Figure 2 - uploaded by Rosemary E Gale
Content may be subject to copyright.
Distribution of the relative FLT3/TKD mutant level in 115 cases. 

Distribution of the relative FLT3/TKD mutant level in 115 cases. 

Source publication
Article
Full-text available
The prognostic impact of tyrosine kinase domain (TKD) mutations of the fms-like tyrosine kinase-3 (FLT3) gene in acute myeloid leukemia (AML) is currently uncertain. To resolve this issue we screened 1107 young adult nonacute promyelocytic leukemia AML patients with known FLT3 internal tandem duplication (ITD) status for FLT3/TKDs; they were detect...

Context in source publication

Context 1
... and therapeutic advances in recent years. 1 A classic example is Fms-like tyrosine kinase 3 (FLT3), a class III TK receptor expressed on the surface of normal hematopoietic progenitors and blast cells from most cases of acute myeloid leukemia (AML), 2 which is constitutively activated in many cases of AML due to the presence of internal tandem duplications (ITDs) within the juxtamembrane (JM) domain. 3-5 FLT3/ITDs occur in approximately 25% of cases of AML in younger adults, and in nonacute promyelocytic leukemia (non-APL) AML they predict for increased relapse rate from complete remission (CR) and reduced survival. 3-7 Subsequent to the identification of FLT3/ITDs in AML, it was noted that primary AML blasts from a number of patients had constitutively phosphorylated FLT3 in the absence of an FLT3/ ITD, 8 raising the possibility of other mechanisms of aberrant FLT3 activation. Shortly afterward, 2 groups independently reported activating mutations affecting codons 835 and 836 in the FLT3 second TK domain (TKD). 9,10 Presence of these mutations has been confirmed by others, and the overall incidence in AML was reported to be approximately 7%. 11-15 A number of different mutations have been reported. Most are point mutations such as substitution of asparate residue 835 with a tyrosine (D835Y, the most frequent mutation), histidine, valine, or glutamate; other alterations include small deletions ( ⌬ 835, ⌬ 836) and insertions. All mutations initially described disrupted an Eco RV restriction enzyme cutting site that facilitated a simple screening technique using digestion of polymerase chain reaction (PCR) products. Subse- quently, however, a number of other point mutations have been reported at codons 839, 841, and 842 16-18 as well as a 2 amino acid insertion between codons 840 and 841, 19 all of which occur outside of the Eco RV restriction digest site. The clinical significance of FLT3/TKD mutations in AML is unclear, and different groups have reported either no significant impact of an FLT3/TKD mutation on overall survival (OS) 9,12,13,15 or an adverse prognostic impact. 11 The differences in outcome in these studies may partly be due to the low incidence of the mutations, such that relatively small numbers of mutant-positive patients have been investi- gated, as well as heterogeneity in both treatment protocols and characteristics of the patients included. A recent meta-analysis of the above studies has suggested that the outcome of FLT3/TKD mutations may be similar to that of FLT3/ITDs. 20 This has important implications for the application of risk-adapted treatment in these patients, particularly in view of the debate regarding optimal therapy for FLT3/ITD ϩ patients 6,21 and the current interest in molecularly targeted therapy with FLT3 inhibitors. 22 To address this issue, we have screened a large cohort of young adult non-APL AML patients with known FLT3/ITD status, treated in the United Kingdom Medical Research Council (MRC) AML10 and AML12 trials, for FLT3/TKD mutations using denaturing high-performance liquid chromatography (dHPLC). We have examined the impact of mutations on clinical outcome and compared this with the impact of FLT3/ITDs. In the total cohort of 1107 patients, 127 (11%) were FLT3/TKD mutant-positive (FLT3/TKD ϩ ) by WAVE screening, of which 122 had a pattern consistent with a single mutant and 5 had evidence of 2 mutants. Mutations were initially determined by direct sequencing; however, it soon became apparent that the WAVE chromatogram pattern was specific to the underlying mutation and, therefore, most were identified by WAVE pattern and confirmed by mutation-specific restriction enzyme digest (Table 2; Figure 1). All mutants were identified and confirmed except those in 7 patients in whom the heteroduplex peak was very small, suggesting that the mutant present was only a small percentage of the total FLT3 alleles (Figure 1A). Overall, 13 different mutations were detected (Table 3). D835Y was the most common, accounting for 50% of mutant-positive patients; D835H, D835V, D835E, and ⌬ 836 were all found at a similar frequency of 6% to 12% of positive patients. Two novel mutations were detected, each in a single case: (1) a 3 bp insertion that replaced aspartate codon 835 with glycine and proline and (2) a 16 bp tandem duplication plus 10 bp deletion within the kinase domain that replaced codons 835 and 836 (DI) with VIPT. Five of the mutations detected (4%) occurred outside the Eco RV restriction digest site. The relative mutant level as a percentage of total FLT3 alleles was quantified in 115 samples using labeled PCR products and size separation after mutation-specific or Eco RV restriction enzyme digestion if required. Quantification was not performed in 12 cases—either because the mutation had not been identified (7 cases) or due to lack of material—and these were scored by visual estimation of the chromatogram. The distribution of the relative mutant level is shown in Figure 2. Mutants were arbitrarily classified as “higher” level, with greater than the median level of 25%, or “lower” level when less than or equal to this. Of the total cohort of 127 mutant-positive cases, a higher level of mutant was present in 62 (49%) (ie, 6% of all patients in the cohort), and the median mutant level in this group was 43% (range, 26% to 93%). Of note, only 2 patients showed evidence of biallelic mutation or loss of WT alleles, with a 88% and 93% mutant level, respectively. A lower level of mutant was present in 65 cases (51%) (ie, 6% of all patients), with a median level of 6% (range, 1% to 25%). In the 5 cases with 2 mutants, 4 of them had 1 major mutant of higher level plus a minor mutant of very low level (approximately 2% of total), and in the remaining case both mutants were lower level (less than 25% of total). Eco RV digestion and dHPLC analysis were both performed in 247 patients, including all 127 with an abnormal WAVE pattern: 120 were WT by both techniques, but 13 (10%) that were mutant by dHPLC were WT by Eco RV digestion. Four of these 13 cases had a mutant of higher level, but the mutations were outside of the Eco RV cutting site. Nine cases had a lower-level mutant by dHPLC. Of these, 5 were identified, all of which disrupted the Eco RV cutting site; 3 were quantified and found to be 3%, 3%, and 1% of total FLT3 alleles, respectively. Consequently, approximately one third of the mutants that were missed by Eco RV digestion were because the mutation did not change this cutting site, and the rest were below the detection level of the Eco RV technique. Details of the presenting features of the 1107 patients in the cohort are given in Table 1. The presence of a mutation was not related to sex or age. Mutations were found in all French-American-British (FAB) subtypes except M7. There was some evidence that FLT3/ TKD mutations were less frequent in patients with secondary AML compared with de novo disease ( P ϭ .02). The presence of an FLT3/TKD mutation correlated with a high presenting white blood cell count (WBC): median WBC, 38.3 ϫ 10 9 /L in mutant-positive patients and 23.0 ϫ 10 9 /L in WT patients ( P ϭ .006). This correlation was restricted to higher-level TKD mutants: median WBC, 49.0 ϫ 10 9 /L in higher-level mutants and 24.4 ϫ 10 9 /L in lower- level mutants ( P ϭ .002). Information on karyotype was available in 882 patients (Table 4). There was a high incidence of FLT3/TKD mutations in patients with inv(16) (24%, P ϭ .005) and a low incidence in cases with adverse cytogenetics (3%, P ϭ .008). There were a number of features associated with FLT3/TKD ϩ cases that differed markedly from those of FLT3/ITD ϩ cases. FLT3/TKD mutations were uncommon in patients with secondary AML, whereas in our cohort the incidence of FLT3/ITDs did not differ significantly between de novo and secondary AML, 23 although others have reported that FLT3/ITDs are less frequent in secondary AML. 11,12,26 Unlike FLT3/ITDs, which are particularly frequent in patients with normal cytogenetics, the only group in which FLT3/TKD mutations were more common were cases with inv(16). Of the 1107 non-APL AML patients, 723 (65%) had WT FLT3, 257 (23%) were FLT3/ITD ϩ only, 100 (9%) FLT3/TKD ϩ only, and 27 (2%) had both mutations. Outcome data were available on the whole cohort with a median follow-up of 7.9 years (range, 6 to 193 months). Of the 127 FLT3/TKD ϩ patients, 14 received an autograft and 17 an allograft. The CR rate for all 1107 patients was 85%, and the presence of an FLT3/TKD mutation did not influence the rate of CR, ID, or RD. In univariate analysis, patients with an FLT3/TKD mutation had a reduced CIR, improved RFS, and improved OS at 5 years (Table 5). Outcome in a significant proportion of the FLT3/TKD mutant- negative cases would be influenced by the presence of an FLT3/ ITD; therefore, the impact in the cohort of both types of mutation was examined. The CIR at 10 years was 68% for FLT3/ITD TKD patients, 62% for FLT3/ITD ϩ TKD Ϫ patients, 46% for FLT3/ ITD Ϫ TKD Ϫ patients, and 37% for FLT3/TKD ϩ ITD Ϫ patients (Figure 3A). OS at 10 years was 37%, 24%, 36%, and 51%, respectively (Figure 3B). A direct comparison of FLT3/TKD ϩ and FLT3/ITD ϩ cases demonstrated a highly significant difference in CIR (OR, 0.45; 95% confidence interval [CI], 0.33 to 0.61; P Ͻ .001) and OS (OR, 0.53; 95% CI, 0.41 to 0.69; P Ͻ .001). There was no evidence that the impact of an FLT3/TKD mutation on OS differed according to FLT3/ITD status (test for interaction, P ϭ .8), although there were only 27 patients with both types of mutation (Figure 3C). Kaplan-Meier curves for OS in the 3 cytogenetic risk groups stratified by FLT3/TKD status are shown in Figure 4A. When the cohort was stratified according to cytogenetic risk group, there was no heterogeneity or trend between the groups in the effect of an FLT3/TKD mutation on OS ( P ϭ .8, Figure 4B). Similarly, stratification according to specific cytogenetic ...

Similar publications

Article
Full-text available
Evaluation of molecular response is important for the diagnosis and monitoring of minimal residual disease in patients with acute promyelocytic leukemia (APL). In this study, we analyzed the molecular response by regular reverse transcription-polymerase chain reaction (RT-PCR) and quantitative real-time RT-PCR in 31 newly diagnosed patients. The re...

Citations

... Another class of FLT3 mutations in AML involves point mutations in the TKD [14][15][16] with the most common being a mutation at aspartic acid residue 835 (D835) [17]. These point mutations cause a permanent open conguration of the activation loop, resulting in constitutive signalling of the FLT3 receptor [18]. ...
Article
Full-text available
FLT3 mutations in Acute Myeloid
... Activating mutations of FMS-like tyrosine kinase 3 (FLT3) account for approximately 30% of acute myeloid leukemia (AML) patients and can be either FLT3 internal tandem duplication (FLT3-ITD) mutations or FLT3 tyrosine kinase domain point mutations (FLT3-TKD) . 1 In addition, mutation of FLT3-ITD occurs in 25% of diagnosed AML patients and which are associated with poor outcomes [1,2]. Whereas, FLT3-TKD have not been associated with a consistent prognostic impact on AML patients [3]. FLT3-ITD lead to receptor dimerization and ligand-independent constitutive activation of downstream signal transduction pathways, such as mitogen-activated protein kinase (MAPK) signal transducer and activator of transcription 5 (STAT5), which maintain the leukemia cells proliferation [4][5][6]. ...
Article
Full-text available
Background Acute myeloid leukemia (AML) with FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation accounts for a large proportion of AML patients and diagnosed with poor prognosis. Although the prognosis of FLT3-ITD AML has been greatly improved, the drug resistance frequently occurred in the treatment of FLT3 targeting drugs. GNF-7, a multitargeted kinase inhibitor, which provided a novel therapeutic strategy for overriding leukemia. In this study, we explored the antitumor activity of GNF-7 against FLT3-ITD and clinically-relevant drug resistance in FLT3 mutant AML. Methods Growth inhibitory assays were performed in AML cell lines and Ba/F3 cells expressing various FLT3 mutants to evaluate the antitumor activity of GNF-7 in vitro. Western blotting was used to examine the inhibitory effect of GNF-7 on FLT3 and its downstream pathways. Molecular docking and cellular thermal shift assay (CETSA) were performed to demonstrate the binding of FLT3 to GNF-7. The survival benefit of GNF-7 in vivo was assessed in mouse models of transformed Ba/F3 cells harboring FLT3-ITD and FLT3-ITD/F691L mutation. Primary patient samples and a patient-derived xenograft (PDX) model were also used to determine the efficacy of GNF-7. Results GNF-7 inhibited the cell proliferation of Ba/F3 cells expressing FLT3-ITD and exhibited potently anti-leukemia activity on primary FLT3-ITD AML samples. Moreover, GNF-7 could bind to FLT3 protein and inhibit the downstream signaling pathway activated by FLT3 including STAT5, PI3K/AKT and MAPK/ERK. In vitro and in vivo studies showed that GNF-7 exhibited potent inhibitory activity against FLT3-ITD/F691L that confers resistant to quizartinib (AC220) or gilteritinib. Importantly, GNF-7 showed potent cytotoxic effect on leukemic stem cells, significantly extend the survival of PDX model and exhibited similar therapy effect compared with gilteritinib. Conclusions Our results show that GNF-7 is a potent FLT3-ITD inhibitor and may become a promising lead compound applied for treating some of the clinically drug resistant patients.
... In addition to ITD, point mutations have been identified in the second TKD, causing the loop to take an active conformation that allows the tyrosine residues to become phosphorylated. The prevalence of these abnormalities is significantly lower than ITD and varies between 7 and 10% of patients newly diagnosed with AML [28,29]. The most frequent observed abnormality substitutes an aspartic acid at codon 835 for a tyrosine (D835Y) but replacements with a histidine (D835H), valine (D835V), or glutamate (D835E) have been identified as well. ...
Article
Full-text available
Acute myeloid leukemia (AML) is a heterogeneous clonal disease characterized overall by an aggressive clinical course. The underlying genetic abnormalities present in leukemic cells contribute significantly to the AML phenotype. Mutations in FMS-like tyrosine kinase 3 (FLT3) are one of the most common genetic abnormalities identified in AML, and the presence of these mutations strongly influences disease presentation and negatively impacts prognosis. Since mutations in FLT3 were identified in AML, they have been recognized as a valid therapeutic target resulting in decades of research to develop effective small molecule inhibitor treatment that could improve outcome for these patients. Despite the approval of several FLT3 inhibitors over the last couple of years, the treatment of patients with FLT3-mutated AML remains challenging and many questions still need to be addressed. This review will provide an up-to-date overview of our current understanding of FLT3-mutated AML and discuss what the current status is of the available FLT3 inhibitors for the day-to-day management of this aggressive disease.
... FLT3-ITD mutations have been consistently shown across most studies to be associated with a more aggressive phenotype and worse survival in AML, while most studies have shown no-or only minimal-prognostic impact of FLT3-TKD mutations [19,20,[27][28][29][30]. In patients with FLT3-ITD-mutated AML, a higher allelic ratio is associated with inferior outcomes, particularly when frontline FLT3 inhibitors are not used [31]. ...
Article
Full-text available
FLT3 is the most frequently mutated gene in acute myeloid leukemia (AML), with FLT3 internal tandem duplication (ITD) mutations being associated with a more aggressive clinical course. While two large, randomized clinical trials have shown a survival benefit with the frontline use of an oral FLT3 inhibitor (midostaurin or quizartinib) in patients with FLT3 -mutated AML, the role of FLT3 inhibitors in older adults with newly diagnosed FLT3 -mutated AML remains unclear. A definitive improvement in survival has not been observed in intensively treated patients over 60 years of age receiving frontline FLT3 inhibitors. Furthermore, many patients with FLT3 -mutated AML are unsuitable for intensive chemotherapy due to age and/or comorbidities, and this population represents a particular unmet need. For these older patients who are unfit for intensive approaches, azacitidine + venetoclax is a new standard of care and is used by many clinicians irrespective of FLT3 mutation status. However, FLT3 -ITD mutations confer resistance to venetoclax and are a well-established mechanism of relapse to lower-intensity venetoclax-based regimens, leading to short durations of remission and poor survival. Preclinical and clinical data suggest synergy between FLT3 inhibitors and venetoclax, providing rationale for their combination. Novel strategies to safely incorporate FLT3 inhibitors into the standard hypomethylating agent + venetoclax backbone are now being explored in this older, less fit population with newly diagnosed FLT3 -mutated AML, with encouraging early results. Herein, we discuss the frontline use of FLT3 inhibitors in older adults with FLT3 -mutated AML, including the potential role of FLT3 inhibitors in combination with intensive chemotherapy and as part of novel, lower-intensity doublet and triplet regimens in this older population.
... Page 3/17 mutations or FLT3 tyrosine kinase point mutations (FLT3-TKD) . 1 In addition, mutation of FLT3-ITD occurs in 25% of diagnosed AML patients and which are associated with poor outcomes [1,2]. Whereas, FLT3-TKD have not been associated with a consistent prognostic impact on AML patients [3]. FLT3-ITD lead to receptor dimerization and ligand-independent constitutive activation of downstream signal transduction pathways, such as mitogen-activated protein kinase (MAPK) signal transducer and activator of transcription 5 (STAT5), which maintain the leukemia cells proliferation [4][5][6]. ...
Preprint
Full-text available
Background Acute myeloid leukemia (AML) with FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation accounts for a large proportion of AML patients and diagnosed with dismay prognosis. Although the prognosis of FLT3-ITD AML has been greatly improved, the drug resistance frequently occurred in the treatment of FLT3 target drugs. GNF-7, a multitargeted kinase inhibitor, which provided a novel therapeutic strategy for overriding leukemia. In this study, we explored the antitumor activity of GNF-7 against FLT3-ITD and clinically-relevant drug resistance in FLT3 mutant AML. Methods Growth inhibitory assays were performed in AML cell lines and Ba/F3 cells expressing various FLT3 mutants to evaluate the antitumor activity of GNF-7 in vitro. Western blotting was used to examine the activity of FLT3 and its downstream pathways. Molecular docking was performed to predict the binding sites of FLT3 to GNF-7. The survival benefit of GNF-7 in vivo was assessed in mouse models of transformed Ba/F3 cells harboring FLT3-ITD and FLT3-ITD/F691L mutation. Primary patient samples and a patient-derived xenograft (PDX) model were also used to determine the efficacy of GNF-7. Results GNF-7 inhibited the cell proliferation of Ba/F3 cells expressing FLT3-ITD and exhibited potently anti-leukemia activity on primary FLT3-ITD AML samples. Moreover, GNF-7 could bind to FLT3 protein and inhibit the phosphorylation of downstream effectors in the FLT3 signaling pathways. In vitro and in vivo studies showed that GNF-7 exhibited a potent inhibitory activity against FLT3-ITD/F691L that confers resistant to quizartinib (AC220) or gilteritinib. Importantly, GNF-7 showed potent cytotoxic effect on leukemic stem cells, significantly extend the survival of PDX model and exhibited similar therapy effect compared with gilteritinib. Conclusions Our results show that GNF-7 is a potent FLT3-ITD inhibitor and may become a promising lead compound applied for treating some of the clinically drug resistant patients.
... [4] Although both mutations promote ligand-independent auto-phosphorylation and constitutive receptor activation, the FLT3-ITD mutation has more clinical impact than the FLT3-TKD mutation. [5,6] It has been proven in previous studies that FLT3-ITD is strongly associated with high blast counts, leucocytosis, increased risk of relapse, shorter overall survival (OS), normal cytogenetics, t(15; 17), t(6; 9), NPM1 and DNMT3A mutations. [7] The allelic ratio of FLT3 mutation also has prognostic significance in that a higher ratio correlates with higher relapse risk. ...
Article
Full-text available
Introduction: Molecular genetic abnormalities in acute myeloid leukaemia (AML) are essential for disease diagnosis and determining prognosis and clinical course. Mutations in FLT3 and nucleophosmin (NPM) genes are the most frequent genetic abnormalities, which are also known to impact disease outcomes. FLT3 mutations have been identified in approximately 30% of de novo AML patients and are associated with poor prognoses. This study aimed to determine the response to induction chemotherapy, overall survival (OS) and relapse rate (RR) in patients with FLT3-positive AML. Materials and methods: In this study, a retrospective analysis was performed of 75 newly diagnosed patients with AML registered between January 2015 and July 2022. Patient demographics and clinical-haematological parameters were noted and molecular analysis for FLT3 ITD/TKD and NPM mutations was performed. All the patients received standard induction chemotherapy and their response to treatment, OS and RR were assessed. Results: A total of 75 cases of AML were analysed. The mean age of the sample was 34.9 years, of which 65.3% were males and 34.7% were females. The patients were stratified into two groups: Those who were positive for FLT3 while negative for NPM (FLT3+/NPM-), representing 17.3% and those who were negative for both FLT3 and NPM (FLT3-/NPM-), representing 82.7% of cases. On day 28 post-induction, the complete remission rate was 69.2% in the FLT3 positive group and 77.4% in the FLT3 negative group. In the FLT3+/NPM- group, 55.6% of cases who were in remission at day 28 subsequently relapsed, compared to 50.0% of FLT3-/NPM- cases. The overall median survival time for the cohort and FLT3+ group was 1467 days, while that of the FLT3-group could not be estimated due to the very high survival rate. Conclusion: No significant differences in outcomes were observed in patients who were FLT3 positive compared to those who were FLT3 negative.
... Interestingly, FLT3-ITD and FLT3-TKD mutations are associated with different prognosis in patients with AML. FLT3-TKD mutations have a significantly better prognosis than FLT3-ITD (Li et al., 2023;Mead et al., 2007). Therefore, it can be seen that inhibition of FLT3 kinase activity might be served as a promising strategy for AML target therapy. ...
Article
Acute myeloid leukemia (AML) is a highly heterogeneous subtype of hematological malignancies with a wide spectrum of cytogenetic and molecular abnormalities, which makes it difficult to manage and cure. Along with the deeper understanding of the molecular mechanisms underlying AML pathogenesis, a large cohort of novel targeted therapeutic approaches has emerged, which considerably expands the medical options and changes the therapeutic landscape of AML. Despite that, resistant and refractory cases caused by genomic mutations or bypass signalling activation remain a great challenge. Therefore, discovery of novel treatment targets, optimization of combination strategies, and development of efficient therapeutics are urgently required. This review provides a detailed and comprehensive discussion on the advantages and limitations of targeted therapies as a single agent or in combination with others. Furthermore, the innovative therapeutic approaches including hyperthermia, monoclonal antibody-based therapy, and CAR-T cell therapy are also introduced, which may provide safe and viable options for the treatment of patients with AML.
... 4 And the presence of FLT3-TKD mutations correlates with a better prognosis than FLT3-ITD mutations. 3,5,6 Fragment analysis (FA) by polymerase chain reaction (PCR) and capillary electrophoresis assay is the current standard approach for identifying FLT3 mutations because this strategy is rapid, easy, affordable and allows screening for ITD mutant to wild-type allele ratio (WT AR) and TKD mutations at codons D835 and I836 according to the 2017 ELN recommendations. 4 However, FA provided limited information for patients' management. ...
Article
Full-text available
Fms‐like tyrosine kinase 3 (FLT3) is frequently mutated in haematological malignancies. Although canonical FLT3 mutations including internal tandem duplications (ITDs) and tyrosine kinase domains (TKDs) have been extensively studied, little is known about the clinical significance of non‐canonical FLT3 mutations. Here, we first profiled the spectrum of FLT3 mutations in 869 consecutively newly diagnosed acute myeloid leukaemia (AML), myelodysplastic syndrome and acute lymphoblastic leukaemia patients. Our results showed four types of non‐canonical FLT3 mutations depending on the affected protein structure: namely non‐canonical point mutations (NCPMs) (19.2%), deletion (0.7%), frameshift (0.8%) and ITD outside the juxtamembrane domain (JMD) and TKD1 regions (0.5%). Furthermore, we found that the survival of patients with high‐frequency (>1%) FLT3‐NCPM in AML was comparable to those with canonical TKD. In vitro studies using seven representative FLT3‐deletion or frameshift mutant constructs showed that the deletion mutants of TKD1 and the FLT3‐ITD mutant of TKD2 had significantly higher kinase activity than wild‐type FLT3, whereas the deletion mutants of JMD had phosphorylation levels comparable with wild‐type FLT3. All tested deletion mutations and ITD were sensitive to AC220 and sorafenib. Collectively, these data enrich our understanding of FLT3 non‐canonical mutations in haematological malignancies. Our results may also facilitate prognostic stratification and targeted therapy of AML with FLT3 non‐canonical mutations.
... Indeed, patients harboring NPM1mut and FLT3-ITDhigh are classified as intermediate risk in ELN2017 [23] . Different is the case for AML with FLT3-TKD; for these patients, a clear impact on prognosis is not established due to conflicting results [24][25][26][27][28][29][30] . However, in the new ELN2022 classification, probably due to the therapeutic benefit of FLT3 inhibitors in the new AML scenario, all patients with FLT3 mutation are classified as intermediate risk regardless of whether the mutation is detected alone or with other co-mutations. ...
Article
Full-text available
FMS-related tyrosine kinase 3 (FLT3) mutations, present in about 25%-30% of acute myeloid leukemia (AML) patients, constitute one of the most frequently detected mutations in these patients. The binding of FLT3L to FLT3 activates the phosphatidylinositol 3-kinase (PI3K) and RAS pathways, producing increased cell proliferation and the inhibition of apoptosis. Two types of FLT3 mutations exist: FLT3-ITD and FLT3-TKD (point mutations in D835 and I836 or deletion of codon I836). A class of drugs, tyrosine-kinase inhibitors (TKI), targeting mutated FLT3, is already available with 1st and 2nd generation molecules, but only midostaurin and gilteritinib are currently approved. However, the emergence of resistance or the selection of clones not responding to FLT3 inhibitors has become an important clinical dilemma, as the duration of clinical responses is generally limited to a few months. This review analyzes the insights into mechanisms of resistance to TKI and poses a particular view on the clinical relevance of this phenomenon. Has resistance been overlooked? Indeed, FLT3 inhibitors have significantly contributed to reducing the negative impact of FLT3 mutations on the prognosis of AML patients who are no longer considered at high risk by the European LeukemiaNet (ELN) 2022. Finally, several ongoing efforts to overcome resistance to FLT3-inhibitors will be presented: new generation FLT3 inhibitors in monotherapy or combined with standard chemotherapy, hypomethylating drugs, or IDH1/2 inhibitors, Bcl2 inhibitors; novel anti-human FLT3 monoclonal antibodies (e.g., FLT3/CD3 bispecific antibodies); FLT3-CAR T-cells; CDK4/6 kinase inhibitor (e.g., palbociclib).
... In contrast, the presence of mutated FLT3-TKD in addition to FLT3-ITD showed very favorable outcomes after allogeneic HSCT, especially in patients with a low FLT3-ITD allelic ratio ( Figure 3C,D and Supplementary Figure S6). While our study is the first to exclusively analyze FLT3-ITD mutated patients, others already suggested improved outcomes in FLT3-TKD mutated AML patients, either alone or when co-mutated NPM1 was present [6,40], but this was not consistent across all reports [7]. ...
Article
Full-text available
Simple Summary The presence of FLT3-ITD is among the most common molecular aberrations in acute myeloid leukemia (AML). Although patients harboring FLT3-ITD are often consolidated by allogeneic hematopoietic stem cell transplantation (HSCT), little is known about how the FLT3-ITD allelic ratio impacts patient outcomes after HSCT. Here, we analyzed the biological and clinical features of these patients in the context of other risk factors, including the ELN2017 risk and the measurable residual disease status (MRD) at HSCT. Our data shows no survival differences between patients with a high or low FLT3-ITD allelic ratio in the context of an allogeneic HSCT, but highlights the importance of pre-HSCT MRD as a prognostic factor. Abstract Although the presence of FLT3-ITD, as well as levels of the FLT3-ITD allelic ratio, have been described as prognostic factors in acute myeloid leukemia (AML), little is known about how the FLT3-ITD allelic ratio impacts patients’ outcomes when receiving an allogeneic hematopoietic stem cell transplantation (HSCT). We analyzed 118 patients (median age at diagnosis 58.3, range 14.3–82.3 years) harboring FLT3-ITD, of whom 94 patients were consolidated with an allogeneic HSCT and included in outcome analyses. A high FLT3-ITD allelic ratio was associated with a higher white blood cell count, higher blood and bone marrow blasts, and worse ELN2017 risk at diagnosis. Patients with a high FLT3-ITD allelic ratio more often had NPM1 mutations, while patients with a low allelic ratio more often had FLT3-TKD mutations. Patients with a high FLT3-ITD allelic ratio were less likely to achieve a measurable residual disease (MRD)-negative remission prior to allogeneic HSCT and had a trend for a shorter time to relapse. However, there was no distinct cumulative incidence of relapse, non-relapse mortality, or overall survival according to the FLT3-ITD allelic ratio in transplanted patients. While co-mutated FLT3-TKD was associated with better outcomes, the MRD status at HSCT was the most significant factor for outcomes. While our data indicates that an allogeneic HSCT may mitigate the adverse effect of a high FLT3-ITD allelic ratio, comparative studies are needed to evaluate which FLT3-ITD mutated patients benefit from which consolidation strategy.