Novel and emerging targeted-based cancer therapy agents and methods
详细信息    查看全文
  • 作者:Mohammad Hojjat-Farsangi (1) (2)

    1. Department of Oncology-Pathology
    ; Immune and Gene therapy Lab ; Cancer Center Karolinska (CCK) ; Karolinska University Hospital Solna and Karolinska Institute ; SE-171 76 ; Stockholm ; Sweden
    2. Department of Immunology
    ; School of Medicine ; Bushehr University of Medical Sciences ; Bushehr ; Iran
  • 关键词:Targeted therapy ; Monoclonal antibody ; Small ; molecule inhibitor ; Tyrosine kinase ; Vaccine
  • 刊名:Tumor Biology
  • 出版年:2015
  • 出版时间:February 2015
  • 年:2015
  • 卷:36
  • 期:2
  • 页码:543-556
  • 全文大小:331 KB
  • 参考文献:1. Zhou, L, Xu, N, Sun, Y, Liu, XM (2014) Targeted biopharmaceuticals for cancer treatment. Cancer Lett 352: pp. 145-51
    2. MacDonald, V (2009) Chemotherapy: managing side effects and safe handling. Can Vet J 50: pp. 665-8
    3. Peng, X, Zhang, MQ, Conserva, F, Hosny, G, Selivanova, G, Bykov, VJ (2013) APR-246/PRIMA-1MET inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase. Cell Death Dis 4: pp. e881
    4. Yap, TA, Popat, S (2014) Toward precision medicine with next-generation EGFR inhibitors in non-small-cell lung cancer. Pharmgenomics Pers Med 7: pp. 285-95
    5. Hojjat-Farsangi, M (2014) Small-molecule inhibitors of the receptor tyrosine kinases: promising tools for targeted cancer therapies. Int J Mol Sci 15: pp. 13768-801
    6. Serviss, JT, Johnsson, P, Grander, D (2014) An emerging role for long non-coding RNAs in cancer metastasis. Front Genet 5: pp. 234
    7. Grander, D, Panaretakis, T (2010) Autophagy: cancer therapy鈥檚 friend or foe?. Futur Med Chem 2: pp. 285-97
    8. Grander, D (2000) How does interferon-alpha exert its antitumour activity in multiple myeloma?. Acta Oncol 39: pp. 801-5
    9. Hojjat-Farsangi, M, Moshfegh, A, Daneshmanesh, AH, Khan, AS, Mikaelsson, E, Osterborg, A (2014) The receptor tyrosine kinase ROR1鈥攁n oncofetal antigen for targeted cancer therapy. Semin Cancer Biol 29C: pp. 21-31
    10. Li, GN, Wang, SP, Xue, X, Qu, XJ, Liu, HP (2013) Monoclonal antibody-related drugs for cancer therapy. Drug Discov Ther 7: pp. 178-84
    11. Fauvel, B, Yasri, A (2014) Antibodies directed against receptor tyrosine kinases: current and future strategies to fight cancer. MAbs 6: pp. 838-51
    12. Ghoreschi, K, Gadina, M (2014) Jackpot! New small molecules in autoimmune and inflammatory diseases. Exp Dermatol 23: pp. 7-11
    13. Wu, X, Liu, X, Koul, S, Lee, CY, Zhang, Z, Halmos, B (2014) AXL kinase as a novel target for cancer therapy. Oncotarget 5: pp. 9546-63
    14. Zhang, H, Nimmer, PM, Tahir, SK, Chen, J, Fryer, RM, Hahn, KR (2007) Bcl-2 family proteins are essential for platelet survival. Cell Death Differ 14: pp. 943-51
    15. Wilson, WH, O鈥機onnor, OA, Czuczman, MS, LaCasce, AS, Gerecitano, JF, Leonard, JP (2010) Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol 11: pp. 1149-59
    16. Rudin, CM, Hann, CL, Garon, EB (2012) Ribeiro de Oliveira M, Bonomi PD, Camidge DR, et al. Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in patients with relapsed small cell lung cancer. Clin Cancer Res 18: pp. 3163-9
    17. Gobessi, S, Laurenti, L, Longo, PG, Carsetti, L, Berno, V, Sica, S (2009) Inhibition of constitutive and BCR-induced Syk activation downregulates Mcl-1 and induces apoptosis in chronic lymphocytic leukemia B cells. Leukemia 23: pp. 686-97
    18. Paterson, A, Mockridge, CI, Adams, JE, Krysov, S, Potter, KN, Duncombe, AS (2012) Mechanisms and clinical significance of BIM phosphorylation in chronic lymphocytic leukemia. Blood 119: pp. 1726-36
