DOI QR코드

DOI QR Code

Survivin, a Promising Gene for Targeted Cancer Treatment

  • 발행 : 2016.08.01

초록

Drawbacks of conventional cancer treatments, with lack of specificity and cytotoxicity using current approaches, underlies the necessity for development of a novel approach, gene-directed cancer therapy. This has provided novel technological opportunities in vitro and in vivo. This review focuses on a member of an apoptosis inhibitor family, survivin, as a valuable target. Not only the gene but also its promoter are applicable in this context. This article is based on a literature survey, with especial attention to RNA interference as well as tumor-specific promoter action. The search engine and databases utilized were Science direct, PubMed, MEDLINE and Google. In addition to cell-cycle modulation, apoptosis inhibition, interaction in cell-signaling pathways, cancer-selective expression, survivin also may be considered as specific target through its promoter as a novel treatment for cancer. Our purpose in writing this article was to create awareness in researchers, emphasizing relation of survivin gene expression to potential cancer treatment. The principal result and major conclusion of this manuscript are that survivin structure, biological functions and applications of RNA interference systems as well as tumor-specific promoter activity are of major interest for cancer gene therapy.

키워드

참고문헌

  1. Altieri DC (2008). Survivin, cancer networks and pathwaydirected drug discovery. Nat Rev Cancer, 8, 61-70. https://doi.org/10.1038/nrc2293
  2. Altieri DC (2013). Targeting survivin in cancer. Cancer lett, 332, 225-8. https://doi.org/10.1016/j.canlet.2012.03.005
  3. Asanuma K, Tsuji N, Endoh T, et al (2004). Survivin enhances Fas ligand expression via up-regulation of specificity protein 1-mediated gene transcription in colon cancer cells. J Immunol, 172, 3922-9. https://doi.org/10.4049/jimmunol.172.6.3922
  4. Bao R, Connolly DC, Murphy M, et al (2002). Activation of cancer-specific gene expression by the survivin promoter. J Natl Cancer I, 94, 522-8. https://doi.org/10.1093/jnci/94.7.522
  5. Baratchi S, Kanwar RK, Kanwar JR (2010). Survivin: a target from brain cancer to neurodegenerative disease. Crit Rev Biochem Mol, 45, 535-54. https://doi.org/10.3109/10409238.2010.516740
  6. Cai J, Fu S, Tu Z, et al (2014). Survivin Gene Functions and Relationships between Expression and Prognosis in Patients with Nasopharyngeal Carcinoma. Asian Pac J Cancer Prev, 16, 2341-5.
  7. Cavalieri F, Beretta GL, Cui J, et al (2015). Redox-sensitive PEG-polypeptide nanoporous particles for survivin silencing in prostate cancer cells. Biomacromolecules, 16, 2168-78. https://doi.org/10.1021/acs.biomac.5b00562
  8. Chan KS, Wong CH, Huang YF, et al (2010). Survivin withdrawal by nuclear export failure as a physiological switch to commit cells to apoptosis. Cell Death Differ, 1, 57. https://doi.org/10.1038/cddis.2010.34
  9. Chen J-S, Liu JC, Shen L, et al (2004). Cancer-specific activation of the survivin promoter and its potential use in gene therapy. Cancer Gene Ther, 11, 740-7. https://doi.org/10.1038/sj.cgt.7700752
  10. Chen J, Li T, Liu Q, et al (2014a). Clinical and prognostic significance of HIF-1alpha, PTEN, CD44v6, and survivin for gastric cancer: a meta-analysis. PloS one, 9, 91842. https://doi.org/10.1371/journal.pone.0091842
  11. Chen P, Zhu J, Liu D-y, et al (2014b). Over-expression of survivin and VEGF in small-cell lung cancer may predict the poorer prognosis. Med Oncol, 31, 1-7.
  12. Chen XQ, Yang S, Li ZY, et al (2012). Effects and mechanism of downregulation of survivin expression by RNA interference on proliferation and apoptosis of lung cancer cells. Mol Med Rep, 5, 917-22. https://doi.org/10.3892/mmr.2012.755
  13. Chen YQ, Zhao CL, Li W (2009). Effect of hypoxia-inducible factor-1${\alpha}$ on transcription of survivin in non-small cell lung cancer. J Exp Clin Canc Res, 28, 29. https://doi.org/10.1186/1756-9966-28-29
  14. Chen Y, Wang X, Li W, et al (2011). Sp1 upregulates survivin expression in adenocarcinoma of lung cell line A549. Anat Rec, 294, 774-80. https://doi.org/10.1002/ar.21378
  15. Cheng SQ, Wang WL, Yan W, et al (2005). Knockdown of survivin gene expression by RNAi induces apoptosis in human hepatocellular carcinoma cell line SMMC-7721. World J Gastroentero, 11, 756-9. https://doi.org/10.3748/wjg.v11.i5.756
  16. Church DN, Talbot DC (2012). Survivin in solid tumors: rationale for development of inhibitors. Curr Oncol Rep, 14, 120-8. https://doi.org/10.1007/s11912-012-0215-2
  17. Croci DO, Cogno IS, Vittar NBR, et al (2008). Silencing survivin gene expression promotes apoptosis of human breast cancer cells through a caspase-independent pathway. J Cell Biochem, 105, 381-90. https://doi.org/10.1002/jcb.21836
  18. Duffy MJ, O D, Norma, Brennan DJ, et al (2007). Survivin: a promising tumor biomarker. Cancer Lett, 249, 49-60. https://doi.org/10.1016/j.canlet.2006.12.020
  19. Eberlein C, Opoku-Ansah G, Legg J, et al (2013). Survivin regulates endothelial cell proliferation, survival and angiogenesis. Cancer Res, 73, 1616. https://doi.org/10.1158/1538-7445.AM2013-1616
  20. Fan Y, Bergmann A (2008). Apoptosis-induced compensatory proliferation. The Cell is dead. Long live the Cell. Trends Cell Biol, 18, 467-73. https://doi.org/10.1016/j.tcb.2008.08.001
  21. Fang L, Shanqu L, Ping G, et al (2012). Gene therapy with RNAi targeting UHRF1 driven by tumor-specific promoter inhibits tumor growth and enhances the sensitivity of chemotherapeutic drug in breast cancer in vitro and in vivo. Cancer Chemoth Pharm, 69, 1079-87. https://doi.org/10.1007/s00280-011-1801-y
  22. Faversani A, Vaira V, Moro GP, et al (2014). Survivin family proteins as novel molecular determinants of doxorubicin resistance in organotypic human breast tumors. Breast Cancer Res, 16, 55.
  23. Felipe AV, Oliveira J, Chang P, et al (2013). RNA interference: a promising therapy for gastric cancer. Asian Pac J Cancer Prev, 15, 5509-15.
  24. Fu J, Bian M, Jiang Q, et al (2007). Roles of Aurora kinases in mitosis and tumorigenesis. Mol Cancer Res, 5, 1-10. https://doi.org/10.1158/1541-7786.MCR-06-0208
  25. Fulda S, Vucic D (2012). Targeting IAP proteins for therapeutic intervention in cancer. Nature Rev Drug Discov, 11, 109-24. https://doi.org/10.1038/nrd3627
  26. Ghosh JC, Dohi T, Kang BH, et al (2008). Hsp60 regulation of tumor cell apoptosis. J Biol Chem, 283, 5188-94. https://doi.org/10.1074/jbc.M705904200
  27. He S-Q, Rehman H, Gong M-g, et al (2007). Inhibiting survivin expression enhances TRAIL-induced tumoricidal activity in human hepatocellular carcinoma via cell cycle arrest. Cancer Biol Ther, 6, 1258-68. https://doi.org/10.4161/cbt.6.8.4444
  28. Hernandez JM, Farma JM, Coppola D, et al (2011). Expression of the antiapoptotic protein survivin in colon cancer. Clin Colorectal Cancer, 10, 188-93. https://doi.org/10.1016/j.clcc.2011.03.014
  29. Jha K, Shukla M, Pandey M (2012). Survivin expression and targeting in breast cancer. Surg Oncol, 21, 125-31. https://doi.org/10.1016/j.suronc.2011.01.001
  30. Kanwar JR, Kamalapuram SK, Kanwar RK (2011). Targeting survivin in cancer: the cell-signalling perspective. Drug Discov Today, 16, 485-94. https://doi.org/10.1016/j.drudis.2011.04.001
  31. Kanwar JR, Shen W-P, Kanwar RK, et al (2001). Effects of survivin antagonists on growth of established tumors and B7-1 immunogene therapy. J Natl Cancer I, 93, 1541-52. https://doi.org/10.1093/jnci/93.20.1541
  32. Kappler M, Bache M, Bartel F, et al (2004). Knockdown of survivin expression by small interfering RNA reduces the clonogenic survival of human sarcoma cell lines independently of p53. Cancer Gene Ther, 11, 186-93. https://doi.org/10.1038/sj.cgt.7700677
  33. Kappler M, Taubert H, Bartel F, et al (2005). Radiosensitization, after a combined treatment of survivin siRNA and irradiation, is correlated with the activation of caspases 3 and 7 in a wt-p53 sarcoma cell line, but not in a mt-p53 sarcoma cell line. Oncol Rep, 13, 167-72.
  34. Karami H, Baradaran B, Esfehani A, et al (2014). Downregulation of Mcl-1 by small interference RNA induces apoptosis and sensitizes HL-60 leukemia cells to etoposide. Asian Pac J Cancer Prev, 15, 629-35. https://doi.org/10.7314/APJCP.2014.15.2.629
  35. Kaufmann T, Strasser A, Jost P (2012). Fas death receptor signalling: roles of Bid and XIAP. Cell Death Differ, 19, 42-50. https://doi.org/10.1038/cdd.2011.121
  36. Kelly RJ, Lopez-Chavez A, Citrin D, et al (2011). Impacting tumor cell-fate by targeting the inhibitor of apoptosis protein survivin. Mol Cancer, 10, 1186.
  37. Knauer SK, Kramer OH, Knosel T, et al (2007). Nuclear export is essential for the tumor-promoting activity of survivin. FASEB J, 21, 207-16. https://doi.org/10.1096/fj.06-5741com
  38. Kunze D, Erdmann K, Froehner M, et al (2013). Enhanced inhibition of bladder cancer cell growth by simultaneous knockdown of antiapoptotic Bcl-xL and survivin in combination with chemotherapy. Int J Mol Sci, 14, 12297- 312. https://doi.org/10.3390/ijms140612297
  39. Li B, Liu X, Fan J, et al (2006). A survivin-mediated oncolytic adenovirus induces non-apoptotic cell death in lung cancer cells and shows antitumoral potential in vivo. J Gene Med, 8, 1232-42. https://doi.org/10.1002/jgm.953
  40. Li F, Altieri D (1999). Transcriptional analysis of human survivin gene expression. Biochem J, 344, 305-11.
  41. Li F, Ambrosini G, Chu EY, et al (1998). Control of apoptosis and mitotic spindle checkpoint by survivin. Nature, 396, 580-4. https://doi.org/10.1038/25141
  42. Li Y, Liu D, Zhou Y, et al (2015a). Silencing of Survivin Expression Leads to Reduced Proliferation and Cell Cycle Arrest in Cancer Cells. J Cancer, 6, 1187. https://doi.org/10.7150/jca.12437
  43. Li Y, Zhou Y, Zheng J, et al (2015b). Downregulation of survivin inhibits proliferation and migration of human gastric carcinoma cells. Int J Clin Exp Pathol, 8, 1731.
  44. Liu G-F, Zhao Q-G, Si L, et al (2009). Effects of survivin interference RNA on non-small cell lung carcinoma. Clin Invest Med, 32, 225-31. https://doi.org/10.25011/cim.v32i6.10656
  45. Liu X, Gao R, Dong Y, et al (2010). Survivin gene silencing sensitizes prostate cancer cells to selenium growth inhibition. BMC Cancer, 10, 418. https://doi.org/10.1186/1471-2407-10-418
  46. Liu Z, Cheng M, Cao MZ (2011). Potential targets for molecular imaging of apoptosis resistance in hepatocellular carcinoma. Biomed Imag Interv J, 7, 5.
  47. Lu B, Mu Y, Cao C, et al (2004). Survivin as a therapeutic target for radiation sensitization in lung cancer. Cancer Res, 64, 2840-5. https://doi.org/10.1158/0008-5472.CAN-03-3547
  48. Mita AC, Mita MM, Nawrocki ST, et al (2008). Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res, 14, 5000-5. https://doi.org/10.1158/1078-0432.CCR-08-0746
  49. Mobahat M, Narendran A, Riabowol K (2014). Survivin as a preferential target for cancer therapy. Int J Mol Sci, 15, 2494-516. https://doi.org/10.3390/ijms15022494
  50. Moraes GN, Vasconcelos FC, Delbue D, et al (2013). Analysis of survivin, XIAP and FoxM1 as potential chemoresistance factors in breast cancer cells. BMC Proc, 7, 27.
  51. Namgung R, Brumbach JH, Jeong JH, et al (2010). Dual bioresponsive gene delivery via reducible poly (amido amine) and survivin-inducible plasmid DNA. Biotechnol Lett, 32, 755-64. https://doi.org/10.1007/s10529-010-0219-7
  52. Ning S, Fuessel S, Kotzsch M, et al (2004). siRNA-mediated down-regulation of survivin inhibits bladder cancer cell growth. Int J Oncol, 25, 1065-136.
  53. Park DS, Hwang KE, Shim H, et al (2012). Elevated survivin is associated with a poor response to chemotherapy and reduced survival in lung cancer with malignant pleural effusions. Clin Exp Metastas, 29, 83-9. https://doi.org/10.1007/s10585-011-9431-7
  54. Pecorino L 2012. Molecular biology of cancer: mechanisms, targets, and therapeutics, Oxford University Press.
  55. Pop C, Salvesen GS (2009). Human caspases: activation, specificity, and regulation. J Biol Chem, 284, 21777-81. https://doi.org/10.1074/jbc.R800084200
  56. Raab M, Kramer A, Hehlgans S, et al (2015). Mitotic arrest and slippage induced by pharmacological inhibition of Polo-like kinase 1. Mol Oncol, 9, 140-54. https://doi.org/10.1016/j.molonc.2014.07.020
  57. Ramachandran PV, Ignacimuthu S (2012). RNA interference as a plausible anticancer therapeutic tool. Asian Pac J Cancer Prev, 13, 2445-52. https://doi.org/10.7314/APJCP.2012.13.6.2445
  58. Roberti A, Macaluso M, Giordano A (2009). Alterations in cell cycle regulatory genes in breast cancer. In 'Breast Cancer in the Post-Genomic Era', Eds Springer, 55-77
  59. Roche H, Vahdat L (2010). Treatment of metastatic breast cancer: second line and beyond. Ann Oncol, 22, 1000-1010.
  60. Ryan BM, O’Donovan N, Duffy MJ (2009). Survivin: a new target for anti-cancer therapy. Cancer Treat Rev, 35, 553-62. https://doi.org/10.1016/j.ctrv.2009.05.003
  61. Salvesen GS, Duckett CS (2002). IAP proteins: blocking the road to death's door. Nature Rev Mol Cell Biol, 3, 401-10.
  62. Shen J, Liu J, Long Y, et al (2009). Knockdown of survivin expression by siRNAs enhances chemosensitivity of prostate cancer cells and attenuates its tumorigenicity. Acta Bioch Bioph Sin, 41, 223-30. https://doi.org/10.1093/abbs/gmp005
  63. Shen J, Sun H, Meng Q, et al (2014). Simultaneous inhibition of tumor growth and angiogenesis for resistant hepatocellular carcinoma by co-delivery of sorafenib and survivin small hairpin RNA. Mol Pharm, 11, 3342-51. https://doi.org/10.1021/mp4006408
  64. Soleimanpour E, Babaei E (2014). Survivin as a potential target for cancer therapy. Asian Pac J Cancer Prev, 16, 6187-91.
  65. Stegh AH (2012). Targeting the p53 signaling pathway in cancer therapy-the promises, challenges and perils. Expert Opin Ther targets, 16, 67-83. https://doi.org/10.1517/14728222.2011.643299
  66. Tarasewicz E, Hamdan R, Straehla J, et al (2014). CDK4 inhibition and doxorubicin mediate breast cancer cell apoptosis through Smad3 and survivin. Cancer Biol Ther, 15, 1301-11. https://doi.org/10.4161/cbt.29693
  67. Thomas S, Quinn BA, Das SK, et al (2013). Targeting the Bcl- 2 family for cancer therapy. Expert Opin Ther targets, 17, 61-75. https://doi.org/10.1517/14728222.2013.733001
  68. Tokino T, Idogawa M, Sasaki Y (2015). P53 pathway and cancer: From bench to clinic. Pers Med Universe, 4, 1-3. https://doi.org/10.1016/j.pmu.2015.03.003
  69. Trabulo S, Cardoso A, Santos-Ferreira T, et al (2011). Survivin silencing as a promising strategy to enhance the sensitivity of cancer cells to chemotherapeutic agents. Mol Pharm, 8, 1120-31. https://doi.org/10.1021/mp100426e
  70. Tu SP, Jiang XH, Lin MC, et al (2003). Suppression of survivin expression inhibits in vivo tumorigenicity and angiogenesis in gastric cancer. Cancer Res, 63, 7724-32.
  71. Uchida H, Tanaka T, Sasaki K, et al (2004). Adenovirus-mediated transfer of siRNA against survivin induced apoptosis and attenuated tumor cell growth in vitro and in vivo. Mol Ther, 10, 162-71. https://doi.org/10.1016/j.ymthe.2004.05.006
  72. Ulasov IV, Zhu ZB, Tyler MA, et al (2007). Survivin-driven and fiber-modified oncolytic adenovirus exhibits potent antitumor activity in established intracranial glioma. Hum Gene Ther, 18, 589-602. https://doi.org/10.1089/hum.2007.002
  73. Unruhe B, Schroder E, Wunsch D, et al (2015). An old flame never dies: survivin in cancer and cellular senescence. Gerontology.
  74. van der Horst A, Lens SM (2014). Cell division: control of the chromosomal passenger complex in time and space. Chromosoma, 123, 25-42. https://doi.org/10.1007/s00412-013-0437-6
  75. Van Houdt WJ, Haviv YS, Lu B, et al (2006). The human survivin promoter: a novel transcriptional targeting strategy for treatment of glioma. J Neurosurg, 104, 583-92. https://doi.org/10.3171/jns.2006.104.4.583
  76. Vanoosthuyse V, Prykhozhij S, Hardwick KG (2007). Shugoshin 2 Regulates Localization of the Chromosomal Passenger proteins in fission yeast mitosis. Mol Biol Cell, 18, 1657-69. https://doi.org/10.1091/mbc.E06-10-0890
  77. Wang IC, Chen YJ, Hughes D, et al (2005). Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2- Cks1) ubiquitin ligase. Mol Cell Biol, 25, 10875-94. https://doi.org/10.1128/MCB.25.24.10875-10894.2005
  78. Wang R, Lin F, Wang X, et al (2007). The therapeutic potential of survivin promoter-driven siRNA on suppressing tumor growth and enhancing radiosensitivity of human cervical carcinoma cells via downregulating hTERT gene expression. Cancer Biol Ther, 6, 1301-7. https://doi.org/10.4161/cbt.6.8.4505
  79. Wang R, Wang X, Li B, et al (2009). Tumor-specific adenovirusmediated PUMA gene transfer using the survivin promoter enhances radiosensitivity of breast cancer cells in vitro and in vivo. Breast Cancer Res TR, 117, 45-54. https://doi.org/10.1007/s10549-008-0163-6
  80. Wang XJ, Li Y, Huang H, et al (2013). A simple and robust vector-based shRNA expression system used for RNA interference. PloS one, 8, 56110. https://doi.org/10.1371/journal.pone.0056110
  81. Wong R (2011). Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res, 30, 87. https://doi.org/10.1186/1756-9966-30-87
  82. Wu J, Apontes P, Song L, et al (2007). Molecular mechanism of upregulation of survivin transcription by the AT-rich DNA-binding ligand, Hoechst33342: evidence for survivin involvement in drug resistance. Nucleic acids Res, 35, 2390-402. https://doi.org/10.1093/nar/gkm149
  83. Xia H, Chen S, Huang H, et al (2015). Survivin over-expression is correlated with a poor prognosis in esophageal cancer patients. Clin Chim Acta, 446, 82-5. https://doi.org/10.1016/j.cca.2015.04.009
  84. Xiang J, Ouyang Y, Cui Y, et al (2012). Silencing of Notch3 Using shRNA driven by survivin promoter inhibits growth and promotes apoptosis of human T-cell acute lymphoblastic leukemia cells. Clin Lymphoma Myeloma Leuk, 12, 59-65. https://doi.org/10.1016/j.clml.2011.07.005
  85. Xie Q, Wu Q, Mack S, et al (2015). CDC20 maintains tumor initiating cells. Oncotarget, 6, 13241-54. https://doi.org/10.18632/oncotarget.3676
  86. Yang CT, Li JM, Weng HH, et al (2010a). Adenovirusmediated transfer of siRNA against survivin enhances the radiosensitivity of human non-small cell lung cancer cells. Cancer Gene Ther, 17, 120-30. https://doi.org/10.1038/cgt.2009.55
  87. Yang D, Song X, Zhang J, et al (2010b). Therapeutic potential of siRNA-mediated combined knockdown of the IAP genes (Livin, XIAP, and Survivin) on human bladder cancer T24 cells. Acta Bioch Bioph Sin, 42, 137-44. https://doi.org/10.1093/abbs/gmp118
  88. Yang L, Cao Z, Li F, et al (2004). Tumor-specific gene expression using the survivin promoter is further increased by hypoxia. Gene Ther, 11, 1215-23. https://doi.org/10.1038/sj.gt.3302280
  89. Yang Y, Gao Y, Chen L, et al (2011). Downregulation of survivin expression and enhanced chemosensitivity of MCF-7 cells to adriamycin by PDMAE/survivin shRNA complex nanoparticles. Int J Pharm, 405, 188-95. https://doi.org/10.1016/j.ijpharm.2010.11.047
  90. Zhang H-Z, Wang Y, Gao P, et al (2006). Silencing stathmin gene expression by survivin promoter-driven siRNA vector to reverse malignant phenotype of tumor cells. Cancer Biol Ther, 5, 1457-61. https://doi.org/10.4161/cbt.5.11.3272
  91. Zhu ZB, Chen Y, Makhija SK, et al (2006a). Survivin promoterbased conditionally replicative adenoviruses target cholangiocarcinoma. Int J Oncol, 29, 1319-29.
  92. Zhu ZB, Makhija SK, Lu B, et al (2004). Transcriptional targeting of tumors with a novel tumor-specific survivin promoter. Cancer Gene Ther, 11, 256-62. https://doi.org/10.1038/sj.cgt.7700679
  93. Zhu ZB, Makhija SK, Lu B, et al (2005). Incorporating the survivin promoter in an infectivity enhanced CRAd-analysis of oncolysis and anti-tumor effects in vitro and in vivo. Int J Oncol, 27, 237-46.
  94. Zhu ZB, Makhija SK, Lu B, et al (2006b). Targeting mesothelioma using an infectivity enhanced survivinconditionally replicative adenoviruses. J Thorac Oncol, 1, 701.