Quizartinib

Quizartinib for the treatment of acute myeloid leukemia

Alejandro Garcia-Horton & Karen Wl Yee

To cite this article: Alejandro Garcia-Horton & Karen Wl Yee (2020): Quizartinib for the treatment of acute myeloid leukemia, Expert Opinion on Pharmacotherapy, DOI: 10.1080/14656566.2020.1801637
To link to this article: https://doi.org/10.1080/14656566.2020.1801637

Published online: 09 Aug 2020.

Submit your article to this journal

Article views: 20
View related articles View Crossmark data

Full Terms & Conditions of access and use can be found at https://www.tandfonline.com/action/journalInformation?journalCode=ieop20

EXPERT OPINION ON PHARMACOTHERAPY

https://doi.org/10.1080/14656566.2020.1801637

DRUG EVALUATION
Quizartinib for the treatment of acute myeloid leukemia
Alejandro Garcia-Horton a and Karen Wl Yeeb
aDivision of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, CANADA; bDivision
of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, CANADA

ABSTRACT
Introduction: . Up to 30% of patients with acute myeloid leukemia (AML) have a mutation in the FLT3 receptor. Molecular targets have acquired a significant interest in the treatment of AML and are changing patient outcomes, including improvement of overall survival (OS) and remission rates. FLT3 inhibitors have obtained a central role in how we treat AML.
Areas covered: . This article reviews the mechanism of action, pharmacology, clinical efficacy, and safety of quizartinib, a FLT3 inhibitor, for the treatment of acute myeloid leukemia.
Expert opinion: . Quizartinib yielded an improvement in OS and complete remission (CR) rates in a
phase 3 trial for relapsed/refractory FLT3-mutated AML. The toxicities are manageable; however, it is associated with significant QTc prolongation and myelosuppression. The FDA and EMA did not grant drug approval to quizartinib in the relapsed/refractory setting due to the lack of a significant benefit – to-risk ratio, safety concerns and concerns with credibility and generalizability of the trial data. Results from the frontline phase 3 study evaluating quizartinib with intensive chemotherapy are eagerly awaited. Ongoing studies are investigating its toxicity and efficacy with other therapeutic agents and will help to clarify its role in the treatment of FLT3-ITD-mutated AML.
ARTICLE HISTORY
Received 24 April 2020
Accepted 22 July 2020
KEYWORDS
Acute myeloid leukemia; ac220; flt3 inhibitor; quizartinib

⦁ Introduction
Acute myeloid leukemia (AML) is a heterogeneous cancer of hematopoietic progenitor cells [1]. It is the most common myeloid malignancy in adults with 21,450 new cases diag- nosed in the USA in 2019 [2]. Overall survival (OS) at 5 years is estimated to be 28.3% [2]; therefore, better treatments are required. Karyotype at time of diagnosis remains one of the most important prognostic factors, but up to 40–50% of patients with AML do not have chromosomal abnormalities [3]. Gene mutations have gained a more prominent role in prognostication and choice of therapeutics over the past dec- ade. Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are present in up to 30% of AML patients [3–5].
FLT3 is a member of the class III receptor tyrosine kinase (RTK) family that includes c-Kit, PDFGR-α, and CSF1 R [6]. Wild type (WT) FLT3 is expressed on bone marrow hematopoietic stem cells, with loss of expression as cells differentiate; hence, it plays a role in cellular growth and normal hematopoiesis [7]. WT FLT3 expression is maintained in leukemic cells in 70–100% of AML patients. Mutated FLT3 leads to constitutive kinase activation and results in proliferation and survival of blasts [8]. Two main types FLT3 of mutations have been identified:
(a) 75% of patients have internal tandem duplications (FLT3- ITD; approximately 25% of all AML cases) and (b) 25% have missense mutations in the tyrosine kinase domain activation loop (FLT3-TKD; approximately 7–10% of all AML cases), most involving codon D835 and to a lesser extent I836 [9]. FLT3-ITD mutations are associated with poor OS and higher risk of
relapse. Furthermore, FLT3-ITD mutant allelic burden, length of the ITD, the insertion site of the ITD and the mutational context (e.g. wild type NPM1) have been shown to impact outcomes [5,10–18].
FLT3 mutations are unstable and can change following che- motherapy and/or targeted therapy. FLT3 mutations detected at diagnosis may become undetectable at the time of relapse, and conversely, FLT3 mutations may be detected at the time of relapse, but not at the time of initial diagnosis. This may be due in part to the sensitivity and diagnostic accuracy of the test, the timing of the test or may indicate that the clone was either eliminated by chemotherapy or failed to re-expand from resi- dual leukemia stem cells or that the clone was present at low levels and selected for by the treatment [12,19–22]. This high- lights the need for serial testing for FLT3 mutations at different time points during the course of the disease [23].

⦁ Overview of the market
Currently there are two FLT3 inhibitors (i.e. midostaurin and gilteritinib) approved for the treatment of FLT3-mutated AML by the USA Food and Drug Administration (FDA), the European Medicines Agency (EMA), and Health Canada (HC), with other agents being tested in a variety of clinical trials in the upfront, relapsed/refractory, and maintenance settings (Table 1) [9]. FLT3 inhibitors can be classified as first genera- tion (e.g. sorafenib and midostaurin) and next generation inhibitors (e.g. crenolanib, quizartinib and gilteritinib) and/or

CONTACT Karen WL Yee [email protected] Division of Medical Oncology and Hematology, University Health Network – Princess Margaret Cancer Centre, Toronto, Ontario, CANADA
© 2020 Informa UK Limited, trading as Taylor & Francis Group

2
A. GARCIA-HORTON AND K. W. YEE
Table 1. Phase 3 trials with FLT3 inhibitors in patients with FLT3-mutated AML.
Agent Age N Regimens Select Eligibility Criteria Primary Outcomes

First-line therapy – Low Intensity

Gilteritinib (ASP2215); (NCT02752035)
≥ 18y 323 Gilteritiniba vs Gilteritinib + azacitidine
vs
Azacitidine
⦁ Untreated AML
⦁ Ineligible for intensive chemotherapy:
⦁ Age ≥ 75 y
⦁ Comorbidities (CHF NYHA ≤ 3; LVEF ≤ 50%; Creatinine > 177 μmol/L; ECOG ≥ 2; decreased DLco &/or on O2 ≤ 2 L/min; prior or current malignancy not requiring therapy; cumulative
anthracycline dose > 400 mg/m2 doxorubicin)
⦁ FLT3-ITD and/or FLT3-TKD [D835 or I836]
Overall survival

First-line therapy – High intensity

Quizartinib (AC220); (NCT02668653;
QuANTUM-First)

Midostaurin (PKC 412);
Crenolanib (CP- 868,596–26; AR-
868,596–26); (NCT03258931)
≥ 18y but
≤ 75y

≥ 18y but
≤ 60y
536 Quizartinib/Placebob + Intensive
chemotherapy followed by continuation/maintenance

510 Crenolanib/Midostaurinc + Intensive chemotherapy followed by continuation/maintenance
● Untreated de novo or secondary AML (prior MDS or MPN)
● Eligible for intensive chemotherapy
● FLT3-ITD (≥ 3% FLT3-ITD/total FLT3)
● No prior FLT3 inhibitors
● Untreated de novo AML
● Eligible for intensive chemotherapy
● FLT3-ITD and/or FLT3-TKD [D835]
Event-free survival

Event-free survival

Salvage therapy – High intensity

Crenolanib (CP- 868,596–26; AR-
868,596–26); (NCT03250338)
≥ 18y but
≤ 75y
322 Crenolanib/placebod + high-dose Ara-C & mitoxantrone (HAM)/FLAG-Ida
⦁ Primary refractory/relapsed to first-line intensive therapy or relapsed/refractory after second line therapy
⦁ FLT3-ITD and/or FLT3-TKD/D835
⦁ No prior crenolanib
Event-free survival

Crenolanib (CP- 868,596–26; AR-
868,596–26); (NCT02298166)
≥ 18y 276 Crenolanib/placebod + high-dose Ara-C
& mitoxantrone (HAM)
⦁ First relapse or primary refractory with/without HCT (after anthracycline + Ara-C-based induction therapy)
⦁ FLT3-ITD/TKD (≥ 5% FLT3-ITD/TKD/total FLT3)
Event-free survival Overall survival

Maintenance therapy – Monotherapy

Gilteritinib (ASP2215); (NCT02927262)

Gilteritinib (ASP2215); (NCT02997202)
≥ 18y 354 Gilteritinib vs Placebo (after induction/
consolidation) in CR1 x 2y

≥ 18y 536 Gilteritinib vs Placebo (after HCT) in CR1 x 2y
⦁ AML in CR1 (incl CRp & CRi)
⦁ No planned alloSCT
⦁ Completed planned no. of consolidations as per institutional standards
⦁ FLT3-ITD at diagnosis
⦁ May have received FLT3 inhibitor during induction &/or consolidation
⦁ AML in CR1 (incl CRp & CRi)
⦁ ≤ 2 inductions to achieve CR1
⦁ ≤ 12 mos from time of CR1 to registration
⦁ FLT3-ITD at diagnosis
Relapse-free survival

Relapse-free survival

Note: CHF – congestive heart failure; DLco – diffusing capacity of the lungs for carbon monoxide; ECOG – Eastern Cooperative Oncology Group performance status; LVEF – left ventricular ejection fraction; mos – months; NYHA –
New York heart Association; y – year
a2:1 randomization in favor of gilteritinib;
badministered during induction up to 2 cycles (idarubicin/daunorubicin + cytarabine), consolidation up to 4 cycles (single agent HiDAc) +/ – HCT & as maintenance for 1 y; c administered during induction (daunorubicin + cytarabine) & consolidation +/ – HCT; d administered during induction, consolidation, post-HCT & as maintenance

as Type I (e.g. midostaurin, crenolanib and gilteritinib) and Type II (e.g. sorafenib and quizartinib) inhibitors. The first generation FLT3 inhibitors are multi-targeted kinase inhibitors. The next generation FLT3 inhibitors were designed to have more specificity and potency against FLT3 with hopefully, fewer off-target effects.
The FLT3 receptor includes an extracellular domain, a trans- membrane region, and an intracellular module that consists of a juxtamembrane domain (JM) and a cytoplasmic tyrosine kinase domain (TKD) interrupted by a kinase insert region [24,25] (Figure 1). FLT3 ligand binds to the extracellular domain of the receptor, causing receptor dimerization and a shift from inactive to active conformation of the TKD. In turn, the intracellular modules move closer, allowing the kinase domain to catalyze phosphate transfer from ATP to the JM. This releases auto-inhibitory interactions in FLT3, which cause further phosphorylation of the intracellular module [24–26]. Together with the TKD, the phosphorylated residues will initi- ate downstream signaling pathways that include the phospha- tidylinositol 3-kinase (PI-3 K), Ras, and STAT5 proliferation pathways [27–29].
Type I FLT3 inhibitors bind to the ATP-binding site of the FLT3 receptor in its active conformation and are able to inhibit both FLT3-ITD and FLT3-TKD mutations; whereas, Type II inhibitors interact with a hydrophobic region adjacent to the ATP-binding site that is only accessible when the FLT3 receptor is in the inactive conformation and thus, prevent FLT3 receptor activation (Figure 2). Therefore, Type II FLT3 inhibitors are only able to inhibit the activity of FLT3-ITD mutations and not FLT3-TKD muta- tions, which result in an active conformation of receptor [9,23].

⦁ Sorafenib
Sorafenib is an oral, multikinase, Type II FLT3 inhibitor with activity against Ras/Raf, c-kit, VEGFR, PDGFR and FLT3 [30]. It is currently approved for treatment of advanced renal cell carcinoma (2005), hepatocellular carcinoma (2007), and thyr- oid carcinoma [2013, 31–33], but has been used off-label in patients with AML since 2007 [34]. Single agent sorafenib has shown responses in the relapsed setting after allogeneic stem cell transplant (alloSCT) with some durable responses [35,36]. Two randomized trials have evaluated sorafenib administered in conjunction with induction and consolida- tion therapy followed by maintenance therapy in younger and older patients with treatment-naïve (TN) AML, irrespec- tive of FLT3 status [37,38]. There were no differences in overall CR rates or OS between the 2 arms in both studies. These findings were consistent in the smaller subgroup of FLT3-ITD-mutated patients. There are no phase 3 studies evaluating standard induction chemotherapy in combination with sorafenib or placebo in FLT3-mutated AML.
Sorafenib has also been evaluated in combination with lower intensity therapy, such as azacitidine (AZA), as first-line therapy in TN older FLT3-mutated AML patients and as salvage therapy in relapsed/refractory FLT3-mutated AML patients [39,40]. Composite CR (CR + CR with incomplete count recov- ery [CRi] + CRp) rates were 70% in untreated patients and 37% in relapsed/refractory patients. Median duration of response
was 14.5 months and 2.3 months, respectively. Median OS was
8.3 months and 6.2 months, respectively.
Sorafenib maintenance after alloSCT in the randomized, phase 2 SORMAIN trial yielded in an improved 2-year relapse-free survival (RFS) in favor of sorafenib compared to placebo (85% vs 53.3%; P = 0.0135) [41]. At 30 months, OS favored the sorafenib arm, with a hazard ratio (HR) of 0.447 (P = 0.03) [42]. However, the study was terminated because of low accrual.

⦁ Midostaurin (PKC412)
Midostaurin is an oral, small molecule tyrosine kinase inhibitor (TKI) with activity against c-Kit, VEGFR, PDGFR, src and FLT3 [43,44]. Single agent midostaurin had limited activity in patients with relapsed/refractory FLT3-mutated or FLT3 WT AML [45,46]. The phase 3 RATIFY trial comparing standard cytarabine and daunorubicin induction and cytarabine conso- lidation plus midostaurin or placebo in adult patients with newly diagnosed FLT3-mutated AML aged 60 years or younger demonstrated comparable CR rates (58.9% vs 53.5%; P = 0.15), but there was an improvement in event-free survival (EFS; HR for event or death, 0.78; one-sided P = 0.002) and median OS (74.7 months vs 25.6 months; HR for death, 0.78; one-sided P = 0.009; 4-year OS 51.4% vs 44.3%), across all FLT3 mutation types, in favor of midostaurin [47]. The side effect profile was tolerable, with main toxicities being flu-like symptoms, mouth sores, unusual bleeding, and bruising [23,48]. This led to the approval of midostaurin with chemotherapy as first-line ther- apy in patients with newly diagnosed FLT3-mutated AML, irrespective of age, in 2017. However, the FDA and HC, unlike the EMA, did not approve midostaurin as maintenance ther- apy given the lack of a second randomization after completion of consolidation and the small number of patients (14–19%) who completed maintenance therapy. An unplanned subset analysis of the maintenance therapy in the RATIFY trial revealed no difference in disease-fee survival (DFS) and OS between the 2 arms (HR of 1.4; 95% CI, 0.63–3.3; P = 0.38 and
HR of 0.96; 95% CI, 0.58–1.59; P = 0.86, for midostaurin,
respectively) [49].

⦁ Gilteritinib (ASP2215)
Gilteritinib is an oral, highly selective, Type I FLT3 inhibitor with activity against AXL and FLT3-ITD and FLT3-TKD mutations [50]. In the phase 3 ADMIRAL study, 371 patients with FLT3-mutated AML who were refractory to (n = 146; 39%) or relapsed after (n = 225; 61%) first-line therapy were randomized 2:1 to gilter- itinib or preselected specific salvage therapy with lower intensity (low dose cytarabine [LDAC] or AZA) or higher intensity therapy (mitoxantrone, etoposide and cytarabine [MEC] or fludarabine, cytarabine, G-CSF and idarubicin [FLAG-Ida]) [51]. Median age was 62 years (range, 19–85). Eighty-eight percent had a FLT3-ITD mutation only and 8% a FLT3-TKD mutation only. Only 12.4% of patients had received prior FLT3 inihibitors (with 5.7% receiving midostaurin). Twenty percent had received prior alloSCT. Twelve percent of patients on the salvage chemotherapy arm did not receive therapy compared with 0.4% on the gilteritnib arm. Seventy-eight (31.6%) patients had gilteritinib doses increased

Figure 1. FLT3 receptor schematic. The areas where internal tandem duplications (ITD) and tyrosine kinase domain (TKD) mutations occur are shown. This figure is reproduced from [8] with permission of Jon Wiley and Sons.

Figure 2. Binding of FLT3 inhibitors to the FLT3 receptor. Type II inhibitors are only able to bind to FLT3 in its inactive conformation. Type I inhibitors bind to FLT3 both in active and inactive conformation, showing activity in ITD and TKD mutations. This figure is reproduced from [23].

to 200 mg/day. Median number of cycles of gilteritinib therapy was 5 (range, 1–33). Single agent gilteritinib demonstrated improved CRc rates (CR + CR with partial hematologic recovery;
34.0% vs 15.3%; risk difference 18.6%; 95% CI, 9.8–27.4) and OS
(9.3 months vs 5.6 months; HR 0.64; 95% CI, 0.49–0.83; two-sided P < 0.001) compared with salvage chemotherapy. EFS was not

significantly different between the 2 arms: 2.8 months in the gilteritinib arm and 0.7 months in the chemotherapy arm (HR for treatment failure or death, 0.79; 95% CI, 0.58–1.09). Responses were observed on both gilteritinib 200 mg/day (n = 78) and
80 mg/day (n = 58) (CRc 32.1% and 55.2%, respectively). Median duration of CRc with gilteritinib was 11 months. Similar responses were seen in ITD- and TKD-mutated FLT3 (CR 20.5% vs 19%, respectively). The main side effects of gilteritinib include diarrhea, fatigue, and transaminitis [23,52]. A higher proportion of patients received an alloSCT in the gilteritinib arm compared to the chemotherapy arm (25.5% vs 15.3%). Quality of life was assessed as an exploratory objective but has not yet been reported. Loss of response was associated with acquisition of new mutations (27/40; 67.5%), involving genes in the Ras/MAPK pathway, FLT3 (including F691 L gatekeeper mutation), WTI, IDH1 and GATA2 [53,54].
This led to the FDA (2018), EMA (2019) and HC (2020) approval of gilteritinib for use as monotherapy in patients with relapsed/refractory FLT3-mutated AML, irrespective of the number of lines of prior therapy (based on evidence of efficacy in a prior phase 1/2 study) [55,56]. As there was no second randomization after alloSCT, the benefit conferred by gilteritinib post-alloSCT could not be determined. However, as continuation of gilteritinib therapy post- alloSCT was part of the overall treatment strategy, there is an option to reinitiate gilteritinib in patients after alloSCT [56].
An open-label, randomized, phase 3 study of gilteritinib, gilteritinib plus AZA or AZA alone in patients with untreated FLT3-mutated AML who are not suitable for intensive che- motherapy is actively enrolling patients (NCT02752035). Gilteritinib (80 or 120 mg per day) in combination with vene- toclax 400 mg/day is also being evaluated in patients with relapsed/refractory AML in a phase 1b trial [57], with active enrollment on the expansion phase with gilteritinib 120 mg/day.
Two phase 3 trials are evaluating gilteritinib monother- apy versus placebo as maintenance therapy in patients with FLT3-mutated AML in CR1 after induction and consolidation chemotherapy (NCT02927262) or alloSCT (NCT02997202).

⦁ Crenolanib (CP-868-596)
Crenolanib is a second-generation Type I FLT3 inhibitor with activity against PDGFRβ, FLT3-ITD and FLT3-TKD. At clinically achievable concentrations, it results in less c-Kit inhibition compared to quizartinib [58–60]. Preliminary results from a phase 2 study evaluated crenolanib administered with stan- dard anthracycline and cytarabine induction and cytarabine consolidation chemotherapy followed by crenolanib mainte- nance (after consolidation or alloSCT) in patients with newly diagnosed FLT3-mutated AML [61–63]. Forty-two patients have been treated. CR rates were 85% and 86%, respectively. Median 1-year OS was 75.4% and 67%, respectively. A rando- mized phase 3 study is comparing standard cytarabine and daunorubicin induction and cytarabine consolidation plus midostaurin or crenolanib in adults aged 18 to 60 years old with newly diagnosed TN FLT3-mutated AML (NCT03258931).
Two randomized, placebo-controlled, phase 3 trials have been initiated combining crenolanib and salvage che- motherapy (with HAM or FLAG-Ida) as first or second sal- vage in patients with relapsed/refractory FLT3-mutated AML (NCT03250338; NCT02298166).

⦁ Quizartinib (AC220)
Quizartinib, previously known as AC220, is a second genera- tion, Type II FLT3 inhibitor with activity also against c-Kit, PDGFR, RET, and CSF1 R (Box 1). The initial compound was synthesized and optimized as a selective FLT3 inhibitor for the treatment of AML [64–66]. Quizartinib demonstrated potent FLT3 inhibition in in vitro and cellular assays, in vivo tumor models, and primary AML cells [64].

⦁ Chemistry
Quizartinib is a benzoimidazothiazole and a member of mor- pholines, isoxazoles and phenylureas. The compound includes urea in which one of the amino groups is substituted by a 5- tert-butyl-1,2-oxazol-3-yl group while the other has been sub- stituted by a phenyl group substituted at the para – position by an imidazo[2,1-b][1,3]benzothiazol-2-yl group that, in turn, is substituted at position 7 by a 2-(morpholin-4-yl)ethoxy group [67]. Its molecular weight is 560.7 g/mol.

⦁ Pharmacodynamics
Quizartinib works by inhibiting FLT3, both the ITD mutant and WT proteins, but not TKD mutants [64,68]. A mutation in the FLT3 gene results in a constitutively active FLT3 RTK. This triggers downstream pathways, as previously described, that results in uncontrolled cell proliferation and inhibition of apoptosis [27,28]. By inhibiting this mutant receptor, proliferation pathways are blocked and apoptotic inhibition reverted. Furthermore, quizar- tinib induces blasts to undergo differentiation in vivo [69]. As a Type II inhibitor, quizartinib binds to the hydrophobic region adjacent to the ATP-binding site that is only accessible when the receptor is in its active conformation. Hence, quizartinib is not active against FLT3-TKD [68,70] (Figure 2).

⦁ Pharmacokinetics and metabolism
Healthy subjects receiving quizartinib at a dose of 60 mg demonstrated a rapid oral absorption of the drug into the circulation with maximum blood concentrations (Cmax) observed at 4h after administration [71]. Quizartinib has a long elimination half-life with a mean of 73 h and accumulates 5-fold after continuous daily dosing. Mean quizartinib phar- macokinetic profiles are similar under fasted and fed condi- tions in healthy subjects [72]. It is metabolized in the liver by cytochrome CYP3A4, warranting caution in patients with hepatic impairment and underscores possible drug-drug inter- actions. Up to 41 metabolites have been identified, with one major active metabolite (AC886) identified in urine and plasma samples [73]. Only 4% of quizartinib is excreted intact, with the remainder of the drug being eliminated as different meta- bolites. Excretion takes place primarily in the feces, with minor renal clearance [71]. Complete FLT3-ITD inhibition in a plasma inhibitory activity (PIA) assay was detected at 2 hours post
60 mg dose on day 1 and maintained throughout the cycle [65].
Quizartinib is converted quickly into AC886, which can be detected in plasma as soon as 15 minutes after administration [65]. AC886 has kinase selectivity and potency as FLT3 inhibi- tor, against both FLT3-ITD and WT FLT3, with half-maximal inhibitory concentration of 0.3 nmol/L [65,71,73]. Similar to quizartinib, AC886 has a very long half-life of 119 hours [65,74]. It is likely that AC886 enhances the pharmacological effects of quizartinib.
Since quizartinib is metabolized by CYP3A, there is a sig- nificant potential interaction with antifungals. When quizarti- nib is administered with a strong CYP3A inhibitor (e.g. ketoconazole), the dose should be reduced from 60 mg to 30 mg and from 30 mg to 20 mg due to the almost 2-fold increase in drug concentrations [75]. When administered with a moderate or weak CYP3A inhibitor (e.g. fluconazole), no dose adjustments are required. Quizartinib has pH-dependent solubility in vitro, but a clinical study showed minimal effects on quizartinib’s pharmacokinetics when co-administered with the proton pump inhibitor, lansoprazole [76].

⦁ Resistance mechanisms
As detailed previously, primary or acquired FLT3-TKD muta- tions in AML render the cell resistant to quizartinib [6,68]. Common mutations are the D835, Y842, and gatekeeper resi- due F691 [68,77,78]. Other mechanisms through which AML can acquire resistance to quizartinib are by: (a) upregulating compensatory signaling pathways that are independent of FLT3 inhibition (e.g. PI3 K/AKT/mTOR pathway); (b) imbalance between the pro-apoptotic BAD promoter and anti-apoptotic proteins BCL-XL, BCL2, and MCL1, leading to activation of antiapoptotic proteins [79]; (c) tumor (bone marrow) micro- environment-mediated resistance, with stromal cells prevent- ing FLT3 inhibition through a combination of soluble factor and direct cell-cell contact [80,81]; (d) upregulation of FLT3 ligand (which increases after chemotherapy) or FLT3 receptor [82]; and (e) concurrent mutations (e.g. CEBPα mutations
appear to confer resistance, as FLT3 inhibitor mediate differ- entiation requires the presence of a functional CEBPα) [69].

⦁ Clinical efficacy
A multicenter, phase 1, open-label, dose escalation trial eval- uated different dose/schedules of quizartinib in 76 patients with relapsed/refractory AML, regardless of FLT3-ITD status [65]. Fifty-one patients received quizartinib on an intermittent schedule (12–450 mg) and 25 on a continuous schedule (200 or 300 mg). MTD was 200 mg on the continuous schedule, with the DLT being grade 3 QTc prolongation. No MTD was found on the intermittent dosing schedule. Responses were observed at doses as low as 40 mg/day. Responses were higher in FLT3-ITD-mutated patients compared to FLT3-ITD negative patients (52.9% vs 13.5%, respectively). The 200 mg dosing was chosen for further clinical trial testing.
The initial phase 2 trial tested quizartinib in 2 different AML cohorts, irrespective of FLT3 status: (a) older (aged ≥ 60 years) SCT ineligible patients (n = 157) and (b) younger (< 60 years) patients (n = 176) [83] (Table 2). After 17 patients were treated on the starting dose of 200 mg/day, the protocol was amended to decrease the starting dose to 135 mg/day for men and 90 mg/day for women due to QTc prolongation. Grade 3 AEs occurred in 5% of the cohort (mainly myelosup- pression and QTc prolongation), with 32% requiring dose reductions. One patient had torsade de pointes and 2 experi- enced hepatic failure. Due to these AEs, lower doses of qui- zartinib were chosen for a subsequent phase 2b trial (30 mg and 60 mg) [84]. CRc was similar between the 2 cohorts and was 56 and 46% in the FLT3-ITD-mutated patients, respec- tively. Median OS was also similar at 25.4 and 24 weeks, respectively.
Similar responses were observed in a randomized phase 2b trial assessing 2 (lower) dosing regimens of quizartinib mono- therapy (30 or 60 mg/day with escalation to 60 or 90 mg/day, respectively) in 76 patients with relapsed/refractory FLT3-ITD- mutated AML [84](Table 2). Median age was 55 years (range, 19–77). Dose escalation occurred in 61% and 14% of the 30 mg and 60 mg groups, respectively. CRc was 47.4% for both groups. Responses were seen rapidly after 1 cycle of quizartinib in 47% of patients in the 60 mg dose cohort. OS and number of patients who received a SCT was higher in the 60 mg dose group. Side effects were similar in the 2 cohorts except for more QTc prolongation at the higher dose.
The phase 3, multicentre, international QuANTUM-R trial randomized 367 patients with relapsed (n = 246) or refractory (n = 121) FLT3-ITD-mutated AML in a 2:1 fashion to either quizartinib (n = 245) daily or investigator’s choice chemother- apy (LDAC (n = 29), MEC (n = 40), or FLAG-IDA (n = 53)) as first salvage [85](Table 2). Twenty-eight patients (23%) did not receive the intended chemotherapy treatment. Quizartinib was prescribed with a lead in dose of 30 mg, and increased to 60 mg if the QTc was ≤450 ms at day 16 of cycle 1. Median age was 55 years (range, 46–65) in the quizartinib arm and
57.5 years (range, 44–66) in the chemotherapy arm. The major- ity of patients had an intermediate-risk cytogenetics. Eighty- nine (24%) patients had received a prior alloSCT. Median

7

Table 2. Clinical outcomes of quizartinib.

Trial

Phase

Population

Study design and treatment

N
Median age (years)

CRc/ORR (%) Median response duration (weeks)

Median OS

No. bridged to SCT

Notes
Cortes et al, 2018 2 Relapsed/refractory primary Single arm; 333; 69 (cohort Cohort 1 = 56% (63/ Cohort Cohort 1 = 25.4 Cohort Results focused on
AML or secondary to MDS Q 135 mg (men) or 90 mg (women) Cohort 1) 112)/77% (86/112) 1 = 12.1 w 2 = 34.5% FLT3-ITD
regardless of FLT3-ITD daily in 28 day cycles 1 = 157 51 (cohort Cohort 2 = 46% (62/ Cohort Cohort 2 = 24 w (47/136) population
status (FLT3- 2) 136)/74% (101/ 2 = 10.6
-Cohort 1: ≥60 years, no ITD = 112); 136)
HSCT, <1 year after any Cohort
treatment 2 = 176
-Cohort 2: ≥18 years and (FLT3-ITD
<85, no HSCT in last 136)
100 days
Cortes et al, 2018 2b Relapsed/refractory FLT3-ITD Randomized; 76; 55 30 mg = 47.4% (18/ 30 mg = 4.2 30 mg = 20.9 w 30 mg = 31.6% Allowed dose
mutated primary AML or 2 dosing cohorts: 30 mg = 38 38)/60.5% (23/38) 60 mg = 9.1 60 mg = 27.3 w (12/38) escalation for lack
secondary to MDS in Q 30 mg daily or 60 mg daily in 28 60 mg = 38 60 mg = 47.4% (18/ 60 mg = 45.1% of response
adults ≥18 years day cycles 38)/71.1% (27/38) (16/38)
Takahashi et al, 2 Relapsed/refractory FLT3-ITD Single arm; 37 65 53.8% (14/26)/77.8% 16.1 34.1 w 37.5% (12/32) Interim analysis
2019 mutated primary AML or Q 30 mg daily (increased to 60 mg on (21/27) report; allowed
secondary to MDS in day 16 if no QTc prolongation) in dose escalation
adults ≥20 years 28 day cycles for lack of
response
Cortes et al, 2019 3 Relapsed/refractory FLT3-ITD Randomized, 2:1; 367; 55 (Q) 48% (118/245) vs 12.1 vs 5 6.2mo vs 4.7mo 32% (78/245) vs
QuANTUM-R mutated primary AML or Q 30 mg daily (increased to 60 mg on Q = 245 57.5 (CT) 27% (33/122)/ 11% (14/122)
secondary to MDS in day 16 if no QTc prolongation) in CT = 122 69.4% (170/245)
adults ≥18 years 28 day cycles vs CT (FLAG-IDA, vs 30.3% (37/122)
MEC, or LDAC)
EXPERT OPINION ON PHARMACOTHERAPY
Note: CRc – composite complete response (CR + CR with incomplete platelet recovery + CR with incomplete hematological recovery); ORR – Overall response rate (CRc + partial response); OS – overall survival; Q – quizartinib; SCT – stem cell transplant; CT – chemotherapy; w – weeks; mo – months

number of cycles of quizartinib was 4 (range, 2–6). Quizartinib was associated with a 24% reduction in the risk of death compared with chemotherapy (HR 0.76; 95%CI 0.58–0.98; P = 0.02). Median OS was 6.2 months with quizartinib com- pared to 4.7 months with chemotherapy with 1-year OS of 27 and 20%, respectively. EFS was 1.4 months for quizartinib arm and 0.9 months for chemotherapy arm (HR 0.90; 95% CI 0.70–
1.6; P = 0.11). CRc was 48.2% in the quizartinib arm (CR 4.1%; CRp 3.7%; CRi 40.4%) vs 27% in the chemotherapy arm (CR 0.8%; CRi 26.2%). Median duration of CRc was 12.1 weeks in the quizartinib arm and 5 weeks in the chemotherapy arm. Thirty-two percent of patients in the quizartinib arm received a SCT (with 62% resuming quizartinib after SCT) compared to 11% in the chemotherapy arm. The trial was not powered to look at outcomes after SCT or quizartinib maintenance. Quality of life was not evaluated; however, a post hoc quality-adjusted time without symptoms or toxicity (Q-TWiST) analysis revealed a significantly prolonged quality-adjusted survival with quizar- tinib compared to chemotherapy [86]. Cause(s) of lack of and loss of response have not yet been reported. A subsequent phase 2 trial conducted in Japan in 37 patients with relapsed/ refractory FLT3-ITD-mutated AML yielded similar findings as in the QuANTUM-R trial [87] (Table 2).
A phase 1 study evaluating 3 different dose/schedules of quizartinib (60 mg/day for 7 days, 60 mg/day for 14 days and
40 mg/day for 14 days) administered in conjunction with standard induction and consolidation chemotherapy followed by single agent quizartinib maintenance or SCT was per- formed in 19 patients with treatment-naïve AML, irrespective of FLT3-ITD status [88]. Median age 43 years (range, 22–60). Nine patients (47%) were FLT3-ITD-mutated. Ten patients com- pleted induction and consolidation. Three patients experi- enced DLTs: 2 with quizartinib 60 mg/day for 14 days (1 pericardial effusion; 1 febrile neutropenia, thrombocytopenia and QTc prolongation) and 1 with 40 mg/day for 14 days (1 pericarditis). CRc rate was 74% (CR 47%; CRp 11%; CRi 16%) with CRc of 67% in FLT3-ITD-mutated patients. RP2D of qui- zartinib was 40 mg/day for 14 days. Based on these results, a placebo-controlled, randomized phase 3 trial comparing stan- dard induction and consolidation chemotherapy plus quizarti- nib or placebo followed by maintenance in patients with treatment-naïve FLT3-ITD-mutated AML (QuANTUM-First) (NCT02668653) was conducted and has completed enroll- ment; however, results are not mature enough to report. The primary endpoint is EFS.
Combination therapy with quizartinib is also being evalu- ated with a number of lower intensity agents, as well as intensive chemotherapy (Table 3). Preliminary results of a phase 1/2 trial evaluating quizartinib in combination with either AZA (n = 38) or LDAC (n = 23) in 61 patients (59 evaluable) with relapsed/refractory (n = 47) or TN (n = 12) AML, MDS or chronic myelomonocytic leukemia (CMML), irre- spective of FLT3-ITD status, have been reported [89,90]. Fifty- five patients (90%) had FLT3-ITD mutations. Eight patients had received prior FLT3 inhibitor (6 with sorafenib, 1 crenolanib, and 1 quizartinib). ORR was 73% (CR 16.9%; CRp 10.2%; CRi 33.9%). Responses were seen in the LDAC (67%) and AZA (76%) arms. ORR was 75% in the FLT3-ITD-mutated patients.
ORR was 92% in the TN arm (CR 50%; CRi 16.7%; CRp 16.7%)
and 68% in the relapsed/refractory arm (CR 8.5%; CRi 40.4%; CRp 8.5%). Median OS was 18.6 months and 11.2 months, respectively.
Early results from a phase 2 study assessing quizartinib with omacetaxine (homoharringtonine) in 29 patients with relapsed/ refractory (n = 22) or TN (n = 7) FLT3-ITD-mutated AML yielded CRc rates of 81% (CR 7%; CRi 74%) (NCT03135054) [91]. All 6
patients who had received prior FLT3 inhibitor (3 each with prior sorafenib or midostaurin) achieved a CR or CRi. Median OS in responders was 11 months.
Quizartinib has shown safety and feasibility in the post-SCT maintenance setting in 13 patients with FLT3-ITD-mutated AML [92]. One DLT was observed in each of the 2 dose levels (40 and 60 mg/day) and consisted of grade 3 gastric hemor- rhage and grade 3 anemia, respectively. The most common adverse events were hematological. MTD was not identified; however, based on data from prior studies, the RP2D was determined to be 60 mg/day.

⦁ Safety and tolerability
Based on the clinical experience with single agent quizartinib, AEs have been reported in over 750 patients with AML; those observed in the phase 2 and 3 trials are summarized in Table 4 [65,74,83–85,87]. The most common AEs were gastrointestinal (abdominal pain, nausea, vomiting, diarrhea), myelosuppression (anemia, neutropenia, febrile neutropenia, and thrombocytope- nia), infections (febrile neutropenia and bacterial infections), constitutional (pyrexia, fatigue, and asthenia) and bleeding and bruising (epistaxis, hematoma, and hemorrhage).
Events of special interest included QTc prolongation (including 1–2% fatal cardiac events) and differentiation syn- drome or acute febrile neutrophil dermatosis (7%) in the QuANTUM-R study [85]. Quizartinib is associated with QTc prolongation (via blockade of the slow delayed rectifier potas- sium current, IKs) and is dose-dependent manner. The inci- dence of grade 3 QTc prolongation (QTc > 500 ms) was 4% in the QuANTUM-R study [85], 15–17% among subjects receiving 90 to 135 mg daily [83] and 28–35% among subjects receiving
200 to 300 mg daily [65,83]. The current ongoing studies being conducted in adults are using doses that range from 20 to 60 mg daily.
A pooled safety analysis of 673 patients on the North American and European trials showed a 26% discontinuation rate due to AEs [93]. The most frequent grade ≥ 3 AEs were febrile neutropenia and myelosuppression. The most common clinical response to quizartinib is CRc, with CR rates being low, in part due to cytopenias which arise from inhibition of c-Kit by quizartinib. c-Kit is present in hematopoietic stem cells and is important for normal hematopoiesis [94].

⦁ Regulatory issues
Based on the results from the QuANTUM-R study, the Ministry of Health, Labor and Welfare of Japan approved quizartinib for use in adults with relapsed/refractory FLT3- ITD-mutated AML in June 2019 [95] whereas, the FDA and

Table 3. Ongoing phase 1/2 trials evaluating quizartinib in patients with AML (not acute promyelocytic leukemia).
Trial Phase Age (y) N Population Treatment Regimen Primary Outcome(s)

Untreated

NCT03723681 1 18–70 18 ● Untreated de novo AML or secondary AML (prior
myelodysplastic syndrome or myeloproliferative
neoplasm)
⦁ Does not need to have FLT3 mutation
⦁ Chinese descent
Quizartinib (20 mg or 40 mg) daily + standard induction and consolidation ● Dose-limiting toxicities
⦁ Adverse events
⦁ Pharmacokinetics

NCT03135054 2 ≥ 18 40 ⦁ Relapsed/Refractory FLT3-ITD-mutated AML or Quizartinib 30 mg/day D1-28 + Omacetaxine (homoharringtonine) 1.5 mg/m2/day ⦁ Complete remission (CR)
⦁ Untreated FLT3-ITD-mutated AML (max 3 mg) D1-7 q28 days
NCT03552029 1 ≥ 18 156 ⦁ Relapsed/Refractory FLT3-ITD-mutated AML (Part 1 & Quizartinib + Milademetan (mdm2 inhibitor) ⦁ Dose-limiting toxicities
Part 1 Cohort 1) ⦁ Adverse events
⦁ Untreated FLT3-ITD-mutated AML (Part 2 Cohort 2) ⦁ Response to therapy
NCT03661307 1/2 ≥ 18 52 ⦁ Relapsed/Refractory FLT3-ITD-mutated AML or MDS Quizartinib D1-28 + Decitabine D1-10 + Venetoclax D1-14 (or D1-21 if persistent ⦁ Maximum tolerated dose
(>10% blasts) or leukemia) q28 days ⦁ Adverse events
⦁ Untreated FLT3-ITD-mutated AML or MDS (>10% ⦁ Overall response rateb

Untreated or Relapsed/Refractory

blasts) ineligible for intensive chemotherapy (age ≥ 60 y)
⦁ NCT04128748 ⦁ 1/2 ⦁ ≥ 18 ⦁ 52 ⦁ Relapsed/Refractory AML or MDS (>10% blasts) or
⦁ Untreated AML or MDS (>10% blasts) ineligible for intensive chemotherapy (age ≥ 60 y)
⦁ Does not need to have FLT3 mutation ⦁ Induction (cycle 1): Quizartinib D6-19 + CPX-351 D1, 3 and 5
⦁ Re-induction (if no response; cycle 2): Quizartinib D6-19 + CPX-351 D1 and 3 Consolidation (up to 2 cycles): Quizartinib D4-28 + CPX-351 D1 and 3 q28 days
⦁ Maintenance: Quizartinib D1-28 q28 days ⦁ Maximum tolerated dose
⦁ Composite CR ratea
⦁ NCT04047641 ⦁ 2 ⦁ ≥ 18 ⦁ 86 ⦁ Relapsed/Refractory AML, MPAL or MDS (>10% blasts) ⦁ Induction (up to 2 cycles): Quizartinib D6-19 + Idarubicin D1-3 + Cladribine D1-5 ⦁ Event-free survival
⦁ ⦁ or ⦁ + Cytarabine D1-5 (or D1-3 if age > 60 y) q28 days ⦁ Adverse events
⦁ Untreated AML, MPAL or MDS (>10% blasts) ineligible ⦁ Consolidation (up to 5 cycles): Quizartinib D4-28 + Idarubicin D1&2 + Cladribine D1-3 ⦁
⦁ for intensive chemotherapy (age 18–65 y) ⦁ + Cytarabine D1-3 q28 days ⦁

⦁ No prior quizartinib

Relapsed/Refractory
⦁ Does not need to have FLT3 mutation
Maintenance (up to 12 cylces): Quizartinib D1-28 q28 days

NCT03735875 1b/2 ≥ 18 32 ● Relapsed/Refractory FLT3-ITD-mutated AML
⦁ No prior quizartinib
Quizartinib D1-28 + Venetoclax D1-28 (D8-28 on cycle 1 only) q28 days (max 24 cycles)
⦁ Maximum tolerated dose
⦁ EXPERT OPINION ON PHARMACOTHERAPY
⦁ Recommended phase II dose
⦁ Composite CR ratea

NCT03793478 1/2 1 mo – 21 y 65 ● Relapsed/Refractory FLT3-ITD-mutated AML Quizartinib D6-28 + Fludarabine 30 mg/m2 IV D1-5 + Cytarabine 2 g/m2/day IV D1-5
or 500 mg/m2/day CIV D1-5 + Etoposide 100 mg/m2 IV D1-5 + IT chemotherapy
Note: CIV – continuous intravenous infusion; D – day; IT – intrathecal; MPAL – mixed phenotypic acute leukemia (or acute biphenotypic leukemia); y – year
aComposite CR (CRc) = CR + CR with incomplete platelet recovery (CRp) + CR with incomplete count recovery (CRi);
bOverall response rate (ORR) = CR + CRp + CRi + partial remission (PR);
9
cComposite CR (CRc) = CR + CR with incomplete count recovery (CRi)
⦁ Composite CR ratec

Table 4. Frequent adverse events of quizartinib in phase 2 or 3 trials.

Trial
N Grade ≥3 (≥10%)
Major (any grade) side effects
QTc grade ≥3
Cortes et al, 2018 333 Febrile neutropenia 41% Nausea 54% 11% (35/333)
Anemia 26% Diarrhea 41% (34 grade 3; 1 grade 4)
Thrombocytopenia 15% Vomiting 39%
Pneumonia 13% QTc prolongation 30%
Neutropenia 11% Peripheral edema 27%
Hypokalemia 19%
Cortes et al, 2018 76; 30 mg: 30 mg: 30 mg:
30 mg = 38 Febrile neutropenia 31.6% QTc prolongation 52.7% 5.3% (2/38)
60 mg = 38 Anemia 36.6% NR¶ 60 mg:
Thrombocytopenia 26.3% 60 mg: 2.8% (1/36)
Pyrexia 10.5% QTc prolongation 63.9% *All events grade 3
60 mg: NR¶
Febrile neutropenia 36.1%
Thrombocytopenia 19.4%
Pneumonia 16.7%
Anemia 16.7%
Neutropenia 13.9%
Elevated ALT 11.1%
Elevated bilirubin 11.1%
Takahashi et al, 2019 37 Febrile neutropenia 37.8% Febrile neutropenia 43.2% 0
Decrease in platelets 29.7% QTc prolongation 35.1%
Anemia 24.7% Nausea 29.7%
Neutropenia 21.6% Vomiting 16.2%
Thrombocytopenia 10.8% Hypokalemia 10.8%
Diarrhea 10.8%
Elevated ALT 10.8%
Cortes et al, 2019 367; Thrombocytopenia 35% Nausea 48% 3.3% (8/241)
QuANTUM-R Quizartinib arm = 241 Neutropenia 32% Fever 38% *All events grade 3
Febrile neutropenia 30% Febrile neutropenia 33%
Anemia 30% Vomiting 33%
Sepsis/shock 19% Hypokalemia 33%
Pneumonia 12% Diarrhea 29%
Hypokalemia 12% QTc prolongation 26%
Peripheral edema 21%
Elevated ALT 14%
Note: NR – not reported Safety population results reported. The main treatment-related adverse event per patient was reported per arm but not all adverse events that the patient could have presented.

EMA rejected the application due to the lack of a significant benefit – to-risk ratio, safety concerns and concerns with credibility and generalizability of the trial data (110, 111). The issues included: (a) lack of corroborating benefit observed in EFS in CR rates between the 2 arms; (b) imbal- ance between the proportion of patients not treated in each arm (23% of patients randomized to the SOC chemotherapy arm were not treated compared to 2% in the quizartinib arm); (c) imbalance between the 2 arms as to proportion censored for the OS endpoint prior to week 8 (7% in the SOC chemotherapy arm compared to 0.4% in the quizartinib arm); (d) subgroup analysis for OS stratified by physician specified low intensity vs high intensity chemotherapy revealed a HR of 0.59 (95% CI, 0.36–0.97) in favor of qui- zartinib for the low intensity group compared with a HR of
0.83 (95% CI, 0.62–1.11) for the high intensity group, such that the overall improvement in OS may be driven by the low intensity group; and (e) difference in post-study treat- ment interventions between the 2 arms (23% of patients in the quizartinib arm received an alloSCT compared to 0% in the SOC chemotherapy arm in the low intensity stratum). Although quizartinib had an acceptable safety profile, car- diac issues, specifically QTc prolongation, was noted as an event of special interest. However, the risk of cardiac toxi- city could be mitigated with a black box warning and education of healthcare providers.

⦁ Conclusion
Quizartinib is a highly selective, well-tolerated, potent FLT3-ITD inhibitor. It has good oral absorption and bioavailability, making it convenient for administration. In the relapsed/refractory FLT3- mutated AML setting, quizartinib demonstrated an improvement in OS and good CRc rates allowing patients to undergo SCT, albeit with some degree of toxicity. Anxieties with QTc prolongation can be mitigated by careful selection of patients, similar to choosing the appropriate BCR-ABL TKI in patients with chronic myeloid leukemia, based on judicious use of concomitant medications and avoidance in patients with cardiac history. However, it is not effective in patients with FLT3-TKD-mutated AML and leukemic cells can develop resistance to quizartinib through diverse mechanisms (section 3.4), including acquisition of secondary FLT3-TKD mutations; thus, limiting its activity. Due to a number of concerns (described in section 3.7), quizartinib did not receive FDA or EMA approval as monotherapy in the relapsed/refractory AML setting. Despite this, quizartinib remains a promising thera- peutic option in the upfront and in the relapsed/refractory setting in combination with other agents and as maintenance therapy.

⦁ Expert opinion
Although the phase 3 QuANTUM-R trial met its primary outcome and showed improvement in OS on an intention-to-treat analysis

⦁ , there were significant concerns with the study (as detailed in section 3.7). In the phase 3 ADMIRAL trial using gilteritinib [51], higher CR rates (21.1%) and median OS (9.3 months) were obtained with gilteritinib compared those obtained on the QuANTUM-R study (CR 4.1%; OS 6.2 months). However, the median duration of CRc with gilteritinib appears comparable to that with quizartinib (11 months vs 12.1 months). In the ADMIRAL study only 12% of patients assigned to salvage chemotherapy were not treated compared to 23% in the QuANTUM-R study. Caution should always be used when comparing two trials as their populations are usually not the same. A key distinction between the two studies, is that a potentially higher risk popula- tion was enrolled in the QuANTUM-R study, where eligible patients had to be primary refractory or relapsed with CR dura- tion ≤ 6 months; the relapsed patients consisted of 60.6% and 67% of the patients enrolled onto the ADMIRAL and QuANTUM-R studies, respectively. Another significant difference between gil- teritinib and quizartinib is the safety profile, with a higher inci- dence/risk of QTc prolongation and myelosuppression observed with quizartinib, which would have impacted on chose of drug, if quizartinib had been approved. Also, it is unclear how effective quizartinib or any of the other FLT3 inhibitors are in patients who have received prior FLT3 inhibitors, as the current phase 3 studies were underway prior to the approval of midostaurin.
Quizartinib may obtain FDA approval in the front-line set- ting depending on the results of the QuANTUM-First study. The use of a placebo-control arm in the QuANTUM-First trial will prevent some of the issues that arose in the QuANTUM-R study where 23% of patients on the SOC chemotherapy were not treated. Combination therapies using quizartinib are being investigated as a strategy to overcome or prevent the devel- opment of resistance and to improve outcomes (Table 3). Other agents to consider in combination with quizartinib are inhibitors against the PI3 k/Akt/mTOR and RAF/Ras/MEK path- ways. Furthermore, its activity in WT FLT3 AML and ability to induce differentiation makes it a candidate for clinical trials in a non-FLT3-ITD-mutated patient population [88].

Funding
This manuscript has not been funded.

Declaration of interest
KWL Yee reports advisory board honorarium from Astellas. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

ORCID
Alejandro Garcia-Horton http://orcid.org/0000-0003-0579-6709
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
⦁ Estey E, Döhner H. Acute myeloid leukaemia. Lancet. 2006;368 (9550):1894–1907.
⦁ SEER NCI-. Cancer stat facts: leukemia – acute myeloid leukemia (AML). [cited 2020 Jan 9]. Available from: ⦁ https://seer.cancer.gov/ ⦁ statfacts/html/amyl.html.
⦁ Schlenk RF, Döhner K, Krauter J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358(18):1909–1918.
⦁ The Cancer Genome Atlas Research Network. Genomic and epige- nomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–2074.
⦁ Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classifica- tion and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209–2221.
⦁ Smith CC. The growing landscape of FLT3 inhibition in AML. Hematology. 2019;2019(1):539–547.
⦁ Adolfsson J, Månsson R, Buza-Vidas N, et al. Identification of Flt3+ lympho-myeloid stem cells lacking Erythro-Megakaryocytic poten- tial: a revised road map for adult blood lineage commitment. Cell. 2005;121(2):295–306.
⦁ Pemmaraju N, Kantarjian H, Ravandi F, et al. FLT3 inhibitors in the treatment of acute myeloid leukemia. Cancer. 2011;117(15):3293–3304.
⦁ Perl AE. Availability of FLT3 inhibitors: how do we use them? Blood. 2019;134(9):741–745.
⦁ Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leuke- mia (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(6):1752–1759.
⦁ 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(23):3441–3449.
⦁ Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correla- tion 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(1):59–66.
⦁ Linch DC, Hills RK, Burnett AK, et al. Impact of FLT3(ITD) mutant allele level on relapse risk in intermediate-risk acute myeloid leu- kemia. Blood. 2014;124(2):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 out- come in patients with intermediate-risk karyotype acute myeloid leukemia. Cancer. 2012;118(24):6110–6117.
⦁ 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(9):3724–3726.
⦁ Liu SB, Dong HJ, Bao XB, et al. Impact of FLT3-ITD length on prognosis of acute myeloid leukemia. Haematologica. 2019;104(1): e9–e12.
⦁ 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(12):2386–2392.
⦁ Dohner K, Thiede C, Jahn N, et al. Impact of NPM1/FLT3-ITD geno- types defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood. 2020;135(5):371–380.
⦁ 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(7):2393–2398.
⦁ Ding L, Ley TJ, Larson DE, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–510.

⦁ Nazha A, Cortes J, Faderl S, et al. Activating internal tandem duplication mutations of the fms-like tyrosine kinase-3 (FLT3-ITD) at complete response and relapse in patients with acute myeloid leukemia. Haematologica. 2012;97(8):1242–1245.
⦁ Ottone T, Zaza S, Divona M, et al. Identification of emerging FLT3 ITD-positive clones during clinical remission and kinetics of disease relapse in acute myeloid leukaemia with mutated nucleophosmin. Br J Haematol. 2013;161(4):533–540.
⦁ Daver N, Schlenk RF, Russell NH, et al. Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. 2019;33(2):299–312.
⦁ Griffith J, Black J, Faerman C, et al. The structural basis for Autoinhibition of FLT3 by the Juxtamembrane domain. Mol Cell. 2004;13(2):169–178.
⦁ Zorn JA, Wang Q, Fujimura E, et al. Crystal structure of the FLT3 Kinase domain bound to the inhibitor Quizartinib (AC220). Plos One. 2015;10(4):e0121177.
⦁ Verstraete K, Vandriessche G, Januar M, et al. Structural insights into the extracellular assembly of the hematopoietic Flt3 signaling complex. Blood. 2011;118(1):60–68.
⦁ Hayakawa F, Towatari M, Kiyoi H, et al. Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines. Oncogene. 2000;19(5):624–631.
⦁ Mizuki M, Fenski R, Halfter H, et al. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood. 2000;96 (12):3907–3914.
⦁ Parcells BW, Ikeda AK, Simms-Waldrip T, et al. 3 in normal hema- topoiesis and acute myeloid leukemia. Stem Cells. 2006;24(5):1174– 1184.
⦁ Wilhelm S, Carter C, Lynch M, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5(10):835–844.
⦁ Kane RC, Farrell AT, Saber H, et al. Sorafenib for the treatment of advanced renal cell carcinoma. Clin Cancer Res. 2006;12(24):7271– 7278.
⦁ Kane RC, Farrell AT, Madabushi R, et al. Sorafenib for the treatment of unresectable hepatocellular carcinoma. Oncologist. 2009;14 (1):95–100.
⦁ Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014;384(9940):319–328.
⦁ Delmonte J, Kantarjian HM, Andreeff M, et al. Update of a phase I study of sorafenib in patients with refractory/relapsed acute mye- loid leukemia or high-risk myelodysplastic syndrome. Blood. 2007;110(11):893.
⦁ Metzelder S, Wang Y, Wollmer E, et al. Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation. Blood. 2009;113(26):6567–6571.
⦁ Metzelder SK, Schroeder T, Lubbert M, et al. Long-term survival of sorafenib-treated FLT3-ITD-positive acute myeloid leukaemia patients relapsing after allogeneic stem cell transplantation. Eur J Cancer. 2017;86:233–239.
⦁ 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 multi- centre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16 (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(25):3110–3118.
⦁ Ohanian M, Garcia-Manero G, Levis M, et al. Sorafenib combined with 5-azacytidine in older patients with untreated FLT3-ITD mutated acute myeloid leukemia. Am J Hematol. 2018;93 (9):1136–1141.
⦁ Ravandi F, Alattar ML, Grunwald MR, et al. Phase 2 study of azacy- tidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation. Blood. 2013;121 (23):4655–4662.
⦁ 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-con- trolled multicentre Sormain trial. Blood. 2018;132(Supplement 1):661.
⦁ Levis MJ, Chen YB, Hamadani M, et al. FLT3 inhibitor maintenance after Allogeneic transplantation: is a placebo-controlled, rando- mized trial ethical? J Clin Oncol. 2019;37(19):1604–1607.
⦁ Fabbro D, Buchdunger E, Wood J, et al. Inhibitors of protein kinases: CGP 41251, a protein kinase inhibitor with potential as an anticancer agent. Pharmacol Ther. 1999;82(2–3):293–301.
⦁ 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(5):433–443.
⦁ Stone RM, DeAngelo DJ, Klimek V, et al. Patients with acute mye- loid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood. 2005;105(1):54–60.
⦁ 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(28):4339–4345.
⦁ Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus che- motherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–464.
⦁ RYDAPT. (Midostaurin) prescribing information. [cited 2020 Jan 17]. Available from: ⦁ https://www.accessdata.fda.gov/drugsatfda_docs/ ⦁ label/2017/207997s000lbl.pdf.
⦁ Larson RA, Mandrekar SJ, Sanford BL, et al. An analysis of main- tenance therapy and post-midostaurin outcomes in the interna- tional prospective randomized, placebo-controlled, double-blind trial (CALGB 10603/RATIFY [Alliance]) for newly diagnosed acute myeloid leukemia (AML) patients with FLT3 mutations. Blood. 2017;130:145.
⦁ Lee LY, Hernandez D, Rajkhowa T, et al. Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood. 2017;129 (2):257–260.
⦁ Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381(18):1728–1740.
⦁ Phase 3 trial of gilteritinib inrelapsed/refractory FLT3 mutated
AML patients
⦁ XOSPATA. (Gilteritinib) prescribing information. [cited 2020 Jan 17]. Available from: ⦁ https://www.accessdata.fda.gov/drugsatfda_docs/ ⦁ label/2018/211349s000lbl.pdf.
⦁ Smith CC, Levis MJ, Perl AE, et al. Emerging mutations at relapse in patients with FLT3-mutated relapsed/refractory acute myeloid leu- kemia who received gilteritinib therapy in the phase 3 admiral trial. Blood. 2019;134:14.
⦁ McMahon CM, Ferng T, Canaani J, et al. Clonal selection with RAS pathway activation mediates secondary clinical resistance to selec- tive FLT3 inhibition in acute myeloid leukemia. Cancer Discov. 2019;9(8):1050–1063.
⦁ 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(8):1061–1075.
⦁ Tzogani K, Roshol H, Olsen HH, et al. The European Medicines Agency review of gilteritinib (Xospata) for the treatment of adult patients with relapsed or refractory acute myeloid leukemia with an FLT3 mutation. Oncologist. 2020;25(7):e1070–e1076.
⦁ Perl AE, Daver NG, Pratz KW, et al. Venetoclax in combination with gilteritinib in patients with relapsed/refractory acute meloid leuke- mia: a phase 1b study. Blood. 2019;134:3910.

⦁ Galanis A, Ma H, Rajkhowa T, et al. Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants. Blood. 2014;123(1):94–100.
⦁ Smith CC, Lasater EA, Lin KC, et al. Crenolanib is a selective type I pan-FLT3 inhibitor. Proc Natl Acad Sci U S A. 2014;111(14):5319– 5324.
⦁ Zhang H, Savage S, Schultz AR, et al. Clinical resistance to crenola- nib in acute myeloid leukemia due to diverse molecular mechan- isms. Nat Commun. 2019;10(1):244.
⦁ Wang ES, Griffiths EA, Walter RB, et al. Tolerability and efficacy of crenolanib and cytarabine/anthracycline chemotherapy in older patients (aged 61 to 75) with newly diagnosed FLT3-mutated acute myeloid leukemia (AML). Blood. 2019;134:3829.
⦁ Goldberg AD, Coombs CC, Wang ES, et al. Younger patients with newly diagnosed FLT3-mutant AML treated with crenolanib plus chemotherapy achieve adequate free crenolanib levels and durable remissions. Blood. 2019;134:1326.
⦁ Oran B, O. CS, Marin D, et al. Safety analysis of intra-patient dose – study of crenolanib maintenance therapy in patients with FLT3 mutant AML following allogeneic hematopoietic stem cell trans- plant. Blood. 2018;132:3426.
⦁ 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(14):2984–2992.
⦁ First report of quizartinib’s activity, development and pharma-
cokinetic/pharmacodynamic properties
⦁ 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–inter- nal tandem duplication status. J Clin Oncol. 2013;31(29):3681–3687.
⦁ Phase 1 trial of quizartinib discussing safety and outcomes in
relapsed/refractory AML
⦁ Chao Q, Sprankle KG, Grotzfeld RM, et al. Identification of N-(5-tert- Butyl-isoxazol-3-yl)-N′-{4-[7-(2-morpholin-4-yl-ethoxy)imidazo[2,1- b][1,3]benzothiazol-2-yl]phenyl}urea dihydrochloride (AC220), a
uniquely potent, selective, and efficacious FMS-like Tyrosine Kinase-3 (FLT3) inhibitor. J Med Chem. 2009;52(23):7808–7816.
⦁ National Center for Biotechnology Information. PubChem data- base. Quizartinib. n.d. [2020 Jan 9]. Available from: ⦁ https://pub ⦁ chem.ncbi.nlm.nih.gov/compound/Quizartinib.
⦁ Smith CC, Lin K, Stecula A, et al. FLT3 D835 mutations confer differential resistance to type II FLT3 inhibitors. Leukemia. 2015;29 (12):2390–2392.
⦁ 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(20):4205–4214.
⦁ Ke YY, Singh VK, Coumar MS, et al. Homology modeling of DFG-in FMS-like tyrosine kinase 3 (FLT3) and structure-based virtual screening for inhibitor identification. Sci Rep. 2015;5:11702.
⦁ Sanga M, James J, Marini J, et al. An open-label, single-dose, phase 1 study of the absorption, metabolism and excretion of quizartinib, a highly selective and potent FLT3 tyrosine kinase inhibitor, in healthy male subjects, for the treatment of acute myeloid leuke- mia. Xenobiotica. 2017;47(10):856–869.
⦁ Li J, Holmes M, Kankam M, et al. Effect of food on the pharmacoki- netics of Quizartinib. Clin Pharmacol Drug Dev. 2020;9(2):277–286.
⦁ James J, Pratz K, Stine A, et al. Clinical pharmacokinetics and FLT3 phosphorylation of AC220, a highly potent and selective inhibitor of FLT3. Blood. 2008;112(11):2637.
⦁ Usuki K, Handa H, Choi I, et al. Safety and pharmacokinetics of quizartinib in Japanese patients with relapsed or refractory acute myeloid leukemia in a phase 1 study. Int J Hematol. 2019;110 (6):654–664.
⦁ Phase 1 trial for safety of quizartinib in the Japanese
population
⦁ Li J, Kankam M, Trone D, et al. Effects of CYP3A inhibitors on the pharmacokinetics of quizartinib, a potent and selective FLT3 inhi- bitor, and its active metabolite. Br J Clin Pharmacol. 2019;85 (9):2108–2117.
⦁ Li J, Trone D, Mendell J, et al. A drug–drug interaction study to assess the potential effect of acid-reducing agent, lansoprazole, on quizartinib pharmacokinetics. Cancer Chemother Pharmacol. 2019;84(4):799–807.
⦁ Smith CC, Wang Q, Chin C-S, et al. Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 2012;485(7397):260–263.
⦁ Smith CC, Zhang C, Lin KC, et al. Characterizing and overriding the structural mechanism of the Quizartinib-Resistant FLT3 “Gatekeeper” F691L mutation with PLX3397. Cancer Discov. 2015;5(6):668–679.
⦁ Naqvi K, Ravandi F. FLT3 inhibitor quizartinib (AC220). Leuk Lymphoma. 2019;60(8):1866–1876.
⦁ Yang X, Sexauer A, Levis M. Bone marrow stroma-mediated resis- tance to FLT3 inhibitors in FLT3-ITD AML is mediated by persistent activation of extracellular regulated kinase. Br J Haematol. 2014;164 (1):61–72.
⦁ 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(13):4696–4706.
⦁ Sato T, Yang X, Knapper S, et al. FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo. Blood. 2011;117(12):3286–3293.
⦁ Cortes J, Perl AE, Döhner 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(7):889–903.
⦁ Phase 2 trial of quizartinib in relapsed/refractory AML patients
⦁ 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(6):598–607.
⦁ Phase 2b trial focused on quizartinib monotherapy in
relapsed/refractory FLT3 mutated AML patients
⦁ Cortes JE, Khaled S, Martinelli G, et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019;20(7):984–997.
•• Phase 3 trial of quizartinib in relapsed/refractory FLT3 mutated
AML patients
⦁ Cortes JE, Ganguly S, Khaled SK, et al. Quality-adjusted time with- out symptoms or toxicity (Q-TWiST) analysis of quizartinib vs sal- vage chemotherapy in patients with relapsed/refractory (R/R) FLT3- ITD acute myeloid leukemia (AML). Blood. 2019;134:382.
⦁ Takahashi T, Usuki K, Matsue K, et al. Efficacy and safety of quizar- tinib in Japanese patients with FLT3-ITD positive relapsed or refrac- tory acute myeloid leukemia in an open-label, phase 2 study. Int J Hematol. 2019;110(6):665–674.
⦁ Phase 2 trial of quizartinib in relapsed/refractory FLT3 mutated
AML patients in Japan
⦁ 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(2):213–221.
⦁ Abdelall W, Kantarjian HM, Borthakur G, 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. 2016;128(22):1642.
⦁ 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 leuke- mias: interim report of a phase I/II trial. Blood. 2017;130 (Supplement 1):723.
⦁ Zhang C, Leung GMK, Tsui SP, et al. A phase II single-arm open- labeled study evaluating combination of quizartinib and omacetax- ine mepesuccinate (QUIZOM) in newly diagnosed or relapsed/ refractory AML carrying FlT3-ITD. Blood. 2019;134:3825.
⦁ Sandmaier BM, Khaled S, Oran B, et al. Results of a phase 1 study of quizartinib as maintenance therapy in subjects with acute myeloid leukemia in remission following allogeneic hematopoietic stem cell transplant. Am J Hematol. 2018;93(2):222–231.

⦁ Cortes JE, Ganguly S, Krämer A, et al. Pooled Safety analysis of Quizartinib monotherapy in patients with relapsed/ refractory (R/R) acute myeloid leukemia (AML). Blood. 2019;134(Supplement_1):1372.
⦁ Galanis A, Levis M. Inhibition of c-Kit by tyrosine kinase inhibitors. Haematologica. 2015;100(3):e77–e9.
⦁ Daiichi-Sankyo. Daiichi Sankyo’s VANFLYTA® receives approval in Japan for the treatment of relapsed/refractory FLT3-ITD AML 2019. [2020 Jan 20]. Available from: ⦁ https://www.daiichisankyo. ⦁ com/media_investors/media_relations/press_releases/detail/ ⦁ 007030.html.