Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-xm8r8 Total loading time: 0 Render date: 2024-06-25T09:04:30.210Z Has data issue: false hasContentIssue false

82 - Tyrosine-kinase inhibitors in oncology

from Part 4 - Pharmacologic targeting of oncogenic pathways

Published online by Cambridge University Press:  05 February 2015

Anne S. Tsao
Affiliation:
University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Vassiliki Papadimitrakopoulou
Affiliation:
University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
Roy S. Herbst
Affiliation:
Yale University School of Medicine, New Haven, CT
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

In the past decade, delivering personalized medicine via molecularly targeted therapies has become a major focus in the field of cancer therapeutics. Tyrosine kinases regulate angiogenesis and cell proliferation, invasion, and apoptosis. Tyrosine-kinase inhibitors (TKIs) are small-molecule inhibitors that permeate through the cell membrane and target specific portions of kinase receptors in cancer cells and/or the surrounding endothelium and vasculature. In this chapter, we review the TKIs currently used to treat cancer, including targeted agents, angiogenesis inhibitors, and Her family inhibitors (Figure 82.1).

Imatinib mesylate

Imatinib mesylate (STI-571; Gleevec/Glivec, Novartis Pharmaceuticals), the first TKI developed for Philadelphia-chromosome-positive (Ph+) chronic myeloid leukemia (CML), specifically targets the translocation that encodes the breakpoint cluster region–Abelson (BCR–ABL) tyrosine kinase (Figure 82.2). Imatinib also inhibits normal ABL. The BCR–ABL fusion gene is found in 90% of patients with CML and 15–30% of patients with acute lymphoblastic leukemia (ALL; 1). BCR–ABL activates multiple cytoplasmic and nuclear signal-transduction pathways, including Ras, phosphatidylinositol-3 kinase (PI3K), protein kinase B (AKT), and Jak/Stat, and up-regulates interleukin-3 and focal adhesion kinase. BCR–ABL is associated with an impaired DNA-repair response that promotes genetic abnormalities (2–10). In addition, imatinib inhibits c-Kit receptor and platelet-derived growth-factor receptor (PDGFR)-α and -β.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 872 - 883
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Rowley, JD. Letter: a new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973;243:290–3.CrossRef
Chai, SK, Nichols, GL, Rothman, P.Constitutive activation of JAKs and STATs in BCR-Abl-expressing cell lines and peripheral blood cells derived from leukemic patients. Journal of Immunology 1997;159:4720–8.Google ScholarPubMed
Cheng, K, Kurzrock, R, Qiu, X, et al. Reduced focal adhesion kinase and paxillin phosphorylation in BCR-ABL-transfected cells. Cancer 2002;95:440–50.CrossRef
Deutsch, E, Dugray, A, AbdulKarim, B, et al. BCR-ABL down-regulates the DNA repair protein DNA-PKcs. Blood 2001; 97:2084–90.CrossRef
Gotoh, A, Miyazawa, K, Ohyashiki, K, et al. Tyrosine phosphorylation and activation of focal adhesion kinase (p125FAK) by BCR-ABL oncoprotein. Experimental Hematology 1995;23:1153–9.
Ilaria, RL, Van Etten, RA.P210 and P190(BCR/ABL) induce the tyrosine phosphorylation and DNA binding activity of multiple specific STAT family members. Journal of Biological Chemistry 1996; 271:31 704–10.CrossRefGoogle ScholarPubMed
Jiang, X, Lopez, A, Holyoake, T, et al. Autocrine production and action of IL-3 and granulocyte colony-stimulating factor in chronic myeloid leukemia. Proceedings of the National Academy of Sciences USA 1999;96:12 804–9.
Skorski, T, Kanakaraj, P, Nieborowska-Skorska, M, et al. Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood 1995;86:726–36.
Slupianek, A, Schmutte, C, Tombline, G, et al. BCR/ABL regulates mammalian RecA homologs, resulting in drug resistance. Molecular Cell 2001;8:795–806.CrossRef
Tauchi, T, Okabe, S, Miyazawa, K, et al. The tetramerization domain-independent Ras activation by BCR-ABL oncoprotein in hematopoietic cells. International Journal of Oncology 1998;12:1269–76.Google ScholarPubMed
Cohen, MH, Johnson, JR, Pazdur, R. U.S. Food and Drug Administration Drug Approval Summary: conversion of imatinib mesylate (STI571; Gleevec) tablets from accelerated approval to full approval. Clinical Cancer Research 2005;11:12–19.
