Skip to main content
Top
Published in: Molecular Cancer 1/2018

Open Access 01-12-2018 | Review

Kinase-targeted cancer therapies: progress, challenges and future directions

Authors: Khushwant S. Bhullar, Naiara Orrego Lagarón, Eileen M. McGowan, Indu Parmar, Amitabh Jha, Basil P. Hubbard, H. P. Vasantha Rupasinghe

Published in: Molecular Cancer | Issue 1/2018

Login to get access

Abstract

The human genome encodes 538 protein kinases that transfer a γ-phosphate group from ATP to serine, threonine, or tyrosine residues. Many of these kinases are associated with human cancer initiation and progression. The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Significantly, protein kinases are the second most targeted group of drug targets, after the G-protein-coupled receptors. Since the development of the first protein kinase inhibitor, in the early 1980s, 37 kinase inhibitors have received FDA approval for treatment of malignancies such as breast and lung cancer. Furthermore, about 150 kinase-targeted drugs are in clinical phase trials, and many kinase-specific inhibitors are in the preclinical stage of drug development. Nevertheless, many factors confound the clinical efficacy of these molecules. Specific tumor genetics, tumor microenvironment, drug resistance, and pharmacogenomics determine how useful a compound will be in the treatment of a given cancer. This review provides an overview of kinase-targeted drug discovery and development in relation to oncology and highlights the challenges and future potential for kinase-targeted cancer therapies.
Literature
1.
go back to reference Coussens L, Parker PJ, Rhee L, Yang-Feng TL, Chen E, Waterfield MD, Francke U, Ullrich A. Multiple, distinct forms of bovine and human protein kinase C suggest diversity in cellular signaling pathways. Science. 1986;233:859–67.PubMedCrossRef Coussens L, Parker PJ, Rhee L, Yang-Feng TL, Chen E, Waterfield MD, Francke U, Ullrich A. Multiple, distinct forms of bovine and human protein kinase C suggest diversity in cellular signaling pathways. Science. 1986;233:859–67.PubMedCrossRef
2.
go back to reference Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–34.PubMedCrossRef Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–34.PubMedCrossRef
4.
go back to reference Köstler WJ, Zielinski CC. Targeting Receptor Tyrosine Kinases in Cancer. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease. New York: Spring; 2015. p. 225–78. Köstler WJ, Zielinski CC. Targeting Receptor Tyrosine Kinases in Cancer. In Receptor Tyrosine Kinases: Structure, Functions and Role in Human Disease. New York: Spring; 2015. p. 225–78.
5.
go back to reference Maurer G, Tarkowski B, Baccarini M. Raf kinases in cancer-roles and therapeutic opportunities. Oncogene. 2011;30:3477–88.PubMedCrossRef Maurer G, Tarkowski B, Baccarini M. Raf kinases in cancer-roles and therapeutic opportunities. Oncogene. 2011;30:3477–88.PubMedCrossRef
6.
go back to reference Kittler H, Tschand P. Driver mutations in the mitogen-activated protein kinase pathway: the seeds of good and evil. Br J Dermat. 2018;178:26-27. Kittler H, Tschand P. Driver mutations in the mitogen-activated protein kinase pathway: the seeds of good and evil. Br J Dermat. 2018;178:26-27.
7.
go back to reference Bardelli A, Parsons DW, Silliman N, Ptak J, Szabo S, Saha S, Markowitz S, Willson JK, Parmigiani G, Kinzler KW. Mutational analysis of the tyrosine kinome in colorectal cancers. Science. 2003;300:949.PubMedCrossRef Bardelli A, Parsons DW, Silliman N, Ptak J, Szabo S, Saha S, Markowitz S, Willson JK, Parmigiani G, Kinzler KW. Mutational analysis of the tyrosine kinome in colorectal cancers. Science. 2003;300:949.PubMedCrossRef
8.
go back to reference Bartram CR, de Klein A, Hagemeijer A, van Agthoven T, van Kessel AG, Bootsma D, Grosveld G, Ferguson-Smith MA, Davies T, Stone M: Translocation of c-abl oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. 1983. Bartram CR, de Klein A, Hagemeijer A, van Agthoven T, van Kessel AG, Bootsma D, Grosveld G, Ferguson-Smith MA, Davies T, Stone M: Translocation of c-abl oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. 1983.
9.
10.
go back to reference Sato S, Sanjo H, Takeda K, Ninomiya-Tsuji J, Yamamoto M, Kawai T, Matsumoto K, Takeuchi O, Akira S. Essential function for the kinase TAK1 in innate and adaptive immune responses. Nat Immunol. 2005;6:1087–95.PubMedCrossRef Sato S, Sanjo H, Takeda K, Ninomiya-Tsuji J, Yamamoto M, Kawai T, Matsumoto K, Takeuchi O, Akira S. Essential function for the kinase TAK1 in innate and adaptive immune responses. Nat Immunol. 2005;6:1087–95.PubMedCrossRef
11.
go back to reference Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 2005;4:387–98.PubMedCrossRef Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 2005;4:387–98.PubMedCrossRef
12.
13.
go back to reference Tabit CE, Shenouda SM, Holbrook M, Fetterman JL, Kiani S, Frame AA, Kluge MA, Held A, Dohadwala MM, Gokce N. Protein kinase C-β contributes to impaired endothelial insulin signaling in humans with diabetes mellitus. Circulation. 2013;127:86–95.PubMedCrossRef Tabit CE, Shenouda SM, Holbrook M, Fetterman JL, Kiani S, Frame AA, Kluge MA, Held A, Dohadwala MM, Gokce N. Protein kinase C-β contributes to impaired endothelial insulin signaling in humans with diabetes mellitus. Circulation. 2013;127:86–95.PubMedCrossRef
14.
go back to reference Shibuya M, Suzuki Y, Sugita K, Saito I, Sasaki T, Takakura K, Nagata I, Kikuchi H, Takemae T, Hidaka H. Effect of AT877 on cerebral vasospasm after aneurysmal subarachnoid hemorrhage: results of a prospective placebo-controlled double-blind trial. J Neurosurg. 1992;76:571–7.PubMedCrossRef Shibuya M, Suzuki Y, Sugita K, Saito I, Sasaki T, Takakura K, Nagata I, Kikuchi H, Takemae T, Hidaka H. Effect of AT877 on cerebral vasospasm after aneurysmal subarachnoid hemorrhage: results of a prospective placebo-controlled double-blind trial. J Neurosurg. 1992;76:571–7.PubMedCrossRef
16.
go back to reference Davies S, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105.PubMedPubMedCentralCrossRef Davies S, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105.PubMedPubMedCentralCrossRef
17.
go back to reference Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–17.PubMedCrossRef Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–17.PubMedCrossRef
18.
go back to reference Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science. 2004;305:399–401.PubMedCrossRef Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science. 2004;305:399–401.PubMedCrossRef
19.
go back to reference Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cornelius LA, Das J, Doweyko AM. Discovery of N-(2-chloro-6-methyl-phenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin-4-ylamino) thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem. 2004;47:6658–61.PubMedCrossRef Lombardo LJ, Lee FY, Chen P, Norris D, Barrish JC, Behnia K, Castaneda S, Cornelius LA, Das J, Doweyko AM. Discovery of N-(2-chloro-6-methyl-phenyl)-2-(6-(4-(2-hydroxyethyl)-piperazin-1-yl)-2-methylpyrimidin-4-ylamino) thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem. 2004;47:6658–61.PubMedCrossRef
20.
go back to reference Fabian MA, Biggs WH, Treiber DK, Atteridge CE, Azimioara MD, Benedetti MG, Carter TA, Ciceri P, Edeen PT, Floyd M. A small molecule–kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;23:329–36.PubMedCrossRef Fabian MA, Biggs WH, Treiber DK, Atteridge CE, Azimioara MD, Benedetti MG, Carter TA, Ciceri P, Edeen PT, Floyd M. A small molecule–kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;23:329–36.PubMedCrossRef
21.
go back to reference Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–32.PubMedCrossRef Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–32.PubMedCrossRef
22.
go back to reference Shukla S, Robey RW, Bates SE, Ambudkar SV. Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Drug Metab Dispos. 2009;37:359–65.PubMedCrossRef Shukla S, Robey RW, Bates SE, Ambudkar SV. Sunitinib (Sutent, SU11248), a small-molecule receptor tyrosine kinase inhibitor, blocks function of the ATP-binding cassette (ABC) transporters P-glycoprotein (ABCB1) and ABCG2. Drug Metab Dispos. 2009;37:359–65.PubMedCrossRef
23.
go back to reference Kirkland LO, McInnes C. Non-ATP competitive protein kinase inhibitors as anti-tumor therapeutics. Biochem Pharmacol. 2009;77:1561–71.PubMedCrossRef Kirkland LO, McInnes C. Non-ATP competitive protein kinase inhibitors as anti-tumor therapeutics. Biochem Pharmacol. 2009;77:1561–71.PubMedCrossRef
24.
go back to reference Tamaoki T, Nomoto H, Takahashi I, Kato Y, Morimoto M, Tomita F. Staurosporine, a potent inhibitor of phospholipidCa++ dependent protein kinase. Biochem Biophys Res Commun. 1986;135:397–402.PubMedCrossRef Tamaoki T, Nomoto H, Takahashi I, Kato Y, Morimoto M, Tomita F. Staurosporine, a potent inhibitor of phospholipidCa++ dependent protein kinase. Biochem Biophys Res Commun. 1986;135:397–402.PubMedCrossRef
25.
go back to reference Shukla S, Chufan E, Singh S, Skoumbourdis A, Kapoor K, Boxer M, Duveau D, Thomas C, Talele T, Ambudkar S. Elucidation of the structural basis of interaction of the BCR-ABL kinase inhibitor, nilotinib (Tasigna) with the human ABC drug transporter P-glycoprotein. Leukemia. 2014; Shukla S, Chufan E, Singh S, Skoumbourdis A, Kapoor K, Boxer M, Duveau D, Thomas C, Talele T, Ambudkar S. Elucidation of the structural basis of interaction of the BCR-ABL kinase inhibitor, nilotinib (Tasigna) with the human ABC drug transporter P-glycoprotein. Leukemia. 2014;
26.
go back to reference Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au H-J, Berry SR, Krahn M, Price T, Simes RJ. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357:2040–8.PubMedCrossRef Jonker DJ, O'Callaghan CJ, Karapetis CS, Zalcberg JR, Tu D, Au H-J, Berry SR, Krahn M, Price T, Simes RJ. Cetuximab for the treatment of colorectal cancer. N Engl J Med. 2007;357:2040–8.PubMedCrossRef
27.
go back to reference Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51.PubMedCrossRef Hudis CA. Trastuzumab—mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51.PubMedCrossRef
28.
go back to reference Noble ME, Endicott JA, Johnson LN. Protein kinase inhibitors: insights into drug design from structure. Science. 2004;303:1800–5.PubMedCrossRef Noble ME, Endicott JA, Johnson LN. Protein kinase inhibitors: insights into drug design from structure. Science. 2004;303:1800–5.PubMedCrossRef
29.
go back to reference Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554.PubMedCrossRef Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554.PubMedCrossRef
30.
go back to reference Ogino S, Lochhead P, Giovannucci E, Meyerhardt J, Fuchs C, Chan A. Discovery of colorectal cancer PIK3CA mutation as potential predictive biomarker: power and promise of molecular pathological epidemiology. Oncogene. 2014;33:2949–55.PubMedCrossRef Ogino S, Lochhead P, Giovannucci E, Meyerhardt J, Fuchs C, Chan A. Discovery of colorectal cancer PIK3CA mutation as potential predictive biomarker: power and promise of molecular pathological epidemiology. Oncogene. 2014;33:2949–55.PubMedCrossRef
31.
go back to reference Vora SR, Juric D, Kim N, Mino-Kenudson M, Huynh T, Costa C, Lockerman EL, Pollack SF, Liu M, Li X. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26:136–49.PubMedPubMedCentralCrossRef Vora SR, Juric D, Kim N, Mino-Kenudson M, Huynh T, Costa C, Lockerman EL, Pollack SF, Liu M, Li X. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26:136–49.PubMedPubMedCentralCrossRef
32.
go back to reference Levine DA, Bogomolniy F, Yee CJ, Lash A, Barakat RR, Borgen PI, Boyd J. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clin Cancer Res. 2005;11:2875–8.PubMedCrossRef Levine DA, Bogomolniy F, Yee CJ, Lash A, Barakat RR, Borgen PI, Boyd J. Frequent mutation of the PIK3CA gene in ovarian and breast cancers. Clin Cancer Res. 2005;11:2875–8.PubMedCrossRef
33.
go back to reference Oda K, Stokoe D, Taketani Y, McCormick F. High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res. 2005;65:10669–73.PubMedCrossRef Oda K, Stokoe D, Taketani Y, McCormick F. High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res. 2005;65:10669–73.PubMedCrossRef
34.
go back to reference Lee JW, Soung YH, Kim SY, Lee HW, Park WS, Nam SW, Kim SH, Lee JY, Yoo NJ, Lee SH. PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene. 2005;24:1477–80.PubMedCrossRef Lee JW, Soung YH, Kim SY, Lee HW, Park WS, Nam SW, Kim SH, Lee JY, Yoo NJ, Lee SH. PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene. 2005;24:1477–80.PubMedCrossRef
36.
go back to reference Mirza AM, Kohn AD, Roth RA, McMahon M. Oncogenic transformation of cells by a conditionally active form of the protein kinase Akt/PKB. Cell Growth Differ Publ Am Assoc Cancer Res. 2000;11:279–92. Mirza AM, Kohn AD, Roth RA, McMahon M. Oncogenic transformation of cells by a conditionally active form of the protein kinase Akt/PKB. Cell Growth Differ Publ Am Assoc Cancer Res. 2000;11:279–92.
37.
go back to reference Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54.PubMedCrossRef Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–54.PubMedCrossRef
38.
go back to reference Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61.PubMedCrossRef Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61.PubMedCrossRef
39.
go back to reference Steensma DP, Dewald GW, Lasho TL, Powell HL, McClure RF, Levine RL, Gilliland DG, Tefferi A. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both “atypical” myeloproliferative disorders and myelodysplastic syndromes. Blood. 2005;106:1207–9.PubMedPubMedCentralCrossRef Steensma DP, Dewald GW, Lasho TL, Powell HL, McClure RF, Levine RL, Gilliland DG, Tefferi A. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both “atypical” myeloproliferative disorders and myelodysplastic syndromes. Blood. 2005;106:1207–9.PubMedPubMedCentralCrossRef
41.
go back to reference Weihua Z, Tsan R, Huang W-C, Wu Q, Chiu C-H, Fidler IJ, Hung M-C. Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell. 2008;13:385–93.PubMedPubMedCentralCrossRef Weihua Z, Tsan R, Huang W-C, Wu Q, Chiu C-H, Fidler IJ, Hung M-C. Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell. 2008;13:385–93.PubMedPubMedCentralCrossRef
42.
go back to reference Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–500.PubMedCrossRef Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–500.PubMedCrossRef
43.
go back to reference Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II, Fong KM, Lee H, Toyooka S, Shimizu N. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005;97:339–46.PubMedCrossRef Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II, Fong KM, Lee H, Toyooka S, Shimizu N. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005;97:339–46.PubMedCrossRef
44.
go back to reference Chen S-J, Luan J, Zhang H-S, Ruan C-P, Xu X-Y, Li Q-Q, Wang N-H. EGFR-mediated G1/S transition contributes to the multidrug resistance in breast cancer cells. Mol Biol Rep. 2012;39:5465–71.PubMedCrossRef Chen S-J, Luan J, Zhang H-S, Ruan C-P, Xu X-Y, Li Q-Q, Wang N-H. EGFR-mediated G1/S transition contributes to the multidrug resistance in breast cancer cells. Mol Biol Rep. 2012;39:5465–71.PubMedCrossRef
45.
go back to reference Sahin Ö, Fröhlich H, Löbke C, Korf U, Burmester S, Majety M, Mattern J, Schupp I, Chaouiya C, Thieffry D. Modeling ERBB receptor-regulated G1/S transition to find novel targets for de novo trastuzumab resistance. BMC Syst Biol. 2009;3:1.PubMedPubMedCentralCrossRef Sahin Ö, Fröhlich H, Löbke C, Korf U, Burmester S, Majety M, Mattern J, Schupp I, Chaouiya C, Thieffry D. Modeling ERBB receptor-regulated G1/S transition to find novel targets for de novo trastuzumab resistance. BMC Syst Biol. 2009;3:1.PubMedPubMedCentralCrossRef
46.
