Little progress has been made in the treatment of acute myeloid leukemia (AML) despite dose intensification of cytotoxic chemotherapy. An alternative approach to treating AML is the incorporation of pro-differentiation agents into standard chemotherapy regimens. In order to identify new AML differentiation agents, our laboratory developed a gene expression-based approach to small molecule screening. We identified gefitinib, an epidermal growth factor (EGFR) inhibitor, as an inducer of AML differentiation. EGFR is not expressed in the tested AML cell lines, thus precluding inhibition of this kinase as the mechanism of AML differentiation. Because multiple EGFR inhibitors induce the phenotype, we hypothesize that a shared off-target kinase is the target in AML differentiation. In order to identify candidate gefitinib targets of AML differentiation, we utilized proteomic and genetic approaches. Spleen tyrosine kinase (Syk) was identified as the top candidate. Syk is a nonreceptor tyrosine kinase, important in normal B-cell differentiation, and implicated in hematological malignancies. We hypothesize that Syk is a target for AML therapy and that loss of Syk will result in differentiation and/or cell death in AML. We confirmed with both pharmacological inhibition (R406) and genetic loss of Syk the induction of differentiation and/or death in a pilot study of AML cells. We now propose to more broadly test this hypothesis with the following Specific Aims.
Specific Aim 1. Characterize the in vitro and in vivo effects of R406 in AML Specific Aim 2. Establish that Syk is the target of R406 activity in AML Specific Aim 3. Determine the downstream effectors of Syk in AML In Aim 1, we will determine the broad potential of Syk inhibition as an anti-AML therapy. We will measure the in vitro effects of a Syk inhibitor (R406) in a large panel of AML cells on differentiation, cell growth, and apoptosis. We will then extend testing to in vivo studies using primary human AML orthotopic models. Next, in Aim 2, we will confirm that Syk is the target of R406 activity with three parallel approaches: A PCR mutagenesis screen for Syk mutants that rescue the effects of R406 anti-AML activity;a genetic approach using RNA interference, and a pharmacological approach evaluating other small molecule inhibitors of Syk.
In Aim 3, we will determine which proteins are critical downstream effectors of Syk signaling in AML using complementary approaches: biochemical, genetic, genomic, pharmacological, and proteomic. The Rigel Pharmaceutical compound, R788, the prodrug of R406, is already in Phase II testing and was recently demonstrated to have activity in autoimmune disease and lymphoma. With Phase I testing now complete and efficacy demonstrated for these diseases, we would be well positioned to rapidly bring R788 to clinical trial. These studies, within the five year time frame of this grant, will have immediate translational relevance and inform the development of a clinical trial testing Syk inhibition in patients with relapsed/refractory AML.

Public Health Relevance

The studies in this proposal will validate the protein spleen tyrosine kinase (Syk) as a target for acute myeloid leukemia (AML) therapy. We will test pharmacological and genetic inhibition of Syk in AML cells in vitro and in vivo models of AML. Because there is already a drug with anti-Syk activity with demonstrated efficacy in lymphoma and autoimmune disease, we anticipate the rapid translation of these studies to a clinical trial testing Syk inhibition for patients with AML.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA140292-05
Application #
8463140
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Arya, Suresh
Project Start
2009-07-07
Project End
2014-04-30
Budget Start
2013-05-01
Budget End
2014-04-30
Support Year
5
Fiscal Year
2013
Total Cost
$331,098
Indirect Cost
$141,899
Name
Dana-Farber Cancer Institute
Department
Type
DUNS #
076580745
City
Boston
State
MA
Country
United States
Zip Code
02215
Wagner, Florence F; Benajiba, Lina; Campbell, Arthur J et al. (2018) Exploiting an Asp-Glu ""switch"" in glycogen synthase kinase 3 to design paralog-selective inhibitors for use in acute myeloid leukemia. Sci Transl Med 10:
Mohr, Sebastian; Doebele, Carmen; Comoglio, Federico et al. (2017) Hoxa9 and Meis1 Cooperatively Induce Addiction to Syk Signaling by Suppressing miR-146a in Acute Myeloid Leukemia. Cancer Cell 31:549-562.e11
Pikman, Yana; Puissant, Alexandre; Alexe, Gabriela et al. (2016) Targeting MTHFD2 in acute myeloid leukemia. J Exp Med 213:1285-306
Lindblad, O; Cordero, E; Puissant, A et al. (2016) Aberrant activation of the PI3K/mTOR pathway promotes resistance to sorafenib in AML. Oncogene 35:5119-31
Boros, Katalin; Puissant, Alexandre; Back, Morgan et al. (2015) Increased SYK activity is associated with unfavorable outcome among patients with acute myeloid leukemia. Oncotarget 6:25575-87
Neumann, Theresa; Benajiba, Lina; Göring, Stefan et al. (2015) Evaluation of Improved Glycogen Synthase Kinase-3? Inhibitors in Models of Acute Myeloid Leukemia. J Med Chem 58:8907-19
Deangelo, Daniel J; Neuberg, Donna; Amrein, Philip C et al. (2014) A phase II study of the EGFR inhibitor gefitinib in patients with acute myeloid leukemia. Leuk Res 38:430-4
Puissant, Alexandre; Fenouille, Nina; Alexe, Gabriela et al. (2014) SYK is a critical regulator of FLT3 in acute myeloid leukemia. Cancer Cell 25:226-42
Carnevale, J; Ross, L; Puissant, A et al. (2013) SYK regulates mTOR signaling in AML. Leukemia 27:2118-28
Banerji, Versha; Frumm, Stacey M; Ross, Kenneth N et al. (2012) The intersection of genetic and chemical genomic screens identifies GSK-3? as a target in human acute myeloid leukemia. J Clin Invest 122:935-47