Nowell, PC, Hungerford, DA.Chromosome studies on normal and leukemic human leukocytes. Journal of the National Cancer Institute 1960;25:85–109.Google ScholarPubMed
Kantarjian, H, Sawyers, C, Hochhaus, A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. New England Journal of Medicine 2002;346:645–52.CrossRefGoogle ScholarPubMed
O’Brien, SG, Guilhot, F, Larson, RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. New England Journal of Medicine 2003;348:994–1004.CrossRefGoogle ScholarPubMed
Druker, BJ, Sawyers, CL, Kantarjian, H, 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. New England Journal of Medicine 2001;344:1038–42.CrossRefGoogle ScholarPubMed
Hochhaus, A, Druker, B, Sawyers, C, et al. Favorable long-term follow-up results over 6 years for response, survival, and safety with imatinib mesylate therapy in chronic-phase chronic myeloid leukemia after failure of interferon-alpha treatment. Blood 2008;111:1039–43.CrossRef
Kantarjian, H, O’Brien, S, Cortes, J, et al. Survival advantage with imatinib mesylate therapy in chronic-phase chronic myelogenous leukemia (CML-CP) after IFN-alpha failure and in late CML-CP, comparison with historical controls. Clinical Cancer Research 2004;10:68–75.CrossRef
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;293:876–80.CrossRef
Branford, S, Rudzki, Z, Walsh, S, et al. High frequency of point mutations clustered within the adenosine triphosphate-binding region of BCR/ABL in patients with chronic myeloid leukemia or Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance. Blood 2002;99:3472–5.CrossRef
Hochhaus, A, Kreil, S, Corbin, AS, et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 2002;16:2190–6.CrossRef
Shah, NP, Nicoll, JM, Nagar, B, et al. 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. Cancer Cell 2002;2:117–25.CrossRef
Branford, S, Rudzki, Z, Walsh, S, et al. Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood 2003;102:276–83.CrossRef
Mahon, FX, Belloc, F, Lagarde, V, et al. MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models. Blood 2003;101:2368–73.CrossRef
Gambacorti-Passerini, CB, Gunby, RH, Piazza, R, et al. Molecular mechanisms of resistance to imatinib in Philadelphia-chromosome-positive leukaemias. Lancet Oncology 2003;4:75–85.CrossRef
Demetri, GD, von Mehren, M, Blanke, CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. New England Journal of Medicine 2002;347:472–80.CrossRefGoogle ScholarPubMed
Heinrich, MC, Corless, CL, Duensing, A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299:708–10.CrossRef
Hirota, S, Isozaki, K, Moriyama, Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science 1998;279:577–80.CrossRef
Verweij, J, Casali, PG, Zalcberg, J, et al. Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet 2004;364:1127–34.CrossRef
Blanke, CD, Rankin, C, Demetri, GD, et al. Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. Journal of Clinical Oncology 2008;26:626–32.CrossRefGoogle ScholarPubMed
Cohen, MH, Farrell, A, Justice, R, et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Oncologist 2009;14:174–80.CrossRef
Shah, NP, Tran, C, Lee, FY, et al. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 2004;305:399–401.CrossRef
Brave, M, Goodman, V, Kaminskas, E, et al. Sprycel for chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia resistant to or intolerant of imatinib mesylate. Clinical Cancer Research 2008;14:352–9.CrossRef
Kantarjian, H, Pasquini, R, Levy, V, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily: two-year follow-up of a randomized phase 2 study (START-R). Cancer 2009;115:4136–47.CrossRef
Kantarjian, H, Pasquini, R, Hamerschlak, N, et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia after failure of first-line imatinib: a randomized phase 2 trial. Blood 2007;109:5143–50.CrossRef
Shah, NP, Kantarjian, HM, Kim, DW, et al. Intermittent target inhibition with dasatinib 100 mg once daily preserves efficacy and improves tolerability in imatinib-resistant and -intolerant chronic-phase chronic myeloid leukemia. Journal of Clinical Oncology 2008;26:3204–12.CrossRefGoogle ScholarPubMed
Aguilera, DG, Tsimberidou, AM.Dasatinib in chronic myeloid leukemia: a review. Journal of Therapeutics and Clinical Risk Management 2009;5:281–9.Google ScholarPubMed
Kantarjian, HM, Giles, F, Gattermann, N, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood 2007;110:3540–6.CrossRef
O’Hare, T, Walters, DK, Stoffregen, EP, et al. In vitro activity of Bcr-Abl inhibitors AMN107 and BMS-354825 against clinically relevant imatinib-resistant Abl kinase domain mutants. Cancer Research 2005;65:4500–5.CrossRef
von Bubnoff, N, Manley, PW, Mestan, J, et al. Bcr-Abl resistance screening predicts a limited spectrum of point mutations to be associated with clinical resistance to the Abl kinase inhibitor nilotinib (AMN107). Blood 2006;108:1328–33.CrossRef
Weisberg, E, Manley, P, Mestan, J, et al. AMN107 (nilotinib): a novel and selective inhibitor of BCR-ABL. British Journal of Cancer 2006;94:1765–9.CrossRefGoogle ScholarPubMed
Weisberg, E, Manley, PW, Breitenstein, W, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005;7:129–41.CrossRef
le Coutre, P, Ottmann, OG, Giles, F, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is active in patients with imatinib-resistant or -intolerant accelerated-phase chronic myelogenous leukemia. Blood 2008;111:1834–9.CrossRef
Duda, DG, Batchelor, TT, Willett, CG, et al. VEGF-targeted cancer therapy strategies: current progress, hurdles and future prospects. Trends in Molecular Medicine 2007;13:223–30.CrossRef
Kiselyov, A, Balakin, KV, Tkachenko, SE. VEGF/VEGFR signalling as a target for inhibiting angiogenesis. Expert Opinion on Investigational Drugs 2007;16:83–107.CrossRef
Grassot, J, Gouy, M, Perriere, G, et al. Origin and molecular evolution of receptor tyrosine kinases with immunoglobulin-like domains. Molecular Biology and Evolution 2006;23:1232–41.CrossRef
Kamba, T, McDonald, DM.Mechanisms of adverse effects of anti-VEGF therapy for cancer. British Journal of Cancer 2007;96:1788–95.CrossRefGoogle ScholarPubMed
Verheul, HM, Pinedo, HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nature Reviews Cancer 2007;7:475–85.CrossRef
Demetri, GD, van Oosterom, AT, Garrett, CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 2006;368:1329–38.CrossRef
Motzer, RJ, Michaelson, MD, Redman, BG, et al. Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology 2006;24:16–24.CrossRefGoogle ScholarPubMed
Motzer, RJ, Rini, BI, Bukowski, RM, et al. Sunitinib in patients with metastatic renal cell carcinoma. Journal of the American Medical Association 2006;295:2516–24.CrossRefGoogle ScholarPubMed
Motzer, RJ, Hutson, TE, Tomczak, P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. New England Journal of Medicine 2007;356:115–24.CrossRefGoogle ScholarPubMed
Wilhelm, S, Carter, C, Lynch, M, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nature Reviews Drug Discovery 2006;5:835–44.CrossRef
Llovet, JM, Ricci, S, Mazzaferro, V, et al. Sorafenib in advanced hepatocellular carcinoma. New England Journal of Medicine 2008;359:378–90.CrossRefGoogle ScholarPubMed
Cheng, AL, Kang, YK, Chen, Z, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a Phase III randomised, double-blind, placebo-controlled trial. Lancet Oncology 2009;10:25–34.CrossRef
Bazley, LA, Gullick, WJ. The epidermal growth factor receptor family. Endocrine-Related Cancer 2005;12:S17–27.
Rusch, V, Klimstra, D, Venkatraman, E, et al. Overexpression of the epidermal growth factor receptor and its ligand transforming growth factor alpha is frequent in resectable non-small cell lung cancer but does not predict tumor progression. Clinical Cancer Research 1997;3:515–22.
Kurie, JM, Shin, HJ, Lee, JS, et al. Increased epidermal growth factor receptor expression in metaplastic bronchial epithelium. Clinical Cancer Research 1996;2:1787–93.
Dittadi, R, Gion, M, Pagan, V, et al. Epidermal growth factor receptor in lung malignancies: comparison between cancer and normal tissue. British Journal of Cancer 1991;64:741–4.CrossRefGoogle ScholarPubMed
Fujino, S, Enokibori, T, Tezuka, N, et al. A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer. European Journal of Cancer 1996;32A:2070–4.CrossRefGoogle Scholar
Rusch, V, Klimstra, D, Linkov, I, et al. Aberrant expression of p53 or the epidermal growth factor receptor is frequent in early bronchial neoplasia and coexpression precedes squamous cell carcinoma development. Cancer Research 1995;55:1365–72.
