FLT3 Mutations in Acute
Myeloid Leukemia: Key Concepts
and Emerging Controversies
Vanessa E. Kennedy
*
and Catherine C. Smith
Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA,
United States
The FLT3 receptor is overexpressed on the majority of acute myeloid leukemia (AML)
blasts. Mutations in FLT3 are the most common genetic alteration in AML, identied in
approximately one third of newly diagnosed patients. FLT3 internal tandem duplication
mutations (FLT3-ITD) are associated with increased relapse and inferior overall survival.
Multiple small molecule inhibitors of FLT3 signaling have been identied, two of which
(midostaurin and gilteritinib) are currently approved in the United States, and many more of
which are in clinical trials. Despite signicant advances, resistance to FLT3 inhibitors
through secondary FLT3 mutations, upregulation of parallel pathways, and extracellular
signaling remains an ongoing challenge. Novel therapeutic strategies to overcome
resistance, i ncluding combining FLT3 inhibitors with other antileukemic agents,
development of new FLT3 inhibitors, and FLT3-directed immunotherapy are in active
clinical development. Multiple questions regarding FLT3-mutated AML remain. In this
review, we highlight several of the current most intriguing controversies in the eld
including the role of FLT3 inhibitors in maintenance therapy, the role of hematopoietic
cell transplantation in FLT3-mutated AML, use of FLT3 inhibitors in FLT3 wild-type
disease, signicance of non-canonical FLT3 mutations, and nally, emerging concerns
regarding clonal evolution.
Keywords: Acute Myeloid Leukemia, FLT3 inhibitor, FLT3 resistance, FLT3 inhibitor maintenance, non-canonical
FLT3 mutation
FLT3 EPIDEMIOLOGY, BIOLOGY, AND PROGNOSTIC
ASSOCIATIONS
Acute Myeloid Leukemia (AML) is an aggressive hematologic malignancy characterized by a
heterogenous genetic landscape and complex clonal evolution (1). Fms-like tyrosine kinase 3
(FLT3), a member of the receptor tyrosine kinase family, is widely expressed in hematopoietic
progenitor cells and is overexpressed on the majority of AML blasts (2). Upon binding to the FLT3
ligand, FLT3 receptors activate and dimerize, leading to conformational change, cellular
proliferation, and inhibition of apoptosis and differentiation (3). Mutations in FLT3 are the most
common genomic alteration in AML, identied in approximately one-third of newly diagnosed
adult patients (4), and are common in pediatric AML as well (5).
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128801
Edited by:
Alessandro Isidori,
AORMN Hospital, Italy
Reviewed by:
Adolfo De La Fuente,
MD Anderson Cancer Center Madrid,
Spain
Hussein A. Abbas,
M D Anderson Cancer Center,
United States
*Correspondence:
Vanessa E. Kennedy
Specialty section:
This article was submitted to
Hematologic Malignancies,
a section of the journal
Frontiers in Oncology
Received: 01 October 2020
Accepted: 19 November 2020
Published: 23 December 2020
Citation:
Kennedy VE and Smith CC (2020)
FLT3 Mutations in Acute Myeloid
Leukemia: Key Concepts and
Emerging Controversies.
Front. Oncol. 10:612880.
doi: 10.3389/fonc.2020.612880
REVIEW
published: 23 December 2020
doi: 10.3389/fonc.2020.612880
FLT3 mutations can be subdivided into internal tandem
duplicates (ITD), present in approximately 25% of patients,
and point mutations in the tyrosine kinase domain (TKD),
present in approximately 5%. Both FLT3-ITD and FLT3-TKD
mutations are constitutively activating, leading to ligand-
independent FLT3 signaling and cellular proliferation (3).
FLT3-ITD AND FLT3-TKD
FLT3-ITD mutations a re in-frame duplications of variable
size, ranging from 3 to >1,000 nucleotides, and are located
within the receptors autoinhibitory juxatamembrane domain.
In wild type (WT) FLT3, the FLT3 juxtamembrane domain
inhibits receptor act ivation; the presence of ITDs disrupts t his
inhibitory effect, resulting in constitutive activation (6).
Clinically, FLT3-ITD muta ted AML is associated with higher
rates of relapse and inferior overall survival, although the full
prognostic im pact is affect ed both by mutant allele burden
and presence of co-existing mutations (7). High allele ratio
(AR; FLT3-ITD
high
), generally dened as a FLT3-ITD to FLT3-
WT ratio of 0.5, is associated with higher disease risk.
Low AR (FLT3-ITD
low
) is associated with favorable risk in
patients with a co-occurrent nucleophosmin 1 (NPM1)
mutations and intermediate risk in patients with NPM1-WT.
These associations are reected in the 2017 European
LeukemiaNet (ELN) risk stratication of AML (8).
FLT3-TKD mutations are point mutations within the
receptors activation loop which stabilize the active kinase
conformation and also result in constitutive kinase activation
(3). In contrast with FLT3-ITD mutant AML, the prognostic
relevance of FLT3-TKD mutations is less clear and may be
dependent on the presence of co-occurring mutations and
cytogenetic changes (9, 10). Currently, the presence of a FLT3-
TKD mutation does not alter formal AML risk assessment (8).
TESTING CONSIDERATIONS
Given the prognostic and treatment implications, testing for
FLT3-ITD in patients with newly diagnosed AML is
recommended by both ELN and National Comprehensive
Cancer Network (NCCN) guidelines (8, 11). In addition, given
the clonal evolution observed in AML, FLT3 mutations can be
gained or lost at disease relapse and progression. In an individual
patient, the presence of FLT3 mutations will often need to be re-
assessed over time.
Currently, there remains considerable variability in FLT3
assay types, associated sensitivity and specicity, and
turnaround time among treating centers (12). There are two
main methods for determining FLT3 status are polymerase chain
reaction (PCR)-based assays and next generation sequencing
(NGS). Due to competition from the FLT3-WT allele, the
sensitivity of standard PCR-based assays may be lower than
NGS-based methods, although this can be overcome using
patient-specic PCR primers (13).
The FLT3 allelic frequency (AF) has also been used to dene
FLT3-ITD positivity, primarily in research studies. Unlike the
AR, which calculates the ratio of the area under the curve (AUC)
of mutant to WT alleles (FLT3-ITD/FLT3-WT), the AF
calculates the fraction of mutant alleles as a percentage of all
FLT3 alleles (FLT3-ITD/FLT3-WT + FLT3-ITD). For example, a
FLT3 AR of 0.5 (0.5 ITD/1.0 WT) would be equal to a FLT3 AR
of 0.33 (0.5 ITD/0.5 ITD + 1.0 WT = 0.33 AF). NGS studies are
typically reported as VAF while PCR assays typically report AR.
Historically, FLT3-ITD has been inherently difcult to detect
using NGS. NGS relies on the reconstruction of short (<300 base
pair) sequences, and longer length ITDs may not be detected (14,
15) although this can be overcome using novel bioinformatic
approaches (16). NGS is becoming more commonly used,
especially in monitoring for FLT3+ minimal residual disease
(MRD) following treatment. An NGS-based FLT3 assay is now
commercially available and is currently used in in an ongoing
trial of gilteritinib maintenance therapy following hematopoietic
cell transplantation (HCT) (NCT02997202).
Both PCR and NGS-based assays are supported by the current
NCCN guidelines (8); however, ELN risk stratication is
dependent upon FLT3 AR, which can only be determined by
PCR. There is currently no standardized methodology or
laboratory referenc e values for AR determination, and the
current cut-off of 0.5 has not been prospectively validated. In
retrospective analyses, higher AR is generally associated with
inferior clinical outcomes (17), although these studies are prior
to the widespread use of FLT3 inhibitors. It is likely that impact
of AR on prognosis exists on a continuum, rather than a discrete
cut-off. Finally, many patients do not receive FLT3 testing at all.
In a large survey by the American College of Pathologists in
2015, only 51% of new AML referrals received
FLT3 testing (18).
OVERVIEW OF CURRENTLY
ESTABLISHED FLT3 INHIBITORS
Given the prevalence and poor prognosis of FLT3-ITD mutated
AML, targeting FLT3 signaling through small molecule
inhibitors is a promising therapeutic strategy. FLT3 inhibitors
can be stratied using two primary schema: rst vs sec ond
generation FLT3 inhibitors and type I vs type II FLT3
inhibitors. FLT3 inhibitors currently in use or in active
development, including toxicity proles, are detailed in Table 1.
Current usage and ongoing trials of established FLT3
inhibitors in newly diagnosed and relapsed/refractor (R/R)
AML are summarized in Tables 2 and 3, respectively.
Stratication of FLT3 Inhibitors
First generation FLT3 inhibitors include sorafenib, midostaurin,
lestaurtinib, sunitinib, and tandutinib. These multi-kinase
inhibitors target not only FLT3 but other kinases as well,
including PKC, SYK, FLK-1, AKT, PKA, KIT, FGR, SRC,
PDGFRa/b, and VEGFR 1/2 (midostaurin) and RAF, VEGFR
1/2/3, PDGFRb, KIT, and RET (sorafenib). The antileukemic
effects of these non-specic inhibitors likely derive not only from
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128802
FLT3 inhibition, but from inhibition of targets in these parallel
pathways as well. Similarly, due to their mul tiple off- target
effects, rst generation FLT3 inhibitor may also be associated
with increased toxicities. In contrast, second generation FLT3
inhibitors are more potent, FLT3-specic and thus have fewer
off-target effects at clinically relevant doses. Second generation
FLT3 inhibitors include gilteritinib, quizartinib, and crenolanib.
FLT3 inhibitors can also be subdivided based upon how they
interact with the intracellular kinase domain (KD) of the FLT3
receptor. In normal physiology, FLT3 ligand binds to the
extracellular domain, causing the FLT3 receptor to dimerize,
assume an enzymatically active conformation and subsequently
activate downstream signaling. Type I FLT3 inhibitors, which
include midostaurin, gilteritinib, lestaurtinib, and crenolanib,
bind the receptor in the active conformation, thus inhibiting
both FLT3-TKD and FLT3-ITD mutated receptors. Type II
inhibitors, including quizartinib and sorafenib, bind to a region
adjacent to the ATP-binding pocket and only inhibit the receptor
TABLE 2 | Select Trials of Established FLT3 inhibitors in newly diagnosed AML.
NCT/Trial
Identier
Phase Treatment Setting Patient Population
Midostaurin
NCT01477606/
AMLSG 16-10
II Combination with induction and consolidation chemotherapy, plus maintenance FLT3-ITD; age 1870
NCT03512197 III Midostaurin vs placebo in combination with induction and consolidation
chemotherapy
FLT3-WT, dened as FLT3-ITD, D835, or I836 AR
< 0.05; age 18; no prior FLT3 inhibitor therapy
NCT03900949 I Combination with induction chemotherapy plus gemtuzumab ozogamicin FLT3-ITD or -TKD, CD33 +; age 18
NCT04385290/
MOSAIC
II Combination with induction chemotherapy plus gemtuzumab ozogamicin FLT3-ITD or -TKD, age 1875
Gilteritinib
NCT02236013 I Combination with induction and consolidation chemotherapy All AML; age 18
NCT04027309/
HOVON 156
III Gilteritinib vs Midostaurin in combination with induction and consolidation
chemotherapy, plus maintenance
FLT3-ITD or -TKD AML; age 18
Quizartinib
NCT02668653/
QuANTUM-First
III Quizartinib vs Placebo in combination with induction and consolidation
chemotherapy, plus maintenance post-chemotherapy and post-HCT
FLT3-ITD AML; age 1875
NCT03723681 I Combination with induction and consolidation chemotherapy All AML; age 1875
NCT03135054 II Combination with Omacetaxine mepesuccinate FLT3-ITD AML; age 18
NCT04047641 II Combination with cladribine, idarubicin, and cytarabine All AML, MDS; age 1865 (rst-line cohort)
NCT04107727 II Quizartinib vs placebo in combination with induction and consolidation chemotherapy FLT3-WT, dened as FLT3-IT AR < 0.03; age 18
70
NCT04128748 I/II Combination with liposomal cytarabine and anthracycline (CPX-351; Vyxeos) All AML; age 60
Crenolanib
NCT03258931 III Crenolanib vs Midostaurin in combination with induction and consolidation
chemotherapy, plus maintenance
FLT3-ITD or -TKD, age 1860
TABLE 1 | Established FLT3 inhibitors and common toxicity proles.
FLT3 Inhibitor Type Common or Notable Toxicities
First Generation
Midostaurin Type I Hematologic: Cytopenias, including febrile neutropenia and abnormal bruising or bleeding; differentiation syndrome
Constitutional: Pyrexia, u-like symptoms
Cardiac: Cardiac failure (rare, <5%)
GI: Abdominal pain, nausea, vomiting, diarrhea, stomatitis, AST or ALT increase (1921)
Sorafenib Type II Hematologic: Cytopenias, usually mild; differentiation syndrome
Constitutional: Fatigue, can be severe in ~6% of patients
Cardiac: Hypertension, cardiac ischemia (rare, <5%)
GI: Diarrhea
Dermatologic: Rash, erythema, hand-foot skin reaction (2224)
Second Generation
Quizartinib Type II Hematologic: Cytopenias, including febrile neutropenia and abnormal bruising or bleeding; differentiation syndrome
Cardiac: QTc prolongation (dose-dependent, can be severe)
GI: Abdominal pain, nausea, anorexia (25, 26)
Gilteritinib Type I Hematologic: Cytopenias, usually mild; differentiation syndrome
GI: Diarrhea, pancreatitis (rare, <5%, but can be severe), AST or ALT increase
Neurologic: Peripheral neuropathy, headache (27, 28)
Crenolanib Type I Hematologic: Differentiation syndrome
GI: Abdominal pain, nausea, AST or ALT increase
Other: Peripheral edema (29, 30)
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128803
in the inactive conformation. Type II inhibitors are inactive
against most FLT3-TKD mutations as these mutations bias the
active kinase conformation of FLT3. This distinction is key in
understanding mechan isms of FLT3 inhibitor resistance, as
discussed later in this review.
