Anti-vascular therapies in ovarian cancer: moving beyond anti-VEGF approaches
详细信息    查看全文
  • 作者:Hyun-Jin Choi (1)
    Guillermo N. Armaiz Pena (1)
    Sunila Pradeep (1)
    Min Soon Cho (4)
    Robert L. Coleman (1)
    Anil K. Sood (1) (2) (3)

    1. Department of Gynecologic Oncology and Reproductive Medicine Unit 1362
    ; The University of Texas MD Anderson Cancer Center ; 1515 Holcombe Boulevard ; Houston ; TX ; 77030 ; USA
    4. Section of Benign Hematology
    ; Department of Pulmonary Medicine ; The University of Texas MD Anderson Cancer Center ; Houston ; TX ; USA
    2. Department of Cancer Biology
    ; The University of Texas MD Anderson Cancer Center ; Houston ; TX ; USA
    3. Center for RNA Interference and Non-Coding RNA
    ; The University of Texas MD Anderson Cancer Center ; Houston ; TX ; USA
  • 关键词:Ovarian cancer ; Targeted therapy ; Angiogenesis ; Anti ; vascular agent ; Resistance to anti ; VEGF therapy
  • 刊名:Cancer and Metastasis Reviews
  • 出版年:2015
  • 出版时间:March 2015
  • 年:2015
  • 卷:34
  • 期:1
  • 页码:19-40
  • 全文大小:508 KB
  • 参考文献:1. Agarwal, R, Kaye, SB (2003) Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nature Reviews Cancer 3: pp. 502-516
    2. Balvert-Locht, HR, Coebergh, JW, Hop, WC (1991) Improved prognosis of ovarian cancer in The Netherlands during the period 1975鈥?985: a registry-based study. Gynecologic Oncology 42: pp. 3-8
    3. Ozols, RF, Bundy, BN, Greer, BE (2003) Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a gynecologic oncology group study. Journal of Clinical Oncology 21: pp. 3194-3200
    4. Bois, A, Neijt, JP, Thigpen, JT (1999) First line chemotherapy with carboplatin plus paclitaxel in advanced ovarian cancer鈥攁 new standard of care?. Annals of Oncology 10: pp. 35-41
    5. Biagi, JJ, Eisenhauer, EA (2003) Systemic treatment policies in ovarian cancer: the next 10 years. International Journal of Gynecological Cancer 13: pp. 231-240
    6. Neijt, JP, Engelholm, SA, Tuxen, MK (2000) Exploratory phase III study of paclitaxel and cisplatin versus paclitaxel and carboplatin in advanced ovarian cancer. Journal of Clinical Oncology 18: pp. 3084-3092
    7. Greenlee, RT, Hill-Harmon, MB, Murray, T, Thun, M (2001) Cancer statistics, 2001. CA: A Cancer Journal for Clinicians 51: pp. 15-36
    8. Gore, ME, Fryatt, I, Wiltshaw, E, Dawson, T (1990) Treatment of relapsed carcinoma of the ovary with cisplatin or carboplatin following initial treatment with these compounds. Gynecologic Oncology 36: pp. 207-211
    9. Wernert, N, Locherbach, C, Wellmann, A, Behrens, P, Hugel, A (2001) Presence of genetic alterations in microdissected stroma of human colon and breast cancers. Anticancer Research 21: pp. 2259-2264
    10. Allinen, M, Beroukhim, R, Cai, L (2004) Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6: pp. 17-32
    11. Fukino, K, Shen, L, Patocs, A, Mutter, GL, Eng, C (2007) Genomic instability within tumor stroma and clinicopathological characteristics of sporadic primary invasive breast carcinoma. JAMA 297: pp. 2103-2111
    12. Folkman, J (1990) What is the evidence that tumors are angiogenesis dependent?. Journal of the National Cancer Institute 82: pp. 4-6
    13. Eskander, RN, Randall, LM (2011) Bevacizumab in the treatment of ovarian cancer. Biologics 5: pp. 1-5
    14. Ferrara, N, Kerbel, RS (2005) Angiogenesis as a therapeutic target. Nature 438: pp. 967-974
    15. Konerding, MA, Fait, E, Gaumann, A (2001) 3D microvascular architecture of pre-cancerous lesions and invasive carcinomas of the colon. British Journal of Cancer 84: pp. 1354-1362
    16. Denekamp, J (1982) Endothelial cell proliferation as a novel approach to targeting tumour therapy. British Journal of Cancer 45: pp. 136-139
    17. Hinnen, P, Eskens, FA (2007) Vascular disrupting agents in clinical development. British Journal of Cancer 96: pp. 1159-1165
    18. Holwell, SE, Cooper, PA, Thompson, MJ (2002) Anti-tumor and anti-vascular effects of the novel tubulin-binding agent combretastatin A-1 phosphate. Anticancer Research 22: pp. 3933-3940
    19. Tozer, GM, Prise, VE, Wilson, J (1999) Combretastatin A-4 phosphate as a tumor vascular-targeting agent: early effects in tumors and normal tissues. Cancer Research 59: pp. 1626-1634
    20. Marysael, T, Ni, Y, Lerut, E, Witte, P (2011) Influence of the vascular damaging agents DMXAA and ZD6126 on hypericin distribution and accumulation in RIF-1 tumors. Journal of Cancer Research and Clinical Oncology 137: pp. 1619-1627
    21. Nathan, P, Zweifel, M, Padhani, AR (2012) Phase I trial of combretastatin A4 phosphate (CA4P) in combination with bevacizumab in patients with advanced cancer. Clinical Cancer Research 18: pp. 3428-3439
    22. Zweifel, M, Jayson, GC, Reed, NS (2011) Phase II trial of combretastatin A4 phosphate, carboplatin, and paclitaxel in patients with platinum-resistant ovarian cancer. Annals of Oncology 22: pp. 2036-2041
    23. Ching, LM, Cao, Z, Kieda, C, Zwain, S, Jameson, MB, Baguley, BC (2002) Induction of endothelial cell apoptosis by the antivascular agent 5,6-Dimethylxanthenone-4-acetic acid. British Journal of Cancer 86: pp. 1937-1942
    24. Fredriksson, L, Li, H, Fieber, C, Li, X, Eriksson, U (2004) Tissue plasminogen activator is a potent activator of PDGF-CC. EMBO Journal 23: pp. 3793-3802
    25. Kazlauskas, A, Cooper, JA (1989) Autophosphorylation of the PDGF receptor in the kinase insert region regulates interactions with cell proteins. Cell 58: pp. 1121-1133