    19. Smith DD, Goldstein L, Cheng M, James DF, Kunkel LA, Fardis M, et al. Modeling absolute lymphocyte counts after treatment of chronic lymphocytic leukemia with ibrutinib. Ann Hematol. 2015;94(2):249鈥?6
    20. Tennant, DA, Duran, RV, Gottlieb, E (2010) Targeting metabolic transformation for cancer therapy. Nat Rev Cancer 10: pp. 267-77
    21. Thomas, HD, Calabrese, CR, Batey, MA, Canan, S, Hostomsky, Z, Kyle, S (2007) Preclinical selection of a novel poly(ADP-ribose) polymerase inhibitor for clinical trial. Mol Cancer Ther 6: pp. 945-56
    22. Plummer, R, Jones, C, Middleton, M, Wilson, R, Evans, J, Olsen, A (2008) Phase I study of the poly(ADP-ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors. Clin Cancer Res 14: pp. 7917-23
    23. Plummer, R, Lorigan, P, Steven, N, Scott, L, Middleton, MR, Wilson, RH (2013) A phase II study of the potent PARP inhibitor, Rucaparib (PF-01367338, AG014699), with temozolomide in patients with metastatic melanoma demonstrating evidence of chemopotentiation. Cancer Chemother Pharmacol 71: pp. 1191-9
    24. Fong, PC, Boss, DS, Yap, TA, Tutt, A, Wu, P, Mergui-Roelvink, M (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361: pp. 123-34
    25. Richardson, PG, Mitsiades, CS, Laubach, JP, Hajek, R, Spicka, I, Dimopoulos, MA (2013) Preclinical data and early clinical experience supporting the use of histone deacetylase inhibitors in multiple myeloma. Leuk Res 37: pp. 829-37
    26. Marquard, L, Poulsen, CB, Gjerdrum, LM, Nully, BP, Christensen, IJ, Jensen, PB (2009) Histone deacetylase 1, 2, 6 and acetylated histone H4 in B- and T-cell lymphomas. Histopathology 54: pp. 688-98
    27. Jazirehi, AR (2010) Regulation of apoptosis-associated genes by histone deacetylase inhibitors: implications in cancer therapy. Anticancer Drugs 21: pp. 805-13
    28. West, AC, Johnstone, RW (2014) New and emerging HDAC inhibitors for cancer treatment. J Clin Invest 124: pp. 30-9
    29. Thaler, F, Mercurio, C (2014) Towards selective inhibition of histone deacetylase isoforms: what has been achieved, where we are and what will be next. Chem Med Chem 9: pp. 523-6
    30. McGraw, AL (2013) Romidepsin for the treatment of T-cell lymphomas. Am J Health Syst Pharm 70: pp. 1115-22
    31. Dong, M, Ning, ZQ, Xing, PY, Xu, JL, Cao, HX, Dou, GF (2012) Phase I study of chidamide (CS055/HBI-8000), a new histone deacetylase inhibitor, in patients with advanced solid tumors and lymphomas. Cancer Chemother Pharmacol 69: pp. 1413-22
    32. Banerji, U, Doorn, L, Papadatos-Pastos, D, Kristeleit, R, Debnam, P, Tall, M (2012) A phase I pharmacokinetic and pharmacodynamic study of CHR-3996, an oral class I selective histone deacetylase inhibitor in refractory solid tumors. Clin Cancer Res 18: pp. 2687-94
    33. Dobashi, Y, Watanabe, Y, Miwa, C, Suzuki, S, Koyama, S (2011) Mammalian target of rapamycin: a central node of complex signaling cascades. Int J Clin Exp Pathol 4: pp. 476-95
    34. Dancey, JE, Monzon, J (2011) Ridaforolimus: a promising drug in the treatment of soft-tissue sarcoma and other malignancies. Future Oncol 7: pp. 827-39
    35. Chen, X, Zhao, M, Hao, M, Sun, X, Wang, J, Mao, Y (2013) Dual inhibition of PI3K and mTOR mitigates compensatory AKT activation and improves tamoxifen response in breast cancer. Mol Cancer Res 11: pp. 1269-78
    36. Laplante, M, Sabatini, DM (2012) mTOR signaling in growth control and disease. Cell 149: pp. 274-93
    37. Lebwohl, D, Anak, O, Sahmoud, T, Klimovsky, J, Elmroth, I, Haas, T (2013) Development of everolimus, a novel oral mTOR inhibitor, across a spectrum of diseases. Ann N Y Acad Sci 1291: pp. 14-32