47.
go back to reference Gritsko TM, Coppola D, Paciga JE, Yang L, Sun M, Shelley SA, Fiorica JV, Nicosia SV, Cheng JQ. Activation and overexpression of centrosome kinase BTAK/aurora-a in human ovarian cancer. Clin Cancer Res. 2003;9:1420–6.PubMed Gritsko TM, Coppola D, Paciga JE, Yang L, Sun M, Shelley SA, Fiorica JV, Nicosia SV, Cheng JQ. Activation and overexpression of centrosome kinase BTAK/aurora-a in human ovarian cancer. Clin Cancer Res. 2003;9:1420–6.PubMed
48.
go back to reference Sen S, Zhou H, White RA. A putative serine/threonine kinase encoding gene BTAK on chromosome 20q13 is amplified and overexpressed in human breast cancer cell lines. Oncogene. 1997;14:2195–200.PubMedCrossRef Sen S, Zhou H, White RA. A putative serine/threonine kinase encoding gene BTAK on chromosome 20q13 is amplified and overexpressed in human breast cancer cell lines. Oncogene. 1997;14:2195–200.PubMedCrossRef
49.
go back to reference Liu Q, Kaneko S, Yang L, Feldman RI, Nicosia SV, Chen J, Cheng JQ. Aurora-a abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem. 2004;279:52175–82.PubMedCrossRef Liu Q, Kaneko S, Yang L, Feldman RI, Nicosia SV, Chen J, Cheng JQ. Aurora-a abrogation of p53 DNA binding and transactivation activity by phosphorylation of serine 215. J Biol Chem. 2004;279:52175–82.PubMedCrossRef
50.
go back to reference Sun C, Chan F, Briassouli P, Linardopoulos S. Aurora kinase inhibition downregulates NF-κB and sensitises tumour cells to chemotherapeutic agents. Biochem Biophys Res Commun. 2007;352:220–5.PubMedCrossRef Sun C, Chan F, Briassouli P, Linardopoulos S. Aurora kinase inhibition downregulates NF-κB and sensitises tumour cells to chemotherapeutic agents. Biochem Biophys Res Commun. 2007;352:220–5.PubMedCrossRef
51.
go back to reference Qi G, Ogawa I, Kudo Y, Miyauchi M, Siriwardena B, Shimamoto F, Tatsuka M, Takata T. Aurora-B expression and its correlation with cell proliferation and metastasis in oral cancer. Virchows Arch. 2007;450:297–302.PubMedCrossRef Qi G, Ogawa I, Kudo Y, Miyauchi M, Siriwardena B, Shimamoto F, Tatsuka M, Takata T. Aurora-B expression and its correlation with cell proliferation and metastasis in oral cancer. Virchows Arch. 2007;450:297–302.PubMedCrossRef
52.
go back to reference Chieffi P, Cozzolino L, Kisslinger A, Libertini S, Staibano S, Mansueto G, De Rosa G, Villacci A, Vitale M, Linardopoulos S. Aurora B expression directly correlates with prostate cancer malignancy and influence prostate cell proliferation. Prostate. 2006;66:326–33.PubMedCrossRef Chieffi P, Cozzolino L, Kisslinger A, Libertini S, Staibano S, Mansueto G, De Rosa G, Villacci A, Vitale M, Linardopoulos S. Aurora B expression directly correlates with prostate cancer malignancy and influence prostate cell proliferation. Prostate. 2006;66:326–33.PubMedCrossRef
53.
go back to reference Khan J, Ezan F, Crémet J-Y, Fautrel A, Gilot D, Lambert M, Benaud C, Troadec M-B, Prigent C. Overexpression of active aurora-C kinase results in cell transformation and tumour formation. PLoS One. 2011;6:e26512.PubMedPubMedCentralCrossRef Khan J, Ezan F, Crémet J-Y, Fautrel A, Gilot D, Lambert M, Benaud C, Troadec M-B, Prigent C. Overexpression of active aurora-C kinase results in cell transformation and tumour formation. PLoS One. 2011;6:e26512.PubMedPubMedCentralCrossRef
54.
go back to reference Keith WM, Kenichi N, Sara W. Recent advances of MEK inhibitors and their clinical progress. Curr Top Med Chem. 2007;7:1364–78.CrossRef Keith WM, Kenichi N, Sara W. Recent advances of MEK inhibitors and their clinical progress. Curr Top Med Chem. 2007;7:1364–78.CrossRef
55.
go back to reference Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5:671–88.PubMedCrossRef Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5:671–88.PubMedCrossRef
56.
go back to reference Hu Y, Fang X, Dunham SM, Prada C, Stachowiak EK, Stachowiak MK. 90-kDa ribosomal S6 kinase is a direct target for the nuclear fibroblast growth factor receptor 1 (FGFR1) ROLE IN FGFR1 SIGNALING. J Biol Chem. 2004;279:29325–35.PubMedCrossRef Hu Y, Fang X, Dunham SM, Prada C, Stachowiak EK, Stachowiak MK. 90-kDa ribosomal S6 kinase is a direct target for the nuclear fibroblast growth factor receptor 1 (FGFR1) ROLE IN FGFR1 SIGNALING. J Biol Chem. 2004;279:29325–35.PubMedCrossRef
57.
go back to reference Kollareddy M, Zheleva D, Dzubak P, Brahmkshatriya PS, Lepsik M, Hajduch M. Aurora kinase inhibitors: progress towards the clinic. Investig New Drugs. 2012;30:2411–32.CrossRef Kollareddy M, Zheleva D, Dzubak P, Brahmkshatriya PS, Lepsik M, Hajduch M. Aurora kinase inhibitors: progress towards the clinic. Investig New Drugs. 2012;30:2411–32.CrossRef
58.
go back to reference Jones DT, Hutter B, Jäger N, Korshunov A, Kool M, Warnatz H-J, Zichner T, Lambert SR, Ryzhova M, Quang DAK. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013;45:927–32.PubMedPubMedCentralCrossRef Jones DT, Hutter B, Jäger N, Korshunov A, Kool M, Warnatz H-J, Zichner T, Lambert SR, Ryzhova M, Quang DAK. Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat Genet. 2013;45:927–32.PubMedPubMedCentralCrossRef
59.
go back to reference Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990;61:203–12.PubMedCrossRef Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990;61:203–12.PubMedCrossRef
60.
go back to reference Ward CW, Lawrence MC, Streltsov VA, Adams TE, McKern NM. The insulin and EGF receptor structures: new insights into ligand-induced receptor activation. Trends Biochem Sci. 2007;32:129–37.PubMedCrossRef Ward CW, Lawrence MC, Streltsov VA, Adams TE, McKern NM. The insulin and EGF receptor structures: new insights into ligand-induced receptor activation. Trends Biochem Sci. 2007;32:129–37.PubMedCrossRef
62.
go back to reference Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–81.PubMedCrossRef Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169–81.PubMedCrossRef
63.
go back to reference Libermann TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H, Whittle N, Waterfield MD, Ullrich A, Schlessinger J: Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. 1985. Libermann TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H, Whittle N, Waterfield MD, Ullrich A, Schlessinger J: Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. 1985.
64.
go back to reference Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707.PubMedCrossRef Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707.PubMedCrossRef
65.
go back to reference Eswarakumar V, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–49.PubMedCrossRef Eswarakumar V, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–49.PubMedCrossRef
66.
go back to reference Brewer MR, Choi SH, Alvarado D, Moravcevic K, Pozzi A, Lemmon MA, Carpenter G. The juxtamembrane region of the EGF receptor functions as an activation domain. Mol Cell. 2009;34:641–51.PubMedCentralCrossRef Brewer MR, Choi SH, Alvarado D, Moravcevic K, Pozzi A, Lemmon MA, Carpenter G. The juxtamembrane region of the EGF receptor functions as an activation domain. Mol Cell. 2009;34:641–51.PubMedCentralCrossRef
67.
68.
go back to reference Castagna M, Takai Y, Kaibuchi K, Sano K, Kikkawa U, Nishizuka Y. Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor-promoting phorbol esters. J Biol Chem. 1982;257:7847–51.PubMed Castagna M, Takai Y, Kaibuchi K, Sano K, Kikkawa U, Nishizuka Y. Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor-promoting phorbol esters. J Biol Chem. 1982;257:7847–51.PubMed
69.
go back to reference Hidaka H, Inagaki M, Kawamoto S, Sasaki Y. Isoquinolinesulfonamides, novel and potent inhibitors of cyclic nucleotide-dependent protein kinase and protein kinase C. Biochemistry. 1984;23:5036–41.PubMedCrossRef Hidaka H, Inagaki M, Kawamoto S, Sasaki Y. Isoquinolinesulfonamides, novel and potent inhibitors of cyclic nucleotide-dependent protein kinase and protein kinase C. Biochemistry. 1984;23:5036–41.PubMedCrossRef
70.
go back to reference Ward NE, O'Brian CA. Kinetic analysis of protein kinase C inhibition by staurosporine: evidence that inhibition entails inhibitor binding at a conserved region of the catalytic domain but not competition with substrates. Mol Pharmacol. 1992;41:387-92. Ward NE, O'Brian CA. Kinetic analysis of protein kinase C inhibition by staurosporine: evidence that inhibition entails inhibitor binding at a conserved region of the catalytic domain but not competition with substrates. Mol Pharmacol. 1992;41:387-92.
71.
go back to reference Knighton DR, Zheng J, Ten Eyck LF, Ashford VA, Xuong N-H, Taylor SS, Sowadski JM. Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science. 1991;253:407–14.PubMedCrossRef Knighton DR, Zheng J, Ten Eyck LF, Ashford VA, Xuong N-H, Taylor SS, Sowadski JM. Crystal structure of the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science. 1991;253:407–14.PubMedCrossRef
72.
go back to reference Zheng J, Knighton DR, Taylor SS, Xuong NH, Sowadski JM, Eyck LF. Crystal structures of the myristylated catalytic subunit of cAMP-dependent protein kinase reveal open and closed conformations. Protein Sci. 1993;2:1559-73. Zheng J, Knighton DR, Taylor SS, Xuong NH, Sowadski JM, Eyck LF. Crystal structures of the myristylated catalytic subunit of cAMP-dependent protein kinase reveal open and closed conformations. Protein Sci. 1993;2:1559-73.
73.
go back to reference Asano T, Ikegaki I, Satoh SI, Seto M, Sasaki Y. A Protein Kinase Inhibitor, Fasudil (AT-877): A Novel Approach to Signal Transduction Therapy. Cardiovasc Drug Rev. 1998;16:76–87. Asano T, Ikegaki I, Satoh SI, Seto M, Sasaki Y. A Protein Kinase Inhibitor, Fasudil (AT-877): A Novel Approach to Signal Transduction Therapy. Cardiovasc Drug Rev. 1998;16:76–87.
74.
go back to reference Liu J, Farmer JD, Lane WS, Friedman J, Weissman I, Schreiber SL. Calcineurin is a common target of cyclophilin-cyclosporin a and FKBP-FK506 complexes. Cell. 1991;66:807–15.PubMedCrossRef Liu J, Farmer JD, Lane WS, Friedman J, Weissman I, Schreiber SL. Calcineurin is a common target of cyclophilin-cyclosporin a and FKBP-FK506 complexes. Cell. 1991;66:807–15.PubMedCrossRef
75.
go back to reference Hopkin MD, Baxendale IR, Ley SV. A flow-based synthesis of Imatinib: the API of Gleevec. Chem Commun. 2010;46:2450–2.CrossRef Hopkin MD, Baxendale IR, Ley SV. A flow-based synthesis of Imatinib: the API of Gleevec. Chem Commun. 2010;46:2450–2.CrossRef
76.
go back to reference Lozzio CB, Lozzio BB. Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood. 1975;45:321–34.PubMed Lozzio CB, Lozzio BB. Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood. 1975;45:321–34.PubMed
77.
go back to reference Schultz KR, Bowman WP, Aledo A, Slayton WB, Sather H, Devidas M, Wang C, Davies SM, Gaynon PS, Trigg M. Improved early event-free survival with imatinib in Philadelphia chromosome–positive acute lymphoblastic leukemia: a children's oncology group study. J Clin Oncol. 2009;27:5175–81.PubMedPubMedCentralCrossRef Schultz KR, Bowman WP, Aledo A, Slayton WB, Sather H, Devidas M, Wang C, Davies SM, Gaynon PS, Trigg M. Improved early event-free survival with imatinib in Philadelphia chromosome–positive acute lymphoblastic leukemia: a children's oncology group study. J Clin Oncol. 2009;27:5175–81.PubMedPubMedCentralCrossRef
78.
go back to reference Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002;1:493–502.PubMedCrossRef Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov. 2002;1:493–502.PubMedCrossRef
79.
go back to reference Steinberg M. Dasatinib: a tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia and philadelphia chromosome—positive acute lymphoblastic leukemia. Clin Ther. 2007;29:2289–308.PubMedCrossRef Steinberg M. Dasatinib: a tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia and philadelphia chromosome—positive acute lymphoblastic leukemia. Clin Ther. 2007;29:2289–308.PubMedCrossRef
80.
go back to reference Kantarjian H, Giles F, Wunderle L, Bhalla K, O'Brien S, Wassmann B, Tanaka C, Manley P, Rae P, Mietlowski W. Nilotinib in imatinib-resistant CML and Philadelphia chromosome–positive ALL. N Engl J Med. 2006;354:2542–51.PubMedCrossRef Kantarjian H, Giles F, Wunderle L, Bhalla K, O'Brien S, Wassmann B, Tanaka C, Manley P, Rae P, Mietlowski W. Nilotinib in imatinib-resistant CML and Philadelphia chromosome–positive ALL. N Engl J Med. 2006;354:2542–51.PubMedCrossRef
81.
go back to reference Motzer RJ, Hoosen S, Bello CL, Christensen JG: Sunitinib malate for the treatment of solid tumours: a review of current clinical data. 2006. Motzer RJ, Hoosen S, Bello CL, Christensen JG: Sunitinib malate for the treatment of solid tumours: a review of current clinical data. 2006.
82.
go back to reference Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, de Oliveira AC, Santoro A, Raoul J-L, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.PubMedCrossRef Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J-F, de Oliveira AC, Santoro A, Raoul J-L, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–90.PubMedCrossRef
83.
go back to reference Sternberg C, Szczylik C, Lee E, Salman P, Mardiak J, Davis I, Pandite L, Chen M, McCann L, Hawkins R. A randomized, double-blind phase III study of pazopanib in treatment-naive and cytokine-pretreated patients with advanced renal cell carcinoma (RCC). In: ASCO annual meeting proceedings; 2009. p. 5021. Sternberg C, Szczylik C, Lee E, Salman P, Mardiak J, Davis I, Pandite L, Chen M, McCann L, Hawkins R. A randomized, double-blind phase III study of pazopanib in treatment-naive and cytokine-pretreated patients with advanced renal cell carcinoma (RCC). In: ASCO annual meeting proceedings; 2009. p. 5021.
84.
go back to reference Pal SK, Azad AA, Bhatia S, Drabkin HA, Costello B, Sarantopoulos J, Kanesvaran R, Lauer R, Starodub AN, Hauke RJ: A phase I/II trial of BNC105P with everolimus in metastatic renal cell carcinoma (mRCC. Clin Cancer Res 2015:clincanres. 3370.2014. Pal SK, Azad AA, Bhatia S, Drabkin HA, Costello B, Sarantopoulos J, Kanesvaran R, Lauer R, Starodub AN, Hauke RJ: A phase I/II trial of BNC105P with everolimus in metastatic renal cell carcinoma (mRCC. Clin Cancer Res 2015:clincanres. 3370.2014.
