FLT3 inhibition and mechanisms of drug resistance in mutant FLT3-positive AML

https://doi.org/10.1016/j.drup.2009.04.001Get rights and content

Abstract

An appealing therapeutic target in AML is constitutively activated, mutant FLT3, which is expressed in a subpopulation of AML patients and is generally a poor prognostic indicator in patients under the age of 65. There are currently several FLT3 inhibitors that are undergoing clinical investigation. However, the discovery of drug-resistant leukemic blast cells in FLT3 inhibitor-treated AML patients has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to circumvent drug resistance. Here, we provide an overview of FLT3 inhibitors under preclinical and clinical investigation, and we discuss mechanisms whereby AML cells develop resistance to FLT3 inhibitors, and the ways in which combination therapy could potentially be utilized to override drug resistance. We discuss how the cross-talk between major downstream signaling pathways, such as PI3K/PTEN/Akt/mTOR, RAS/Raf/MEK/ERK, and Jak/STAT, can be exploited for therapeutic purposes by targeting key signaling molecules with selective inhibitors, such as mTOR inhibitors, HSP90 inhibitors, or farnesyltransferase inhibitors, and identifying those agents with the ability to positively combine with inhibitors of FLT3, such as PKC412 and sunitinib. With the widespread onset of drug resistance associated with tyrosine kinase inhibitors, due to mechanisms involving development of point mutations or gene amplification of target proteins, the use of a multi-targeted therapeutic approach is of potential clinical benefit.

Introduction

There are around 15,000 newly diagnosed acute myelocytic leukemia (AML) patients in the US each year. This hematopoietic malignancy is characterized by aberrant proliferation of myeloid progenitor cells, coupled by a partial block in cellular differentiation (McKenzie, 2005). Permeation of bone marrow and peripheral blood with immature leukemic myeloblasts is the outcome of the abnormal survival advantage of leukemic cells, and causes such symptoms as bleeding, anemia, and infection.

Current therapies for AML often do not succeed because of therapy-induced mortality or drug resistance (Estey, 2001). The use of conventional chemotherapeutic agents as a single treatment approach is coupled to a low therapy-induced mortality, however a high risk of relapse due to drug resistance (Mathews and DiPersio, 2004). In contrast, allogeneic transplantation (alloBMT) has a high therapy-induced mortality, and yet a lower risk of relapse (Mathews and DiPersio, 2004). Due to the fact that alloBMT shows more promise in younger patients, it has an overall small impact on the majority of AML patients, who tend to be aged 65 and older (Witherspoon and Deeg, 1999).

In AML, the activation of signaling pathways results from a range of genetic modifications leading to mutation of signaling molecules, such as receptor tyrosine kinases. Approximately 30% of AML patients, as well as a portion of ALL patients, harbor a mutant form of the class III receptor tyrosine kinase, FLT3 (Fms-Like Tyrosine kinase-3; STK-1, human Stem Cell Tyrosine Kinase-1; or FLK-2, Fetal Liver Kinase-2) (Stirewalt and Radich, 2003). Internal tandem duplications (ITD) within the juxtamembrane domain represent the most common form of constitutively activated FLT3, occurring in approximately 20–25% of AML patients and in less than 5% of myelodysplastic syndrome (MDS) patients (Nakao et al., 1996, Horiike et al., 1997, Kiyoi et al., 1998, Kondo et al., 1999, Rombouts et al., 2000, Kelly et al., 2002a, Kelly et al., 2002b). Indeed, a rapidly lethal myeloproliferative disorder in mice results from the in vivo transplantation of murine bone marrow cells infected with a FLT3-ITD-expressing retrovirus (Kelly et al., 2002a, Kelly et al., 2002b).

Also identified in AML patients are point mutations within the “activation loop” of FLT3 (Yamamoto et al., 2001). For example, a missense mutation in the aspartic acid residue at position 835 is believed to induce the activation loop into an “activated” configuration. Additional, albeit less prevalent, mutations in the kinase domain includes N841I (Jiang et al., 2004) and Y842C (Kindler et al., 2005).

Generally, the existence of a FLT3-ITD mutation translates into a poorer prognosis in both disease-free survival and overall survival (Mattison et al., 2007). In fact, patients harboring both a nucleophosmin 1 (NMP1) mutation, which is typically a positive prognostic indicator, and mutant FLT3 tend to have poorer outcomes (Mattison et al., 2007).

There are several inhibitors of FLT3 currently in clinical trials, and a number of novel inhibitors under preclinical investigation. However, the FLT3 inhibitors tested thus far clinically generally induce only partial and transient responses in patients when used as single agents. This suggests a need for the development of novel agents conferring higher potency and/or less toxicity that can either be used effectively as single agents or that can be effectively combined with other agents to suppress disease progression and prolong the lifespan of patients.

