21,430
Views
12
CrossRef citations to date
0
Altmetric
Review

The importance of FLT3 mutational analysis in acute myeloid leukemia

ORCID Icon
Pages 2273-2286 | Received 19 Jul 2017, Accepted 24 Oct 2017, Published online: 22 Nov 2017

Abstract

Activating mutations in FMS-like tyrosine kinase 3 (FLT3), including internal tandem duplications (ITDs) and tyrosine kinase domain (TKD) mutations, are common in patients with acute myeloid leukemia (AML). FLT3-ITD is a negative prognostic factor that remains prognostically relevant even after intensive chemotherapy and/or stem cell transplant. FLT3 testing was historically viewed as being purely prognostic; however, with the advent of FLT3 inhibitors, it will likely be seen as both prognostic and predictive. The multikinase inhibitor midostaurin, in combination with chemotherapy, is the first targeted agent to significantly prolong survival in patients with newly diagnosed FLT3-mutated AML and was recently approved by health authorities. Recently, the European LeukemiaNet recommended FLT3 testing (both TKD and ITD) for all patients with AML, with results required within 3 days. The need for optimized, multigene platform testing incorporating FLT3 mutations will increase as knowledge of interactions between FLT3 and other myeloid-relevant mutations grows.

FLT3 mutations in AML

FMS-like tyrosine kinase 3 (FLT3), a member of the type III receptor tyrosine kinase family [Citation1,Citation2], is expressed in ≈90% of leukemic blasts of patients with acute myeloid leukemia (AML) [Citation3,Citation4]. FLT3 mutations occur in approximately one-third of patients with AML () [Citation5–13]. In-frame duplications of 3 to >400 base pairs (bp), known as internal tandem duplications (ITDs), are the most common, occurring in up to 30% of adult patients with de novo AML [Citation5,Citation6,Citation14]. However, FLT3-ITD is not expressed equally among patients with FLT3-ITD–positive (FLT3-ITD+) disease [Citation15]. Differences in expression levels, measured using the FLT3-ITD-to-wild-type (WT) allelic ratio, impact prognosis [Citation16]. This ratio is a measure of the relative signal intensity derived from the fluorescently labeled products amplified from the FLT3-ITD and FLT3-WT alleles using a polymerase chain reaction (PCR) assay [Citation17,Citation18]. Consensus is that a high FLT3-ITD-to-WT allelic ratio is a negative prognostic factor [Citation17,Citation19–25]; however, until recently, no standard definition existed as to what cutoff distinguished a low vs high allelic ratio. The 2017 European LeukemiaNet (ELN) guidelines defined 0.5 as the cutoff between low (FLT3-ITDlow; <0.5) and high (FLT3-ITDhigh; ≥0.5) allelic ratios [Citation16].

Figure 1. FMS-like tyrosine kinase 3 (FLT3) contains 5 functional domains: an immunoglobulin-like extracellular domain, a transmembrane domain, a juxtamembrane domain (JMD), an interrupted tyrosine kinase domain (TKD), and a small C-terminal domain. Internal tandem duplications (ITDs), insertions of 3 to >400 base pairs (bp), are the most common mutations in FLT3. ITDs occur in up to 30% of patients with acute myeloid leukemia (AML); of these, 69.5% are located in the JMD and 30.5% are located in the TKD (25.8% in the beta1-sheet and 4.6% in other regions). Activating mutations within the TKD occur in up to 14% of patients with AML; of these, 90.5% are located within the activation loop of the TKD2 and 9.5% are located within the TKD1. Additional activating mutations have been identified at the very low frequency within the extracellular domain (<1% of cases) and the JMD (<1–2% of cases) [Citation5–13]. aAdditional point mutations that have been identified in patients with AML – but have not been found to be activating mutations in vitro – include mutations within the extracellular domain (e.g. T167, V194, D324, Y364, and V491), transmembrane domain (e.g. I548 and V557), JMD (e.g. V579 and E598), TKD1 (e.g. A680 and M737), and TKD2 (e.g. V816, A814, and T784) [Citation6,Citation8,Citation9]. bThe majority of mutations within the TKD are point mutations that result in amino acid changes; however, activating mutations caused by insertions (e.g. insertion of glycine and serine between residues S840 and N841 [S840GS]) and deletions (e.g. ΔI836 and ΔE598/Y599) have also been identified in the TKD [Citation6,Citation10,Citation11,Citation13].

Figure 1. FMS-like tyrosine kinase 3 (FLT3) contains 5 functional domains: an immunoglobulin-like extracellular domain, a transmembrane domain, a juxtamembrane domain (JMD), an interrupted tyrosine kinase domain (TKD), and a small C-terminal domain. Internal tandem duplications (ITDs), insertions of 3 to >400 base pairs (bp), are the most common mutations in FLT3. ITDs occur in up to 30% of patients with acute myeloid leukemia (AML); of these, 69.5% are located in the JMD and 30.5% are located in the TKD (25.8% in the beta1-sheet and 4.6% in other regions). Activating mutations within the TKD occur in up to 14% of patients with AML; of these, 90.5% are located within the activation loop of the TKD2 and 9.5% are located within the TKD1. Additional activating mutations have been identified at the very low frequency within the extracellular domain (<1% of cases) and the JMD (<1–2% of cases) [Citation5–13]. aAdditional point mutations that have been identified in patients with AML – but have not been found to be activating mutations in vitro – include mutations within the extracellular domain (e.g. T167, V194, D324, Y364, and V491), transmembrane domain (e.g. I548 and V557), JMD (e.g. V579 and E598), TKD1 (e.g. A680 and M737), and TKD2 (e.g. V816, A814, and T784) [Citation6,Citation8,Citation9]. bThe majority of mutations within the TKD are point mutations that result in amino acid changes; however, activating mutations caused by insertions (e.g. insertion of glycine and serine between residues S840 and N841 [S840GS]) and deletions (e.g. ΔI836 and ΔE598/Y599) have also been identified in the TKD [Citation6,Citation10,Citation11,Citation13].

Mutations within the tyrosine kinase domain (TKD) are the second most common type of FLT3 mutation in AML (occurring in up to 14% of adult patients with AML) [Citation13,Citation17,Citation26]. Mutations within the TKD are primarily point mutations within the activation loop (e.g. residues D835, I836, and Y842) of the TKD2 [Citation6,Citation13,Citation18,Citation27] and, to a lesser extent, within the TKD1 (e.g. residues N676 and F691) [Citation12,Citation27]. Other point mutations and smaller insertions/deletions have also been identified within the TKD and other domains (e.g. extracellular and juxtamembrane domains [occurring in ≈2% of patients with AML]) [Citation8,Citation9,Citation11,Citation13]. The prognostic significance of FLT3-TKD mutations in the overall AML population and the impact of the FLT3-TKD allelic ratio are still debatable and may depend on additional mutations as well as the cytogenetic background [Citation13,Citation24].

Both FLT3-ITD and FLT3-TKD mutations are common in patients with AML with normal karyotype (30–39% and 6–14%, respectively), but they are also associated with karyotypic abnormalities, such as t(15;17)/PML-RARA (30–39% and 8–9%, respectively) and core binding factor AML (5–8% and 4–14%, respectively) [Citation13,Citation17,Citation26,Citation28,Citation29]. FLT3-ITD is also frequently associated with t(6;9) [DEK-NUP214] abnormalities (in up to 90% of patients) [Citation17,Citation30,Citation31]. Importantly, the prognostic impact of FLT3 mutations can vary by cytogenetic group. For example, in patients with t(15;17) abnormalities, there was no difference in outcome between those with and without FLT3-ITD mutations; however, patients with FLT3-TKD had significantly worse outcomes (compared with those with FLT3-WT) [Citation13,Citation17,Citation32]. Furthermore, recent advances indicate that the prognosis for patients with FLT3 mutations can be affected by the presence or absence of additional mutations [Citation14,Citation16,Citation33]. For example, patients who are FLT3-ITD negative (FLT3-ITD−) or FLT3-ITDlow and positive for nucleophosmin 1 mutations (NPM1+) have a favorable prognosis, whereas those who are FLT3-ITD − or FLT3-ITDlow with NPM1-WT or FLT3-ITD + and NPM1+ have an intermediate prognosis. Patients who are FLT3-ITDhigh with NPM1-WT have a poor prognosis [Citation16] and are less likely to achieve complete remission (CR) with induction chemotherapy than patients with other FLT3/NPM1 combinations (p < .005) [Citation34].

FLT3 testing: a prognostic marker

Current FLT3 testing landscape

Historically, patients with AML were stratified into risk groups based on age, performance status, white blood cell count, and cytogenetics [Citation35]. Subsequently, gene mutations (e.g. NPM1, FLT3, TP53, and CEBPA) were recognized as important prognostic factors and thus included in testing recommendations in the United States and Europe [Citation16,Citation36]. Until recently, FLT3 testing was recommended as a prognostic marker only in patients with cytogenetically normal AML. However, new recommendations for FLT3 testing in all patients with AML are a result of the approval of the first FLT3-targeted therapy, midostaurin, and the recognition that FLT3 is a negative prognostic marker, regardless of cytogenetics [Citation16,Citation36–37]. Importantly, results of FLT3 testing should be made available within 48–72 h after the initial diagnosis of AML so that targeted therapy can be initiated in a timely manner [Citation16].

Little information exists on the real-world FLT3 testing rates in patients with AML, but a retrospective chart review suggests that despite the recommendations, FLT3 testing is not always performed, even in patients with cytogenetically normal AML. According to a retrospective registry review of molecular marker testing performed at a single referral center between 2010 and 2012, only 77% of patients with cytogenetically normal AML were routinely tested for FLT3 [Citation38]. Furthermore, there is a gap in molecular testing rates (including FLT3) between academic centers and community referral sites, as suggested by the results of a single-institution retrospective chart review that analyzed molecular testing rates over time (2008–2012). Despite an increase in testing over time, testing rates were significantly higher at academic centers than at community sites (93% vs 41%; p < .001) [Citation39]. Routine testing for FLT3 in patients with cytogenetically normal AML had been recommended since at least 2010 [Citation40], which corresponds to the time at which molecular testing was routinely performed in 100% of patients at academic centers but not at community sites [Citation39]. This suggests that there is a lack of awareness or knowledge about the importance of molecular testing at community sites. More recently (2015), 294 members of professional societies in the United States and Europe were surveyed about their testing practices. Among responders, 51 and 46% indicated that they tested for FLT3-ITD in all patients and selected patients, respectively [Citation41]. This survey was intended to provide a baseline for testing prior to the release of the diagnostic workup guidelines jointly issued by the College of American Pathologists and the American Society of Hematology in 2017 [Citation36]. It would be expected that testing rates, particularly those for FLT3, will soon increase given that FLT3-targeted therapies are entering the market. One potential hurdle to widespread FLT3 testing in the past was the lack of commercially available tests. It will be interesting to see whether testing rates at community sites will catch up to those at academic centers – especially now that commercially developed FLT3 testing assays are routinely incorporated into clinical trials and are beginning to hit the market [Citation42,Citation43].

Methods for testing FLT3

The first method for the prognostic identification of FLT3-ITD mutations involved PCR amplification and subsequent sequencing of the juxtamembrane domain region within the FLT3 gene [Citation44]. Since then, several methods have been developed or adapted for identifying mutations and aberrant karyotypes () [Citation45–51]. These methods vary in their sensitivity, turnaround time, and development stage [Citation52]. Some methods have been used in the clinic for >10 years, while others are still being validated.

Table 1. Comparison between FLT3 testing methods.

The first method to be readily adopted and widely used in clinical trials is a modified PCR technique that uses capillary electrophoresis to resolve fluorescently labeled PCR products and can measure the FLT3-ITD-to-WT allelic ratio [Citation18]. Subsequently, a multiplex PCR assay was developed that uses two sets of fluorescently labeled primers to simultaneously amplify the ITD and D835 mutant regions [Citation50]. The resulting PCR products are then digested with EcoRV restriction endonuclease and resolved using capillary electrophoresis. FLT3-ITD mutations are identified by comparing the size of the amplification products (the reference WT product is 330 bp; ITDs are >330 bp). Mutations in D835 and I836 remove a naturally occurring EcoRV restriction endonuclease site in the WT amplification product, resulting in a larger fluorescently labeled fragment (129 bp; the WT product is 80 bp). Real-time quantitative PCR (RT-qPCR)–based tests have been proposed as alternatives for detecting FLT3-ITD, FLT3-TKD, and other point mutations [Citation53,Citation54] and can also be used for monitoring disease progression (see Role in detection of minimal residual disease). PCR-based methods have short turnaround times [Citation50,Citation51] and are highly selective. Their major limitation is that very few FLT3-TKD point mutations can be detected unless the PCR products are sequenced.

More recently, next-generation sequencing (NGS) approaches have been developed that are capable of screening many molecular markers. These NGS approaches can be broadly divided into two large groups: whole-genome sequencing, which captures the entire genome; and whole-exome sequencing, which selects for protein coding regions within the genome [Citation46]. Despite their tremendous potential, NGS approaches are currently not suitable for the clinic: they generate large amounts of data that can be overwhelming for hematologists and may not provide additional value for the diagnosis and treatment of patients with AML. They also have long turnaround times. Additionally, FLT3-ITD is inherently difficult to detect using NGS approaches [Citation46,Citation47,Citation55].

Multiplex-targeted NGS approaches, also known as gene panels, are more suitable for the clinic because they have rapid turnaround times and are highly sensitive for detecting variant alleles [Citation46]. Using a recently validated 54-gene panel, researchers identified FLT3-ITDs of varying lengths and insertion sites at lower thresholds than conventional methods could detect [Citation48]. Karyogene, a recently developed diagnostic tool that uses DNA capture to enrich for specific genes and cytogenetic abnormalities sequenced by high-throughput sequencing and analyzed with open-source software, was able to detect 49 predefined recurrent gene mutations, four chromosomal rearrangements, and several copy number aberrations in 62 samples from patients with AML [Citation45]. Adopting a technology such as Karyogene has its advantages (e.g. it integrates cytogenetic and molecular diagnosis into a single method and has a relatively short turnaround time [<10 d]) [Citation45] and disadvantages (e.g. it requires specialized high-throughput sequencing equipment and technical knowledge and skills).

Similarly, the use of gene panels for FLT3 testing has both advantages and disadvantages. An advantage is that this technology can detect rare mutations and could aid in enrolling patient subgroups into clinical trials to better understand the impact of such mutations. For example, this technology would be useful to determine the prognostic and therapeutic impact of the recently identified, rare N767 mutation that confers resistance to certain FLT3 inhibitors in vitro [Citation12,Citation56]. A potential disadvantage is that gene panel testing can have longer turnaround times (3–20 d) [Citation46] than conventional PCR-based methods (48–72 h) currently used to screen patients in clinical trials [Citation16,Citation57,Citation58].

Treatment for patients with FLT3-mutated AML

Until recently, the standard of care for patients with AML – induction and consolidation chemotherapy – remained unchanged for >25 years [Citation59,Citation60]. Outside the context of a clinical trial, therapy for patients with newly diagnosed AML depends on age, fitness level, and eligibility to receive intensive induction chemotherapy [Citation16,Citation59]. Most fit patients generally receive intensive anthracycline- and cytarabine-based induction chemotherapy, whereas older or unfit patients may receive lower-intensity induction chemotherapies (e.g. low-dose cytarabine or hypomethylating agents). For patients who achieve CR, the choice of consolidation therapy depends on their risk stratification group (i.e. favorable, intermediate, or unfavorable): patients with favorable risk receive high-dose cytarabine, whereas patients with intermediate or unfavorable risk in first complete remission (CR1) often undergo allogeneic hematopoietic stem cell transplant (alloHSCT), if eligible [Citation16,Citation59]. Before now, no targeted therapies were approved for patients with FLT3-mutated AML [Citation37,Citation61]. Despite this, outcomes in patients with FLT3 mutations have improved over the past 15 years [Citation62]. In a retrospective study of patients with AML evaluated at a single institution from 2000 to 2014, an increasing number underwent HSCT over time; and those who underwent HSCT, particularly in CR1, had improved survival compared with patients who did not receive an HSCT. In the study, a trend toward better response rates was seen in patients who received first-line chemotherapy in combination with FLT3 inhibitors (mostly in the setting of a clinical trial) compared with those who did not [Citation62].

Role of alloHSCT therapy

Transplant rates have increased significantly over the past 20 years, accompanied by increasing survival rates in patients with AML [Citation62,Citation63]. AlloHSCT is usually recommended for patients with FLT3-ITD mutations in CR1 who are eligible for transplant therapy and have a suitable donor [Citation16,Citation59]. These recommendations are supported by data from retrospective analyses [Citation19,Citation64–70] but have yet to be validated in prospective trials.

Among patients with FLT3-ITD mutations in CR1, those who undergo alloHSCT have significantly better outcomes (e.g. prolonged survival and decreased risk of relapse) than those who receive chemotherapy alone [Citation65]. Patients who have FLT3-ITDhigh [Citation19,Citation64,Citation70] or FLT3-ITDlow with NPM1-WT derive the most benefit from alloHSCT [Citation19,Citation64]. Despite this, FLT3-ITD remains a poor prognostic factor following alloHSCT [Citation63,Citation71]. Results from early-phase and retrospective studies suggest that patients with FLT3-ITD AML may benefit from the use of FLT3 tyrosine kinase inhibitors (TKIs) as maintenance therapy to prevent relapse following alloHSCT [Citation19,Citation72–75] – a hypothesis currently being investigated in clinical trials [Citation76–80]. In the United States, the TKI sorafenib is often used off-label as post-transplant maintenance therapy [Citation81].

Importantly, an analysis of alloHSCT rates outside the clinical trial setting revealed that less than half of patients who achieved CR went on to receive alloHSCT in CR1 (49.1%) [Citation82], suggesting that real-world transplant strategies need streamlining.

FLT3 inhibitors

Multiple small-molecule TKIs that target FLT3 are in development for the treatment of patients with AML () and have demonstrated clinical activity as a single agent or in combination with chemotherapy [Citation19,Citation27,Citation58,Citation72–75,Citation83–98]. Several FLT3 TKIs – including the multikinase inhibitors midostaurin and sorafenib and the more-selective FLT3 inhibitors crenolanib, gilteritinib, and quizartinib – are currently being evaluated or have completed evaluation in phase 3 clinical trials () [Citation58,Citation78,Citation80,Citation99–108]. Each of these FLT3 TKIs has advantages and disadvantages. It has recently been proposed that multikinase inhibitors, such as midostaurin and sorafenib, are better suited as first-line therapy because of the polyclonal nature of AML, whereas more-selective agents, such as crenolanib, gilteritinib, and quizartinib, are more appropriate in the relapsed/refractory (R/R) setting [Citation81]. Furthermore, even though all FLT3 TKIs have demonstrated inhibitory activity against ITD mutations, not all of them target important TKD mutations, such as the F691L ‘gatekeeper’ resistance mutation [Citation27,Citation84–86,Citation91,Citation92,Citation95,Citation109].

Table 2. Comparison of FLT3 inhibitors in late-phase clinical trials.

Table 3. Ongoing phase 3 clinical studies evaluating FLT3 inhibitors in patients with AML.

In the Randomized AML Trial in FLT3 patients <60 Years old (RATIFY), the largest study conducted to date in adult patients (aged 18 to <60 years) with newly diagnosed AML with FLT3 mutations (ITD and TKD), midostaurin in combination with intensive induction and consolidation chemotherapy and as single-agent maintenance therapy reduced the risk of death compared with placebo by 22% and improved event-free survival (EFS) and disease-free survival [Citation58]. The benefit in overall survival (OS) and EFS was independent of HSCT and FLT3 mutation status (FLT3-ITDhigh [≥0.7], FLT3-ITDlow [<0.7], or FLT3-TKD). Grade 3/4 adverse events were comparable between the two arms except for rash, which was more common in the midostaurin arm. Midostaurin, in combination with induction and consolidation chemotherapy, became the first FLT3 TKI approved in the United States [Citation37] and is listed as a potential therapy for patients with FLT3-mutated AML beginning in version 2 of the National Comprehensive Cancer Network guidelines [Citation59] and the 2017 ELN recommendations [Citation16]. Additional ongoing studies are evaluating midostaurin as frontline treatment for FLT3-ITD + AML (patients aged 18–70 years) in combination with lower-intensity therapies and as maintenance therapy following HSCT [Citation76,Citation79,Citation110,Citation111].

Sorafenib, in combination with standard chemotherapy, was evaluated in adults (aged 18–60 years) with newly diagnosed AML in the randomized, placebo-controlled, phase 2 Sorafenib in AML in patients ≤60 years (SORAML) trial [Citation112]. Sorafenib demonstrated significant improvement compared with placebo in EFS (p = .013) and relapse-free survival (p = .017) but not OS (p = .382) in all patients; a similar trend in improvement, albeit not significant, was observed in patients with FLT3-ITD mutations (only 17% of patients had FLT3-ITD mutations). Sorafenib was associated with an increased risk of bleeding, fever, and hand-foot syndrome [Citation112]. Addition of sorafenib to intensive chemotherapy did not result in clinical benefit (no significant improvements were observed in EFS or OS compared with placebo, and there was an increased rate of early death compared with placebo) in older patients (aged 61–80 years) [Citation113]. Sorafenib was the first agent to demonstrate single-agent activity as maintenance therapy following HSCT. Promising results from several phase 1 and retrospective studies in patients with FLT3-ITD + AML [Citation19,Citation72–75,Citation83] have led to a flurry of new studies evaluating FLT3 TKIs in this setting (). Sorafenib showed promising activity in combination with azacitidine and decitabine (phase 2 and single-institution retrospective studies, respectively) in patients with FLT3-ITD + R/R AML [Citation114,Citation115]. Sorafenib is currently being investigated as frontline treatment for AML in combination with azacitidine in patients not eligible for standard chemotherapy and as single-agent maintenance therapy following alloHSCT [Citation80,Citation107,Citation116]. In the United States, sorafenib is routinely used off-label as single-agent maintenance therapy following transplant [Citation81] and in combination with hypomethylating agents as salvage therapy in patients with FLT3-mutated R/R AML [Citation59,Citation81].

Crenolanib, gilteritinib, and quizartinib have demonstrated single-agent activity in patients with R/R AML with FLT3 mutations [Citation89,Citation94,Citation98]. Among these agents, quizartinib is the most selective FLT3-ITD inhibitor and has shown the strongest single-agent activity in this patient population (). Despite initial safety concerns about QT prolongation with quizartinib in early studies, it has not been an issue in subsequent studies evaluating lower doses in which high response rates have been maintained [Citation89]. Quizartinib is being evaluated in a phase 3, randomized study compared with salvage chemotherapy in patients with FLT3-ITD + R/R AML [Citation102] and as frontline treatment for patients with FLT3-ITD + AML [Citation106].

Crenolanib is currently being investigated in combination with salvage chemotherapy in two randomized, placebo-controlled, phase 3 studies in patients with R/R AML [Citation100,Citation103]. Crenolanib has shown promising activity in combination with intensive induction (cytarabine + daunorubicin or idarubicin) and consolidation chemotherapy in newly diagnosed FLT3-mutated AML, as demonstrated by the high overall response rates (CR/CR with incomplete blood count recovery: 88%) observed in preliminary analyses of an ongoing phase 2 study [Citation97]. Crenolanib is also being evaluated in a phase 3 study (compared with midostaurin) in combination with induction and consolidation chemotherapy in newly diagnosed FLT3-mutated AML [Citation108].

Gilteritinib, a highly selective FLT3-mutant inhibitor (including F691L), has undergone rapid development after initial promising single-agent activity [Citation94]; there are currently five ongoing phase 3 trials. The first two trials are evaluating gilteritinib compared with salvage chemotherapy in patients with R/R AML [Citation101,Citation104]. A third trial is evaluating gilteritinib alone or in combination with azacitidine compared with azacitidine alone in patients with newly diagnosed AML who are not eligible for intensive chemotherapy [Citation105]. Two additional trials are evaluating single-agent gilteritinib maintenance therapy following either alloHSCT or induction/consolidation chemotherapy in patients with FLT3-ITD + AML [Citation78,Citation99].

Additional studies of FLT3 inhibitors include early-phase trials of single-agent FLX925 (NCT02335814) and TAK-659 (NCT02323113) in R/R AML. A study of E6201 in patients with FLT3-mutated R/R AML or older patients (aged ≥60 years) with newly diagnosed AML who are not eligible for standard chemotherapy (NCT02418000) was recently terminated. Ponatinib, originally developed as a BCR-ABL1 inhibitor, has shown preclinical activity in FLT3-mutated AML models in vitro [Citation117]; ongoing early-phase trials are evaluating ponatinib as frontline treatment for AML (NCT02779283) and as maintenance therapy in patients with FLT3-ITD AML in CR1 (NCT02428543).

New role of FLT3 testing: diagnostic marker that drives therapy

Given the increasing knowledge of AML pathobiology and advances in FLT3 testing methods, the current FLT3 testing paradigm is likely to evolve. New risk-stratification models have been proposed that integrate the identification of additional molecular markers into the routine diagnostic workup [Citation14,Citation118]. More-radical proposals forgo cytogenetic testing and suggest implementing molecular markers as the sole determinant of risk stratification [Citation119]. Adopting such a model would increase FLT3 testing rates. Currently, some guidelines recommend FLT3 testing for patients with normal cytogenetics only [Citation120], but patients with abnormal cytogenetics also harbor FLT3 mutations [Citation6].

FLT3 testing will continue to be an important prognostic determinant and can guide therapeutic decisions [Citation16,Citation37]; thus, demand for rapid FLT3 testing will likely increase in the future. There are three major areas that are critical to ensure that FLT3 testing is clinically relevant: (1) universal adoption, (2) rapid turnaround times, and (3) harmonization. First, barriers to adoption can be overcome by increasing awareness about and access to FLT3 testing. As previously mentioned, FLT3 testing is now recommended for all patients with AML, and commercial kits are now available. Second, rapid turnaround times (<8 d) are required for patients with newly diagnosed AML to be able to receive midostaurin (the only approved FLT3 inhibitor to date) in combination with chemotherapy [Citation37,Citation61]. Current recommendations requiring FLT3 testing results within 72 h [Citation16] are well within these rapid turnaround times. However, it is not clear whether this benchmark will be met in the real-world setting. Third, given that the FLT3-ITD-to-WT allelic ratio is a determinant of risk stratification [Citation16], harmonization of FLT3 testing will be important to ensure that comparable results are achieved regardless of measurement procedure, time, or location of testing [Citation121,Citation122]. Currently, harmonization of FLT3 testing will likely focus on PCR-based methods; however, in the future, NGS approaches that incorporate multigene panels could be the norm.

Role in detection of minimal residual disease

The term ‘minimal (or measurable) residual disease’ (MRD) is used to define the low levels of leukemic clones that may persist in patients who achieve a morphological CR and have a higher risk of relapse. These leukemic clones are not detectable by conventional microscopy but can be detected by more-sensitive techniques, including RT-qPCR, multiparameter flow cytometry, and even NGS [Citation123,Citation124]. Despite its importance as a prognostic marker, FLT3-ITD was long seen as an unsuitable marker for MRD monitoring because of patient-to-patient heterogeneity (e.g. length, insertion site, and allelic ratio) and inherent instability during the course of the disease [Citation125–129]. However, more-sensitive PCR- and NGS-based techniques have recently been developed [Citation43,Citation130–132] and are becoming commercially available [Citation133]. Nevertheless, the clinical application of these techniques needs to be validated in randomized clinical trials, as suggested by current recommendations [Citation16]. Several ongoing, phase 3 clinical trials evaluating FLT3 TKIs now include MRD as an endpoint [Citation78,Citation99,Citation103].

NPM1 has also emerged as a reliable marker for MRD monitoring because (1) NPM1 levels remain stable throughout disease progression and (2) NPM1 MRD levels have been clinically shown to correlate with therapeutic response. However, MRD monitoring has not yet been incorporated into AML disease management [Citation59], given that no standard methods or definitive markers for MRD monitoring have been established [Citation16]. The ELN is working on developing recommendations for MRD monitoring, which will likely include a combination of multiparameter flow cytometry and molecular-based assays.

Conclusion

Because of the recent results observed with FLT3-targeted therapies, the FLT3 testing paradigm may shift from FLT3 being regarded as a prognostic marker to being viewed as a diagnostic marker that can guide therapy choice. FLT3 testing guidelines are beginning to change, including requirements for faster turnaround times (48–72 h), testing for both ITD and TKD mutations, and testing regardless of karyotype [Citation16]. These changes will likely be adopted in the United States, requiring a shift in the order in which FLT3 testing is performed. Currently, in many centers, cytogenetic and FLT3 testing is done sequentially (i.e. FLT3 testing follows cytogenetic testing); in the future, FLT3 testing should be done in parallel with cytogenetic testing, as recommended in current diagnostic guidelines. This parallel approach will require education on the importance of FLT3 testing, particularly in community oncology centers, to ensure widespread and timely testing. As we gain more insight into the prognostic impact of complex gene-gene interactions and molecular-cytogenetic abnormalities – and as new targeted therapies potentially become available – the diagnostic and therapeutic landscape of AML is likely to see major changes. Additional challenges in FLT3 testing will include the need for harmonization of screening and MRD assays. Nevertheless, it is exciting to know that these changes and challenges are driven by gains in the development of therapeutic agents (evidenced by the large number of phase 3 trials evaluating FLT3 TKIs) for this high–unmet need patient population.

Potential conflict of interest

Disclosure forms provided by the author is available with the full text of this article online at https://doi.org/10.1080/10428194.2017.1399312.

Supplemental material

ICMJE Form for Disclosure of Potential Conflicts of Interest

Download PDF (1.2 MB)

Acknowledgments

Editorial assistance was provided by Katherine Mills-Lujan, PhD, CMPP, and Pamela Tuttle, PhD, CMPP, of ArticulateScience LLC, and was funded by Novartis Pharmaceuticals Corporation. This work was supported by the National Center for Advancing Translational Sciences under CTSA Grant Number KL2 TR000136. Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health.

References

  • Rosnet O, Mattei MG, Marchetto S, et al. Isolation and chromosomal localization of a novel FMS-like tyrosine kinase gene. Genomics. 1991;9:380–385.
  • Agnes F, Shamoon B, Dina C, et al. Genomic structure of the downstream part of the human FLT3 gene: exon/intron structure conservation among genes encoding receptor tyrosine kinases (RTK) of subclass III. Gene. 1994;145:283–288.
  • Rosnet O, Buhring HJ, Marchetto S, et al. Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells. Leukemia. 1996;10:238–248.
  • Carow CE, Levenstein M, Kaufmann SH, et al. Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood. 1996;87:1089–1096.
  • Kayser S, Schlenk RF, Londono MC, et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood. 2009;114:2386–2392.
  • Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209–2221.
  • Stirewalt DL, Meshinchi S, Kussick SJ, et al. Novel FLT3 point mutations within exon 14 found in patients with acute myeloid leukaemia. Br J Haematol. 2004;124:481–484.
  • Reindl C, Bagrintseva K, Vempati S, et al. Point mutations in the juxtamembrane domain of FLT3 define a new class of activating mutations in AML. Blood. 2006;107:3700–3707.
  • Fröhling S, Scholl C, Levine RL, et al. Identification of driver and passenger mutations of FLT3 by high-throughput DNA sequence analysis and functional assessment of candidate alleles. Cancer Cell. 2007;12:501–513.
  • Spiekermann K, Bagrintseva K, Schoch C, et al. A new and recurrent activating length mutation in exon 20 of the FLT3 gene in acute myeloid leukemia. Blood. 2002;100:3423–3425.
  • Chatain N, Perera RC, Rossetti G, et al. Rare FLT3 deletion mutants may provide additional treatment options to patients with AML: an approach to individualized medicine. Leukemia. 2015;29:2434–2438.
  • Opatz S, Polzer H, Herold T, et al. Exome sequencing identifies recurring FLT3 N676K mutations in core-binding factor leukemia. Blood. 2013;122:1761–1769.
  • Bacher U, Haferlach C, Kern W, et al. Prognostic relevance of FLT3–TKD mutations in AML: the combination matters-an analysis of 3082 patients. Blood. 2008;111:2527–2537.
  • Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366:1079–1089.
  • Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a Cancer and Leukemia Group B study. Cancer Res. 2001;61:7233–7239.
  • Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–447.
  • Thiede C, Steudel C, Mohr B, et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood. 2002;99:4326–4335.
  • Stirewalt DL, Radich JP. The role of FLT3 in haematopoietic malignancies. Nat Rev Cancer. 2003;3:650–665.
  • Pratcorona M, Brunet S, Nomdedeu J, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121:2734–2738.
  • Gale RE, Green C, Allen C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111:2776–2784.
  • Linch DC, Hills RK, Burnett AK, et al. Impact of FLT3(ITD) mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia. Blood. 2014;124:273–276.
  • How J, Sykes J, Gupta V, et al. Influence of FLT3-internal tandem duplication allele burden and white blood cell count on the outcome in patients with intermediate-risk karyotype acute myeloid leukemia. Cancer. 2012;118:6110–6117.
  • Schneider F, Hoster E, Unterhalt M, et al. The FLT3ITD mRNA level has a high prognostic impact in NPM1 mutated, but not in NPM1 unmutated, AML with a normal karyotype. Blood. 2012;119:4383–4386.
  • Allen C, Hills RK, Lamb K, et al. The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia. Leukemia. 2013;27:1891–1901.
  • Koszarska M, Meggyesi N, Bors A, et al. Medium-sized FLT3 internal tandem duplications confer worse prognosis than short and long duplications in a non-elderly acute myeloid leukemia cohort. Leuk Lymphoma. 2014;55:1510–1517.
  • Fröhling S, Schlenk RF, Breitruck J, et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm. Blood. 2002;100:4372–4380.
  • Smith CC, Wang Q, Chin CS, et al. Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 2012;485:260–263.
  • Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98:1752–1759.
  • Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood. 2002;100:59–66.
  • Oyarzo MP, Lin P, Glassman A, et al. Acute myeloid leukemia with t(6;9) (p23;q34) is associated with dysplasia and a high frequency of flt3 gene mutations. Am J Clin Pathol. 2004;122:348–358.
  • Slovak ML, Gundacker H, Bloomfield CD, et al. A retrospective study of 69 patients with t(6;9)(p23;q34) AML emphasizes the need for a prospective, multicenter initiative for rare ‘poor prognosis’ myeloid malignancies. Leukemia. 2006;20:1295–1297.
  • Kainz B, Heintel D, Marculescu R, et al. Variable prognostic value of FLT3 internal tandem duplications in patients with de novo AML and a normal karyotype, t(15;17), t(8;21) or inv(16). Hematol J. 2002;3:283–289.
  • Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group study. Blood. 2000;96:4075–4083.
  • Walter RB, Othus M, Burnett AK, et al. Resistance prediction in AML: analysis of 4601 patients from MRC/NCRI, HOVON/SAKK, SWOG and MD Anderson Cancer Center. Leukemia.2015;29:312–320.
  • Cheson BD, Bennett JM, Kopecky KJ, et al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21:4642–4649.
  • Arber DA, Borowitz MJ, Cessna M, et al. Initial diagnostic workup of acute leukemia: guideline from the College of American Pathologists and the American Society of Hematology. Arch Pathol Lab Med. 2017;141:1342–1393.
  • Rydapt (midostaurin) [prescribing information]. East Hanover (NJ): Novartis Pharmaceuticals Corporation; April 2017.
  • Jones C, LeDay TV, Miller AM. Acute myelogenous leukemia at Baylor Charles A. Sammons Cancer Center, 2010 to 2012: retrospective analysis of molecular genetic evaluation. Proc (Bayl Univ Med Cent). 2014;27:299–304.
  • Lin TL, Williams T, He J, et al. Rates of complete diagnostic testing for patients with acute myeloid leukemia. Cancer Med. 2015;4:519–522.
  • Döhner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–474.
  • George TI, Tworek JA, Thomas NE, et al. Evaluation of testing of acute leukemia samples: survey result from the College of American Pathologists. Arch Pathol Lab Med. 2017;141:1101–1106.
  • Invivoscribe. LeukoStrat® CDx FLT3 mutation assay [Internet]. San Diego (CA): Invivoscribe; 2017 [cited 2017 Sep 8]. Available from: https://invivoscribe.com/product/leukostrat-cdx-flt3-mutation-kit
  • Altman JK, Perl AE, Cortes JE, et al. Deep molecular response to gilteritinib improves survival in FLT3 mutation-positive relapsed/refractory acute myeloid leukemia. Paper presented at: European Hematology Association 22nd Congress: 2017 Jun 22–25; Madrid, Spain.
  • Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood. 1999;93:3074–3080.
  • McKerrell T, Moreno T, Ponstingl H, et al. Development and validation of a comprehensive genomic diagnostic tool for myeloid malignancies. Blood. 2016;128:e1–e9.
  • Duncavage EJ, Tandon B. The utility of next-generation sequencing in diagnosis and monitoring of acute myeloid leukemia and myelodysplastic syndromes. Int J Lab Hematol. 2015;37(Suppl 1):115–121.
  • Spencer DH, Abel HJ, Lockwood CM, et al. Detection of FLT3 internal tandem duplication in targeted, short-read-length, next-generation sequencing data. J Mol Diagn. 2013;15:81–93.
  • Au CH, Wa A, Ho DN, et al. Clinical evaluation of panel testing by next-generation sequencing (NGS) for gene mutations in myeloid neoplasms. Diagn Pathol. 2016;11:11.
  • Guan YF, Li GR, Wang RJ, et al. Application of next-generation sequencing in clinical oncology to advance personalized treatment of cancer. Chin J Cancer. 2012;31:463–470.
  • Murphy KM, Levis M, Hafez MJ, et al. Detection of FLT3 internal tandem duplication and D835 mutations by a multiplex polymerase chain reaction and capillary electrophoresis assay. J Mol Diagn. 2003;5:96–102.
  • Liu HE, Ko C, Lam F, et al. Establishment of a cost-effective method to detect FLT-ITD and D835 mutations in acute myeloid leukemia patients in the Taiwanese population. Tzu Chi Med J. 2015;27:18–24.
  • Gupta A, Viswanatha DS, Patnaik MM. FLT3 mutation testing in acute myeloid leukemia. JAMA Oncol. 2017;3:991–992.
  • Stirewalt DL, Willman CL, Radich JP. Quantitative, real-time polymerase chain reactions for FLT3 internal tandem duplications are highly sensitive and specific. Leuk Res. 2001;25:1085–1088.
  • Scholl S, Krause C, Loncarevic IF, et al. Specific detection of Flt3 point mutations by highly sensitive real-time polymerase chain reaction in acute myeloid leukemia. J Lab Clin Med. 2005;145:295–304.
  • Prasad V, Gale RP. Precision medicine in acute myeloid leukemia: hope, hype or both? Leuk Res. 2016;48:73–77.
  • Huang K, Yang M, Pan Z, et al. Leukemogenic potency of the novel FLT3-N676K mutant. Ann Hematol. 2016;95:783–791.
  • Schlenk RF, Döhner K, Salih H, et al. Midostaurin in combination with intensive induction and as single agent maintenance therapy after consolidation therapy with allogeneic hematopoietic stem cell transplantation or high-dose cytarabine (NCT01477606). Paper presented at: 57th American Society of Hematology Annual Meeting and Exposition; 2015 Dec 5–8; Orlando, FL.
  • Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377:454–464.
  • NCCN Clinical Practice Guidelines in Oncology. Acute Myeloid Leukemia. V3.2017 [Internet]. Fort Washington (PA): National Comprehensive Cancer Network; [cited 2017 Sep 8]. Available from: https://www.nccn.org/professionals/physician_gls/PDF/aml.pdf
  • Lin TL, Levy MY. Acute myeloid leukemia: focus on novel therapeutic strategies. Clin Med Insights Oncol. 2012;6:205–217.
  • Rydapt (midostaurin) [summary of product characteristics]. Basel, Switzerland: Novartis Pharmaceuticals AG; September 2017.
  • Badar T, Kantarjian HM, Nogueras-Gonzalez GM, et al. Improvement in clinical outcome of FLT3 ITD mutated acute myeloid leukemia patients over the last one and a half decade. Am J Hematol. 2015;90:1065–1070.
  • Schiller GJ, Tuttle P, Desai P. Allogeneic hematopoietic stem cell transplantation in FLT3-ITD-positive acute myelogenous leukemia: the role for FLT3 tyrosine kinase inhibitors post-transplantation. Biol Blood Marrow Transplant. 2016;22:982–990.
  • Ho AD, Schetelig J, Bochtler T, et al. Allogeneic stem cell transplantation improves survival in patients with AML characterized by a high allelic ratio of mutant FLT3-ITD. Biol Blood Marrow Transplant. 2016;22:462–469.
  • Oran B, Cortes J, Beitinjaneh A, et al. Allogeneic transplantation in first remission improves outcomes irrespective of FLT3-ITD allelic ratio in FLT3-ITD-positive acute myelogenous leukemia. Biol Blood Marrow Transplant. 2016;22:1218–1226.
  • Gale RE, Hills R, Kottaridis PD, et al. No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials. Blood. 2005;106:3658–3665.
  • Bornhauser M, Illmer T, Schaich M, et al. Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML. Blood. 2007;109:2264–2265.
  • Schlenk RF, Dohner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1909–1918.
  • Brunet S, Labopin M, Esteve J, et al. Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis. J Clin Oncol. 2012;30:735–741.
  • Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124:3441–3449.
  • Song Y, Magenau J, Li Y, et al. FLT3 mutational status is an independent risk factor for adverse outcomes after allogeneic transplantation in AML. Bone Marrow Transplant. 2016;51:511–520.
  • Chen Y, Li S, Lane AA, et al. Phase I trial of maintenance sorafenib after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20:2042–2048.
  • Antar A, Kharfan-Dabaja MA, Mahfouz R, et al. Sorafenib maintenance appears safe and improves clinical outcomes in FLT3-ITD acute myeloid leukemia after allogeneic hematopoietic cell transplantation. Clin. Lymphoma Myeloma Leuk. 2015;15:298–302.
  • Battipaglia G, Ruggeri A, Jestin M, et al. Efficacy and feasibility of sorafenib as a maintenance agent after allogeneic hematopoietic stem cell transplantation for fms-like tyrosine kinase 3 mutated acute myeloid leukemia. Poster presented at: 58th American Society of Hematology Annual Meeting and Exposition; 2016 Dec 3–6; San Diego, CA.
  • Brunner AM, Li S, Fathi AT, et al. Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission. Br J Haematol. 2016;175:496–504.
  • Maziarz RT, Patnaik MM, Scott BL, et al. Radius: a phase 2, randomized trial of standard of care with or without midostaurin to prevent relapse following allogeneic hematopoietic stem cell transplant in patients with FLT3-ITD–mutated acute myeloid leukemia. Poster presented at: 58th American Society of Hematology Annual Meeting and Exposition; 2016 Dec 3–6; San Diego, CA.
  • ClinicalTrials.gov. Crenolanib maintenance following allogeneic stem cell transplantation in FLT3-positive acute myeloid leukemia patients [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 8 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02400255
  • ClinicalTrials.gov. A trial of the FMS-like tyrosine kinase 3 (FLT3) inhibitor gilteritinib administered as maintenance therapy following allogeneic transplant for patients with FLT3/internal tandem duplications (ITD) acute myeloid leukemia (AML) [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 7 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02997202
  • ClinicalTrials.gov. Standard of care +/- midostaurin to prevent relapse post stem cell transplant in patients with FLT3-ITD mutated AML (RADIUS) [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Mar 21 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT01883362
  • ClinicalTrials.gov. Sorafenib for prophylaxis of leukemia relapse in allo-HSCT recipients with FLT3-ITD positive AML [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Jun 11 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02474290
  • Pratz KW, Levis M. How I treat FLT3-mutated AML. Blood. 2017;129:565–571.
  • Kansagra AJ, Alkhateeb HB, Hefazi M, et al. Feasibility of allogeneic hematopoietic stem cell transplant for high risk FLT3-ITD mutant patients with acute myeloid leukemia in CR1—a real world analysis. Poster presented at: 58th American Society of Hematology Annual Meeting and Exposition; 2016 Dec 3–6; San Diego, CA.
  • Antar A, Otrock ZK, El-Cheikh J, et al. Inhibition of FLT3 in AML: a focus on sorafenib. Bone Marrow Transplant. 2017;52:344–351.
  • Zhang W, Konopleva M, Shi YX, et al. Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst. 2008;100:184–198.
  • Mori M, Kaneko N, Ueno Y, et al. Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia. Invest New Drugs. 2017;35:556–565.
  • Zarrinkar PP, Gunawardane RN, Cramer MD, et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML). Blood. 2009;114:2984–2992.
  • Cortes JE, Perl AE, Dombret H, et al. Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients > = 60 years of age with FLT3 ITD positive or negative relapsed/refractory acute myeloid leukemia. Paper presented at: 54th American Society of Hematology Annual Meeting and Exposition; 2012 Dec 8–11; Atlanta, GA.
  • Levis MJ, Perl AE, Dombret H, et al. Final results of a phase 2 open-label, monotherapy efficacy and safety study of quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory acute myeloid leukemia after second-line chemotherapy or hematopoietic stem cell transplantation. Paper presented at: 54th American Society of Hematology Annual Meeting and Exposition; 2012 Dec 8–11; Atlanta, GA.
  • Schiller GJ, Tallman MS, Goldberg SL, et al. Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia. Poster presented at: 2014 American Society of Clinical Oncology Annual Meeting; 2014 May 30–Jun 3; Chicago, IL.
  • Altman JK, Foran JM, Pratz KW, et al. Results of a phase 1 study of quizartinib (AC220, ASP2689) in combination with induction and consolidation chemotherapy in younger patients with newly diagnosed acute myeloid leukemia. Paper presented at: 55th American Society of Hematology Annual Meeting and Exposition; 2013 Dec 7–10; New Orleans, LA.
  • Weisberg E, Boulton C, Kelly LM, et al. Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell. 2002;1:433–443.
  • von Bubnoff N, Engh RA, Åberg E, et al. FMS-like tyrosine kinase 3-internal tandem duplication tyrosine kinase inhibitors display a nonoverlapping profile of resistance mutations in vitro. Cancer Res. 2009;69:3032–3041.
  • Fischer T, Stone RM, DeAngelo DJ, et al. Phase IIB trial of oral midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin Oncol. 2010;28:4339–4345.
  • Perl AE, Altman JK, Cortes J, et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1–2 study. Lancet Oncol. 2017;18:1061–1075.
  • Collins R, Kantarjian HM, Levis MJ, et al. Clinical activity of crenolanib in patients with D835 mutant FLT3-positive relapsed/refractory acute myeloid leukemia (AML). Poster presented at: 2014 American Society of Clinical Oncology Annual Meeting; 2014 May 30–Jun 3; Chicago, IL.
  • Randhawa JK, Kantarjian HM, Borthakur G, et al. Results of a phase II study of crenolanib in relapsed/refractory acute myeloid leukemia patients (pts) with activating FLT3 mutations. Paper presented at: 56th American Society of Hematology Annual Meeting and Exposition; 2014 Dec 6–9; San Francisco, CA.
  • Wang ES, Stone RM, Tallman MS, et al. Crenolanib, a type I FLT3 TKI, can be safely combined with cytarabine and anthracycline induction chemotherapy and results in high response rates in patients with newly diagnosed FLT3 mutant acute myeloid leukemia (AML). Paper presented at: 58th American Society of Hematology Annual Meeting and Exposition; 2016 Dec 3–6; San Diego, CA.
  • Cortes JE, Kantarjian HM, Kadia TM, et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, demonstrates clinical activity in multiple relapsed FLT3-ITD and D835 AML. Poster presented at: 2016 American Society of Clinical Oncology Annual Meeting; 2016 Jun 3–7; Chicago, IL.
  • ClinicalTrials.gov. A study of ASP2215 (gilteritinib), administered as maintenance therapy following induction/consolidation therapy for subjects with FMS-like tyrosine kinase 3 (FLT3/ITD) acute myeloid leukemia (AML) in first complete remission [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 18 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02927262
  • ClinicalTrials.gov. Study of crenolanib in combination with chemotherapy in patients with relapsed or refractory acute myeloid leukemia and activating FLT3 mutations [Internet]. Bethesda (MD): US National Institutes of Health; 2017 May 15 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02298166
  • ClinicalTrials.gov. A study of ASP2215 versus salvage chemotherapy in patients with relapsed or refractory acute myeloid leukemia (AML) with FMS-like tyrosine kinase (FLT3) mutation [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 23 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02421939
  • ClinicalTrials.gov. (QuANTUM-R): an open-label study of quizartinib monotherapy vs salvage chemotherapy in acute myeloid leukemia (AML) subjects who are FLT3-ITD positive [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Jun 23 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02039726
  • ClinicalTrials.gov. Study investigating the efficacy of crenolanib with chemotherapy vs chemotherapy alone in R/R FLT3 mutated AML [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 14 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT03250338
  • ClinicalTrials.gov. A study of ASP2215 versus salvage chemotherapy in patients with relapsed or refractory acute myeloid leukemia (AML) with FLT3 mutation [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Jun 22 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT03182244
  • ClinicalTrials.gov. A study of ASP2215 (gilteritinib), combination of ASP2215 plus azacitidine and azacitidine alone in the treatment of newly diagnosed acute myeloid leukemia with FMS-like tyrosine kinase (FLT3) mutation in patients not eligible for intensive induction chemotherapy [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 10 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02752035
  • ClinicalTrials.gov. Quizartinib with standard of care chemotherapy and as maintenance therapy in patients with newly diagnosed FLT3-ITD (+) acute myeloid leukemia (AML) (QuANTUM-First) [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 30 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02668653
  • ClinicalTrials.gov. Bortezomib and sorafenib tosylate in treating patients with newly diagnosed acute myeloid leukemia [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Sep 4 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT01371981
  • ClinicalTrials.gov. Study of crenolanib vs midostaurin following induction chemotherapy and consolidation therapy in newly diagnosed FLT3 mutated AML [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Aug 21 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT03258931
  • Albers C, Leischner H, Verbeek M, et al. The secondary FLT3-ITD F691L mutation induces resistance to AC220 in FLT3-ITD(+) AML but retains in vitro sensitivity to PKC412 and sunitinib. Leukemia. 2013;27:1416–1418.
  • Gallogly MM, Lazarus HM. Midostaurin: an emerging treatment for acute myeloid leukemia patients. J Blood Med. 2016;7:73–83.
  • ClinicalTrials.gov. Midostaurin and azacitidine in treating elderly patients with acute myelogenous leukemia [Internet]. Bethesda (MD): US National Institutes of Health; 2016 Jun 8 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT01093573
  • Röllig C, Serve H, Huttmann A, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16:1691–1699.
  • Serve H, Krug U, Wagner R, et al. Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial. J Clin Oncol. 2013;31:3110–3118.
  • Ravandi F, Alattar ML, Grunwald MR, et al. Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121:4655–4662.
  • Muppidi MR, Portwood S, Griffiths EA, et al. Decitabine and sorafenib therapy in FLT-3 ITD-mutant acute myeloid leukemia. Clin Lymphoma Myeloma Leuk. 2015;15(Suppl):S73–S79.
  • ClinicalTrials.gov. Sorafenib plus 5-azacitidine initial therapy of patients with acute myeloid leukemia (AML) and high risk myelodysplastic syndrome (MS) with FLT3-ITD mutation [Internet]. Bethesda (MD): US National Institutes of Health; 2017 Mar 9 [cited 2017 Sep 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02196857
  • Smith CC, Lasater EA, Zhu X, et al. Activity of ponatinib against clinically-relevant AC220-resistant kinase domain mutants of FLT3-ITD. Blood. 2013;121:3165–3171.
  • Kihara R, Nagata Y, Kiyoi H, et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia. 2014;28:1586–1595.
  • Grossmann V, Schnittger S, Kohlmann A, et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood. 2012;120:2963–2972.
  • Fey MF, Buske C, ESMO Guidelines Working Group. Acute myeloblastic leukaemias in adult patients: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013;24(Suppl 6):vi138–vi143.
  • Myers GL, Miller WG. The International Consortium for Harmonization of Clinical Laboratory Results (ICHCLR) – a pathway for harmonization. EJIFCC. 2016;27:30–36.
  • Plebani M. Harmonization of clinical laboratory information–current and future strategies. EJIFCC. 2016;27:15–22.
  • Kayser S, Walter RB, Stock W, et al. Minimal residual disease in acute myeloid leukemia—current status and future perspectives. Curr Hematol Malig Rep. 2015;10:132–144.
  • Ossenkoppele G, Schuurhuis GJ. MRD in AML: does it already guide therapy decision-making? Hematology Am Soc Hematol Educ Program. 2016;2016:356–365.
  • Shih LY, Huang CF, Wu JH, et al. Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse. Blood. 2002;100:2387–2392.
  • Kottaridis PD, Gale RE, Langabeer SE, et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood. 2002;100:2393–2398.
  • Schnittger S, Schoch C, Kern W, et al. FLT3 length mutations as marker for follow-up studies in acute myeloid leukaemia. Acta Haematol. 2004;112:68–78.
  • Cloos J, Goemans BF, Hess CJ, et al. Stability and prognostic influence of FLT3 mutations in paired initial and relapsed AML samples. Leukemia. 2006;20:1217–1220.
  • Palmisano M, Grafone T, Ottaviani E, et al. NPM1 mutations are more stable than FLT3 mutations during the course of disease in patients with acute myeloid leukemia. Haematologica. 2007;92:1268–1269.
  • Grunwald MR, Tseng LH, Lin MT, et al. Improved FLT3 internal tandem duplication PCR assay predicts outcome after allogeneic transplant for acute myeloid leukemia. Biol Blood Marrow Transplant. 2014;20:1989–1995.
  • Zuffa E, Franchini E, Papayannidis C, et al. Revealing very small FLT3 ITD mutated clones by ultra-deep sequencing analysis has important clinical implications in AML patients. Oncotarget. 2015;6:31284–31294.
  • Lin MT, Tseng LH, Dudley JC, et al. A novel tandem duplication assay to detect minimal residual disease in FLT3/ITD AML. Mol Diagn Ther. 2015;19:409–417.
  • Invivoscribe. FLT3 ITD MRD testing by NGS [Internet]. San Diego (CA): Invivoscribe; 2017 [cited 2017 Sep 11]. Available from: https://invivoscribe.com/clinical-services/flt3-itd-mrd-testing-by-ngs