85.
go back to reference Massarweh S, Romond E, Black EP, Van Meter E, Shelton B, Kadamyan-Melkumian V, Stevens M, Elledge R. Erratum to: a phase II study of combined fulvestrant and everolimus in patients with metastatic estrogen receptor (ER)-positive breast cancer after aromatase inhibitor (AI) failure. Breast Cancer Res Treat. 2015:1–1. Massarweh S, Romond E, Black EP, Van Meter E, Shelton B, Kadamyan-Melkumian V, Stevens M, Elledge R. Erratum to: a phase II study of combined fulvestrant and everolimus in patients with metastatic estrogen receptor (ER)-positive breast cancer after aromatase inhibitor (AI) failure. Breast Cancer Res Treat. 2015:1–1.
86.
go back to reference Abdel-Rahman O, Fouad M. Everolimus-based combination for the treatment of advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): biological rationale and critical review of published data. Tumor Biol. 2015:1–12. Abdel-Rahman O, Fouad M. Everolimus-based combination for the treatment of advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): biological rationale and critical review of published data. Tumor Biol. 2015:1–12.
87.
go back to reference Scholtens A, Foppen MG, Blank C, van Thienen J, van Tinteren H, Haanen J. Vemurafenib for BRAF V600 mutated advanced melanoma: results of treatment beyond progression. Eur J Cancer. 2015; Scholtens A, Foppen MG, Blank C, van Thienen J, van Tinteren H, Haanen J. Vemurafenib for BRAF V600 mutated advanced melanoma: results of treatment beyond progression. Eur J Cancer. 2015;
88.
go back to reference Chougnet CN, Borget I, Leboulleux S, de la Fouchardiere C, Bonichon F, Criniere L, Niccoli P, Bardet S, Schneegans O, Zanetta S. Vandetanib for the treatment of advanced medullary thyroid cancer outside a clinical trial: results from a French cohort. Thyroid. 2015; Chougnet CN, Borget I, Leboulleux S, de la Fouchardiere C, Bonichon F, Criniere L, Niccoli P, Bardet S, Schneegans O, Zanetta S. Vandetanib for the treatment of advanced medullary thyroid cancer outside a clinical trial: results from a French cohort. Thyroid. 2015;
89.
go back to reference Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT: Three-year efficacy, overall survival, and safety of ruxolitinib therapy in patients with myelofibrosis from the COMFORT-I study. Haematologica 2015:haematol. 2014.115840. Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT: Three-year efficacy, overall survival, and safety of ruxolitinib therapy in patients with myelofibrosis from the COMFORT-I study. Haematologica 2015:haematol. 2014.115840.
90.
go back to reference Costa DB, Shaw AT, Ou S-HI, Solomon BJ, Riely GJ, Ahn M-J, Zhou C, Shreeve SM, Selaru P, Polli A: Clinical experience with Crizotinib in patients with advanced ALK-rearranged non–small-cell lung cancer and brain metastases. J Clin Oncol 2015:JCO. 2014.2059. 0539. Costa DB, Shaw AT, Ou S-HI, Solomon BJ, Riely GJ, Ahn M-J, Zhou C, Shreeve SM, Selaru P, Polli A: Clinical experience with Crizotinib in patients with advanced ALK-rearranged non–small-cell lung cancer and brain metastases. J Clin Oncol 2015:JCO. 2014.2059. 0539.
91.
go back to reference Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys P-Y, Awad MM, Yanagitani N, Kim S. The ALK inhibitor ceritinib overcomes crizotinib resistance in non–small cell lung cancer. Cancer Discovery. 2014;4:662–73.PubMedPubMedCentralCrossRef Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys P-Y, Awad MM, Yanagitani N, Kim S. The ALK inhibitor ceritinib overcomes crizotinib resistance in non–small cell lung cancer. Cancer Discovery. 2014;4:662–73.PubMedPubMedCentralCrossRef
92.
go back to reference Shaw AT, Kim D-W, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T. Ceritinib in ALK-rearranged non–small-cell lung cancer. N Engl J Med. 2014;370:1189–97.PubMedPubMedCentralCrossRef Shaw AT, Kim D-W, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T. Ceritinib in ALK-rearranged non–small-cell lung cancer. N Engl J Med. 2014;370:1189–97.PubMedPubMedCentralCrossRef
93.
go back to reference Dar AC, Shokat KM. The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annu Rev Biochem. 2011;80:769–95.PubMedCrossRef Dar AC, Shokat KM. The evolution of protein kinase inhibitors from antagonists to agonists of cellular signaling. Annu Rev Biochem. 2011;80:769–95.PubMedCrossRef
94.
go back to reference Roskoski R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol Res. 2016;103:26–48.PubMedCrossRef Roskoski R. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol Res. 2016;103:26–48.PubMedCrossRef
95.
go back to reference Zuccotto F, Ardini E, Casale E, Angiolini M. Through the “gatekeeper door”: exploiting the active kinase conformation. J Med Chem. 2009;53:2681–94.CrossRef Zuccotto F, Ardini E, Casale E, Angiolini M. Through the “gatekeeper door”: exploiting the active kinase conformation. J Med Chem. 2009;53:2681–94.CrossRef
96.
go back to reference Gavrin LK, Saiah E. Approaches to discover non-ATP site kinase inhibitors. Med Chem Comm. 2013;4:41–51.CrossRef Gavrin LK, Saiah E. Approaches to discover non-ATP site kinase inhibitors. Med Chem Comm. 2013;4:41–51.CrossRef
97.
go back to reference Lamba V, Ghosh I. New directions in targeting protein kinases: focusing upon true allosteric and bivalent inhibitors. Curr Pharm Des. 2012;18:2936–45.PubMedCrossRef Lamba V, Ghosh I. New directions in targeting protein kinases: focusing upon true allosteric and bivalent inhibitors. Curr Pharm Des. 2012;18:2936–45.PubMedCrossRef
98.
go back to reference Liu Y, Gray NS. Rational design of inhibitors that bind to inactive kinase conformations. Nat Chem Biol. 2006;2:358–64.PubMedCrossRef Liu Y, Gray NS. Rational design of inhibitors that bind to inactive kinase conformations. Nat Chem Biol. 2006;2:358–64.PubMedCrossRef
99.
go back to reference Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer. 2009;9:28–39.PubMedCrossRef Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer. 2009;9:28–39.PubMedCrossRef
100.
go back to reference Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, Hocker M, Treiber DK, Zarrinkar PP. Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol. 2011;29:1046–51.PubMedCrossRef Davis MI, Hunt JP, Herrgard S, Ciceri P, Wodicka LM, Pallares G, Hocker M, Treiber DK, Zarrinkar PP. Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol. 2011;29:1046–51.PubMedCrossRef
101.
go back to reference Force T, Kolaja KL. Cardiotoxicity of kinase inhibitors: the prediction and translation of preclinical models to clinical outcomes. Nat Rev Drug Discov. 2011;10:111–26.PubMedCrossRef Force T, Kolaja KL. Cardiotoxicity of kinase inhibitors: the prediction and translation of preclinical models to clinical outcomes. Nat Rev Drug Discov. 2011;10:111–26.PubMedCrossRef
102.
go back to reference Hasinoff BB. The cardiotoxicity and myocyte damage caused by small molecule anticancer tyrosine kinase inhibitors is correlated with lack of target specificity. Toxicol Appl Pharmacol. 2010;244:190–5.PubMedCrossRef Hasinoff BB. The cardiotoxicity and myocyte damage caused by small molecule anticancer tyrosine kinase inhibitors is correlated with lack of target specificity. Toxicol Appl Pharmacol. 2010;244:190–5.PubMedCrossRef
103.
go back to reference Kufareva I, Abagyan R. Type-II kinase inhibitor docking, screening, and profiling using modified structures of active kinase states. J Med Chem. 2008;51:7921–32.PubMedPubMedCentralCrossRef Kufareva I, Abagyan R. Type-II kinase inhibitor docking, screening, and profiling using modified structures of active kinase states. J Med Chem. 2008;51:7921–32.PubMedPubMedCentralCrossRef
105.
go back to reference Berndt N, Karim RM, Schönbrunn E. Advances of small molecule targeting of kinases. Curr Opin Chem Biol. 2017;39:126–32.PubMedCrossRef Berndt N, Karim RM, Schönbrunn E. Advances of small molecule targeting of kinases. Curr Opin Chem Biol. 2017;39:126–32.PubMedCrossRef
106.
go back to reference Eglen R, Reisine T. Drug discovery and the human kinome: recent trends. Pharmacol Ther. 2011;130:144–56.PubMedCrossRef Eglen R, Reisine T. Drug discovery and the human kinome: recent trends. Pharmacol Ther. 2011;130:144–56.PubMedCrossRef
107.
go back to reference Allen LF, Sebolt-Leopold J, Meyer MB. CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK). In Seminars in oncology. Amsterdam: Elsevier; 2003. p. 105–16. Allen LF, Sebolt-Leopold J, Meyer MB. CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK). In Seminars in oncology. Amsterdam: Elsevier; 2003. p. 105–16.
108.
go back to reference Klaeger S, Heinzlmeir S, Wilhelm M, Polzer H, Vick B, Koenig P-A, Reinecke M, Ruprecht B, Petzoldt S, Meng C. The target landscape of clinical kinase drugs. Science. 2017;358:eaan4368.PubMedCrossRef Klaeger S, Heinzlmeir S, Wilhelm M, Polzer H, Vick B, Koenig P-A, Reinecke M, Ruprecht B, Petzoldt S, Meng C. The target landscape of clinical kinase drugs. Science. 2017;358:eaan4368.PubMedCrossRef
109.
go back to reference Adrián FJ, Ding Q, Sim T, Velentza A, Sloan C, Liu Y, Zhang G, Hur W, Ding S, Manley P. Allosteric inhibitors of Bcr-abl–dependent cell proliferation. Nat Chem Biol. 2006;2:95–102.PubMedCrossRef Adrián FJ, Ding Q, Sim T, Velentza A, Sloan C, Liu Y, Zhang G, Hur W, Ding S, Manley P. Allosteric inhibitors of Bcr-abl–dependent cell proliferation. Nat Chem Biol. 2006;2:95–102.PubMedCrossRef
110.
go back to reference Dong Q, Dougan DR, Gong X, Halkowycz P, Jin B, Kanouni T, O’Connell SM, Scorah N, Shi L, Wallace MB. Discovery of TAK-733, a potent and selective MEK allosteric site inhibitor for the treatment of cancer. Bioorg Med Chem Lett. 2011;21:1315–9.PubMedCrossRef Dong Q, Dougan DR, Gong X, Halkowycz P, Jin B, Kanouni T, O’Connell SM, Scorah N, Shi L, Wallace MB. Discovery of TAK-733, a potent and selective MEK allosteric site inhibitor for the treatment of cancer. Bioorg Med Chem Lett. 2011;21:1315–9.PubMedCrossRef
111.
go back to reference Grimsby J, Sarabu R, Corbett WL, Haynes N-E, Bizzarro FT, Coffey JW, Guertin KR, Hilliard DW, Kester RF, Mahaney PE. Allosteric activators of glucokinase: potential role in diabetes therapy. Science. 2003;301:370–3.PubMedCrossRef Grimsby J, Sarabu R, Corbett WL, Haynes N-E, Bizzarro FT, Coffey JW, Guertin KR, Hilliard DW, Kester RF, Mahaney PE. Allosteric activators of glucokinase: potential role in diabetes therapy. Science. 2003;301:370–3.PubMedCrossRef
112.
go back to reference Guertin KR, Grimsby J. Small molecule glucokinase activators as glucose lowering agents: a new paradigm for diabetes therapy. Curr Med Chem. 2006;13:1839–43.PubMedCrossRef Guertin KR, Grimsby J. Small molecule glucokinase activators as glucose lowering agents: a new paradigm for diabetes therapy. Curr Med Chem. 2006;13:1839–43.PubMedCrossRef
113.
go back to reference Blanc J, Geney R, Menet C. Type II kinase inhibitors: an opportunity in cancer for rational design. Anti-Cancer Agents Med Chem (Formerly Current Medicinal Chemistry-Anti-Cancer Agents). 2013;13:731–47.CrossRef Blanc J, Geney R, Menet C. Type II kinase inhibitors: an opportunity in cancer for rational design. Anti-Cancer Agents Med Chem (Formerly Current Medicinal Chemistry-Anti-Cancer Agents). 2013;13:731–47.CrossRef
114.
go back to reference Maekawa T, Ashihara E, Kimura S. The Bcr-Abl tyrosine kinase inhibitor imatinib and promising new agents against Philadelphia chromosome-positive leukemias. Int J Clin Oncol. 2007;12:327–40.PubMedCrossRef Maekawa T, Ashihara E, Kimura S. The Bcr-Abl tyrosine kinase inhibitor imatinib and promising new agents against Philadelphia chromosome-positive leukemias. Int J Clin Oncol. 2007;12:327–40.PubMedCrossRef
115.
go back to reference Gumireddy K, Baker SJ, Cosenza SC, John P, Kang AD, Robell KA, Reddy MR, Reddy EP. A non-ATP-competitive inhibitor of BCR-ABL overrides imatinib resistance. Proc Natl Acad Sci U S A. 2005;102:1992–7.PubMedPubMedCentralCrossRef Gumireddy K, Baker SJ, Cosenza SC, John P, Kang AD, Robell KA, Reddy MR, Reddy EP. A non-ATP-competitive inhibitor of BCR-ABL overrides imatinib resistance. Proc Natl Acad Sci U S A. 2005;102:1992–7.PubMedPubMedCentralCrossRef
116.
go back to reference Cohen MS, Zhang C, Shokat KM, Taunton J. Structural bioinformatics-based design of selective, irreversible kinase inhibitors. Science. 2005;308:1318–21.PubMedPubMedCentralCrossRef Cohen MS, Zhang C, Shokat KM, Taunton J. Structural bioinformatics-based design of selective, irreversible kinase inhibitors. Science. 2005;308:1318–21.PubMedPubMedCentralCrossRef
117.
go back to reference Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci U S A. 2005;102:7665–70.PubMedPubMedCentralCrossRef Kwak EL, Sordella R, Bell DW, Godin-Heymann N, Okimoto RA, Brannigan BW, Harris PL, Driscoll DR, Fidias P, Lynch TJ. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proc Natl Acad Sci U S A. 2005;102:7665–70.PubMedPubMedCentralCrossRef
118.
go back to reference Leproult E, Barluenga S, Moras D, Wurtz J-M, Winssinger N. Cysteine mapping in conformationally distinct kinase nucleotide binding sites: application to the design of selective covalent inhibitors. J Med Chem. 2011;54:1347–55.PubMedCrossRef Leproult E, Barluenga S, Moras D, Wurtz J-M, Winssinger N. Cysteine mapping in conformationally distinct kinase nucleotide binding sites: application to the design of selective covalent inhibitors. J Med Chem. 2011;54:1347–55.PubMedCrossRef
119.
go back to reference Liu Q, Sabnis Y, Zhao Z, Zhang T, Buhrlage SJ, Jones LH, Gray NS. Developing irreversible inhibitors of the protein kinase cysteinome. Chem Biol. 2013;20:146–59.PubMedPubMedCentralCrossRef Liu Q, Sabnis Y, Zhao Z, Zhang T, Buhrlage SJ, Jones LH, Gray NS. Developing irreversible inhibitors of the protein kinase cysteinome. Chem Biol. 2013;20:146–59.PubMedPubMedCentralCrossRef
120.
go back to reference Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10:307–17.PubMedCrossRef Singh J, Petter RC, Baillie TA, Whitty A. The resurgence of covalent drugs. Nat Rev Drug Discov. 2011;10:307–17.PubMedCrossRef
121.
go back to reference Barf T, Kaptein A. Irreversible protein kinase inhibitors: balancing the benefits and risks. J Med Chem. 2012;55:6243–62.PubMedCrossRef Barf T, Kaptein A. Irreversible protein kinase inhibitors: balancing the benefits and risks. J Med Chem. 2012;55:6243–62.PubMedCrossRef
122.
