Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.


Abstract

Effective targeted cancer therapeutic development depends upon distinguishing disease-associated 鈥榙river鈥?mutations, which have causative roles in malignancy pathogenesis, from 鈥榩assenger鈥?mutations, which are dispensable for cancer initiation and maintenance. Translational studies of clinically active targeted therapeutics can definitively discriminate driver from passenger lesions and provide valuable insights into human cancer biology. Activating internal tandem duplication (ITD) mutations in FLT3 (FLT3-ITD) are detected in approximately 20% of acute myeloid leukaemia (AML) patients and are associated with a poor prognosis1. Abundant scientific2 and clinical evidence1,3, including the lack of convincing clinical activity of early FLT3 inhibitors4,5, suggests that FLT3-ITD probably represents a passenger lesion. Here we report point mutations at three residues within the kinase domain of FLT3-ITD that confer substantial in vitro resistance to AC220 (quizartinib), an active investigational inhibitor of FLT3, KIT, PDGFRA, PDGFRB and RET6,7; evolution of AC220-resistant substitutions at two of these amino acid positions was observed in eight of eight FLT3-ITD-positive AML patients with acquired resistance to AC220. Our findings demonstrate that FLT3-ITD can represent a driver lesion and valid therapeutic target in human AML. AC220-resistant FLT3 kinase domain mutants represent high-value targets for future FLT3 inhibitor development efforts.


Subscription info for Chinese customers

We have a dedicated website for our Chinese customers. Please go to naturechina.com to subscribe to this journal.

Go to naturechina.com

Rent or Buy article

Get time limited or full article access on ReadCube.

from$8.99

Rent or Buy

All prices are NET prices.


Data deposits

SMRT sequencing data is deposited online at http:// www.ncbi.nlm.nih.gov/sra under accession number SRA050226.1.

References1

Thiede, C. et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 99, 4326鈥?335 (2002)

CASArticle Google Scholar2

Lee, B. H. et al. FLT3 internal tandem duplication mutations induce myeloproliferative or lymphoid disease in a transgenic mouse model. Oncogene 24, 7882鈥?892 (2005)

CASArticle Google Scholar3

Shih, L. Y. et al. Acquisition of FLT3 or N-ras mutations is frequently associated with progression of myelodysplastic syndrome to acute myeloid leukemia. Leukemia 18, 466鈥?75 (2004)

CASArticle Google Scholar4

Knapper, S. et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 108, 3262鈥?270 (2006)

CASArticle Google Scholar5

Fischer, T. et al. Phase IIB trial of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J. Clin. Oncol. 28, 4339鈥?345 (2010)

CASArticle Google Scholar6

Zarrinkar, P. P. et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood 114, 2984鈥?992 (2009)

CASArticle Google Scholar7

Cortes, J. et al. in 16th Congress of the European Hematology Association (Haematologica, 2011)

8

Heidel, F. et al. Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood 107, 293鈥?00 (2006)

CASArticle Google Scholar9

Metzelder, S. et al. Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation. Blood 113, 6567鈥?571 (2009)

CASArticle Google Scholar10

Scholl, S. et al. Secondary resistance to sorafenib in two patients with acute myeloid leukemia (AML) harboring FLT3-ITD mutations. Ann. Hematol. 90, 473鈥?75 (2011)

Article Google Scholar11

Azam, M., Latek, R. R. Daley, G. Q. Mechanisms of autoinhibition and STI-571/imatinib resistance revealed by mutagenesis of BCR-ABL. Cell 112, 831鈥?43 (2003)

CASArticle Google Scholar12

Shah, N. P. et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2, 117鈥?25 (2002)

CASArticle Google Scholar13

Travers, K. J., Chin, C. S., Rank, D. R., Eid, J. S. Turner, S. W. A flexible and efficient template format for circular consensus sequencing and SNP detection. Nucleic Acids Res. 38, e159 (2010)

Article Google Scholar14

Griffith, J. et al. The structural basis for autoinhibition of FLT3 by the juxtamembrane domain. Mol. Cell 13, 169鈥?78 (2004)

CASArticle Google Scholar15

Levinson, N. M. et al. A Src-like inactive conformation in the Abl tyrosine kinase domain. PLoS Biol. 4, e144 (2006)

Article Google Scholar16

Mol, C. D. et al. Structural basis for the autoinhibition and STI-571 inhibition of c-Kit tyrosine kinase. J. Biol. Chem. 279, 31655鈥?1663 (2004)

CASArticle Google Scholar17

Hubbard, S. R., Wei, L., Ellis, L. Hendrickson, W. A. Crystal structure of the tyrosine kinase domain of the human insulin receptor. Nature 372, 746鈥?54 (1994)

CASADSArticle Google Scholar18

Mol, C. D. et al. Structure of a c-Kit product complex reveals the basis for kinase transactivation. J. Biol. Chem. 278, 31461鈥?1464 (2003)

CASArticle Google Scholar19

Hubbard, S. R. Crystal structure of the activated insulin receptor tyrosine kinase in complex with peptide substrate and ATP analog. EMBO J. 16, 5572鈥?581 (1997)

CASArticle Google Scholar20

Wodicka, L. M. et al. Activation state-dependent binding of small molecule kinase inhibitors: structural insights from biochemistry. Chem. Biol. 17, 1241鈥?249 (2010)

CASArticle Google Scholar21

Burley, S. K. Petsko, G. A. Aromatic-aromatic interaction: a mechanism of protein structure stabilization. Science 229, 23鈥?8 (1985)

CASADSArticle Google Scholar22

Wan, P. T. et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116, 855鈥?67 (2004)

CASArticle Google Scholar23

Choudhary, C. et al. Activation mechanisms of STAT5 by oncogenic Flt3-ITD. Blood 110, 370鈥?74 (2007)

CASArticle Google Scholar24

Pao, W. et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2, e73 (2005)

Article Google Scholar25

Barbie, D. A. Deangelo, D. J. Systemic mastocytosis: current classification and novel therapeutic options. Clin. Adv. Hematol. Oncol. 4, 768鈥?75 (2006)

PubMed Google Scholar26

Anderson, K. et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature 469, 356鈥?61 (2011)

CASADSArticle Google Scholar27

Kralovics, R. et al. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood 108, 1377鈥?380 (2006)

CASArticle Google Scholar28

Nazarian, R. et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 468, 973鈥?77 (2010)

CASADSArticle Google Scholar29

Benson, G. Tandem repeats finder: a program to analyze DNA sequences. Nucleic Acids Res. 27, 573鈥?80 (1999)

CASArticle Google Scholar30

Yates, F. Tests of significance for 2鈥壝椻€? contingency tables. J. R. Stat. Soc. A 147, 426鈥?63 (1984)

MathSciNetArticle Google Scholar31

Barnard, G. A. Must clinical trials be large? The interpretation of p-values and the combination of test results. Stat. Med. 9, 601鈥?14 (1990)

CASArticle Google Scholar32

Morris, G. M. et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J. Comput. Chem. 30, 2785鈥?791 (2009)

CASArticle Google Scholar33

Sch眉ttelkopf, A. W. van Aalten, D. M. PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D 60, 1355鈥?363 (2004)

Article Google Scholar

Download references

Acknowledgements

We thank K. Lin for technical assistance. This work was funded by grants from the Leukemia and Lymphoma Society (to C.C.S. and N.P.S.), the Doris Duke Charitable Foundation (to N.P.S.), NCI Leukemia SPORE P50 CA100632-06 (to M.J.L.), NCI R01 CA12886 (to M.J.L.) and the NIH T-32 Molecular Mechanisms of Cancer (to C.C.S.). C.C.S. would like to acknowledge the EHA/ASH Translational Research Training Institute. N.P.S. would like to thank Art and Alison Kern and the Edward S. Ageno family for their support.

Author informationAuthor notesJeremy P. Hunt,聽Patrick P. Zarrinkar,聽Eric E. Schadt聽 聽Andrew Kasarskis

Present address: Present addresses: KINOMEscan Division of DiscoveRx Corporation, San Diego, California 92121, USA (J.P.H.); Blueprint Medicines Corporation, Cambridge, Massachusetts 02142, USA (P.P.Z.); Institute for Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York 10029, USA (E.E.S. and A.K.).,

AffiliationsDivision of Hematology/Oncology, University of California, San Francisco, 94143, California, USACatherine C. Smith,聽Sara Salerno,聽Lauren E. Damon聽 聽Neil P. ShahDepartment of Molecular and Cell Biology, University of California, Berkeley, 94720, California, USAQi Wang聽 聽John KuriyanPacific Biosciences, Menlo Park, 94025, California, USAChen-Shan Chin,聽Kevin J. Travers,聽Susana Wang,聽Eric E. Schadt聽 聽Andrew KasarskisDepartment of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, 21231, Maryland, USAMark J. LevisAbramson Cancer Center of the University of Pennsylvania, Philadelphia, 19104, Pennsylvania, USAAlexander E. PerlAmbit Biosciences, San Diego, 92121, California, USAJeremy P. Hunt聽 聽Patrick P. ZarrinkarHelen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 94115, California, USANeil P. ShahAuthorsCatherine C. SmithView author publicationsYou can also search for this author in PubMed Google Scholar Qi WangView author publicationsYou can also search for this author in PubMed Google Scholar Chen-Shan ChinView author publicationsYou can also search for this author in PubMed Google Scholar Sara SalernoView author publicationsYou can also search for this author in PubMed Google Scholar Lauren E. DamonView author publicationsYou can also search for this author in PubMed Google Scholar Mark J. LevisView author publicationsYou can also search for this author in PubMed Google Scholar Alexander E. PerlView author publicationsYou can also search for this author in PubMed Google Scholar Kevin J. TraversView author publicationsYou can also search for this author in PubMed Google Scholar Susana WangView author publicationsYou can also search for this author in PubMed Google Scholar Jeremy P. HuntView author publicationsYou can also search for this author in PubMed Google Scholar Patrick P. ZarrinkarView author publicationsYou can also search for this author in PubMed Google Scholar Eric E. SchadtView author publicationsYou can also search for this author in PubMed Google Scholar Andrew KasarskisView author publicationsYou can also search for this author in PubMed Google Scholar John KuriyanView author publicationsYou can also search for this author in PubMed Google Scholar Neil P. ShahView author publicationsYou can also search for this author in PubMed Google Scholar Contributions

C.C.S., Q.W., C.-S.C., K.J.T., A.K., E.E.S. and J.K. designed experiments, performed research, analysed data and wrote the manuscript. N.P.S. designed experiments, analysed data and wrote the manuscript. L.E.D., S.W., J.P.H. and S.S. performed experiments and reviewed the manuscript. P.P.Z. was involved in study design and reviewed the manuscript. A.E.P. and M.J.L. provided reagents, performed research and reviewed the manuscript.

Corresponding author

Correspondence to Neil P. Shah.

Ethics declarations Competing interests

The following authors declare competing financial interests: C.C.S. (consultancy, Astellas Pharma); M.J.L. (consultancy, Ambit Biosciences); J.P.H. and P.P.Z. (employment, Ambit Biosciences) E.E.S. and A.K. (equity ownership and employment, Pacific Biosciences); C.-S.C., K.J.T. and S.W. (employment, Pacific Biosciences).

Supplementary information Supplementary Information

This file contains Supplementary Figures 1-6 and Supplementary Tables 1-5. (PDF 1527 kb)

PowerPoint slides
PowerPoint slide for Fig. 1PowerPoint slide for Fig. 2Rights and permissions

Reprints and Permissions

About this articleCite this article

Smith, C., Wang, Q., Chin, C. et al. Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature 485, 260鈥?63 (2012). https://doi.org/10.1038/nature11016

Download citation

Received: 25 August 2011

Accepted: 05 March 2012

Published: 15 April 2012

Issue Date: 10 May 2012

DOI: https://doi.org/10.1038/nature11016


Sarah Bertoli, Pierre-Yves Dumas, Emilie B茅rard, Laetitia Largeaud, Audrey Bidet, Eric Delabesse, Suzanne Tavitian, No茅mie Gadaud, Thibaut Leguay, Harmony Leroy, Jean-Baptiste Rieu, Jean-Philippe Vial, Fran莽ois Vergez, Nicolas Lechevalier, Isabelle Luquet, Emilie Klein, Audrey Sarry, Anne-Charlotte De Grande, Christian R茅cher Arnaud Pigneux Cancers (2020)
Jiyin Bao, Haichun Liu, Yanle Zhi, Wenqianzi Yang, Jiawei Zhang, Tao Lu, Yue Wang Shuai Lu Bioorganic Chemistry (2020)
Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.


Editorial SummaryFLT3 a target in leukaemia

Activating internal tandem duplication FLT3 mutations are relatively frequent in acute myeloid leukaemia (AML). Although they are associated with poor prognosis, it has remained unclear whether they play a causal part in the development of AML. In a study of AC220 (quizartinib), an FLT3 kinase inhibitor that is currently being evaluated in clinical trials for people with AML, Neil Shah and colleagues have identified secondary mutations in FLT3 that confer resistance to AC220. The mutations are present in cell lines and in people treated with AC220 who have relapsed after an initial response, and seem to act by preventing FLT3 inhibitor binding. This study establishes FLT3 as a potential target in a subset of AML cases.

\"Advertisement\"