In addition to identifying and developing potent FLT3 inhibitors representative of novel and unique structural classes, there is a push toward gaining a better understanding of the mechanisms underlying drug resistance in AML. Clinical trial data with tyrosine kinase inhibitors show that while the peripheral blood blasts decline well, bone marrow responses are less common. Stromal cells have been implicated in this mode of resistance, as they provide viability signals to leukemic cells that protect them from the effects of the inhibitor. Other mechanisms of drug resistance include the emergence of point mutations in the target protein, and deregulation of signaling molecules associated with apoptotic signaling leading to a survival advantage in leukemic cells.

There are several strategies that may be effective in preventing relapse due to the emergence of point mutations in target proteins, as well as in overcoming drug resistance believed to be caused by stromal-mediated viability signals or deregulation of apoptotic signaling. These include the combined use of more than one FLT3 inhibitor, providing their interaction with FLT3 signaling or the FLT3 protein target is distinct enough for the two inhibitors to synergize. Alternatively, FLT3 inhibitors can be combined with small molecule inhibitors that interact with key components of major signaling pathways that play a significant role in AML. Finally, FLT3 inhibitors can be combined with standard chemotherapy as an approach to achieve maximum efficacy in patients.

Section snippets

Classes of FLT3 inhibitors

The structural classes of prominent FLT3 inhibitors in clinical trials or under preclinical investigation are shown in Fig. 1. The N-indolocarbazole PKC412 (midostaurin; N-benzoyl-staurosporine; Novartis Pharma AG) is one of several FLT3 inhibitors that is undergoing clinical testing, and which is currently in late-stage clinical trials. PKC412 is a broad spectrum, orally bioavailable inhibitor of FLT3, as well as platelet-derived growth factor β (PDGFRβ), c-KIT, and c-FMS (Weisberg et al., 2002

Clinical resistance to FLT3 inhibition

While small molecule inhibitors of FLT3 are showing promise clinically for AML, thus far none has elicited sustained cytogenic responses as a single agent. For instance, quantitative measurement of FLT3 inhibition in patients treated with KW-2449 in a Phase I trial showed that inhibition of FLT3 occurred transiently to less than 20% of baseline (Pratz et al., 2009). It is possible that such incomplete and only temporary inhibition of FLT3 can be generalized to other FLT3 inhibitors under

Combination therapy

The FLT3 inhibitors tested thus far generally induce only partial and transient responses in patients when administered as single agents. Thus, there is a need for the discovery and development of novel, more efficacious and less toxic inhibitors of FLT3 that could potentially be used effectively as single agents. There is also a need to test these, as well as FLT3 inhibitors under investigation, in combination with other therapeutics already in clinical use for leukemia.

Detection of

Conclusion

There is an urgent need for development of new treatment strategies that could lead to improved therapeutic efficacy in AML patients. Existing therapeutic approaches include the discovery and development of novel agents with unique structures conferring higher potency and selectivity toward FLT3 as a target. Such characteristics may allow for more complete inhibition of the FLT3 kinase protein target as compared to that of existing therapies in preclinical and clinical development. Elucidation

References (110)

  • J. Jiang et al.

    Identifying and characterizing a novel activating mutation of the FLT3 tyrosine kinase in AML

    Blood

    (2004)
  • R.K. Kancha et al.

    Sensitivity toward sorafenib and sunitinib varies between different activating and drug-resistant FLT3-ITD mutations

    Exp. Hematol.

    (2007)
  • S. Kasper et al.

    The kinase inhibitor LS104 induces apoptosis, enhances cytotoxic effects of chemotherapeutic drugs and is targeting the receptor tyrosine kinase FLT3 in acute myeloid leukemia

    Leuk. Res.

    (2008)
  • L.M. Kelly et al.

    FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model

    Blood

    (2002)
  • L.M. Kelly et al.

    CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML)

    Cancer Cell

    (2002)
  • T. Kindler et al.

    Identification of a novel activating mutation (Y842C) within the activation loop of FLT3 in patients with acute myeloid leukemia (AML)

    Blood

    (2005)
  • S. Knapper 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

    (2006)
  • S. Knapper et al.

    The effects of lestaurtinib (CEP701) and PKC412 on primary AML blasts: the induction of cytotoxicity varies with dependence on FLT3 signaling in both FLT3-mutated and wild-type cases

    Blood

    (2006)
  • S.M. Kornblau et al.

    Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia

    Blood

    (2006)
  • M. Levis et al.

    A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo

    Blood

    (2002)
  • M. Levis et al.

    In vitro studies of a FLT3 inhibitor combined with chemotherapy: sequence of administration is important to achieve synergistic cytotoxic effects

    Blood

    (2004)
  • M. Levis et al.

    A FLT3 tyrosine kinase inhibitor is selectively cytotoxic to acute myeloid leukemia blasts harboring FLT3 internal tandem duplication mutations

    Blood

    (2001)
  • J. LoPiccolo et al.

    Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations

    Drug Resist. Updates

    (2008)
  • K. Murata et al.