go back to reference Zhao Z, Liu Q, Bliven S, Xie L, Bourne PE. Determining cysteines available for covalent inhibition across the human kinome. J Med Chem. 2017;60:2879–89.PubMedPubMedCentralCrossRef Zhao Z, Liu Q, Bliven S, Xie L, Bourne PE. Determining cysteines available for covalent inhibition across the human kinome. J Med Chem. 2017;60:2879–89.PubMedPubMedCentralCrossRef
123.
go back to reference Liao B-C, Lin C-C, Yang JC-H. Second and third-generation epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer. Curr Opin Oncol. 2015;27:94–101.PubMedCrossRef Liao B-C, Lin C-C, Yang JC-H. Second and third-generation epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer. Curr Opin Oncol. 2015;27:94–101.PubMedCrossRef
124.
go back to reference Wang A, Yan X-E, Wu H, Wang W, Hu C, Chen C, Zhao Z, Zhao P, Li X, Wang L. Ibrutinib targets mutant-EGFR kinase with a distinct binding conformation. Oncotarget. 2016;7:69760.PubMedPubMedCentral Wang A, Yan X-E, Wu H, Wang W, Hu C, Chen C, Zhao Z, Zhao P, Li X, Wang L. Ibrutinib targets mutant-EGFR kinase with a distinct binding conformation. Oncotarget. 2016;7:69760.PubMedPubMedCentral
125.
go back to reference Schwartz PA, Murray BW. Protein kinase biochemistry and drug discovery. Bioorg Chem. 2011;39:192–210.PubMedCrossRef Schwartz PA, Murray BW. Protein kinase biochemistry and drug discovery. Bioorg Chem. 2011;39:192–210.PubMedCrossRef
126.
go back to reference Roskoski R. Ibrutinib inhibition of Bruton protein-tyrosine kinase (BTK) in the treatment of B cell neoplasms. Pharmacol Res. 2016;113:395–408.PubMedCrossRef Roskoski R. Ibrutinib inhibition of Bruton protein-tyrosine kinase (BTK) in the treatment of B cell neoplasms. Pharmacol Res. 2016;113:395–408.PubMedCrossRef
127.
go back to reference Rabindran SK, Discafani CM, Rosfjord EC, Baxter M, Floyd MB, Golas J, Hallett WA, Johnson BD, Nilakantan R, Overbeek E. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Res. 2004;64:3958–65.PubMedCrossRef Rabindran SK, Discafani CM, Rosfjord EC, Baxter M, Floyd MB, Golas J, Hallett WA, Johnson BD, Nilakantan R, Overbeek E. Antitumor activity of HKI-272, an orally active, irreversible inhibitor of the HER-2 tyrosine kinase. Cancer Res. 2004;64:3958–65.PubMedCrossRef
128.
go back to reference Kobayashi S, Ji H, Yuza Y, Meyerson M, Wong K-K, Tenen DG, Halmos B. An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor. Cancer Res. 2005;65:7096–101.PubMedCrossRef Kobayashi S, Ji H, Yuza Y, Meyerson M, Wong K-K, Tenen DG, Halmos B. An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor. Cancer Res. 2005;65:7096–101.PubMedCrossRef
129.
go back to reference Moebitz H. The ABC of protein kinase conformations. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2015;1854:1555–66.CrossRef Moebitz H. The ABC of protein kinase conformations. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2015;1854:1555–66.CrossRef
130.
go back to reference Cohen P. Protein kinases—the major drug targets of the twenty-first century? Nat Rev Drug Discov. 2002;1:309–15.PubMedCrossRef Cohen P. Protein kinases—the major drug targets of the twenty-first century? Nat Rev Drug Discov. 2002;1:309–15.PubMedCrossRef
131.
go back to reference Vieth M, Sutherland JJ, Robertson DH, Campbell RM. Kinomics: characterizing the therapeutically validated kinase space. Drug Discov Today. 2005;10:839–46.PubMedCrossRef Vieth M, Sutherland JJ, Robertson DH, Campbell RM. Kinomics: characterizing the therapeutically validated kinase space. Drug Discov Today. 2005;10:839–46.PubMedCrossRef
132.
go back to reference Chen H, Ma J, Li W, Eliseenkova AV, Xu C, Neubert TA, Miller WT, Mohammadi M. A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell. 2007;27:717–30.PubMedPubMedCentralCrossRef Chen H, Ma J, Li W, Eliseenkova AV, Xu C, Neubert TA, Miller WT, Mohammadi M. A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell. 2007;27:717–30.PubMedPubMedCentralCrossRef
133.
go back to reference Cowan-Jacob SW, Ramage P, Stark W, Fendrich G, Jahnke W. Structural biology of protein tyrosine kinases. In: Protein Tyrosine Kinases. New York: Springer; 2006. p. 187–230. Cowan-Jacob SW, Ramage P, Stark W, Fendrich G, Jahnke W. Structural biology of protein tyrosine kinases. In: Protein Tyrosine Kinases. New York: Springer; 2006. p. 187–230.
134.
go back to reference Nolen B, Taylor S, Ghosh G. Regulation of protein kinases: controlling activity through activation segment conformation. Mol Cell. 2004;15:661–75.PubMedCrossRef Nolen B, Taylor S, Ghosh G. Regulation of protein kinases: controlling activity through activation segment conformation. Mol Cell. 2004;15:661–75.PubMedCrossRef
135.
go back to reference Johnson LN, Noble ME, Owen DJ. Active and inactive protein kinases: structural basis for regulation. Cell. 1996;85:149–58.PubMedCrossRef Johnson LN, Noble ME, Owen DJ. Active and inactive protein kinases: structural basis for regulation. Cell. 1996;85:149–58.PubMedCrossRef
136.
go back to reference Scapin G. Protein kinase inhibition: different approaches to selective inhibitor design. Curr Drug Targets. 2006;7:1443–54.PubMedCrossRef Scapin G. Protein kinase inhibition: different approaches to selective inhibitor design. Curr Drug Targets. 2006;7:1443–54.PubMedCrossRef
137.
go back to reference Angell RM, Angell TD, Bamborough P, Bamford MJ, Chung C-W, Cockerill SG, Flack SS, Jones KL, Laine DI, Longstaff T. Biphenyl amide p38 kinase inhibitors 4: DFG-in and DFG-out binding modes. Bioorg Med Chem Lett. 2008;18:4433–7.PubMedCrossRef Angell RM, Angell TD, Bamborough P, Bamford MJ, Chung C-W, Cockerill SG, Flack SS, Jones KL, Laine DI, Longstaff T. Biphenyl amide p38 kinase inhibitors 4: DFG-in and DFG-out binding modes. Bioorg Med Chem Lett. 2008;18:4433–7.PubMedCrossRef
138.
go back to reference Han S, Mistry A, Chang JS, Cunningham D, Griffor M, Bonnette PC, Wang H, Chrunyk BA, Aspnes GE, Walker DP. Structural characterization of proline-rich tyrosine kinase 2 (PYK2) reveals a unique (DFG-out) conformation and enables inhibitor design. J Biol Chem. 2009;284:13193–201.PubMedPubMedCentralCrossRef Han S, Mistry A, Chang JS, Cunningham D, Griffor M, Bonnette PC, Wang H, Chrunyk BA, Aspnes GE, Walker DP. Structural characterization of proline-rich tyrosine kinase 2 (PYK2) reveals a unique (DFG-out) conformation and enables inhibitor design. J Biol Chem. 2009;284:13193–201.PubMedPubMedCentralCrossRef
139.
go back to reference Traxler P, Bold G, Buchdunger E, Caravatti G, Furet P, Manley P, O'Reilly T, Wood J, Zimmermann J. Tyrosine kinase inhibitors: from rational design to clinical trials. Med Res Rev. 2001;21:499–512.PubMedCrossRef Traxler P, Bold G, Buchdunger E, Caravatti G, Furet P, Manley P, O'Reilly T, Wood J, Zimmermann J. Tyrosine kinase inhibitors: from rational design to clinical trials. Med Res Rev. 2001;21:499–512.PubMedCrossRef
140.
go back to reference Fabbro D, Ruetz S, Buchdunger E, Cowan-Jacob SW, Fendrich G, Liebetanz J, Mestan J, O'Reilly T, Traxler P, Chaudhuri B. Protein kinases as targets for anticancer agents: from inhibitors to useful drugs. Pharmacol Ther. 2002;93:79–98.PubMedCrossRef Fabbro D, Ruetz S, Buchdunger E, Cowan-Jacob SW, Fendrich G, Liebetanz J, Mestan J, O'Reilly T, Traxler P, Chaudhuri B. Protein kinases as targets for anticancer agents: from inhibitors to useful drugs. Pharmacol Ther. 2002;93:79–98.PubMedCrossRef
141.
go back to reference Garuti L, Roberti M, Bottegoni G. Non-ATP competitive protein kinase inhibitors. Curr Med Chem. 2010;17:2804–21.PubMedCrossRef Garuti L, Roberti M, Bottegoni G. Non-ATP competitive protein kinase inhibitors. Curr Med Chem. 2010;17:2804–21.PubMedCrossRef
142.
go back to reference Garuti L, Roberti M, Bottegoni G. Irreversible protein kinase inhibitors. Curr Med Chem. 2011;18:2981–94.PubMedCrossRef Garuti L, Roberti M, Bottegoni G. Irreversible protein kinase inhibitors. Curr Med Chem. 2011;18:2981–94.PubMedCrossRef
143.
go back to reference Pan Z, Scheerens H, Li SJ, Schultz BE, Sprengeler PA, Burrill LC, Mendonca RV, Sweeney MD, Scott KC, Grothaus PG. Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem. 2007;2:58–61.PubMedCrossRef Pan Z, Scheerens H, Li SJ, Schultz BE, Sprengeler PA, Burrill LC, Mendonca RV, Sweeney MD, Scott KC, Grothaus PG. Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem. 2007;2:58–61.PubMedCrossRef
144.
go back to reference Filippakopoulos P, Kofler M, Hantschel O, Gish GD, Grebien F, Salah E, Neudecker P, Kay LE, Turk BE, Superti-Furga G. Structural coupling of SH2-kinase domains links Fes and Abl substrate recognition and kinase activation. Cell. 2008;134:793–803.PubMedPubMedCentralCrossRef Filippakopoulos P, Kofler M, Hantschel O, Gish GD, Grebien F, Salah E, Neudecker P, Kay LE, Turk BE, Superti-Furga G. Structural coupling of SH2-kinase domains links Fes and Abl substrate recognition and kinase activation. Cell. 2008;134:793–803.PubMedPubMedCentralCrossRef
145.
go back to reference Wissner A, Fraser HL, Ingalls CL, Dushin RG, Floyd MB, Cheung K, Nittoli T, Ravi MR, Tan X, Loganzo F. Dual irreversible kinase inhibitors: quinazoline-based inhibitors incorporating two independent reactive centers with each targeting different cysteine residues in the kinase domains of EGFR and VEGFR-2. Bioorg Med Chem. 2007;15:3635–48.PubMedCrossRef Wissner A, Fraser HL, Ingalls CL, Dushin RG, Floyd MB, Cheung K, Nittoli T, Ravi MR, Tan X, Loganzo F. Dual irreversible kinase inhibitors: quinazoline-based inhibitors incorporating two independent reactive centers with each targeting different cysteine residues in the kinase domains of EGFR and VEGFR-2. Bioorg Med Chem. 2007;15:3635–48.PubMedCrossRef
146.
147.
go back to reference Faivre S, Djelloul S, Raymond E. New paradigms in anticancer therapy: targeting multiple signaling pathways with kinase inhibitors. Amsterdam: Seminars in oncology Elsevier; 2006. p. 407–20. Faivre S, Djelloul S, Raymond E. New paradigms in anticancer therapy: targeting multiple signaling pathways with kinase inhibitors. Amsterdam: Seminars in oncology Elsevier; 2006. p. 407–20.
148.
go back to reference García-Echeverría C, Traxler P, Evans DB. ATP site-directed competitive and irreversible inhibitors of protein kinases. Med Res Rev. 2000;20:28–57.PubMedCrossRef García-Echeverría C, Traxler P, Evans DB. ATP site-directed competitive and irreversible inhibitors of protein kinases. Med Res Rev. 2000;20:28–57.PubMedCrossRef
149.
go back to reference Cervantes F, Vannucchi AM, Kiladjian J-J, Al-Ali HK, Sirulnik A, Stalbovskaya V, McQuitty M, Hunter DS, Levy RS, Passamonti F. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122:4047–53.PubMedCrossRef Cervantes F, Vannucchi AM, Kiladjian J-J, Al-Ali HK, Sirulnik A, Stalbovskaya V, McQuitty M, Hunter DS, Levy RS, Passamonti F. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122:4047–53.PubMedCrossRef
150.
go back to reference Walker I, Newell H. Do molecularly targeted agents in oncology have reduced attrition rates? Nat Rev Drug Discov. 2009;8:15–6.PubMedCrossRef Walker I, Newell H. Do molecularly targeted agents in oncology have reduced attrition rates? Nat Rev Drug Discov. 2009;8:15–6.PubMedCrossRef
151.
go back to reference Toniatti C, Jones P, Graham H, Pagliara B, Draetta G. Oncology drug discovery: planning a turnaround. Cancer Discovery. 2014;4:397–404.PubMedCrossRef Toniatti C, Jones P, Graham H, Pagliara B, Draetta G. Oncology drug discovery: planning a turnaround. Cancer Discovery. 2014;4:397–404.PubMedCrossRef
152.
go back to reference Yr L, Zhu W, Jl Z, Jq H, Zhao YZ, Zhang W, Han BH, Yao YH, Jiang LY, Li SQ. The evaluation of efficacy and safety of sunitinib on EGFR-TKI pretreated advanced non-small cell lung cancer patients in China. Clin Respir J. 2014;8:206–12.CrossRef Yr L, Zhu W, Jl Z, Jq H, Zhao YZ, Zhang W, Han BH, Yao YH, Jiang LY, Li SQ. The evaluation of efficacy and safety of sunitinib on EGFR-TKI pretreated advanced non-small cell lung cancer patients in China. Clin Respir J. 2014;8:206–12.CrossRef
153.
go back to reference Takeda M, Nakagawa K. Toxicity profile of epidermal growth factor receptor tyrosine kinase inhibitors in patients with epidermal growth factor receptor gene mutation-positive lung cancer. Mole Clin Oncol. 2017;6:3–6.CrossRef Takeda M, Nakagawa K. Toxicity profile of epidermal growth factor receptor tyrosine kinase inhibitors in patients with epidermal growth factor receptor gene mutation-positive lung cancer. Mole Clin Oncol. 2017;6:3–6.CrossRef
154.
go back to reference Fakih M, Vincent M. Adverse events associated with anti-EGFR therapies for the treatment of metastatic colorectal cancer. Curr Oncol. 2010;17:S18.PubMedPubMedCentral Fakih M, Vincent M. Adverse events associated with anti-EGFR therapies for the treatment of metastatic colorectal cancer. Curr Oncol. 2010;17:S18.PubMedPubMedCentral
156.
go back to reference Hilton JF, Shapiro GI. Aurora kinase inhibition as an anticancer strategy. J Clin Oncol. 2014;32:57–9.PubMedCrossRef Hilton JF, Shapiro GI. Aurora kinase inhibition as an anticancer strategy. J Clin Oncol. 2014;32:57–9.PubMedCrossRef
157.
go back to reference Ahmed K, Unger G, Kren BT, Trembley JH. Targeting CK2 for cancer therapy using a nanomedicine approach. In Protein kinase CK2 cellular function in normal and disease states. New York: Springer; 2015. p. 299–315. Ahmed K, Unger G, Kren BT, Trembley JH. Targeting CK2 for cancer therapy using a nanomedicine approach. In Protein kinase CK2 cellular function in normal and disease states. New York: Springer; 2015. p. 299–315.
158.
go back to reference Gallorini M, Cataldi A, di Giacomo V. Cyclin-dependent kinase modulators and cancer therapy. BioDrugs. 2012;26:377–91.PubMedCrossRef Gallorini M, Cataldi A, di Giacomo V. Cyclin-dependent kinase modulators and cancer therapy. BioDrugs. 2012;26:377–91.PubMedCrossRef
159.
go back to reference Cance WG, Kurenova E, Marlowe T, Golubovskaya V. Disrupting the scaffold to improve focal adhesion kinase–targeted cancer therapeutics. Sci Signal. 2013;6:pe10.PubMedPubMedCentralCrossRef Cance WG, Kurenova E, Marlowe T, Golubovskaya V. Disrupting the scaffold to improve focal adhesion kinase–targeted cancer therapeutics. Sci Signal. 2013;6:pe10.PubMedPubMedCentralCrossRef
160.
go back to reference Fayard E, Xue G, Parcellier A, Bozulic L, Hemmings BA. Protein kinase B (PKB/Akt), a key mediator of the PI3K signaling pathway. In: Phosphoinositide 3-kinase in health and disease. New York: Springer; 2011. p. 31–56. Fayard E, Xue G, Parcellier A, Bozulic L, Hemmings BA. Protein kinase B (PKB/Akt), a key mediator of the PI3K signaling pathway. In: Phosphoinositide 3-kinase in health and disease. New York: Springer; 2011. p. 31–56.
161.
162.
go back to reference WeiSZ L, Efferth T. Polo-like kinase 1 as target for cancer therapy. Exp Hematol Oncol. 2012;1:38.CrossRef WeiSZ L, Efferth T. Polo-like kinase 1 as target for cancer therapy. Exp Hematol Oncol. 2012;1:38.CrossRef
163.
go back to reference Krisenko MO, Geahlen RL. Calling in SYK: SYK's dual role as a tumor promoter and tumor suppressor in cancer. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2015;1853:254–63.CrossRef Krisenko MO, Geahlen RL. Calling in SYK: SYK's dual role as a tumor promoter and tumor suppressor in cancer. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2015;1853:254–63.CrossRef
164.
go back to reference Anagnostopoulou V, Pediaditakis I, Alkahtani S, Alarifi SA, Schmidt E-M, Lang F, Gravanis A, Charalampopoulos I, Stournaras C. Differential effects of dehydroepiandrosterone and testosterone in prostate and colon cancer cell apoptosis: the role of nerve growth factor (NGF) receptors. Endocrinology. 2013;154:2446–56.PubMedCrossRef Anagnostopoulou V, Pediaditakis I, Alkahtani S, Alarifi SA, Schmidt E-M, Lang F, Gravanis A, Charalampopoulos I, Stournaras C. Differential effects of dehydroepiandrosterone and testosterone in prostate and colon cancer cell apoptosis: the role of nerve growth factor (NGF) receptors. Endocrinology. 2013;154:2446–56.PubMedCrossRef
165.
go back to reference Sharma A, Madhunapantula SV, Gowda R, Berg A, Neves RI, Robertson GP. Identification of aurora kinase B and Wee1-like protein kinase as downstream targets of V600E B-RAF in melanoma. Am J Pathol. 2013;182:1151–62.PubMedPubMedCentralCrossRef Sharma A, Madhunapantula SV, Gowda R, Berg A, Neves RI, Robertson GP. Identification of aurora kinase B and Wee1-like protein kinase as downstream targets of V600E B-RAF in melanoma. Am J Pathol. 2013;182:1151–62.PubMedPubMedCentralCrossRef
166.
167.
go back to reference Katayama H, Brinkley WR, Sen S. The aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev. 2003;22:451–64.PubMedCrossRef Katayama H, Brinkley WR, Sen S. The aurora kinases: role in cell transformation and tumorigenesis. Cancer Metastasis Rev. 2003;22:451–64.PubMedCrossRef
168.
go back to reference Knapp S, Sundström M. Recently targeted kinases and their inhibitors—the path to clinical trials. Curr Opin Pharmacol. 2014;17:58–63.PubMedCrossRef Knapp S, Sundström M. Recently targeted kinases and their inhibitors—the path to clinical trials. Curr Opin Pharmacol. 2014;17:58–63.PubMedCrossRef
169.
go back to reference Morgensztern D, Campo MJ, Dahlberg SE, Doebele RC, Garon E, Gerber DE, Goldberg SB, Hammerman PS, Heist RS, Hensing T. Molecularly targeted therapies in non–small-cell lung cancer annual update 2014. J Thorac Oncol. 2015;10:S1–S63.PubMedPubMedCentralCrossRef Morgensztern D, Campo MJ, Dahlberg SE, Doebele RC, Garon E, Gerber DE, Goldberg SB, Hammerman PS, Heist RS, Hensing T. Molecularly targeted therapies in non–small-cell lung cancer annual update 2014. J Thorac Oncol. 2015;10:S1–S63.PubMedPubMedCentralCrossRef
170.
go back to reference Sequist LV, Yang JC-H, Yamamoto N, O'Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai C-M, Boyer M. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:3327–34.PubMedCrossRef Sequist LV, Yang JC-H, Yamamoto N, O'Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai C-M, Boyer M. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:3327–34.PubMedCrossRef
171.
172.
go back to reference Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol. 2014;11:473–81.PubMedCrossRef Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: learning from lung cancer. Nat Rev Clin Oncol. 2014;11:473–81.PubMedCrossRef
175.
go back to reference von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, Suter T, Arahmani A, Rouchet N, Clark E. Adjuvant Pertuzumab and Trastuzumab in early HER2-positive breast cancer. N Engl J Med. 2017; von Minckwitz G, Procter M, de Azambuja E, Zardavas D, Benyunes M, Viale G, Suter T, Arahmani A, Rouchet N, Clark E. Adjuvant Pertuzumab and Trastuzumab in early HER2-positive breast cancer. N Engl J Med. 2017;
176.
go back to reference Hayashi T, Seiler R, Oo HZ, Jäger W, Moskalev I, Awrey S, Dejima T, Todenhöfer T, Li N, Fazli L. Targeting HER2 with T-DM1, an antibody cytotoxic drug conjugate, is effective in HER2 over expressing bladder cancer. J Urol. 2015;194:1120–31.PubMedCrossRef Hayashi T, Seiler R, Oo HZ, Jäger W, Moskalev I, Awrey S, Dejima T, Todenhöfer T, Li N, Fazli L. Targeting HER2 with T-DM1, an antibody cytotoxic drug conjugate, is effective in HER2 over expressing bladder cancer. J Urol. 2015;194:1120–31.PubMedCrossRef
177.
go back to reference Ferrara N, Hillan KJ, Gerber H-P, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391.PubMedCrossRef Ferrara N, Hillan KJ, Gerber H-P, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391.PubMedCrossRef
178.
go back to reference Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Verslype C. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.PubMedCrossRef Cunningham D, Humblet Y, Siena S, Khayat D, Bleiberg H, Santoro A, Bets D, Mueser M, Harstrick A, Verslype C. Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory metastatic colorectal cancer. N Engl J Med. 2004;351:337–45.PubMedCrossRef
179.
go back to reference Sokolowska-Wedzina A, Chodaczek G, Chudzian J, Borek A, Zakrzewska M, Otlewski J: High-affinity internalizing human scFv-fc antibody for targeting FGFR1-overexpressing lung cancer. Mol Cancer Res 2017:molcanres. 0136.2016. Sokolowska-Wedzina A, Chodaczek G, Chudzian J, Borek A, Zakrzewska M, Otlewski J: High-affinity internalizing human scFv-fc antibody for targeting FGFR1-overexpressing lung cancer. Mol Cancer Res 2017:molcanres. 0136.2016.
180.
go back to reference Mohammadi M, Nejatollahi F, Ghasemi Y, Faraji SN. Anti-metastatic and anti-invasion effects of a specific anti-MUC18 scFv antibody on breast cancer cells. Appl Biochem Biotechnol. 2017;181:379–90.PubMedCrossRef Mohammadi M, Nejatollahi F, Ghasemi Y, Faraji SN. Anti-metastatic and anti-invasion effects of a specific anti-MUC18 scFv antibody on breast cancer cells. Appl Biochem Biotechnol. 2017;181:379–90.PubMedCrossRef
181.
go back to reference Sandberg D, Tolmachev V, Velikyan I, Olofsson H, Wennborg A, Feldwisch J, Carlsson J, Lindman H, Sörensen J. Intra-image referencing for simplified assessment of HER2-expression in breast cancer metastases using the Affibody molecule ABY-025 with PET and SPECT. Eur J Nucl Med Mol Imaging. 2017:1–10. Sandberg D, Tolmachev V, Velikyan I, Olofsson H, Wennborg A, Feldwisch J, Carlsson J, Lindman H, Sörensen J. Intra-image referencing for simplified assessment of HER2-expression in breast cancer metastases using the Affibody molecule ABY-025 with PET and SPECT. Eur J Nucl Med Mol Imaging. 2017:1–10.
182.
go back to reference Hu S-z, Shively L, Raubitschek A, Sherman M, Williams LE, Wong JY, Shively JE, Wu AM. Minibody: a novel engineered anti-carcinoembryonic antigen antibody fragment (single-chain Fv-C H 3) which exhibits rapid, high-level targeting of xenografts. Cancer Res. 1996;56:3055–61.PubMed Hu S-z, Shively L, Raubitschek A, Sherman M, Williams LE, Wong JY, Shively JE, Wu AM. Minibody: a novel engineered anti-carcinoembryonic antigen antibody fragment (single-chain Fv-C H 3) which exhibits rapid, high-level targeting of xenografts. Cancer Res. 1996;56:3055–61.PubMed
183.
go back to reference Jafari R, M Zolbanin N, Rafatpanah H, Majidi J, Kazemi T. Fc-fusion proteins in therapy: An updated view. Curr Med Chem. 2017;24:1228–37. Jafari R, M Zolbanin N, Rafatpanah H, Majidi J, Kazemi T. Fc-fusion proteins in therapy: An updated view. Curr Med Chem. 2017;24:1228–37.
184.
go back to reference Mukherjee B, Tomimatsu N, Amancherla K, Camacho CV, Pichamoorthy N, Burma S. The dual PI3K/mTOR inhibitor NVP-BEZ235 is a potent inhibitor of ATM-and DNA-PKCs-mediated DNA damage responses. Neoplasia. 2012;14:34–IN38.PubMedPubMedCentralCrossRef Mukherjee B, Tomimatsu N, Amancherla K, Camacho CV, Pichamoorthy N, Burma S. The dual PI3K/mTOR inhibitor NVP-BEZ235 is a potent inhibitor of ATM-and DNA-PKCs-mediated DNA damage responses. Neoplasia. 2012;14:34–IN38.PubMedPubMedCentralCrossRef
185.
go back to reference Yu P, Laird AD, Du X, Wu J, Won K-A, Yamaguchi K, Hsu PP, Qian F, Jaeger CT, Zhang W. Characterization of the activity of the PI3K/mTOR inhibitor XL765 (SAR245409) in tumor models with diverse genetic alterations affecting the PI3K pathway. Mol Cancer Ther. 2014;13:1078–91.PubMedCrossRef Yu P, Laird AD, Du X, Wu J, Won K-A, Yamaguchi K, Hsu PP, Qian F, Jaeger CT, Zhang W. Characterization of the activity of the PI3K/mTOR inhibitor XL765 (SAR245409) in tumor models with diverse genetic alterations affecting the PI3K pathway. Mol Cancer Ther. 2014;13:1078–91.PubMedCrossRef
186.
go back to reference Powles T, Oudard S, Escudier BJ, Brown JE, Hawkins RE, Castellano DE, Ravaud A, Staehler MD, Rini BI, Lin W: A randomized phase II study of GDC-0980 versus everolimus in metastatic renal cell carcinoma (mRCC) patients (pts) after VEGF-targeted therapy (VEGF-TT). In ASCO annual meeting proceedings 2014: 4525. Powles T, Oudard S, Escudier BJ, Brown JE, Hawkins RE, Castellano DE, Ravaud A, Staehler MD, Rini BI, Lin W: A randomized phase II study of GDC-0980 versus everolimus in metastatic renal cell carcinoma (mRCC) patients (pts) after VEGF-targeted therapy (VEGF-TT). In ASCO annual meeting proceedings 2014: 4525.
187.
go back to reference Dowsett M, Koehler M, Millham R, Borzillo G, A'Hern R, Pierce K, Barton J, Giorgetti C. Phase Ii Randomized Study Of Pre-Operative Pf-04691502 Plus Letrozole Compared With Letrozole (L) In Patients With Estrogen Receptor-Positive, Her2-Negative Early Breast Cancer (BC). In: Annals Of Oncology. Oxford Univ Press Great Clarendon St, Oxford; 2012: 44–44. Dowsett M, Koehler M, Millham R, Borzillo G, A'Hern R, Pierce K, Barton J, Giorgetti C. Phase Ii Randomized Study Of Pre-Operative Pf-04691502 Plus Letrozole Compared With Letrozole (L) In Patients With Estrogen Receptor-Positive, Her2-Negative Early Breast Cancer (BC). In: Annals Of Oncology. Oxford Univ Press Great Clarendon St, Oxford; 2012: 44–44.
188.
go back to reference Bedard PL, Grilley-Olson JE, Cornfeld M, Cartee L, Warwick S, Razak AA, Stayner L-A, Wu Y, Greenwood R, Viana-Gilmartin V. Abstract CT205: a phase I dose-escalation study of trametinib (T) in combination with continuous or intermittent GSK2126458 (GSK458) in patients (pts) with advanced solid tumors. Cancer Res. 2014;74:CT205.CrossRef Bedard PL, Grilley-Olson JE, Cornfeld M, Cartee L, Warwick S, Razak AA, Stayner L-A, Wu Y, Greenwood R, Viana-Gilmartin V. Abstract CT205: a phase I dose-escalation study of trametinib (T) in combination with continuous or intermittent GSK2126458 (GSK458) in patients (pts) with advanced solid tumors. Cancer Res. 2014;74:CT205.CrossRef
189.
go back to reference Haïk S, Marcon G, Mallet A, Tettamanti M, Welaratne A, Giaccone G, Azimi S, Pietrini V, Fabreguettes J-R, Imperiale D. Doxycycline in Creutzfeldt-Jakob disease: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13:150–8.PubMedCrossRef Haïk S, Marcon G, Mallet A, Tettamanti M, Welaratne A, Giaccone G, Azimi S, Pietrini V, Fabreguettes J-R, Imperiale D. Doxycycline in Creutzfeldt-Jakob disease: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13:150–8.PubMedCrossRef
190.
go back to reference Eriksson A, Österroos A, Hassan S, Gullbo J, Rickardson L, Jarvius M, Nygren P, Fryknäs M, Höglund M, Larsson R. Drug screen in patient cells suggests quinacrine to be repositioned for treatment of acute myeloid leukemia. Blood Cancer J. 2015;5:e307.PubMedPubMedCentralCrossRef Eriksson A, Österroos A, Hassan S, Gullbo J, Rickardson L, Jarvius M, Nygren P, Fryknäs M, Höglund M, Larsson R. Drug screen in patient cells suggests quinacrine to be repositioned for treatment of acute myeloid leukemia. Blood Cancer J. 2015;5:e307.PubMedPubMedCentralCrossRef
191.
go back to reference Shapiro GI, Bell-McGuinn KM, Molina JR, Bendell J, Spicer J, Kwak EL, Pandya SS, Millham R, Borzillo G, Pierce KJ. First-in-human study of PF-05212384 (PKI-587), a small-molecule, intravenous, dual inhibitor of PI3K and mTOR in patients with advanced cancer. Clin Cancer Res. 2015. Shapiro GI, Bell-McGuinn KM, Molina JR, Bendell J, Spicer J, Kwak EL, Pandya SS, Millham R, Borzillo G, Pierce KJ. First-in-human study of PF-05212384 (PKI-587), a small-molecule, intravenous, dual inhibitor of PI3K and mTOR in patients with advanced cancer. Clin Cancer Res. 2015.
