帳號:guest(44.223.31.148)          離開系統
字體大小: 字級放大   字級縮小   預設字形  

詳目顯示

以作者查詢圖書館館藏以作者查詢臺灣博碩士論文系統以作者查詢全國書目
作者(中文):葉室亨
作者(外文):Yeh, Shih-Heng
論文名稱(中文):A Network Flow Approach to Predict Drug Targets from Microarray Data
論文名稱(外文):藉由生物晶片與網路流量方法來預測藥物標的
指導教授(中文):蘇豐文
指導教授(外文):Soo, Von-Wun
學位類別:碩士
校院名稱:國立清華大學
系所名稱:資訊系統與應用研究所
學號:9765535
出版年(民國):99
畢業學年度:98
語文別:英文
論文頁數:50
中文關鍵詞:藥物標的網路流量攝護腺癌生物晶片
外文關鍵詞:Drug targetNetwork flowProstate cancerMicroarray data
相關次數:
  • 推薦推薦:0
  • 點閱點閱:80
  • 評分評分:*****
  • 下載下載:2
  • 收藏收藏:0
在系統生物學中,具備系統方法的藥物發展是逐漸新起的領域,其目的是整合大規模的互動資料與實驗來闡述疾病的現象。在治療癌症的研究議題之中的藥物開發過程,尤其以如何發現與設計藥物標的為重要課題。然而,以往的藥物標的本身是一個試誤的臨床試驗,面對複合性疾病而言,將會花費較多時間與成本,而開發一個具有系統性的方法去預測可能的藥物標的組合成為一個具有挑戰性的任務。
本研究將針對設計藥物標的方法,提出新的系統架構,利用基於網路流量的方法下,去辨識出有效的藥物標的,以及呈現藥物標的之不同組合下,減少與其他不相關的組合比較次數的搜尋空間。我們使用攝護腺癌的生物晶片與DrugBank生物網站來做為我們的實驗測試來源與定義。藉由此方法,我們成功辨識出潛在的藥物標的具有非常強烈地與已知的攝護腺癌的治療藥物有所相關,並且發現更多其他潛在藥物標的與他們的藥物組合,是現今正吸引著生物學學家來探討。
System approach for medicine discovery is an emerging discipline in systems biology that aims at integrating large scale of the interaction data and experimental data to elucidate diseases. It also raises new issues in the drug discovery and design in development process for cancer treatment. However, drug target are still a trial-and-error experimental stage in clinical testing and it is a challenging task to develop a prediction model that can systematically detect the possible drug targets and their combinations to deal with a complex disease. We present a network flow-based approach to identify the effective drug targets and reduce the search space for drug target combination comparing with exhaustive search. We use the prostate cancer microarray data and DrugBank database as our test domain. We successfully identify potential drug targets which are strongly related to the well known drugs for prostate cancer treatment and also discover more potential drug targets and their combinations which attract the attention to biologists at present.
摘要.................................................................................................................................i
Abstraction………………………………………………………………………….....ii
1 Introduction 1
2 System Architecture 7
2.1 Network reconstruction from microarray data and protein-protein interactions database 8
2.2 The network flow approach 12
2.3 Filtering search space of drug target combination based on heuristic rules 20
3 results 22
3.1 Networks 23
3.2 Drug target discovery 25
3.3 The maximum flow and biological processes in recent prostate cancer drugs 31
3.4 The discovery of combination of drug targets 35
3.5 The effect of the partially-directed and directed graph in our method 39
3.6 The execution time of our methods 42
4 Conclusions 43
Acknowledgements 44
References 45
Appendix 50
□ Appendix 1 : The all disease-related genes (include disease-causing genes) 50
□ Appendix 2 :The flow and damage of drug targets 50
□ Appendix 3 : Top 5% drug targets flow to each disease-related genes 50
□ Appendix 4 : Prostate Cancer Drugs in the network appeared 50
Appendix 1 : All disease-related genes (include disease-causing genes) 1
Appendix 2 : The flow and damage of drug targets 2
Appendix 3 : Top 5% drug targets flow to each disease-related genes 1
Appendix 4 : Prostate Cancer Drugs in the network appeared 5
[1] B. Vogelstein, and K.W. Kinzler, Cancer genes and the pathways they control. Nat Med 10(8) (2004), 789-99.