    38. Molina, AM, Motzer, RJ, Heng, DY (2013) Systemic treatment options for untreated patients with metastatic clear cell renal cancer. Semin Oncol 40: pp. 436-43
    39. Wan, X, Shen, N, Mendoza, A, Khanna, C, Helman, LJ (2006) CCI-779 inhibits rhabdomyosarcoma xenograft growth by an antiangiogenic mechanism linked to the targeting of mTOR/Hif-1alpha/VEGF signaling. Neoplasia 8: pp. 394-401
    40. Mayer, I (2013) Role of mTOR inhibition in preventing resistance and restoring sensitivity to hormone-targeted and HER2-targeted therapies in breast cancer. Clin Adv Hematol Oncol 11: pp. 217-24
    41. Kabir, A, Suresh, KG (2014) Targeting double-stranded RNA with spermine, 1-naphthylacetyl spermine and spermidine: a comparative biophysical investigation. J Phys Chem B 118: pp. 11050-64
    42. Pan, Q, Shai, O, Lee, LJ, Frey, BJ, Blencowe, BJ (2008) Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet 40: pp. 1413-5
    43. Pawellek A, McElroy S, Samatov T, Mitchell L, Woodland A, Ryder U, et al. Identification of small molecule inhibitors of pre-mRNA splicing. J Biol Chem. 2014;289(50):34683鈥?8.
    44. Thomas, T, Balabhadrapathruni, S, Gallo, MA, Thomas, TJ (2002) Development of polyamine analogs as cancer therapeutic agents. Oncol Res 13: pp. 123-35
    45. Casero, RA, Marton, LJ (2007) Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 6: pp. 373-90
    46. Fan, L, Lagisetti, C, Edwards, CC, Webb, TR, Potter, PM (2011) Sudemycins, novel small molecule analogues of FR901464, induce alternative gene splicing. ACS Chem Biol 6: pp. 582-9
    47. Kotake, Y, Sagane, K, Owa, T, Mimori-Kiyosue, Y, Shimizu, H, Uesugi, M (2007) Splicing factor SF3b as a target of the antitumor natural product pladienolide. Nat Chem Biol 3: pp. 570-5
    48. Hong, DS, Kurzrock, R, Naing, A, Wheler, JJ, Falchook, GS, Schiffman, JS (2014) A phase I, open-label, single-arm, dose-escalation study of E7107, a precursor messenger ribonucleic acid (pre-mRNA) splicesome inhibitor administered intravenously on days 1 and 8 every 21聽days to patients with solid tumors. Invest New Drugs 32: pp. 436-44
    49. Eskens, FA, Ramos, FJ, Burger, H, O鈥橞rien, JP, Piera, A, Jonge, MJ (2013) Phase I pharmacokinetic and pharmacodynamic study of the first-in-class spliceosome inhibitor E7107 in patients with advanced solid tumors. Clin Cancer Res 19: pp. 6296-304
    50. Jayaraj GG, Nahar S, Maiti S. Nonconventional chemical inhibitors of microRNA: therapeutic scope. Chem Commun (Camb). 2015;51(5):820鈥?1.
    51. Zhou, M, Chen, J, Zhou, L, Chen, W, Ding, G, Cao, L (2014) Pancreatic cancer derived exosomes regulate the expression of TLR4 in dendritic cells via miR-203. Cell Immunol 292: pp. 65-9
    52. Caramuta, S, Lee, L, Ozata, DM, Akcakaya, P, Georgii-Hemming, P, Xie, H (2013) Role of microRNAs and microRNA machinery in the pathogenesis of diffuse large B-cell lymphoma. Blood Cancer J 3: pp. e152
    53. Liu, R, Liu, X, Zheng, Y, Gu, J, Xiong, S, Jiang, P (2014) MicroRNA-7 sensitizes non-small cell lung cancer cells to paclitaxel. Oncol Lett 8: pp. 2193-200
    54. Shen G, Rong X, Zhao J, Yang X, Li H, Jiang H, et al. MicroRNA-105 suppresses cell proliferation and inhibits PI3K/AKT signaling in human hepatocellular carcinoma. Carcinogenesis. 2014;35(12):2748鈥?5.
    55. Liu C, Yang H, Xu Z, Li D, Zhou M, Xiao K, et al. microRNA-548 l is involved in the migration and invasion of non-small cell lung cancer by targeting the AKT1 signaling pathway. J Cancer Res Clin Oncol. 2014. doi:10.1007/s00432-014-1836-7 .