192.
go back to reference Bauer TM, Patel MR, Infante JR. Targeting PI3 kinase in cancer. Pharmacol Ther. 2015;146:53-60. Bauer TM, Patel MR, Infante JR. Targeting PI3 kinase in cancer. Pharmacol Ther. 2015;146:53-60.
193.
go back to reference Eradat HA, Coutre SE, Barrientos JC, Rai KR, Farber CM, Hillmen P, Sharman JP, Ghia P, Coiffier B, Walewski JA. A phase III, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of idelalisib (GS-1101) in combination with bendamustine and rituximab for previously treated chronic lymphocytic leukemia (CLL). In: ASCO annual meeting proceedings; 2013. p. TPS7133. Eradat HA, Coutre SE, Barrientos JC, Rai KR, Farber CM, Hillmen P, Sharman JP, Ghia P, Coiffier B, Walewski JA. A phase III, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of idelalisib (GS-1101) in combination with bendamustine and rituximab for previously treated chronic lymphocytic leukemia (CLL). In: ASCO annual meeting proceedings; 2013. p. TPS7133.
194.
go back to reference Mikhail S, Albanese C, Pishvaian MJ. Cyclin-dependent kinase inhibitors and the treatment of gastrointestinal cancers. Am J Pathol. 2015;85:1185-97. Mikhail S, Albanese C, Pishvaian MJ. Cyclin-dependent kinase inhibitors and the treatment of gastrointestinal cancers. Am J Pathol. 2015;85:1185-97.
195.
go back to reference Marotta V, Sciammarella C, Vitale M, Colao A, Faggiano A. The evolving field of kinase inhibitors in thyroid cancer. Crit Rev Oncol Hematol. 2015;93:60–73.PubMedCrossRef Marotta V, Sciammarella C, Vitale M, Colao A, Faggiano A. The evolving field of kinase inhibitors in thyroid cancer. Crit Rev Oncol Hematol. 2015;93:60–73.PubMedCrossRef
196.
go back to reference Mohamed A, Krajewski K, Cakar B, Ma CX. Targeted therapy for breast cancer. Am J Pathol. 2013;183:1096–112.PubMedCrossRef Mohamed A, Krajewski K, Cakar B, Ma CX. Targeted therapy for breast cancer. Am J Pathol. 2013;183:1096–112.PubMedCrossRef
197.
go back to reference Marotta V, Franzese MD, Del Prete M, Chiofalo MG, Ramundo V, Esposito R, Marciello F, Pezzullo L, Carratù A, Vitale M. Targeted therapy with kinase inhibitors in aggressive endocrine tumors. Expert Opin Pharmacother. 2013;14:1187–203.PubMedCrossRef Marotta V, Franzese MD, Del Prete M, Chiofalo MG, Ramundo V, Esposito R, Marciello F, Pezzullo L, Carratù A, Vitale M. Targeted therapy with kinase inhibitors in aggressive endocrine tumors. Expert Opin Pharmacother. 2013;14:1187–203.PubMedCrossRef
198.
go back to reference Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367:1694–703.PubMedPubMedCentralCrossRef Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012;367:1694–703.PubMedPubMedCentralCrossRef
199.
go back to reference Yadav V, Chen SH, Yue YG, Buchanan S, Beckmann RP, Peng SB. Co-targeting BRAF and cyclin dependent kinases 4/6 for BRAF mutant cancers. Pharmacol Ther. 2015;149:139-49. Yadav V, Chen SH, Yue YG, Buchanan S, Beckmann RP, Peng SB. Co-targeting BRAF and cyclin dependent kinases 4/6 for BRAF mutant cancers. Pharmacol Ther. 2015;149:139-49.
200.
go back to reference Nicholson R, Gee J, Harper M. EGFR and cancer prognosis. Eur J Cancer. 2001;37:9–15.CrossRef Nicholson R, Gee J, Harper M. EGFR and cancer prognosis. Eur J Cancer. 2001;37:9–15.CrossRef
201.
go back to reference Ménard S, Pupa SM, Campiglio M, Tagliabue E. Biologic and therapeutic role of HER2 in cancer. Oncogene. 2003;22:6570–8.PubMedCrossRef Ménard S, Pupa SM, Campiglio M, Tagliabue E. Biologic and therapeutic role of HER2 in cancer. Oncogene. 2003;22:6570–8.PubMedCrossRef
202.
go back to reference Jeong KJ, Cho KH, Panupinthu N, Kim H, Kang J, Park CG, Mills GB, Lee HY. EGFR mediates LPA-induced proteolytic enzyme expression and ovarian cancer invasion: inhibition by resveratrol. Mol Oncol. 2013;7:121–9.PubMedCrossRef Jeong KJ, Cho KH, Panupinthu N, Kim H, Kang J, Park CG, Mills GB, Lee HY. EGFR mediates LPA-induced proteolytic enzyme expression and ovarian cancer invasion: inhibition by resveratrol. Mol Oncol. 2013;7:121–9.PubMedCrossRef
203.
go back to reference Huang C-Y, Chan C-Y, Chou I-T, Lien C-H, Hung H-C, Lee M-F. Quercetin induces growth arrest through activation of FOXO1 transcription factor in EGFR-overexpressing oral cancer cells. J Nutr Biochem. 2013;24:1596–603.PubMedCrossRef Huang C-Y, Chan C-Y, Chou I-T, Lien C-H, Hung H-C, Lee M-F. Quercetin induces growth arrest through activation of FOXO1 transcription factor in EGFR-overexpressing oral cancer cells. J Nutr Biochem. 2013;24:1596–603.PubMedCrossRef
204.
go back to reference Sun X-D, Liu X-E, Huang D-S. Curcumin induces apoptosis of triple-negative breast cancer cells by inhibition of EGFR expression. Mol Med Rep. 2012;6:1267–70.PubMedCrossRef Sun X-D, Liu X-E, Huang D-S. Curcumin induces apoptosis of triple-negative breast cancer cells by inhibition of EGFR expression. Mol Med Rep. 2012;6:1267–70.PubMedCrossRef
205.
go back to reference Masuda M, Wakasaki T, Toh S, Shimizu M, Adachi S. Chemoprevention of head and neck cancer by green tea extract: EGCG—the role of EGFR signaling and “lipid raft”. J Oncol. 2011;2011. Masuda M, Wakasaki T, Toh S, Shimizu M, Adachi S. Chemoprevention of head and neck cancer by green tea extract: EGCG—the role of EGFR signaling and “lipid raft”. J Oncol. 2011;2011.
206.
go back to reference Thomas T, Venkiteswaran S. Polyphenolic phytochemical curcumin suppresses HER-2 protein phosphorylation and gene expression in breast cancer cells. Cancer Res. 2012;72:5445.CrossRef Thomas T, Venkiteswaran S. Polyphenolic phytochemical curcumin suppresses HER-2 protein phosphorylation and gene expression in breast cancer cells. Cancer Res. 2012;72:5445.CrossRef
207.
go back to reference Boly R, Gras T, Lamkami T, Guissou P, Serteyn D, Kiss R, Dubois J. Quercetin inhibits a large panel of kinases implicated in cancer cell biology. Int J Oncol. 2011;38:833–42.PubMed Boly R, Gras T, Lamkami T, Guissou P, Serteyn D, Kiss R, Dubois J. Quercetin inhibits a large panel of kinases implicated in cancer cell biology. Int J Oncol. 2011;38:833–42.PubMed
208.
go back to reference Khan A, Aljarbou AN, Aldebasi YH, Faisal SM, Khan MA. Resveratrol suppresses the proliferation of breast cancer cells by inhibiting fatty acid synthase signaling pathway. Cancer Epidemiol. 2014;38:765–72.PubMedCrossRef Khan A, Aljarbou AN, Aldebasi YH, Faisal SM, Khan MA. Resveratrol suppresses the proliferation of breast cancer cells by inhibiting fatty acid synthase signaling pathway. Cancer Epidemiol. 2014;38:765–72.PubMedCrossRef
209.
go back to reference Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev. 2014;34:280–300.PubMedCrossRef Katoh M, Nakagama H. FGF receptors: cancer biology and therapeutics. Med Res Rev. 2014;34:280–300.PubMedCrossRef
210.
211.
go back to reference Chadalapaka G, Jutooru I, Safe S. Celastrol decreases specificity proteins (Sp) and fibroblast growth factor receptor-3 (FGFR3) in bladder cancer cells. Carcinogenesis. 2012;33:886–94.PubMedPubMedCentralCrossRef Chadalapaka G, Jutooru I, Safe S. Celastrol decreases specificity proteins (Sp) and fibroblast growth factor receptor-3 (FGFR3) in bladder cancer cells. Carcinogenesis. 2012;33:886–94.PubMedPubMedCentralCrossRef
212.
go back to reference Sakurai R, Villarreal P, Husain S, Liu J, Sakurai T, Tou E, Torday JS, Rehan VK. Curcumin protects the developing lung against long-term hyperoxic injury. Am J Phys Lung Cell Mol Phys. 2013;305:L301–11. Sakurai R, Villarreal P, Husain S, Liu J, Sakurai T, Tou E, Torday JS, Rehan VK. Curcumin protects the developing lung against long-term hyperoxic injury. Am J Phys Lung Cell Mol Phys. 2013;305:L301–11.
213.
go back to reference Narasimhan M, Ammanamanchi S. Curcumin blocks RON tyrosine kinase–mediated invasion of breast carcinoma cells. Cancer Res. 2008;68:5185–92.PubMedCrossRef Narasimhan M, Ammanamanchi S. Curcumin blocks RON tyrosine kinase–mediated invasion of breast carcinoma cells. Cancer Res. 2008;68:5185–92.PubMedCrossRef
214.
go back to reference Xia Y, Lian S, Khoi PN, Yoon HJ, Han JY, Chay KO, Kim KK, Jung YD. Chrysin inhibits cell invasion by inhibition of Recepteur d'origine Nantais via suppressing early growth response-1 and NF-κB transcription factor activities in gastric cancer cells. Int J Oncol. Xia Y, Lian S, Khoi PN, Yoon HJ, Han JY, Chay KO, Kim KK, Jung YD. Chrysin inhibits cell invasion by inhibition of Recepteur d'origine Nantais via suppressing early growth response-1 and NF-κB transcription factor activities in gastric cancer cells. Int J Oncol.
215.
go back to reference Irby RB, Yeatman TJ. Role of Src expression and activation in human cancer. Oncogene. 2000;19:5636–42.PubMedCrossRef Irby RB, Yeatman TJ. Role of Src expression and activation in human cancer. Oncogene. 2000;19:5636–42.PubMedCrossRef
216.
go back to reference Greuber EK, Smith-Pearson P, Wang J, Pendergast AM. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat Rev Cancer. 2013;13:559–71.PubMedPubMedCentralCrossRef Greuber EK, Smith-Pearson P, Wang J, Pendergast AM. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat Rev Cancer. 2013;13:559–71.PubMedPubMedCentralCrossRef
218.
go back to reference Byun S, Lee KW, Jung SK, Lee EJ, Hwang MK, Lim SH, Bode AM, Lee HJ, Dong Z. Luteolin inhibits protein kinase Cε and c-Src activities and UVB-induced skin cancer. Cancer Res. 2010;70:2415–23.PubMedCrossRef Byun S, Lee KW, Jung SK, Lee EJ, Hwang MK, Lim SH, Bode AM, Lee HJ, Dong Z. Luteolin inhibits protein kinase Cε and c-Src activities and UVB-induced skin cancer. Cancer Res. 2010;70:2415–23.PubMedCrossRef
219.
go back to reference Wolanin K, Magalska A, Mosieniak G, Klinger R, Mckenna S, Vejda S, Sikora E, Piwocka K. Curcumin affects components of the chromosomal passenger complex and induces mitotic catastrophe in apoptosis-resistant Bcr-Abl-expressing cells. Mol Cancer Res. 2006;4:457–69.PubMedCrossRef Wolanin K, Magalska A, Mosieniak G, Klinger R, Mckenna S, Vejda S, Sikora E, Piwocka K. Curcumin affects components of the chromosomal passenger complex and induces mitotic catastrophe in apoptosis-resistant Bcr-Abl-expressing cells. Mol Cancer Res. 2006;4:457–69.PubMedCrossRef
220.
go back to reference Milligan SA, Burke P, Coleman DT, Bigelow RL, Steffan JJ, Carroll JL, Williams BJ, Cardelli JA. The green tea polyphenol EGCG potentiates the Antiproliferative activity of c-met and epidermal growth factor receptor inhibitors in non–small cell lung cancer cells. Clin Cancer Res. 2009;15:4885–94.PubMedPubMedCentralCrossRef Milligan SA, Burke P, Coleman DT, Bigelow RL, Steffan JJ, Carroll JL, Williams BJ, Cardelli JA. The green tea polyphenol EGCG potentiates the Antiproliferative activity of c-met and epidermal growth factor receptor inhibitors in non–small cell lung cancer cells. Clin Cancer Res. 2009;15:4885–94.PubMedPubMedCentralCrossRef
221.
go back to reference Rajasingh J, Raikwar HP, Muthian G, Johnson C, Bright JJ. Curcumin induces growth-arrest and apoptosis in association with the inhibition of constitutively active JAK–STAT pathway in T cell leukemia. Biochem Biophys Res Commun. 2006;340:359–68.PubMedCrossRef Rajasingh J, Raikwar HP, Muthian G, Johnson C, Bright JJ. Curcumin induces growth-arrest and apoptosis in association with the inhibition of constitutively active JAK–STAT pathway in T cell leukemia. Biochem Biophys Res Commun. 2006;340:359–68.PubMedCrossRef
222.
go back to reference Wang G, Wang J, Chen X, Du S, Li D, Pei Z, Lan H, Wu L. The JAK2/STAT3 and mitochondrial pathways are essential for quercetin nanoliposome-induced C6 glioma cell death. Cell Death Dis. 2013;4:e746.PubMedPubMedCentralCrossRef Wang G, Wang J, Chen X, Du S, Li D, Pei Z, Lan H, Wu L. The JAK2/STAT3 and mitochondrial pathways are essential for quercetin nanoliposome-induced C6 glioma cell death. Cell Death Dis. 2013;4:e746.PubMedPubMedCentralCrossRef
223.
go back to reference Vara JÁF, Casado E, de Castro J, Cejas P, Belda-Iniesta C, González-Barón M. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 2004;30:193–204.CrossRef Vara JÁF, Casado E, de Castro J, Cejas P, Belda-Iniesta C, González-Barón M. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 2004;30:193–204.CrossRef
224.
go back to reference Li Y, Liu J, Liu X, Xing K, Wang Y, Li F, Yao L. Resveratrol-induced cell inhibition of growth and apoptosis in MCF7 human breast cancer cells are associated with modulation of phosphorylated Akt and caspase-9. Appl Biochem Biotechnol. 2006;135:181–92.PubMedCrossRef Li Y, Liu J, Liu X, Xing K, Wang Y, Li F, Yao L. Resveratrol-induced cell inhibition of growth and apoptosis in MCF7 human breast cancer cells are associated with modulation of phosphorylated Akt and caspase-9. Appl Biochem Biotechnol. 2006;135:181–92.PubMedCrossRef
225.
go back to reference Sexton É, Van Themsche C, Leblanc K, Parent S, Lemoine P, Asselin E. Resveratrol interferes with AKT activity and triggers apoptosis in human uterine cancer cells. Mol Cancer. 2006;5:45.PubMedPubMedCentralCrossRef Sexton É, Van Themsche C, Leblanc K, Parent S, Lemoine P, Asselin E. Resveratrol interferes with AKT activity and triggers apoptosis in human uterine cancer cells. Mol Cancer. 2006;5:45.PubMedPubMedCentralCrossRef
226.
go back to reference Aziz MH, Nihal M, Fu VX, Jarrard DF, Ahmad N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol Cancer Ther. 2006;5:1335–41.PubMedCrossRef Aziz MH, Nihal M, Fu VX, Jarrard DF, Ahmad N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol Cancer Ther. 2006;5:1335–41.PubMedCrossRef
227.