[2] C. Smith, Hitting the target, Nature 422 (2003) 341-347
[3] J. Drews, Drug Discovery: A historical perspective, Science 287 (2000) 1960-1964
[4] J. Lamb, E. Crawford, D. Peck, J. Modell, I. Blat, M. Wrobel, J. Lerner, J. Brunet, A. Subramanian, K. Ross, M. Reich, H. Hieronymus, G.Wei, S.Arm- strong, S. Haggarty, P. Clemons, R.Wei, S. Carr, E. Lander, T. Golub, The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313 (2006) 1929-35
[5] M. Campillos, M. Kuhn, A. Gavin, L. Jensen, P. Bork, Drug target identification using side-effect similarity, Science 321 (2008) 263-6
[6] T. Takenaka, Classical vx. Reverse pharmacology in drug discovery, BJU International, 88 (2001) 7-10
[7] S. Imoto and Tamada, Computational strategy for discovering druggable gene networks from genome-wide RNA expression profiles, Pacific Symposium on Biocomputing (2006)
[8] G.R. Zimmermann, J. Lehár and C.T. Keith, Multi-target therapeutics: when the whole is greater than the sum of the parts, Drug Discovery Today 12(1-2) (2007), 34-42
[9] S. Loewe, The problem of synergism and antagonism of combined drugs. Arzneimittel-Forschung 3 (1953)285-90.
[10] C. Bliss, e toxicity of poisons applied jointly. Ann Appl Biol (1939) 26: 585–615
[11] T.C. Chou, Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies. Pharmacol Rev. 58 (2006) 621-680.
[12] S. Huang, Rational drug discovery: what can we learn from regulatory networks? Drug Discovery Today 7(20)(2002) , 163-169
[13] P. Csermely, Strong links are important, but weak links stabilize them, Trends Biochem Sci 29(7)(2004), 331-334.
[14] F. Hormozdiari, R. Salari ,V. Bafna, and S.C. Sahinalp , Protein-protein interaction network evaluation for identifying potential drug targets, J Comput Biol 17(5) (2010) , 669-684
[15] N. Lemke, F. Herdia, C.K. Barcellos.dos, A.N. Reis and Mom-bach J.C.M. Essentiality and damage in metabolic networks. Bioinformatics, 20(1) (2004), 115-119
[16] P. Sridhar, B. Song, T. Kahveci and S. Ranka, Mining metabolic networks for optimal drug targets. Pacific Symposium on Biocomputing 13 (2008) 291-302.
[17] D. Brown and G. Superti-Furga , Rediscovering the sweet spot in drug discovery, Drug Discov Today 8(23)(2003), 1067-1077.
[18] X. Wu, R. Jiang, M.Q. Zhang, S. Li, Network-based global inference of human disease genes. Mol Syst Biol 4 (2008) 189
[19] A.JM Walhout, Unraveling transcription regulatory networks by protein–DNA and protein–protein interaction mapping, Genome Research, 16 (2006) 1445-1454.
[20] C.Y. Wang and B.S. Chen, Integrated cellular network of transcription regulations and protein-protein interactions, BMC Systems Biology (2010)
[21] O. Troyanskaya, M. Cantor, G. Sherlock, P. Brown, T. Hastie, R. Tibshirani, D. Botstein, R.B. Altman Missing value estimation methods for DNA microarrays, Bioinformatics, 17 (2001) 520–525.
[22] H.Y. Yeh, S.W. Cheng, Y.C Lin, C.Y. Yeh, S.F. Lin, V.W. Soo, Identifying significant genetic regulatory networks in the prostate cancer from microarray data based on transcription factor analysis and conditional independency, BMC Med Genomics(2009) 70.
[23] A. Bairoch, The ENZYME database in 2000, Nucleic Acids Res. 28 (2000) 304–305.
[24] V. Curwen, E. Eyras, T.D. Andrews, L. Clarke, E. Mongin, S.M. Searle and M. Clamp, The Ensembl Automatic Gene Annotation System, Genome Research 14 (2004) 942–950.