    26. Antoniades, HN, Hunkapiller, MW (1983) Human platelet-derived growth factor (PDGF): amino-terminal amino acid sequence. Science 220: pp. 963-965
    27. Andrae, J, Gallini, R, Betsholtz, C (2008) Role of platelet-derived growth factors in physiology and medicine. Genes and Development 22: pp. 1276-1312
    28. Jain, RK (2003) Molecular regulation of vessel maturation. Nature Medicine 9: pp. 685-693
    29. Abramsson, A, Kurup, S, Busse, M (2007) Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development. Genes and Development 21: pp. 316-331
    30. Benjamin, LE, Hemo, I, Keshet, E (1998) A plasticity window for blood vessel remodelling is defined by pericyte coverage of the preformed endothelial network and is regulated by PDGF-B and VEGF. Development 125: pp. 1591-1598
    31. Oikawa, T, Onozawa, C, Sakaguchi, M, Morita, I, Murota, S (1994) Three isoforms of platelet-derived growth factors all have the capability to induce angiogenesis in vivo. Biological and Pharmaceutical Bulletin 17: pp. 1686-1688
    32. Lu, C, Thaker, PH, Lin, YG (2008) Impact of vessel maturation on antiangiogenic therapy in ovarian cancer. American Journal of Obstetrics and Gynecology 198: pp. e471-e479
    33. Valius, M, Kazlauskas, A (1993) Phospholipase C-gamma 1 and phosphatidylinositol 3 kinase are the downstream mediators of the PDGF receptor鈥檚 mitogenic signal. Cell 73: pp. 321-334
    34. Coughlin, SR, Escobedo, JA, Williams, LT (1989) Role of phosphatidylinositol kinase in PDGF receptor signal transduction. Science 243: pp. 1191-1194
    35. Heldin, CH, Ostman, A, Ronnstrand, L (1998) Signal transduction via platelet-derived growth factor receptors. Biochimica et Biophysica Acta 1378: pp. F79-F113
    36. Yao, R, Cooper, GM (1995) Requirement for phosphatidylinositol-3 kinase in the prevention of apoptosis by nerve growth factor. Science 267: pp. 2003-2006
    37. Huang, JS, Huang, SS, Deuel, TF (1984) Transforming protein of simian sarcoma virus stimulates autocrine growth of SSV-transformed cells through PDGF cell-surface receptors. Cell 39: pp. 79-87
    38. Greenhalgh, DG, Sprugel, KH, Murray, MJ, Ross, R (1990) PDGF and FGF stimulate wound healing in the genetically diabetic mouse. American Journal of Pathology 136: pp. 1235-1246
    39. Hellberg, C, Ostman, A, Heldin, CH (2010) PDGF and vessel maturation. Recent Results in Cancer Research 180: pp. 103-114
    40. Gaengel, K, Genove, G, Armulik, A, Betsholtz, C (2009) Endothelial-mural cell signaling in vascular development and angiogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 29: pp. 630-638
    41. Quaegebeur, A, Segura, I, Carmeliet, P (2010) Pericytes: blood-brain barrier safeguards against neurodegeneration?. Neuron 68: pp. 321-323
    42. Ribatti, D, Vacca, A, Roccaro, AM, Crivellato, E, Presta, M (2003) Erythropoietin as an angiogenic factor. European Journal of Clinical Investigation 33: pp. 891-896
    43. Crawford, Y, Kasman, I, Yu, L (2009) PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell 15: pp. 21-34
    44. Bergers, G, Song, S, Meyer-Morse, N, Bergsland, E, Hanahan, D (2003) Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. Journal of Clinical Investigation 111: pp. 1287-1295
    45. Erber, R, Thurnher, A, Katsen, AD (2004) Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte-mediated endothelial cell survival mechanisms. FASEB Journal 18: pp. 338-340
    46. Jo, N, Mailhos, C, Ju, M (2006) Inhibition of platelet-derived growth factor B signaling enhances the efficacy of anti-vascular endothelial growth factor therapy in multiple models of ocular neovascularization. American Journal of Pathology 168: pp. 2036-2053
    47. Hasumi, Y, Klosowska-Wardega, A, Furuhashi, M, Ostman, A, Heldin, CH, Hellberg, C (2007) Identification of a subset of pericytes that respond to combination therapy targeting PDGF and VEGF signaling. International Journal of Cancer 121: pp. 2606-2614
    48. Gerhardt, H, Semb, H (2008) Pericytes: gatekeepers in tumour cell metastasis?. Journal of Molecular Medicine (Berl) 86: pp. 135-144
    49. Alberts, DS, Liu, PY, Wilczynski, SP (2007) Phase II trial of imatinib mesylate in recurrent, biomarker positive, ovarian cancer (Southwest Oncology Group Protocol S0211). International Journal of Gynecological Cancer 17: pp. 784-788
    50. Coleman, RL, Broaddus, RR, Bodurka, DC (2006) Phase II trial of imatinib mesylate in patients with recurrent platinum- and taxane-resistant epithelial ovarian and primary peritoneal cancers. Gynecologic Oncology 101: pp. 126-131
    51. Posadas, EM, Kwitkowski, V, Kotz, HL (2007) A prospective analysis of imatinib-induced c-KIT modulation in ovarian cancer: a phase II clinical study with proteomic profiling. Cancer 110: pp. 309-317
    52. Safra, T, Andreopoulou, E, Levinson, B (2010) Weekly paclitaxel with intermittent imatinib mesylate (Gleevec): tolerance and activity in recurrent epithelial ovarian cancer. Anticancer Research 30: pp. 3243-3247
    53. Matulonis, UA, Berlin, S, Ivy, P (2009) Cediranib, an oral inhibitor of vascular endothelial growth factor receptor kinases, is an active drug in recurrent epithelial ovarian, fallopian tube, and peritoneal cancer. Journal of Clinical Oncology 27: pp. 5601-5606
    54. Raja, FA, Griffin, CL, Qian, W (2011) Initial toxicity assessment of ICON6: a randomised trial of cediranib plus chemotherapy in platinum-sensitive relapsed ovarian cancer. British Journal of Cancer 105: pp. 884-889
    55. Wilhelm, S, Carter, C, Lynch, M (2006) Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nature Reviews Drug Discovery 5: pp. 835-844
    56. Kane, RC, Farrell, AT, Saber, H (2006) Sorafenib for the treatment of advanced renal cell carcinoma. Clinical Cancer Research 12: pp. 7271-7278
    57. Kane, RC, Farrell, AT, Madabushi, R (2009) Sorafenib for the treatment of unresectable hepatocellular carcinoma. The Oncologist 14: pp. 95-100
    58. Matei, D, Sill, MW, Lankes, HA (2011) Activity of sorafenib in recurrent ovarian cancer and primary peritoneal carcinomatosis: a gynecologic oncology group trial. Journal of Clinical Oncology 29: pp. 69-75
    59. Welch, SA, Hirte, HW, Elit, L (2010) Sorafenib in combination with gemcitabine in recurrent epithelial ovarian cancer: a study of the Princess Margaret Hospital Phase II Consortium. International Journal of Gynecological Cancer 20: pp. 787-793
    60. Ramasubbaiah, R, Perkins, SM, Schilder, J (2011) Sorafenib in combination with weekly topotecan in recurrent ovarian cancer, a phase I/II study of the Hoosier Oncology Group. Gynecologic Oncology 123: pp. 499-504
    61. Herzog, TJ, Scambia, G, Kim, BG (2013) A randomized phase II trial of maintenance therapy with sorafenib in front-line ovarian carcinoma. Gynecologic Oncology 130: pp. 25-30
    62. Ledermann, JA, Hackshaw, A, Kaye, S (2011) Randomized phase II placebo-controlled trial of maintenance therapy using the oral triple angiokinase inhibitor BIBF 1120 after chemotherapy for relapsed ovarian cancer. Journal of Clinical Oncology 29: pp. 3798-3804
    63. Izzedine, H, Buhaescu, I, Rixe, O, Deray, G (2007) Sunitinib malate. Cancer Chemotheraphy and Pharmacology 60: pp. 357-364
    64. Biagi, JJ, Oza, AM, Chalchal, HI (2011) A phase II study of sunitinib in patients with recurrent epithelial ovarian and primary peritoneal carcinoma: an NCIC Clinical Trials Group Study. Annals of Oncology 22: pp. 335-340
    65. Friedlander, M, Hancock, KC, Rischin, D (2010) A phase II, open-label study evaluating pazopanib in patients with recurrent ovarian cancer. Gynecologic Oncology 119: pp. 32-37
    66. Normanno, N, Luca, A, Bianco, C (2006) Epidermal growth factor receptor (EGFR) signaling in cancer. Gene 366: pp. 2-16
    67. Yarden, Y, Sliwkowski, MX (2001) Untangling the ErbB signalling network. Nature Reviews Molecular Cell Biology 2: pp. 127-137
    68. Cascone, T, Herynk, MH, Xu, L (2011) Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor-resistant human lung adenocarcinoma. Journal of Clinical Investigation 121: pp. 1313-1328
    69. Viloria-Petit, A, Crombet, T, Jothy, S (2001) Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: a role for altered tumor angiogenesis. Cancer Research 61: pp. 5090-5101
    70. Vergote, IB, Jimeno, A, Joly, F (2014) Randomized phase III study of erlotinib versus observation in patients with no evidence of disease progression after first-line platin-based chemotherapy for ovarian carcinoma: a European Organisation for Research and Treatment of Cancer-Gynaecological Cancer Group, and Gynecologic Cancer Intergroup Study. Journal of Clinical Oncology 32: pp. 320-326
    71. Pakkala, S, Ramalingam, SS (2009) Combined inhibition of vascular endothelial growth factor and epidermal growth factor signaling in non-small-cell lung cancer therapy. Clinical Lung Cancer 10: pp. S17-S23
    72. Kimelman, D, Kirschner, M (1987) Synergistic induction of mesoderm by FGF and TGF-beta and the identification of an mRNA coding for FGF in the early Xenopus embryo. Cell 51: pp. 869-877
    73. Moerlooze, L, Spencer-Dene, B, Revest, JM, Hajihosseini, M, Rosewell, I, Dickson, C (2000) An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Development 127: pp. 483-492
    74. Beenken, A, Mohammadi, M (2009) The FGF family: biology, pathophysiology and therapy. Nature Reviews Drug Discovery 8: pp. 235-253
    75. Johnson, DE, Williams, LT (1993) Structural and functional diversity in the FGF receptor multigene family. Advances in Cancer Research 60: pp. 1-41
    76. Bae, JH, Schlessinger, J (2010) Asymmetric tyrosine kinase arrangements in activation or autophosphorylation of receptor tyrosine kinases. Molecules and Cells 29: pp. 443-448
    77. Eswarakumar, VP, Lax, I, Schlessinger, J (2005) Cellular signaling by fibroblast growth factor receptors. Cytokine and Growth Factor Reviews 16: pp. 139-149
    78. Cunningham, DL, Sweet, SM, Cooper, HJ, Heath, JK (2010) Differential phosphoproteomics of fibroblast growth factor signaling: identification of Src family kinase-mediated phosphorylation events. Journal of Proteome Research 9: pp. 2317-2328
    79. Klint, P, Claesson-Welsh, L (1999) Signal transduction by fibroblast growth factor receptors. Frontiers in Bioscience 4: pp. D165-D177
    80. Presta, M, Dell鈥橢ra, P, Mitola, S, Moroni, E, Ronca, R, Rusnati, M (2005) Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine and Growth Factor Reviews 16: pp. 159-178
    81. Cross, MJ, Claesson-Welsh, L (2001) FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends in Pharmacological Sciences 22: pp. 201-207
    82. Presta, M, Tiberio, L, Rusnati, M, Dell鈥橢ra, P, Ragnotti, G (1991) Basic fibroblast growth factor requires a long-lasting activation of protein kinase C to induce cell proliferation in transformed fetal bovine aortic endothelial cells. Cell Regulation 2: pp. 719-726
    83. Shono, T, Kanetake, H, Kanda, S (2001) The role of mitogen-activated protein kinase activation within focal adhesions in chemotaxis toward FGF-2 by murine brain capillary endothelial cells. Experimental Cell Research 264: pp. 275-283
    84. Casanovas, O, Hicklin, DJ, Bergers, G, Hanahan, D (2005) Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8: pp. 299-309
    85. Compagni, A, Wilgenbus, P, Impagnatiello, MA, Cotten, M, Christofori, G (2000) Fibroblast growth factors are required for efficient tumor angiogenesis. Cancer Research 60: pp. 7163-7169
    86. Giavazzi, R, Sennino, B, Coltrini, D (2003) Distinct role of fibroblast growth factor-2 and vascular endothelial growth factor on tumor growth and angiogenesis. American Journal of Pathology 162: pp. 1913-1926
    87. Nissen, LJ, Cao, R, Hedlund, EM (2007) Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. Journal of Clinical Investigation 117: pp. 2766-2777
    88. Lieu, C, Heymach, J, Overman, M, Tran, H, Kopetz, S (2011) Beyond VEGF: inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clinical Cancer Research 17: pp. 6130-6139
    89. Fujii, T, Kuwano, H (2010) Regulation of the expression balance of angiopoietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cellular and Developmental Biology - Animal 46: pp. 487-491
    90. Pepper, MS, Ferrara, N, Orci, L, Montesano, R (1992) Potent synergism between vascular endothelial growth factor and basic fibroblast growth factor in the induction of angiogenesis in vitro. Biochemical and Biophysical Research Communications 189: pp. 824-831
    91. Kopetz, S, Hoff, PM, Morris, JS (2010) Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance. Journal of Clinical Oncology 28: pp. 453-459
    92. Batchelor, TT, Sorensen, AG, Tomaso, E (2007) AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 11: pp. 83-95
    93. Carmeliet, P, Jain, RK (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature 473: pp. 298-307
    94. Augustin, HG, Koh, GY, Thurston, G, Alitalo, K (2009) Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nature Reviews Molecular Cell Biology 10: pp. 165-177
    95. Sundberg, C, Kowanetz, M, Brown, LF, Detmar, M, Dvorak, HF (2002) Stable expression of angiopoietin-1 and other markers by cultured pericytes: phenotypic similarities to a subpopulation of cells in maturing vessels during later stages of angiogenesis in vivo. Laboratory Investigation 82: pp. 387-401
    96. Winkler, F, Kozin, SV, Tong, RT (2004) Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 6: pp. 553-563
    97. Maisonpierre, PC, Suri, C, Jones, PF (1997) Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277: pp. 55-60
    98. Scharpfenecker, M, Fiedler, U, Reiss, Y, Augustin, HG (2005) The Tie-2 ligand angiopoietin-2 destabilizes quiescent endothelium through an internal autocrine loop mechanism. Journal of Cell Science 118: pp. 771-780
    99. Bach, F, Uddin, FJ, Burke, D (2007) Angiopoietins in malignancy. European Journal of Surgical Oncology 33: pp. 7-15
    100. Carmeliet, P, Jain, RK (2011) Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nature Reviews Drug Discovery 10: pp. 417-427
    101. Falcon, BL, Hashizume, H, Koumoutsakos, P (2009) Contrasting actions of selective inhibitors of angiopoietin-1 and angiopoietin-2 on the normalization of tumor blood vessels. American Journal of Pathology 175: pp. 2159-2170
    102. Koh, YJ, Kim, HZ, Hwang, SI (2010) Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage. Cancer Cell 18: pp. 171-184
    103. Herbst, RS, Hong, D, Chap, L (2009) Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. Journal of Clinical Oncology 27: pp. 3557-3565
    104. Birchmeier, C, Birchmeier, W, Gherardi, E, Vande Woude, GF (2003) Met, metastasis, motility and more. Nature Reviews Molecular Cell Biology 4: pp. 915-925
    105. Funakoshi, H, Nakamura, T (2003) Hepatocyte growth factor: from diagnosis to clinical applications. Clinica Chimica Acta 327: pp. 1-23
    106. Bottaro, DP, Rubin, JS, Faletto, DL (1991) Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 251: pp. 802-804
    107. Bolanos-Garcia, VM (2005) MET meet adaptors: functional and structural implications in downstream signalling mediated by the Met receptor. Molecular and Cellular Biochemistry 276: pp. 149-157
    108. Yu, J, Miehlke, S, Ebert, MP (2000) Frequency of TPR-MET rearrangement in patients with gastric carcinoma and in first-degree relatives. Cancer 88: pp. 1801-1806
    109. Dharmawardana, PG, Giubellino, A, Bottaro, DP (2004) Hereditary papillary renal carcinoma type I. Current Molecular Medicine 4: pp. 855-868
    110. Bussolino, F, Renzo, MF, Ziche, M (1992) Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. Journal of Cell Biology 119: pp. 629-641
    111. Kitajima, Y, Ide, T, Ohtsuka, T, Miyazaki, K (2008) Induction of hepatocyte growth factor activator gene expression under hypoxia activates the hepatocyte growth factor/c-Met system via hypoxia inducible factor-1 in pancreatic cancer. Cancer Science 99: pp. 1341-1347
    112. Kubota, T, Taiyoh, H, Matsumura, A (2009) NK4, an HGF antagonist, prevents hematogenous pulmonary metastasis by inhibiting adhesion of CT26 cells to endothelial cells. Clinical and Experimental Metastasis 26: pp. 447-456
    113. Sulpice, E, Ding, S, Muscatelli-Groux, B (2009) Cross-talk between the VEGF-A and HGF signalling pathways in endothelial cells. Biology of the Cell 101: pp. 525-539
    114. Puri, N, Khramtsov, A, Ahmed, S (2007) A selective small molecule inhibitor of c-Met, PHA665752, inhibits tumorigenicity and angiogenesis in mouse lung cancer xenografts. Cancer Research 67: pp. 3529-3534
    115. Cantelmo, AR, Cammarota, R, Noonan, DM (2010) Cell delivery of Met docking site peptides inhibit angiogenesis and vascular tumor growth. Oncogene 29: pp. 5286-5298
    116. Gherardi, E, Birchmeier, W, Birchmeier, C, Woude, GV (2012) Targeting MET in cancer: rationale and progress. Nature Reviews Cancer 12: pp. 89-103
    117. Hara, S, Nakashiro, K, Klosek, SK, Ishikawa, T, Shintani, S, Hamakawa, H (2006) Hypoxia enhances c-Met/HGF receptor expression and signaling by activating HIF-1alpha in human salivary gland cancer cells. Oral Oncology 42: pp. 593-598
    118. Ide, T, Kitajima, Y, Miyoshi, A (2006) Tumor-stromal cell interaction under hypoxia increases the invasiveness of pancreatic cancer cells through the hepatocyte growth factor/c-Met pathway. International Journal of Cancer 119: pp. 2750-2759
    119. Pennacchietti, S, Michieli, P, Galluzzo, M, Mazzone, M, Giordano, S, Comoglio, PM (2003) Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 3: pp. 347-361
    120. Qian, F, Engst, S, Yamaguchi, K (2009) Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Research 69: pp. 8009-8016
    121. Nakagawa, T, Tohyama, O, Yamaguchi, A (2010) E7050: a dual c-Met and VEGFR-2 tyrosine kinase inhibitor promotes tumor regression and prolongs survival in mouse xenograft models. Cancer Science 101: pp. 210-215
    122. You, WK, McDonald, DM (2008) The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Reports 41: pp. 833-839
    123. Shojaei, F, Lee, JH, Simmons, BH (2010) HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Cancer Research 70: pp. 10090-10100
    124. Tomioka, D, Maehara, N, Kuba, K (2001) Inhibition of growth, invasion, and metastasis of human pancreatic carcinoma cells by NK4 in an orthotopic mouse model. Cancer Research 61: pp. 7518-7524
    125. Burgess, T, Coxon, A, Meyer, S (2006) Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/c-Met-dependent human tumors. Cancer Research 66: pp. 1721-1729
    126. Martin, LP, Sill, M, Shahin, MS (2014) A phase II evaluation of AMG 102 (rilotumumab) in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecologic Oncology 132: pp. 526-530
    127. Buckanovich, R. J., Berger, R., Sella, A., et al. (2011). Results from phase II randomized discontinuation trial. / Journal of Clinical Oncology ASCO Annual Meeting. 29, abstract 5008.
    128. Pasquale, EB (2008) Eph-ephrin bidirectional signaling in physiology and disease. Cell 133: pp. 38-52
    129. Walker-Daniels, J, Hess, AR, Hendrix, MJ, Kinch, MS (2003) Differential regulation of EphA2 in normal and malignant cells. American Journal of Pathology 162: pp. 1037-1042
    130. Pasquale, EB (1997) The Eph family of receptors. Current Opinion in Cell Biology 9: pp. 608-615
    131. Thaker, PH, Deavers, M, Celestino, J (2004) EphA2 expression is associated with aggressive features in ovarian carcinoma. Clinical Cancer Research 10: pp. 5145-5150
    132. Cheng, N, Brantley, DM, Liu, H (2002) Blockade of EphA receptor tyrosine kinase activation inhibits vascular endothelial cell growth factor-induced angiogenesis. Molecular Cancer Research 1: pp. 2-11
    133. Spannuth, WA, Sood, AK, Coleman, RL (2008) Angiogenesis as a strategic target for ovarian cancer therapy. Nature Clinical Practice Oncology 5: pp. 194-204
    134. Lu, XS, Sun, W, Ge, CY, Zhang, WZ, Fan, YZ (2013) Contribution of the PI3K/MMPs/Ln-5gamma2 and EphA2/FAK/Paxillin signaling pathways to tumor growth and vasculogenic mimicry of gallbladder carcinomas. International Journal of Oncology 42: pp. 2103-2115
    135. Hess, AR, Seftor, EA, Gardner, LM (2001) Molecular regulation of tumor cell vasculogenic mimicry by tyrosine phosphorylation: role of epithelial cell kinase (Eck/EphA2). Cancer Research 61: pp. 3250-3255
    136. Landen, CN, Chavez-Reyes, A, Bucana, C (2005) Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Research 65: pp. 6910-6918
    137. Adam, MG, Berger, C, Feldner, A (2013) Synaptojanin-2 binding protein stabilizes the Notch ligands DLL1 and DLL4 and inhibits sprouting angiogenesis. Circulation Research 113: pp. 1206-1218
    138. Hu, W, Lu, C, Dong, HH (2011) Biological roles of the Delta family Notch ligand Dll4 in tumor and endothelial cells in ovarian cancer. Cancer Research 71: pp. 6030-6039
    139. Gale, NW, Dominguez, MG, Noguera, I (2004) Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proceedings of the National Academy of Sciences of the United States of America 101: pp. 15949-15954
    140. Lobov, IB, Renard, RA, Papadopoulos, N (2007) Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proceedings of the National Academy of Sciences of the United States of America 104: pp. 3219-3224
    141. Thurston, G, Noguera-Troise, I, Yancopoulos, GD (2007) The Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth. Nature Reviews Cancer 7: pp. 327-331
    142. Ridgway, J, Zhang, G, Wu, Y (2006) Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature 444: pp. 1083-1087
    143. Li, JL, Sainson, RC, Shi, W (2007) Delta-like 4 Notch ligand regulates tumor angiogenesis, improves tumor vascular function, and promotes tumor growth in vivo. Cancer Research 67: pp. 11244-11253
    144. Tolcher, AW, Messersmith, WA, Mikulski, SM (2012) Phase I study of RO4929097, a gamma secretase inhibitor of notch signaling, in patients with refractory metastatic or locally advanced solid tumors. Journal of Clinical Oncology 30: pp. 2348-2353
    145. Sahebjam, S, Bedard, PL, Castonguay, V (2013) A phase i study of the combination of ro4929097 and cediranib in patients with advanced solid tumours (PJC-004/NCI 8503). British Journal of Cancer 109: pp. 943-949
    146. Diaz-Padilla, I, Hirte, H, Oza, AM (2013) A phase Ib combination study of RO4929097, a gamma-secretase inhibitor, and temsirolimus in patients with advanced solid tumors. Investigational New Drugs 31: pp. 1182-1191
    147. Strosberg, JR, Yeatman, T, Weber, J (2012) A phase II study of RO4929097 in metastatic colorectal cancer. European Journal of Cancer 48: pp. 997-1003
    148. Summy, JM, Gallick, GE (2003) Src family kinases in tumor progression and metastasis. Cancer Metastasis Reviews 22: pp. 337-358
    149. Ma, WW, Adjei, AA (2009) Novel agents on the horizon for cancer therapy. CA: A Cancer Journal for Clinicians 59: pp. 111-137
    150. Frame, MC (2002) Src in cancer: deregulation and consequences for cell behaviour. Biochimica et Biophysica Acta 1602: pp. 114-130
    151. Trevino, JG, Summy, JM, Gray, MJ (2005) Expression and activity of SRC regulate interleukin-8 expression in pancreatic adenocarcinoma cells: implications for angiogenesis. Cancer Research 65: pp. 7214-7222
    152. Kanda, S, Miyata, Y, Kanetake, H, Smithgall, TE (2007) Non-receptor protein-tyrosine kinases as molecular targets for antiangiogenic therapy (review). International Journal of Molecular Medicine 20: pp. 113-121
    153. Labrecque, L, Royal, I, Surprenant, DS, Patterson, C, Gingras, D, B茅liveau, R (2003) Regulation of vascular endothelial growth factor receptor-2 activity by caveolin-1 and plasma membrane cholesterol. Molecular Biology of the Cell 14: pp. 334-347
    154. Eliceiri, BP, Puente, XS, Hood, JD (2002) Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. Journal of Cell Biology 157: pp. 149-160
    155. Kim, YM, Lee, YM, Kim, HS (2002) TNF-related activation-induced cytokine (TRANCE) induces angiogenesis through the activation of Src and phospholipase C (PLC) in human endothelial cells. Journal of Biological Chemistry 277: pp. 6799-6805
    156. Laird, AD, Li, G, Moss, KG (2003) Src family kinase activity is required for signal tranducer and activator of transcription 3 and focal adhesion kinase phosphorylation and vascular endothelial growth factor signaling in vivo and for anchorage-dependent and -independent growth of human tumor cells. Molecular Cancer Therapeutics 2: pp. 461-469
    157. Bankhead, C. (2010). ESMO: failed trials dominate gyn cancer session. Accessed 14 Oct 2010.
    158. McNeish, I. A., Ledermann, J. A., Webber, L. C., et al. (2013). A randomized placebo-controlled trial of saracatinib (AZD0530) plus weekly paclitaxel in platinum-resistant ovarian, fallopian-tube, or primary peritoneal cancer (SaPPrOC). / Journal of Clinical Oncology, 31.
    159. Hermann, C, Assmus, B, Urbich, C, Zeiher, AM, Dimmeler, S (2000) Insulin-mediated stimulation of protein kinase Akt: a potent survival signaling cascade for endothelial cells. Arteriosclerosis, Thrombosis, and Vascular Biology 20: pp. 402-409
    160. Granville, CA, Memmott, RM, Gills, JJ, Dennis, PA (2006) Handicapping the race to develop inhibitors of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway. Clinical Cancer Research 12: pp. 679-689
    161. Frisch, SM, Ruoslahti, E (1997) Integrins and anoikis. Current Opinion in Cell Biology 9: pp. 701-706
    162. Gerber, HP, McMurtrey, A, Kowalski, J (1998) Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3鈥?kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. Journal of Biological Chemistry 273: pp. 30336-30343
    163. Brunet, A, Bonni, A, Zigmond, MJ (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: pp. 857-868
    164. Yang, D, Sun, Y, Hu, L (2013) Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell 23: pp. 186-199
    165. Bianco, R, Garofalo, S, Rosa, R (2008) Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs. British Journal of Cancer 98: pp. 923-930
    166. Jin, H, Varner, J (2004) Integrins: roles in cancer development and as treatment targets. British Journal of Cancer 90: pp. 561-565
    167. Ruegg, C, Mariotti, A (2003) Vascular integrins: pleiotropic adhesion and signaling molecules in vascular homeostasis and angiogenesis. Cellular and Molecular Life Sciences 60: pp. 1135-1157
    168. Silva, RG, Tavora, B, Robinson, SD (2010) Endothelial alpha3beta1-integrin represses pathological angiogenesis and sustains endothelial-VEGF. American Journal of Pathology 177: pp. 1534-1548
    169. Assoian, RK (1997) Anchorage-dependent cell cycle progression. Journal of Cell Biology 136: pp. 1-4
    170. Urbich, C, Dernbach, E, Reissner, A, Vasa, M, Zeiher, AM, Dimmeler, S (2002) Shear stress-induced endothelial cell migration involves integrin signaling via the fibronectin receptor subunits alpha(5) and beta(1). Arteriosclerosis, Thrombosis, and Vascular Biology 22: pp. 69-75
    171. Abedi, H, Zachary, I (1997) Vascular endothelial growth factor stimulates tyrosine phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and paxillin in endothelial cells. Journal of Biological Chemistry 272: pp. 15442-15451
    172. Brooks, PC, Stromblad, S, Klemke, R, Visscher, D, Sarkar, FH, Cheresh, DA (1995) Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. Journal of Clinical Investigation 96: pp. 1815-1822
    173. Gutheil, JC, Campbell, TN, Pierce, PR (2000) Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clinical Cancer Research 6: pp. 3056-3061
    174. Stupp, R, Ruegg, C (2007) Integrin inhibitors reaching the clinic. Journal of Clinical Oncology 25: pp. 1637-1638
    175. Shibata, K, Kikkawa, F, Nawa, A, Suganuma, N, Hamaguchi, M (1997) Fibronectin secretion from human peritoneal tissue induces Mr 92,000 type IV collagenase expression and invasion in ovarian cancer cell lines. Cancer Research 57: pp. 5416-5420
    176. Sawada, K, Mitra, AK, Radjabi, AR (2008) Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Research 68: pp. 2329-2339
    177. Park, CC, Zhang, H, Pallavicini, M (2006) Beta1 integrin inhibitory antibody induces apoptosis of breast cancer cells, inhibits growth, and distinguishes malignant from normal phenotype in three dimensional cultures and in vivo. Cancer Research 66: pp. 1526-1535
    178. Bhaskar, V, Zhang, D, Fox, M (2007) A function blocking anti-mouse integrin alpha5beta1 antibody inhibits angiogenesis and impedes tumor growth in vivo. Journal of Translational Medicine 5: pp. 61
    179. Bhaskar, V, Fox, M, Breinberg, D (2008) Volociximab, a chimeric integrin alpha5beta1 antibody, inhibits the growth of VX2 tumors in rabbits. Investigational New Drugs 26: pp. 7-12
    180. Ramakrishnan, V, Bhaskar, V, Law, DA (2006) Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. Journal of Experimental Therapeutics and Oncology 5: pp. 273-286
    181. Bell-McGuinn, KM, Matthews, CM, Ho, SN (2011) A phase II, single-arm study of the anti-alpha5beta1 integrin antibody volociximab as monotherapy in patients with platinum-resistant advanced epithelial ovarian or primary peritoneal cancer. Gynecologic Oncology 121: pp. 273-279
    182. Naylor, MS, Stamp, GW, Foulkes, WD, Eccles, D, Balkwill, FR (1993) Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. Journal of Clinical Investigation 91: pp. 2194-2206
    183. Wu, S, Boyer, CM, Whitaker, RS (1993) Tumor necrosis factor alpha as an autocrine and paracrine growth factor for ovarian cancer: monokine induction of tumor cell proliferation and tumor necrosis factor alpha expression. Cancer Research 53: pp. 1939-1944
    184. Kulbe, H, Thompson, R, Wilson, JL (2007) The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Research 67: pp. 585-592
    185. Jin, DK, Shido, K, Kopp, HG (2006) Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nature Medicine 12: pp. 557-567
    186. Kaplan, RN, Riba, RD, Zacharoulis, S (2005) VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438: pp. 820-827
    187. Kryczek, I, Lange, A, Mottram, P (2005) CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Research 65: pp. 465-472
    188. Aguayo, A, Kantarjian, H, Manshouri, T (2000) Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood 96: pp. 2240-2245
    189. Charles, KA, Kulbe, H, Soper, R (2009) The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. Journal of Clinical Investigation 119: pp. 3011-3023
    190. Kulbe, H, Chakravarty, P, Leinster, DA (2012) A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment. Cancer Research 72: pp. 66-75
    191. Madhusudan, S, Muthuramalingam, SR, Braybrooke, JP (2005) Study of etanercept, a tumor necrosis factor-alpha inhibitor, in recurrent ovarian cancer. Journal of Clinical Oncology 23: pp. 5950-5959
    192. Giuntoli, RL, Webb, TJ, Zoso, A (2009) Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Research 29: pp. 2875-2884
    193. Lane, D, Matte, I, Rancourt, C, Piche, A (2011) Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer 11: pp. 210
    194. Dankbar, B, Padro, T, Leo, R (2000) Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood 95: pp. 2630-2636
    195. Nilsson, MB, Langley, RR, Fidler, IJ (2005) Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Research 65: pp. 10794-10800
    196. Rabinovich, A, Medina, L, Piura, B, Segal, S, Huleihel, M (2007) Regulation of ovarian carcinoma SKOV-3 cell proliferation and secretion of MMPs by autocrine IL-6. Anticancer Research 27: pp. 267-272
    197. Scambia, G, Testa, U, Benedetti Panici, P (1995) Prognostic significance of interleukin 6 serum levels in patients with ovarian cancer. British Journal of Cancer 71: pp. 354-356
    198. Guo, Y, Nemeth, J, O鈥橞rien, C (2010) Effects of siltuximab on the IL-6-induced signaling pathway in ovarian cancer. Clinical Cancer Research 16: pp. 5759-5769
    199. Coward, J, Kulbe, H, Chakravarty, P (2011) Interleukin-6 as a therapeutic target in human ovarian cancer. Clinical Cancer Research 17: pp. 6083-6096
    200. Fire, A, Xu, S, Montgomery, MK, Kostas, SA, Driver, SE, Mello, CC (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 391: pp. 806-811
    201. Hammond, SM, Bernstein, E, Beach, D, Hannon, GJ (2000) An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature 404: pp. 293-296
    202. Elbashir, SM, Harborth, J, Lendeckel, W, Yalcin, A, Weber, K, Tuschl, T (2001) Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411: pp. 494-498
    203. Fattal, E, Bochot, A (2006) Ocular delivery of nucleic acids: antisense oligonucleotides, aptamers and siRNA. Advanced Drug Delivery Reviews 58: pp. 1203-1223
    204. Bitko, V, Musiyenko, A, Shulyayeva, O, Barik, S (2005) Inhibition of respiratory viruses by nasally administered siRNA. Nature Medicine 11: pp. 50-55
    205. Ozpolat, B, Sood, AK, Lopez-Berestein, G (2010) Nanomedicine based approaches for the delivery of siRNA in cancer. Journal of Internal Medicine 267: pp. 44-53
    206. Zhou, J, Shum, KT, Burnett, JC, Rossi, JJ (2013) Nanoparticle-based delivery of RNAi therapeutics: progress and challenges. Pharmaceuticals (Basel) 6: pp. 85-107
    207. Tan, WB, Jiang, S, Zhang, Y (2007) Quantum-dot based nanoparticles for targeted silencing of HER2/neu gene via RNA interference. Biomaterials 28: pp. 1565-1571
    208. Lee, JH, Lee, K, Moon, SH, Lee, Y, Park, TG, Cheon, J (2009) All-in-one target-cell-specific magnetic nanoparticles for simultaneous molecular imaging and siRNA delivery. Angewandte Chemie International Edition in English 48: pp. 4174-4179
    209. Yu, D, Peng, P, Dharap, SS (2005) Antitumor activity of poly(ethylene glycol)-camptothecin conjugate: the inhibition of tumor growth in vivo. Journal of Controlled Release 110: pp. 90-102
    210. Maeda, H (2001) The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Advances in Enzyme Regulation 41: pp. 189-207
    211. Nikitenko, NA, Prassolov, VS (2013) Non-viral delivery and therapeutic application of small interfering RNAs. Acta Naturae 5: pp. 35-53
    212. Davis, ME, Zuckerman, JE, Choi, CH (2010) Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464: pp. 1067-1070
    213. Heidel, JD, Yu, Z, Liu, JY (2007) Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proceedings of the National Academy of Sciences of the United States of America 104: pp. 5715-5721
    214. Matei, D, Emerson, RE, Schilder, J (2008) Imatinib mesylate in combination with docetaxel for the treatment of patients with advanced, platinum-resistant ovarian cancer and primary peritoneal carcinomatosis: a Hoosier Oncology Group trial. Cancer 113: pp. 723-732
    215. Juretzka, M, Hensley, ML, Tew, W (2008) A phase 2 trial of oral imatinib in patients with epithelial ovarian, fallopian tube, or peritoneal carcinoma in second or greater remission. European Journal of Gynaecological Oncology 29: pp. 568-572
    216. Liu, JF, Tolaney, SM, Birrer, M (2013) A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. European Journal of Cancer 49: pp. 2972-2978
    217. Hjalmarson, A (1990) Heart rate and beta-adrenergic mechanisms in acute myocardial infarction. Basic Research in Cardiology 85: pp. 325-333
    218. Bodnar, L, Gornas, M, Szczylik, C (2011) Sorafenib as a third line therapy in patients with epithelial ovarian cancer or primary peritoneal cancer: a phase II study. Gynecologic Oncology 123: pp. 33-36
    219. Campos, SM, Penson, RT, Matulonis, U (2013) A phase II trial of sunitinib malate in recurrent and refractory ovarian, fallopian tube and peritoneal carcinoma. Gynecologic Oncology 128: pp. 215-220
    220. Baumann, KH, Bois, A, Meier, W (2012) A phase II trial (AGO 2.11) in platinum-resistant ovarian cancer: a randomized multicenter trial with sunitinib (SU11248) to evaluate dosage, schedule, tolerability, toxicity and effectiveness of a multitargeted receptor tyrosine kinase inhibitor monotherapy. Annals of Oncology 23: pp. 2265-2271
    221. Karlan, BY, Oza, AM, Richardson, GE (2012) Randomized, double-blind, placebo-controlled phase II study of AMG 386 combined with weekly paclitaxel in patients with recurrent ovarian cancer. Journal of Clinical Oncology 30: pp. 362-371
    222. Secord, AA, Teoh, DK, Barry, WT (2012) A phase I trial of dasatinib, an SRC-family kinase inhibitor, in combination with paclitaxel and carboplatin in patients with advanced or recurrent ovarian cancer. Clinical Cancer Research 18: pp. 5489-5498
    223. Schilder, RJ, Brady, WE, Lankes, HA (2012) Phase II evaluation of dasatinib in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecologic Oncology 127: pp. 70-74
    224. Behbakht, K, Sill, MW, Darcy, KM (2011) Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a Gynecologic Oncology Group study. Gynecologic Oncology 123: pp. 19-26
    225. Temkin, SM, Yamada, SD, Fleming, GF (2010) A phase I study of weekly temsirolimus and topotecan in the treatment of advanced and/or recurrent gynecologic malignancies. Gynecologic Oncology 117: pp. 473-476
    226. Kollmannsberger, C, Hirte, H, Siu, LL (2012) Temsirolimus in combination with carboplatin and paclitaxel in patients with advanced solid tumors: a NCIC-CTG, phase I, open-label dose-escalation study (IND 179). Annals of Oncology 23: pp. 238-244
  • 刊物类别:Medicine
  • 刊物主题:Medicine & Public Health
    Oncology
    Cancer Research
  • 出版者:Springer Netherlands
  • ISSN:1573-7233
文摘
Resistance to chemotherapy is among the most important issues in the management of ovarian cancer. Unlike cancer cells, which are heterogeneous as a result of remarkable genetic instability, stromal cells are considered relatively homogeneous. Thus, targeting the tumor microenvironment is an attractive approach for cancer therapy. Arguably, anti-vascular endothelial growth factor (anti-VEGF) therapies hold great promise, but their efficacy has been modest, likely owing to redundant and complementary angiogenic pathways. Components of platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), and other pathways may compensate for VEGF blockade and allow angiogenesis to occur despite anti-VEGF treatment. In addition, hypoxia induced by anti-angiogenesis therapy modifies signaling pathways in tumor and stromal cells, which induces resistance to therapy. Because of tumor cell heterogeneity and angiogenic pathway redundancy, combining cytotoxic and targeted therapies or combining therapies targeting different pathways can potentially overcome resistance. Although targeted therapy is showing promise, much more work is needed to maximize its impact, including the discovery of new targets and identification of individuals most likely to benefit from such therapies.
NGLC 2004-2010.National Geological Library of China All Rights Reserved.
Add:29 Xueyuan Rd,Haidian District,Beijing,PRC. Mail Add: 8324 mailbox 100083
For exchange or info please contact us via email.