Next Article in Journal
Pterygium and Ocular Surface Squamous Neoplasia: Optical Biopsy Using a Novel Autofluorescence Multispectral Imaging Technique
Next Article in Special Issue
Are We Moving the Needle for Patients with TP53-Mutated Acute Myeloid Leukemia?
Previous Article in Journal
Targeting an MDM2/MYC Axis to Overcome Drug Resistance in Multiple Myeloma
Previous Article in Special Issue
Comparison of Melphalan Combined with Treosulfan or Busulfan as High-Dose Chemotherapy before Autologous Stem Cell Transplantation in AML
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Multi-Kinase Inhibitor EC-70124 Is a Promising Candidate for the Treatment of FLT3-ITD-Positive Acute Myeloid Leukemia

by Belen Lopez-Millan 1,2,3,*, Paula Costales 4, Francisco Gutiérrez-Agüera 1, Rafael Díaz de la Guardia 1,2, Heleia Roca-Ho 1, Meritxell Vinyoles 1,3, Alba Rubio-Gayarre 1,2, Rémi Safi 1,3, Julio Castaño 1, Paola Alejandra Romecín 1,3, Manuel Ramírez-Orellana 3,5, Eduardo Anguita 6, Irmela Jeremias 7, Lurdes Zamora 1,8, Juan Carlos Rodríguez-Manzaneque 2, Clara Bueno 1,3,9, Francisco Morís 4 and Pablo Menendez 1,3,9,10,*
1
Department of Biomedicine, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
2
GENYO, Centre for Genomics and Oncological Research, Pfizer, Universidad de Granada, Junta de Andalucía, 18016 Granada, Spain
3
Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
4
EntreChemSL, 33011 Oviedo, Spain
5
Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
6
Servicio de Hematología, Hospital Clínico San Carlos, IdISSC, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
7
Research Unit Apoptosis in Hematopoietic Stem Cells, Helmholtz Zentrum München, 85764 Munich, Germany
8
Hematology Department, ICO-Hospital Germans Trias i Pujol, 08916 Barcelona, Spain
9
Centro de Investigación Biomédica en Red–Oncología (CIBERONC), 28029 Madrid, Spain
10
Instituciò Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
*
Authors to whom correspondence should be addressed.
Submission received: 3 March 2022 / Accepted: 11 March 2022 / Published: 21 March 2022
(This article belongs to the Special Issue New Therapeutic Strategies for Acute Myeloid Leukemia)

Abstract

:

Simple Summary

Patients with AML harboring constitutively active mutations in the FLT3 receptor generally have a poor prognosis (FLT3-ITDMUT). Despite the fact that several FLT3 inhibitors have been developed, clinical responses are commonly partial or not durable, highlighting the need for new molecules targeting FLT3-ITDMUT. Here, we tested EC-70124, a hybrid indolocarbazole analog from the same chemical space as midostaurin (a well-known FLT3 inhibitor). Our in vitro and in vivo experiments showed that EC-70124 exerts a robust and specific antileukemia activity against FLT3-ITDMUT AML cells while sparing healthy hematopoietic cells. Collectively, EC-70124 is a promising and safe agent for the treatment of this aggressive type of AML.

Abstract

Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Patients with AML harboring a constitutively active internal tandem duplication mutation (ITDMUT) in the FMS-like kinase tyrosine kinase (FLT3) receptor generally have a poor prognosis. Several tyrosine kinase/FLT3 inhibitors have been developed and tested clinically, but very few (midostaurin and gilteritinib) have thus far been FDA/EMA-approved for patients with newly diagnosed or relapse/refractory FLT3-ITDMUT AML. Disappointingly, clinical responses are commonly partial or not durable, highlighting the need for new molecules targeting FLT3-ITDMUT AML. Here, we tested EC-70124, a hybrid indolocarbazole analog from the same chemical space as midostaurin with a potent and selective inhibitory effect on FLT3. In vitro, EC-70124 exerted a robust and specific antileukemia activity against FLT3-ITDMUT AML primary cells and cell lines with respect to cytotoxicity, CFU capacity, apoptosis and cell cycle while sparing healthy hematopoietic (stem/progenitor) cells. We also analyzed its efficacy in vivo as monotherapy using two different xenograft models: an aggressive and systemic model based on MOLM-13 cells and a patient-derived xenograft model. Orally disposable EC-70124 exerted a potent inhibitory effect on the growth of FLT3-ITDMUT AML cells, delaying disease progression and debulking the leukemia. Collectively, our findings show that EC-70124 is a promising and safe agent for the treatment of AML with FLT3-ITDMUT.

1. Introduction

Acute myeloid leukemia (AML), the most common acute leukemia in adults, is a clinically and biologically heterogeneous disease characterized by the rapid proliferation and accumulation of immature myeloid blasts in bone marrow (BM) [1,2,3]. Internal tandem duplication (ITD) or kinase domain mutations of the Fms-like tyrosine kinase 3 (FLT3) gene represent genetic alterations frequently found in AML (~a third of patients) [4]. These mutations constitutively activate the tyrosine kinase receptor FLT3 and promote cell growth, survival and antiapoptotic signaling via several downstream targets including PI3K/AKT/mTOR and STAT5 [5,6]. FLT3-ITD mutations (FLT3-ITDMUT) represent approximately 80% of all FLT3 mutations and are associated with a high rate of relapse and poor clinical outcome [1,7,8,9]. As a consequence, there have been increasing efforts to develop potent targeted inhibitors of the FLT3 tyrosine kinase [10]. Despite initial challenges with the first-generation tyrosine kinase inhibitors (TKIs), which were associated with systemic toxicities, off-target effects and suboptimal target inhibition, there has been recent clinical success, with some TKIs including midostaurin and gilteritinib being clinically approved by the USA Food and Drug Administration (FDA) and the European Medicines Agency (EMA) as either monotherapy or combined with conventional chemotherapy for patients with de novo or relapse/refractory FLT3-ITDMUT AML [11,12,13,14]. Unfortunately, however, most clinical responses to such TKIs are either partial or transient, and many patients ultimately relapse with the common acquisition of secondary mutations in FLT3 or the upregulation of compensatory oncogenic pathways [15,16]. In addition, midostaurin is currently administered in combination with cytarabine and daunorubicin, thus preventing its administration in elderly patients and/or in those with comorbidities [12]. Accordingly, there is a clinical need for the development of new FLT3 inhibitors to broaden the range of therapeutic arsenal for patients with FLT3-TDMUT AML.
EC-70124 is a hybrid indolocarbazole analog from the same chemical space as midostaurin which has a potent multi-kinase inhibitory spectrum that impacts key signaling kinases implicated in pro-survival and proliferative cellular pathways [17]. The antitumor activity of EC-70124 has been demonstrated in solid tumors including sarcoma, glioblastoma, colorectal, prostate and breast cancers, associated with the inhibition of a wide variety of targets including components of PI3K/AKT/mTOR or JAK/STAT pathways [18,19,20,21,22,23]. In addition, EC-70124 was recently demonstrated to exert a potent and selective inhibitory effect on AML-related kinases such as FLT3 that is even higher than that of midostaurin [21]. Here, we sought to further characterize the antileukemia activity of EC-70124 as monotherapy against FLT3-ITDMUT AML primary cells in both in vitro and in vivo models. We show that EC-70124 has a potent and selective cytotoxic effect on FLT3-ITDMUT cells while sparing FLT3-ITDWT cells and healthy hematopoietic (stem/progenitor) cells (HSPC). Our results support EC-70124 as a novel and safe TKI in FLT3-ITDMUT.

2. Materials and Methods

2.1. AML Cell Lines and Primary AML and Healthy HSPC

The FLT3-ITDMUT AML cell line MV4;11 was obtained from the DSMZ (Braunschweig, Germany). The FLT3-ITDWT cell line HL60 was kindly provided by Prof. Luciano Di Croce (CRG, Barcelona, Spain). The MOLM-13-Luc+ (MOML-13) and Baf3 cell lines were kindly provided by Prof. Michael Andreeff (MD Anderson Cancer Center, Houston, TX, USA). All cell lines were cultured in RPMI medium supplemented with fetal bovine serum and antibiotics (Gibco, Invitrogen, Paisley, UK). A luciferase-expressing FLT3-ITDMUT AML patient-derived xenograft (PDX) (FLT3-ITDMUT-Luc+ AML-640 PDX) was generated and characterized as previously described by Jeremias’ Laboratory (Helmholtz Zentrum, München, Germany) [24,25]. Primary AML cells were obtained from fresh bone marrow (BM) aspirates from the Hospital Clínico San Carlos and the Hospital Infantil Niño Jesús (Madrid, Spain). BM-derived leukemic cells were isolated using Ficoll-Paque Plus (GE Healthcare) density gradient centrifugation. Table 1 summarizes the main clinical/cytogenetic/molecular characteristics of the patients’ cells. Fresh peripheral blood (PB) and cord blood (CB) units were obtained from healthy donors from the Catalonia Blood Tissue Bank following the institutional guidelines approved by our local Institutional Review Board. PB- and CB-derived mononuclear cells were also isolated using Ficoll-Paque density gradient centrifugation. CB-derived CD34+ HSPCs were magnetic-activated cell sorting (MACS)-purified using the human CD34 MicroBead kit and the AutoMACS device (Miltenyi Biotec, Madrid, Spain), as previously reported [26,27,28]. The purity of the CD34+ fraction was consistently >95%. Primary cells were cultured in StemSpan medium (Stem Cell Technologies, Vancouver, BC, Canada) supplemented with the hematopoietic cytokines Stem Cell Factor (100 ng/mL), FLT3 ligand (100 ng/mL), IL3 (10 ng/mL, all from PeproTech, London, UK) and antibiotics (Gibco). Cultures were maintained in a humidified atmosphere with 5% CO2 at 37 °C. The study was approved by the Institutional Review Board of the Clinic Hospital of Barcelona (HCB/2014/0687), and samples were accessed upon signed informed consent.

2.2. Drugs

EC-70124 was synthesized by EntreChem S.L. (Oviedo, Spain) using a proprietary process [21]. Midostaurin (PKC-412) was synthesized from staurosporine (Biomar Microbial Technologies, León, Spain), and the identity of the isolated product was verified by comparison with an authentic sample using high-performance liquid chromatography and nuclear magnetic resonance [21]. EC-70124 and midostaurin were reconstituted in DMSO and stored at −20 °C.

2.3. Cytotoxicity and Apoptosis Assays

The cytotoxic effect of the drugs on AML cell lines and Baf3 cells was assessed using the Cell Counting Kit-8 (which is based on reduction of the sulfonated tetrazolium salt WST-8) (Sigma-Aldrich, St. Lous, MO, USA) [28] and MTT assay, respectively, as described [28]. Briefly, 5000 AML cells or 60,000 Baf3 cells were plated in a 96-well plate and incubated with increasing concentrations of the corresponding drug for 48 and 96 h, respectively. Primary samples were incubated with EC-70124 (or midostaurin) at 200 nM (~IC50 average of all cell lines assayed in Puente-Moncada et al. [21]) for 48 h, and cell viability was measured using the Annexin-V/7-AAD Apoptosis Detection Kit (BD Biosciences, Madrid, Spain) on a FACS Canto-II cytometer, as described in [29]. Myeloid, B cell and T cell populations from HD-PBMCs were immunophenotyped by flow cytometry using the monoclonal antibodies CD33-APC, CD19-BV421 and CD3-PE, respectively.

2.4. Colony-Forming Unit (CFU) and Cell Cycle Assays

Cells were treated with 200 nM EC-70124 overnight (CB-CD34+ and primary AML cells) or for 48 h (AML cell lines). For the CFU formation assay, a total of 5 × 104 primary AML cells, 5 × 102 CB-CD34+ cells or 1 × 103 AML cells (cell lines) were plated onto methylcellulose H4434 medium (StemCell Technologies, Saint Égrève, France). Colonies were counted and scored at day 14 (CB-CD34+ and primary AML cells) or day 7 (AML cell lines) using standard morphologic criteria, as described in [27,29,30,31]. For cell cycle analysis, cells were fixed with 70% ice-cold ethanol after treatments. DNA was stained with a propidium iodide (PI) staining solution and cells were analyzed by flow cytometry (FACSCanto II, Becton Dickinson, Madrid, Spain), as described in [32]. Data were evaluated using ModFit (VeritySoftware).

2.5. Western Blot Analysis

A total of 107 MOLM-13 cells were treated with the different drugs at the indicated concentrations for 4 h. Cells were then lysed in 1% Triton X-100 lysis buffer supplemented with a complete protease inhibitor cocktail (Roche, Diagnostic, Basel, Switzerland) and phosphatase inhibitors (Sigma-Aldrich, St. Lous, MO, USA), as detailed in [33]. Proteins were separated by SDS-PAGE and transferred to polyvinylidene difluoride membranes (Amersham Bioscience, Merck KGaA, Darmstadt, Alemania). Primary antibodies (Cell Signaling Technology, Danvers, MA, USA) were used against the phosphorylated (p) and total forms of Stat5 and Akt (1:1000), S6, BAD, Erk and Akt (1:2000 and 1:1000, for phosphorylated and total forms, respectively). For signaling detection, appropriate secondary (anti-rabbit IgG peroxidase-conjugated and anti-mouse IgG peroxidase 1:3000, Calbiochem) antibodies were used and the reaction was visualized using the Bio-Rad Chemidoc MP Imaging system.

2.6. NSG Mice Xenotransplantation and Engraftment Analysis

All experimental procedures were approved by the Animal Care Committee of The Barcelona Biomedical Research Park (HRH-17-0007). We used 8–14-week-old NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice housed under pathogen-free conditions. EC-70124 was dosed daily by oral gavage at 20 mg/kg [21] and tumor burden was monitored at the indicated time points by bioluminescence using the Xenogen in vivo imaging system (IVIS, Perkin Elmer). For the MOLM-13 model, 103 cells (n = 10 mice) were intra-BM (IBM) transplanted into non-irradiated NSG mice [34,35]. On the next day, mice were randomized into treatment groups and were treated for 15 days. At day 20, mice were sacrificed and cells from the BM and PB were stained with anti-human HLA-ABC-FITC, CD33-PE and CD45-APC antibodies (BD Biosciences, Madrid, Spain) to analyze leukemic engraftment by flow cytometry. For the FLT3-ITDMUT-AML PDX model, 105 cells were transplanted intravenously (i.v.) into sublethally (2 Gy) irradiated mice (n = 14 mice), as described in [36]. Engraftment was monitored weekly by IVIS until it was detectable in most mice (week 7). At this time, BM and PB were collected by aspiration and facial vein bleeding, respectively, and human grafts were immunophenotyped by flow cytometry using HLA-ABC-PE, CD45-FITC and CD33-APC antibodies. Mice were homogeneously divided into the treatment groups and were treated for 18 days. BM and PB were also collected at the end of the treatment and mice were left untreated for 65 days to follow-up potential relapse by IVIS. Mice were sacrificed when disease symptoms were evident and/or IVIS signal was incompatible with life.

2.7. Targeted Next Generation Sequencing (NGSeq)

A targeted NGSeq study was performed on ex vivo FACS-sorted AML cells retrieved from four mice (AML-640 PDX), treated (two mice) or untreated with EC-70124 (two mice) at the endpoint. The NGS panel included 76 genes related to myeloid disorders. Briefly, DNA was extracted from the AML cells and the libraries were prepared using the Haematology OncoKitDx (Imegen–Healthincode group®). Target regions were then captured and amplified by hybridizing the DNA library with the above-mentioned kit (Imegen, Healthincode®). The captured and amplified DNA was sequenced using NextSeq550 platform (Illumina®). The results were analyzed using DataGenomics platform as described elsewhere [37,38,39].

2.8. Statistical Analyses

Data are expressed as mean ± SEM of different independent experiments unless otherwise specified. Statistical comparisons were performed using unpaired Student’s t test, except for comparison between treatments for the same patient (paired Student’s test). Differences between treatments along the days in PDX model were performed using multiple t test. Statistical analyses, normality test (Shapiro–Wilk) and IC50 were performed using GraphPad Prism v6.0 (GraphPad Software Inc. San Diego, CA, USA). Statistical significance was defined as a p-value < 0.05.

3. Results

3.1. EC-70124 Has a Potent Cytotoxic Effect on FLT3-ITDMUT AML Cells In Vitro

EC-70124 is an indolocarbazole from the same chemical space as midostaurin, with a recently demonstrated inhibitory activity against AML-related kinases and a greater potency than midostaurin for wild-type FLT3 and FLT3-ITD [21]. As these latter findings were based on an assay platform evaluating the dissociation constants of both compounds for selected kinases and disease relevant mutants, we were prompted to further functionally evaluate and compare the efficacy of EC-70124 and midostaurin against FLT3-ITDMUT AML cells. Cytotoxicity assays based on tetrazolium dye reduction (WST-8) revealed a comparable efficacy for both inhibitors, with similar levels of half-maximal inhibitory concentration (IC50) in FLT3-ITDMUT AML cells (17 nM and 14 nM in MOLM-13 and MV4;11 cells, respectively, for EC-70124; and 8 nM and 13 nM in MOLM-13 and MV4;11 cells, respectively, for midostaurin) (Figure 1a). EC-70124 treatment of primary FLT3-ITDMUT AML patient cells had a stronger effect on cell viability than midostaurin (both 200 nM, during 48 h, Figure 1b). The inhibitory activity of EC-70124 was also assessed by CFU assays in FLT3-ITDMUT AML patient cells and cell lines (Figure 1b,c). Up to a 50% decrease in the clonogenic potential after overnight pre-treatment with EC-70124 was observed with primary AML cells (17 ± 5 vs. 9 ± 3 CFUs in untreated and EC-70124-treated, respectively) (Figure 1b, right panel). This inhibitory activity was five- to six-fold more pronounced in both MOLM-13 and MV4;11 FLT3-ITDMUT AML cell lines (Figure 1c). Mechanistically, and very similar to midostaurin, EC-70124-mediated cytotoxicity was associated with an induction of apoptosis and severe cell cycle arrest through specific inhibition (dephosphorylation) of p-ERK and STAT5, one of the most important downstream targets in constitutively-activated FLT3 [40] (Figure 1d–f). Although EC-70124 induced a mild decrease in pAKT, we detected a strong inhibition of master downstream components including pS6 and pBAD (Figure 1f). Lysates from cells treated with cytarabine and idarubicin (standard-of-care treatment in AML) were included as controls to confirm the specificity of EC-70124 on FLT3 signaling. Indeed, cytarabine and idarubicin increased the phosphorylation of most components of the FLT3 signaling, likely reflecting a damage-sensing cellular response (Figure 1f). Taken together, these data indicate that EC-70124 exerts a potent and specific cytotoxic effect on FLT3-ITDMUT AML cells through inhibition of FLT3 downstream signaling.

3.2. EC-70124 Has No Evident Effect on FLT3-ITDWT AML Cells and Spares Healthy HSPCs and Mature Blood Cells

We next tested the specificity of EC-70124 by assaying its effects on FLT3-ITDWT AML cells using both FLT3-ITDWT AML primary cells and the cell lines HL60 and THP1. Cytotoxicity analysis revealed that the EC-70124 IC50 was considerably higher in FLT3-ITDWT cells than in FLT3-ITDMUT cells (72 µM (HL60) and 77 nM (THP1) vs. 17 nM (MOLM-13) and 14 nM (MV4:11)) (Figure 1g). While EC-70124 treatment caused a decrease in cell viability of primary FLT3-ITDWT AML patient cells (Figure 1h), the effect was much less pronounced than that observed in FLT3-ITDMUT cells (Figure 1b). Treatment with EC-70124 did not affect the clonogenic potential (CFUs) of either FLT3-ITDWT AML patient cells (Figure 1h) or FLT3-ITDWT AML cell lines (Figure 1i), Accordingly, EC-70124 had no effect on apoptosis or cell cycle progression in FLT3-ITDWT cells (Figure 1j,k). Finally, we performed a cytotoxicity assay on Baf3-WT or FLT3-ITD-overexpressing BaF cells (Baf3-ITD) treated with increasing concentrations of EC-70124 for 96 h (Figure 1l). The IC50 of EC-70124 was way lower in Baf3-ITD cells than in Baf3-WT cells (0.2 mM vs. 180 mM), confirming the specificity of EC-70124 for FLT3-ITDMUT AML cells.
We next evaluated the cytotoxicity of EC-70124 on healthy mature B cells, myeloid cells and T cells (Figure 2a) and on CD34+ HSPCs (Figure 2b) by treating cells overnight with 200 nM EC-70124. EC-70124 revealed no cytotoxicity on either healthy mature circulating blood cells or HSPCs (Figure 2a,b). EC-70124 did not affect either the clonogenic capacity of the healthy CD34+ HSPCs (Figure 2b, right panel), demonstrating that EC-70124 has no detectable cytotoxic effects on normal hematopoiesis.

3.3. EC-70124 Exhibits Antileukemic Activity in FLT3-ITDMUT AML Cells In Vivo

The oral availability and pharmacodynamics of EC-70124 were recently evaluated in a subcutaneous model of AML [21]. To extend and expand these observations, we tested its efficacy in two preclinical models of AML: an aggressive and systemic model using MOLM-13 cells and a robust FLT3-ITDMUT-Luc+ PDX model. For the MOLM-13 model, 1000 cells were IBM-transplanted into non-irradiated NSG mice, and due to the aggressiveness of this model, mice were randomized into groups and treatment was started the next day. Mice were dosed orally once daily with either vehicle or EC-70124 (20 mg/kg) for 15 days and were monitored by IVIS once weekly (Figure 3a). Mice were sacrificed at day 20 and engraftment levels were analyzed in BM and PB by flow cytometry. During the treatment period, EC-70124-treated mice showed significantly lower signals of leukemia than mice vehicle-treated mice, as measured by IVIS. At the endpoint, mice treated with EC-70124 had a significantly lower AML burden in both BM and PB than mice dosed with vehicle (Figure 3b). Next, we tested the antileukemic activity of EC-70124 using a PDX model with an FLT3-ITDMUT-Luc+ AML sample. A total of 100,000 PDX cells were i.v. injected into irradiated NSG mice and engraftment was monitored by IVIS weekly until mice showed a detectable signal. At this time, mice were randomized into two groups and were dosed orally once daily for 15 days. BM samples were taken by aspirate at the beginning and at the end of the treatment, and graft levels were analyzed by flow cytometry (Figure 3c). Unlike control mice, EC-70124-treated mice displayed the same low levels of engraftment at the beginning and at the end of the treatment, as monitored by IVIS and flow cytometry (Figure 3c,d). Quantification of the IVIS signal revealed that mice dosed with EC-70124 had a significantly lower leukemia burden than vehicle-treated mice at the end of the treatment (day 18). Importantly, EC-70124 treatment inhibited/delayed AML cell growth for ~24 days after withdrawal of treatment, but disease reoccurrence was eventually observed after this timepoint (Figure 3e). These data reveal that orally dosed EC-70124 inhibits FLT3-ITDMUT AML cell growth in vivo, but withdrawal of the inhibitor results in leukemia relapse, suggesting that outgrowth of EC-70124 resistant cells.
We next performed targeted NGSeq on ex vivo FACS-sorted AML cells retrieved from PDXs treated (n = 2) or untreated with EC-70124 (n = 2) at the endpoint in order to genetically characterize the EC-70124 resistant outgrowing cells observed at relapse. The variant allelic frequency (VAF) of FLT3-ITD after EC-70124 or vehicle treatment in vivo remained almost identical to that at disease presentation (45.47 vs. 46.27), indicating that a FLT3-IDTMUT clone remains the major clone responsible for disease reoccurrence after EC-70124 discontinuation. Interestingly, only one mutation in EZH2 (VAFs: 6%), a gene recently involved in AML pathogenesis, was specifically found in EC-7012-treated mice (Table 2). These data indicate that AML reoccurrence after suspension of EC-70124 treatment is driven by the major FLT3-ITDMUT AML clone. The biological relevance of the EZH2 mutation in 12% of the FLT3-ITD AML cells requires further prospective experimental work.

4. Discussion

AML is the most common hematological malignancy in adults, and the presence of FLT3-ITDMUT confers poor outcome in terms of overall and relapse-free survival in AML patients [41,42]. Indeed, recent results suggest that FLT3-ITDMUT may function as an important oncogenic hit accelerating disease progression [43]. Accordingly, few TKIs disrupting the oncogenic signaling initiated by FLT3 are already FDA-approved (midostaurin and gilteritinib), and several others are under development, offering promising therapeutic strategies for patients with FLT3-ITDMUT AML [44]. In line with this aim, we here evaluated the efficacy of EC-70124, a selective inhibitor from the same chemical space as midostaurin, on FLT3-ITDMUT AML cells. We confirm in vitro the cytotoxic effect of EC-70124 on primary FLT3-ITDMUT AML patient cells and cell lines, revealing an even greater toxicity than midostaurin, which is in line with recent data by Puentes-Moncada et al. [21]. These authors have previously shown that EC-70124 exerts a higher inhibitory activity for FLT3-ITDMUT than midostaurin using kinase affinity assays, pointing to EC-70124 as a novel therapeutic for AML.
EC-70124 is a potent multi-kinase inhibitor that can inhibit a broad spectrum of kinases including FLT3, JAK, SYK and PIM [18,19,20,21,22,23]. FLT3-ITD induces several downstream pro-survival pathways including MAPK, PI3K/Akt and STAT5, triggering proliferation and suppressing apoptosis [45]. EC-70124 treatment increased apoptosis and induced cell cycle arrest in FLT3-ITDMUT cell lines, and these effects were mediated through inhibition of FLT3 signaling. We found that EC-70124 inhibited p-AKT, p-ERK and pSTAT5, a master target in constitutively activated FLT3, and also inhibited the phosphorylation of S6 and BAD, which are downstream components of MAPK or PI3K pathways. This indicates a specific inhibitory function of EC-70124 on FLT3 signaling.
Of note, it has been reported that EC-70124 has more affinity for FLT3 mutants in AML than both wild-type FLT3 and other kinases involved in the development of healthy hematopoietic cells, such as KIT, suggesting safety and specificity for FLT3-ITDMUT [21,46]. Indeed, EC-70124 had no cytotoxic effects on FLT3-ITDWT cell lines, and barely (not significant) impacts FLT3-ITDWT AML patient cells, indicating a high specificity for FLT3-ITDMUT versus FLT3-ITDWT AML cells. This affinity may confer an advantage for treatment with FLT3 inhibitors, as it may uncouple specificity and potency from toxicity caused by off-target activity, the main limitation of first-generation inhibitors [47]. Very importantly, EC-70124 spares healthy HSPCs and mature blood lymphoid and myeloid cells, underlying its safety and non-myeloablative profile on normal hematopoiesis.
Finally, in vivo analysis revealed that EC-70124 has antitumor activity and suppresses disease progression in both MOLM-13 and FT3-ITDMUT PDX models. However, after treatment withdrawal, mice transplanted with FT3-ITDMUT PDX relapsed, with an action window of ~24 days, suggesting the eventual outgrowth of EC-70124-resistant AML cells. These results are in line with previous studies demonstrating that FT3-ITDMUT is subclonal and absent in the most immature AML-initiating/preleukemic cells, reinforcing that therapeutic targeting of subclonal mutations such as FLT3-ITDMUT might only have a transient clinical benefit in debulking the leukemia, but is unlikely to be curative, since it will not target the roots of the disease [48]. In fact, the VAF of FLT3-ITD after EC-70124 treatment in vivo remained almost identical to that at disease presentation, indicating that a FLT3-IDTMUT clone remains the major clone responsible for disease reoccurrence after EC-70124 discontinuation. Emergence of resistance poses a significant challenge in TKI-based treatments [44]. FLT3-TKD point mutations in the FLT3 drug binding site have been previously associated with resistance to FLT3 inhibitors [11]; however, we failed to detect such mutations in the FLT3 gene in EC-70124-resistant AML cells. Acquisition of new mutations in other genes and activation of alternative signaling pathways represent a common but complex mechanism underlying resistance to FLT3 inhibitors [44,49]. Interestingly, only one mutation in EZH2 (VAFs: 6%), a gene recently involved in AML pathogenesis [50,51], was specifically found in EC-7012-treated mice. The biological relevance of the EZH2 mutation in 12% of the FLT3-ITD AML cells requires further prospective experimental work. In sum, our results suggest that wise treatment schemes should be developed to administer TKIs as co-adjuvant treatment with other therapeutic strategies and/or allogeneic HSC transplantation aimed to target the AML-initiating cells and suppress disease progression.

5. Conclusions

Here, we show that EC-70124, a multi-kinase inhibitor, has a potent and selective cytotoxic effect on FLT3-ITDMUT cells in vitro and in vivo, delaying disease progression and sparing FLT3-ITDWT cells and healthy mature hematopoietic cells and HSPCs. Collectively, our results support EC-70124 as a novel and safe TKI in FLT3-ITDMUT AML.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers14061593/s1, Figure S1: Uncropped blots.

Author Contributions

B.L.-M. conceived the study, designed and performed experiments, analyzed data and wrote the manuscript. P.C., F.G.-A., R.D.d.l.G., H.R.-H., M.V., A.R.-G., R.S., J.C., P.A.R., L.Z. and C.B. performed experiments and analyzed data. M.R.-O. and E.A. provided primary leukemia samples. I.J., J.C.R.-M. and F.M. contributed intellectually and with key reagents/resources, and financially supported the study. P.M. conceived the study, designed experiments, wrote the manuscript and financially supported the study. All authors have read and agreed to the published version of the manuscript.

Funding

We thank CERCA/Generalitat de Catalunya and Fundació Josep Carreras-Obra Social la Caixa for institutional support. This research was funded by the Spanish Ministry of Economy and Competitiveness (RTC-2016-4603-1 in collaboration with Entrechem, PID2019-108160RB-I00/AEI/10.13039/501100011033), the Interreg V-A program (POCTEFA) 2014–2020 (grant PROTEOblood EFA360/19), Health Canada (H4080-144541) and Deutsche Josep Carreras Leukämie Stiftung in collaboration with I.J. to P.M. (15R/2021). P.M. and M.R.-O. also acknowledge the support from ISCIII-RICORS within the Next Generation EU program (plan de recuperación, transformación y resiliencia). Additional funding was provided by Consejería de Salud y Familia (PI-0119-2019) to RDG, the Health Institute Carlos III (FIS PI20/00822), Asociación Española Contra el Cáncer (PRYGN211192BUEN) and Ministerio de Ciencia e Innovacion (PLE2021-007518 and PI20/00822) to C.B. M.V. was supported by Juan de la Cierva fellowship (IJCI-2017-33172). B.L.-M. was supported by the Asociación Española Contra el Cáncer (INVES20011LÓPE) and Consejería de Salud y Familia (PEER-0028-2020).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of the Clinic Hospital of Barcelona (HCB/2014/0687). All experimental procedures were approved by the Animal Care Committee of The Barcelona Biomedical Research Park (HRH-17-0007).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

P.M. is the co-founder of OneChain Immunotherapeutics, an IJC spin-off which has no connection or conflict of interest with the present research. The remaining authors declare no conflict of interest.

References

  1. Dohner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Buchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Grimwade, D.; Ivey, A.; Huntly, B.J. Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 2016, 127, 29–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Porwit, A.; Bene, M.C. Acute leukemias of ambiguous origin. Am. J. Clin. Pathol. 2015, 144, 361–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
  5. Gunawardane, R.N.; Nepomuceno, R.R.; Rooks, A.M.; Hunt, J.P.; Ricono, J.M.; Belli, B.; Armstrong, R.C. Transient exposure to quizartinib mediates sustained inhibition of FLT3 signaling while specifically inducing apoptosis in FLT3-activated leukemia cells. Mol. Cancer. Ther. 2013, 12, 438–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hayakawa, F.; Towatari, M.; Kiyoi, H.; Tanimoto, M.; Kitamura, T.; Saito, H.; Naoe, T. Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines. Oncogene 2000, 19, 624–631. [Google Scholar] [CrossRef] [Green Version]
  7. Wagner, K.; Damm, F.; Thol, F.; Gohring, G.; Gorlich, K.; Heuser, M.; Schafer, I.; Schlegelberger, B.; Heil, G.; Ganser, A.; et al. FLT3-internal tandem duplication and age are the major prognostic factors in patients with relapsed acute myeloid leukemia with normal karyotype. Haematologica 2011, 96, 681–686. [Google Scholar] [CrossRef] [Green Version]
  8. Ravandi, F.; Kantarjian, H.; Faderl, S.; Garcia-Manero, G.; O’Brien, S.; Koller, C.; Pierce, S.; Brandt, M.; Kennedy, D.; Cortes, J.; et al. Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk. Res. 2010, 34, 752–756. [Google Scholar] [CrossRef] [Green Version]
  9. Rucker, F.G.; Du, L.; Luck, T.J.; Benner, A.; Krzykalla, J.; Gathmann, I.; Voso, M.T.; Amadori, S.; Prior, T.W.; Brandwein, J.M.; et al. Molecular landscape and prognostic impact of FLT3-ITD insertion site in acute myeloid leukemia: RATIFY study results. Leukemia 2021, 36, 90–99. [Google Scholar] [CrossRef]
  10. Fathi, A.T.; Chen, Y.B. The role of FLT3 inhibitors in the treatment of FLT3-mutated acute myeloid leukemia. Eur. J. Haematol. 2017, 98, 330–336. [Google Scholar] [CrossRef]
  11. Antar, A.I.; Otrock, Z.K.; Jabbour, E.; Mohty, M.; Bazarbachi, A. FLT3 inhibitors in acute myeloid leukemia: Ten frequently asked questions. Leukemia 2020, 34, 682–696. [Google Scholar] [CrossRef] [PubMed]
  12. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Dohner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef] [PubMed]
  13. Pulte, E.D.; Norsworthy, K.J.; Wang, Y.; Xu, Q.; Qosa, H.; Gudi, R.; Przepiorka, D.; Fu, W.; Okusanya, O.O.; Goldberg, K.B.; et al. FDA Approval Summary: Gilteritinib for Relapsed or Refractory Acute Myeloid Leukemia with a FLT3 Mutation. Clin. Cancer Res. 2021, 27, 3515–3521. [Google Scholar] [CrossRef] [PubMed]
  14. Hunter, B.D. Quizartinib for Relapsed/Refractory FLT3-Mutated Acute Myeloid Leukemia: More Patients Make It to Transplant, but Are There Any Other Benefits? Transplant Cell Ther. 2021, 27, 99–100. [Google Scholar] [CrossRef] [PubMed]
  15. Man, C.H.; Fung, T.K.; Ho, C.; Han, H.H.; Chow, H.C.; Ma, A.C.; Choi, W.W.; Lok, S.; Cheung, A.M.; Eaves, C.; et al. Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia: Favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation. Blood 2012, 119, 5133–5143. [Google Scholar] [CrossRef]
  16. Dumas, P.Y.; Villacreces, A.; Guitart, A.V.; El-Habhab, A.; Massara, L.; Mansier, O.; Bidet, A.; Martineau, D.; Fernandez, S.; Leguay, T.; et al. Dual Inhibition of FLT3 and AXL by Gilteritinib Overcomes Hematopoietic Niche-Driven Resistance Mechanisms in FLT3-ITD Acute Myeloid Leukemia. Clin. Cancer. Res. 2021, 27, 6012–6025. [Google Scholar] [CrossRef]
  17. Sanchez, C.; Salas, A.P.; Brana, A.F.; Palomino, M.; Pineda-Lucena, A.; Carbajo, R.J.; Mendez, C.; Moris, F.; Salas, J.A. Generation of potent and selective kinase inhibitors by combinatorial biosynthesis of glycosylated indolocarbazoles. Chem. Commun. 2009, 27, 4118–4120. [Google Scholar] [CrossRef]
  18. Civenni, G.; Longoni, N.; Costales, P.; Dallavalle, C.; Garcia Inclan, C.; Albino, D.; Nunez, L.E.; Moris, F.; Carbone, G.M.; Catapano, C.V. EC-70124, a Novel Glycosylated Indolocarbazole Multikinase Inhibitor, Reverts Tumorigenic and Stem Cell Properties in Prostate Cancer by Inhibiting STAT3 and NF-kappaB. Mol. Cancer. Ther. 2016, 15, 806–818. [Google Scholar] [CrossRef] [Green Version]
  19. Cuenca-Lopez, M.D.; Serrano-Heras, G.; Montero, J.C.; Corrales-Sanchez, V.; Gomez-Juarez, M.; Gascon-Escribano, M.J.; Morales, J.C.; Voisin, V.; Nunez, L.E.; Moris, F.; et al. Antitumor activity of the novel multi-kinase inhibitor EC-70124 in triple negative breast cancer. Oncotarget 2015, 6, 27923–27937. [Google Scholar] [CrossRef] [Green Version]
  20. Nogueira, L.; Ruiz-Ontanon, P.; Vazquez-Barquero, A.; Lafarga, M.; Berciano, M.T.; Aldaz, B.; Grande, L.; Casafont, I.; Segura, V.; Robles, E.F.; et al. Blockade of the NFkappaB pathway drives differentiating glioblastoma-initiating cells into senescence both in vitro and in vivo. Oncogene 2011, 30, 3537–3548. [Google Scholar] [CrossRef]
  21. Puente-Moncada, N.; Costales, P.; Antolin, I.; Nunez, L.E.; Oro, P.; Hermosilla, M.A.; Perez-Escuredo, J.; Rios-Lombardia, N.; Sanchez-Sanchez, A.M.; Luno, E.; et al. Inhibition of FLT3 and PIM Kinases by EC-70124 Exerts Potent Activity in Preclinical Models of Acute Myeloid Leukemia. Mol. Cancer. Ther. 2018, 17, 614–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Serrano-Heras, G.; Cuenca-Lopez, M.D.; Montero, J.C.; Corrales-Sanchez, V.; Morales, J.C.; Nunez, L.E.; Moris, F.; Pandiella, A.; Ocana, A. Phospho-kinase profile of colorectal tumors guides in the selection of multi-kinase inhibitors. Oncotarget 2015, 6, 31272–31283. [Google Scholar] [CrossRef] [PubMed]
  23. Estupinan, O.; Santos, L.; Rodriguez, A.; Fernandez-Nevado, L.; Costales, P.; Perez-Escuredo, J.; Hermosilla, M.A.; Oro, P.; Rey, V.; Tornin, J.; et al. The multikinase inhibitor EC-70124 synergistically increased the antitumor activity of doxorubicin in sarcomas. Int. J. Cancer 2019, 145, 254–266. [Google Scholar] [CrossRef] [Green Version]
  24. Vick, B.; Rothenberg, M.; Sandhofer, N.; Carlet, M.; Finkenzeller, C.; Krupka, C.; Grunert, M.; Trumpp, A.; Corbacioglu, S.; Ebinger, M.; et al. An advanced preclinical mouse model for acute myeloid leukemia using patients’ cells of various genetic subgroups and in vivo bioluminescence imaging. PLoS ONE 2015, 10, e0120925. [Google Scholar] [CrossRef]
  25. Reiter, K.; Polzer, H.; Krupka, C.; Maiser, A.; Vick, B.; Rothenberg-Thurley, M.; Metzeler, K.H.; Dorfel, D.; Salih, H.R.; Jung, G.; et al. Tyrosine kinase inhibition increases the cell surface localization of FLT3-ITD and enhances FLT3-directed immunotherapy of acute myeloid leukemia. Leukemia 2018, 32, 313–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bueno, C.; Montes, R.; Martin, L.; Prat, I.; Hernandez, M.C.; Orfao, A.; Menendez, P. NG2 antigen is expressed in CD34+ HPCs and plasmacytoid dendritic cell precursors: Is NG2 expression in leukemia dependent on the target cell where leukemogenesis is triggered? Leukemia 2008, 22, 1475–1478. [Google Scholar] [CrossRef] [Green Version]
  27. Bueno, C.; Montes, R.; Menendez, P. The ROCK inhibitor Y-27632 negatively affects the expansion/survival of both fresh and cryopreserved cord blood-derived CD34+ hematopoietic progenitor cells: Y-27632 negatively affects the expansion/survival of CD34+HSPCs. Stem Cell Rev. Rep. 2010, 6, 215–223. [Google Scholar] [CrossRef]
  28. Lopez-Millan, B.; Diaz de la Guardia, R.; Roca-Ho, H.; Anguita, E.; Islam, A.; Romero-Moya, D.; Prieto, C.; Gutierrez-Aguera, F.; Bejarano-Garcia, J.A.; Perez-Simon, J.A.; et al. IMiDs mobilize acute myeloid leukemia blasts to peripheral blood through downregulation of CXCR4 but fail to potentiate AraC/Idarubicin activity in preclinical models of non del5q/5q- AML. Oncoimmunology 2018, 7, e1477460. [Google Scholar] [CrossRef] [Green Version]
  29. Romero-Moya, D.; Bueno, C.; Montes, R.; Navarro-Montero, O.; Iborra, F.J.; Lopez, L.C.; Martin, M.; Menendez, P. Cord blood-derived CD34+ hematopoietic cells with low mitochondrial mass are enriched in hematopoietic repopulating stem cell function. Haematologica 2013, 98, 1022–1029. [Google Scholar] [CrossRef] [Green Version]
  30. de la Guardia, R.D.; Lopez-Millan, B.; Roca-Ho, H.; Bueno, C.; Gutierrez-Aguera, F.; Fuster, J.L.; Anguita, E.; Zanetti, S.R.; Vives, S.; Nomdedeu, J.; et al. Bone marrow mesenchymal stem/stromal cells from risk-stratified acute myeloid leukemia patients are anti-inflammatory in in vivo preclinical models of hematopoietic reconstitution and severe colitis. Haematologica 2019, 104, e54–e58. [Google Scholar] [CrossRef] [Green Version]
  31. Montes, R.; Ayllon, V.; Gutierrez-Aranda, I.; Prat, I.; Hernandez-Lamas, M.C.; Ponce, L.; Bresolin, S.; Te Kronnie, G.; Greaves, M.; Bueno, C.; et al. Enforced expression of MLL-AF4 fusion in cord blood CD34+ cells enhances the hematopoietic repopulating cell function and clonogenic potential but is not sufficient to initiate leukemia. Blood 2011, 117, 4746–4758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Molina, O.; Vinyoles, M.; Granada, I.; Roca-Ho, H.; Gutierrez-Aguera, F.; Valledor, L.; Lopez-Lopez, C.M.; Rodriguez-Gonzalez, P.; Trincado, J.L.; Menendez, S.T.; et al. Impaired condensin complex and Aurora B kinase underlie mitotic and chromosomal defects in hyperdiploid B-cell ALL. Blood 2020, 136, 313–327. [Google Scholar] [CrossRef] [PubMed]
  33. Montes, R.; Ayllon, V.; Prieto, C.; Bursen, A.; Prelle, C.; Romero-Moya, D.; Real, P.J.; Navarro-Montero, O.; Chillon, C.; Marschalek, R.; et al. Ligand-independent FLT3 activation does not cooperate with MLL-AF4 to immortalize/transform cord blood CD34+ cells. Leukemia 2014, 28, 666–674. [Google Scholar] [CrossRef]
  34. Prieto, C.; Lopez-Millan, B.; Roca-Ho, H.; Stam, R.W.; Romero-Moya, D.; Rodriguez-Baena, F.J.; Sanjuan-Pla, A.; Ayllon, V.; Ramirez, M.; Bardini, M.; et al. NG2 antigen is involved in leukemia invasiveness and central nervous system infiltration in MLL-rearranged infant B-ALL. Leukemia 2018, 32, 633–644. [Google Scholar] [CrossRef] [PubMed]
  35. Rodriguez, R.; Rosu-Myles, M.; Arauzo-Bravo, M.; Horrillo, A.; Pan, Q.; Gonzalez-Rey, E.; Delgado, M.; Menendez, P. Human bone marrow stromal cells lose immunosuppressive and anti-inflammatory properties upon oncogenic transformation. Stem Cell Rep. 2014, 3, 606–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lopez-Millan, B.; Sanchez-Martinez, D.; Roca-Ho, H.; Gutierrez-Aguera, F.; Molina, O.; Diaz de la Guardia, R.; Torres-Ruiz, R.; Fuster, J.L.; Ballerini, P.; Suessbier, U.; et al. NG2 antigen is a therapeutic target for MLL-rearranged B-cell acute lymphoblastic leukemia. Leukemia 2019, 33, 1557–1569. [Google Scholar] [CrossRef] [Green Version]
  37. Diaz de la Guardia, R.; Velasco-Hernandez, T.; Gutierrez-Aguera, F.; Roca-Ho, H.; Molina, O.; Nombela-Arrieta, C.; Bataller, A.; Fuster, J.L.; Anguita, E.; Vives, S.; et al. Engraftment characterization of risk-stratified AML in NSGS mice. Blood Adv. 2021, 5, 4842–4854. [Google Scholar] [CrossRef]
  38. Espasa, A.; Torrents, S.; Morales-Indiano, C.; Rico, L.G.; Bardina, J.; Ancochea, A.; Bistue-Rovira, A.; Linio, R.; Raya, M.; Vergara, S.; et al. Diagnostic performance of the ClearLLab 10C B cell tube. Cytometry B Clin. Cytom. 2021, 100, 519–530. [Google Scholar] [CrossRef]
  39. Huguet, M.; Zamora, L.; Granada, I.; Orna, E.; Mesa, A.; Tapia, G.; Sorigue, M.; Fernandez-Sanmartin, M.A.; Xicoy, B.; Navarro, J.T. Polycythemia Vera Evolution to Chronic Myelomocytic Leukemia: The Prognostic Value of Next Generation Sequencing. Hemasphere 2020, 4, e466. [Google Scholar] [CrossRef]
  40. Zimmerman, E.I.; Turner, D.C.; Buaboonnam, J.; Hu, S.; Orwick, S.; Roberts, M.S.; Janke, L.J.; Ramachandran, A.; Stewart, C.F.; Inaba, H.; et al. Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia. Blood 2013, 122, 3607–3615. [Google Scholar] [CrossRef] [Green Version]
  41. O’Donnell, M.R.; Tallman, M.S.; Abboud, C.N.; Altman, J.K.; Appelbaum, F.R.; Arber, D.A.; Bhatt, V.; Bixby, D.; Blum, W.; Coutre, S.E.; et al. Acute Myeloid Leukemia, Version 3.2017, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2017, 15, 926–957. [Google Scholar] [CrossRef] [PubMed]
  42. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef] [PubMed]
  43. Kronke, J.; Bullinger, L.; Teleanu, V.; Tschurtz, F.; Gaidzik, V.I.; Kuhn, M.W.; Rucker, F.G.; Holzmann, K.; Paschka, P.; Kapp-Schworer, S.; et al. Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood 2013, 122, 100–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Daver, N.; Schlenk, R.F.; Russell, N.H.; Levis, M.J. Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia 2019, 33, 299–312. [Google Scholar] [CrossRef] [Green Version]
  45. Leung, A.Y.; Man, C.H.; Kwong, Y.L. FLT3 inhibition: A moving and evolving target in acute myeloid leukaemia. Leukemia 2013, 27, 260–268. [Google Scholar] [CrossRef] [Green Version]
  46. Warkentin, A.A.; Lopez, M.S.; Lasater, E.A.; Lin, K.; He, B.L.; Leung, A.Y.; Smith, C.C.; Shah, N.P.; Shokat, K.M. Overcoming myelosuppression due to synthetic lethal toxicity for FLT3-targeted acute myeloid leukemia therapy. eLife 2014, 3, e03445. [Google Scholar] [CrossRef] [Green Version]
  47. Weisberg, E.; Roesel, J.; Furet, P.; Bold, G.; Imbach, P.; Florsheimer, A.; Caravatti, G.; Jiang, J.; Manley, P.; Ray, A.; et al. Antileukemic Effects of Novel First- and Second-Generation FLT3 Inhibitors: Structure-Affinity Comparison. Genes Cancer 2010, 1, 1021–1032. [Google Scholar] [CrossRef] [Green Version]
  48. Guardia, R.D.; Gonzalez-Silva, L.; Lopez-Millan, B.; Rodriguez-Sevilla, J.J.; Baroni, M.L.; Bueno, C.; Anguita, E.; Vives, S.; Palomo, L.; Lapillonne, H.; et al. Bone Marrow Clonogenic Myeloid Progenitors from NPM1-Mutated AML Patients Do Not Harbor the NPM1 Mutation: Implication for the Cell-Of-Origin of NPM1+ AML. Genes 2020, 11, 73. [Google Scholar] [CrossRef] [Green Version]
  49. Piloto, O.; Wright, M.; Brown, P.; Kim, K.T.; Levis, M.; Small, D. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood 2007, 109, 1643–1652. [Google Scholar] [CrossRef] [Green Version]
  50. Panuzzo, C.; Signorino, E.; Calabrese, C.; Ali, M.S.; Petiti, J.; Bracco, E.; Cilloni, D. Landscape of Tumor Suppressor Mutations in Acute Myeloid Leukemia. J. Clin. Med. 2020, 9, 802. [Google Scholar] [CrossRef] [Green Version]
  51. Lund, K.; Adams, P.D.; Copland, M. EZH2 in normal and malignant hematopoiesis. Leukemia 2014, 28, 44–49. [Google Scholar] [CrossRef] [PubMed]
Figure 1. EC-70124 exerts profound cytotoxic effects in FLT3-ITDMUT AML cells. (a) Cytotoxicity dose–response curves of midostaurin (right panel) and EC-70124 (left panel) on FLT3-ITDMUT AML cell lines. (b) Effect of EC-70124 (EC) and midostaurin (MID) on cell viability (left panel) and CFU formation (right panel) in primary FLT3-ITDMUT AML patient cells. Viability data are relative to the untreated group (Ctrl). Patient samples are color-codified, and black lines represent the mean. (ce) Effect of EC-70124 on CFU formation (c), apoptosis (d) and cell cycle (e) in FLT3-ITDMUT AML cell lines. Representative CFUs are shown as inset in C. Representative cell cycle FACS analysis is shown E (right panel). (f) Representative Western blots showing the effect of EC-70124 and midostaurin on the main targets related to FLT3 signaling (Figure S1). Lysates from cells treated with cytarabine (AraC) and idarubicin (Ida) were included as controls. Red asterisks depict the specific effect of EC-70124 or midostaurin on the indicated phosphorylation. Blue numbers depict the ratio phosphorylated/total protein for each band. (g) Cytotoxicity dose–response curves of EC-70124 on FLT3-ITDWT AML cell lines. (h) Effect of EC-70124 on cell viability cell (left panel) and CFU formation (right panel) in primary FLT3-ITDWT AML patient samples. Viability data are relative to Ctrl group. Patient samples are color-codified, and black lines represent the mean. (ik) Effect of EC-70124 on CFU formation (i), apoptosis (j) and cell cycle (k) in FLT3-ITDWT AML cell lines (HL60 and THP1). Representative CFUs are shown as inset in (i). Representative cell cycle FACS analysis is shown (k) (right panel). (l) Cytotoxicity dose–response curves of EC-70124 on Baf3-WT and BaF-ITD cells (n = 3). Data represent mean ± SEM. * p < 0.05; ** p < 0.01; *** p < 0.001; ns, not significant (Student’s t test). Abbreviations: EC, EC-70124; MID, midostaurin.
Figure 1. EC-70124 exerts profound cytotoxic effects in FLT3-ITDMUT AML cells. (a) Cytotoxicity dose–response curves of midostaurin (right panel) and EC-70124 (left panel) on FLT3-ITDMUT AML cell lines. (b) Effect of EC-70124 (EC) and midostaurin (MID) on cell viability (left panel) and CFU formation (right panel) in primary FLT3-ITDMUT AML patient cells. Viability data are relative to the untreated group (Ctrl). Patient samples are color-codified, and black lines represent the mean. (ce) Effect of EC-70124 on CFU formation (c), apoptosis (d) and cell cycle (e) in FLT3-ITDMUT AML cell lines. Representative CFUs are shown as inset in C. Representative cell cycle FACS analysis is shown E (right panel). (f) Representative Western blots showing the effect of EC-70124 and midostaurin on the main targets related to FLT3 signaling (Figure S1). Lysates from cells treated with cytarabine (AraC) and idarubicin (Ida) were included as controls. Red asterisks depict the specific effect of EC-70124 or midostaurin on the indicated phosphorylation. Blue numbers depict the ratio phosphorylated/total protein for each band. (g) Cytotoxicity dose–response curves of EC-70124 on FLT3-ITDWT AML cell lines. (h) Effect of EC-70124 on cell viability cell (left panel) and CFU formation (right panel) in primary FLT3-ITDWT AML patient samples. Viability data are relative to Ctrl group. Patient samples are color-codified, and black lines represent the mean. (ik) Effect of EC-70124 on CFU formation (i), apoptosis (j) and cell cycle (k) in FLT3-ITDWT AML cell lines (HL60 and THP1). Representative CFUs are shown as inset in (i). Representative cell cycle FACS analysis is shown (k) (right panel). (l) Cytotoxicity dose–response curves of EC-70124 on Baf3-WT and BaF-ITD cells (n = 3). Data represent mean ± SEM. * p < 0.05; ** p < 0.01; *** p < 0.001; ns, not significant (Student’s t test). Abbreviations: EC, EC-70124; MID, midostaurin.
Cancers 14 01593 g001
Figure 2. EC-70124 has no cytotoxic effects on healthy hematopoietic (stem/progenitor) cells. (a) Cytotoxic effect of EC-70124 on HD-PBMCs (B, T and myeloid cells). (b) Number of CFUs from CB-CD34+ cells after treatment with EC-70124. Right panel, scoring of the colony types. * p < 0.05 (Student’s t test).
Figure 2. EC-70124 has no cytotoxic effects on healthy hematopoietic (stem/progenitor) cells. (a) Cytotoxic effect of EC-70124 on HD-PBMCs (B, T and myeloid cells). (b) Number of CFUs from CB-CD34+ cells after treatment with EC-70124. Right panel, scoring of the colony types. * p < 0.05 (Student’s t test).
Cancers 14 01593 g002
Figure 3. Oral administration of EC-70124 delays the growth of FLT3-ITDMUT AML cells in NSG mice. (a) Experimental design for the MOLM-13 leukemic model. (b) Upper panel, IVIS imaging of tumor burden monitored by bioluminescence at the indicated time points in the MOLM-13 leukemic model. Bottom panel, percentage of MOLM-13 cells at the endpoint in the BM and PB of vehicle- and EC-70124-treated mice. (c) Upper panel, experimental design for the FLT3-ITDMUTLuc+ AML-640 PDX model. Bottom panel, IVIS imaging of tumor burden by bioluminescence at the indicated time points in the Luc-FLT3-ITDMUT PDX model. (d) Percentage of FLT3-ITDMUT AML blasts in BM at the beginning (D0) and the end (D17) of the treatment in the FLT3-ITDMUT AML PDX model. Each point depicts a mouse. (e) Total radiance quantification at the indicated time points. Ec-70124 was stopped on day 17. * p < 0.05; ** p < 0.01 (Multiple t test).
Figure 3. Oral administration of EC-70124 delays the growth of FLT3-ITDMUT AML cells in NSG mice. (a) Experimental design for the MOLM-13 leukemic model. (b) Upper panel, IVIS imaging of tumor burden monitored by bioluminescence at the indicated time points in the MOLM-13 leukemic model. Bottom panel, percentage of MOLM-13 cells at the endpoint in the BM and PB of vehicle- and EC-70124-treated mice. (c) Upper panel, experimental design for the FLT3-ITDMUTLuc+ AML-640 PDX model. Bottom panel, IVIS imaging of tumor burden by bioluminescence at the indicated time points in the Luc-FLT3-ITDMUT PDX model. (d) Percentage of FLT3-ITDMUT AML blasts in BM at the beginning (D0) and the end (D17) of the treatment in the FLT3-ITDMUT AML PDX model. Each point depicts a mouse. (e) Total radiance quantification at the indicated time points. Ec-70124 was stopped on day 17. * p < 0.05; ** p < 0.01 (Multiple t test).
Cancers 14 01593 g003
Table 1. Biological and cytogenetic-molecular characteristics of patients with AML used in this study.
Table 1. Biological and cytogenetic-molecular characteristics of patients with AML used in this study.
Patient IDDiagnosticCytogeneticsMolecularBlasts (%)Age (y)Sex
#AML1AML-M446,XX, + 8NPM1MUT, FLT3-ITD7063F
#AML2 AML-M446,XY [20]FLT3-ITD, DNMT3, NRASn.a.64M
#AML3AML-M5n.pNPM1MUT, FLT3-ITD>9077M
#AML4AML46,XX,del(5)(q31) [2]/45,XXdel5(q31),-7 [7]FLT3-ITD6076F
#AML5AML-M446, XYNPM1MUT, FLT3-ITD8848M
#AML6AML-M4n.pFLT3-ITD8816M
#AML7AML-M245,XX,-7 [9]/46,XX [13]-6833F
#AML8AMLn.pNPM1MUT>9086F
#AML9Relapse AML46,XX [20]-4052F
#AML10AML-M448,XX, + 8, + 13,inv16(p13;p22) [4]/
47,XX, + 8inv16(p13;p22) [4]
CBF/MYH119044F
#AML11AML-M1n.p-9081M
Abbreviations: y, years; M, male; F, female; n.p, not performed due to absence of metaphases; -, no mutations found in TP53, FLT3, NPM1, cEBPa, WT and IDH1; n.a. not analyzed.
Table 2. Specific mutations (and their VAF) commonly found in myeloid genes in AML patients analyzed by targeted NGS in ex vivo FACS-sorted AML cells retrieved from PDXs treated (n = 2 mice) or untreated with EC-70124 (n = 2 mice).
Table 2. Specific mutations (and their VAF) commonly found in myeloid genes in AML patients analyzed by targeted NGS in ex vivo FACS-sorted AML cells retrieved from PDXs treated (n = 2 mice) or untreated with EC-70124 (n = 2 mice).
VehicleEC70124
GenecHgvsMouse #1
VAF (%)
Mouse #2
VAF (%)
Mouse #1
VAF (%)
Mouse #2
VAF (%)
DNMT3Ac.2173 + 1G > A44.7348.9044.5447.86
DNMT3Ac.1937-8T > A49.3247.4845.0947.29
IDH1c.395G > A47.9346.9044.9449.01
NPM1c.860_863dup32.1132.2736.3234.47
FLT3c.1774_1833dup44.7946.1548.3144.22
EZH2c.179G > A--9.652.47
Abbreviations: VAF: variant allele frequency; -: no pathogenic variants found; black and blue numbers: depict mutations in myeloid genes commonly found in AML in both vehicle- and EC-70124-treated mice (black) or exclusively in EC-70124-treated mice (blue).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lopez-Millan, B.; Costales, P.; Gutiérrez-Agüera, F.; Díaz de la Guardia, R.; Roca-Ho, H.; Vinyoles, M.; Rubio-Gayarre, A.; Safi, R.; Castaño, J.; Romecín, P.A.; et al. The Multi-Kinase Inhibitor EC-70124 Is a Promising Candidate for the Treatment of FLT3-ITD-Positive Acute Myeloid Leukemia. Cancers 2022, 14, 1593. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14061593

AMA Style

Lopez-Millan B, Costales P, Gutiérrez-Agüera F, Díaz de la Guardia R, Roca-Ho H, Vinyoles M, Rubio-Gayarre A, Safi R, Castaño J, Romecín PA, et al. The Multi-Kinase Inhibitor EC-70124 Is a Promising Candidate for the Treatment of FLT3-ITD-Positive Acute Myeloid Leukemia. Cancers. 2022; 14(6):1593. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14061593

Chicago/Turabian Style

Lopez-Millan, Belen, Paula Costales, Francisco Gutiérrez-Agüera, Rafael Díaz de la Guardia, Heleia Roca-Ho, Meritxell Vinyoles, Alba Rubio-Gayarre, Rémi Safi, Julio Castaño, Paola Alejandra Romecín, and et al. 2022. "The Multi-Kinase Inhibitor EC-70124 Is a Promising Candidate for the Treatment of FLT3-ITD-Positive Acute Myeloid Leukemia" Cancers 14, no. 6: 1593. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14061593

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop