Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors

Edited by Katerina Politi, Yale University, New Haven, CT, and accepted by the Editorial Board October 7, 2014 (received for review February 25, 2014)
October 27, 2014
111 (45) E4869-E4877

Significance

Inhibitors of the FGF receptors (FGFRs) are currently under clinical investigation for the treatment of various cancers. All currently approved kinase inhibitors eventually are rendered useless by the emergence of drug-resistant tumors. We used structure-based drug design to develop the first, to our knowledge, selective, next-generation covalent FGFR inhibitors that can overcome the most common form of kinase inhibitor resistance, the mutation of the so-called “gatekeeper” residue located in the ATP-binding pocket. We also describe a novel kinase inhibitor design strategy that uses a single electrophile to target covalently cysteines that are located in different positions within the ATP-binding pocket. These results have important implications for the design of covalent FGFR inhibitors that can overcome clinical resistance.

Abstract

The human FGF receptors (FGFRs) play critical roles in various human cancers, and several FGFR inhibitors are currently under clinical investigation. Resistance usually results from selection for mutant kinases that are impervious to the action of the drug or from up-regulation of compensatory signaling pathways. Preclinical studies have demonstrated that resistance to FGFR inhibitors can be acquired through mutations in the FGFR gatekeeper residue, as clinically observed for FGFR4 in embryonal rhabdomyosarcoma and neuroendocrine breast carcinomas. Here we report on the use of a structure-based drug design to develop two selective, next-generation covalent FGFR inhibitors, the FGFR irreversible inhibitors 2 (FIIN-2) and 3 (FIIN-3). To our knowledge, FIIN-2 and FIIN-3 are the first inhibitors that can potently inhibit the proliferation of cells dependent upon the gatekeeper mutants of FGFR1 or FGFR2, which confer resistance to first-generation clinical FGFR inhibitors such as NVP-BGJ398 and AZD4547. Because of the conformational flexibility of the reactive acrylamide substituent, FIIN-3 has the unprecedented ability to inhibit both the EGF receptor (EGFR) and FGFR covalently by targeting two distinct cysteine residues. We report the cocrystal structure of FGFR4 with FIIN-2, which unexpectedly exhibits a “DFG-out” covalent binding mode. The structural basis for dual FGFR and EGFR targeting by FIIN3 also is illustrated by crystal structures of FIIN-3 bound with FGFR4 V550L and EGFR L858R. These results have important implications for the design of covalent FGFR inhibitors that can overcome clinical resistance and provide the first example, to our knowledge, of a kinase inhibitor that covalently targets cysteines located in different positions within the ATP-binding pocket.

Continue Reading

Data Availability

Data deposition: Crystallography, atomic coordinates, and structure factors reported in this paper have been deposited in the Protein Data Bank, www.pdb.org (PDB ID codes 4QQC, 4R6V, and 4R5S).

Acknowledgments

We thank Liping Wang for technical assistance and Dr. Sara Buhrlage for proofreading. This work was supported by Lung Specialized Programs of Research Excellence Grant 5 P50 CA090578-10 (to P.A.J. and N.S.G.) and National Institute of Dental and Craniofacial Research Grant R01 DE13686 (to M.M.).

Supporting Information

Appendix (PDF)
Supporting Information

References

1
M Breccia, G Alimena, Nilotinib: A second-generation tyrosine kinase inhibitor for chronic myeloid leukemia. Leuk Res 34, 129–134 (2010).
2
T O’Hare, et al., Inhibition of wild-type and mutant Bcr-Abl by AP23464, a potent ATP-based oncogenic protein kinase inhibitor: Implications for CML. Blood 104, 2532–2539 (2004).
3
F Solca, et al., Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker. J Pharmacol Exp Ther 343, 342–350 (2012).
4
W Zhou, et al., Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature 462, 1070–1074 (2009).
5
KH Tiong, LY Mah, CO Leong, Functional roles of fibroblast growth factor receptors (FGFRs) signaling in human cancers. Apoptosis 18, 1447–1468 (2013).
6
AN Brooks, E Kilgour, PD Smith, Molecular pathways: Fibroblast growth factor signaling: A new therapeutic opportunity in cancer. Clin Cancer Res 18, 1855–1862 (2012).
7
P Richette, T Bardin, C Stheneur, Achondroplasia: From genotype to phenotype. Joint Bone Spine 75, 125–130 (2008).
8
MJ Harding, AV Nechiporuk, Fgfr-Ras-MAPK signaling is required for apical constriction via apical positioning of Rho-associated kinase during mechanosensory organ formation. Development 139, 3130–3135 (2012).
9
S Ota, N Tonou-Fujimori, K Yamasu, The roles of the FGF signal in zebrafish embryos analyzed using constitutive activation and dominant-negative suppression of different FGF receptors. Mech Dev 126, 1–17 (2009).
10
RT Böttcher, C Niehrs, Fibroblast growth factor signaling during early vertebrate development. Endocr Rev 26, 63–77 (2005).
11
MV Dieci, M Arnedos, F Andre, JC Soria, Fibroblast growth factor receptor inhibitors as a cancer treatment: From a biologic rationale to medical perspectives. Cancer Discov 3, 264–279 (2013).
12
A Etienne, et al., Combined translocation with ZNF198-FGFR1 gene fusion and deletion of potential tumor suppressors in a myeloproliferative disorder. Cancer Genet Cytogenet 173, 154–158 (2007).
13
D Singh, et al., Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 337, 1231–1235 (2012).
14
Y Kim, et al., Integrative and comparative genomic analysis of lung squamous cell carcinomas in East Asian patients. J Clin Oncol 32, 121–128 (2014).
15
YM Wu, et al., Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov 3, 636–647 (2013).
16
IJ Majewski, et al., Identification of recurrent FGFR3 fusion genes in lung cancer through kinome-centred RNA sequencing. J Pathol 230, 270–276 (2013).
17
RG Liao, et al., Inhibitor-sensitive FGFR2 and FGFR3 mutations in lung squamous cell carcinoma. Cancer Res 73, 5195–5205 (2013).
18
S Gatius, et al., FGFR2 alterations in endometrial carcinoma. Mod Pathol 24, 1500–1510 (2011).
19
SA Byron, et al., The N550K/H mutations in FGFR2 confer differential resistance to PD173074, dovitinib, and ponatinib ATP-competitive inhibitors. Neoplasia 15, 975–988 (2013).
20
A Roidl, et al., The FGFR4 Y367C mutant is a dominant oncogene in MDA-MB453 breast cancer cells. Oncogene 29, 1543–1552 (2010).
21
YY Shen, et al., Fibroblast growth factor receptor 4 Gly388Arg polymorphism in Chinese gastric cancer patients. World J Gastroenterol 19, 4568–4575 (2013).
22
S Ezzat, et al., The cancer-associated FGFR4-G388R polymorphism enhances pancreatic insulin secretion and modifies the risk of diabetes. Cell Metab 17, 929–940 (2013).
23
6th JG Taylor, et al., Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. J Clin Invest 119, 3395–3407 (2009).
24
M Katoh, H Nakagama, FGF receptors: Cancer biology and therapeutics. Med Res Rev 34, 280–300 (2014).
25
J Weiss, et al., Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med 2, 62ra93 (2010).
26
I Tsimafeyeu, L Demidov, E Stepanova, N Wynn, H Ta, Overexpression of fibroblast growth factor receptors FGFR1 and FGFR2 in renal cell carcinoma. Scand J Urol Nephrol 45, 190–195 (2011).
27
T Sato, et al., Overexpression of the fibroblast growth factor receptor-1 gene correlates with liver metastasis in colorectal cancer. Oncol Rep 21, 211–216 (2009).
28
F Cottoni, et al., Overexpression of the fibroblast growth factor receptor 2-IIIc in Kaposi’s sarcoma. J Dermatol Sci 53, 65–68 (2009).
29
Y Matsuda, M Hagio, T Seya, T Ishiwata, Fibroblast growth factor receptor 2 IIIc as a therapeutic target for colorectal cancer cells. Mol Cancer Ther 11, 2010–2020 (2012).
30
JJ Gómez-Román, et al., Fibroblast growth factor receptor 3 is overexpressed in urinary tract carcinomas and modulates the neoplastic cell growth. Clin Cancer Res 11, 459–465 (2005).
31
BJ Henson, SM Gollin, Overexpression of KLF13 and FGFR3 in oral cancer cells. Cytogenet Genome Res 128, 192–198 (2010).
32
A Kalff, A Spencer, The t(4;14) translocation and FGFR3 overexpression in multiple myeloma: Prognostic implications and current clinical strategies. Blood Cancer J 2, e89 (2012).
33
HK Ho, et al., Fibroblast growth factor receptor 4 regulates proliferation, anti-apoptosis and alpha-fetoprotein secretion during hepatocellular carcinoma progression and represents a potential target for therapeutic intervention. J Hepatol 50, 118–127 (2009).
34
N Motoda, et al., Overexpression of fibroblast growth factor receptor 4 in high-grade pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma. Int J Oncol 38, 133–143 (2011).
35
W Poh, et al., Klotho-beta overexpression as a novel target for suppressing proliferation and fibroblast growth factor receptor-4 signaling in hepatocellular carcinoma. Mol Cancer 11, 1–10 (2012).
36
TM Zaid, et al., Identification of FGFR4 as a potential therapeutic target for advanced-stage, high-grade serous ovarian cancer. Clin Cancer Res 19, 809–820 (2013).
37
YW Ye, et al., Fibroblast growth factor receptor 4 regulates proliferation and antiapoptosis during gastric cancer progression. Cancer 117, 5304–5313 (2011).
38
HJ Lee, et al., Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell 26, 207–221 (2014).
39
V Guagnano, et al., Discovery of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl-1-methyl-urea (NVP-BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. J Med Chem 54, 7066–7083 (2011).
40
B Wasag, E Lierman, P Meeus, J Cools, P Vandenberghe, The kinase inhibitor TKI258 is active against the novel CUX1-FGFR1 fusion detected in a patient with T-lymphoblastic leukemia/lymphoma and t(7;8)(q22;p11). Haematologica 96, 922–926 (2011).
41
G Zhao, et al., A novel, selective inhibitor of fibroblast growth factor receptors that shows a potent broad spectrum of antitumor activity in several tumor xenograft models. Mol Cancer Ther 10, 2200–2210 (2011).
42
PR Gavine, et al., AZD4547: An orally bioavailable, potent, and selective inhibitor of the fibroblast growth factor receptor tyrosine kinase family. Cancer Res 72, 2045–2056 (2012).
43
W Zhou, et al., A structure-guided approach to creating covalent FGFR inhibitors. Chem Biol 17, 285–295 (2010).
44
D Bixby, M Talpaz, Mechanisms of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia and recent therapeutic strategies to overcome resistance. Hematology (Am Soc Hematol Educ Program) 2009, 461–476 (2009).
45
E Weisberg, et al., Discovery of a small-molecule type II inhibitor of wild-type and gatekeeper mutants of BCR-ABL, PDGFRalpha, Kit, and Src kinases: Novel type II inhibitor of gatekeeper mutants. Blood 115, 4206–4216 (2010).
46
CH Yun, et al., The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci USA 105, 2070–2075 (2008).
47
AW Schüttelkopf, DM van Aalten, PRODRG: A tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr D Biol Crystallogr 60, 1355–1363 (2004).
48
V Chell, et al., 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 32, 3059–3070 (2013).
49
N Shukla, et al., Oncogene mutation profiling of pediatric solid tumors reveals significant subsets of embryonal rhabdomyosarcoma and neuroblastoma with mutated genes in growth signaling pathways. Clin Cancer Res 18, 748–757 (2012).
50
D Ang, et al., Novel mutations in neuroendocrine carcinoma of the breast: Possible therapeutic targets. Diagn Mol Pathol, 2014).
51
N Kammasud, et al., Novel inhibitor for fibroblast growth factor receptor tyrosine kinase. Bioorg Med Chem Lett 17, 4812–4818 (2007).
52
A Jansma, et al., Verification of a designed intramolecular hydrogen bond in a drug scaffold by nuclear magnetic resonance spectroscopy. J Med Chem 50, 5875–5877 (2007).
53
DM Goldstein, NS Gray, PP Zarrinkar, High-throughput kinase profiling as a platform for drug discovery. Nat Rev Drug Discov 7, 391–397 (2008).
54
CV Miduturu, et al., High-throughput kinase profiling: A more efficient approach toward the discovery of new kinase inhibitors. Chem Biol 18, 868–879 (2011).
55
CS Lebakken, et al., Development and applications of a broad-coverage, TR-FRET-based kinase binding assay platform. J Biomol Screen 14, 924–935 (2009).
56
M Warmuth, S Kim, XJ Gu, G Xia, F Adrián, Ba/F3 cells and their use in kinase drug discovery. Curr Opin Oncol 19, 55–60 (2007).
57
N Reintjes, et al., Activating somatic FGFR2 mutations in breast cancer. PLoS ONE 8, e60264 (2013).
58
D Li, et al., BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 27, 4702–4711 (2008).
59
E Leproult, S Barluenga, D Moras, JM Wurtz, N Winssinger, Cysteine mapping in conformationally distinct kinase nucleotide binding sites: Application to the design of selective covalent inhibitors. J Med Chem 54, 1347–1355 (2011).
60
Y Liu, NS Gray, Rational design of inhibitors that bind to inactive kinase conformations. Nat Chem Biol 2, 358–364 (2006).
61
CH Yun, et al., Structures of lung cancer-derived EGFR mutants and inhibitor complexes: Mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell 11, 217–227 (2007).
62
S Chang, et al., Design, synthesis, and biological evaluation of novel conformationally constrained inhibitors targeting epidermal growth factor receptor threonine790 → methionine790 mutant. J Med Chem 55, 2711–2723 (2012).
63
JH Dey, et al., Targeting fibroblast growth factor receptors blocks PI3K/AKT signaling, induces apoptosis, and impairs mammary tumor outgrowth and metastasis. Cancer Res 70, 4151–4162 (2010).
64
C Cole, et al., Inhibition of FGFR2 and FGFR1 increases cisplatin sensitivity in ovarian cancer. Cancer Biol Ther 10, 495–504 (2010).
65
JM Sewell, KG Macleod, A Ritchie, JF Smyth, SP Langdon, Targeting the EGF receptor in ovarian cancer with the tyrosine kinase inhibitor ZD 1839 (“Iressa”). Br J Cancer 86, 456–462 (2002).
66
AR Aref, et al., Screening therapeutic EMT blocking agents in a three-dimensional microenvironment. Integr Biol (Camb) 5, 381–389 (2013).
67
N Itoh, The Fgf families in humans, mice, and zebrafish: Their evolutional processes and roles in development, metabolism, and disease. Biol Pharm Bull 30, 1819–1825 (2007).
68
RC Turkington, et al., Fibroblast growth factor receptor 4 (FGFR4): A targetable regulator of drug resistance in colorectal cancer. Cell Death Dis 5, e1046 (2014).
69
S Miura, et al., Fibroblast growth factor 19 expression correlates with tumor progression and poorer prognosis of hepatocellular carcinoma. BMC Cancer 12, 1–15 (2012).
70
A Peláez-García, et al., FGFR4 role in epithelial-mesenchymal transition and its therapeutic value in colorectal cancer. PLoS ONE 8, e63695 (2013).
71
D Agarwal, et al., FGF receptor genes and breast cancer susceptibility: Results from the Breast Cancer Association Consortium. Br J Cancer; kConFab Investigators; Australian Ovarian Cancer Study Group; GENICA Network; TNBCC 110, 1088–1100 (2014).
72
AL Cheng, YC Shen, AX Zhu, Targeting fibroblast growth factor receptor signaling in hepatocellular carcinoma. Oncology 81, 372–380 (2011).
73
DM French, et al., Targeting FGFR4 inhibits hepatocellular carcinoma in preclinical mouse models. PLoS ONE 7, e36713 (2012).
74
F Ito, Foreword. Target therapy for cancer: Anti-cancer drugs targeting growth-factor signaling molecules. Biol Pharm Bull 34, 1773 (2011).
75
ML Slattery, et al., Associations with growth factor genes (FGF1, FGF2, PDGFB, FGFR2, NRG2, EGF, ERBB2) with breast cancer risk and survival: The Breast Cancer Health Disparities Study. Breast Cancer Res Treat 140, 587–601 (2013).
76
SA Kono, ME Marshall, KE Ware, LE Heasley, The fibroblast growth factor receptor signaling pathway as a mediator of intrinsic resistance to EGFR-specific tyrosine kinase inhibitors in non-small cell lung cancer. Drug Resist Updat 12, 95–102 (2009).
77
AL Tai, et al., Co-overexpression of fibroblast growth factor 3 and epidermal growth factor receptor is correlated with the development of nonsmall cell lung carcinoma. Cancer 106, 146–155 (2006).
78
H Terai, et al., Activation of the FGF2-FGFR1 autocrine pathway: A novel mechanism of acquired resistance to gefitinib in NSCLC. Mol Cancer Res 11, 759–767 (2013).
79
KE Ware, et al., Rapidly acquired resistance to EGFR tyrosine kinase inhibitors in NSCLC cell lines through de-repression of FGFR2 and FGFR3 expression. PLoS ONE 5, e14117 (2010).
80
MT Herrera-Abreu, et al., Parallel RNA interference screens identify EGFR activation as an escape mechanism in FGFR3-mutant cancer. Cancer Discov 3, 1058–1071 (2013).
81
J Wang, et al., Ligand-associated ERBB2/3 activation confers acquired resistance to FGFR inhibition in FGFR3-dependent cancer cells. Oncogene, 2014).
82
A Issa, et al., Combinatorial targeting of FGF and ErbB receptors blocks growth and metastatic spread of breast cancer models. Breast Cancer Res 15, 1–16 (2013).
83
T Shimizu, et al., The clinical effect of the dual-targeting strategy involving PI3K/AKT/mTOR and RAS/MEK/ERK pathways in patients with advanced cancer. Clin Cancer Res 18, 2316–2325 (2012).
84
A Wissner, et al., 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 15, 3635–3648 (2007).
85
D Lipson, et al., Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 18, 382–384 (2012).
86
Z Huang, et al., Structural mimicry of a-loop tyrosine phosphorylation by a pathogenic FGF receptor 3 mutation. Structure 21, 1889–1896 (2013).
87
W Minor, M Cymborowski, Z Otwinowski, M Chruszcz, HKL-3000: The integration of data reduction and structure solution—from diffraction images to an initial model in minutes. Acta Crystallogr D Biol Crystallogr 62, 859–866 (2006).
88
PD Adams, et al., PHENIX: A comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr 66, 213–221 (2010).
89
H Chen, et al., A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell 27, 717–730 (2007).
90
P Emsley, B Lohkamp, WG Scott, K Cowtan, Features and development of Coot. Acta Crystallogr D Biol Crystallogr 66, 486–501 (2010).
91
W Kabsch, Solution for Best Rotation to Relate 2 Sets of Vectors. Acta Crystallogr A 32, 922–923 (1976).
92
; Anonymous, The CCP4 suite: Programs for protein crystallography. Acta Crystallogr D Biol Crystallogr; Collaborative Computational Project, Number 4 50, 760–763 (1994).
93
WL DeLano The PyMOL User's Manual (DeLano Scientific, San Carlos, CA, 2002).
94
WA Farahat, et al., Ensemble analysis of angiogenic growth in three-dimensional microfluidic cell cultures. PLoS ONE 7, e37333 (2012).
95
DC Duffy, JC McDonald, OJ Schueller, GM Whitesides, Rapid prototyping of microfluidic systems in poly(dimethylsiloxane). Anal Chem 70, 4974–4984 (1998).

Information & Authors

Information

Published in

Go to Proceedings of the National Academy of Sciences
Proceedings of the National Academy of Sciences
Vol. 111 | No. 45
November 11, 2014
PubMed: 25349422

Classifications

Data Availability

Data deposition: Crystallography, atomic coordinates, and structure factors reported in this paper have been deposited in the Protein Data Bank, www.pdb.org (PDB ID codes 4QQC, 4R6V, and 4R5S).

Submission history

Published online: October 27, 2014
Published in issue: November 11, 2014

Keywords

  1. drug discovery
  2. cancer drug resistance
  3. kinase inhibitor
  4. structure-based drug design

Acknowledgments

We thank Liping Wang for technical assistance and Dr. Sara Buhrlage for proofreading. This work was supported by Lung Specialized Programs of Research Excellence Grant 5 P50 CA090578-10 (to P.A.J. and N.S.G.) and National Institute of Dental and Craniofacial Research Grant R01 DE13686 (to M.M.).

Notes

This article is a PNAS Direct Submission. K.P. is a guest editor invited by the Editorial Board.

Authors

Affiliations

Li Tan1
Departments of aBiological Chemistry and Molecular Pharmacology,
Departments of bCancer Biology and
Jun Wang1
Junko Tanizaki1
Medical Oncology,
The Lowe Center for Thoracic Oncology,
Zhifeng Huang1
Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016;
School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China;
Amir R. Aref1
Genetics, and
Medical Oncology,
Maria Rusan
Medical Oncology,
Department of Clinical Medicine, Aarhus University, Aarhus, 8200 Denmark;
Su-Jie Zhu
Peking University Health Science Center, Beijing, 100191, China;
Yiyun Zhang
Medicine, Harvard Medical School, Boston, MA 02115;
Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129;
Dalia Ercan
Medical Oncology,
The Lowe Center for Thoracic Oncology,
Rachel G. Liao
Medical Oncology,
Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02141;
Marzia Capelletti
Medical Oncology,
The Lowe Center for Thoracic Oncology,
Wenjun Zhou
Departments of aBiological Chemistry and Molecular Pharmacology,
Departments of bCancer Biology and
Wooyoung Hur
Departments of aBiological Chemistry and Molecular Pharmacology,
Departments of bCancer Biology and
Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 136-791 Republic of Korea;
NamDoo Kim
New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 706-010 Republic of Korea; and
Taebo Sim
Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 136-791 Republic of Korea;
Korea University-Korean Institute of Science and Technology Graduate School of Converging Science and Technology, Seoul, 136-713 Republic of Korea
Suzanne Gaudet
Genetics, and
Departments of bCancer Biology and
Center for Systems Cancer Biology, and
David A. Barbie
Jing-Ruey Joanna Yeh
Medicine, Harvard Medical School, Boston, MA 02115;
Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA 02129;
Cai-Hong Yun
Peking University Health Science Center, Beijing, 100191, China;
Peter S. Hammerman2 [email protected]
Medical Oncology,
Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02141;
Moosa Mohammadi2 [email protected]
Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016;
Pasi A. Jänne2 [email protected]
Medical Oncology,
The Lowe Center for Thoracic Oncology,
Belfer Institute for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA 02215;
Nathanael S. Gray2 [email protected]
Departments of aBiological Chemistry and Molecular Pharmacology,
Departments of bCancer Biology and

Notes

2
To whom correspondence may be addressed. Email: [email protected], [email protected], [email protected], or [email protected].
Author contributions: L.T., P.S.H., P.A.J., and N.S.G. designed research; L.T., J.W., J.T., Z.H., A.R.A., M.R., S.-J.Z., Y.Z., D.E., R.G.L., W.Z., W.H., N.K., T.S., S.G., D.A.B., J.-R.J.Y., C.-H.Y., P.S.H., M.M., P.A.J., and N.S.G. performed research; L.T., J.W., J.T., Z.H., A.R.A., M.R., S.-J.Z., Y.Z., D.E., R.G.L., M.C., W.Z., S.G., D.A.B., J.-R.J.Y., C.-H.Y., P.S.H., M.M., P.A.J., and N.S.G. contributed new reagents/analytic tools; L.T., J.W., J.T., Z.H., A.R.A., M.R., Y.Z., D.E., R.G.L., W.H., N.K., T.S., S.G., D.A.B., J.-R.J.Y., C.-H.Y., P.S.H., M.M., P.A.J., and N.S.G. analyzed data; and L.T. and N.S.G. wrote the paper.
1
L.T., J.W., J.T., Z.H., and A.R.A. contributed equally to this work.

Competing Interests

The authors declare no conflict of interest.

Metrics & Citations

Metrics

Note: The article usage is presented with a three- to four-day delay and will update daily once available. Due to ths delay, usage data will not appear immediately following publication. Citation information is sourced from Crossref Cited-by service.


Citation statements

Altmetrics

Citations

Export the article citation data by selecting a format from the list below and clicking Export.

Cited by

    Loading...

    View Options

    View options

    PDF format

    Download this article as a PDF file

    DOWNLOAD PDF

    Login options

    Check if you have access through your login credentials or your institution to get full access on this article.

    Personal login Institutional Login

    Recommend to a librarian

    Recommend PNAS to a Librarian

    Purchase options

    Purchase this article to access the full text.

    Single Article Purchase

    Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors
    Proceedings of the National Academy of Sciences
    • Vol. 111
    • No. 45
    • pp. 15853-16225

    Media

    Figures

    Tables

    Other

    Share

    Share

    Share article link

    Share on social media

    Further reading in this issue