high doses of methotrexate with citrovorum factor in patients with lung cancer. Cancer, 1972. 30(1): p. 22-30.
    208. Fan, X.M., et al., Five-lipoxygenase-activating protein inhibitor MK-886 induces apoptosis in gastric cancer through upregulation of p27kip1 and bax. J Gastroenterol Hepatol, 2004. 19(1): p. 31-7.
    209. Zhou, W., et al., The antipsychotic drug pimozide inhibits cell growth in prostate cancer through suppression of STAT3 activation. Int J Oncol, 2016. 48(1): p. 322-8.
    210. Rossi, T., et al., Mepacrine antagonises tumour cell growth induced by natural polyamines. Anticancer Res, 2008. 28(5A): p. 2765-8.
    211. El Sharkawi, F.Z., H.A. El Shemy, and H.M. Khaled, Possible anticancer activity of rosuvastatine, doxazosin, repaglinide and oxcarbazepin. Asian Pac J Cancer Prev, 2014. 15(1): p. 199-203.
    212. Borden, K.L. and B. Culjkovic-Kraljacic, Ribavirin as an anti-cancer therapy: acute myeloid leukemia and beyond? Leuk Lymphoma, 2010. 51(10): p. 1805-15.
    213. Gao, Z., et al., Procaine and procainamide inhibit the Wnt canonical pathway by promoter demethylation of WIF-1 in lung cancer cells. Oncol Rep, 2009. 22(6): p. 1479-84.
    214. Bilir, A., et al., Potentiation of cytotoxicity by combination of imatinib and chlorimipramine in glioma. Int J Oncol, 2008. 32(4): p. 829-39.
    215. Daley, E., et al., Chlorimipramine: a novel anticancer agent with a mitochondrial target. Biochem Biophys Res Commun, 2005. 328(2): p. 623-32.
    216. Chen, T.C., et al., The type IV phosphodiesterase inhibitor rolipram induces expression of the cell cycle inhibitors p21(Cip1) and p27(Kip1), resulting in growth inhibition, increased differentiation, and subsequent apoptosis of malignant A-172 glioma cells. Cancer Biol Ther, 2002. 1(3): p. 268-76.
    217. Armour, A.A. and C.L. Watkins, The challenge of targeting EGFR: experience with gefitinib in nonsmall cell lung cancer. Eur Respir Rev, 2010. 19(117): p. 186-96.
    218. Nurwidya, F., F. Takahashi, and K. Takahashi, Gefitinib in the treatment of nonsmall cell lung cancer with activating epidermal growth factor receptor mutation. J Nat Sci Biol Med, 2016. 7(2): p. 119-23.
    219. Chang, A., et al., Gefitinib (IRESSA) in patients of Asian origin with refractory advanced non-small cell lung cancer: subset analysis from the ISEL study. J Thorac Oncol, 2006. 1(8): p. 847-55.
    220. Jiang, H., Overview of gefitinib in non-small cell lung cancer: an Asian perspective. Jpn J Clin Oncol, 2009. 39(3): p. 137-50.
    221. Scagliotti, G., et al., The differential efficacy of pemetrexed according to NSCLC histology: a review of two Phase III studies. Oncologist, 2009. 14(3): p. 253-63.
    222. Tomasini, P., et al., Pemetrexed for advanced stage nonsquamous non-small cell lung cancer: latest evidence about its extended use and outcomes. Ther Adv Med Oncol, 2016. 8(3): p. 198-208.

    QR CODE