    Selective cytotoxic mechanism of GTP-14564, a novel tyrosine kinase inhibitor in leukemia cells expressing a constitutively active Fms-like tyrosine kinase 3 (FLT3)

    J. Biol. Chem.

    (2003)
  • U. Nishiyama et al.

    Antineoplastic effect of a single oral dose of the novel Flt3 inhibitor KRN383 on xenografted human leukemic cells harboring Flt3-activating mutations

    Leuk. Res.

    (2006)
  • C. Nishioka et al.

    Ki11502, a novel multitargeted receptor tyrosine kinase inhibitor, induces growth arrest and apoptosis of human leukemia cells in vitro and in vivo

    Blood

    (2008)
  • C. Nishioka et al.

    Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene

    Leuk. Res.

    (2008)
  • C. Nishioka et al.

    MS-275, a novel histone deacetylase inhibitor with selectivity against HDAC1, induces degradation of FLT3 via inhibition of chaperone function of heat shock protein 90 in AML cells

    Leuk. Res.

    (2008)
  • A.M. O’Farrell et al.

    SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo

    Blood

    (2003)
  • A.M. O’Farrell et al.

    Effects of SU5416, a small molecule tyrosine kinase receptor inhibitor, on FLT3 expression and phosphorylation in patients with refractory acute myeloid leukemia

    Leuk. Res.

    (2004)
  • K.W. Pratz et al.

    A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response

    Blood

    (2009)
  • C. Recher et al.

    Antileukemic activity of rapamycin in acute myeloid leukemia

    Blood

    (2005)
  • J.L. Rocnik et al.

    Roles of tyrosine 589 and 591 in STAT5 activation and transformation mediated by FLT3-ITD

    Blood

    (2006)
  • N.P. Shah et al.

    Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia

    Cancer Cell

    (2002)
  • D.B. Shankar et al.

    ABT-869, a multitargeted receptor tyrosine kinase inhibitor: inhibition of FLT3 phosphorylation and signaling in acute myeloid leukemia

    Blood

    (2007)
  • Y. Shiotsu

    KW-2449, a novel multi-kinase inhibitor against FLT3, Abl, FGFR1 and aurora suppresses the growth of AML both in vitro and in vivo

    Blood

    (2007)
  • B.D. Smith et al.

    Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia

    Blood

    (2004)
  • R.M. Stone et al.

    Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412

    Blood

    (2005)
  • R.M. Stone et al.

    Phase 1B study of PKC412, an oral FLT3 kinase inhibitor, in sequential and simultaneous combinations with daunorubicin and cytarabine (DA) induction and high-dose cytarabine consolidation in newly diagnosed patients with AML

    Blood

    (2005)
  • S. Trudel et al.

    CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma

    Blood

    (2005)
  • E. Weisberg et al.

    Potentiation of antileukemic therapies by the dual PI3K/PDK-1 inhibitor. BAG956: effects on BCR-ABL and mutant FLT3-expressing cells

    Blood

    (2008)
  • E. Weisberg et al.

    Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412

    Cancer Cell

    (2002)
  • E. Weisberg et al.

    Antileukemic effects of the novel, mutant FLT3 inhibitor NVP-AST487: effects on PKC412-sensitive and -resistant FLT3-expressing cells

    Blood

    (2008)
  • D.H. Albert et al.

    Preclinical activity of ABT-869: a multitargeted receptor tyrosine kinase inhibitor

    Mol. Cancer Ther.

    (2006)
  • L. Al Shaer et al.

    Heat shock protein 90 inhibition is cytotoxic to primary AML cells expressing mutant FLT3 and results in altered downstream signaling

    Br. J. Haematol.

    (2008)
  • D. Auclair et al.

    Antitumor activity of sorafenib in FLT3-driven leukemic cells

    Leukemia

    (2007)
  • C.H. Brandts et al.

    Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation

    Cancer Res.

    (2005)
  • Y. Cheng et al.

    Tandutinib, an oral, small-molecule inhibitor of FLT3 for the treatment of AML and other cancer indications

    IDrugs

    (2008)
  • J. Cools et al.

    A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome

    New Engl. J. Med.

    (2003)
  • J. Cortes et al.

    A phase I dose escalation study of KW-2449, an oral multi-kinase inhibitor against FLT3, Abl, FGFR1 and aurora in patients with relapsed/refractory AML, ALL and MDS or resistant/intolerant CML

    Blood

    (2008)
  • Cited by (93)

    • Combining structure- and property-based optimization to identify selective FLT3-ITD inhibitors with good antitumor efficacy in AML cell inoculated mouse xenograft model

      2019, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Moreover, compound 46 almost showed no inhibitory activity against CDKs that were among the targets of FN-1501. As reported [31,32], some FLT3 inhibitors were subjected to drug resistance due to FLT3 mutation. Therefore, we tested inhibitory activities of compound 46 against a variety of commonly occurred FLT3 mutants.

    View all citing articles on Scopus
    View full text