[25] H.C. Liu, C. R. Arias and V.W. Soo, BioIR: An approach to public domain resource integration of human protein-protein interaction, The proceeding of the Asia Pacific Bioinformatics Conference (APBC) (2009)
[26] X. Ren and X.S. Zhang, A Novel Approach for Pathway Inference Based on Network Flow. The Third International Symposium on Optimization and Systems Biology (2009) 468–474.
[27] A. Goldberg and R. Tarjan A new approach to the maximum-flow problem, Journal of the ACM (JACM), 35(4) (1988), 921 – 940
[28] D.S. Wishart, C. Knox, A.C Guo, D. Cheng, S. Shrivastava, D. Tzur, B. Gautam, M. Hassanali, DrugBank: a knowledgebase for drugs, drug actions and drug targets, Nucleic acids research. 36(Database issue) (2008) D901-6.
[29] B.V. Cherkassky and AV. Golderg, On implementing push relabel method for the maximum flow problem, Algorithmica 19 (1994) 390-410,
[30] R. W. Floyd, R.Algorithm 97: Shortest Path, Communications of the ACM 5 (1992) 345
[31] P. Sridhar, T. Kahveci and S. Ranka, An iterative algorithm for metabolic network-based drug target identification. Pacific Symposium on Biocomputing 12 (2007) 88-99.
[32] J. Lapointe, C. Li, J. P. Higgins, M. V. de Rijn, E. Bair, K. Montgomery, M.Ferrari, L. Egevad, W. Rayford, U. Bergerheim, P. Ekman, A. M. DeMarzo, R. Tibshirani, D. Botstein, P. O Brown, J. D Brooks, and J. R. Pollacka, Gene expression profiling identifies clinically relevant subtypes of prostate cancer, The National Academy of Sciences., 101 (2004) 811–881
[33] G. Sherlock, T. H. Boussard, A. Kasarskis, G. Binkley, J. C. Matese, S. S. Dwight, M. Kaloper, S. Weng, H. Jin, C. A. Ball, M. B. Eisen, P. T. Spellman, D. Brown, P. O. Botstein, and C. J. Michael, The Stanford Microarray Database, Nucleic Acids Research 29 (2001) 152-155.
[34] M. Kanehisa, S. Goto, S. Kawashima, Y. Okuno and M. Hattori, “The KEGG resource for deciphering the genome,” Nucleic Acids Res. vol. 1, (2004), pp. 277-80.
[35] V.A. McKusuck,”Mendelian inheritance in man and its online version,”Am J Hum Genet, 80(2007) 588-604.
[36] L.C. Li, H. Zhao, H. Shiina, C.J.Kane and R. Dahiya, ”PGDB: a curated and integrated database of genes related to the prostate,” Nucleic Acids Res, vol.31, (2003), pp. 291-293.
[37] H.Savli,A. Szendroi,l. Romics and B.Nagy, ”Gene network and canonical pathway analysis in prostate cancer: a microarray study,” Experimental and Molecular Medicine,vol.40 no.2, (2008)176-185.
[38] H.Wang, D.Yu, S. Agrawal and R. Zhang, Experimental therapy of human prostate cancer y inhibiting mdm2 expression with novel mixed-backbone antisense oligonucletides: in vitro and in vivo activities and mechanisms, Prostate, 54 (2003) 194-205.
[39] J.H. Kim, C. Xu, Y.S. Keum, B. Reddy, A. Conney and ,A.N. Tony Kong, Inhibition of EGFR signaling in human prostate cancer PC-3 cells by combination treatment with ß-phenylethyl isothiocyanate and curcumin. Carcinogenesis, 27 (2006) 475–482.
[40] C. Festuccia, G.L. Gravina, L. Biordi, S. D'Ascenzo, V. Dolo, C. Ficorella, E. Ricevuto and V. Tombolini Effects of EGFR tyrosine kinase inhibitor erlotinib in prostate cancer cells in vitro. The Prostate, 69 (2009) 1529-37.
[41] M.A. Rochester, J. Riedemann, G.O. Hellawell, S.F. Brewster and V.M. Macaulay, Silencing of the IGF1R gene enhances sensitivity to DNA-damaging agents in both PTEN wild-type and mutant human prostate cancer. Cancer Gene Therapy 12 (2005) 90–100.
[42] F. Junya, J.W. Christopher, P. M. Brett, E.G. Martin and E.C. Michael, ATL1101 Prostate Cancer Drug Tumour Suppression Data to be Presented at US Cancer Meeting, Antisense Therapeutics (2010).
[43] A. Chen, YW. Tsau and CH. Lin, Novel methods to identify biologically relevant genes for leukemia and prostate cancer from gene expression profiles, BMC Genomics 11 (2010), 274.
[44] A. Subramanian, P. Tamayo, V.K. Mootha, S. Mukherjee, B.L. Ebert, M.A. Gillette, A. Paulovich, S.L. Pomeroy, T.R. Golub, E.S. Lander and J.P. Mesirov, Gene set enrichment analysis: A knowledge-based approach for interpreting genome, Proc Natl Acad Sci U S A. (2005) 15545-50.
[45] S. Hardy and M.L. Tremblay, Protein tyrosine phosphatases: new markers and targets in oncology? Curr Oncol, 15(1) (2008) 5-8.
[46] R. Delsite and D. Djakiew, Anti-proliferative effect of the kinase inhibitor K252a on human prostatic carcinoma cell lines, Journal of Andrology 17 (1996), 481-490
[47] M. Marcelli, M. Marani, X. Li, L. Sturgis, S.J. Haidacher, J.A. Trial, R. Mannucci, I. Nicoletti and L. Denner, Heterogeneous apoptotic responses of prostate cancer cell lines identify an association between sensitivity to staurosporine-induced apoptosis, expression of Bcl-2 family members, and caspase activation, Prostate 42(4) (2000) 260-73.
[48] D. Trudel, Y. Fradet, F. Meyer, F. Harel and B. Têtu , Significance of MMP-2 expression in prostate cancer: an immunohistochemical study, Cancer Res 63 (23) (2003) 8511-8515.
[49] O. Gautschi , J. Heighway, P.C. Mack, P.R. Purnell, P.N. Jr. Lara and D.R. Gandara, Aurora kinases as anticancer drug targets, Clin Cancer Res 14 (6) (2008) 1639-48.
[50] V. Flamand, H. Zhao and D.M. Peehl, Targeting monoamine oxidase A in advanced prostate cancer , J Cancer Res Clin Oncol. (2010).
[51] C.S. Mantzoros, A. Tzonou, L.B. Signorello, M. Stampfer, D. Trichopoulos and H.O. Adami, Insulin-like growth factor 1 in relation to prostate cancer and benign prostatic hyperplasia, Br J Cancer ;76(9) (1997) 1115-1118.
[52] S. Sarfaraz, F.Afaq, VM. Adhami, A.Malik and H.Mukhtar, Cannabinoid receptor agonist-induced apoptosis of human prostate cancer cells Incap proceeds through sustained activation of erk1/2 leading to g1 cell cycle arrest, J. Biol, Chem., 281, (2006) 39480-39491.
[53] C. Gausterer, M. Muller and B. Strobl, In Vivo Target Validation: Methodology and Case Studies on the Janus Kinase Tyk2, Anti-Inflammatory & Anti-Allergy Agents in Medicinal Chemistry 6 (2007) 29-45.
[54] I. Tannock, M. Gosbodarowicz, and W. Meakin, Treatment of metastatic prostate cancer with low dose prednisone: Evaluation of pain and quality of life as pragmatic indices of response, J Clin Oncol 7 (1989) 590-7.
[55] K. Rao, S. Goodin, M.J. Levitt, N. Dave, W.J. Shin, Y. Lin, T. Capanna, S. Doyle-Lindrud, P. Juvidian and R.S. Dipaola, A phase II trial of imatinib mesylate in patients with prostate specific antigen progression after local therapy for prostate cancer. The Prostate, 62 (2005) 115-122.
[56] T.L. Gillison, L.J. Appleman, D.M Friedland, T.L. Evans, P.N. Lara, W.E. Gooding, D.E. Lenzner, H.M. Strausser, J.R. Gingrich and G.S. Chatta, Docetaxel and imatinib every 21 days for castration resistant prostate cancer: A phase II trial, American Society of Clinical Oncology (2009).
 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *