Abstract
Mutations in the fms-like tyrosine kinase 3 (FLT3) gene are detected in approximately one-third of patients with newly diagnosed acute myeloid leukemia (AML). These consist of the more common FLT3-internal tandem duplication (ITD) in approximately 20–25% of AML cases, and point mutations in the tyrosine kinase domain (TKD) in approximately 5–10%. FLT3 mutations, especially FLT3-ITD, are associated with proliferative disease, increased risk of relapse, and inferior overall survival when treated with conventional regimens. However, the recent development of well tolerated and active FLT3 inhibitors has significantly improved the outcomes of this aggressive subtype of AML. The multikinase inhibitor midostaurin was approved by the United States Food and Drug Administration (US FDA) in April 2017 for the frontline treatment of patients with FLT3-mutated (either ITD or TKD) AML in combination with induction chemotherapy, representing the first new drug approval in AML in nearly two decades. In November 2018, the US FDA also approved the second-generation FLT3 inhibitor gilteritinib as a single agent for patients with relapsed or refractory FLT3-mutated AML. Promising phase I and II efficacy data for quizartinib is likely to lead to a third regulatory approval in relapsed/refractory AML in the near future. However, despite the significant progress made in managing FLT3-mutated AML, many questions remain regarding the best approach to integrate these inhibitors into combination regimens, and also the optimal sequencing of different FLT3 inhibitors in various clinical settings. This review comprehensively examines the FLT3 inhibitors currently in clinical development, with an emphasis on their spectra of activity against different FLT3 mutations and other kinases, clinical safety and efficacy data, and their current and future roles in the management of AML. The mechanisms of resistance to FLT3 inhibitors and potential combination strategies to overcome such resistance pathways are also discussed.
Keywords: acute myeloid leukemia, FLT3, gilteritinib, midostaurin, quizartinib, resistance, sorafenib, tyrosine kinase inhibitors
Introduction
Acute myeloid leukemia (AML) is a genomically heterogeneous disease characterized by a number of well-described recurrent mutations that drive disease phenotype, response to therapy, and risk for relapse.1 Mutations of the fms-like tyrosine kinase 3 (FLT3) gene are the most common genomic alterations in AML, identified in approximately one-third of newly diagnosed patients.2–4 FLT3-internal tandem duplication (ITD)-mutated AML often presents with proliferative features such as leukocytosis and increased peripheral blasts. When treated with combination chemotherapy alone, FLT3-ITD mutated AML is associated with a higher rate of relapse and inferior overall survival than FLT3 wildtype disease.5–11 In contrast the prognostic impact of FLT3 tyrosine kinase domain (TKD) mutations is less clear. This has led to intense interest in the development of targeted agents against the FLT3 mutant protein, eventually culminating in both United States Food and Drug Administration (US FDA) and European Medicines Agency (EMA) approvals of the multikinase tyrosine kinase inhibitor (TKI) midostaurin in combination with standard cytarabine and daunorubicin induction and cytarabine consolidation in adults with newly diagnosed FLT3-mutated AML. This was the first AML drug to receive regulatory approval in the US since 2000. Following the midostaurin approval, in November 2018, the second-generation FLT3 inhibitor gilteritinib was approved by the US FDA for use as a single agent for adults with relapsed or refractory FLT3-mutated AML. Several other FLT3 inhibitors are currently in advanced clinical development with anticipated approval in different AML settings and are likely to change the therapeutic approach to this subtype of AML in the very near future.
In this review, we will discuss the landscape of FLT3 mutations in AML and how these mutations impact prognosis. We will also review the available clinical data on the several FLT3 inhibitors currently in development and describe the potential role that each of these FLT3 TKIs may place in the future management of FLT3-mutated AML, with particular emphasis on their distinct mechanism and spectra of action. Finally, we will discuss the ongoing studies of these various inhibitors and how rationally designed combination therapies may overcome the known mechanisms of resistance to FLT3 inhibitors in AML.
Wildtype and mutated FLT3 in AML
The FLT3 protein is a receptor tyrosine kinase widely expressed in hematopoietic progenitor cells. Upon binding to the cytokine FLT3 ligand, FLT3 receptors dimerize; this dimerization leads to conformational changes in the FLT3 proteins that expose their adenosine triphosphate (ATP) pockets and trigger autophosphorylation and signal transduction. Downstream effects of this intracellular signaling include the promotion of cellular proliferation and survival and inhibition of differentiation.12,13 Independent of the presence or absence of FLT3 mutations, FLT3 is commonly overexpressed in lineage-restricted AML blasts and may be associated with worse outcomes, suggesting that it may serve as a therapeutic target irrespective of FLT3 mutation status.14,15
FLT3 mutations (ITD, TKD, or both) are identified in approximately one-third of patients with newly diagnosed AML.2–4 These mutations are particularly enriched in patients with a normal karyotype.8,16 However, as they are a later event in leukemogenesis and generally not a primary, initiating event, FLT3 mutations are not restricted to any particular AML subgroups.17 ITD mutations are identified in 20–25% of patients with newly diagnosed AML, whereas point mutations in the TKD are identified in 5–10%, approximately half of which occur at D835 in the activation loop.18,19 Regardless of the type of mutation, both cause spontaneous dimerization and ligand-dependent growth. Importantly, the mutated receptor remains responsive to the FLT3 ligand, which is capable of further modulating signaling from the mutant kinase.20
Prognostic impact of FLT3 mutations
Prior to the advent of FLT3 inhibitors, studies have consistently shown that FLT3-ITD-mutated AML is associated with an increased risk of relapse and worse survival compared with wildtype disease.5–11 In contrast, the clinical relevance of TKD mutations is less clear,21–24 although it is generally accepted that their prognostic impact is neutral and that the presence of a TKD mutation should not alter risk assessment.25 Among patients with FLT3-ITD mutations, co-mutations further influence outcomes, particularly the presence of a co-existing NPM1 mutation, which is associated with a decreased risk of relapse and improved survival in FLT3-ITD-mutated AML, especially in patients who have a low allele burden FLT3-ITD.2,4,26–29 The allelic ratio of FLT3-ITD to wildtype FLT3 has been shown to strongly influences outcomes in several studies of chemotherapy-based induction therapy (3+7 or similar) in patients with newly diagnosed FLT3-ITD-mutated AML.29–31 The FLT3-ITD allelic ratio is generally defined as the ratio of the area under the curve of ‘FLT3-ITD’ divided by the area under the curve of ‘FLT3-wildtype’ using a semi-quantitative DNA fragment analysis.25 A higher FLT3-ITD ratio (generally defined as ⩾0.5 or ⩾0.7, depending on the study) is generally associated with worse survival than lower ratios, likely reflecting increased FLT3 dependency in the cases with high allelic ratios. It is important to note that most of these studies on the impact of FLT3-ITD allelic ratio did not incorporate an FLT3 inhibitor as part of the induction, consolidation, or maintenance therapy; however, one study that included midostaurin in the treatment regimen failed to show a difference in the cumulative incidence of relapse according to FLT3-ITD allelic ratio of <0.5 versus ⩾0.5.32 Given consistent evidence of the cooperative role of NPM1 mutations and the FLT3-ITD allelic ratio in influencing outcomes in AML across most studies, both have been incorporated into consensus recommendations on genomic-based risk stratification.25 It has been suggested that a longer ITD length may be associated with a higher risk of relapse, although studies have yielding conflicting results16,33,34 and therefore ITD length assessment is not recommended for routine risk stratification.
The primary importance of accurate risk stratification in AML is to determine appropriate post-remission strategies, particularly to decide between consolidative chemotherapy or allogeneic hematopoietic stem cell transplantation (HSCT) in first remission.1 With the exception of one study that was limited by small numbers of FLT3-mutated patients who underwent HSCT and an unexpectedly high transplant-related mortality,35 the majority of evidence suggests that HSCT improves outcomes for patients with FLT3-ITD-mutated AML, when patients were analyzed agnostic of NPM1 status and allelic ratio.36–40 Furthermore, the RATIFY study showed that the addition of midostaurin to chemotherapy proportionally improved overall survival in FLT3 allele low (<0.5) and FLT3 allele high (⩾0.5) patients, albeit the improvement in these subgroups was not statistically significant (p = 0.19 for both analyses).41 Therefore, at most centers, fit patients with FLT3-ITD mutations are generally considered for HSCT in first remission, with the possible exception of NPM1-mutated patients with a low allelic burden FLT3-ITD mutation, as this constitutes a relatively favorable risk group in the pre-TKI era.25 In the absence of robust prognostic analysis in patients treated with FLT3 inhibitors in induction/consolidation, the current standard at our institution and at many academic centers in the US has been to add a multikinase FLT3 inhibitor (midostaurin or sorafenib) to induction and consolidation in patients with FLT3-mutated AML (defined as FLT3 mutation on next-generation sequencing or FLT3 allele burden >3% on polymerase chain reaction) and to proceed to HSCT in first remission for patients with FLT3-ITD-mutated AML that is classified as intermediate or adverse risk by European LeukemiaNet guidelines.25 However, studies reassessing the role of HSCT in patients with FLT3-mutated AML who receive frontline FLT3 inhibitors are needed. Randomized clinical trial data supports the addition of midostaurin,41 which is US FDA approved for this indication, while we generally add off-label sorafenib (unless a TKD mutation is present) based on data from phase II studies showing its safety and efficacy in combination with chemotherapy.42–44 We also have routinely used post-HSCT maintenance with a FLT3 inhibitor, either sorafenib (most commonly used as maintenance in our center) or crenolanib (in the setting of a previous clinical trial with post-HSCT crenolanib). Recent randomized data from ASH 2018 suggest that post-HSCT sorafenib maintenance significantly improved overall survival (OS) in FLT3-ITD-mutated patients who underwent HSCT.45
Characteristics of FLT3 inhibitors and principles of use
Given the established pathobiological and prognostic role that FLT3-ITD mutations play in AML, mutant FLT3 is an attractive therapeutic target for leukemia-directed therapies.46 To this end, several FLT3 inhibitors have been developed and are currently in clinical trials (Table 1). These agents work largely through competitive inhibition of ATP-binding sites in the FLT3 receptor, leading to cell cycle arrest and differentiation.47 In addition, FLT3 TKIs vary in their ability to target non-FLT3 signaling pathways, which influences both the tolerability and the efficacy of different agents. For example, first-generation FLT3 inhibitors (e.g. lestaurtinib, sorafenib, and midostaurin) are less specific for FLT3 and have broad kinome profiles with more off-target toxicities; in contrast, second-generation FLT3 inhibitors (e.g. quizartinib, crenolanib, and gilteritinib) are more specific and potent at inhibiting FLT3 with narrower kinome profiles.48,49
Table 1.
FLT3 inhibitor | Non-FLT3 targets | FLT3-TKD mutation activity | Single-agent CRc rates in R/R FLT3-mutated AML | Dose | Major toxicities | Approval status |
---|---|---|---|---|---|---|
Sorafenib | c-KIT, PDGFR, RAF, VEGFR | No | <10% | 400 mg bid | Rash, hemorrhage, myelosuppression | Available off-label (US FDA approved for hepatocellular, renal cell, and differentiated thyroid cancer) |
Midostaurin | c-KIT, PKC, PDGFR, VEGFR | Yes | <10% | 50 mg bid | GI toxicity, myelosuppression | US FDA and EMA approved for adults with newly diagnosed FLT3-mutated AML in combination with intensive chemotherapy (improves overall survival versus chemotherapy alone) |
Quizartinib | c-KIT, PDGFR, RET | No | 24–47% | 30–60 mg daily | QTc prolongation, myelosuppression | US FDA approval sought for use in relapsed/refractory setting (improves overall survival versus chemotherapy) |
Crenolanib | PDGFR | Yes | 17–39% | 100 mg tid | GI toxicity | Drug development plan is focused on chemotherapy-based combination |
Gilteritinib | AXL | Yes | 37–41% | 120 mg daily | Elevated transaminases, diarrhea | US FDA approved for adults with relapsed/refractory FLT3-mutated AML (full data not yet released) |
AML, acute myeloid leukemia; bid, twice daily; CRc, composite complete remission; EMA, European Medicines Agency; GI, gastrointestinal; PDGFR, platelet-derived growth factor receptor; R/R, relapsed/refractory; tid, three times daily; TKD, tyrosine kinase domain; US FDA, United States Food and Drug Administration; VEGFR, vascular endothelial growth factor receptor.
In addition to their off-target effects, FLT3 TKIs have varying potencies against different FLT3 mutations.50 Type I inhibitors (e.g. lestaurtinib, midostaurin, gilteritinib, and crenolanib) bind to the gatekeeper domain of FLT3 near the activation loop or the ATP-binding pocket regardless of receptor conformation, while type II inhibitors (e.g. sorafenib and quizartinib) bind to the hydrophobic region directly adjacent to the ATP-binding domain when the protein is in its inactive conformation. Notably, type II FLT3 inhibitors do not have significant activity against TKD mutations, as these mutations favor the active protein conformation.51 As FLT3-TKD mutations may be present at the time of diagnosis or emerge as a mechanism of resistance under the therapeutic pressure of chemotherapy or FLT3 inhibitor therapy, the inhibitory activity of these agents against TKD mutations should be a consideration when selecting the optimal FLT3 inhibitor for a particular patient, especially in the setting of an established TKD mutation.
While several FLT3 inhibitors have single-agent activity, there is preclinical rationale supporting the combination of these agents with cytotoxic chemotherapy, particularly given concomitantly or immediately after chemotherapy.52 Several studies have evaluated such combinations, most notably the randomized phase III RATIFY study of standard chemotherapy with or without midostaurin for patients with newly diagnosed FLT3-mutated AML, which showed an OS benefit with the addition of midostaurin to standard therapy.41
The efficacy of FLT3 inhibition in AML may also be influenced by the properties of the specific TKI and whether the leukemia is previously untreated or relapsed/refractory. Previously untreated FLT3-mutated AML is often a polyclonal disease, of which the FLT3 mutation may be present only in one subclone, whereas relapsed/refractory AML is a more monoclonal disease.53 In the former setting in which aberrant FLT3 signaling may be only one of several factors driving the disease biology and phenotype, it has been hypothesized that agents capable of inhibiting multiple kinases in addition to FLT3 (e.g. sorafenib and midostaurin) may be particularly beneficial. In contrast, in relapsed/refractory FLT3-mutated AML, there is often a higher FLT3 allelic burden and increased addiction to aberrant FLT3 signaling.54,55 In this setting, the use of more selective and potent second-generation FLT3 inhibitors may be ideal. However, the clinical data at this time do not clearly indicate whether a broad or selective kinase FLT3 inhibitor would be better during induction and relapse, respectively. A number of clinical trials combining selective FLT3 inhibitors (e.g. quizartinib, gilteritinib, and crenolanib) with induction therapy in patients with newly diagnosed FLT3-mutated AML are ongoing and will likely help answer this question (ClinicalTrials.gov identifiers: NCT02668653, NCT02752035, NCT02283177, NCT03258931)
Clinical experience with FLT3 inhibitors
Lestaurtinib
Lestaurtinib is a first-generation FLT3 inhibitor and one of the first to be studied in clinical trials. It has activity against FLT3, as well as JAK2 and Trk tyrosine kinases. In early studies of single-agent lestaurtinib, including a phase I/II study in relapsed/refractory AML (n = 17) and a phase II study of older adults with newly diagnosed AML (n = 29, including 5 patients with an activating FLT3 mutation), transient reductions of bone marrow blasts were observed. No patients achieved complete remission (CR) or complete remission with incomplete count recovery (CRi).56,57 These studies were followed by two randomized phase III trials evaluating chemotherapy with or without the addition of lestaurtinib in patients with FLT3-mutated AML, including one study in patients in first relapse58 and one in younger patients with newly diagnosed AML.59 Neither of these trials showed an improvement in either response rates or survival with the addition of lestaurtinib to chemotherapy in the intention to treat population, which may have in part been due to suboptimal FLT3 inhibition. Among patients in the frontline lestaurtinib plus chemotherapy trial in whom >85% FLT3 inhibition was sustained with lestaurtinib therapy, a benefit in relapse rate and OS was observed, supporting the potential for FLT3 inhibitor-based regimens in this context.59 However, given these negative phase III study results, lestaurtinib is no longer in clinical development.
Sorafenib
Sorafenib is another first-generation FLT3 inhibitor that has a wide variety of kinase inhibitory activity, including c-KIT, platelet-derived growth factor receptor (PDGFR), and vascular endothelial growth factor receptor (VEGFR). As a type II inhibitor, it notably has no clinically significant TKD activity, which limits its utility in patients harboring FLT3-D835 and other FLT3 point mutations. Like lestaurtinib, sorafenib had limited single-agent activity in AML, with marrow remissions observed in <10% of patients treated with sorafenib monotherapy.60–62
The clinical results with sorafenib have been more promising when used in combination, both with cytotoxic chemotherapy and with the hypomethylating agent azacitidine. In a phase II study of 62 adults with newly diagnosed AML (median age 53 years; range, 18–66 years), the combination of sorafenib with idarubicin and cytarabine (IA regimen) resulted in an overall CR or CR with incomplete platelet recovery (CRp) rate of 87%.42,43 A total of 23 patients (37%) harbored a FLT3-ITD mutation, and the CR/CRp rates for those with and without mutation were 95% and 84%, respectively. There was no difference in survival outcomes between patients with and without FLT3-ITD mutation, suggesting that the addition of sorafenib benefitted these patients by improving the survival in traditionally adverse risk FLT3-ITD patients to that of non-FLT3-ITD-mutated patients. The phase II CALGB 11001 study evaluated standard cytarabine and anthracycline-based induction and consolidation chemotherapy in combination with sorafenib in 54 patients age ⩾60 with newly diagnosed FLT3-mutated AML. Among patients with FLT3-ITD mutations (71% of the cohort), the median OS was 15.0 months and the 1-year OS rate was 62%, which was significantly better than the historical control group (1-year OS rate 30%, p < 0.0001). Together, these studies suggest that the combination of sorafenib with intensive chemotherapy is safe and may improve outcomes in both younger and older adults with FLT3-ITD-mutated AML.
The combination of sorafenib with azacitidine has also been shown to be well tolerated and effective. In a study of this combination in 43 adults with predominantly relapsed/refractory FLT3-ITD-mutated AML, the overall response rate was 46%.63 Notably, among the 40 patients with FLT3-ITD mutations, 9 had prior treatment with at least one FLT3 TKI. Despite the encouraging response rate, the median duration of response was only 2.3 months and the median OS was 6.2 months. Additional translational work showed that FLT3 ligand levels did not rise appreciably with exposure to the combination regimen. This finding is important as increased FLT3 ligand has been observed in patients treated with chemotherapy and has been suggested to be a mechanism of FLT3 inhibitor resistance in these patients.20 Despite the short duration of response and survival, these correlative studies suggest that hypomethylating agent and FLT3 inhibitor-based combinations may potentially overcome this mechanism of resistance, although the clinical significance remains to be determined. In a similar study in the frontline setting in 27 older adults with FLT3-ITD-mutated AML who were not candidates for intensive induction therapy (median age 74 years; range, 61–86 years), azacitidine plus sorafenib resulted in an overall response rate of 78% with a median duration of response of 14.5 months and median OS of 8.3 months.64
Given its multikinase activity, sorafenib has also been evaluated in combination with cytotoxic chemotherapy in patients with AML irrespective of FLT3 status. The SORAML study randomized 267 patients ⩽60 years of age with newly diagnosed AML to standard intensive induction and consolidation chemotherapy or to the same chemotherapy regimen in combination with sorafenib followed by 1 year of sorafenib maintenance.44 Only 17% of the randomized patients had a FLT3-ITD mutation at diagnosis. Patients randomized to the sorafenib-containing regimen had an improvement in both event-free survival (EFS) and relapse-free survival (RFS) compared with the placebo arm (3-year EFS rate: 22% versus 40%, p = 0.01; 3-year RFS rate: 56% versus 38%, p = 0.017), although no OS benefit was observed. The addition of sorafenib was associated with increased adverse events, particularly diarrhea, bleeding, cardiac events, hand-foot-skin reaction and rash but the 30-day mortality was comparable between the two arms (2% in the sorafenib arm and 1% in the placebo arm). In an update with longer follow up (median: 78 months), EFS and RFS benefits with the addition of sorafenib continued to be observed.65 Additionally, there appeared to be a late separation in the OS curves after 2 years, with a 5-year OS rate of 61% and 52% (p = 0.28) for the sorafenib and placebo arms, respectively. Overall, the results from the SORAML study suggest that there may be a benefit to adding a multikinase inhibitor such as sorafenib to induction even among patients without FLT3-ITD mutations. In contrast, a similar randomized study in an older population of patients with newly diagnosed AML (median age 68 years; range, 61–80 years) showed no EFS or OS benefit in this elderly population, likely driven by increased early deaths in the sorafenib arm (17% versus 7% with the placebo arm).66 Thus, the benefit of adding sorafenib to chemotherapy regardless of FLT3 status in frontline treatment regimens may be most prominent in younger, fitter patients who likely have a higher tolerance and resilience to addition of therapies to induction.
Midostaurin
Midostaurin is a first-generation FLT3 inhibitor capable of targeting ITD and TKD mutations with broad-spectrum inhibitory activity against VEGFR, protein kinase C, c-KIT, and PDGFR-β.67 In a study of 95 patients, midostaurin single-agent therapy in relapsed/refractory AML resulted in blast reductions in 71% of patients with FLT3-mutant AML and 42% of patients with FLT3-wildtype, although no CR or CRis were observed.68 In a smaller study of 20 patients with relapsed/refractory FLT3-mutated AML, 2 patients (10%) achieved bone marrow blast reduction <5%. In a phase Ib study in 69 younger adults with AML, the addition of midostaurin to standard cytarabine and anthracycline induction and consolidation was found to be safe and effective, with a CR rate of 80% (74% for FLT3 wildtype versus 92% for FLT3-mutated).44 OS was similar regardless of FLT3 status, suggesting the addition of midostaurin overcame the poor prognostic impact of the FLT3 mutation.
The encouraging findings of this dose-finding study led to a large, multinational, randomized phase III RATIFY (CALBG 10603) study.69 In this study, 717 adults <60 years of age with newly diagnosed FLT3-mutated AML (either ITD or TKD) were randomized to receive standard induction followed by consolidation chemotherapy (or HSCT, if indicated) with either midostaurin or placebo. Midostaurin or placebo were given on Days 8–21 of each induction and consolidation cycle, followed by up to an additional one year of maintenance with midostaurin or placebo. A total of 77% of patients had an ITD mutation and 23% had a TKD mutation. Patients in the midostaurin arm had a significant improvement in OS (4-year OS rate: 51.4% versus 44.3%; median OS: 74.7 months versus 25.6 months; p = 0.009), an effect that was seen regardless of type of FLT3 mutation or the ITD allelic burden (i.e. <0.5 or ⩾0.5).
Based on these results, midostaurin was approved by the US FDA in April 2017 and by the EMA in September 2017 for the treatment of adults with newly diagnosed FLT3-mutated AML in combination with standard cytarabine and daunorubicin induction and cytarabine consolidation. Despite use of up to 1 year of maintenance midostaurin in the RATIFY trial, no US FDA approval was given for midostaurin maintenance therapy, whereas use of midostaurin as maintenance was included in the EMA approval. Notably, along with the approval of midostaurin, the US FDA also approved the LeukoStrat CDx FLT3 Mutation Assay (Invivoscribe, Inc. San Diego, CA) as a companion diagnostic test for the detection of FLT3 mutations.
In the RATIFY trial, the largest survival benefit was observed in patients who underwent subsequent HSCT in first remission, which was performed in approximately 25% of the entire cohort (28.1% in the midostaurin group and 22.7% in the placebo group, p = 0.10). The 4-year OS with midostaurin followed by HSCT in first remission was 63.7%, compared with 55.7% for patients who received placebo followed by HSCT in first remission (p = 0.08), which translated to a 24.3% lower risk of death in the midostaurin group in the context of HSCT in first remission. Although no formal measurable residual disease (MRD) data have been reported as part of the RATIFY study, the finding that midostaurin improves outcomes for patients who received HSCT in first remission suggests that it may result in deeper remissions (i.e. more patients achieving MRD negativity) than placebo, as MRD negativity prior to HSCT has been shown to be a primary predictor of better post-HSCT outcomes in AML.70
Based on the results from this large, multicenter, randomized trial, rapid assessment and identification of a FLT3-ITD or TKD mutation, addition of midostaurin to induction and consolidation, and HSCT in first remission should be the goals of therapy for most patients. In the absence of strong data to the contrary from future analyses, it is reasonable to defer HSCT in first remission for those patients with low allelic burden FLT3-ITD mutation and concomitant NPM1 mutation, which is a favorable risk group in the pre-FLT3 inhibitor era. However, confirmation of these biomarkers in patients who receive FLT3 (Invivoscribe, Inc, San Diego, CA) inhibitor-based therapies is needed.29
Quizartinib
Quizartinib is a second-generation TKI that is more selective for FLT3 than the first-generation inhibitors, although it still has activity against other receptor tyrosine kinases such as c-KIT and PDGFR. Its more potent FLT3 inhibition was evident in studies of patients with relapsed/refractory AML treated with single-agent quizartinib, in which composite CR (CRc) rates >40%, including CRs, have been observed.71–73 This activity is in clear distinction to the first-generation FLT3 inhibitors, in which single-agent use in the relapsed/refractory setting resulted in transient blast decreases, generally without CR/CRis. In contrast with midostaurin, crenolanib and gilteritinib, quizartinib does not have significant activity against TKD mutations, which is an established mechanism of resistance in quizartinib-treated patients. In one small study, the development of resistance TKD mutations was observed in eight of eight patients with acquired resistance to quizartinib, although these findings remain to be confirmed in larger trials.46
In a phase I study of quizartinib in 76 patients with relapsed/refractory AML irrespective of FLT3 status, the overall response rate was 30%, including a CRc rate of 13%.71 Notably, responses were more frequently observed in FLT3-ITD mutated patients compared with FLT3 wildtype (overall response rate: 53% versus 14%; CRc rate: 24% versus 5%, respectively). The median duration of response was 13.3 weeks. The primary safety concern with quizartinib was QTc prolongation, which was observed in 12% of patients (any grade). Similar response rates were observed in patients with relapsed/refractory AML treated in a subsequent phase II study of quizartinib unselected for FLT3 status.72 Notably, in this study of 333 patients, a quizartinib dose of 200 mg daily was initially implemented, based on the recommended phase II dose determined in the earlier phase I study. However, due to a higher than acceptable rate of grade 3 QTc prolongation at this dose, the study was later amended to reduce the dose of quizartinib to 135 mg daily for men and 90 mg daily for women.
Given the uncertainty about the optimal dosing of quizartinib, a randomized phase IIb study was undertaken to evaluate two lower doses of quizartinib that had shown efficacy in the phase I trial, in patients with relapsed/refractory FLT3-ITD-mutated AML.73 A total of 76 patients were randomized to receive quizartinib at a dose of either 30 mg or 60 mg daily, which could be escalated to 60 mg or 90 mg, respectively, due to lack of response or loss of response. The CRc rate in both groups was 47%, and grade 3 QTc prolongation rates were only 3–5% suggesting equivalent CRc rates with significantly lower QTc prolongation than was seen with the higher doses of quizartinib (grade 3 QTc prolongation rate: 10%). Overall, the 60 mg group had a longer duration of remission (9.1 weeks versus 4.2 weeks), higher HSCT rate (42% versus 32%) and longer median OS (27.3 months versus 20.9 months), leading this dose to be selected for future quizartinib studies.
The phase III QuANTUM-R study evaluated single-agent quizartinib versus salvage chemotherapy in AML with FLT3-ITD mutation (ClinicalTrials.gov identifier: NCT02039726). A total of 367 patients were randomized in a 2:1 ratio to receive either single-agent quizartinib (30 mg lead-in, then 60 mg daily after QTc assessment) or investigator’s choice of salvage chemotherapy [this included low-dose cytarabine, MEC (mitoxantrone, etoposide, and cytarabine) or FLAG-Ida (fludarabine, cytarabine, idarubicin, and G-CSF)]. Quizartinib prolonged OS compared with chemotherapy (median OS: 27.0 weeks versus 20.4 weeks; 1-year OS rate: 27% versus 20%; p = 0.0177). These findings represent the first study of an FLT3 inhibitor shown to improve OS in the salvage setting; US FDA approval has been sought by the company that manufactures quizartinib, although a final regulatory decision is still pending.
Quizartinib has also been studied in the frontline and relapsed/refractory settings in combination with chemotherapy or hypomethylating agents. In a phase I/II study of quizartinib with azacitidine or low-dose cytarabine, the combination resulted in an overall response rate of 75% in patients with relapsed/refractory AML harboring a FLT3-ITD mutation; these results appear superior to those observed with quizartinib monotherapy in phase II and III studies.74 In the same study, 11 of 12 (92%) older patients >60 years of age in the frontline arm achieved response (CRc rate: 83%) with the combination with a median OS of 18.6 months. These results suggest that both response rates and duration of response may be meaningfully improved by combining quizartinib with hypomethylating agents (as was seen with azacitidine and sorafenib) and should be further evaluated in larger studies. A phase I study in 19 patients confirmed the safety and efficacy of quizartinib in combination with intensive chemotherapy in patients with newly diagnosed AML ⩽ 60 years of age.75 These data served as the basis for the randomized, multinational, phase III QuANTUM-First that is testing intensive chemotherapy with quizartinib or with placebo during induction, consolidation, and up to 1 year of maintenance in patients with FLT3-ITD-mutated AML (ClinicalTrials.gov identifier: NCT02668653).
Crenolanib
Crenolanib is a second-generation FLT3 TKI that is capable of inhibiting both ITD and TKD mutations, with activity against other signaling pathways including PDGFR. In a phase I study in relapsed/refractory FLT3-mutated AML, an overall response rate of 50% (CRc rate: 39%) was achieved among 18 patients with no prior FLT3 inhibitor exposure; among 36 patients who had received prior FLT3 inhibitors, the overall response rate was 31% (CRc rate: 17%).76 Activity was noted among patients with both ITD and TKD mutations.
The developmental strategy of crenolanib has focused on evaluating this agent in combination with cytotoxic chemotherapy, both in the frontline and the relapse setting. A study of the IA regimen plus crenolanib in 13 patients with relapsed/refractory FLT3-mutated AML resulted in an overall response rate of 36%.77 In a frontline study of crenolanib in combination with standard ‘7+3’ induction and high-dose cytarabine consolidation in patients with a median age of 55 years (range, 22–74 years), CR/CRi was achieved in 24 of 25 evaluable patients (96%), with 88% of patients achieving CR.78 In a later analysis of the 29 patients ⩽60 years of age treated on the study, only 2 patients had relapsed (1 systemic, 1 central nervous system only) with a median duration of follow up of 14 months, suggesting durable responses with the combination. A randomized phase III study of chemotherapy with mitoxantrone and cytarabine with or without crenolanib in patients with relapsed/refractory AML with activating FLT3 mutation is currently accruing (ClinicalTrials.gov identifier: NCT02298166). A frontline, randomized, multicenter phase III study evaluating standard 3+7 induction with either crenolanib or midostaurin during induction, consolidation and up to 1 year of maintenance has also been initiated (ClinicalTrials.gov identifier: NCT03258931)
Gilteritinib
Gilteritinib is a dual FLT3/AXL inhibitor that also has clinical activity against TKD mutations, and does not inhibit KIT.79 Notably, increased Axl-1 expression has been implicated as a resistance mechanism with other FLT3 inhibitors, including midostaurin and quizartinib.80,81 In a phase I/II study of 252 patients with relapsed/refractory AML, including FLT3-wildtype and FLT3-mutated, the overall response rate with gilteritinib was 40% (CRc rate: 30%).82 An overall response rate of 49% was observed in FLT3-mutated patients (ITD, D835, or both; CRc rate: 37%) compared with 12% in FLT3-wildtype AML. A total of 37% of patients who failed at least one prior FLT3 inhibitor achieved a response, suggesting the potential for gilteritinib to overcome resistance to other TKIs and suggesting that gilteritinib would still be active in patients relapsed/refractory to induction with midostaurin. Among patients with a FLT3 mutation who received ⩾80 mg daily (n = 169), the overall response rate was 52% (CRc rate: 41%), and the median OS was 31 weeks. Gilteritinib was well tolerated, and the maximum tolerated dose was established at 300 mg daily. However, given uniform in vivo target inhibition across all the dose levels studied and a high proportion of patients responding to the 120 mg/day dose, this lower dose was selected for further evaluation. A phase II study of gilteritinib, gilteritinib plus azacitidine, or azacitidine alone for patients with newly diagnosed FLT3-mutated AML who are unfit for intensive chemotherapy is currently accruing patients (ClinicalTrials.gov identifier: NCT02752035). A randomized phase III study of gilteritinib versus salvage chemotherapy in relapsed/refractory FLT3-mutated AML has recently completed accrual (ADMIRAL study; ClinicalTrials.gov identifier: NCT03182244). Based on an interim analysis of this trial that showed a CRc rate of 21% and median time to best response of approximately 3.5 months, the US FDA approved gilteritinib as single-agent therapy for adults with relapsed/refractory FLT3-mutated AML in November 2018; full details of the study findings, including survival data, are eagerly awaited.
Mechanisms of FLT3 inhibitor resistance and potential novel targets
Despite encouraging response rates with FLT3 TKI-based regimens in both the frontline and relapsed/refractory settings, many patients still fail to respond to FLT3 inhibitor therapy or subsequently relapse. To improve these outcomes, an increased understanding of the mechanisms underpinning FLT3 inhibitor resistance is needed. A full description of the numerous mechanisms of FLT3 inhibitor resistance is outside the scope of this manuscript; however, here we outline some of the best described pathways of resistance reported in preclinical and clinical studies.
One such mechanism of resistance is the development of secondary mutations in the FLT3 gene.83 These mutations are often single amino acid exchanges in the activating loop residues (e.g. D835, I836, D839, Y842) or gatekeeper residues (e.g. F691).84,85 This resistance mechanism is a particular issue for patients treated with the type II inhibitors (e.g. sorafenib or quizartinib) that do not have significant activity against TKD mutations.46,51,86 However, while treatment-emergent TKD mutations are an important mechanism of FLT3 inhibitor resistance, these mutations may not be the major modality of FLT3 inhibitor resistance. For example, in one study, only 22% of patients treated with FLT3 inhibitors developed a new detectable TKD mutation at the time of progression.87 Other extrinsic alterations also play an important role in the development of FLT3 inhibitor resistance.
Various intracellular and extracellular pathways have been implicated in FLT3 TKI resistance. These serve as rational targets for FLT3 inhibitor-based combination regimens, several of which are currently undergoing clinical testing (Table 2). Bone marrow microenvironment and stroma-mediated processes, including increased basic fibroblast growth factor (FGF2) and CXCL12/CXCR4 signaling, have been described in FLT3 inhibitor-resistant patient cases and may protect FLT3-mutated progenitors.88–92 Increased activity of several parallel prosurvival pathways also contribute to resistance. FLT3-independent activation of the RAS/RAF/MEK/ERK pathway may serve as a potential therapeutic target.93 The PI3K/AKT/mTOR pathway is also upregulated in FLT3 TKI-resistant cases, suggesting that AKT or mTOR inhibitors may be useful in this setting.12,94–96 Constant signaling through FLT3 leads to downstream activation of STAT5 and the oncogenic serine/threonine kinase Pim-1, leading to a positive feedback loop that consolidates aberrant FLT3 signaling.97,98 A Pim kinase inhibitor is currently being studied in an early phase trial in patients with relapsed/refractory AML (ClinicalTrials.gov identifier: NCT02078609) and there is also interest in the use of STAT5 inhibitors in patients harboring FLT3 mutations.97,99,100 CDK4 and CDK6 regulate transcription of both FLT3 and Pim-1, and therefore inhibition of these kinases may target multiple pathogeneic pathways that are important in FLT3-mutated AML.101 An ongoing trial combining sorafenib with the CDK4/6 inhibition is currently ongoing and seeks to test this hypothesis. Additionally, upregulation of anti-apoptotic proteins (e.g. Bcl-2, Bcl-xL and Mcl-1) is often observed in TKI-resistant cases and may contribute to a resistant phenotype.102–105 Notably, this process may in part be driven by aberrant Pim-1 kinase activity.106–108 Supporting these apoptotic pathways as a potential therapeutic target in FLT3-mutated AML, in vitro synergy between FLT3 and Bcl-2 inhibitors has been reported.104 Clinical trials combining second-generation FLT3 inhibitors with venetoclax are currently planned (ClinicalTrials.gov identifier: NCT03625505).
Table 2.
Combination regimen | Mechanism of action of combination agent | Mechanistic rationale for combination | Clinicaltrials.gov identifier |
---|---|---|---|
LGH447 + midostaurin | Pim kinase inhibitor | Pim kinase activity mediates FLT3 inhibitor resistance; combination increases apoptosis97,99 | NCT02078609 |
Milademetan (DS-3032b) + quizartinib | MDM2 inhibitor | MDM2 inhibitors restore p53 tumor suppression function109 | NCT03552029 |
Omacetaxine mepesuccinate + sorafenib Omacetaxine mepesuccinate + quizartinib |
Protein synthesis inhibitor | Synergistic with FLT3 inhibitors to suppress leukemic proliferation110,111 | NCT03170895 NCT03135054 |
Palbociclib + sorafenib | CDK4/6 inhibitor | CDK4/6 regulate transcription of FLT3 and Pim kinases (mechanism of FLT3 inhibitor resistance)101 | NCT03132454 |
SEL24 (dual pan-Pim/FLT3 inhibitor) | Pim kinase inhibitor | Pim kinase activity mediates FLT3 inhibitor resistance; combination increases apoptosis97,99 | NCT03008187 |
Venetoclax + gilteritinib | Bcl-2 inhibitor | Upregulation of anti-apoptotic proteins (e.g. Bcl-2, BCL-xL and Mcl-1) mediates FLT3 inhibitor resistance104–108 | NCT03625505 |
Vorinostat + bortezomib + sorafenib | Histone deacetylase inhibitor (vorinostat) Proteasome inhibitor (bortezomib) |
Histone deacetylase inhibitors synergistically induce apoptosis with FLT3 inhibitors;112 proteasome inhibitors induce FLT3-ITD degradation through autophagy113 | NCT01534260 |
Conventional chemotherapy and hypomethylating agent-based combinations are excluded.
Future directions
The development of FLT3 inhibitors has revolutionized the standard of care for FLT3-mutated AML, and has improved the outcomes of these patients both in the frontline and relapsed/refractory settings.41,114 With the rapid development of new, active second-generation FLT3 inhibitors with varying properties, important questions remain. In the frontline setting, the addition of the multikinase FLT3 inhibitor midostaurin to induction therapy has been shown to improve OS in younger patients with FLT3-mutated (ITD and D835) AML; however, whether better or worse results would be obtained with more specific, second-generation FLT3 TKIs such as quizartinib, gilteritinib, or crenolanib is currently unknown. Theoretically, these more specific FLT3 inhibitors may play a larger role in the relapsed/refractory setting when the disease is more monoclonal and dependent on FLT3 signaling for survival.53 This hypothesis is being explored in ongoing randomized studies of frontline induction with second-generation FLT3 TKIs (including quizartinib, crenolanib, and gilteritinib). It is also unclear how the widespread use of FLT3 inhibitors in the frontline setting will impact the need for HSCT in first remission for these patients. While in the RATIFY study the best outcomes were observed in patients who received midostaurin with induction and then underwent subsequent HSCT in remission, it is possible that longer FLT3 inhibitor maintenance beyond 1 year or the use of alternative FLT3 inhibitors may alter our current risk stratification of patients with FLT3-mutated AML, particularly when other established prognostic factors are considered (e.g. NPM1 status, type of FLT3 mutation, and FLT3 allelic ratio). However, at this time, the recommended approach in newly diagnosed FLT3-mutated AML in fit patients remains the addition of midostaurin to intensive chemotherapy, with a plan to go to HSCT in first remission.
For those patients who do undergo HSCT in first remission, the role of FLT3 inhibitors as maintenance therapy is unclear. Studies of the first-generation TKIs sorafenib and midostaurin have suggested that they may be effective and associated with improved post-HSCT EFS and OS but may be associated with more toxicities and require frequent and liberal dose reduction in this setting.32,115–118 In contrast, preliminary evidence suggests that quizartinib, a more specific FLT3 inhibitor, is well tolerated and effective as post-HSCT maintenance.119 A randomized study of gilteritinib versus placebo administered after HSCT in FLT3-mutated AML is ongoing and may help to more definitively address the benefit of FLT3 inhibition in this setting (BMT CTN 1506; ClinicalTrials.gov identifier: NCT02997202).
Many investigators have evaluated FLT3 inhibitors with standard chemotherapy or hypomethyla-ting agents, both of which commonly serve as backbone regimens for the study of new AML drugs. However, with an increased understanding of the intra- and extracellular mechanisms responsible for TKI resistance, rationally designed combinations with agents targeting specific resistance pathways will hopefully lead to further improved outcomes. For example, a hypomethylating agent plus the Bcl-2 inhibitor venetoclax has shown very promising activity in AML and is emerging as a potential new standard of care for the frontline treatment of older adults with newly diagnosed AML not candidates for intensive chemotherapy;120 however, despite the preclinical rationale for Bcl-2 inhibition in FLT3-mutated AML, the role of venetoclax in this setting remains largely unknown and needs to be evaluated in rationally designed combinatorial trials. While combination studies with FLT3 inhibitors and other targeted agents such as venetoclax will initially be performed predominantly in patients with relapsed/refractory disease, promising data in this setting may ultimately support the evaluation of these novel combinations in the frontline setting possibly with the addition of a hypomethylating agent backbone.
Finally, the role of FLT3 inhibitors in patients with AML harboring wild type FLT3 remains an open question. The SORAML study showed an EFS benefit and a trend to OS benefit with longer follow up with the addition of sorafenib to standard chemotherapy in younger patients with newly diagnosed AML irrespective of FLT3 status; however, as sorafenib targets multiple kinases, it is not clear what specific properties of sorafenib were the primary drivers for these outcomes.44,65 A similar randomized phase III study of a different multikinase inhibitor, midostaurin in combination with induction chemotherapy in patients with FLT3 wildtype AML is currently ongoing, and may provide further support for the use of FLT3 inhibitors in the frontline management of patients without FLT3 mutations (ClinicalTrials.gov identifier: NCT03512197).
Footnotes
Funding: This study was supported by the MD Anderson Cancer Center Support Grant CA016672, the MD Anderson Cancer Center Leukemia SPORE CA100632, the Dick Clark Leukemia Research Fund, and the MD Anderson Moon Shots Program.
Conflict of interest statement: ND has received research funding from Daiichi-Sankyo, Novartis, Astellas and served as an advisor/consultant to Daiichi-Sankyo, Novartis, Astellas, and Arog pharmaceuticals. HK has served as an advisor/consultant to Daiichi-Sankyo. FR has served on an advisor board and received honoraria from Astellas. NS has no relevant conflicts of interest to disclose.
ORCID iD: Naval Daver https://orcid.org/0000-0002-2983-2738
Contributor Information
Nicholas J. Short, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA
Hagop Kantarjian, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Farhad Ravandi, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Naval Daver, Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
References
- 1. Short NJ, Rytting ME, Cortes JE. Acute myeloid leukaemia. Lancet 2018; 593–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 2016; 374: 2209–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368: 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. 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. [PubMed] [Google Scholar]
- 6. 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. [DOI] [PubMed] [Google Scholar]
- 7. Yanada M, Matsuo K, Suzuki T, et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis. Leukemia 2005; 19: 1345–1349. [DOI] [PubMed] [Google Scholar]
- 8. 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. [DOI] [PubMed] [Google Scholar]
- 9. 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. [PubMed] [Google Scholar]
- 10. Frohling 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. [DOI] [PubMed] [Google Scholar]
- 11. 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. [DOI] [PubMed] [Google Scholar]
- 12. Brandts CH, Sargin B, Rode M, et al. Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res 2005; 65: 9643–9650. [DOI] [PubMed] [Google Scholar]
- 13. Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood 2002; 100: 1532–1542. [DOI] [PubMed] [Google Scholar]
- 14. 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. [PubMed] [Google Scholar]
- 15. Kuchenbauer F, Kern W, Schoch C, et al. Detailed analysis of FLT3 expression levels in acute myeloid leukemia. Haematologica 2005; 90: 1617–1625. [PubMed] [Google Scholar]
- 16. 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. [DOI] [PubMed] [Google Scholar]
- 17. 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. [DOI] [PubMed] [Google Scholar]
- 18. Abu-Duhier FM, Goodeve AC, Wilson GA, et al. Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia. Br J Haematol 2001; 113: 983–988. [DOI] [PubMed] [Google Scholar]
- 19. Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 2001; 97: 2434–2439. [DOI] [PubMed] [Google Scholar]
- 20. Sato T, Yang X, Knapper S, et al. FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo. Blood 2011; 117: 3286–3293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Mead AJ, Linch DC, Hills RK, et al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood 2007; 110: 1262–1270. [DOI] [PubMed] [Google Scholar]
- 22. Whitman SP, Ruppert AS, Radmacher MD, et al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood 2008; 111: 1552–1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Moreno I, Martin G, Bolufer P, et al. Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia. Haematologica 2003; 88: 19–24. [PubMed] [Google Scholar]
- 24. 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. [DOI] [PubMed] [Google Scholar]
- 25. Dohner 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Garg M, Nagata Y, Kanojia D, et al. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood 2015; 126: 2491–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. 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. [DOI] [PubMed] [Google Scholar]
- 28. Thiede C, Koch S, Creutzig E, et al. Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML). Blood 2006; 107: 4011–4020. [DOI] [PubMed] [Google Scholar]
- 29. 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. [DOI] [PubMed] [Google Scholar]
- 30. 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. [DOI] [PubMed] [Google Scholar]
- 31. Ho AD, Schetelig J, Bochtler T, et al. Allogeneic stem cell transplantation improves survival in patients with acute myeloid leukemia characterized by a high allelic ratio of mutant FLT3-ITD. Biol Blood Marrow Transplant 2016; 22: 462–469. [DOI] [PubMed] [Google Scholar]
- 32. Schlenk R, 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). Blood 2015; 126: 322–322. [Google Scholar]
- 33. Stirewalt DL, Kopecky KJ, Meshinchi S, et al. Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia. Blood 2006; 107: 3724–3726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Ponziani V, Gianfaldoni G, Mannelli F, et al. The size of duplication does not add to the prognostic significance of FLT3 internal tandem duplication in acute myeloid leukemia patients. Leukemia 2006; 20: 2074–2076. [DOI] [PubMed] [Google Scholar]
- 35. 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. [DOI] [PubMed] [Google Scholar]
- 36. 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. [DOI] [PubMed] [Google Scholar]
- 37. Ma Y, Wu Y, Shen Z, et al. Is allogeneic transplantation really the best treatment for FLT3/ITD-positive acute myeloid leukemia? A systematic review. Clin Transplant 2015; 29: 149–160. [DOI] [PubMed] [Google Scholar]
- 38. Doubek M, Muzik J, Szotkowski T, et al. Is FLT3 internal tandem duplication significant indicator for allogeneic transplantation in acute myeloid leukemia? An analysis of patients from the Czech Acute Leukemia Clinical Register (ALERT). Neoplasma 2007; 54: 89–94. [PubMed] [Google Scholar]
- 39. DeZern AE, Sung A, Kim S, et al. Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution. Biol Blood Marrow Transplant 2011; 17: 1404–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Lin PH, Lin CC, Yang HI, et al. Prognostic impact of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia patients with internal tandem duplication of FLT3. Leuk Res 2013; 37: 287–292. [DOI] [PubMed] [Google Scholar]
- 41. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Ravandi F, Cortes JE, Jones D, et al. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010; 28: 1856–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Ravandi F, Arana Yi C, Cortes JE, et al. Final report of phase II study of sorafenib, cytarabine and idarubicin for initial therapy in younger patients with acute myeloid leukemia. Leukemia 2014; 28: 1543–1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Rollig 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. [DOI] [PubMed] [Google Scholar]
- 45. Burchert A, Bug G, Finke J, et al. Sorafenib as maintenance therapy post allogeneic stem cell transplantation for FLT3-ITD positive AML: results from the randomized, double-blind, placebo-controlled multicentre sormain trial. Blood 2018; 132: 661–661. [Google Scholar]
- 46. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Sexauer A, Perl A, Yang X, et al. Terminal myeloid differentiation in vivo is induced by FLT3 inhibition in FLT3/ITD AML. Blood 2012; 120: 4205–4214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Karaman MW, Herrgard S, Treiber DK, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol 2008; 26: 127–132. [DOI] [PubMed] [Google Scholar]
- 49. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Grundler R, Thiede C, Miething C, et al. Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor. Blood 2003; 102: 646–651. [DOI] [PubMed] [Google Scholar]
- 51. Smith CC, Lin K, Stecula A, et al. FLT3 D835 mutations confer differential resistance to type II FLT3 inhibitors. Leukemia 2015; 29: 2390–2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Levis M, Pham R, Smith BD, et al. In vitro studies of a FLT3 inhibitor combined with chemotherapy: sequence of administration is important to achieve synergistic cytotoxic effects. Blood 2004; 104: 1145–1150. [DOI] [PubMed] [Google Scholar]
- 53. Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 2012; 481: 506–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Pratz KW, Sato T, Murphy KM, et al. FLT3-mutant allelic burden and clinical status are predictive of response to FLT3 inhibitors in AML. Blood 2010; 115: 1425–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. 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. [DOI] [PubMed] [Google Scholar]
- 56. Smith BD, Levis M, Beran M, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 2004; 103: 3669–3676. [DOI] [PubMed] [Google Scholar]
- 57. Knapper S, Burnett AK, Littlewood T, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 2006; 108: 3262–3270. [DOI] [PubMed] [Google Scholar]
- 58. Levis M, Ravandi F, Wang ES, et al. Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse. Blood 2011; 117: 3294–3301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Knapper S, Russell N, Gilkes A, et al. A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML. Blood 2017; 129: 1143–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Pratz KW, Cho E, Levis MJ, et al. A pharmacodynamic study of sorafenib in patients with relapsed and refractory acute leukemias. Leukemia 2010; 24: 1437–1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Crump M, Hedley D, Kamel-Reid S, et al. A randomized phase I clinical and biologic study of two schedules of sorafenib in patients with myelodysplastic syndrome or acute myeloid leukemia: a NCIC (National Cancer Institute of Canada) Clinical Trials Group Study. Leuk Lymphoma 2010; 51: 252–260. [DOI] [PubMed] [Google Scholar]
- 62. Borthakur G, Kantarjian H, Ravandi F, et al. Phase I study of sorafenib in patients with refractory or relapsed acute leukemias. Haematologica 2011; 96: 62–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Ohanian M, Garcia-Manero G, Levis M, et al. Sorafenib combined with 5-azacytidine (AZA) in older patients with untreated FLT3-ITD mutated acute myeloid leukemia (AML). Am J Hematol 2018; 93: 1136–1141. [DOI] [PubMed] [Google Scholar]
- 65. Rollig C, Serve H, Hüttmann A, et al. The addition of sorafenib to standard AML treatment results in a substantial reduction in relapse risk and improved survival. updated results from long-term follow-up of the randomized-controlled soraml trial. Blood 2017; 130: 721–721. [Google Scholar]
- 66. 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. [DOI] [PubMed] [Google Scholar]
- 67. 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. [DOI] [PubMed] [Google Scholar]
- 68. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Stone RM, Fischer T, Paquette R, et al. Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia. Leukemia 2012; 26: 2061–2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Araki D, Wood BL, Othus M, et al. Allogeneic hematopoietic cell transplantation for acute myeloid leukemia: time to move toward a minimal residual disease-based definition of complete remission? J Clin Oncol 2016; 34: 329–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Cortes JE, Kantarjian H, Foran JM, et al. Phase I study of quizartinib administered daily to patients with relapsed or refractory acute myeloid leukemia irrespective of fms-like tyrosine kinase 3-internal tandem duplication status. J Clin Oncol 2013; 31: 3681–3687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Cortes J, Perl AE, Dohner H, et al. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 2018; 19: 889–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Cortes JE, Tallman MS, Schiller GJ, et al. Phase 2b study of 2 dosing regimens of quizartinib monotherapy in FLT3-ITD-mutated, relapsed or refractory AML. Blood 2018; 132: 598–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Swaminathan M, Kantarjian HM, Daver N, et al. The combination of quizartinib with azacitidine or low dose cytarabine is highly active in patients (Pts) with FLT3-ITD mutated myeloid leukemias: interim report of a phase I/II trial. Blood 2017; 130: 723–723. [Google Scholar]
- 75. Altman JK, Foran JM, Pratz KW, et al. Phase 1 study of quizartinib in combination with induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia. Am J Hematol 2018; 93: 213–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Cortes JE, Kantarjian HM, Kadia TM, et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, to demonstrate clinical activity in multiply relapsed FLT3-ITD and D835 AML. J Clin Oncol 2016; 34: 7008–7008. [Google Scholar]
- 77. Ohanian M, Kantarjian HM, Borthakur G, et al. Efficacy of a type I FLT3 inhibitor, crenolanib, with idarubicin and high-dose ara-c in multiply relapsed/refractory FLT3+ AML. Blood 2016; 128: 2744–2744.27932331 [Google Scholar]
- 78. 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). Blood 2016; 128: 1071–1071. [Google Scholar]
- 79. Lee LY, Hernandez D, Rajkhowa T, et al. Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood 2017; 129: 257–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Park IK, Mundy-Bosse B, Whitman SP, et al. Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia. Leukemia 2015; 29: 2382–2389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Park IK, Mishra A, Chandler J, et al. Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target. Blood 2013; 121: 2064–2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Daver N, Cortes J, Ravandi F, et al. Secondary mutations as mediators of resistance to targeted therapy in leukemia. Blood 2015; 125: 3236–3245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Cools J, Mentens N, Furet P, et al. Prediction of resistance to small molecule FLT3 inhibitors: implications for molecularly targeted therapy of acute leukemia. Cancer Res 2004; 64: 6385–6389. [DOI] [PubMed] [Google Scholar]
- 85. Heidel F, Solem FK, Breitenbuecher F, et al. Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood 2006; 107: 293–300. [DOI] [PubMed] [Google Scholar]
- 86. Man CH, Fung TK, Ho C, et al. Sorafenib treatment of FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms of subsequent nonresponsiveness associated with the emergence of a D835 mutation. Blood 2012; 119: 5133–5143. [DOI] [PubMed] [Google Scholar]
- 87. Alvarado Y, Kantarjian HM, Luthra R, et al. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer 2014; 120: 2142–2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Traer E, Martinez J, Javidi-Sharifi N, et al. FGF2 from marrow microenvironment promotes resistance to FLT3 inhibitors in acute myeloid leukemia. Cancer Res 2016; 76: 6471–6482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kojima K, McQueen T, Chen Y, et al. p53 activation of mesenchymal stromal cells partially abrogates microenvironment-mediated resistance to FLT3 inhibition in AML through HIF-1alpha-mediated down-regulation of CXCL12. Blood 2011; 118: 4431–4439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Onishi C, Mori-Kimachi S, Hirade T, et al. Internal tandem duplication mutations in FLT3 gene augment chemotaxis to Cxcl12 protein by blocking the down-regulation of the Rho-associated kinase via the Cxcl12/Cxcr4 signaling axis. Journal Biol Chem 2014; 289: 31053–31065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Yang X, Sexauer A, Levis M. Bone marrow stroma-mediated resistance to FLT3 inhibitors in FLT3-ITD AML is mediated by persistent activation of extracellular regulated kinase. Br J Haematol 2014; 164: 61–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Parmar A, Marz S, Rushton S, et al. Stromal niche cells protect early leukemic FLT3-ITD+ progenitor cells against first-generation FLT3 tyrosine kinase inhibitors. Cancer Res 2011; 71: 4696–4706. [DOI] [PubMed] [Google Scholar]
- 93. Zhang W, Borthakur G, Gao C, et al. The dual MEK/FLT3 inhibitor E6201 exerts cytotoxic activity against acute myeloid leukemia cells harboring resistance-conferring FLT3 mutations. Cancer Res 2016; 76: 1528–1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Lindblad O, Cordero E, Puissant A, et al. Aberrant activation of the PI3K/mTOR pathway promotes resistance to sorafenib in AML. Oncogene 2016; 35: 5119–5131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Mohi MG, Boulton C, Gu TL, et al. Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci U S A 2004; 101: 3130–3135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Weisberg E, Liu Q, Zhang X, et al. Selective Akt inhibitors synergize with tyrosine kinase inhibitors and effectively override stroma-associated cytoprotection of mutant FLT3-positive AML cells. PLoS One 2013; 8: e56473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Green AS, Maciel TT, Hospital MA, et al. Pim kinases modulate resistance to FLT3 tyrosine kinase inhibitors in FLT3-ITD acute myeloid leukemia. Sci Adv 2015; 1: e1500221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Natarajan K, Xie Y, Burcu M, et al. Pim-1 kinase phosphorylates and stabilizes 130 kDa FLT3 and promotes aberrant STAT5 signaling in acute myeloid leukemia with FLT3 internal tandem duplication. PLoS One 2013; 8: e74653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Kapoor S, Natarajan K, Baldwin PR, et al. Concurrent Inhibition of Pim and FLT3 Kinases Enhances Apoptosis of FLT3-ITD Acute Myeloid Leukemia Cells through Increased Mcl-1 Proteasomal Degradation. Clin Cancer Res 2018; 24: 234–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Nelson EA, Walker SR, Xiang M, et al. The STAT5 inhibitor pimozide displays efficacy in models of acute myelogenous leukemia driven by FLT3 mutations. Genes Cancer 2012; 3: 503–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Uras IZ, Walter GJ, Scheicher R, et al. Palbociclib treatment of FLT3-ITD+ AML cells uncovers a kinase-dependent transcriptional regulation of FLT3 and PIM1 by CDK6. Blood 2016; 127: 2890–2902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Breitenbuecher F, Markova B, Kasper S, et al. A novel molecular mechanism of primary resistance to FLT3-kinase inhibitors in AML. Blood 2009; 113: 4063–4073. [DOI] [PubMed] [Google Scholar]
- 103. Bagrintseva K, Geisenhof S, Kern R, et al. FLT3-ITD-TKD dual mutants associated with AML confer resistance to FLT3 PTK inhibitors and cytotoxic agents by overexpression of Bcl-x(L). Blood 2005; 105: 3679–3685. [DOI] [PubMed] [Google Scholar]
- 104. Kohl TM, Hellinger C, Ahmed F, et al. BH3 mimetic ABT-737 neutralizes resistance to FLT3 inhibitor treatment mediated by FLT3-independent expression of BCL2 in primary AML blasts. Leukemia 2007; 21: 1763–1772. [DOI] [PubMed] [Google Scholar]
- 105. Yoshimoto G, Miyamoto T, Jabbarzadeh-Tabrizi S, et al. FLT3-ITD up-regulates MCL-1 to promote survival of stem cells in acute myeloid leukemia via FLT3-ITD-specific STAT5 activation. Blood 2009; 114: 5034–5043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Kim KT, Baird K, Ahn JY, et al. Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival. Blood 2005; 105: 1759–1767. [DOI] [PubMed] [Google Scholar]
- 107. Kim KT, Levis M, Small D. Constitutively activated FLT3 phosphorylates BAD partially through pim-1. Br J Haematol 2006; 134: 500–509. [DOI] [PubMed] [Google Scholar]
- 108. Adam M, Pogacic V, Bendit M, et al. Targeting PIM kinases impairs survival of hematopoietic cells transformed by kinase inhibitor-sensitive and kinase inhibitor-resistant forms of Fms-like tyrosine kinase 3 and BCR/ABL. Cancer Res 2006; 66: 3828–3835. [DOI] [PubMed] [Google Scholar]
- 109. Seipel K, Marques MAT, Sidler C, et al. MDM2- and FLT3-inhibitors in the treatment of FLT3-ITD acute myeloid leukemia, specificity and efficacy of NVP-HDM201 and midostaurin. Haematologica 2018; 103: 1862–1872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Xu G, Mao L, Liu H, et al. Sorafenib in combination with low-dose-homoharringtonine as a salvage therapy in primary refractory FLT3-ITD-positive AML: a case report and review of literature. Int J Clin Exp Med 2015; 8: 19891–19894. [PMC free article] [PubMed] [Google Scholar]
- 111. Lam SS, Ho ES, He BL, et al. Homoharringtonine (omacetaxine mepesuccinate) as an adjunct for FLT3-ITD acute myeloid leukemia. Sci Transl Med 2016; 8: 359ra129. [DOI] [PubMed] [Google Scholar]
- 112. Pietschmann K, Bolck HA, Buchwald M, et al. Breakdown of the FLT3-ITD/STAT5 axis and synergistic apoptosis induction by the histone deacetylase inhibitor panobinostat and FLT3-specific inhibitors. Mol Cancer Ther 2012; 11: 2373–2383. [DOI] [PubMed] [Google Scholar]
- 113. Larrue C, Saland E, Boutzen H, et al. Proteasome inhibitors induce FLT3-ITD degradation through autophagy in AML cells. Blood 2016; 127: 882–892. [DOI] [PubMed] [Google Scholar]
- 114. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Chen YB, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Pratz KW, Gojo I, Karp JE, et al. Prospective study of peri-transplant use of sorafenib as remission maintenance for FLT3-ITD patients undergoing allogeneic transplantation. Blood 2015; 126: 3164–3164. [Google Scholar]
- 117. Schlenk RF, Fiedler W, Salih HR, et al. Impact of age and midostaurin-dose on response and outcome in acute myeloid leukemia with FLT3-ITD: interim-analyses of the AMLSG 16-10 trial. Blood 2016; 128: 449–449.27268085 [Google Scholar]
- 118. Maziarz RT, Patnaik MM, Scott BL, et al. Radius: a phase 2, randomized trial of standard of care (SOC) with or without midostaurin to prevent relapse following allogeneic hematopoietic stem cell transplant (alloHSCT) in patients (pts) with FLT3-Itd-mutated acute myeloid leukemia (AML). Blood 2016; 128: 2248–2248. [Google Scholar]
- 119. Sandmaier BM, Khaled SK, Oran B, et al. Results of a phase 1 study of quizartinib (AC220) as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic cell transplantation. Blood 2014; 124: 428–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. DiNardo CD, Pratz KW, Letai A, et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol 2018; 19: 216–228. [DOI] [PubMed] [Google Scholar]