Skip to main content

Protein Tyrosine Kinases as Targets for Cancer and Other Indications

  • Chapter

Part of the book series: Cancer Drug Discovery and Development ((CDD&D))

Abstract

The identification and characterization of the members of individual signal transduction cascades, and advances in understanding how these signals are integrated in normal and pathological conditions have provided new strategies for therapeutic intervention. Rapid progress has occurred in last few years in the development of inhibitors that target protein tyrosine kinases (PTKs), enzymes that transfer the γ-phosphate group of adenosine triphosphate (ATP) to the hydroxyl group of tyrosine residues on target proteins. Although PTKs represent a small percentage of the total number of kinases in the “kinome,” 90 of 518, a disproportional number of inhibitors currently in clinical trials are directed against them; e.g., more than 20 different tyrosine kinases are being evaluated as potential targets in oncology. There are a number of reasons why tyrosine kinases have been considered to be good targets. Epistatically, PTKs are located upstream and downstream of tumor suppressor genes or oncogenes and have been demonstrated to play central roles in apoptosis, proliferation, invasion, and differentiation (1). Aberrant activation of tyrosine kinases, owing to mutation or overexpression, is sufficient for them to become transforming in cellular and animal models. The majority of targets are receptor protein tyrosine kinases (RPTKs), as deregulating mutations of over half of the known RPTKs have been associated with different human malignancies; see Table 1 for examples. Finally, and equally as important as the epidemiological and biochemical data, the prevalence of PTKs as targets is because of the fact that they are considered druggable.

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   169.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   219.00
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Blume-Jensen P, Hunter T. Oncogenic kinase signalling [Review]. Nature 2001; 411: 355–365.

    PubMed  CAS  Google Scholar 

  2. Noble ME, Endicott JA, Johnson LN. Protein kinase inhibitors: insights into drug design from structure. Science 2004; 303:1800–1805.

    PubMed  CAS  Google Scholar 

  3. Krystal-Imatinib mesylate (STI571) for myeloid malignancies other than CML. Leuk Res 2004; 28(Suppl 1):53–59.

    Google Scholar 

  4. Druker BJ, Sawyers CL, Kantarjian HR, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med 2001; 344:1038–1042.

    PubMed  CAS  Google Scholar 

  5. Heinrich MC, Griffith DJ, Druker BJ, et al. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood 2000; 96:925–932.

    PubMed  CAS  Google Scholar 

  6. Daley GQ, Van Etten RA, Baltimore D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science 1990; 247:824–830.

    PubMed  CAS  Google Scholar 

  7. Savage DG, and Antman KH. Imatinib mesylate; a new oral targeted therapy. N Engl J Med 2002; 346:683–693.

    PubMed  CAS  Google Scholar 

  8. Griffin JD. Resistance to targeted therapy in leukaemia. Lancet 2002; 359:458–459.

    PubMed  Google Scholar 

  9. Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 2001; 293:876–880.

    PubMed  CAS  Google Scholar 

  10. Gorre ME, Sawyers CL. Molecular mechanisms of resistance to STI571 in chronic myeloid leukemia. Curr Opin Hematol 2002; 9:303–307.

    PubMed  Google Scholar 

  11. Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002; 347:472–480.

    PubMed  CAS  Google Scholar 

  12. van Oosterom AJ, Judson I, Verweij J, et al. Safety and efficacy of imatinib (STI571) in metastatic gastrointestinal stromal tumours: a phase I study. Lancet 2001; 358:1421–1423.

    PubMed  Google Scholar 

  13. Heinrich MC, Corless CL, Demetri GD, et al. Kinase mutations and imatinib response in patients with metastatic gastrointestinal stromal tumor. [Report]. J Clin Oncol 2003; 21:4342–4349.

    PubMed  CAS  Google Scholar 

  14. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA-activating mutations in gastrointestinal stromal tumors. Science 2003; 299:708–710.

    PubMed  CAS  Google Scholar 

  15. Rubin BP, Schuetze SM, Eary JF, et al. Molecular targeting of platelet-derived growth factor B by imatinib mesylate in a patient with metastatic dermatofibrosarcoma protuberans [see comment]. J Clin Oncol 2002; 20:3586–3591.

    PubMed  CAS  Google Scholar 

  16. Sawyers CL. Imatinib GIST keeps finding new indications: successful treatment of dermatofibrosarcoma protuberans by targeted inhibition of the platelet-derived growth factor receptor [comment]. J Clin Oncol 2002; 20:3568–3569.

    PubMed  Google Scholar 

  17. Apperley JF, Gardembas M, Melo JV, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. N Engl J Med 2002; 347:481–487.

    PubMed  CAS  Google Scholar 

  18. Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome [see comment]. N Engl J Med 2003; 348:1201–1214.

    PubMed  CAS  Google Scholar 

  19. Kemmer K, Corless CL, Fletcher JA, et al. KIT mutations are common in testicular seminomas. Am J Pathol 2004; 164:305–313.

    PubMed  CAS  Google Scholar 

  20. Pardanani A, Elliott M, Reeder T. Imatinib for systemic mast-cell disease. Lancet 2003; 362:535–536.

    PubMed  CAS  Google Scholar 

  21. Tefferi A, Mesa RA, Gray LA, et al. Phase 2 trial of imatinib mesylate in myelofibrosis with myeloid metaplasia. Blood 2002; 99:3854–3856.

    PubMed  CAS  Google Scholar 

  22. Pietras K, Ostman A, Sjoquist M, et al. Inhibition of platelet-derived growth factor receptors reduces interstitial hypertension and increases transcapillary transport in tumors. Cancer Res 1902; 61:2929–2934.

    Google Scholar 

  23. Myllarniemi M, Frosen J, Calderon Ramirez LG, et al. Selective tyrosine kinase inhibitor for the platelet-derived growth factor receptor in vitro inhibits smooth muscle cell proliferation after reinjury of arterial intima in vivo. Cardiovasc Drugs Ther 1999; 13:159–168.

    PubMed  CAS  Google Scholar 

  24. Savikko J, Taskinen E, Von Willebrand E. Chronic allograft nephropathy is prevented by inhibition of platelet-derived growth factor receptor: tyrosine kinase inhibitors as a potential therapy. Transplantation 2003; 75:1147–1153.

    PubMed  CAS  Google Scholar 

  25. Lassila M, Allen TJ, Cao, Z, et al. Imatinib attenuates diabetes-associated atherosclerosis. Arterioscler Thromb Vasc Biol 2004; 24:935–942.

    PubMed  CAS  Google Scholar 

  26. Sihvola RK, Tikkanen JM, Krebs R, et al. Platelet-derived growth factor receptor inhibition reduces allograft arteriosclerosis of heart and aorta in cholesterol-fed rabbits. Transplantation 2003; 75:334–339.

    PubMed  CAS  Google Scholar 

  27. Ahuja H, Bar-Eli M, Advani SH, Benchimol S, Cline MJ. Alterations in the p53 gene and the clonal evolution of the blast crisis of chronic myelocytic leukemia. Proc Nat Acad Sci US Am 1989; 86:6783–6787.

    CAS  Google Scholar 

  28. Foti A, Ahuja HG, Allen SL, et al. Correlation between molecular and clinical events in the evolution of chronic myelocytic leukemia to blast crisis. Blood 1991; 77:2441–2444.

    PubMed  CAS  Google Scholar 

  29. Sill H, Goldman JM, Cross NC. Homozygous deletions of the p16 tumor-suppressor gene are associated with lymphoid transformation of chronic myeloid leukemia. Blood 1995; 85:2013–2016.

    PubMed  CAS  Google Scholar 

  30. Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood 1999; 93:3074–3080.

    PubMed  CAS  Google Scholar 

  31. Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 1996; 10:1911–1918.

    PubMed  CAS  Google Scholar 

  32. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia [Review]. Blood 2002; 100:1532–1542.

    PubMed  CAS  Google Scholar 

  33. Carnicer MJ, Nomdedeu JF, Lasa A, et al. FLT3 mutations are associated with other molecular lesions in AML. Leuk Res 2004; 28:19–23.

    PubMed  CAS  Google Scholar 

  34. Sohal J, Phan VT, Chan PV. A model of APL with FLT3 mutation is responsive to retinoic acid and a receptor tyrosine kinase inhibitor, SU11657. Blood 2003; 101:3188–3197.

    PubMed  CAS  Google Scholar 

  35. Levis M, Allebach J, Tse KF, et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood 2002; 99:3885–3891.

    PubMed  CAS  Google Scholar 

  36. Fabbro D, Ruetz S, Bodis S, et al. PKC412—a protein kinase inhibitor with a broad therapeutic potential. Anticancer Drug Des 2000; 15:17–28.

    PubMed  CAS  Google Scholar 

  37. Weisberg E, Boulton C, Kelly LM, et al. Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitors PKC412. Cancer Cell 2002; 1:433–443.

    PubMed  CAS  Google Scholar 

  38. Abrams TJ, Lee LB, Murray LJ, Mendel DB, Cherrington JM. Inhibition of Kit-positive SCLC growth by SU11248, a novel tyrosine kinase inhibitors. 93rd Annual Meeting of the AACR Symposium on Molecular Targets and Cancer Therapeutics 2002.

    Google Scholar 

  39. Mendel DB, Laird AD, Xin X, et al. In vivo antitumor and mechanism of action studies of SU11248, a potent and selective inhibito of the VEGF and PDGF receptors. 93rd Annual Meeting of the AACR Symposium on Molecular Targets and Cancer Therapeutics 2002; 93:Abstr. 5349.

    Google Scholar 

  40. Kelly LM, Yu JC, Boulton CL, et al. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML). Cancer Cell 2002; 1:421–432.

    PubMed  CAS  Google Scholar 

  41. O’Farrell AM, Abrams TJ, Yuen HA, et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003; 101:3597–3605.

    PubMed  CAS  Google Scholar 

  42. Smith BD, Levis M, Beran M, et al. Single agent CEP-701, a novel FLT3 inhibitor, shows initial response in patients with refractory acute myeloid leukemia. Proc Am Soc Clin Oncol 2003; 194.

    Google Scholar 

  43. Smith BD, Levis M, Beran M, et al. Single agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 2004; 2003–2011.

    Google Scholar 

  44. Mendelsohn J, Baselga J. The EGF receptor family as targets for cancer therapy. Oncogene 2000; 19:6550–6565.

    PubMed  CAS  Google Scholar 

  45. Velu TJ. Structure, function and transforming potential of the epidermal growth factor receptor [Review]. Mol Cell Endocrinol 1990; 70:205–216.

    PubMed  CAS  Google Scholar 

  46. Yarden Y, Slimkowski MX. Untangling the erbB signalling network. Nat Rev Mol Cell Biol 2001; 2:127–137.

    PubMed  CAS  Google Scholar 

  47. Yarden Y. The EGFR family and its lignad in human cancer signalling mechanisms and therapeutic opportunities. Eur J Cancer 2001; 37:S3–S8.

    PubMed  CAS  Google Scholar 

  48. Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol 1995; 19:183–232.

    PubMed  CAS  Google Scholar 

  49. Garcia IE, Adams, GP, Sundareshan P, et al. Expression of mutated epidermal growth factor receptor by non-small cell lung carcinomas. Cancer Res 1993; 53:3217–3220.

    Google Scholar 

  50. Libermann TA, Nusbaum HR, Razon N, Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature 1985; 313:144–147.

    PubMed  CAS  Google Scholar 

  51. Sridhar SS, Seymour L, Shepherd FA. Inhibitors of epidermal-growth-factor receptors: a review of clinical research with a focus on non-small-cell lung cancer [Review]. Lancet Oncol 2003; 4:397–406.

    PubMed  CAS  Google Scholar 

  52. Hirsch FR, Langer CJ. The role of HER2/neu expression and trastuzumab in non-small cell lung cancer [Review]. Semin Oncol 2004; 31:Suppl 82.

    Google Scholar 

  53. Baselga J, Pfister D, Cooper MR, et al. Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin. J Clin Oncol 2000; 18:904–914.

    PubMed  CAS  Google Scholar 

  54. Foon KA, Yang XD, Weiner LM, et al. Preclinical and clinical evaluations of ABX-EGF, a fully human anti-epidermal growth factor receptor antibody [Review]. Int J Radiat Oncol Biol Phys 2004;58:984–990.

    PubMed  CAS  Google Scholar 

  55. Lynch DH, Yang XD. Therapeutic potential of ABX-EGF: a fully human anti-epidermal growth factor receptor monoclonal antibody for cancer treatment. Semin Oncol 2002; 29:47–50.

    PubMed  CAS  Google Scholar 

  56. Yang XD, Jia XC, Corvalan JR, Wang P, Davis CG. Development of ABX-EGF, a fully human anti-EGF receptor monoclonal antibody, for cancer therapy. Crit Rev Oncol Hematol 2001; 38:17–23.

    PubMed  CAS  Google Scholar 

  57. Repp R, van Ojik HH, Valerius T, et al. Phase I clinical trial of the bispecific antibody MDX-H210 (anti-FcgammaRI × anti-HER-2/neu) in combination with Filgrastim (G-CSF) for treatment of advanced breast cancer. Br J Cancer 2003; 89:2234–2243.

    PubMed  CAS  Google Scholar 

  58. Wallace PK, Romet-Lemonne JL, Chokri M, Kasper LH, Fanger MW, Fadul CE. Production of macrophage-activated killer cells for targeting of glioblastoma cells with bispecific antibody to FcgammaRI and the epidermal growth factor receptor. Cancer Immunol Immunother 2000; 49:493–503.

    PubMed  CAS  Google Scholar 

  59. Dancey J, Sausville EA. Issues and progress with protein kinase inhibitors for cancer treatment [Review]. Nat Rev Drug Discov 2003; 2:296–313.

    PubMed  CAS  Google Scholar 

  60. Dancey J. Epidermal growth factor receptor inhibitors in clinical development [Review]. Int J Radiat Oncol Biol Phys 2004; 58:1003–1007.

    PubMed  CAS  Google Scholar 

  61. Dancey J, Freidlin B. Targeting epidermal growth factor receptor—are we missing the mark? [Review]. Lancet 2003; 362:62–64.

    PubMed  CAS  Google Scholar 

  62. Herbst RS, Giaccone G, Schiller JH, et al. Gefitinib in combination with paclitaxel and carboplatin in advanced non-small-cell lung cancer: a phase III trial—INTACT 2 [see comment]. J Clin Oncol 2004; 22:785–794.

    PubMed  CAS  Google Scholar 

  63. Rich JN, Reardon DA, Peery T, et al. Phase II trial of gefitinib in recurrent glioblastoma. J Clin Oncol 2004; 22:133–142.

    PubMed  CAS  Google Scholar 

  64. Sanders. Investigations into the mechanism for suramin as an inhibitor of cAMP-dependent protein kinase. 1996; BIOL-035.

    Google Scholar 

  65. Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS, Siu LL. Multicenter phase II study of erlotinib, an oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with recurrent or metastatic squamous cell cancer of the head and neck. J Clin Oncol 2004; 22:77–85.

    PubMed  CAS  Google Scholar 

  66. Baselga J, Rischin D, Ranson M, et al. Phase I safety, pharmacokinetic, and pharmacodynamic trial of ZD1839, a selective oral epidermal growth factor receptor tyrosine kinase inhibitor, in patients with five selected solid tumor types [see comment]. J Clin Oncol 2002; 20:4292–4302.

    PubMed  CAS  Google Scholar 

  67. Cortes-Funes H, Soto Parra H. Extensive experience of disease control with gefitinib and the role of prognostic markers. Br J Cancer 2003; 89:Suppl 8.

    Google Scholar 

  68. Santoro A, Cavina R, Latteri F, et al. Activity of a specific inhibitor, gefitinib (Iressa, ZD1839), of epidermal growth factor receptor in refractory non-small-cell lung cancer. Ann Oncol 2004; 15(1):33–37.

    PubMed  CAS  Google Scholar 

  69. Phase III trials of Tarceva(TM) plus chemotherapy in first-line non-small cell lung cancer do not meet primary efficacy endpoint. Genentech press release; 2003.

    Google Scholar 

  70. Allen LF, Lenehan PF, Eiseman IA, Elliott WL, Fry DW. Potential benefits of the irreversible pan-erbB inhibitor, CI-1033, in the treatment of breast cancer. Semin Oncol 2002; 3:11–21.

    Google Scholar 

  71. Allen LF, Eiseman IA, Fry DW, Lenehan PF. CI-1033, an irreversible pan-erbB receptor inhibitor and its potential application for the treatment of breast cancer. Semin Oncol 2003; 5:65–78.

    Google Scholar 

  72. Wissner A, Overbeek E, Reich MF, et al. Synthesis and structure-activity relationships of 6,7-disubstituted 4-anilinoquinoline-3-carbonitriles. The design of an orally activa, irreversible inhibitor of the tyrosine kinase activity of the epidermal growth factor receptor (EGFR) and the human epidermal growth factor receptor-2 (HER-2). J Med Chem 2003; 46:49–63.

    PubMed  CAS  Google Scholar 

  73. Shin D, Nemunaitis J, Zinner RG, et al. A phase I clinical and biomarker study of CI-1033, a novel pan-ErbB tyrosine inhibitor in patients with solid tumors. Proc Am Soc Clin Oncol 2001; 20:Abst 324.

    Google Scholar 

  74. Anido J, Matar P, Albanell J, et al. ZD1839, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, induces the formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers and prevents heregulin signaling in HER2-overexpressing breast cancer cells. Clin Cancer Res 2003; 9:1274–1283.

    PubMed  CAS  Google Scholar 

  75. Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors [Review]. Nat Med 2003; 9:669–676.

    PubMed  CAS  Google Scholar 

  76. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch [Review]. Nat Rev Cancer 2003; 3:401–410.

    PubMed  CAS  Google Scholar 

  77. Folkman J. Anti-angiogenesis: new concept for therapy of solid tumors. Ann Surg 1972; 175:409–419.

    PubMed  CAS  Google Scholar 

  78. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer [see comment] N Engl J Med 2003; 349:427–434.

    PubMed  CAS  Google Scholar 

  79. Cobleigh MA, Langmuir VK, Sledge GW, et al. A phase I/II dose-escalation trial of bevacizumab in previously treated metastatic breast cancer. Sem Oncol 2003; 30:Suppl 24.

    Google Scholar 

  80. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer [see comment]. J Clin Oncol 2003; 21:60–65.

    PubMed  CAS  Google Scholar 

  81. FDA approves avastin, a targeted therapy for first-line metastatic colorectal cancer patients. Genentech press release; 2004.

    Google Scholar 

  82. Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med 2004; 10:145–147.

    PubMed  CAS  Google Scholar 

  83. Posey JA, Ng TC, Yang B, et al. A phase I study of anti-kinase insert domain-containing receptor antibody, IMC-1C11, in patients with liver metastases from colorectal carcinoma. Clin Cancer Res 2003; 9:1323–1332.

    PubMed  CAS  Google Scholar 

  84. Manley PW, Bold G, Fendrich G, et al. Advances in the structural biology, design and clinical development of VEGF-R kinase inhibitors for the treatment of angiogenesis. Biochim Biophys Acta 2004; 1697(1–2):17–27.

    PubMed  CAS  Google Scholar 

  85. Sandberg JA, Parker VP, Blanchard KS, et al. Pharmacokinetics and tolerability of an antiangiogenic ribozyme (ANGIOZYME) in healthy volunteers. J Clin Pharmacol 2000; 40:1462–1469.

    PubMed  CAS  Google Scholar 

  86. Kuenen BC, Tabernero J, Baselga J, et al. Efficacy and toxicity of the angiogenesis inhibitor SU5416 as a single agent in patients with advanced renal cell carcinoma, melanoma, and soft tissue sarcoma. Clin Cancer Res 2003; 9:1648–1655.

    PubMed  CAS  Google Scholar 

  87. Mendel DB, Laird AD, Smolich BD, et al. Development of SU5416, a selective small molecule inhibitor of VEGFR receptor tyrosine kinase activity, as an anti-angiogenesis agent. Anticancer Drug Des 2000; 15:29–41.

    PubMed  CAS  Google Scholar 

  88. Fabbro D, Manley PW. Su-6668.SUGEN. Curr Opin Investig Drugs 2001; 2:1142–1148.

    PubMed  CAS  Google Scholar 

  89. Mendel DB, Laird AD, Xin X, et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003; 9:327–337.

    PubMed  CAS  Google Scholar 

  90. O’Farrell AM, Abrams TJ, Yuen HA, et al. M.SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003; 101:3597–3605.

    PubMed  CAS  Google Scholar 

  91. Wedge SR, Ogilvie DJ, Dukes M, et al. VEGF receptor tyrosine kinase inhibitors as potential anti-tumor agents. Proc Am Assoc Cancer Res 2000; 41:3610.

    Google Scholar 

  92. Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induce responses and tumor growth after oral administration. Cancer Res 2000; 60:2178–2189.

    PubMed  CAS  Google Scholar 

  93. Wood JM, Bold G, Buchdunger E, et al. PTK787/ZK 222584, a novel and potent inhibitor of vascular endothelial growth factor receptor tyrosine kinases, impairs vascular endothelial growth factor-induced responses and tumor growth after oral administration. Cancer Res 2000; 60:2178–2189.

    PubMed  CAS  Google Scholar 

  94. Morgan B, Thomas AL, Drevs JN, et al. Dynamic contrast-enhanced magnetic resonance imaging as a biomarker for the pharmacological response of PTK787/ZK 222584, an inhibitor of the vascular endothelial growth factor receptor tyrosine kinases, in patients with advanced colorectal cancer and liver metastases: results from two phase I studies. J Clin Oncol 2003; 21:3955–3964.

    PubMed  CAS  Google Scholar 

  95. Ciardiello F, Caputo R, Damiano V, et al. Antitumor effects of ZD6474, a small molecule vascular endothelial growth factor receptor tyrosine kinase inhibitor, with additional activity against epidermal growth factor receptor tyrosine kinase [see comment]. Clin Cancer Res 2003; 9:1546–1556.

    PubMed  CAS  Google Scholar 

  96. Hurwitz HI, Eckhardt SG, Holden SN, et al. A phase I study of ZD6474, an oral VEGF receptor tyrosine kinase inhibitor, in patients with solid tumors. Proceedings of the 12th AACR-NCI-EORTC International Conference, Molecular Targets and Cancer Therapeutics, Discovery, Biology and Clinical Applications 2001; 7:5.

    Google Scholar 

  97. Hurwitz HI, Eckhardt SG, Holden SN, et al. Phase I pharmacokinetic and biological study of the angiogenesis inhibitor ZD6474, in patients with solid tumors. Proc Am Soc Clin Oncol 2001; 20:396.

    Google Scholar 

  98. Hampton T. Scientists take aim at angiogenesis to treat degenerative eye diseases. JAMA 2004; 291:1309–1310.

    PubMed  CAS  Google Scholar 

  99. Vinores SA. Technology evaluation: pegaptanib, Eyetech/Pfizer. Curr Opin Mol Ther 2003; 5:673–679.

    PubMed  CAS  Google Scholar 

  100. Ferrara N. Vascular endothelial growth factor. Trends Cardiovasc Med 1993; 3:244–250.

    CAS  Google Scholar 

  101. Sauder DN, DeKoven J, Champagne P, Croteau D, Dupont E. Neovastat (AE-941), an inhibitor of angiogenesis: randomized phase I/II clinical trial results in patients with plaque psoriasis. J Am Acad Dermatol 2002; 47:535–541.

    PubMed  Google Scholar 

  102. Grosios K, Wood J, Esser R, Raychaudhuri A, Dawson J. Angiogenesis inhibition by the novel VEGF receptor tyrosine kinase inhibitor, PTK787/ZK222584, causes significant antiarthritic effects in models of rheumatoid arthritis. Inflamm Res 2004; 53(4):133–142.

    PubMed  CAS  Google Scholar 

  103. Jeffers M, LaRochelle WJ, Lichenstein HS. Fibroblast growth factors in cancer: therapeutic possibilities. Expert Opin Ther Targets 2002; 6(4):469–482.

    PubMed  CAS  Google Scholar 

  104. Baserga R, Hongo A, Rubini M, Prisco M, Valentinis B. The IGF-I receptor in cell growth, transformation and apoptosis. Biochim Biophys Acta 1997; 1332:F105–F126.

    PubMed  CAS  Google Scholar 

  105. Baserga R. The contradictions of the insulin-like growth factor 1 receptor [Review]. Oncogene 2000; 19:5574–5581.

    PubMed  CAS  Google Scholar 

  106. Werner H, Leroith D. The role of the insulin-like growth factor system in human cancer [Review]. Adv Cancer Res 1996; 68:183–223.

    PubMed  CAS  Google Scholar 

  107. Furstenberger G, Senn HJ. Insulin-like growth factors and cancer [Review]. Lancet Oncol 2002; 3:298–302.

    PubMed  CAS  Google Scholar 

  108. Grimberg A, Cohen P. Role of insulin-like growth factors and their binding proteins in growth control and carcinogenesis [Review]. J Cell Physiol 2000; 183:1–9.

    PubMed  CAS  Google Scholar 

  109. Yu H, Rohan T. Role of the insulin-like growth factor family in cancer development and progression. J Natl Cancer Inst 2000; 92:1472–1489.

    PubMed  CAS  Google Scholar 

  110. Kalebic T, Blakesley V, Slade C, Plasschaert S, Leroith D, Helman LJ. Expression of a kinase-deficient IGF-I-R suppresses tumorigenicity of rhabdomyosarcoma cells constitutively expressing a wild type IGF-I-R. Int J Cancer 1998; 76:223–227.

    PubMed  CAS  Google Scholar 

  111. Reiss K, D’Ambrosio C, Tu X, Tu C, Baserga R. Inhibition of tumor growth by a dominant negative mutant of the insulin-like growth factor I receptor with a bystander effect. Clin Cancer Res 1998; 4:2647–2655.

    PubMed  CAS  Google Scholar 

  112. Scotlandi K, Benini S, Nanni P, et al. Blockage of insulin-like growth factor-I receptor inhibits the growth of Ewing’s sarcoma in athymic mice. Cancer Res 1998; 58:4127–4131.

    PubMed  CAS  Google Scholar 

  113. Scotlandi K, Avnet S, Benini S, et al. Expression of an IGF-I receptor dominant negative mutant induces apoptosis, inhibits tumorigenesis and enhances chemosensitivity in Ewing’s sarcoma cells. Int J Cancer 2002; 101:11–16.

    PubMed  CAS  Google Scholar 

  114. Scotlandi K, Maini C, Manara MC, et al. Effectiveness of insulin-like growth factor I receptor antisense strategy against Ewing’s sarcoma cells. Cancer Gene Ther 2002; 9:296–307.

    PubMed  CAS  Google Scholar 

  115. Burtrum D, Zhu Z, Lu D, et al. A fully human monoclonal antibody to the insulin-like growth factor I receptor blocks ligand-dependent signaling and inhibits human tumor growth in vivo. Cancer Res 2003; 63:8912–8921.

    PubMed  CAS  Google Scholar 

  116. Garcia-Echeverria C, Pearson MA, Marti A, et al. In vivo antitumor activity of NVP-AEW541—A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell 2004; 5(3):231–239.

    PubMed  CAS  Google Scholar 

  117. Mitsiades CS, Mitsiades NS, McMullan CJ, et al. Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell 2004; 5(3):221–230.

    PubMed  CAS  Google Scholar 

  118. Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more [Review]. Nat Rev Mol Cell Biol 2003; 4:915–925.

    PubMed  CAS  Google Scholar 

  119. Birchmeier W. Signaling by HGF/SF and the receptor tyrosine kinase Met. Eur J Biochem 2003; 270:Suppl 1:8–9.

    Google Scholar 

  120. Longati P, Comoglio PM, Bardelli A. Receptor tyrosine kinases as therapeutic targets: the model of the Met oncogene. Curr Drug Targets 2001; 2:41–55.

    PubMed  CAS  Google Scholar 

  121. Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the Met proto-oncogene in papillary renal carcinomas. Nat Gen 1997; 16:68–73.

    CAS  Google Scholar 

  122. Lorenzato A, Olivero M, Patane S, et al. Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res 2002; 62:7025–7030.

    PubMed  CAS  Google Scholar 

  123. Lee JH, Han SU, Cho H, et al. A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene 2000; 19:4947–4953.

    PubMed  CAS  Google Scholar 

  124. Yamashita J, Ogawa M, Yamashita S, et al. Immunoreactive hepatocyte growth factor is a strong and independent predictor of recurrence and survival in human breast cancer. Cancer Res 1994; 54:1630–1633.

    PubMed  CAS  Google Scholar 

  125. Abounader R, Ranganathan S, Lal B, et al. Reversion of human glioblastoma malignancy by U1 small nuclear RNA/ribozyme targeting of scatter factor/hepatocyte growth factor and c-met expression. J Natl Cancer Inst 1999; 91:1548–1556.

    PubMed  CAS  Google Scholar 

  126. Herynk MH, Stoeltzing O, Reinmuth N, et al. Down-regulation of c-met inhibits growth in the liver of human colorectal carcinoma cells. Cancer Res 2003; 63:2990–2996.

    PubMed  CAS  Google Scholar 

  127. Jiang WG, Grimshaw D, Lane J, et al. A hammerhead ribozyme suppresses expression of hepatocyte growth factor/scatter factor receptor c-MET and reduces migration and invasiveness of breast cancer cells. Clin Cancer Res 2001; 7:2555–2562.

    PubMed  CAS  Google Scholar 

  128. Jiang WG, Grimshaw D, Martin TA, et al. Reduction of stromal fibroblast-induced mammary tumor growth, by retroviral ribozyme transgenes to hepatocyte growth factor/scatter factor and its receptor, c-MET. Clin Cancer Res 2003; 9:4274–4281.

    PubMed  CAS  Google Scholar 

  129. Furge KA, Kiewlich D, Le P, et al. Suppression of Ras-mediated tumorigenicity and metastasis through inhibition of the Met receptor tyrosine kinase. Proc Natl Acad Sci USA 2001; 98:10722–10727.

    PubMed  CAS  Google Scholar 

  130. Maemondo M, Narumi K, Saijo Y, et al. Targeting angiogenesis and HGF function using an adenoviral vector expressing the HGF antagonist NK4 for cancer therapy. Mol Ther 2002; 5:177–185.

    PubMed  CAS  Google Scholar 

  131. Matsumoto, and Nakamura T. Suppression of tumor malignancy by NK4/malignostatin: a new cancer therapy by inhibition of tumor invasion-metastasis and angiogenesis. Saishin Igaku 2000; 55:1960–1968.

    CAS  Google Scholar 

  132. Morotti A, Mila S, Accornero P, Tagliabue E, Ponzetto C. K252a inhibits the oncogenic properties of Met, the HGF receptor. Oncogene 2002; 21:4885–4893.

    PubMed  CAS  Google Scholar 

  133. Christensen JG, Schreck R, Burrows J, et al. A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res 2003; 63:7345–7355.

    PubMed  CAS  Google Scholar 

  134. Sattler M, Pride YB, Ma P, et al. A novel small molecule Met inhibitor induces apoptosis in cells transformed by the oncogenic Tpr-Met Tyrosine kinase. Cancer Res 2003; 63:5462–5469.

    PubMed  CAS  Google Scholar 

  135. Wang X, Le P, Liang C, et al. Potent and selective inhibitors of the Met [hepatocyte growth factor/scatter factor (HGF/SF) receptor] tyrosine kinase block HGF/SF-induced tumor cell growth and invasion. Mol Cancer Ther 2003; 2(11):1085–1092.

    PubMed  CAS  Google Scholar 

  136. Liu WM, Stimson LA, Joel SP. The in vitro activity of the tyrosine kinase inhibitor STI571 in BCR-ABL positive chronic myeloid leukaemia cells: synergistic interactions with anti-leukaemic agents. Br J Cancer 2002; 86:1472–1478.

    PubMed  CAS  Google Scholar 

  137. Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system [Review]. Nat Rev Immunol 2003; 3:900–911.

    PubMed  CAS  Google Scholar 

  138. Igaz A, Toth S, Falus A. Biological and clinical significance of the JAK-STAT pathway; lessons from knockout mice [Review]. Inflamm Res 2001; 50:435–441.

    PubMed  CAS  Google Scholar 

  139. Ihle JN, Stravapodis D, Parganas E, et al. The roles of Jaks and Stats in cytokine signaling. [Review] [49 refs]. Cancer J Sci Am 1998; 4:Suppl 91.

    Google Scholar 

  140. Macchi P, Villa A, Giliani S, et al. J. Mutations of Jak-3 gene in patients with autosomal severe combined immune deficiency (SCID). Nature 1995; 377:65–68.

    PubMed  CAS  Google Scholar 

  141. Notarangelo LD, Mella P, Jones A, et al. Mutations in severe combined immune deficiency (SCID) due to JAK3 deficiency [Review]. Hum Mutat 2001; 18:255–263.

    PubMed  CAS  Google Scholar 

  142. Russell SM, Tayebi N, Nakajima H, et al. Mutation of Jak3 in a patient with SCID: essential role of Jak3 in lymphoid development. Science 1995; 270:797–800.

    PubMed  CAS  Google Scholar 

  143. Tsuge I, Matsuoka H, Abe T, Kamachi Y, Torii S. Interleukin-2 receptor gamma-chain mutations in severe combined immunodeficiency with B-lymphocytes. Eur J Pediatr 1996; 155:1018–1024.

    PubMed  CAS  Google Scholar 

  144. Changelian PS, Flanagan ME, Ball DJ, et al. Prevention of organ allograft rejection by a specific Janus kinase 3 inhibitor. Science 2003; 302:875–878.

    PubMed  CAS  Google Scholar 

  145. Cetkovic-Cvrlje M, Dragt AL, Vassilev A, Liu XP, Uckun FM. Targeting JAK3 with JANEX-1 for prevention of autoimmune type 1 diabetes in NOD mice. Clin Immunol 2003; 106:213–225.

    PubMed  CAS  Google Scholar 

  146. Lacronique V, Boureux A, Valle VD, et al. A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia. Science 1997; 278:1309–1312.

    PubMed  CAS  Google Scholar 

  147. Ruchatz H, Coluccia AM, Stano P, Marchesi E, Gambacorti-Passerini C. Constitutive activation of Jak2 contributes to proliferation and resistance to apoptosis in NPM/ALK-transformed cells. Exp Hematol 2003; 31:309–315.

    PubMed  CAS  Google Scholar 

  148. Sun X, Layton JE, Elefanty A, Lieschke GJ. Comparison of effects of the tyrosine kinase inhibitors AG957, AG490, and STI571 on BCR-ABL-expressing cells, demonstrating synergy between AG490 and STI571. Blood 2001; 97:2008–2015.

    PubMed  CAS  Google Scholar 

  149. Fukushima N, Sato N, Sahin F, Su GH, Hruban RH, Goggins M. Aberrant methylation of suppressor of cytokine signalling-1 (SOCS-1) gene in pancreatic ductal neoplasms. Br J Cancer 2003; 89:338–343.

    PubMed  CAS  Google Scholar 

  150. Yoshikawa H, Matsubara K, Qian GS, et al. SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity [see comment]. Nat Gen 2001; 28:29–35.

    CAS  Google Scholar 

  151. Burke WM, Jin X, Lin HJ, et al. Inhibition of constitutively active Stat3 suppresses growth of human ovarian and breast cancer cells. Oncogene 2001; 20:7925–7934.

    PubMed  CAS  Google Scholar 

  152. Li L, Shaw PE. Autocrine-mediated activation of STAT3 correlates with cell proliferation in breast carcinoma lines. J Biol Chem 2002; 277:17397–17405.

    PubMed  CAS  Google Scholar 

  153. Sriuranpong V, Park JI, Amornphimoltham P, et al. Epidermal growth factor receptor-independent constitutive activation of STAT3 in head and neck squamous cell carcinoma is mediated by the autocrine/paracrine stimulation of the interleukin 6/gp130 cytokine system. Cancer Res 2003; 63:2948–2956.

    PubMed  CAS  Google Scholar 

  154. Toyonaga T, Nakano K, Nagano M, et al. Blockade of constitutively activated Janus kinase/signal transducer and activator of transcription-3 pathway inhibits growth of human pancreatic cancer. Cancer Lett 2003; 201:107–116.

    PubMed  CAS  Google Scholar 

  155. Uckun FM, Sudbeck EA, Mao C, et al. Structure-based design of novel anticancer agents [Review]. Curr Cancer Drug Targets 2001; 1:59–71.

    PubMed  CAS  Google Scholar 

  156. Hunt JL. Molecular mutations in thyroid carcinogenesis [Review]. Am J Clin Pathol 2002; 118:Suppl 27.

    Google Scholar 

  157. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer [see comment]. Nature 2002; 417:949–954.

    PubMed  CAS  Google Scholar 

  158. DiGiovanna MP, Chu P, Davison TL, et al. active signaling by HER-2/neu in a subpopulation of HER-2/neu-overexpressing ductal carcinoma in situ: clinicopathological correlates. Cancer Res 2002; 62:6667–6673.

    PubMed  CAS  Google Scholar 

  159. Thor AD, Liu S, Edgerton S, et al. Activation (tyrosine phosphorylation) of ErbB-2 (HER-2/neu): a study of incidence and correlation with outcome in breast cancer. J Clin Oncol 2000; 18:3230–3239.

    PubMed  CAS  Google Scholar 

  160. She QB, Solit D, Basso A, Moasser MM. Resistance to Gefitinib in PTEN-null HER-overexpressing tumor cells can be overcome through restoration of PTEN function or pharmacologic modulation of constitutive phosphatidylinositol 3′-Kinase/Akt pathway signaling. Clin Cancer Res 2003; 9:4340–4346.

    PubMed  CAS  Google Scholar 

  161. Sawyers CL. Opportunities and challenges in the development of kinase inhibitor therapy for cancer [Review]. Genes Dev 2003; 17:2998–3010.

    PubMed  CAS  Google Scholar 

  162. Shah NP, Nicoll JM, Nagar B, et al. L.Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia [comment]. Cancer Cell 2002; 2:117–125.

    PubMed  CAS  Google Scholar 

  163. Shah NP, Sawyers CL. Mechanisms of resistance to STI571 in Philadelphia chromosome-associated leukemias [Review]. Oncogene 2003; 22:7389–7395.

    PubMed  CAS  Google Scholar 

  164. Nagar B, Hantschel O, Young MA, et al. Structural basis for the autoinhibition of c-Abl tyrosine kinase [see comment]. Cell 2003; 112:859–871.

    PubMed  CAS  Google Scholar 

  165. Schindler T, Bornmann W, Pellicena P, et al. Structural mechanism for STI-571 inhibition of Abelson tyrosine kinase. Science 2000; 289:1938–1942.

    PubMed  CAS  Google Scholar 

  166. Azam M, Latek RR, Daley GQ. Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL [see comment]. Cell 2003; 112:831–843.

    PubMed  CAS  Google Scholar 

  167. Cools J, Stover EH, Boulton CL, et al. PKC412 overcomes resistance to imatinib in a murine model of FIP1L1-PDGFRalpha-induced myeloproliferative disease [see comment]. Cancer Cell 2003; 3:459–469.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2006 Humana Press Inc., Totowa, NJ

About this chapter

Cite this chapter

Pearson, M., García-Echeverría, C., Fabbro, D. (2006). Protein Tyrosine Kinases as Targets for Cancer and Other Indications. In: Fabbro, D., McCormick, F. (eds) Protein Tyrosine Kinases. Cancer Drug Discovery and Development. Humana Press. https://doi.org/10.1385/1-59259-962-1:001

Download citation

Publish with us

Policies and ethics