go back to reference Sheth S, Jajoo S, Kaur T, Mukherjea D, Sheehan K, Rybak LP, Ramkumar V. Resveratrol reduces prostate cancer growth and metastasis by inhibiting the Akt/MicroRNA-21 pathway. PLoS One. 2012;7:e51655.PubMedPubMedCentralCrossRef Sheth S, Jajoo S, Kaur T, Mukherjea D, Sheehan K, Rybak LP, Ramkumar V. Resveratrol reduces prostate cancer growth and metastasis by inhibiting the Akt/MicroRNA-21 pathway. PLoS One. 2012;7:e51655.PubMedPubMedCentralCrossRef
228.
go back to reference Roy P, Kalra N, Prasad S, George J, Shukla Y. Chemopreventive potential of resveratrol in mouse skin tumors through regulation of mitochondrial and PI3K/AKT signaling pathways. Pharm Res. 2009;26:211–7.PubMedCrossRef Roy P, Kalra N, Prasad S, George J, Shukla Y. Chemopreventive potential of resveratrol in mouse skin tumors through regulation of mitochondrial and PI3K/AKT signaling pathways. Pharm Res. 2009;26:211–7.PubMedCrossRef
229.
go back to reference Jiang H, Shang X, Wu H, Gautam SC, Al-Holou S, Li C, Kuo J, Zhang L, Chopp M. Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. J Exp Ther Oncol. 2009;8:25.PubMedPubMedCentral Jiang H, Shang X, Wu H, Gautam SC, Al-Holou S, Li C, Kuo J, Zhang L, Chopp M. Resveratrol downregulates PI3K/Akt/mTOR signaling pathways in human U251 glioma cells. J Exp Ther Oncol. 2009;8:25.PubMedPubMedCentral
231.
go back to reference Park D, Jeong H, Lee MN, Koh A, Kwon O, Yang YR, Noh J, Suh P-G, Park H, Ryu SH. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci Rep. 2016;6:21772.PubMedPubMedCentralCrossRef Park D, Jeong H, Lee MN, Koh A, Kwon O, Yang YR, Noh J, Suh P-G, Park H, Ryu SH. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci Rep. 2016;6:21772.PubMedPubMedCentralCrossRef
232.
go back to reference Kumamoto T, Fujii M, Hou D-X. Akt is a direct target for myricetin to inhibit cell transformation. Mol Cell Biochem. 2009;332:33–41.PubMedCrossRef Kumamoto T, Fujii M, Hou D-X. Akt is a direct target for myricetin to inhibit cell transformation. Mol Cell Biochem. 2009;332:33–41.PubMedCrossRef
233.
go back to reference Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, Tridandapani S, Anant S, Kuppusamy P. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biol Therapy. 2007;6:178–84.CrossRef Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, Tridandapani S, Anant S, Kuppusamy P. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biol Therapy. 2007;6:178–84.CrossRef
234.
go back to reference Granado-Serrano AB, Martín MA, Bravo L, Goya L, Ramos S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J Nutr. 2006;136:2715–21.PubMedCrossRef Granado-Serrano AB, Martín MA, Bravo L, Goya L, Ramos S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J Nutr. 2006;136:2715–21.PubMedCrossRef
235.
go back to reference Gulati N, Laudet B, Zohrabian VM, Murali R, Jhanwar-Uniyal M. The antiproliferative effect of quercetin in cancer cells is mediated via inhibition of the PI3K-Akt/PKB pathway. Anticancer Res. 2006;26:1177–81.PubMed Gulati N, Laudet B, Zohrabian VM, Murali R, Jhanwar-Uniyal M. The antiproliferative effect of quercetin in cancer cells is mediated via inhibition of the PI3K-Akt/PKB pathway. Anticancer Res. 2006;26:1177–81.PubMed
236.
go back to reference Tang FY, Nguyen N, Meydani M. Green tea catechins inhibit VEGF-induced angiogenesis in vitro through suppression of VE-cadherin phosphorylation and inactivation of Akt molecule. Int J Cancer. 2003;106:871–8.PubMedCrossRef Tang FY, Nguyen N, Meydani M. Green tea catechins inhibit VEGF-induced angiogenesis in vitro through suppression of VE-cadherin phosphorylation and inactivation of Akt molecule. Int J Cancer. 2003;106:871–8.PubMedCrossRef
237.
go back to reference Shin DY, Lee WS, Lu JN, Kang MH, Ryu CH, Kim GY, Kang HS, Shin SC, Choi YH. Induction of apoptosis in human colon cancer HCT-116 cells by anthocyanins through suppression of Akt and activation of p38-MAPK. Int J Oncol. 2009;35:1499–504.PubMed Shin DY, Lee WS, Lu JN, Kang MH, Ryu CH, Kim GY, Kang HS, Shin SC, Choi YH. Induction of apoptosis in human colon cancer HCT-116 cells by anthocyanins through suppression of Akt and activation of p38-MAPK. Int J Oncol. 2009;35:1499–504.PubMed
238.
go back to reference Kavitha K, Kowshik J, Kishore TKK, Baba AB, Nagini S. Astaxanthin inhibits NF-κB and Wnt/β-catenin signaling pathways via inactivation of Erk/MAPK and PI3K/Akt to induce intrinsic apoptosis in a hamster model of oral cancer. Biochimica et Biophysica Acta (BBA)-General Subjects. 2013;1830:4433–44.CrossRef Kavitha K, Kowshik J, Kishore TKK, Baba AB, Nagini S. Astaxanthin inhibits NF-κB and Wnt/β-catenin signaling pathways via inactivation of Erk/MAPK and PI3K/Akt to induce intrinsic apoptosis in a hamster model of oral cancer. Biochimica et Biophysica Acta (BBA)-General Subjects. 2013;1830:4433–44.CrossRef
239.
240.
go back to reference Kaur P, Shukla S, Gupta S. Plant flavonoid apigenin inactivates Akt to trigger apoptosis in human prostate cancer: an in vitro and in vivo study. Carcinogenesis. 2008;29:2210–7.PubMedPubMedCentralCrossRef Kaur P, Shukla S, Gupta S. Plant flavonoid apigenin inactivates Akt to trigger apoptosis in human prostate cancer: an in vitro and in vivo study. Carcinogenesis. 2008;29:2210–7.PubMedPubMedCentralCrossRef
241.
go back to reference Sharma PS, Sharma R, Tyagi R. Inhibitors of cyclin dependent kinases: useful targets for cancer treatment. Curr Cancer Drug Targets. 2008;8:53–75.PubMedCrossRef Sharma PS, Sharma R, Tyagi R. Inhibitors of cyclin dependent kinases: useful targets for cancer treatment. Curr Cancer Drug Targets. 2008;8:53–75.PubMedCrossRef
242.
go back to reference Wolter F, Akoglu B, Clausnitzer A, Stein J. Downregulation of the cyclin D1/Cdk4 complex occurs during resveratrol-induced cell cycle arrest in colon cancer cell lines. J Nutr. 2001;131:2197–203.PubMedCrossRef Wolter F, Akoglu B, Clausnitzer A, Stein J. Downregulation of the cyclin D1/Cdk4 complex occurs during resveratrol-induced cell cycle arrest in colon cancer cell lines. J Nutr. 2001;131:2197–203.PubMedCrossRef
243.
go back to reference Shan B-E, Wang M-X, Li R-Q. Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/β-catenin signaling pathway. Cancer Investig. 2009;27:604–12.CrossRef Shan B-E, Wang M-X, Li R-Q. Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/β-catenin signaling pathway. Cancer Investig. 2009;27:604–12.CrossRef
244.
go back to reference Mukhopadhyay A, Banerjee S, Stafford LJ, Xia C, Liu M, Aggarwal BB. Curcumin-induced suppression of cell proliferation correlates with down-regulation of cyclin D1 expression and CDK4-mediated retinoblastoma protein phosphorylation. Oncogene. 2002;21:8852–61.PubMedCrossRef Mukhopadhyay A, Banerjee S, Stafford LJ, Xia C, Liu M, Aggarwal BB. Curcumin-induced suppression of cell proliferation correlates with down-regulation of cyclin D1 expression and CDK4-mediated retinoblastoma protein phosphorylation. Oncogene. 2002;21:8852–61.PubMedCrossRef
245.
go back to reference Aggarwal BB, Banerjee S, Bharadwaj U, Sung B, Shishodia S, Sethi G. Curcumin induces the degradation of cyclin E expression through ubiquitin-dependent pathway and up-regulates cyclin-dependent kinase inhibitors p21 and p27 in multiple human tumor cell lines. Biochem Pharmacol. 2007;73:1024–32.PubMedCrossRef Aggarwal BB, Banerjee S, Bharadwaj U, Sung B, Shishodia S, Sethi G. Curcumin induces the degradation of cyclin E expression through ubiquitin-dependent pathway and up-regulates cyclin-dependent kinase inhibitors p21 and p27 in multiple human tumor cell lines. Biochem Pharmacol. 2007;73:1024–32.PubMedCrossRef
246.
go back to reference Thangapazham RL, Singh AK, Sharma A, Warren J, Gaddipati JP, Maheshwari RK. Green tea polyphenols and its constituent epigallocatechin gallate inhibits proliferation of human breast cancer cells in vitro and in vivo. Cancer Lett. 2007;245:232–41.PubMedCrossRef Thangapazham RL, Singh AK, Sharma A, Warren J, Gaddipati JP, Maheshwari RK. Green tea polyphenols and its constituent epigallocatechin gallate inhibits proliferation of human breast cancer cells in vitro and in vivo. Cancer Lett. 2007;245:232–41.PubMedCrossRef
247.
go back to reference Van Aller GS, Carson JD, Tang W, Peng H, Zhao L, Copeland RA, Tummino PJ, Luo L. Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res Commun. 2011;406:194–9.PubMedCrossRef Van Aller GS, Carson JD, Tang W, Peng H, Zhao L, Copeland RA, Tummino PJ, Luo L. Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res Commun. 2011;406:194–9.PubMedCrossRef
248.
go back to reference Li M, Zhang Z, Hill DL, Wang H, Zhang R. Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway. Cancer Res. 2007;67:1988–96.PubMedCrossRef Li M, Zhang Z, Hill DL, Wang H, Zhang R. Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway. Cancer Res. 2007;67:1988–96.PubMedCrossRef
249.
go back to reference Adhami VM, Syed DN, Khan N, Mukhtar H. Dietary flavonoid fisetin: a novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem Pharmacol. 2012;84:1277–81.PubMedCrossRef Adhami VM, Syed DN, Khan N, Mukhtar H. Dietary flavonoid fisetin: a novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem Pharmacol. 2012;84:1277–81.PubMedCrossRef
250.
go back to reference Seo YJ, Kim BS, Chun SY, Park YK, Kang KS, Kwon TG. Apoptotic effects of genistein, biochanin-a and apigenin on LNCaP and PC-3 cells by p21 through transcriptional inhibition of polo-like kinase-1. J Korean Med Sci. 2011;26:1489–94.PubMedPubMedCentralCrossRef Seo YJ, Kim BS, Chun SY, Park YK, Kang KS, Kwon TG. Apoptotic effects of genistein, biochanin-a and apigenin on LNCaP and PC-3 cells by p21 through transcriptional inhibition of polo-like kinase-1. J Korean Med Sci. 2011;26:1489–94.PubMedPubMedCentralCrossRef
251.
go back to reference Xie F, Lang Q, Zhou M, Zhang H, Zhang Z, Zhang Y, Wan B, Huang Q, Yu L. The dietary flavonoid luteolin inhibits aurora B kinase activity and blocks proliferation of cancer cells. Eur J Pharm Sci. 2012;46:388–96.PubMedCrossRef Xie F, Lang Q, Zhou M, Zhang H, Zhang Z, Zhang Y, Wan B, Huang Q, Yu L. The dietary flavonoid luteolin inhibits aurora B kinase activity and blocks proliferation of cancer cells. Eur J Pharm Sci. 2012;46:388–96.PubMedCrossRef
252.
254.
go back to reference Xavier CP, Lima CF, Preto A, Seruca R, Fernandes-Ferreira M, Pereira-Wilson C. Luteolin, quercetin and ursolic acid are potent inhibitors of proliferation and inducers of apoptosis in both KRAS and BRAF mutated human colorectal cancer cells. Cancer Lett. 2009;281:162–70.PubMedCrossRef Xavier CP, Lima CF, Preto A, Seruca R, Fernandes-Ferreira M, Pereira-Wilson C. Luteolin, quercetin and ursolic acid are potent inhibitors of proliferation and inducers of apoptosis in both KRAS and BRAF mutated human colorectal cancer cells. Cancer Lett. 2009;281:162–70.PubMedCrossRef
255.
256.
go back to reference Chen L-X, He Y-J, Zhao S-Z, Wu J-G, Wang J-T, Zhu L-M, Lin T-T, Sun B-C, Li X-R. Inhibition of tumor growth and vasculogenic mimicry by curcumin through down-regulation of the EphA2/PI3K/MMP pathway in a murine choroidal melanoma model. Cancer Biol Therapy. 2011;11:229–35.CrossRef Chen L-X, He Y-J, Zhao S-Z, Wu J-G, Wang J-T, Zhu L-M, Lin T-T, Sun B-C, Li X-R. Inhibition of tumor growth and vasculogenic mimicry by curcumin through down-regulation of the EphA2/PI3K/MMP pathway in a murine choroidal melanoma model. Cancer Biol Therapy. 2011;11:229–35.CrossRef
257.
go back to reference Khan N, Afaq F, Saleem M, Ahmad N, Mukhtar H. Targeting multiple signaling pathways by green tea polyphenol (−)-epigallocatechin-3-gallate. Cancer Res. 2006;66:2500–5.PubMedCrossRef Khan N, Afaq F, Saleem M, Ahmad N, Mukhtar H. Targeting multiple signaling pathways by green tea polyphenol (−)-epigallocatechin-3-gallate. Cancer Res. 2006;66:2500–5.PubMedCrossRef
258.
go back to reference Wu JC, Lai CS, Badmaev V, Nagabhushanam K, Ho CT, Pan MH. Tetrahydrocurcumin, a major metabolite of curcumin, induced autophagic cell death through coordinative modulation of PI3K/Akt-mTOR and MAPK signaling pathways in human leukemia HL-60 cells. Mol Nutr Food Res. 2011;55:1646–54.PubMedCrossRef Wu JC, Lai CS, Badmaev V, Nagabhushanam K, Ho CT, Pan MH. Tetrahydrocurcumin, a major metabolite of curcumin, induced autophagic cell death through coordinative modulation of PI3K/Akt-mTOR and MAPK signaling pathways in human leukemia HL-60 cells. Mol Nutr Food Res. 2011;55:1646–54.PubMedCrossRef
259.
go back to reference Heffernan-Stroud LA, Obeid LM. Sphingosine kinase 1 in cancer. Adv Cancer Res. 2012;201. Heffernan-Stroud LA, Obeid LM. Sphingosine kinase 1 in cancer. Adv Cancer Res. 2012;201.