    56. Moskwa P, Zinn PO, Choi YE, Shukla SA, Fendler W, Chen CC, et al. A functional screen identifies miRs that induce radioresistance in glioblastomas. Mol Cancer Res. 2014.
    57. Hojjat-Farsangi, M, Ghaemimanesh, F, Daneshmanesh, AH, Bayat, AA, Mahmoudian, J, Jeddi-Tehrani, M (2013) Inhibition of the receptor tyrosine kinase ROR1 by anti-ROR1 monoclonal antibodies and siRNA induced apoptosis of melanoma cells. PLoS One 8: pp. e61167
    58. Daneshmanesh, AH, Hojjat-Farsangi, M, Khan, AS, Jeddi-Tehrani, M, Akhondi, MM, Bayat, AA (2012) Monoclonal antibodies against ROR1 induce apoptosis of chronic lymphocytic leukemia (CLL) cells. Leukemia 26: pp. 1348-55
    59. Staff, C, Magnusson, CG, Hojjat-Farsangi, M, Mosolits, S, Liljefors, M, Frodin, JE (2012) Induction of IgM, IgA and IgE antibodies in colorectal cancer patients vaccinated with a recombinant CEA protein. J Clin Immunol 32: pp. 855-65
    60. Riley, JK, Sliwkowski, MX (2000) CD20: a gene in search of a function. Semin Oncol 27: pp. 17-24
    61. Fang, D, Nguyen, TK, Leishear, K, Finko, R, Kulp, AN, Hotz, S (2005) A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 65: pp. 9328-37
    62. Bologna, L, Gotti, E, Roit, F, Intermesoli, T, Rambaldi, A, Introna, M (2013) Ofatumumab is more efficient than rituximab in lysing B chronic lymphocytic leukemia cells in whole blood and in combination with chemotherapy. J Immunol 190: pp. 231-9
    63. Fabbri A, Cencini E, Rigacci L, Bartalucci G, Puccini B, Dottori R, et al. Efficacy and safety of rituximab plus low-dose oral fludarabine and cyclophosphamide as first-line treatment of Elderly patients with indolentnon Hodgkin lymphomas. Leuk Lymphoma. 2014;55(4):781鈥?.
    64. Robak, T (2012) Rituximab for chronic lymphocytic leukemia. Expert Opin Biol Ther 12: pp. 503-15
    65. Bauer, K, Rancea, M, Roloff, V, Elter, T, Hallek, M, Engert, A (2012) Rituximab, ofatumumab and other monoclonal anti-CD20 antibodies for chronic lymphocytic leukaemia. Cochrane Database Syst Rev 11:
    66. Goteri, G, Olivieri, A, Ranaldi, R, Lucesole, M, Filosa, A, Capretti, R (2006) Bone marrow histopathological and molecular changes of small B-cell lymphomas after rituximab therapy: comparison with clinical response and patients outcome. Int J Immunopathol Pharmacol 19: pp. 421-31
    67. Wierda, WG, Kipps, TJ, Mayer, J, Stilgenbauer, S, Williams, CD, Hellmann, A (2010) Ofatumumab as single-agent CD20 immunotherapy in fludarabine-refractory chronic lymphocytic leukemia. J Clin Oncol 28: pp. 1749-55
    68. Smolej, L, Doubek, M, Panovska, A, Simkovic, M, Brychtova, Y, Belada, D (2012) Rituximab in combination with high-dose dexamethasone for the treatment of relapsed/refractory chronic lymphocytic leukemia. Leuk Res 36: pp. 1278-82
    69. Abou-Nassar, K, Brown, JR (2010) Novel agents for the treatment of chronic lymphocytic leukemia. Clin Adv Hematol Oncol 8: pp. 886-95
    70. Goede, V, Hallek, M (2011) Optimal pharmacotherapeutic management of chronic lymphocytic leukaemia: considerations in the elderly. Drugs Aging 28: pp. 163-76
    71. McCoyd, M (2013) Update on therapeutic options for multiple sclerosis. Neurol Clin 31: pp. 827-45
    72. Tsimberidou, AM, Keating, MJ (2009) Treatment of fludarabine-refractory chronic lymphocytic leukemia. Cancer 115: pp. 2824-36
    73. Osterborg, A, Foa, R, Bezares, RF, Dearden, C, Dyer, MJ, Geisler, C (2009) Management guidelines for the use of alemtuzumab in chronic lymphocytic leukemia. Leukemia 23: pp. 1980-8
    74. Hale, G (2001) The CD52 antigen and development of the CAMPATH antibodies. Cytotherapy 3: pp. 137-43
    75. Sorokin, P (2001) Campath-1H. Clin J Oncol Nurs 5: pp. 65-6
    76. Ravandi, F, O鈥橞rien, S (2005) Alemtuzumab. Expert Rev Anticancer Ther 5: pp. 39-51
    77. Karlsson, C, Lundin, J, Kimby, E, Kennedy, B, Moreton, P, Hillmen, P (2009) Phase II study of subcutaneous alemtuzumab without dose escalation in patients with advanced-stage, relapsed chronic lymphocytic leukaemia. Br J Haematol 144: pp. 78-85
    78. Myrvang, H (2011) Transplantation: alemtuzumab induction is safe for renal transplant recipients. Nat Rev Nephrol 7: pp. 362
    79. Hanaway, MJ, Woodle, ES, Mulgaonkar, S, Peddi, VR, Kaufman, DB, First, MR (2011) Alemtuzumab induction in renal transplantation. N Engl J Med 364: pp. 1909-19
    80. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer Therapy. J Clin Oncol. 2015. doi:10.1200/JCO.2014.59.4358 .
    81. Blank, CU (2014) The perspective of immunotherapy: new molecules and new mechanisms of action in immune modulation. Curr Opin Oncol 26: pp. 204-14
    82. Finkelstein, SE, Salenius, S, Mantz, CA, Shore, ND, Fernandez, EB, Shulman, J (2015) Combining immunotherapy and radiation for prostate cancer. Clin Genitourin Cancer 13: pp. 1-9
    83. Vasekar, M, Liu, X, Zheng, H, Belani, CP (2014) Targeted immunotherapy for non-small cell lung cancer. World J Clin Oncol 5: pp. 39-47
    84. Langer, LF, Clay, TM, Morse, MA (2007) Update on anti-CTLA-4 antibodies in clinical trials. Expert Opin Biol Ther 7: pp. 1245-56
    85. Kothari, AN, Mi, Z, Zapf, M, Kuo, PC (2014) Novel clinical therapeutics targeting the epithelial to mesenchymal transition. Clin Transl Med 3: pp. 35
    86. Becker, KF, Rosivatz, E, Blechschmidt, K, Kremmer, E, Sarbia, M, Hofler, H (2007) Analysis of the E-cadherin repressor Snail in primary human cancers. Cells Tissues Organs 185: pp. 204-12
    87. Sanchez-Tillo, E, Liu, Y, Barrios, O, Siles, L, Fanlo, L, Cuatrecasas, M (2012) EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 69: pp. 3429-56
    88. Arteaga, CL (2006) Inhibition of TGFbeta signaling in cancer therapy. Curr Opin Genet Dev 16: pp. 30-7
    89. Fabregat, I, Fernando, J, Mainez, J, Sancho, P (2014) TGF-beta signaling in cancer treatment. Curr Pharm Des 20: pp. 2934-47
    90. Yang, Y, Shaffer, AL, Emre, NC, Ceribelli, M, Zhang, M, Wright, G (2012) Exploiting synthetic lethality for the therapy of ABC diffuse large B cell lymphoma. Cancer Cell 21: pp. 723-37
    91. Semeraro, M, Vacchelli, E, Eggermont, A, Galon, J, Zitvogel, L, Kroemer, G (2013) Trial Watch: lenalidomide-based immunochemotherapy. Oncoimmunology 2: pp. e26494
    92. LeBlanc, R, Hideshima, T, Catley, LP, Shringarpure, R, Burger, R, Mitsiades, N (2004) Immunomodulatory drug costimulates T cells via the B7-CD28 pathway. Blood 103: pp. 1787-90
    93. Crane, E, List, A (2005) Immunomodulatory drugs. Cancer Invest 23: pp. 625-34
    94. Song, W, Vliet, HJ, Tai, YT, Prabhala, R, Wang, R, Podar, K (2008) Generation of antitumor invariant natural killer T cell lines in multiple myeloma and promotion of their functions via lenalidomide: a strategy for immunotherapy. Clin Cancer Res 14: pp. 6955-62
    95. Harada, T, Ozaki, S, Oda, A, Fujii, S, Nakamura, S, Miki, H (2013) Association of Th1 and Th2 cytokines with transient inflammatory reaction during lenalidomide plus dexamethasone therapy in multiple myeloma. Int J Hematol 97: pp. 743-8
    96. Gonzalez-Rodriguez, AP, Payer, AR, Acebes-Huerta, A, Huergo-Zapico, L, Villa-Alvarez, M, Gonzalez-Garcia, E (2013) Lenalidomide and chronic lymphocytic leukemia. Biomed Res Int 2013: pp. 932010
    97. Ferrajoli, A, Lee, BN, Schlette, EJ, O鈥橞rien, SM, Gao, H, Wen, S (2008) Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia. Blood 111: pp. 5291-7
    98. Kourelis TV, Kumar SK, Srivastava G, Gertz MA, Lacy MQ, Buadi FK, et al. Long term response to lenalidomide in patients with newly diagnosed multiple myeloma. Leukemia 2014;28(2):455鈥?.
    99. Song K, Herzog BH, Sheng M, Fu J, McDaniel JM, Ruan J, et al. Lenalidomide inhibits lymphangiogenesis in preclinical models of mantle cell lymphoma. Cancer Res. 2013;73(24):7254鈥?4.
    100. Provencio M, Sanchez A, Sanchez-Beato M. New drugs and targeted treatments in Hodgkin鈥檚 lymphoma. Cancer Treat Rev. 2014;40(3):457鈥?4.
    101. Alvarez-Fernandez, S, Ortiz-Ruiz, MJ, Parrott, T, Zaknoen, S, Ocio, EM, San Miguel, J (2013) Potent antimyeloma activity of a novel ERK5/CDK inhibitor. Clin Cancer Res 19: pp. 2677-87
    102. Minnema, MC, Veer, MS, Aarts, T, Emmelot, M, Mutis, T, Lokhorst, HM (2009) Lenalidomide alone or in combination with dexamethasone is highly effective in patients with relapsed multiple myeloma following allogeneic stem cell transplantation and increases the frequency of CD4鈥?鈥塅oxp3+ T cells. Leukemia 23: pp. 605-7
    103. Pan, Z, Scheerens, H, Li, SJ, Schultz, BE, Sprengeler, PA, Burrill, LC (2007) Discovery of selective irreversible inhibitors for Bruton鈥檚 tyrosine kinase. Chem Med Chem 2: pp. 58-61
    104. Honigberg, LA, Smith, AM, Sirisawad, M, Verner, E, Loury, D, Chang, B (2010) The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A 107: pp. 13075-80
    105. Xing, L, Huang, A (2014) Bruton鈥檚 TK inhibitors: structural insights and evolution of clinical candidates. Futur Med Chem 6: pp. 675-95
    106. Zheng, X, Ding, N, Song, Y, Feng, L, Zhu, J (2014) Different sensitivity of germinal center B cell-like diffuse large B cell lymphoma cells towards ibrutinib treatment. Cancer Cell Int 14: pp. 32
    107. Dubovsky, JA, Beckwith, KA, Natarajan, G, Woyach, JA, Jaglowski, S, Zhong, Y (2013) Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood 122: pp. 2539-49
    108. Ohtsubo, K, Marth, JD (2006) Glycosylation in cellular mechanisms of health and disease. Cell 126: pp. 855-67
    109. Ruhaak, LR, Miyamoto, S, Lebrilla, CB (2013) Developments in the identification of glycan biomarkers for the detection of cancer. Mol Cell Proteomics 12: pp. 846-55
    110. Takahashi, T, Johnson, TD, Nishinaka, Y, Morton, DL, Irie, RF (1999) IgM anti-ganglioside antibodies induced by melanoma cell vaccine correlate with survival of melanoma patients. J Invest Dermatol 112: pp. 205-9
    111. Irie, RF, Ollila, DW, O鈥橠ay, S, Morton, DL (2004) Phase I pilot clinical trial of human IgM monoclonal antibody to ganglioside GM3 in patients with metastatic melanoma. Cancer Immunol Immunother 53: pp. 110-7
    112. Zhang, S, Zhang, HS, Cordon-Cardo, C, Reuter, VE, Singhal, AK, Lloyd, KO (1997) Selection of tumor antigens as targets for immune attack using immunohistochemistry: II. Blood group-related antigens. Int J Cancer 73: pp. 50-6
    113. Ashford, TP, Porter, KR (1962) Cytoplasmic components in hepatic cell lysosomes. J Cell Biol 12: pp. 198-202
    114. Lee, J, Giordano, S, Zhang, J (2012) Autophagy, mitochondria and oxidative stress: cross-talk and redox signalling. Biochem J 441: pp. 523-40
    115. Zhang, J, Yang, Z, Xie, L, Xu, L, Xu, D, Liu, X (2013) Statins, autophagy and cancer metastasis. Int J Biochem Cell Biol 45: pp. 745-52
    116. Fullgrabe, J, Heldring, N, Hermanson, O, Joseph, B (2014) Cracking the survival code: autophagy-related histone modifications. Autophagy 10: pp. 556-61
    117. Viry E, Paggetti J, Baginska J, Mgrditchian T, Berchem G, Moussay E, et al. Autophagy: an adaptive metabolic response to stress shaping the antitumor immunity. Biochem Pharmacol. 2014;92(1):31鈥?2.
    118. Guan, JL, Simon, AK, Prescott, M, Menendez, JA, Liu, F, Wang, F (2013) Autophagy in stem cells. Autophagy 9: pp. 830-49
    119. Burroughs, SK, Kaluz, S, Wang, D, Wang, K, Meir, EG, Wang, B (2013) Hypoxia inducible factor pathway inhibitors as anticancer therapeutics. Futur Med Chem 5: pp. 553-72
    120. Ward, C, Langdon, SP, Mullen, P, Harris, AL, Harrison, DJ, Supuran, CT (2013) New strategies for targeting the hypoxic tumour microenvironment in breast cancer. Cancer Treat Rev 39: pp. 171-9
    121. Moeller, BJ, Dreher, MR, Rabbani, ZN, Schroeder, T, Cao, Y, Li, CY (2005) Pleiotropic effects of HIF-1 blockade on tumor radiosensitivity. Cancer Cell 8: pp. 99-110
    122. Yeo, EJ, Chun, YS, Park, JW (2004) New anticancer strategies targeting HIF-1. Biochem Pharmacol 68: pp. 1061-9
    123. Poon, E, Harris, AL, Ashcroft, M (2009) Targeting the hypoxia-inducible factor (HIF) pathway in cancer. Expert Rev Mol Med 11: pp. e26
    124. Chen, J, Zhao, S, Nakada, K, Kuge, Y, Tamaki, N, Okada, F (2003) Dominant-negative hypoxia-inducible factor-1 alpha reduces tumorigenicity of pancreatic cancer cells through the suppression of glucose metabolism. Am J Pathol 162: pp. 1283-91
    125. Zinzalla, G, Thurston, DE (2009) Targeting protein-protein interactions for therapeutic intervention: a challenge for the future. Futur Med Chem 1: pp. 65-93
    126. Kung, AL, Zabludoff, SD, France, DS, Freedman, SJ, Tanner, EA, Vieira, A (2004) Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 6: pp. 33-43
    127. Greenberger, LM, Horak, ID, Filpula, D, Sapra, P, Westergaard, M, Frydenlund, HF (2008) A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor cell growth. Mol Cancer Ther 7: pp. 3598-608
    128. Jones, DT, Harris, AL (2012) Small-molecule inhibitors of the HIF pathway and synthetic lethal interactions. Expert Opin Ther Targets 16: pp. 463-80
    129. Connors, TA, Whisson, ME (1966) Cure of mice bearing advanced plasma cell tumours with aniline mustard: the relationship between glucuronidase activity and tumour sensitivity. Nature 210: pp. 866-7
    130. Whisson, ME, Connors, TA (1965) Cure of mice bearing advanced plasma cell tumours with aniline mustard. Nature 206: pp. 689-91
    131. Bryant, JL, Meredith, SL, Williams, KJ, White, A (2014) Targeting hypoxia in the treatment of small cell lung cancer. Lung Cancer 86: pp. 126-32
    132. Ghattass, K, Assah, R, El-Sabban, M, Gali-Muhtasib, H (2013) Targeting hypoxia for sensitization of tumors to radio- and chemotherapy. Curr Cancer Drug Targets 13: pp. 670-85
    133. Wardman, P, Rothkamm, K, Folkes, LK, Woodcock, M, Johnston, PJ (2007) Radiosensitization by nitric oxide at low radiation doses. Radiat Res 167: pp. 475-84
    134. Marchwicka, A, Cebrat, M, Sampath, P, Sniezewski, L, Marcinkowska, E (2014) Perspectives of differentiation therapies of acute myeloid leukemia: the search for the molecular basis of patients鈥?variable responses to 1,25-dihydroxyvitamin d and vitamin d analogs. Front Oncol 4: pp. 125
    135. Strickland, S, Mahdavi, V (1978) The induction of differentiation in teratocarcinoma stem cells by retinoic acid. Cell 15: pp. 393-403
    136. Werneck, MB (2012) Endogenous anticancer mechanism: differentiation. Front Biosci (Schol Ed) 4: pp. 1518-38
    137. Chen, H, Dong, JM, Liu, Y, Chiu, JF (1999) Identification of a cis-acting element in the rat alpha-fetoprotein gene and its specific binding proteins in F9 cells during retinoic acid-induced differentiation. J Cell Biochem 72: pp. 25-34
    138. Bunaciu, RP, Yen, A (2011) Activation of the aryl hydrocarbon receptor AhR promotes retinoic acid-induced differentiation of myeloblastic leukemia cells by restricting expression of the stem cell transcription factor Oct4. Cancer Res 71: pp. 2371-80
    139. Tang, XH, Gudas, LJ (2011) Retinoids, retinoic acid receptors, and cancer. Annu Rev Pathol 6: pp. 345-64
    140. Fuchs, E, Green, H (1981) Regulation of terminal differentiation of cultured human keratinocytes by vitamin A. Cell 25: pp. 617-25
    141. Matthay, KK, Reynolds, CP, Seeger, RC, Shimada, H, Adkins, ES, Haas-Kogan, D (2009) Long-term results for children with high-risk neuroblastoma treated on a randomized trial of myeloablative therapy followed by 13-cis-retinoic acid: a children鈥檚 oncology group study. J Clin Oncol 27: pp. 1007-13
    142. Warrell, RP, Frankel, SR, Miller, WH, Scheinberg, DA, Itri, LM, Hittelman, WN (1991) Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid). N Engl J Med 324: pp. 1385-93
    143. Duong, V, Rochette-Egly, C (2011) The molecular physiology of nuclear retinoic acid receptors. From health to disease. Biochim Biophys Acta 1812: pp. 1023-31
    144. Touma, SE, Goldberg, JS, Moench, P, Guo, X, Tickoo, SK, Gudas, LJ (2005) Retinoic acid and the histone deacetylase inhibitor trichostatin a inhibit the proliferation of human renal cell carcinoma in a xenograft tumor model. Clin Cancer Res 11: pp. 3558-66
    145. Farias, EF, Petrie, K, Leibovitch, B, Murtagh, J, Chornet, MB, Schenk, T (2010) Interference with Sin3 function induces epigenetic reprogramming and differentiation in breast cancer cells. Proc Natl Acad Sci U S A 107: pp. 11811-6
    146. Tavares, TS, Nanus, D, Yang, XJ, Gudas, LJ (2008) Gene microarray analysis of human renal cell carcinoma: the effects of HDAC inhibition and retinoid treatment. Cancer Biol Ther 7: pp. 1607-18
    147. Garcia-Manero, G, Kantarjian, HM, Sanchez-Gonzalez, B, Yang, H, Rosner, G, Verstovsek, S (2006) Phase 1/2 study of the combination of 5-aza-2鈥?deoxycytidine with valproic acid in patients with leukemia. Blood 108: pp. 3271-9
  • 刊物主题:Cancer Research;
  • 出版者:Springer Netherlands
  • ISSN:1423-0380
文摘
After several decades of uncovering the cancer features and following the improvement of therapeutic agents, however cancer remains as one of the major reasons of mortality. Chemotherapy is one of the main treatment options and has significantly improved the overall survival of cancer patients, but chemotherapeutic agents are highly toxic for normal cells. Therefore, there is a great unmet medical need to develop new therapeutic principles and agents. Targeted-based cancer therapy (TBCT) agents and methods have revolutionized the cancer treatment efficacy. Monoclonal antibodies (mAbs) and small molecule inhibitors (SMIs) are among the most effective agents of TBCT. These drugs have improved the prognosis and survival of cancer patients; however, the therapeutic resistance has subdued the effects. Several mechanisms lead to drug resistance such as mutations in the drug targets, activation of compensatory pathways, and intrinsic or acquired resistance of cancer stem cells. Therefore, new modalities, improving current generation of inhibitors and mAbs, and optimizing the combinational therapy regimens are necessary to decrease the current obstacles in front of TBCT. Moreover, the success of new TBCT agents such as mAbs, SMIs, and immunomodulatory agents has sparked further therapeutic modalities with novel targets to inhibit. Due to the lack of cumulative information describing different agents and methods of TBCT, this review focuses on the most important agents and methods of TBCT that are currently under investigation.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700