Salomon, DS, Brandt, R, Ciardiello, F, et al. Epidermal growth factor-related peptides and their receptors in human malignancies. Critical Reviews in Oncology and Hematology 1995;19:183–232.CrossRef
Fernandes, AM, Hamburger, AW, Gerwin, BI.Dominance of ERBB-1 heterodimers in lung epithelial cells overexpressing ERBB-2. Both ERBB-1 and ERBB-2 contribute significantly to tumorigenicity. American Journal of Respiratory Cell and Molecular Biology 1999;21:701–9.CrossRefGoogle ScholarPubMed
Sibilia, M, Fleischmann, A, Behrens, A, et al. The EGF receptor provides an essential survival signal for SOS-dependent skin tumor development. Cell 2000;102:211–20.CrossRef
Rusch, V, Mendelsohn, J, Dmitrovsky, E. The epidermal growth factor receptor and its ligands as therapeutic targets in human tumors. Cytokine and Growth Factor Reviews 1996;7:133–41.CrossRef
Podsypanina, K, Ellenson, LH, Nemes, A, et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proceedings of the National Academy of Sciences USA 1999;96:1563–8.CrossRef
Bellacosa, A, Testa, JR, Staal, SP, et al. A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science 1991;254:274–7.CrossRef
Hutchinson, J, Jin, J, Cardiff, RD, et al. Activation of Akt (protein kinase B) in mammary epithelium provides a critical cell survival signal required for tumor progression. Molecular and Cellular Biology 2001;21:2203–12.CrossRef
Di Cristofano, A, Pesce, B, Cordon-Cardo, C, et al. Pten is essential for embryonic development and tumour suppression. Nature Genetics 1998;19:348–55.CrossRef
Chang, HW, Aoki, M, Fruman, D, et al. Transformation of chicken cells by the gene encoding the catalytic subunit of PI 3-kinase. Science 1997;276:1848–50.CrossRef
Sharma, SV, Bell, DW, Settleman, J, et al. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews Cancer 2007;7:169–81.CrossRef
Wakeling, AE, Guy, SP, Woodburn, JR, et al. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Research 2002;62:5749–54.
Moyer, JD, Barbacci, EG, Iwata, KK, et al. Induction of apoptosis and cell cycle arrest by CP-358,774, an inhibitor of epidermal growth factor receptor tyrosine kinase. Cancer Research 1997;57:4838–48.
Kris, MG, Natale, RB, Herbst, RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. Journal of the American Medical Association 2003;290:2149–58.CrossRefGoogle ScholarPubMed
Kim, ES, Hirsh, V, Mok, T, et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): a randomised Phase III trial. Lancet 2008;372:1809–18.CrossRef
Mok, TS, Wu, YL, Thongprasert, S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. New England Journal of Medicine 2009;361:947–57.CrossRefGoogle ScholarPubMed
Lynch, TJ, Bell, DW, Sordella, R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2004;350:2129–39.CrossRefGoogle ScholarPubMed
Metro, G, Finocchiaro, G, Cappuzzo, F. Anti-cancer therapy with EGFR inhibitors: factors of prognostic and predictive significance. Annals of Oncology 2006;17:842–5.
Shepherd, FA, Rodrigues Pereira, J, Ciuleanu, T, et al. Erlotinib in previously treated non-small-cell lung cancer. New England Journal of Medicine 2005;353:123–32.CrossRefGoogle ScholarPubMed
Fukuoka, M, Yano, S, Giaccone, G, et al. Multi-institutional randomized Phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer (The IDEAL 1 Trial) [corrected]. Journal of Clinical Oncology 2003;21:2237–46.CrossRefGoogle Scholar
Perez-Soler, R, Chachoua, A, Hammond, LA, et al. Determinants of tumor response and survival with erlotinib in patients with non-small-cell lung cancer. Journal of Clinical Oncology 2004;22:3238–47.CrossRefGoogle ScholarPubMed
Miller, VA, Zakowski, M, Riely, GJ, et al. EGFR mutation and copy number, EGFR protein expression and KRAS mutation as predictors of outcome with erlotinib in bronchioloalveolar cell carcinoma (BAC): results of a prospective Phase II trial. ASCO Meeting Abstracts 2006;24:364s.
Kwak, EL, Sordella, R, Bell, DW, et al. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proceedings of the National Academy of Sciences USA 2005;102:7665–70.CrossRef
Greulich, H, Chen, TH, Feng, W, et al. Oncogenic transformation by inhibitor-sensitive and -resistant EGFR mutants. PLoS Medicine 2005;2:e313.
Pao, W, Miller, VA, Politi, KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Medicine 2005;2:e73.
Prudkin, L, Tang, X, Wistuba, II.Germ-line and somatic presentations of the EGFR T790M mutation in lung cancer. Journal of Thoracic Oncology 2009;4:139–41.CrossRefGoogle ScholarPubMed
Shih, JY, Gow, CH, Yang, PC.EGFR mutation conferring primary resistance to gefitinib in non-small-cell lung cancer. New England Journal of Medicine 2005;353:207–8.CrossRefGoogle ScholarPubMed
Massarelli, E, Varella-Garcia, M, Tang, X, et al. KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clinical Cancer Research 2007;13:2890–6.CrossRef
Tsao, M, Zhu, C, Sakurada, A, et al. An analysis of the prognostic and predictive importance of K-ras mutation status in the National Cancer Institute of Canada Clinical Trials Group BR.21 study of erlotinib versus placebo in the treatment of non-small cell lung cancer. ASCO Meeting Abstracts 2006;24:365s.
Mok, T, Wu, Y-L, Thongprasert, S, et al. Phase III, randomized, open-label, first-line study of gefitinib (G) versus carboplatin/paclitaxel (C/P) in clinically selected patients (pts) with advanced non-small-cell lung cancer (NSCLC). Annals of Oncology 2008;19:1–4.
Kobayashi, S, Boggon, TJ, Dayaram, T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2005;352:786–92.CrossRefGoogle ScholarPubMed
Kumar, A, Petri, ET, Halmos, B, et al. Structure and clinical relevance of the epidermal growth factor receptor in human cancer. Journal of Clinical Oncology 2008;26:1742–51.CrossRefGoogle ScholarPubMed
Engelman, JA, Janne, PA. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clinical Cancer Research 2008;14:2895–9.CrossRef
Carter, TA, Wodicka, LM, Shah, NP, et al. Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases. Proceedings of the National Academy of Sciences USA 2005;102:11 011–16.
Maheswaran, S, Sequist, LV, Nagrath, S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. New England Journal of Medicine 2008;359:366–77.CrossRefGoogle ScholarPubMed
Engelman, JA, Zejnullahu, K, Mitsudomi, T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43.CrossRef
Habib, AA, Chun, SJ, Neel, BG, et al. Increased expression of epidermal growth factor receptor induces sequestration of extracellular signal-related kinases and selective attenuation of specific epidermal growth factor-mediated signal transduction pathways. Molecular Cancer Research 2003;1:219–33.
Elkind, NB, Szentpetery, Z, Apati, A, et al. Multidrug transporter ABCG2 prevents tumor cell death induced by the epidermal growth factor receptor inhibitor Iressa (ZD1839, Gefitinib). Cancer Research 2005;65:1770–7.CrossRef
Guilhot, F, Apperley, J, Kim, DW, et al. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase. Blood 2007;109:4143–50.CrossRef
Apperley, JF, Cortes, JE, Kim, DW, et al. Dasatinib in the treatment of chronic myeloid leukemia in accelerated phase after imatinib failure: the START-A trial. Journal of Clinical Oncology 2009;27:3472–9.CrossRefGoogle Scholar
Cortes, J, Kim, DW, Raffoux, E, et al. Efficacy and safety of dasatinib in imatinib-resistant or -intolerant patients with chronic myeloid leukemia in blast phase. Leukemia 2008;22:2176–83.CrossRef
Cortes, J, Rousselot, P, Kim, DW, et al. Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in blast crisis. Blood 2007;109:3207–13.CrossRef
Hochhaus, A. Dasatinib for the treatment of Philadelphia chromosome-positive chronic myelogenous leukaemia after imatinib failure. Expert Opinion in Pharmacotherapy 2007;8:3257–64.CrossRef
Thatcher, N, Chang, A, Parikh, P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer). Lancet 2005;366:1527–37.CrossRef
Cappuzzo, FCB, Wierzbicki, R. Efficacy and safety of erlotinib as first-line maintenance in NSCLC following non-progression with chemotherapy: results from the Phase III SATURN study. 2009 IASLC World Conference on Lung Cancer, San Francisco, CA; 2009.

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×