260.
go back to reference Lim KG, Gray AI, Pyne S, Pyne NJ. Resveratrol dimers are novel sphingosine kinase 1 inhibitors and affect sphingosine kinase 1 expression and cancer cell growth and survival. Br J Pharmacol. 2012;166:1605–16.PubMedPubMedCentralCrossRef Lim KG, Gray AI, Pyne S, Pyne NJ. Resveratrol dimers are novel sphingosine kinase 1 inhibitors and affect sphingosine kinase 1 expression and cancer cell growth and survival. Br J Pharmacol. 2012;166:1605–16.PubMedPubMedCentralCrossRef
261.
go back to reference Kim Y, Kim W-J, Cha E-J. Quercetin-induced growth inhibition in human bladder cancer cells is associated with an increase in Ca2+−activated K+ channels. Korean J Physiol Pharmacol. 2011;15:279–83.PubMedPubMedCentralCrossRef Kim Y, Kim W-J, Cha E-J. Quercetin-induced growth inhibition in human bladder cancer cells is associated with an increase in Ca2+−activated K+ channels. Korean J Physiol Pharmacol. 2011;15:279–83.PubMedPubMedCentralCrossRef
262.
go back to reference Brizuela L, Dayon A, Doumerc N, Ader I, Golzio M, Izard J-C, Hara Y, Malavaud B, Cuvillier O. The sphingosine kinase-1 survival pathway is a molecular target for the tumor-suppressive tea and wine polyphenols in prostate cancer. FASEB J. 2010;24:3882–94.PubMedCrossRef Brizuela L, Dayon A, Doumerc N, Ader I, Golzio M, Izard J-C, Hara Y, Malavaud B, Cuvillier O. The sphingosine kinase-1 survival pathway is a molecular target for the tumor-suppressive tea and wine polyphenols in prostate cancer. FASEB J. 2010;24:3882–94.PubMedCrossRef
263.
go back to reference Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–9.PubMedPubMedCentralCrossRef Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–9.PubMedPubMedCentralCrossRef
264.
go back to reference Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nat Med. 2013;19:1401–9.PubMedCrossRef Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF inhibitors. Nat Med. 2013;19:1401–9.PubMedCrossRef
265.
go back to reference Chell V, Balmanno K, Little A, Wilson M, Andrews S, Blockley L, Hampson M, Gavine P, Cook S. Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene. 2013;32:3059–70.PubMedCrossRef Chell V, Balmanno K, Little A, Wilson M, Andrews S, Blockley L, Hampson M, Gavine P, Cook S. Tumour cell responses to new fibroblast growth factor receptor tyrosine kinase inhibitors and identification of a gatekeeper mutation in FGFR3 as a mechanism of acquired resistance. Oncogene. 2013;32:3059–70.PubMedCrossRef
266.
go back to reference Foguel D, Cordeiro Y, Ventura S, Graña-Montes AA, Cortines J. The importance of a gatekeeper residue on. 2014. Foguel D, Cordeiro Y, Ventura S, Graña-Montes AA, Cortines J. The importance of a gatekeeper residue on. 2014.
267.
go back to reference Chen R, An T, Zhao J, Duan J, Wang Z, Zhuo M, Wang S, Wu M, Li Z, Yang X. Non-invasive, Dynamic and Quantitative Analysis of T790M mutation in matched plasma DNA of pre-and post-EGFR-TKIs treatment for advanced NSCLC. 第 13 届全国肺癌学术大会论文汇编. 2013. Chen R, An T, Zhao J, Duan J, Wang Z, Zhuo M, Wang S, Wu M, Li Z, Yang X. Non-invasive, Dynamic and Quantitative Analysis of T790M mutation in matched plasma DNA of pre-and post-EGFR-TKIs treatment for advanced NSCLC. 第 13 届全国肺癌学术大会论文汇编. 2013.
268.
go back to reference Pauwels D, Sweron B, Cools J. The N676D and G697R mutations in the kinase domain of FLT3 confer resistance to the inhibitor AC220. Haematologica. 2012;97:1773–4.PubMedPubMedCentralCrossRef Pauwels D, Sweron B, Cools J. The N676D and G697R mutations in the kinase domain of FLT3 confer resistance to the inhibitor AC220. Haematologica. 2012;97:1773–4.PubMedPubMedCentralCrossRef
269.
go back to reference Gibbons DL, Pricl S, Kantarjian H, Cortes J, Quintás-Cardama A. The rise and fall of gatekeeper mutations? The BCR-ABL1 T315I paradigm. Cancer. 2012;118:293–9.PubMedCrossRef Gibbons DL, Pricl S, Kantarjian H, Cortes J, Quintás-Cardama A. The rise and fall of gatekeeper mutations? The BCR-ABL1 T315I paradigm. Cancer. 2012;118:293–9.PubMedCrossRef
270.
go back to reference Metzgeroth G, Erben P, Martin H, Mousset S, Teichmann M, Walz C, Klippstein T, Hochhaus A, Cross N, Hofmann W. Limited clinical activity of nilotinib and sorafenib in FIP1L1-PDGFRA positive chronic eosinophilic leukemia with imatinib-resistant T674I mutation. Leukemia. 2012;26:162–4.PubMedCrossRef Metzgeroth G, Erben P, Martin H, Mousset S, Teichmann M, Walz C, Klippstein T, Hochhaus A, Cross N, Hofmann W. Limited clinical activity of nilotinib and sorafenib in FIP1L1-PDGFRA positive chronic eosinophilic leukemia with imatinib-resistant T674I mutation. Leukemia. 2012;26:162–4.PubMedCrossRef
271.
go back to reference Heinrich MC, Corless CL, Blanke CD, Demetri GD, Joensuu H, Roberts PJ, Eisenberg BL, von Mehren M, Fletcher CD, Sandau K. Molecular correlates of imatinib resistance in gastrointestinal stromal tumors. J Clin Oncol. 2006;24:4764–74.PubMedCrossRef Heinrich MC, Corless CL, Blanke CD, Demetri GD, Joensuu H, Roberts PJ, Eisenberg BL, von Mehren M, Fletcher CD, Sandau K. Molecular correlates of imatinib resistance in gastrointestinal stromal tumors. J Clin Oncol. 2006;24:4764–74.PubMedCrossRef
272.
go back to reference Yun C-H, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong K-K, Meyerson M, Eck MJ. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci. 2008;105:2070–5.PubMedPubMedCentralCrossRef Yun C-H, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong K-K, Meyerson M, Eck MJ. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci. 2008;105:2070–5.PubMedPubMedCentralCrossRef
273.
go back to reference Suda K, Onozato R, Yatabe Y, Mitsudomi T. EGFR T790M mutation: a double role in lung cancer cell survival? J Thorac Oncol. 2009;4:1–4.PubMedCrossRef Suda K, Onozato R, Yatabe Y, Mitsudomi T. EGFR T790M mutation: a double role in lung cancer cell survival? J Thorac Oncol. 2009;4:1–4.PubMedCrossRef
274.
go back to reference Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale C-M, Zhao X, Christensen J. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale C-M, Zhao X, Christensen J. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–43.PubMedCrossRef
275.
go back to reference Janku F, Wheler JJ, Westin SN, Moulder SL, Naing A, Tsimberidou AM, Fu S, Falchook GS, Hong DS, Garrido-Laguna I. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol. 2012;30:777–82.PubMedPubMedCentralCrossRef Janku F, Wheler JJ, Westin SN, Moulder SL, Naing A, Tsimberidou AM, Fu S, Falchook GS, Hong DS, Garrido-Laguna I. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol. 2012;30:777–82.PubMedPubMedCentralCrossRef
276.
go back to reference Blencke S, Zech B, Engkvist O, Greff Z, Őrfi L, Horváth Z, Kéri G, Ullrich A, Daub H. Characterization of a conserved structural determinant controlling protein kinase sensitivity to selective inhibitors. Chem Biol. 2004;11:691–701.PubMedCrossRef Blencke S, Zech B, Engkvist O, Greff Z, Őrfi L, Horváth Z, Kéri G, Ullrich A, Daub H. Characterization of a conserved structural determinant controlling protein kinase sensitivity to selective inhibitors. Chem Biol. 2004;11:691–701.PubMedCrossRef
277.
go back to reference Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, Kris MG, Varmus H. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73.PubMedPubMedCentralCrossRef Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, Kris MG, Varmus H. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73.PubMedPubMedCentralCrossRef
278.
go back to reference Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, Johnson BE, Eck MJ, Tenen DG, Halmos B. EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786–92.PubMedCrossRef Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, Johnson BE, Eck MJ, Tenen DG, Halmos B. EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786–92.PubMedCrossRef
279.
go back to reference Heymach JV, Nilsson M, Blumenschein G, Papadimitrakopoulou V, Herbst R. Epidermal growth factor receptor inhibitors in development for the treatment of non–small cell lung cancer. Clin Cancer Res. 2006;12:4441s–5s.PubMedCrossRef Heymach JV, Nilsson M, Blumenschein G, Papadimitrakopoulou V, Herbst R. Epidermal growth factor receptor inhibitors in development for the treatment of non–small cell lung cancer. Clin Cancer Res. 2006;12:4441s–5s.PubMedCrossRef
280.
go back to reference Patel RY, Doerksen RJ. Protein kinase− inhibitor database: structural variability of and inhibitor interactions with the protein kinase P-loop. J Proteome Res. 2010;9:4433–42.PubMedPubMedCentralCrossRef Patel RY, Doerksen RJ. Protein kinase− inhibitor database: structural variability of and inhibitor interactions with the protein kinase P-loop. J Proteome Res. 2010;9:4433–42.PubMedPubMedCentralCrossRef
282.
go back to reference Warburton C, Dragowska WH, Gelmon K, Chia S, Yan H, Masin D, Denyssevych T, Wallis AE, Bally MB. Treatment of HER-2/neu overexpressing breast cancer xenograft models with trastuzumab (Herceptin) and gefitinib (ZD1839): drug combination effects on tumor growth, HER-2/neu and epidermal growth factor receptor expression, and viable hypoxic cell fraction. Clin Cancer Res. 2004;10:2512–24.PubMedCrossRef Warburton C, Dragowska WH, Gelmon K, Chia S, Yan H, Masin D, Denyssevych T, Wallis AE, Bally MB. Treatment of HER-2/neu overexpressing breast cancer xenograft models with trastuzumab (Herceptin) and gefitinib (ZD1839): drug combination effects on tumor growth, HER-2/neu and epidermal growth factor receptor expression, and viable hypoxic cell fraction. Clin Cancer Res. 2004;10:2512–24.PubMedCrossRef
283.
go back to reference Normanno N, Campiglio M, Perrone F, De Luca A, Menard S. Is the gefitinib plus trastuzumab combination feasible in breast cancer patients? Ann Oncol. 2005;16:1709.PubMedCrossRef Normanno N, Campiglio M, Perrone F, De Luca A, Menard S. Is the gefitinib plus trastuzumab combination feasible in breast cancer patients? Ann Oncol. 2005;16:1709.PubMedCrossRef
284.
go back to reference Shah DR, Shah RR, Morganroth J. Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Saf. 2013;36:413–26.PubMedCrossRef Shah DR, Shah RR, Morganroth J. Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Saf. 2013;36:413–26.PubMedCrossRef
285.
go back to reference Orphanos GS, Ioannidis GN, Ardavanis AG. Cardiotoxicity induced by tyrosine kinase inhibitors. Acta Oncol. 2009;48:964–70.PubMedCrossRef Orphanos GS, Ioannidis GN, Ardavanis AG. Cardiotoxicity induced by tyrosine kinase inhibitors. Acta Oncol. 2009;48:964–70.PubMedCrossRef
287.
288.
go back to reference Bernards R, Brummelkamp TR, Beijersbergen RL. shRNA libraries and their use in cancer genetics. Nat Methods. 2006;3:701–6.PubMedCrossRef Bernards R, Brummelkamp TR, Beijersbergen RL. shRNA libraries and their use in cancer genetics. Nat Methods. 2006;3:701–6.PubMedCrossRef
289.
go back to reference Modugno M, Casale E, Soncini C, Rosettani P, Colombo R, Lupi R, Rusconi L, Fancelli D, Carpinelli P, Cameron AD. Crystal structure of the T315I Abl mutant in complex with the aurora kinases inhibitor PHA-739358. Cancer Res. 2007;67:7987–90.PubMedCrossRef Modugno M, Casale E, Soncini C, Rosettani P, Colombo R, Lupi R, Rusconi L, Fancelli D, Carpinelli P, Cameron AD. Crystal structure of the T315I Abl mutant in complex with the aurora kinases inhibitor PHA-739358. Cancer Res. 2007;67:7987–90.PubMedCrossRef
290.
go back to reference Weisberg E, Manley PW, Breitenstein W, Brüggen J, Cowan-Jacob SW, Ray A, Huntly B, Fabbro D, Fendrich G, Hall-Meyers E. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005;7:129–41.PubMedCrossRef Weisberg E, Manley PW, Breitenstein W, Brüggen J, Cowan-Jacob SW, Ray A, Huntly B, Fabbro D, Fendrich G, Hall-Meyers E. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005;7:129–41.PubMedCrossRef
291.
go back to reference Gumireddy K, Reddy MR, Cosenza SC, Nathan RB, Baker SJ, Papathi N, Jiang J, Holland J, Reddy EP. ON01910, a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell. 2005;7:275–86.PubMedCrossRef Gumireddy K, Reddy MR, Cosenza SC, Nathan RB, Baker SJ, Papathi N, Jiang J, Holland J, Reddy EP. ON01910, a non-ATP-competitive small molecule inhibitor of Plk1, is a potent anticancer agent. Cancer Cell. 2005;7:275–86.PubMedCrossRef
292.
go back to reference Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A, Lee FY, Weinmann R, Holyoake TL. BMS-214662 potently induces apoptosis of chronic myeloid leukemia stem and progenitor cells and synergizes with tyrosine kinase inhibitors. Blood. 2008;111:2843–53.PubMedCrossRef Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A, Lee FY, Weinmann R, Holyoake TL. BMS-214662 potently induces apoptosis of chronic myeloid leukemia stem and progenitor cells and synergizes with tyrosine kinase inhibitors. Blood. 2008;111:2843–53.PubMedCrossRef
293.
go back to reference Lindsley CW, Zhao Z, Leister WH, Robinson RG, Barnett SF, Defeo-Jones D, Jones RE, Hartman GD, Huff JR, Huber HE. Allosteric Akt (PKB) inhibitors: discovery and SAR of isozyme selective inhibitors. Bioorg Med Chem Lett. 2005;15:761–4.PubMedCrossRef Lindsley CW, Zhao Z, Leister WH, Robinson RG, Barnett SF, Defeo-Jones D, Jones RE, Hartman GD, Huff JR, Huber HE. Allosteric Akt (PKB) inhibitors: discovery and SAR of isozyme selective inhibitors. Bioorg Med Chem Lett. 2005;15:761–4.PubMedCrossRef
294.
go back to reference Barnett S, Defeo-Jones D, Fu S, Hancock P, Haskell K, Jones R, Kahana J, Kral A, Leander K, Lee L. Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors. Biochem J. 2005;385:399–408.PubMedPubMedCentralCrossRef Barnett S, Defeo-Jones D, Fu S, Hancock P, Haskell K, Jones R, Kahana J, Kral A, Leander K, Lee L. Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors. Biochem J. 2005;385:399–408.PubMedPubMedCentralCrossRef
295.
go back to reference Converso A, Hartingh T, Garbaccio RM, Tasber E, Rickert K, Fraley ME, Yan Y, Kreatsoulas C, Stirdivant S, Drakas B. Development of thioquinazolinones, allosteric Chk1 kinase inhibitors. Bioorg Med Chem Lett. 2009;19:1240–4.PubMedCrossRef Converso A, Hartingh T, Garbaccio RM, Tasber E, Rickert K, Fraley ME, Yan Y, Kreatsoulas C, Stirdivant S, Drakas B. Development of thioquinazolinones, allosteric Chk1 kinase inhibitors. Bioorg Med Chem Lett. 2009;19:1240–4.PubMedCrossRef
296.
go back to reference Fabbro D, Manley PW, Jahnke W, Liebetanz J, Szyttenholm A, Fendrich G, Strauss A, Zhang J, Gray NS, Adrian F. Inhibitors of the Abl kinase directed at either the ATP-or myristate-binding site. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2010;1804:454–62.CrossRef Fabbro D, Manley PW, Jahnke W, Liebetanz J, Szyttenholm A, Fendrich G, Strauss A, Zhang J, Gray NS, Adrian F. Inhibitors of the Abl kinase directed at either the ATP-or myristate-binding site. Biochimica et Biophysica Acta (BBA)-Proteins and Proteomics. 2010;1804:454–62.CrossRef
Metadata
Title
Kinase-targeted cancer therapies: progress, challenges and future directions
Authors
Khushwant S. Bhullar
Naiara Orrego Lagarón
Eileen M. McGowan
Indu Parmar
Amitabh Jha
Basil P. Hubbard
H. P. Vasantha Rupasinghe
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2018
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0804-2

Other articles of this Issue 1/2018

Molecular Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine