DOI QR코드

DOI QR Code

Endpoint of Cancer Treatment: Targeted Therapies

  • Topcul, Mehmet (Department of Biology, Faculty of Science, Istanbul University) ;
  • Cetin, Idil (Department of Radiobiology, Institute of Science, Istanbul University)
  • Published : 2014.06.15

Abstract

Nowadays there are several limitations in cancer treatment. One of these is the use of conventional medicines which not only target cancer cells and thus also cause high toxicity precluding effective treatment. Recent elucidation of mechanisms that cause cancer has led to discovery of novel key molecules and pathways which have have become successful targets for the treatments that eliminate only cancer cells. These so-called targeted therapies offer new hope for millions of cancer patients, as briefly reveiwed here focusing on different types of agents, like PARP, CDK, tyrosine kinase, farnysyl transferase and proteasome inhibitors, monoclonal antibodies and antiangiogenic agents.

Keywords

References

  1. Abdalla MY (2011). Glutathione as potential target for cancer therapy; more or less is good? Jordan J Biol Sci, 4, 119-24.
  2. Advani SH (2010). Targeting mTOR pathway: a new concept in cancer therapy. Indian J Med Paediatr Oncol, 31, 132-6.
  3. Agrawal AG, Somani RR (2011). Farnesyltransferase inhibitor sin cancer treatment. In 'Current Cancer Treatment - Novel Beyond Conventional Approaches', Ed. Ozdemir O. InTech, Croatia, pp149-172.
  4. Alanazi M, Pathan AAK, Shaik JP, Amri AA, Parine NR (2013). The C allele of a synonymous SNP (rs1805414, Ala284Ala) in PARP1 is a risk factor for susceptibility to breast cancer in Saudi patients. Asian Pac J Cancer Prev, 14, 3051-6. https://doi.org/10.7314/APJCP.2013.14.5.3051
  5. Aqeilan RI, Zanesi N, Croce CM (2009). Environmental, genetic, and viral causes of cancer. In 'The biology and treatment of cancer', Eds. Pardee AB, Stein GS. John Wiley & Sons, Inc, New Jersey, pp 35-56.
  6. Arora A, Scholar EM (2005). Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther, 315, 971-9. https://doi.org/10.1124/jpet.105.084145
  7. Blagden S, Bono JD (2005). Drugging cell cycle kinases in cancer therapy. Curr Drug Targets, 6, 325-35. https://doi.org/10.2174/1389450053765824
  8. Bolden JE, Peart MJ, Johnstone RW (2006). Anticancer activities of histone deacetylase inhibitors. Nature Rev Drug Discovery, 5, 769-84. https://doi.org/10.1038/nrd2133
  9. Canavese M, Santo L, Raje N (2012). Cyclin dependent kinases in cancer: Potential for therapeutic intervention. Cancer Biol Ther, 13, 451-7. https://doi.org/10.4161/cbt.19589
  10. Carter G, Lemoine NR (1993). Antisense technology for cancer therapy: does it make sense? Br J Cancer, 67, 869-76. https://doi.org/10.1038/bjc.1993.164
  11. Cetin I, Topcul M (2012). Cancer stem cells in oncology. J BUON, 17, 644-8.
  12. Chauhan D, Hideshima T, Mitsiades C, Richardson P, Anderson KC (2005). Proteasome inhibitor therapy in multiple myeloma. Mol Cancer Ther, 4, 686-92.
  13. Cheetham GM (2004). Novel protein kinases and molecular mechanisms of autoinhibition. Curr Opin Struct Biol, 14, 700-5. https://doi.org/10.1016/j.sbi.2004.10.011
  14. Cheng L, Zhang S, Davidson DD, Montironi R, Lopez-Beltran A (2009). Implications of cancer stem cells for cancer therapy. In 'Cancer drug discovery and development', Eds. Bagley RG, Teicher BA. Humana Press, USA, pp 255-262.
  15. Chumsri S, Howes T, Bao T, Sabnis G, Brodie A (2011). Aromatase, aromatase inhibitors, and breast cancer. J Steroid Biochem Mol Biol, 125, 13-22. https://doi.org/10.1016/j.jsbmb.2011.02.001
  16. Clemens JA, Bennett DR, Black LJ, Jones CD (1983). Effects of a new anti-estrogen, keoxifene (LY156758), on growth of carcinogen-induced mammary tumors and on LH and prolactin secretion. Life Sci, 32, 2869-75. https://doi.org/10.1016/0024-3205(83)90323-5
  17. D'Amours D, Desnoyers S, D'Silva I, Poirier GG (1999). Poly (ADPribosyl) ation reactions in the regulation of nuclear functions. Biochem J, 342, 249-68. https://doi.org/10.1042/0264-6021:3420249
  18. Diaz-Padilla I, Siu LL, Duran I (2009). Cyclin-dependent kinase inhibitors as potential targeted anticancer agents. Investigational New Drugs, 27, 586-94. https://doi.org/10.1007/s10637-009-9236-6
  19. Duan D, Li SL, Zhu YQ, et al (2012). Radioimmunoimaging with mixed monoclonal antibodies of nude mice bearing human lung adenocarcinoma xenografts. Asian Pac J Cancer Prev, 13, 4255-61. https://doi.org/10.7314/APJCP.2012.13.9.4255
  20. Foreman KE, Rizzo P, Osipo C, Miele L (2009). The cancer stem cell hypothesis. In 'Cancer drug discovery and development', Eds. Bagley RG, Teicher BA. Humana Press, USA, pp 3-14.
  21. Gao XH, Yang XQ, Wang BC, Liu SP, Wang FB (2013). Overexpression of twist and matrix metalloproteinase-9 with metastasis and prognosis in gastric cancer. Asian Pac J Cancer Prev, 14, 5055-60. https://doi.org/10.7314/APJCP.2013.14.9.5055
  22. Genin E, Reboud-Ravaux M, Vidal J (2010). Proteasome inhibitors: recent advances and new perspectives in medicinal chemistry. Current Topics in Medicinal Chemistry, 10, 232-56. https://doi.org/10.2174/156802610790725515
  23. Gollob JA, Wilhelm S, Carter C, Kelley SL (2006). Role of Raf kinase in cancer: therapeutic potential of targeting the Raf/MEK/ERK signal transduction pathway. Seminars in Oncology, 33, 392-406. https://doi.org/10.1053/j.seminoncol.2006.04.002
  24. Gryder BE, Sodji QH, Oyelere AK (2012). Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Medicinal Chemistry, 4, 505-24. https://doi.org/10.4155/fmc.12.3
  25. Guo YT, Hou QY, Wang N (2011). Monoclonal antibodies in cancer therapy. Clin Oncol Cancer Res, 8, 215-9. https://doi.org/10.1007/s11805-011-0583-7
  26. Gupta S, Singh RP, Rabadia N, Patel G, Panchal H (2011). Antisense technology. Int J Pharmaceutical Sci Review and Res, 9, 38-45.
  27. Hamanaka RB, Chandel NS (2012). Targeting glucose metabolism for cancer therapy. J Exp Med, 209, 211-5. https://doi.org/10.1084/jem.20120162
  28. Hebert JR (2009). Epidemiology: identifying cancer's causes. In 'The biology and treatment of cancer', Eds. Pardee AB, Stein GS. John Wiley & Sons Inc, New Jersey, pp 223-255.
  29. Hejmadi M (2009). How cancer arises? In 'Introduction to cancer biology', Ed. Hejmadi M. Ventus Publishing ApS, United Kingdom, pp 6-12.
  30. Hidalgo M, Eckhardt SG (2001). Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst, 93, 178-93. https://doi.org/10.1093/jnci/93.3.178
  31. Howell A (2012). Genesis and outcome of a breast cancer trial to develop the aromatase inhibitor anastrozole. Clin Chem, 58, 782-3. https://doi.org/10.1373/clinchem.2011.181206
  32. Hurwitz HI (2004). Introduction: targeting angiogenesis in cancer therapy. The Oncologist, 9, 1.
  33. Jensen EV, Jordan VC (2003). The estrogen receptor: a model for molecular medicine. Clin Cancer Res, 9, 1980-9.
  34. Johnston SRD (2001). Farnesyl transferase inhibitors: a novel targeted therapy for cancer. Lancet Oncol, 2, 18-26. https://doi.org/10.1016/S1470-2045(00)00191-1
  35. Jones NP, Schulze A (2012). Targeting cancer metabolism-aiming at a tumour's sweet-spot. Drug Discov Today, 17, 232-41. https://doi.org/10.1016/j.drudis.2011.12.017
  36. Jordan VC, Collins MM, Rowsby L, Prestwich G (1977). A monohydroxylated metabolite of tamoxifen with potent antioestrogen activity. J Endocrinol, 75, 305-16. https://doi.org/10.1677/joe.0.0750305
  37. Karamysheva AF (2008). Mechanisms of angiogenesis. Biochem, 73, 751-62.
  38. Knockaert M, Greengard P, Meije L (2002). Pharmacological inhibitors of cyclin-dependent kinases. Trends in Pharmacoll Sci, 23, 417-25. https://doi.org/10.1016/S0165-6147(02)02071-0
  39. Kondapalli L, Soltani K, Lacouture ME (2005). The promise of molecular targeted therapies: protein kinase inhibitors in the treatment of cutaneous malignancies. J Am Acad Dermatol, 53, 291-302. https://doi.org/10.1016/j.jaad.2005.02.011
  40. Kushner DM, Silverman RH (2000). Antisense cancer therapy: the state of the science. Curr Oncol Reports, 2, 23-30. https://doi.org/10.1007/s11912-000-0007-y
  41. Lindahl T, Satoh MS, Poirier GG, Klungland A (1995). Post-translational modification of poly(ADP-ribose)polymerase induced by DNA strand breaks. Trends Biochem Sci, 20, 405-12. https://doi.org/10.1016/S0968-0004(00)89089-1
  42. Liu Z, Wang L, Zhang LN, et al (2012). Expression and clinical significance of mTOR in surgically resected non-small cell lung cancer tissues: a case control study. Asian Pac J Cancer Prev, 13, 6139-44. https://doi.org/10.7314/APJCP.2012.13.12.6139
  43. London CA (2012). Tyrosine kinase inhibitors in veterinary medicine. Top Companion Anim Mede, 24, 106-12.
  44. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S (2002). The protein kinase complement of the human genome. Science, 298, 1912-34. https://doi.org/10.1126/science.1075762
  45. Massova I, Kotra LP, Fridman R, Mobashery S (1998). Matrix metalloproteinases: structures, evolution, and diversification. FASEB J, 12, 1075-95.
  46. Meiyanto E, Hermawan A, Anindyajati (2012). Natural products for cancer-targeted therapy: citrus flavonoids as potent chemopreventive agents. Asian Pac J Cancer Prev, 13, 427-36. https://doi.org/10.7314/APJCP.2012.13.2.427
  47. Mukanganyama S, Widersten M, Naik YS, et al (2002). Inhibition of glutathione S-transferases by antimalarial drugs possible implications for circumventing anticancer drug resistance. Int J Cancer, 97, 700-5. https://doi.org/10.1002/ijc.10054
  48. Narashimamurthy J, Rao AR, Sastry GN (2004). Aromatase inhibitors: a new paradigm in breast cancer treatment. Curr Med Chem Anticancer Agents, 4, 523-34. https://doi.org/10.2174/1568011043352669
  49. Nguewa PA, Fuertes MA, Alonso C, Perez JM (2003). Pharmacological modulation of poly (ADP-ribose) polymerase- mediated cell death: exploitation in cancer chemotherapy. Mol Pharmacol, 64, 1007-14. https://doi.org/10.1124/mol.64.5.1007
  50. O'Regan RM, Khuri FR (2004). Farnesyl transferase inhibitors: the next targeted therapies for breast cancer? Endocrin Relat Cancer, 11, 191-205. https://doi.org/10.1677/erc.0.0110191
  51. Pan JB, Hou YH, Zhang GJ (2013). Correlation between EGFR mutations and serum tumor markers in lung adenocarcinoma patients. Asian Pac J Cancer Prev, 14, 695-700. https://doi.org/10.7314/APJCP.2013.14.2.695
  52. Papeo G, Casale E, Montagnoli A, Cirla A (2013). PARP inhibitors in cancer therapy: an update. Expert Opinion on Therapeutic Patents, 23, 503-14. https://doi.org/10.1517/13543776.2013.768615
  53. Pardee AB (2009). What goes wrong in cancer. In 'The biology and treatment of cancer', Eds. Pardee AB, Stein GS. John Wiley & Sons, Inc, New Jersey, pp 3-19.
  54. Paul MK, Mukhopadhyay AK (2004). Tyrosine kinase-role and significance in cancer. Int J Med Sci, 1, 101-15.
  55. Pearson M, Garcia-Echeverria C, Fabbro D (2006). Protein tyrosine kinases as targets for cancer and other indications. In 'Protein tyrosine kinases', Eds. Fabbro D, Mc Cormick F. Humana Press Inc, Totowa, New Jersey, pp 1-29.
  56. Pitot HC, Loeb DD (2002). Some basic and applied principles of cancer chemotherapy. In 'Fundamentals of Oncology', Eds. Pitot HC, Loeb DD. Marcel Dekker Inc, New York, pp 901-945.
  57. Popovic R, Licht JD (2012). Emerging epigenetic targets and therapies in cancer medicine. Cancer Discov, 2, 405-13. https://doi.org/10.1158/2159-8290.CD-12-0076
  58. Populo H, Lopes JM, Soares P (2012). The mTOR signalling pathway in human cancer. Int J Mol Sci, 13, 1886-918. https://doi.org/10.3390/ijms13021886
  59. Posada JG, Frankel AE (2008). Monoclonal antibody therapy of cancer. In 'The molecular basis of cancer', Eds. Mendelsohn AC, Howley A, Israel S, Gray JE, Lindsten T. Elsevier, Philadelphia, pp 671-678.
  60. Rajkumar SV, Richardson PG, Hideshima T, Anderson KC (2005). Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol, 23, 630-9.
  61. Rayburn ER, Zhang R (2008). Antisense, RNAi, and gene silencing strategies for therapy: mission possible or impossible? Drug Discov Today, 13, 513-21. https://doi.org/10.1016/j.drudis.2008.03.014
  62. Ren R (2005). Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer, 5, 172-83. https://doi.org/10.1038/nrc1567
  63. Reya T, Morrison SJ, Clarke MF, Weissman IL (2001). Stem cells, cancer, and cancer stem cells. Nature, 414, 105-11. https://doi.org/10.1038/35102167
  64. Rowinsky EK, Windle JJ, Von Hoff DD (1999). Ras protein farnesyltransferase: a strategic target for anticancer therapeutic development. J Clin Oncol, 17, 3631-52.
  65. Roy R, Yang J, Moses MA (2009). Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J Clin Oncol, 27, 5287-97. https://doi.org/10.1200/JCO.2009.23.5556
  66. Satpute PS, Hazarey V, Ahmed R, Yadav L (2013). Cancer stem cells in head and neck squamous cell carcinoma: a review. Asian Pac J Cancer Prev, 14, 5579-87. https://doi.org/10.7314/APJCP.2013.14.10.5579
  67. Sawyers C (2004). Targeted cancer therapy. Nature, 432, 294-7. https://doi.org/10.1038/nature03095
  68. Schwartz GK, Shah MA (2005). Targeting the cell cycle: a new approach to cancer therapy. J Clin Oncol, 23, 9408-21. https://doi.org/10.1200/JCO.2005.01.5594
  69. Scott AM, Allison JP, Wolchok JD (2012). Monoclonal antibodies in cancer therapy. Cancer Immun, 14, 1-8.
  70. Sell S (2006). Potential gene therapy strategies for cancer stem cells. Curr Gene Ther, 6, 579-91. https://doi.org/10.2174/156652306778520674
  71. Sell S (2009). Stem cells and cancer: An introduction. In 'Stem cells and cancer', Ed. Majumder S. Springer, USA, pp 1-31.
  72. Sharova NP (2005). How does a cell repair damaged DNA? Biochemistry, 70, 275-91.
  73. Shchemelinin L, Sefc EN (2006). Protein kinases, their function and implication in cancer and other diseases. Folia Biologica (Praha), 52, 81-100.
  74. Sotgia F, Martinez-Outschoorn UE, Pavlides S, et al (2011). Understanding the warburg effect and the prognostic value of stromal caveolin-1 as a marker of a lethal tumor microenvironment. Breast Cancer Res, 13, 213-26. https://doi.org/10.1186/bcr2892
  75. Sridhar SS, Hedley D, Siu LL (2005). Raf kinase as a target for anticancer therapeutics. Mol Cancer Ther, 4, 677-85. https://doi.org/10.1158/1535-7163.MCT-04-0297
  76. Sun QC, Liu MB, Shen HJ, et al (2013). Inhibition by imatinib of expression of O-glycan-related glycosyltransferases and tumor-associated carbohydrate antigens in the K562 human leukemia cell line. Asian Pac J Cancer Prev, 14, 2447-51. https://doi.org/10.7314/APJCP.2013.14.4.2447
  77. Suter R, Marcum JA (2007). The molecular genetics of breast cancer and targeted therapy. Biologics, 1, 241-58.
  78. Tew KD, Gate L (2001). Glutathione S-transferases as emerging therapeutic targets. Expert Opinion on Therapeutic Targets, 5, 477-89. https://doi.org/10.1517/14728222.5.4.477
  79. Tu Y, Chen C, Pan J, et al (2012). The ubiquitin proteasome pathway (UPP) in the regulation of cell cycle control and DNA damage repair and its implication in tumorigenesis. Int J Clin Exp Pathol, 5, 726-38.
  80. Turella P, Cerella C, Filomeni G, et al, (2005) Proapoptotic activity of new glutathione S-transferase inhibitors. Cancer Res, 65, 3751-61. https://doi.org/10.1158/0008-5472.CAN-04-3903
  81. Vendetti FP, Rudin CM (2013). Epigenetic therapy in non-small-cell lung cancer: targeting DNA methyltransferases and histone deacetylases. Expert Opinion on Biological Therapy, 13, 1273-85. https://doi.org/10.1517/14712598.2013.819337
  82. Voorhees PM, Dees EC, O'Neil B, Orlowski RZ (2003). The proteasome as a target for cancer therapy. Clin Cancer Res, 9, 6316-25.
  83. Voorhees PM, Orlowski RZ (2006). The proteasome and proteasome inhibitors in cancer therapy. Ann Rev Pharmacol Toxicol, 46, 189-213. https://doi.org/10.1146/annurev.pharmtox.46.120604.141300
  84. Wright D (2007). Cytotoxic chemotherapy. In 'The biology of cancer', Ed. Gabriel JN. John Wiley& Sons Ltd, England, pp 45-53.
  85. Wu HC, Huang CT, Chang DK (2008). Anti-angiogenic therapeutic drugs for treatment of human cancer. J Cancer Molecules, 4, 37-45.
  86. Yuan R, Kay A, Berg WJ, Lebwohl D (2009). Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol, 2, 1-12. https://doi.org/10.1186/1756-8722-2-1
  87. Zheng L, Li J, Luo Y (2012). Glucose metabolism and cancer. In 'Biochemistry', Ed. Ekinci D. InTech, Croatia, pp 289-304.

Cited by

  1. FoxM1 as a Novel Therapeutic Target for Cancer Drug Therapy vol.16, pp.1, 2015, https://doi.org/10.7314/APJCP.2015.16.1.23