FIRST-GENERATION FLT3 INHIBITORS
Midostaurin
Midostaurin was one of the rst FLT3 inhibitors to be studied in
AML. Early studies of single-agent midostaurin in R/R AML
demonstrated the drug was well-tolerated, but had limited
efcacy (19, 31).
Results of midostaurin in combination therapy have been
more promising. In the phase III RATIFY trial, midostaurin was
evaluated in combination with standard induction and
consolidation chemotherapy in adults <60 years with FLT3-
mutated AML. This combination demonstrated sign icant
improvement in the primary endpoint of overall survival (OS),
with a median OS of 74.7 m onths in patients receiv ing
midostaurin plus chemotherapy vs 25.6 mont hs in patients
receiving placebo plus chemotherapy (p = 0.009) (20). In 2017,
over two decades since FLT3 mutations were rst described in
AML, midostaurin gained US Food and Drug Administration
(FDA) approval, becoming rst agent to achieve FDA approval
for AML since the year 2000.
While the resul ts of RATIFY were promising, there are
some important caveats to consider. In RATIFY, 23% of the
study population had a FLT3-TKD mutation, signicantly
larger than that seen in the general population, and perhaps
biasing the results toward this less-aggressive disease
subtype. In addition, while patients in RATIFY were younger
(median age 48), FDA approval was extended to all age groups
(20). The phase II AMLSG 16-10 trial is currently evaluating
midostaurin in combination with induction and consolidation
chemotherapy in adults up to age 70 (NCT01477606), with initial
results suggesting that older age does not signicantly impact
outcomes, despite the majority of patients requiring midostaurin
dose reduction (32).
Sorafenib
Unlike RATIFY, trials of sorafenib in combination with
chemotherapy have not been conducted in only FLT3-mutant
patients and can thus be more challenging to interpret. Early
phase I/II results of sorafenib in combination with induction
chemotherapy demonstrated prom ising results, with a 75%
complete response (CR) rate in all patients and a 93% CR rate
in the subset with FLT3-ITD mutant AML (22). These ndings,
however, were not replicated in follow-up randomized studies. In
2013, sorafenib plus chemotherapy was evaluated in older adults
(6180 years), but did not show benet in either the primary
endpoint of EFS or in OS, including in FLT3-ITD subgroup
analysis (median EFS 7 vs 5 months, p = 0.12). Furthermore,
sorafenib demonstrated marked toxicity, presumably due to off-
target effects (33).
In 2015, sorafenib plus chemotherapy was evaluated in adults
<60 years in the randomized phase II SORAML trial. While
sorafenib demonstrated improvement in the primary endpoint of
EFS compared to placebo in all patients regardless of FLT3 status
(median EFS 21 vs 9 months, p = 0.013) (23), there was
ultimately no difference in OS (5y OS 61 vs 52%, p = 0.23) (34).
Sorafenib may have efcacy as a single agent. Early phase I
and retrospective studies of sorafenib monotherapy in R/R AML
irrespective of FLT3 status demonstrated acceptable toxicity
proles but mixed CR rates, ranging from 10 to 48% (35, 36).
In studies of FLT3-ITD R/R AML, response rates of sorafenib
monotherapy were 2392% (3739) with some post-transplant
patients achieving sustained remission for multiple years (39,
40). Sorafenib does not have regulatory approval for AML but
can be used off-label in the US as it is approve d for
hepatocellular, renal cell, and thyroid cancer.
Sunitinib, Lestaurtinib, Tandutinib
Other rst-generation FLT3 inhibitors have demonstrated
limited antileukemic effect as monotherapy and mixed results
when combined with chemotherapy. As single-agent therapy for
patients with R/R disease, sunitinib (41), lestaurtinib (42), and
tandutinib (43) have all demonstrated limited and short-lived
responses. In combination with chemotherapy, a phase I/II study
of sunitinib plus frontline chemotherapy in adults >60
TABLE 3 | Select Trials of Established FLT3 inhibitors in R/R AML.
NCT/Trial
Identier
Phase Treatment Setting Patient Population
Quizartinib
NCT03989713/
Q-HAM
II Combination with salvage chemotherapy (Ara-C, Mitoxantrone); R/R after rst-line treatment, including HCT FLT3-ITD AML; age 18
75
NCT04047641 II Combination with cladribine, idarubicin, and cytarabine; any previous treatment All AML, MDS; age 18
(R/R cohort)
NCT04209725 II Combination with liposomal cytarabine and anthracycline (CPX-351; Vyxeos); R/R after any prior treatment, rst-line
treatment must have been standard induction chemotherapy
FLT3-ITD AML; age 18
75
NCT04128748 I/II Combination with liposomal cytarabine and anthracycline (CPX-351; Vyxeos); R/R to rst, second, third, or fourth
line therapy
All AML, MDS; age 18
NCT04112589 I/II Combination with FLAG-Ida (udarabine, cytarabine, idarubicin, GCSF); R/R to frontline standard induction
chemotherapy
All AML; age 1870
Crenolanib
NCT03250338 III Crenolanib vs Placebo plus salvage chemotherapy; R/R after rst or second-line treatment, including HCT FLT3-ITD and -D835;
age 1875
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128804
demonstrated a 59% CR rate; however, multiple patients
experienced d ose-limiting toxicities (44). To contrast, a
randomized phase III trial of lestaurtinib plus frontline
induction and consolidation chemotherapy in patients with
FLT3-mutated AML demonstrated no difference in prim ary
endpoints of OS (5-year OS 46% lestaurtinib vs 45% control,
p = 0.3) or RFS (5-year RFS 40 vs 36%, p = 0.30) (45). Similarly, a
randomized phase III trial of chemotherapy with or without
lestaurtinib in patients with FLT3-mutated AML in rst relapse
demonstrat ed no di fference in the primary endpoint of CR
rates (26 vs 21%, p = 0.35) (42). Currently, none of these
agents are approved for AML and development in AML has
been abandoned.
SECOND-GENERATION FLT3 INHIBITORS
Unlike midostaurin and sorafenib, second-generation FLT3
inhibitors are more specic to the FLT3 receptor, exhibit
greater potency, and have thus been far more efcacious as
single-agent therapy. Both gilteritinib and crenolanib are type I
inhibitors, active against both the active and inactive
conformation of the FLT3 receptor, while quizartinib is a type
II inhibitor, active only against the inactive form.
Quizartinib
Quizartinib has demonstrated improved potency and selectivity
against FLT3-ITD in preclinical cellular assays, and targets KIT
and PDGFR as well as FLT3 (46). Early phase II studies of
quizartinib monotherapy in R/R disease were highly promising,
with CR rates of 4656% and median OS of 25 weeks in FLT3-
ITD mutated AML (25, 47). Quizartinib also demonstrated an
acceptable safety pro le aside from QTc prolongation, leading to
an additional phase IIb study which explored dose reduction to
60 and 90 mg daily vs up to 200 mg daily (47).
Quizartinib was subsequently evaluated in the phase III
randomized QuAN TUM-R trial, which randomized patients
with R/R FLT3-ITD AML to single-agent quizartinib vs salvage
chemotherapy (26). Quizartinib was associated with a
signicantly longer primary endpoint of overall survival (6.2 vs
4.7 months, p = 0.02), and a greater proportion of patients were
able to proceed to hematopoietic cell transplantation (HCT; 32 vs
11%). Total treatment-associated toxicities were similar between
the two arms, although 2% of patients receiving quizartinib
experienced Grade 3 QTc prolongation. Based on these results,
the authors of QuANTUM-R concluded that quizarti nib
monotherapy should be considered a standard of care option
in R/R FLT3-ITD mutated AML (26).
Despite these positive results, in 2019, both the FDA and the
European Medicines Agency (EMA) rejected approval for
quizartinib, although regulatory approval was granted in Japan.
Although the FDA Oncologic Drug Advisory Committee
(ODAC) raised concerns that up to four deaths in the
quizartinib arm were attributed to cardiotoxicity, the decision
to decline was ultimately due to concerns regarding trial design
(48). These concerns included (1) an imbalance in patients who
were randomized but not treated, with 23% of patients
randomized to chemotherapy not receiving treatment vs 11%
of patients randomized to quizartinib and (2) in stratied
analysis, a signicant survival benet only when quizartinib
was compared against low-intensity therapy (low-dose
cytarabine) and not against high-intensity therapy (MEC or
FLAG-Ida). The ODAC concluded that, while a modest
survival benet of 6 weeks was still meaningful, especially if
more patients were bridged to HCT, ultimately additional data
would be needed for quizartinib to be approved in this setting
(48) . The phase II Q-HAM tri al, which has not yet begun
recruiting, will evaluate quizartinib in combination with
salvage chemotherapy in R/R FLT3 -ITD AML (NCT03989713).
In newly di agnos ed AML, quizartinib is currently being
evaluated in the randomized phase III QuANTUM-First trial,
which will compare quizartinib vs placebo in combination with
induction and consolidation chemot herapy (NCT02668653).
Additional phase I/II studies evaluating quizartinib in
combination with frontline cytotoxic chemotherapy are
ongoing as well (NCT03723681, NCT03135054, NCT04047641).
Gilteritinib
Gilteritinib is a selective and potent type I FLT3 inhibitor which
also has activity against AXL, ALT, and ALK (49). Gilteritinib is a
particularly promising agent due to its ability to target KD
mutation s, including the D835 residue, the developm ent of
which is a key mechanism of both quizartinib (50)and
sorafenib (51) resistance. In the phase I/II CHRYSALIS trial,
gilteritinib demonstrated promising results as monotherapy in
R/R FLT3-mutant AML with an overall response rate (ORR) of
40% and a median OS of 25 weeks (52). Notably, patients with
FLT3-D835 mutations also responded to gilteritinib, albeit at
lower rates than patients with FLT3-ITD mutations, with an
ORR of 55% in FLT-ITD-mutated patients, 17% in FLT3-D835-
mutated patients, and 62% in patients with both FLT3-ITD and
FLT-D835 mutations (52).
Following these results, the randomized phase III ADMIRAL
trial compared single-agent gilteritinib vs salvage chemotherapy
in R/R FLT3-mutated AML (27) with co-primary endpoints of
CR rate and OS. Compared to standard salvage chemotherapy,
gilteritinib demonstrated signicantly greater CR rate (34 vs 15%,
p = 0.0001) and improvement in OS (9.3 vs 5.6 months, p <
0.001). While prior use of midostaurin or sorafenib was allowed,
as the trial enrolled prior to midostaurins approval and
widespread use, the majority (88%) of patients had not
received a prior FLT3 inhibitor, limiting the ability of this trial
to evaluate the ability of gilteritinib to overcome midostaurin
resistance; however, results were similar in patients with FLT3-
ITD and FLT3- TKD mutated disease (27). Based on a pre-
planned interim analysis, in 2018, the FDA approved
gilteritinib as single-agent therapy for R/R FLT3-mutated AML.
It is unknown whether gilteritinib is similarly benecial in
newly diagnosed AML. Gilteritinib is currently being studied in a
phase I study in combination with induction and consolidation
chemotherapy in newly diagnosed AML (NCT02236013); interim
results indicate this approach is safe and feasible (53). The phase
III HOVON 156 trial, which is actively accruing, will compare
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128805
gilteritinib vs midostaurin in combination with chemotherapy
followed by FLT3 inhibitor maintenance (NCT04027309).
Crenolanib
Crenolanib is a potent type I inhibitor with activity against
PDGFRb, FLT3-ITD, and FLT3-TKD mutations, including at
D835 (54). Two smaller phase II studies have demonstrated
efcacy of single-agent crenolanib in R/R FLT3-mutated AML,
with a CR rate of 2339% in patients naïve to FLT3 inhibitors,
and 5% patients with prior FLT3 exposure (29, 30).
Crenolanib also demonstrates promising results in
combination with chemotherapy. A phase II trial of crenolanib
plus chemotherapy in newly diagnosed FLT3-mutated AML
demonstrated a high CR rate of 85%; notably, 70% of patients
remained alive and disease free with a median follow-up of 29.3
months (55). Crenolanib plus chemotherapy is also efcacious in
older adults. In a phase II trial of adults 6175 years with newly
diagnosed FLT3-mutated AML, crenolanib plus chemotherapy
demonstrated a CR rate of 86% and was relatively well-tolerated,
although four of 14 patients did require dose-reductions due to
toxicity (56). A phase III trial of crenolanib vs midostaurin plus
chemotherapy in newly diagnosed FLT3-mutated AML is
currently recruiting (NCT03258931).
RESISTANCE TO FLT3 INHIBITORS
Despite the relative success of established FLT3 inhibitors,
responses are frequently short-lived and therapeutic resistance
poses an ongoing challenge. Mechanisms of FLT3 inhibitor
resistance differ based on drug type, but broadly can be
subdivided into cell intrinsic and extrinsic mechanisms.
Intrinsic resistance can be further sub-divided into on-target
secondary mutations within FLT3 and off-target mutations in
downstream or parallel signaling pathways. These mutations
may develop de novo or via expansion of pre-existing
subclones (57). Extrinsic resistance can occur through altered
expression or metabolism of the FLT3 ligand or changes in the
interactions between the leukemic blast and the bone marrow
microenvir onment. Figure 1 illustrates common resistance
pathways and mechanisms.
On-Target Secondary Mutations
One common mechanism of FLT3 inhibitor resistance is
development of a secondary FLT3 mutation, most commonly
in the KD (58). These mutations commonly occur at gatekeep
F691 and activation loop (AL) D835 residues, but can involve
other KD residues I836, D839, and Y842, among others (59).
This mechanism is most relevant for type II FLT3 inhibitors,
which interact weakly with the active receptor formation caused
by some KD mutations. Type I inhibitors gilteritinib and
crenolanib are both active against D835 mutations (60, 61).
To contrast, the gatekeeper F691L mutation confers
resistance not only to quizartinib and sorafenib, but also to
gilteritinib and crenolanib as well (51, 6062). In practice, the
impact of F691L mutations on type I inhibitor resistance may be
relatively minor. In studies of both single-agent gilteritinib (62,
63) and single-agent crenolanib (64) resistance in patients with
FIGURE 1 | Proposed Intrinsic and Extrinsic Mechanisms of FLT3 Inhibitor Resistance. Schematic of FLT3 inhibitor resistance mechanisms, including on-target
secondary FLT3 mutations, off-target mutations in parallel and/or downstream signaling pathways, and extrinsic alterations in drug metabolism and the bone marrow
microenvironment.
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128806
R/R FLT3-mutated AML, only 12% of patients receiving
gilteritinib, and 11% of patients receiving crenolanib developed
F691L mutations at the time of resistance.
In most cases, on-target mutations are not detected prior to
FLT3 inhibitor treatment (65). In a recent study of 11 patients
treated with quizartinib monotherapy, single-cell sequencing
revealed 7/11 patients developed KD mutations at relapse and
no patients had these mutations prior to FLT3 inhibition,
suggesting TK mutations typically evolve de novo (66) or exist
at levels undetectable by current sequencing methods.
Off-Target Resistance in Parallel
or Downstream Pathways
KD mutations only partly explain FLT3 inhibitor resistance, and
expansion or emergence of non-FLT3 mutant clones is a key
resistance mechanism. In patients treated with gilteritinib and
crenolanib, sequencing of paired patient samples pre- and post-
therapy demonstrated a wide variety of mutations at resistance,
including genes involved in the RAS pathways (NRAS, KRAS,
PTPN11), ASXL1, TP53, TET2, and IDH1/2. These mutations
occurred not only in cells expressing the FLT3-mutant allele, but
in FLT3-WT cells as well (63, 64). These off-target mutations are
not limited to type I inhibitors. In a study using single-cell
sequencing to analyze 11 patients treated with quizartinib, 3/11
demonstrated off-target mutations at relapse, all of which were
present in small clonal populations prior to FLT3 inhibitor
therapy (66).
Of these off-target pathways, activation of the downstream
Ras and associated PI3K/Akt/mTOR and MAPK/Erk pathways
are particularly common in clinical gilteritinib and crenolanib
resistance (6264), and in vitro studies have demonstrated
mutations in these pathways can confer resistance to FLT3
inhibitors in FLT3-mutant cell lines (67, 68). Development of
the BCR-ABL1 fusion gene has been describe in patients with
gilteritinib resistance (63, 69).
Upregulation of parallel AXL tyrosine kinase signaling is
another mechanism of FLT3 inhibitor resistance. In one study,
blasts from a patient with FLT3-ITD AML were exposed to both
midostaurin and quizartinib and were found to have increased
AXL phosphorylation up on development of FLT3 inhibitor
resistance. This resistance could be overcome with use of the
AXL inhibitor TP-0903 (70, 71) and a phase I trial of TP-0903
with or without azacitidine in FLT3-mutated AML has recently
opened (NCT04518345).
Together, these studies suggest off-target resistance
mechanisms are common to all FLT3 inhibitors and frequently
arise via selection of pre-existing sub-clones harboring survival
advantages under the selective pressure of FLT3 inhibition.
Extrinsic Mechanisms of Resistance
The majority of leukemic blasts, regardless of mutational status,
express the FLT3 receptor and proliferate in response to FLT3
ligand. Increased levels of FLT3 ligand in the bone marrow
microenvironment have been demonstrated during induction
and consolidation chemotherapy, and can lead to increased
signaling via the native FLT3 receptor, even in the presence of
FLT3 inhibitors (72, 73). FLT3 inhib itors may also have
decreased efcacy due to decreased drug availability, either due
to enhanced CYP34A expression on BM stromal cells (74)or
iatrogenic drug-drug interactions (75).
The bone marrow microenvironment c an also directly
contribute to FLT3 inhibitor resistance. In addition to directly
secreting FLT3 ligand, bone marrow stro mal cells can also
upregulate Ras/MAPK signaling independent of the FLT3
receptor via secretion of FGF2 (76). In one study of patients
treated with quizartinib, stromal cell expression of FGF2 conferred
FLT3 inhibitor resistance, and could be overcame by FGFR
inhibition (76, 77). Multiple dual FLT3/FGFR inhibitors are in
pre-clinical development (78, 79) with MAX-40279 currently in
phase I clinical trials (NCT03412292, NCT04187495). In addition,
ponatinib, a multikinase inhibitor approved in CML, has activity
against both FLT3 and FGFR (80).
NOVEL FLT3 INHIBITOR COMBINATIONS
One strategy to overcome resistance and provide durable
remissions is to use FLT3 inhibitors in novel combinations
with other antileukemic agents (Tables 4, 5; Figure 2).
Hypomethylating Agents
Aside from conventional cytotoxic chemotherapy, the most well-
studied FLT3 inhibitor combination is with the hypomethylating
agents (HMA) azacitidine or decitabine. This combination is
particularly attractive, both due to the synergistic cytotoxicity in
preclinical studies as well as the established tolerability and
durable responses of HMAs in older adults (81, 82).
Multiple phase I/II trials have demonstrated the combination
of midostaurin and HMA to be feasible in adults with FLT-
mutated AML who are unt for traditional chemotherapy in the
frontline setting (83) and in R/R disease (82, 84 ). Additional
trials of midost aurin plus HMA for older/untadultshave
opened, but have terminated due to inability to accrue
(NCT01846624. NCT02634827). There is an ongoing trial of
midostaurin plus azacitidine for newly diagnosed AML
regardless of FLT3 mutational status (NCT01093573), w ith
primary endpoints of tolerability and ORR.
Sorafenib plus HMA have also been shown to be safe and
efcacious in single-arm and retrospective studies in both the
R/R (85, 86) and frontline settings (87). In the frontline setting,
sorafenib plus HMA demonstrated an ORR of 78%, the studys
primary endpoint, and a and median response duration of 14.5
(87). Similarly, a phase I/II study of sorfenib plus azacitidine
demonstrated an ORR of 46%, the secondary endpoint for the
trials phase II portion, and median response duration of 2.3
months (85). Notably, FLT3 ligand levels remained low with this
combination, which is intriguing as increase in ligand expression
has been suggested as a possible mechanism of FLT3 inhibitor
resistance (72, 85).
Of the second generation FLT3 inhibitors, an interim analysis
of a phase I/II trial of quizartinib plus azacitidine in unt patients
with newly diagnosed or in R/R FLT3-ITD AML demonstrated
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128807
an ORR of 75%, the secondary endpoint for the trials phase II
portion, including in four/ve patients with prior FLT3 inhibitor
exposure (88). A phase II study of gilteritinib plus azacitidine in
unt patients with newly diagnosed FLT3-ITD AML with
primary endpoint of OS is currently accruing (NCT02752035)
with interim results of secondary endpoints demonstrating CR
and ORR rates of 67 and 80%, respectively (89).
Venetoclax
Venetoclax, an inhibitor of the anti-apoptotic protein Bcl-2, is
particularly intriguing in combinatio n with FLT3 inhibitors.
Preclinical studies have shown FLT3-ITD mutated blasts have
higher Bcl-2 expression compared to FLT3-WT blasts (90) and
upregulation of antiapoptotic proteins is a mechanism of FLT3
inhibitor resistance (91). In preclinical mouse models, venetoclax
has demonstrated s ynergistic antileukemic activity with
midostaurin (92), gilteritinib (92), and quizartinib (93). In
addition, FLT3-ITD mutations were frequently observed at
progression in trials of venetoclax monotherapy in AML (94, 95).
Gilteritinib is currently being studied in combination with
venetoclax in R/R AML (NCT03625505). Quizartinib is being
evaluated in combination with venetoclax and decitabine in
poor-risk newly diagnosed or R/R FLT3- mutated AML
(NCT03661307) and in combination with venetoclax alone in
R/R FLT3-ITD AML (NCT03735875).
Proteosome Inhibitors
Proteosome inhibitors, including bortezomib, have
demonstrated cytotoxicity toward FLT3-ITD mutant cells in
preclinical studies (96) and a phase I study of midostaurin in
combination with bortezomib and chemotherapy in R/R AML
demonstrated activity, albeit with marked toxicity ( 97). Sorafenib
TABLE 4 | Select Trials of Established FLT3 Inhibitors in Combination Therapy.
Combination
Agent
FLT3
inhibitor
NCT/Trial
Identied
Phase Treatment Setting Patient Population
Hypomethylating Agents
Azacitidine Midostaurin NCT01093573 I/II Newly diagnosed All AML; age 18 and unt for
chemotherapy
Decitabine Midostaurin NCT04097470;
HO-155
II Midostaurin plus decitabine vs decitabine alone, newly
diagnosed
All AML, age 18100 and unt for
chemotherapy
Azacitidine Sorafenib NCT02196857 II Newly diagnosed FLT3-ITD, TKD AML, MDS; age 60 or 18
60 and unt for chemotherapy
Azacitidine Gilteritinib NCT02752035;
LACEWING
II/III Gilteritinib monotherapy vs azacitidine monotherapy vs
gilteritinib plus azacitidine; newly diagnosed AML
FLT3-ITD, TKD; age 65 or 1865 and
unt for chemotherapy
Azacitidine or
Low-Dose AraC
Quizartinib NCT01892371 I/II Quizartinib plus azacitidine or cytarabine; Newly diagnosed or
R/R after rst or second-line treatment, including HCT
All AML, MDS, CMML; age 60 (all
settings) or age 18 (R/R only)
Venetoclax +/- HMA
Venetoclax Gilteritinib NCT03625505 I R/R to at least one prior therapy All AML; age 18
Venetoclax +
Azacitidine
Gilteritinib NCT04140487 I/II Newly diagnosed or R/R FLT3-ITD or D835 AML; age 18
Venetoclax +
Decitabine
Quizartinib NCT03661307 I/II Newly diagnosed; R/R and not eligible for salvage
chemotherapy or HCT
FLT3-ITD or FLT3-ITD/TKD co-mutations;
age 60 or age 18 and unt for
chemotherapy
Venetoclax Quizartinib NCT03735875 Ib/II R/R up to 4 prior therapies FLT3-ITD; age 18
Proteosome Inhibitors
Bortezomib Sorafenib NCT01371981 III Bortezomib plus sorafenib plus chemotherapy vs sorafenib
plus chemotherapy; Newly diagnosed
FLT3-ITD; age < 29
Bortezomib,
then decitabine
Sorafenib NCT01861314 I Newly diagnosed, R/R to at least one prior therapy All AML; age 60 or 18 and unt for
chemotherapy
Bortezomib,
Vorinostat
Sorafenib NCT01534260 I/II R/R to at least one prior therapy FLT3-mutated or poor-risk cytogenetics;
age 18
Targeted Agents
Pim kinase
inhibitor
(LGH447)
Midostaurin NCT02078609 I R/R after rst or second-line treatment All AML, MDS; age 18
mTOR inhibitor
(Everolimus)
Midostaurin NCT00819546 I R/R to at least one prior therapy All AML, MDS; age 18
HDM2 inhibitor
(HDM201)
Midostaurin NCT04496999 I R/R to at least one prior therapy FLT3-ITD or FLT3-TKD and TP53-WT; age
18
CDK 4/6
inhibitor
(Palbociclib)
Sorafenib NCT03132454 I Palbociclib in combination with sorafenib vs decitabine vs
dexamethasone, newly diagnosed
All AML, ALL; age 15
MDM2 inhibitor
(Milademetan)
Quizartinib NCT03552029 I Newly diagnosed and ineligible for intensive therapy; R/R to at
least one prior therapy
FLT3-ITD; age 18
Immunotherapy
Atezolizumab Gilteritinib NCT03730012 I/II R/R to at least one prior therapy FLT3-mutated AML; age 18
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128808
plus bortezomib was also studi ed in a combination with
vorinostat, a histone deacetylase inhibitor, in a phase I/II trial
of R/R AML and demonstrated a modest ORR of 28% in all
patients and 60% in patients with FLT3-ITD AML (98). Ongoing
studies of this combination include a phase III trial of sorafenib
plus bortezomib in younger adults (up to age 29) with newly
diagnosed FLT3-ITD AML (NCT01371981) and sorafenib plus
bortezomib followed by decitabine in newly diagnosed or R/R
AML regardless of FLT3 status (NCT01861314).
Additional Targeted Agents
Multiple agents targeting signaling pathways downstream of
FLT3 signaling, including those frequently implicated in FLT3
inhibitor resistance, have been studied in combination therapies.
TABLE 5 | Select Trials of Novel FLT3-Directed Therapies.
Combination Agent NCT Phase Treatment Setting Study Population
Multikinase Inhibitors
Ponatinib NCT02428543 I/II AML in rst CR, following induction and consolidation
with standard cytotoxic chemotherapy
FLT3-ITD with AR > 10%; age 1870
Ponatinib NCT03690115;
PONALLO
II AML in CR, following allo-HCT FLT3-ITD; age 18 - 70
Novel Dual Agents
SEL24/MEN1703: Dual FLT3/Pim
kinase inhibitor
NCT03008187 I R/R, no standard treatment options available All AML; age 18
MAX-040279: Dual FGFR/FLT3
inhibitor
NCT03412292 I R/R, no standard treatment options available All AML; age 18
MAX-040279: Dual FGFR/FLT3
inhibitor
NCT04187495 I R/R, no standard treatment options available All AML; age 18
CG-806: Dual BTK/FLT3 inhibitor NCT04477291 Ia/b R/R to at least one prior therapy All AML; age 18
Novel FLT3 Inhibitors
FF-10101 NCT03194685 I/IIa R/R to at least one prior therapy All AML; age 18
HM43239 NCT03850574 I/II R/R to at least one prior therapy All AML; age 18
Biologic Agents
FLYNSYN: Anti-FLT3 IgG1
Antibody
NCT02789254;
FLYSYN-101
I/II AML in and hematologic CR but MRD+ after
chemotherapy but not HCT
All AML, but leukemic cells must express
FLT3 by ow cytometry; age 18
ASP1235 (AGS62P1): anti-FL3
antibody-drug-conjugate
NCT02864290 I R/R to rst, second, or third therapy All AML; age 18 and not candidate for
salvage chemotherapy
AMG 553: FLT3 CART NCT03904069 I R/R, no standard treatment options available All AML, but leukemic cells must express
FLT3 by ow cytometry; age 12
FIGURE 2 | Established and In-Development FLT3 inhibitors, dual inhibitors, and combination agents. Schematic detailing mechanisms of action of established and
in-development FLT3 inhibitors, dual and multikinase inhibitors, and combination agents.
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 6128809
JAK/STAT5/Pim-1 is a key signaling pathway parallel and
downstream of FLT3. Pim kinase inhibitors inhibit Pim-1, a
kinase which promotes FLT3 signaling via positive feedback (99,
100). In preclinical models, Pim kinase and FLT3 inhibitors
demonstrate synergistic cytotoxicity (100, 101). The Pim kinase
inhibitor L GH447 is b eing studie d in com bination wit h
midostaurin in a phase I trial (NCT02078609), as is the novel
dual Pim kinase/FLT3 inhibitor SEL24 (NCT03008187). Other
agents, including the dual JAK/FLT3 inhibitor pacritinib, have
demonstrated efcacy in phase I trials as well (102).
mTOR inhibitors, such as everolimus, target the PI3K/AKT/
mTOR pathways, which is similarly downstream of FLT3 signaling.
Concomitant inhibition of both mTOR and FLT3 demonstrate
synergistic cytotoxicity (103)andmTORisupregulatedinAML
blasts resistant to FLT3 inhibitors (68). A phase I study of
midostaurinpluseverolimusisactive, but not currently recruiting
(NCT00819546). In addition, metformin, a drug long approved in
diabetes, can also down-regulate the P31K/Akt/mTO R pathways,
and has shown to act synergistically with sorafenib in FLT3-
mutated cell lines (104).
Cyclin-dependent kinase 6 (CKD6) is a key regulator of cell cycle
progression, part of a transcriptional complex that promotes
leukemogenesis, and is found at the promoter of both FLT3 and
PIM1 genes (105, 106). A phase I study of the CDK4/6 inhibitor
palbociclib, which is approved in breast cancer, in combination with
sorafenib is actively recruiting (NCT03132454). In add ition,
multiple novel dual FLT3/CDK4 inhibitors are in active
preclinical development, including AMS-925 (107) (Keegan),
ETH-155008 (108), and FLX925 (109), which recently completed
a phase I dose-escalation trial (NCT02335814).
Finally, the tumor suppressor p53 is increasingly recognized as a
mechanism of FLT3 inhibitor resistance, particularly to crenolanib
(64). Milademetan, an inhibitor of the p53-regulatory protein
MDM2, has demonstrated synergistic anti-leukemic activity with
quizartinib in FLT3-mutant cell lines (110)andaphaseItrialis
actively recruiting (NCT03552029). Similarly, HDM-201, an
inhibitor of the p53-regulatory protein HDM2,isactively
investigated in combination with midostaurin (NCT04496999).
Immunotherapy Combinations
Compared to lymphoid and many solid malignancies, AML has
thus far demonstrated limited response to immunotherapies.
Exploratory studies have indicated that elevated programmed
cell death 1 (PD-1) and PD-1 ligand (PD-L1) are associated with
inferior OS in AML, including in patients with FLT3- mutated
disease (111). A phase I/II trial of gilteritinib in combination with
the checkpoint inhibitor atezolizumab in R/R FLT-mutated AML
is ongoing (NCT03730012).
NOVEL FLT3 INHIBITORS
IN DEVELOPMENT
In addition to novel FLT3 inhibitor combinations, multiple novel
FLT3 inhibitors are in active preclinical and clinical development
(Table 5; Figure 2). These include multikinase inhibitors
approved for non-AML malignancies as well as specic, next-
generation agents. In addition to overcoming pre-established
mechanisms of resistance, including F691 mutations, these novel
agents also offer alternative and potentially more desirable
toxicity proles.
Multikinase Inhibitors
Ponatinib is approved to target BCR-ABL in chronic myelogenous
leukemia (CML), but is also a type II FLT3 inhibitor with
activity against F691L (112). Ponatinib demonstrated modest
efcacy in a phase I trial of heavily-pretreated R/R AML patients
(113), and is being actively investigated in combination with
chemotherapy (NCT02428543).
Cabozantinib is a multikinase inhibitor currently approved
for medullary thyroid and renal cell carcinomas. Cabozantinib is
selectively cytotoxic to FLT3-ITD mutant cells in culture (114)
and a phase I trial demonstrated sustained inhibition of FLT3-
ITD and -F691 mutant cells, although no treated patients had a
formal disease response (115).
The multikinase inhibitor pexidartinib (PLX3397) has been
studied in multiple solid tumors and also demonstrates activity
against FLT3, including F691L (50). Recently, a phase I/II trial of
single-agent pexidartinib in R/R FLT3-ITD AML demonstrated
an ORR of 21% and CR rate of 11%; furthermore, several patients
were successfully bridged to transplant ( 116 ).
Finally, the BTK inhibitor ibrutinib, currently approved for
lymphoid malignancies, also demonstrates type II FLT3
inhibitory effects (117). A phase I trial of CG-806, a dual BTK/
FLT3 inhibitor, recently opened for patients with R/R
AML (NCT04477291).
Novel FLT3 Inhibitors
One of the most promising novel agents is FF-10101, the rst
covalently-binding FLT3 inhibitor. In pre-clinical studies, FF-
10101 demonstrated potent activity against quizartinib-resistant
AL and gatekeeper F691 muta tions (118).Otheragentsin
development include TTT-3002, G-749, MZH-29, and
HM43239, all highly se lective FLT3 inhibitors with activity
against D835 and F691 residues (119122). In preclinical
studies, these agents have demonstrated activity against AML
blasts resistant to sorafenib and quizartinib (119) or midostaurin
and quizartinib (120), and may represent options for refractory
disease. Phase I/II trials of both FF-10101 and HM43239 are
ongoing (NCT03194685, NCT03850574).
Biologic Agents
To date, pharmacologic targeting of FLT3-mutant AML has
primarily focused on signaling inhibition via small molecules;
however, multiple immunotherapeutic approaches are
in development.
Ongoing trials are investigating targeting FLT3 through an
IgG1 antibody (FLYSYN; NCT02789254), with promising
preliminary efcacy data (123), as well as via an anti-FLT3
antibody drug conjugate (124) (NCT02864290). In addition, an
anti-FLT3/anti-CD3 bi-specic antibody has shown promise in
preclinical models as well (125). Finally, FLT3 may represent a
potential target for chimeric antigen receptor T cell (CART)
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288010
therapy (126), including in combination with established FLT3
inhibitors (127). A phase I trial of a FLT3-directed CART has
recently opened (NCT03904069).
ONGOING QUESTIONS
AND CONTROVERSIES
Despite the considerable advances in trea ting FLT3-mutant
AML, many outstanding questions and controversies remain.
We have summarized a few of the most intriguing and timely
questions below.
What is the Benet of Maintenance
Therapy
in FLT3-Mutant AML?
Of all the questions regarding the clinical use of FLT3 inhibitors,
one of the most pressing is the role of maintenance therapy. Use
of FLT3 inhibitor maintenanc e, either during remission for
patients who do not undergo HCT or during post-HCT
remission, is currently not standard of care; however, there are
data to suggest this may be a promising approach. Table 6
describes ongoing clinical trials of maintenance therapy.
Post-Chemotherapy Maintenance
Perhaps most notably, in the experimental arm of the
phase III RATIFY study, patients could receive up to one year
of midostaurin maintenance following induction and
consolidation chemotherapy plus midostaurin (20). In
unplanned post-hoc analysis of the subset of p atients who
received either midostaurin or placebo maintenance, there was
no benet seen with midostaurin, although it was well-tolerated
(128). Importantly, the m edian duration of midostaurin
exposure was 3 months, as the majority (59%) of p atients
randomized to midostaurin received allogeneic HCT and thus
received only two to three cycles of therapy, limiting the ability to
draw conclusions from midostau rin maintenance data (20).
Maintenance midostaurin after chemotherapy did not receive
US FDA approval; however, regulatory approval for
maintenance midostaurin was granted by the EMA.
Currently, both the single-arm AMLSG 1610 trials, which
will also evaluate midostaurin in combination with
chemotherapy in older adults, and the phase III HOVON 156
AML trial, which will compare midostaurin vs gilteritinib plus
chemotherapy in newly diagnosed AML, will each evaluate up to
one year of midostaurin maintenance following chemotherapy
(NCT01477606, NCT04027309). Of note, neither of these trials
will directly compare maintenance midostaurin against placebo,
which will make it difcult to isolate the true benet of post-
chemotherapy midostaurin maintenance.
Multiple ongoing trials are also evaluating second generation
FLT3 inhibitors as post-chemotherapy maintenance. In the
HOVON 156 AML trial, patients may receive up to one year
of gilteritinib maintenance, although again, this study will not
compare gilteritinib against a placebo control. A separate
randomized phase III trial will compare gilteritinib vs placebo
maintenance for up to two years following induction and
consolidation therapy in patients who are not proceeding to
HCT accrual (NCT02927262). Similarly, the QuANTUM-First
trial will include up to three years of post-chemotherapy
quizartinib maintenance and will be placebo-controlled.
Post-HCT Maintenance
In the post-HCT setting, midostaurin was evaluated in a phase II
trial as single-agent maintenance following midostaurin plus
chemotherapy and subsequent HCT. Of the 56% of patients
who ultimately received HCT and subsequent maintenance,
mido staurin maintenance was associated with improved OS
compared to a historical control group; however, given this
comparison was relative to historic controls it must be
interpreted with caution (129). Midostaurin maintenance was
also associated with signicant toxicities, particularly in
older adults.
In the phase II RADIUS trial, patients were ran domized
to receive up to one year of maintenanc e midostaurin vs
TABLE 6 | Select Trials of FLT3 Inhibitors as Post-Chemotherapy or Post-HCT Maintenance Therapy.
FLT3 inhibitor NCT/Trial
Identier
Phase Treatment Setting Patient Population
Midostaurin NCT01477606/
AMLSG 1610
II Combination with induction and consolidation chemotherapy, plus maintenance FLT3-ITD; age <70
Midostaurin/
Gilteritinib
NCT04027309/
HOVON 156
III Gilteritinib vs Midostaurin in combination with induction and consolidation chemotherapy,
plus maintenance
FLT3-ITD or -TKD AML;
age 18
Midostaurin NCT03951961/
MAURITUS
II Midostaurin maintenance post-HCT; MRD + disease FLT3-mutated; age 18
Midostaurin/
Crenolanib
NCT03258931 III Midostaurin vs Crenolanib post-HCT; MRD+ disease FLT3-mutated; age 18
60
Gilteritinib NCT02927262 III Gilteritinib vs Placebo for up to 2 years following induction and consolidation
chemotherapy
FLT3-ITD; age 18 with
no plan for HCT
Gilteritinib NCT02997202/BMT
CTN 1506
III Gilteritinib vs. Placebo following HCT; AML in CR1 or CRi1 FLT3-ITDi; age 18
Quizartinib NCT02668653/
QuANTUM-First
III Quizartinib vs Placebo in combination with induction and consolidation chemotherapy,
plus maintenance post-chemotherapy and post-HCT
FLT3-ITD AML; age 18
75
Crenolanib NCT02400255 II Crenolanib following HCT; AML in any CR by morphologic assessment FLT3-ITD and -D835; age
18
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288011
standard care following HCT. Although preliminary reports
suggested that addition of midostaurin could reduce risk of
relapse at 18 months post-HCT by 46%, 63% of patients
receiving midostaurin required dose modications and 25%
discontinued midostaurin due to toxicity. RADIUS ultimately
had inadequate enrollment to detect a statistically signicant
difference in 18-month relapse-free survival (RFS), the studys
primary endpoint (estimated 18-month RFS 89% in midostaurin
arm vs 76% in standard-of-care arm, p = 0.27) (130).
Similarly, post-HCT sorafenib maintenance was shown to be
tolerable and potentially efcacious in both retrospective and
prospective phase I/II studies, although frequent dose
adjustments were needed ( 131, 132 ). More recently, the phase
II SORMAIN trial randomized patients with FLT3-ITD AML in
remission following HCT to maintenance with two years of
sorafenib vs placebo (133). The HR for RFS, the primary
endpoint, demonstrated a signicant benet with sorafenib vs
placebo (HR 0.39, 95% CI 0.180.85, p = 0.013); however, only 9/
43 (21%) of patients receiving sorafenib had received a frontline
FLT3 inhibitor, so it remains unclear whether the same benet
would be seen in patients who received a FLT3 inhibitor in the
frontline setting. Similar to the RADIUS trial, study drug
discontinuation was more common in the sorafenib arm (22 vs
5%), although this difference was not signicant. Finally, and
again similar to the RADIUS trail, SORMAIN did not reach is
target accrual and was terminated prematurely.
Building on the results of RADIUS and SORMAIN, there the
multiple ongoing trails evaluating post-HCT maintenance
therapy. The phase II, single-arm MAURITUS trial will
evaluate midostaurin maintenance following HCT in MRD-
positive FLT3-mutated AML (NCT03951961) and a phase III
trial with compare post-HCT midostaurin vs cren olanib
maintenance (NCT03258931).
For the second generation FLT3 inhibitors, BMT CTN 1506, a
randomized phase III trial of gilteritinib vs placebo for FLT3-ITD
mutated AML following HCT is ongoing (NCT02997202).
Importantly, although prior FLT3 inhibitor treatment is not an
inclusion criterion, since enrollment occurred after the
widespread use of midostaurin in the frontline setting, the
majority of patients enrolled on BMT CTN 1506 will likely
have received prior FLT3 inhibitor therapy, answering a key
question raised by SORMAIN. In addition, quizartinib was
shown to b e well-tolerated as single-agent maintenance
following HCT in a phase I study (134) and this strategy will
be further explored in the QuANTUM-First trial. Finally, a non-
randomized trial is evaluating the efcacy of crenolanib post-
HCT in FLT3-mutated AML (NCT02400255).
Maintenance Therapy: Where Do We Go From Here?
The role of FLT3 inhibitor maintenance, while promising,
remains unknown, and placebo-controlled randomized trials
are necessary to establish the efcacy of this approach.
Multiple questions remain, including: How can we identify the
patients for whic h FLT3 inhibitor mainten ance is most
benecial? In SORMAIN, the strongest benet from sorafenib
maintenance was in patients with undetectable MRD pre-HCT
and detectable MRD post-HCT (133). Notably, the ongoing
BMT 1506 trial will include correlative MRD analysis to
determine if the presence of MRD is predictive of benet from
FLT3 inhibitor maintenance. What is the optimal duration of
maintenance the rapy ? In a correlative analysis of RADIUS,
decreased levels of phosphorylated FLT3 were associated with
improved OS (135), suggesting this may be one potential
biomarker for determining maintenance duration. What type
of FLT3 inhibitor is most efcacious as a maintenance agent, a
multikinase, rst-generation agent, or a more targeted, second
generation one? In the post-chemotherapy setting, do es
mainten ance serve as a bridge to transplant, or obv iate the
need? Finally, what is the effect of maintenance therapy on
health-associat ed quality of life? Given that t his patient
population may not have active disease, studies sp ecically
investigating health-associated quality of life are ne eded to
fully inform the benet of prolonged maintenance therapy.
What Is the Role of Transplant in FLT3-
Mutant AML?
Historically, FLT3-mutated AML has been considered adverse
risk disease, and patients with FLT3-ITD have been
recommen ded to undergo HCT in the rst CR (136, 137).
More recently, the ELN has re-classied FLT3-mutated AML
such that patients with FLT3-ITD
low
(AR < 0.5), normal
cytogenetics, and mutated NPM1 are considered low risk,
suggesting these patients may have good prognosis without
HCT (138). This is not widely accepted, however, and
retrospective data have demonstrated that low risk FLT3-ITD
AML, with FLT3- IT low AR and positive NPM1 mutational
status, still conveys poor prognosis, with a 5-year OS of 41.3%,
and OS is improved by HCT (139). A similar retrospective study
found that HCT in rst CR improves OS in all FLT3-ITD AML,
regardless of AR or NPM1 status (140). Importantly, both these
studies and the ELN guidelines were written prior to widespread
FLT3 inhibitor use, and the role of HCT in FLT3-mutated AML
today remains an open question.
In a retrospective analysis of the RATIFY trial, the benecial
effect of midostaurin was seen across all ELN subgroups;
however, the benet of HCT was only seen in patients with
adverse risk disease (138). This should be interprete d with
caution as RATIFY was not powered for these sub group
analyses. Nonetheless, this study provides support that in low
or even intermediate risk FLT3-ITD AML, HCT could
potentially be delayed until rst relapse or MRD positivity. Of
note, in RATIFY, patients that did not receive HCT did receive
post-consolidation midostaurin maintenance, an indication that
was not approved as discussed above.
More recently, in a phase II study of crenolanib plus
chemotherapy followed by crenolanib maintenance in newly
diagnosed FLT3-mutated AML, 85% of patients achieved CR.
Of the 27 patients on trial, 7/27 received consolidation but not
HCT; of those, 6/7 remained in long-term remission. OS was
similar between patients who underwent HCT vs those who did
not (55). While the number of patients is small and the ELN risk
category not specied, this again raises the question of whether
HCT is needed in all patients with FLT3-mutated AML.
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288012
In a recent position statement by the European Society for
Blood and Marrow Transplantation (EBMT), HCT in rst CR
was recommended for patients with intermediate or adverse risk
FLT3-ITD AML and could be considered in low risk disease.
Furthermore, in the absence of GVHD, post-HCT F LT3
inhibitor maintenance was recommended, ideally on a clinical
trial (141). Randomized trials are needed to further clarify these
algorithms. Multiple trials of FLT3 inhibitors plus chemotherapy
in newly diagnosed AML are actively accruing, and it will be
interesting to see the role of HCT in these studies.
Given New FLT3-Directed Therapies, Will
We Need to Re-Think Risk Stratication?
The current ELN criteria were developed before FLT3 inhibitors
were routinely used. Prior to widespread FLT3 inhibitor use, the
prognosis for FLT3-ITD AML was dismal. Historically, while
patients with FLT3-ITD responded to induction chemotherapy
with similar remission rates as other AML subtypes, patients
were more likely to relapse and had inferior OS (7, 142).
In a retrospective analysis of the RATIFY trial, for all enrolled
patients, OS differed signicantly among ELN risk groups. In all
risk groups, midostaurin signicantly improved OS, with 5-year
OS probabilities for the midostaurin arm of 0.53 and 0.52 for
intermediate- and adverse-risk, respectively (138). More recently
crenolanib plus chemotherapy in newly diagnosed AML has
demonstrated a 3-year OS of 0.76 in adults 60 ( 55) and a 1-year
OS of 0.67 in ad ults 6175 (56). As a comparison, in at
retrospective validation of ELN risk stratication in newly-
diagnosed patients receiving conventional chemotherapy, 5-year
OS probabilities were 0.36 for FLT3-ITD
low
/NPM1
WT
and 0.29 for
FLT3-ITD
high
/NPM1
mutated
(intermediate risk) and 0.09 for FLT3-
ITDhigh/NPM-WT (adverse risk) (143). While historic comparisons
must be interpreted with caution, this may suggest FLT3 inhibitors
have shifted the risk associated with FLT3-mutated AML.
Given the multiple FLT3-directed therapies both approved
and in development, the prognosis of FLT3-mutated AML may
be changing. Will FLT3 mutations in AML e ventually be
analogous to HER2 amp licatio n in breast cancer or BCR-
ABL1 fusions in acute lymphoblastic leukemia? In both of
these cases, the development of targeted therapies has
dramatically improved the prognosis of a previously poor-risk
subtype, and a similar pattern may evolve with FLT3 as well.
What Is the Prognostic and Therapeutic
Impact of Non-Canonical FLT3 Mutations?
As sequencing technology improves, FLT3 mutations outside of
the ITD and D835 regions have been described. These non-
canonical (NC) mutations are frequently in exon 14 of the
juxtamembrane (JM) domain, where ITDs occur, or in the KD
domain adjacent to D835; however NC mutations in other sites,
including the extracellular (EC) domain, have been described as
well. Select NC mutations identied in the clinical literature are
summarized in Table 7.
In the pediatric literature, whole genome sequencing of 799
pediatric AML patients revealed a 7.6% prevalence of NC FLT3
point mutations and insertions-deletions compared to a 23%
total prevalence of all FLT3 mutations (including FLT-ITD and
D835). This included 9 JM mutations with activating potential
(E598D, E573D/G, L576R, T582N, D586Y, Y589H, E596K/G,
Y599C, D600G), many which occurred as co-mutations with
FLT3-ITD (145). Similarly, in a large study of 1,540 adult AML
patients, targeting genetic sequencing identied four NC FLT3
driver mutation, including two EC (S451F and V491L) and two
JM (V592A, E598D) mutations (4).
TABLE 7 | Non-Canonical Mutations Identied in Clinical Studies.
Mutation Exon Domain Clinical Activity
D200N 5 EC Maintained through crenolanib treatment (64)
T227M 6 EC Confers resistance to sorafenib (68)
K429E 10 EC Maintained through crenolanib treatment (64)
S451F 11 EC Pediatric AML (5); Adult AML (4); minimal midostaurin sensitivity (144)
V491L 12 EC Pediatric AML (5); Adult AML (4)
Y572C 14 JM High midostaurin sensitivity (144); maintained through crenolanib treatment (64)
E573D/G 14 JM Pediatric AML (145)
L576R 14 JM Pediatric AML (145)
T582N 14 JM Pediatric AML (145)
D586Y 14 JM Pediatric AML (145)
Y589H 14 JM Pediatric AML (145)
V592A/G 14 JM Pediatric AML (5); Adult AML (4); high midostaurin sensitivity (144); clinical sorafenib response (146)
E596K/G 14 JM Pediatric AML (145)
E598D 14 JM Adult AML (4); Pediatric AML (5, 145); found after relapse on Giltertininb (62)
Y599C 14 JM Pediatric AML (145)
D600G 14 JM Pediatric AML (145)
L601F 14 JM Mutation maintained through crenolanib treatment (64)
N676D/K 16 KD Clinical sorafenib response (146), Resistance to midostaurin (147); resistance to quizartinib (148)
G697R 16 KD Resistance to quizartinib (148)
A833S 20 AL Eliminated with crenolanib treatment (64)
R834Q 20 AL High Midostaurin sensitivity (144)
D839Y/G 20 AL Eliminated with crenolanib treatment (64)
N841K 20 AL Eliminated with crenolanib treatment (64)
Y842C 20 AL Eliminated with crenolanib treatment (
64)
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288013
It is unclear to whether NC mutations confer FLT3 inhibitor
susceptibility, resistance, or are simply passenger mutations
unimportant to disease biology. In one study of 222 AML
patients without FLT3-ITD or -D835 mutations, four NC
driver mutations were identied which had variable sensitivity
to midostaurin (144 ). Similarly, in a study of 18 patients treated
with crenolanib, pre- and post-treatment sequencing identied
small populations of four NC mutations at baseline which were
eliminated over the course of treatment (A833S, D839Y/G,
N841K, Y842C) (64). In a recent in vitro study, gilteritinib was
active against multiple NC mutations, including mutations like
N676, which are associated with resistance to midostaurin and
quizartinib (149). Finally, in a recent case series, two patients
with non-FLT3-ITD or D835 AML were found to have JM FLT3-
V592G and KD FLT3-N676K mutations, both of which clinically
responded to sorafenib (146).
To contrast, maintenance or development of NC mutations
have also been observed in conjunction with FLT3 inhibitor
resistance. In patients treated with crenolanib, four NC FLT3
mutations (D200N, K429E, Y572C, L601F) were maintained at
time of relapse (64). In an analysis of 40 patients with FLT3-
mutated AML treatment with gilteritinib on the ADMIRAL trial,
six patients had new FLT3 mutations at time of relapse, ve of
which were in gatekeeper F691L and two at the NC site JM
E598D (62).
Many questions remain regarding the nature of NC
mutations, including their true prevalence (as they are not
detected outside of whole-exome or genome sequencing),
prognostic impact, and role in the FLT3 inhibitor susceptibility
and resistance.
Is There a Role for FLT3 Inhibitors in FLT3-
WT Disease?
Although only one-quarter of AML patients harbor FLT3
mutations, the FLT3 receptor is over-expressed on leukemic
blast regardless of mutational status (2), and early studies of
FLT3 inhibitors observed blast reduction in patients with FLT3-
WT disease (19).
Early trials of FLT3 inhibitors were not limited to patients
with FLT3-mutated AML, and results from these studies may
indicate benet in targeting FLT3-WT. In the RATIFY trial,
using an arbitrary AR cut-off of 0.7, post-hoc analysis noted a
similar OS benet withFLT3-ITD
low
, FLT3-ITD
high
, and FLT3-
TKD AML (20). Similarly, in the SORAML trial, EFS benet and
trend toward OS benet were demonstrated irrespective of FLT3
mutation status (23, 34).
Given that both midostaurin and sorafenib are multikinase
inhibitors, it is possible these results are due to inhibition of
alternative kinase-dependent pathways. For example, both
sorafenib and midostaurin also inhibit KIT, and KIT mutations
areseenin3046% of core binding factor (CBF) AML and may
impact prognosis (150, 151). The use of midostaurin in CBF AML is
currently being explored in a phase II study (NCT03686345), and a
trial of midostaurin in c-KIT or FLT3-ITD mutated t(8,21) AML
recently completed accrual (NCT01830361).
Efcacy in FLT3-WT disease has also been demonstrated in
second generation FLT3 inhibitors, which are more specicto
the FLT3 receptor. While the initial phase I/II study of gilteritinib
demonstrated strongest antileukemic effect in patients with
FLT3-mutated disease, a 12% ORR was see n in FLT3-WT
AML (52). Similarly, quizartinib monotherapy demonstrated
54 and 32% ORR in patients with FLT3-ITD and FLT3-WT
disease, respectively (152).
Trials of FLT3 inhibitors in FLT3 -WT AML are ongoing,
including a randomized phase III trial of chemotherapy +/
midostaurin (NCT03512197) and chemotherapy +/ quizartinib
(NCT04107727), both in patients with newly-diagnosed FLT3-
WT AML. In addition, many phase I trials of novel combination
therapies, dual FLT3 inhibitors, and biologic agents are not
restricted to FLT3-mutant disease.
How Should We Approach Tumor
Heterogeneity in FLT3-Mutant AML?
Increasingly, AML is understood as a heterogenous disease with
multiple genetically distinct subclones, dynamically evolving
under pressure of therapy. Clonal evolution is particularly
relevant in FLT3-mutated AML as FLT3 mutations can be
gained with disease progression, and development of a new
FLT3-ITD is an independent negative prognostic factor (153).
Developm ent of FLT3-mutated clones can arise under
targeted therapy. In patients with R/R AML treated with
venetoclax, analy sis of pre- and post-treatment sam ples
demonstrated 4/20 patients developed new FLT3-ITD clones
following therapy (94). In a larger study of 81 patients treated
with frontline venetoclax-based combinations, 5/25 patients
showed increased FLT3-ITD clonal burden at relapse, two of
which had newly acquired FLT3-ITD clones (95). Similarly, in
patients with IDH1-mutated AML treated with ivosidenib, bulk
NGS at time of progression identied multiple patients with new
FLT3-ITD or -TKD mutations not present at therapy initiation
(154), and in patients with IDH2-mutated AML treated with
enasidenib, analysis of paired pre- and post-treatment samples
described several cases with increased FLT3 variant allele
frequency at relapse (155).
Complex clonal evolution has also been observed following
FLT3 inhibition. In an analysis of patients treated with single
agent gilteritinib on the phase III ADMIRAL or phase II
CHRYSALIS trial, targeted NGS identied emerging clones with
mutations activating RAS/MAPK signaling, including NRAS and
KRAS. Serial single-cell sequencing conrmed early selection for
RAS-mutant subclones under gilteritinib pressure (63). In an
analysis of paired pre- and post-treatment samples of patients
treated with quizartinib, NRAS development was similarly noted
at relapse; furthermore, single cell sequencing conrmed these
distinct subclones existed in small populations prior to therapy
and expanded under FLT3 inhibition (66).
It is unclear how to best address clonal evolution and
associated adaptive resistance in AML. Is the treatment of
multiple clones best appro ached through blunt approaches,
such as cytotoxic chemotherapy or broad, triple-therapy
combinations, such as HMA/Venetoclax/Gilteritinib?
Alternatively, would it be more benecial to target individual
subclones sequentially, perhaps focusing on the highest-risk or
fastest-growing subclones rst? As genomic technologies such as
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288014
single cell sequencing become more widely adopted, available
genomic information will increase dramatically. L ooking
forward, innovative bioinformatic and machine learning-based
approaches may be employed to rationally treat the complex
clonal architecture in FLT3-mutated AML.
TREATING FLT3-MUTATED AML TODAY
The current standard of care for an adult with newly diagnosed
FLT3-mutated AML with AR 0.05 who is eligible for treatment
is induction and consolidation chemotherapy in combination
with midostaurin. As FLT3 AR is not universally reported, if not
available the presence of a FLT3-ITD or TKD mutation should
warrant treatment with a FLT3 inhi bitor. This is generally
followed by HCT, as the largest survival benet in the RATIFY
trial was observed in patients who underwent HCT in rst
remission (20), although whether this approach holds in
patients with low-AR FLT3-ITD, FLT3-TKD, or concomitant
NPM1 mutations is debatable.
While not standard of care, use of single-agent maintenance
FLT3 inhibitors, either in remission for patients not going to HCT
or in post-HCT remission can be considered. This would ideally be
done in a clinical trial setting, although off-label use of sorafenib or
midostaurin maintenance is routinely practiced in the US (156,
157). We await the results of ongoing clinical trials to help
determine in which settings FLT3 inhibitor maintenance is
most useful.
Should a patient with FLT3-ITD AML relapse or a patient with
FLT3-WT AML develop a new FLT3-mutated clone, then single-agent
gilteritinib is standard salvage therapy world-wide and single-agent
quizartinib could be considered in certain practice locations. If response
if achieved, HCT would be recommended in all tpatients.Ifapatient
remains refractory or develops relapse, then multiple clinical trials could
be considered, including FLT3 inhibitor combinations, novel FLT3
inhibitors, or FLT3-directed biologics (Figure 3). Although both
gilteritinib and venetoclax are FDA-approved, and there is promising
pre-clinical data on this combination (92), th is regimen can be
associated with marked myelosuppression and we would not
recommend it outside of a clinical trial. Similarly, the triplet
combination of HMA/Gilteritinib/Venetoclax should only be used as
part of clinical trial until the toxicities associated with this novel
combination are better understood.
Finally, special consideration should be given to older adults,
who may be intolerant of intensive induction chemotherapy as well
as the side effects associated with multikinase inhibitors. As
described above, multiple studies are actively investigating FLT3
inhibitors in combination with lower-intensity therapies currently
approved as frontline therapy for older adults, including HMAs as
well as liposomal cytarabine and anthracycline (CPX-351, Vyxeos).
CONCLUDING REMARKS
While this provides a baseline treatment paradig m, survi val
remains poor in FLT3-mutated AML, and additional treatment
options are needed. This includes increasing the diversity of
approved FLT3, investigating FLT3 inhibitors in new
combinations and treatment settings, and development of
novel agents. Given the diversity in FLT3 inhibitor potency,
FIGURE 3 | Current Standard of Care for Treating FLT3-mutated AML in the Fit Patient.
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288015
resistance patterns, and toxicity proles, ideally the oncologist
will have a range of FLT3-directed therapies to choose from,
similar to TKI selection in chronic myelogenous leukemia, so
that a particular FLT3 inhibitor could be matched to an
individual patients needs.
AUTHOR CONTRIBUTIONS
VK and CS both contributed to the writing of this manuscript.
All authors contributed to the article and approved the
submitted version.
REFERENCES
1. Dohner H, Weisdorf DJ, Bloomeld CD. Acute Myeloid Leukemia. N Engl J
Med (2015) 373(12):113652. doi: 10.1056/NEJMra1406184
2. Carow CE, Levenstein M, Kaufmann SH, Chen J, Amin S, Rockwell P, et al.
Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2)
in human leukemias. Bloo d (1996) 87(3):1089 96. doi: 10.1182/
blood.V87.3.1089.bloodjournal8731089
3. Gilliland DG, Grifn JD. The roles of FLT3 in hematopoiesis and leukemia.
Blood (2002) 100(5):153242. doi: 10.1182/blood-2002-02-0492
4. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts
ND, et al. Genomic Classication and Prognosis in Acute Myeloid
Leukemia. NEnglJMed(2016) 374(23):2209 21. doi: 10.10 56/
NEJMoa1516192
5. Bolouri H, Farrar JE, Triche TJr., Ries RE, Lim EL, Alonzo TA, et al. The
molecular landscape of pediatric acute myeloid leukemia reveals recurrent
structural alterations and age-specic mutational interactions. Nat Med
(2018) 24(1):10312. doi: 10.1038/nm.4439
6. Schnittger S, Bacher U, Haferlach C, Alpermann T, Kern W, Haferlach T.
Diversity of the juxtamembrane and TKD1 mutations (exons 13-15) in the
FLT3 gene with regards to mutant load, sequence, length, localization, and
correlation with biological data. Genes Chromos omes Cancer (2012) 51
(10):91024. doi: 10.1002/gcc.21975
7. Yanada M, Matsuo K, Suzuki T, Kiyoi H, Naoe T. Prognostic signicance of
FLT3 internal tandem duplication and tyrosine kinase domain mutations
for acute myeloid leukemia: a meta-analysis. Leukemia (2005) 19(8):13459.
doi: 10.1038/sj.leu.2403838
8. Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T,
et al. Diagnosis and management of AML in adults: 2017 ELN
recommendations from an international expert panel. Blood (2017) 129
(4):42447. doi: 10.1182/blood-2016-08-733196
9. Mead AJ, Linch DC, Hills RK, Wheatley K, Burnett AK, Gale RE. FLT3
tyrosine kinase domain mutations are biologically distinct from and have a
signicantly more favorable prognosis than FLT3 internal tandem
duplications in patients with acute myeloid leukemia. Blood (2007) 110
(4):126270. doi: 10.1182/blood-2006-04-015826
10. Bacher U, Haferlach C, Kern W, Haferlach T, Schnittger S. Prognostic
relevance of FLT3-TKD mutations in AML: the combination mattersan
analysis of 3082 patients. Blood (2008) 111(5):252737. doi: 10.1182/blood-
2007-05-091215
11. Tallman MS, Wang ES, Altman JK, Appelbaum FR, Bhatt VR, Bixby D, et al.
Acute Myeloid Leukemia, Version 3.2019, NCCN Clinical Practice
Guidelines in Oncology. J Natl Compr Canc Netw (2019) 17(6):72149.
doi: 10.6004/jnccn.2019.0028
12. Daver N, Schlenk RF, Russell NH, Levis MJ. Targeting FLT3 mutations in
AML: review of current knowledge and evidence. Leukemia (2019) 33
(2):299
312. doi: 10.1038/s41375-018-0357-9
13. Levis MJ, Perl AE, Altman JK, Gocke CD, Bahceci E, Hill J, et al. A next-
generation sequencing-based assay for minimal residual disease assessment
in AML patients with FLT3-ITD mutations. Blood Adv (2018) 2(8):82531.
doi: 10.1182/bloodadvances.2018015925
14. Spencer DH, Abel HJ, Lockwood CM, Payton JE, Szankasi P, Kelley TW,
et al. Detection of FLT3 internal tandem duplication in targeted, short-read-
length, next-generation sequencing data. J Mol Diagn (2013) 15(1):8193.
doi: 10.1016/j.jmoldx.2012.08.001
15. Bo lli N, Manes N, McKerrell T, Chi J, Park N , Gundem G, et al.
Characterization of gene mutations and copy number changes in acute
myeloid leukemia using a rapid target enrichment protocol. Haematologica
(2015) 100(2):21422. doi: 10.3324/haematol.2014.113381
16. Au CH, Wa A, Ho DN, Chan TL, Ma ES. Clinical evaluation of panel testing
by next -generation sequencing (NGS) for gene mutations in myeloid
neoplasms. Diagn Pathol (2016) 11:11. doi: 10.1186/s13000-016-0456-8
17. Schlenk RF, Kayser S, Bullinger L, Kobbe G, Casper J, Ringhoffer M, 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
9. doi: 10.1182/blood-2014-05-578070
18. George TI, Tworek JA, Thomas NE, Fatheree LA, Souers RJ, Nakhleh RE,
et al. Evaluation of Testing of Acute Leukemia Samples: Survey Result From
the College of American Pathologists. Arch Pathol Lab Med (2017) 141
(8):11016. doi: 10.5858/arpa.2016-0398-CP
19. Fischer T, Stone RM, Deangelo DJ, Galinsky I, Estey E, Lanza C, 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):433945. doi: 10.1200/
JCO.2010.28.9678
20. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomeld CD,
et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a
FLT3 Mutation. NEnglJMed(2017) 377(5):45464. doi: 10.1056/
NEJMoa1614359
21. Novartis Pharmaceuticals Corporation. RYDAPT
®
product monograph. East
Hanover, New Jersey (2018).
22. Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY,
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(11):185662. doi: 10.1200/JCO.2009.25.4888
23. Rollig C, Serve H, Huttmann A, Noppeney R, Muller-Tidow C, Krug U, 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(16):16919. doi: 10.1016/S1470-2045(15)00362-9
24. Astellas Pharma US Inc. XOSPATA
®
product monograph . Northbrook,
Illinois, USA (2019).
25. Cortes J, Perl AE, Dohner H, Kantarjian H, Martinelli G, Kovacsovics T,
et al. Quizartinib, an FLT3 inhibitor, as monotherapy in patients with
relapsed or refractory acute m yeloid leukaemia: an open-label,
multicentre, single-arm, phase 2 trial. Lancet Oncol (2018) 19(7):889903.
doi: 10.1016/S1470-2045(18)30240-7
26. Cortes JE, Khaled S, Martinelli G, Perl AE, Ganguly S, Russell N, 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):98497.
doi: 10.1016/S1470-2045(19)30150-0
27. Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al.
Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated
AML. N Engl J Med (2019) 381(18):172840. doi: 10.1056/NEJMoa1902688
28. Inc. APU. XOSPATA
®
product monograph. Illinoise, USA: Northbrook (2019).
29. Cortes JE, Kantarzian HM, Kadia TM, Borthakur G, Konopleva M, Garcia-
Manero G, et al. Crenolanib besylate, a type I pan-FLT3 inhibitor, to
demonstrate clinical activity in multiply relapsed FLT3-ITD and D835
AML. JClinOncol(2016) 34(15_suppl):70087008. doi: 10.1200/
JCO.2016.34.15_suppl.7008
30. Randhawa JK KH, Borthakur G, Thompson PA, Konopleva M, Daver N,
et al. Results of a Phase II Study of Crenolanib in Relapsed/Refractory Acute
Myeloid Leukemia Patients (Pts) with Activating FLT3 Mutations. Blood
(2014) 124(21):389. doi: 10.1182/blood.V124.21.389.389
31. Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD, et al.
Patients with acute myeloid leukemia and an activating mutation in FLT3
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288016
respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood
(2005) 105(1):5460. doi: 10.1182/blood-2004-03-0891
32. Schlenk RF, Fiedler W, Salih HR, Wulf G, Thol F, Kundgen A, 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. doi: 10.1182/blood.V128.22.449.449
33. Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U, 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):31108. doi: 10.1200/JCO.2012.46.4990
34. Rollig C, Serve H, Huttmann A, Noppeney R, Muller-Tidow C, Krug U, et al.
The addition of sorafenib to standard AML treatment results in substantial
reduction in relapse risks and improved survival. Updated results from long-
term follow-up of the randomized-controlled SORAML trial. Blood (2017)
130:7211. doi: 10.1182/blood.V130.Suppl_1.721.721
35. Crump M, Hedley D, Kamel-Reid S, Leber B, Wells R, Brandwein J, 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(2):25260. doi: 10.3109/10428190903585286
36. Borthakur G, Kantarjian H, Ravandi F, Zhang W, Konopleva M, Wright JJ, et al.
Phase I study of sorafenib in patients with refractory or relapsed acute leukemias.
Haematolo gica (2011) 96(1):628. doi: 10.3324/haematol.2010.030452
37. Metzelder SK, Schroeder T, Finck A, Scholl S, Fey M, Gotze K, et al. High
activity of sorafenib in FLT3-ITD-positive acute myeloid leukemia
synergizes with allo-immune effects to induce sustained responses.
Leukemia (2012) 26(11):23539. doi: 10.1038/leu.2012.105
38. De Freitas T, Marktel S, Piemontese S, Carrabba MG, Tresoldi C, Messina C,
et al. High rate of hematological responses to sorafenib in FLT3-ITD acute
myeloid leukemia relapsed after allogeneic hematopoietic stem cell
transplantation. Eur J Haematol (2016) 96(6):629 36. doi: 10.1111/ejh.12647
39. Tschan-Plessl A, Halter JP, Heim D, Medinger M, Passweg JR, Gerull S.
Synergistic effect of sorafenib and cGvHD in patients with high-risk FLT3-
ITD+AML allows long-term disease control after allogeneic transplantation.
Ann Hematol (2015) 94(11):1899905. doi: 10.1007/s00277-015-2461-5
40. Metzelder SK, Schroeder T, Lubbert M, Ditschkowski M, Gotze K, Scholl S, 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) 86:2339. doi: 10.1016/j.ejca.2017.09.016
41. FiedlerW,ServeH,DohnerH,SchwittayM,OttmannOG,OFarre ll AM, et al. A
phase 1 study of SU11248 in the treatment of patients with refractory or resistant
acute myeloid leukemia (AML) or not amenable to conventional therapy for the
disease. Blood (2005) 105(3):98693. doi: 10.1182/blood-2004-05-1846
42. Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S, et al. Results from
a randomized trial of salvage chemotherapy followed by lestaurtinib for
patients with FLT3 mutant AML in rst relapse. Blood (2011) 117(12):3294
301. doi: 10.1182/blood-2010-08-301796
43. DeAngelo DJ, Stone RM, Heaney ML, Nimer SD, Paquette RL, Klisovic RB, et al.
Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in
patients with acute myelogenous leukemia or high-risk myelodysplastic
syndrome: safety, pharmacoki netics, and pharmacodynamics. Blood (2006) 108
(12):367481. doi: 10.1182/blood-2006-02-005702
44. Fiedler W, Kayser S, Kebenko M, Janning M, Krauter J, Schittenhelm M,
et al. A phase I/II study of sunitinib and intensive chemotherapy in patients
over 60 years of age with acute myeloid leukaemia and activating FLT3
mutations. Br J Haematol (2015) 169(5):694700. doi: 10.1111/bjh.13353
45. Knapper S, Russell N, Gilkes A, Hills RK, Gale RE, Cavenagh JD, et al. A
randomized assessment of adding the kinase inhibitor lestaurtinib to rst-
line chemotherapy for FLT3-mutated AML. Blood (2017) 129(9):114354.
doi: 10.1182/blood-2016-07-730648
46. Zarrinkar PP, Gunawardane RN, Cramer MD, Gardner MF, Brigham D,
Belli B, et al. AC220 is a uniquely potent and selective inhibitor of FLT3 for
the treatment of acute myeloid leukemia (AML). Blood (2009) 114
(14):298492. doi: 10.1182/blood-2009-05-222034
47. Cortes JE, Tallman MS, Schiller GJ, Trone D, Gammon G, Goldberg SL, et al.
Phase 2b study of 2 dosing regimens of quizartinib monotherapy in FLT3-
ITD-mutated, relapsed or refractory AML. Blood (2018) 132(6):598607.
doi: 10.1182/blood-2018-01-821629
48. 21266 N. Quizartininb. (2019). FDA Brieng Document of the Oncologic
Drugs Advisory Committee (ODAC).
49. Ueno Y, Kaneko N, Saito R, Kondoh Y, Shimada I, Mori M, et al. ASP2215, a
novel FL T3/AXL inhibitor: Preclinical evaluation in combinatio n with
cytarabine and anthracycline in acute myeloid leukemia (AML). J Clin
Oncol (2014) 3 2(15_suppl):7071 7071. doi: 10.1200/jco.2014 .32.
15_suppl.7071
50.SmithCC,ZhangC,LinKC,LasaterEA,ZhangY,MassiE,etal.
Characterizing and Overriding the Structural Mechanism of the
Quizartinib-Resistant FLT3 Gatekeeper F691L Mutation with PLX3397.
Cancer Discov (2015) 5(6):66879. doi: 10.1158/2159-8290.CD-15-0060
51. Baker SD, Zimmerman EI, Wang YD, Orwick S, Zatechka DS, Buaboonnam J,
et al. Emergence of polyclonal FLT3 tyrosine kinase domain mutations during
sequential therapy with sorafenib and sunitinib in FLT3-ITD-positive acute
myeloid leukemia. Clin Cancer Res (2013) 19(20):575868. doi: 10.1158/1078-
0432.CCR-13-1323
52. Perl AE, Altman JK, Cortes J, Smith C, Litzow M, Baer MR, et al. Selective
inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid
leukaemia: a multicentre, rst-in-human, open- labe l, phase 1-2 study.
Lancet Oncol (2017) 18(8):106175. doi: 10.1016/S1470-2045(17)30416-3
53. Pratz KCM, Altman JK, Cooper B, Cruz JC, Juric JG, Levis MJ, et al. Updated
Results from a Phase 1 Study of Gilteritininb in Combination with Induction
and Consolidation Chemotherapy in Subjects with Newly Diagnosed Acute
Myeloid L eukemia (AML). Blood (2018) 132(Supplement 1):564. doi:
10.1182/blood-2018-99-110975
54. Galanis A, Ma H, Rajkhowa T, Ramachandran A, Small D, Cortes J, et al.
Crenolanib is a potent inhibitor of FLT3 with activity against resistance-
conferring point mutants. Blood (2014) 123(1):94100. doi: 10.1182/blood-
2013-10-529313
55. Goldberg AD, Coombs CC, Wang ES, Walter RB, Karanes C, Vigil CE, 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(Supplement_1):13266.
doi: 10.1182/blood-2019-130863
56. Wang ES, Grifths EA, Walter RB, Tallman MS, Goldberg AD, Messahel B,
et al. Tolerability and Efcacy 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
(Supplement_1):38299. doi: 10.1182/blood-2019-130536
57. Leung AY, Man CH, Kwong YL. FLT3 inhibition: a moving and evolving
target in acute myeloid leukaemia. Leukemia (2013) 27(2):2608.
doi: 10.1038/leu.2012.195
58. DaverN,CortesJ,RavandiF,PatelKP,BurgerJA,KonoplevaM,etal.Secondary
mutations as mediators of resistance to targeted therapy in leukemia. Blood
(2015) 125(21):323645. doi: 10.1182/blood-2014-10-605808
59. Cools J, Mentens N, Furet P, Fabbro D, Clark JJ, Grifn JD, et al. Prediction
of resistance to small molecule FLT3 inhibitors: implications for molecularly
targeted therapy of acute leukemia. Cancer Res (2004) 64(18):63859.
doi: 10.1158/0008-5472.CAN-04-2148
60. Lee LY, Hernandez D, Rajkhowa T, Smith SC, Raman JR, Nguyen B, et al.
Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor. Blood
(2017) 129(2):25760. doi: 10.1182/blood-2016-10-745133
61. Smith CC, Lasater EA, Lin KC, Wang Q, McCreery MQ, Stewart WK, et al.
Crenolanib is a selective type I pan-FLT3 inhibitor. Proc Natl Acad Sci U S A
(2014) 111(14):531924. doi: 10.1073/pnas.1320661111
62. Smith CC, Levis MJ, Perl AE, Martinelli G, Neubauer A, Berman E, et al.
Emerging Mutations at Relapse in Patients with FLT3-Mutated Relapsed/
Refractory Acute Myeloid Leukemia Who Received Gilteritinib Therapy in
the Phase 3 Admiral Trial. Blood (2019) 134(Supplement_1):144.
doi: 10.1182/blood-2019-122620
63. McMahon CM, Ferng T, Canaani J, Wang ES, Morrissette JJD, Eastburn DJ,
et al. Clonal Selection with RAS Pathway Activation Mediates Secondary
Clinical Resistance to Selective FLT3 Inhibition in Acute Myeloid Leukemia.
Cancer Discov (2019) 9(8):105063. doi: 10.1158/2159-8290.CD-18-1453
64. Zhang H, Savage S, Schultz AR, Bottomly D, White L, Segerdell E, et al.
Clinical resistance to crenolanib in acute myeloid leukemia due to diverse
molecular mechanisms. Nat Commun (2019) 10(1):244. doi: 10.1038/
s41467-018-08263-x
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288017
65. Smith CC, Wang Q, Chin CS, Salerno S, Damon LE, Levis MJ, et al. Validation
of ITD mutations in FLT3 as a therapeutic target in human acute myeloid
leukaemia. Nature (2012) 485(7397):2603. doi: 10.1038/nature11016
66. Cohler Peretz CA ML, Kumar TF, Jackson LH, Jacob J, Durruthy R, et al.
Single Cell Sequencing Reveals Evolution of Tumor Heterogeneity of Acute
Myeloid Leukemia on Quizartinib. Blood (2019) 134(Supplement_1):1440.
doi: 10.1182/blood-2019-132105
67. Piloto O, Wright M, Brown P, Kim KT, Levis M, Small D. Prolonged
exposure to FLT3 inhibitors leads to resistance via activation of parallel
signaling pathways. Blood (2007) 109(4):164352. doi: 10.1182/blood-2006-
05-023804
68. Lindblad O, Cordero E, Puissant A, Macaulay L, Ramos A, Kabir NN, et al.
Aberrant activation of the PI3K/mTOR pathway promotes resistance to
sorafenib in AML. Oncogene (2016) 35(39):511931. doi: 10.1038/
onc.2016.41
69. Kasi PM, Litzow MR, Patnaik MM, Hashmi SK, Gangat N. Clonal evolution
of AML on novel FMS-like tyrosine kinase-3 (FLT3) inhibitor therapy with
evolving actionable targets. Leuk Res Rep (2016) 5:710. doi: 10.1016/
j.lrr.2016.01.002
70. Park IK, Mundy-Bosse B, Whitman SP, Zhang X, Warner SL, Bearss DJ,
et al. Receptor tyrosine kinase Axl is required for resistance of leukemic cells
to FLT3-targeted therapy in acute myeloid leukemia. Leukemia (2015) 29
(12):23829. doi: 10.1038/leu.2015.147
71. Jeon JY, Garrison D, Thomas MZ, Buelow D, Whatcott C, Warner S, et al. Activity of
TP-0903 in FLT3 inhibitor resistant AML models. FASEB J (2019) 33
(1_supplement):675.4-675.4. doi: 10.1096/fasebj.2019.33.1_supplement.675.4
72. Sato T, Yang X, Knapper S, White P, Smith BD, Galkin S, et al. FLT3 ligand
impedes the efcacy of FLT3 inhibitors in vitro and in vivo. Blood (2011) 117
(12):328693. doi: 10.1182/blood-2010-01-266742
73. 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(1):6172.
doi: 10.1111/bjh.12599
74. Chang YT HD, Ghiaur G, Levis MJ, Jones RJ. Bone Marrow Stroma Protects
FLT3 Acute Myeloid Leukemia (AML) through CYP3A4-Mediated Drug
Metabolization of FLT3 Tyrosine Kinase Inhibitors (TKIs). Blood (2017) 130
(Supplement 1):2519. doi: 10.1182/blood.V130.Suppl_1.2519.2519
75. Dutreix C, Munarini F, Lorenzo S, Roesel J, Wang Y. Investigation into
CYP3A4-mediated drug-drug interactions on midostaurin in healthy
volunteers. Cancer Chemother Pharmacol (2013) 72(6):1223 34.
doi: 10.1007/s00280-013-2287-6
76. Traer E, Martinez J, Javidi-Shari N, Agarwal A, Dunlap J, English I, et al.
FGF2 from Marrow Microenvironment Promotes Resistance to FLT3
Inhibitors in Acute Myeloid Leukemia. Cancer Res (2016) 76(22):647182.
doi: 10.1158/0008-5472.CAN-15-3569
77. Javidi-Shari N,MartinezJ,EnglishI,JoshiSK,Scopim-RibeiroR,ViolaSK,etal.
FGF2-FGFR1 signaling regulates release of Leukemia-Protective exosomes from
bone marrow stromal cells. Elife (2019) 8:8. doi: 10.7554/eLife.40033
78. Wang Y BH, Zhang N, Wang F, He Y, Wu T. Preclinical Evaluation of MAX-
40279, a FLT3/FGFR Dual Kinase Inhibitor for Treatment of Acute Myeloid
Leukemia. BLood (2018) 132(Supplement 1):3997. doi: 10.1182/blood-2018-
99-110414
79. Wang Y, Zhang L, Tang X, Luo J, Tu Z, Jiang K, et al. GZD824 as a FLT3,
FGFR1 and PDGFRalpha Inhibitor Against Leukemia In Vitro and In Vivo.
Transl Oncol (2020) 13(4):100766. doi: 10.1016/j.tranon.2020.100766
80. Gozgit JM, Wong MJ, Wardwell S, Tyner JW, Loriaux MM, Mohemmad
QK, et al. Potent activity of ponatinib (AP24534) in models of FLT3-driven
acute myeloid leukemia and other hematologic malignancies. Mol Cancer
Ther (2011) 10(6):102835. doi: 10.1158/1535-7163.MCT-10-1044
81. Chang E, Ganguly S, Rajkhowa T, Gocke CD, Levis M, Konig H. The
combination of FLT3 and DNA methyltransferase inhibition is
synergistic ally cytotox ic to FLT3/ITD acute myeloid leukemia cells.
Leukemia (2016) 30(5):102532. doi: 10.1038/leu.2015.346
82. Williams CB, Kambhampati S, Fiskus W, Wick J, Dutreix C, Ganguly S, et al.
Preclinical and phase I results of decitabine in combination with
midostaurin (PKC412) for newly diagnosed elderly or relapsed/refractory
adult patients with acute myeloid leukemia. Pharmacotherapy (2013) 33
(12):134152. doi: 10.1002/phar.1316
83. Tomlinson BK, Gallogly MM, Kane DM, Metheny L, Lazarus HM, William
BM, et al. A Phase II Study of Midostaurin and 5-Azacitidine for Untreated
Elderly and Unt Patients With FLT3 Wild-type Acute Myelogenous
Leukemia. Clin Lymphoma Myeloma Leuk (2020) 20( 4):22633.e1.
doi: 10.1016/j.clml.2019.10.018
84. Strati P, Kantarjian H, Ravandi F, Nazha A, Borthakur G, Daver N, et al.
Phase I/II trial of the combination of m idostaurin (PKC412) and 5-
azacytidine for patients with acute myeloid leukemia and myelodysplastic
syndrome. Am J Hematol (2015) 90(4):27681. doi: 10.1002/ajh.23924
85. Ravandi F, Alattar ML, Grunwald MR, Rudek MA, Rajkhowa T, Richie MA,
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(23):465562. doi: 10.1182/blood-2013-01-480228
86. Muppidi MR GE, Thompson JE, Ford LA, Freyer CW, Wetzler M, Wang ES.
Decitabine and Sorafenib Therapy in Patients with FLT3-ITD Mutant Acute
Myeloid Leukemia Is Associated with High Response RatesaSingle
Institute Experience. Blood (2014) (12421):5284. doi: 10.1182/blood.
V124.21.5284.5284
87. Ohanian M, Garcia-Manero G, Levis M, Jabbour E, Daver N, Borthakur G,
et al. Sorafenib Combined with 5-azacytidine in Older Patients with
Untreated FLT3-ITD Mutated Acute Myeloid Leukemia. Am J Hematol
(2018) 93(9):113641. doi: 10.1002/ajh.25198
88. Swaminathan M, Kantarjian H, Daver N, Borthakur G, Ohanian M, Kadia T,
et al. The Combin ation of Quizartinib with Azacit idine 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
(Supplement 1):723. doi: 10.1182/blood.V130.Suppl_1.723.723
89. Esteve J SR, Del Castillo TB, Lee J, Wang E, Dinner S, Minden M, et al.
Open-label study of gilteritinib, gilteritinib plus azacitidine, or azacitidine
alone in newly diagnosed FLT3-mutated AML patients ineligible for
intensive chemotherapy: results from the safety cohort. In: European
Hematology Association Annual Meeting. EHA Library (2019). 266682;
PS1065. doi: 10.1097/01.HS9.0000562556.40615.f8
90. Bagrintseva K, Geisenhof S, Kern R, Eichenlaub S, Reindl C, Ellwart JW,
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(9):367985. doi: 10.1182/blood-2004-06-2459
91. Kohl TM, Hellinger C, Ahmed F, Buske C, Hiddemann W, Bohlander SK,
et al. BH3 mimetic ABT-737 neutralizes resist ance to FLT3 inhibitor
treatment mediated by FLT3-independent expression of BCL2 in primary
AML blasts. Leukemia
(2007) 21(8):176372. doi: 10.1038/sj.leu.2404776
92. Ma J, Zhao S, Qiao X, Knight T, Edwards H, Polin L, et al. Inhibition of Bcl-2
Synergistically Enhances the Antileukemic Activity of Midostaurin and
Gilteritinib in Preclinical Models of FLT3-Mutated Acute Myeloid Leukemia.
Clin Cancer Res (2019) 25(22):681526. doi: 10.1158/1078-0432.CCR-19-0832
93. Mali RS, Zhang Q, DeFilippis R, Cavazos A, Kuruvilla VM, Raman J, et al.
Venetoclax combines synergistically with FLT3 inhibition to effectively
target leukemic cells in FLT3-ITD+ acute myeloid leukemia models.
Haematologic a (2020) haematol.2019.244020. doi: 10.3324/haematol.
2019.244020
94. Chyla B, Daver N, Doyle K, McKeegan E, Huang X, Ruvolo V, et al. Genetic
Biomarkers Of Sensitivity and Resistance to Venetoclax Monotherapy in
Patients With Relapsed Acute Myeloid Leukemia. Am J Hematol (2018) 93
(8):E2025. doi: 10.1002/ajh.25146
95. DiNardo CD, Tiong IS, Quaglieri A, MacRaild S, Loghavi S, Brown FC, et al.
Molecular patterns of response and treatment failure after frontline
venetoclax combinations in older patients with AML. Blood (2020) 135
(11):791803. doi: 10.1182/blood.2019003988
96. Larrue C, Saland E, Boutzen H, Vergez F, David M, Joffre C, et al.
Proteasome inhibitors induce FLT3-ITD degradation through autophagy
in AML cells. Blood (2016) 127(7):88292. doi: 10.1182/blood-2015-05-
646497
97. Walker AR, Wang H, Walsh K, Bhatnagar B, Vasu S, Garzon R, et al.
Midostaurin, bortezomib and MEC in relapsed/refractory acute myeloid
leukemia. Leuk Lymphoma (2016) 57(9):21008. doi: 10.3109/10428194.
2015.1135435
98. Saliba AN, Boswell HS, Cripe LD, Abu Zaid MI, Weisenbach J, Sayar H.
Final Results of Phase I/II Study of Combination of Sorafenib, Vorinostat,
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288018
and Bortezomib in Acute Myeloid Leukemia with FLT3-ITD Mutation or
Poor-Risk Cytogenetics . Blood (2017) 130(Supplement 1):3897 7.
doi: 10.1182/blood.V130.Suppl_1.3897.3897
99. Kim KT, Baird K, Ahn JY, Meltzer P, Lilly M, Levis M, et al. Pim-1 is up-
regulated by constitutively activated FLT3 and plays a role in FLT3-mediated
cell survival. Blood (2005) 105(4):175967. doi: 10.1182/blood-2004-05-2006
100. Natarajan K, Xie Y, Burcu M, Linn DE, Qiu Y, Baer MR. 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(9):e74653. doi: 10.1371/journal.pone.0074653
101. Kapoor S, Natarajan K, Baldwin PR, Doshi KA, Lapidus RG, Mathias TJ,
et al. Concurrent Inhibition of Pim and FLT3 Kinases Enhances Apoptosis of
FLT3-ITD Acute Myeloid Leukemia Cells through Increased Mcl-1
Proteaso mal Degrad ation. Clin Cancer Res (2018) 24(1):234 47.
doi: 10.1158/1078-0432.CCR-17-1629
102. Jeon JY, Zhao Q, Buelow DR, Phelps M, Walker AR, Mims AS, et al.
Preclinical activity and a pilot phase I study of pacritinib, an oral JAK2/FLT3
inhibitor, and chemotherapy in FLT3-ITD-positive AML. Invest New Drugs
(2020) 38(2):3409. doi: 10.1007/s10637-019-00786-4
103. Mohi MG, Boulton C, Gu TL, Sternberg DW, Neuberg D, Grifn JD, et al.
Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for
the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci
USA(2004) 101(9):31305. doi: 10.1073/pnas.0400063101
104. Wang F, Liu Z, Zeng J, Zhu H, Li J, Cheng X, et al. Metformin synergistically
sensitizes FLT3-ITD-positive acute myeloid leukemia to sorafenib by
promoting mTOR-mediated apoptosis and autophagy. Leuk Res (2015) 39
(12):14217. doi: 10.1016/j.leukres.2015.09.016
105. Kollmann K, Heller G, Schneckenleithner C, Warsch W, Scheicher R, Ott
RG, et al. A Kinase-Independent Function of CDK6 Links the Cell Cycle to
Tumor Angiogenesis. Cancer Cell (2016) 30(2):35960. doi: 10.1016/
j.ccell.2016.07.003
106. Uras IZ, Walter GJ, Scheicher R, Bellutti F, Prchal-Murphy M, Tigan AS,
et al. Palbociclib treatment of FLT3-ITD+ AML cells uncovers a kinase-
dependent transcriptional regulation of FLT3 and PIM1 by CDK6. Blood
(2016) 127(23):2890902. doi: 10.1182/blood-2015-11-683581
107. Keegan K, Li C, Li Z, Ma J, Ragains M, Coberly S, et al. Preclinical evaluation
of AMG 925, a FLT3/CDK4 dual kinase inhibitor for treating acute myeloid
leukemia. Mol Cancer Ther (2014) 13(4):8809 . doi: 10.115 8/1535-
7163.MCT-13-0858
108. Liu C LB, Xu C, Zhang P, Li B, Ji B, Zhang B, et al. ETH-155008, a Novel
Selective Dual Inhibitor of FLT3 and CDK4/6 in Preclinical Treatment of
Acute Myeloid Leuke mia. Blood (2019) 134(Suppleme nt_1):5141. doi:
10.1182/blood-2019-123589
109. Daver N, Pollyea DA, Rizzieri DA, Palmer J, Rampal RK, Dinner S, et al. A
Phase I Study of FLX925, a Dual FLT3 and CDK4/6 Inhibitor in Patients
with Relapsed or Refractory Acute Myeloid Leukemia (AML). Blood (2017)
130(Supplement 1):1343. doi: 10.1182/blood.V130.Suppl_1.1343.1343
110. Andreeff M ZW, Kumar P, Zernovak O, Daver NG, Isoyama T, Iwanaga K,
et al. Synergistic Anti-Leukemic Activity with Combination of FLT3
Inhibitor Quizartinib and MDM2 Inhibitor Milademet an in FLT3-ITD
Mutant/p53 Wild-Type Acute Myeloid Leukemia Models. Blood (2018)
132(Supplement 1):2720. doi: 10.1182/blood-2018-99-115183
111. Brodska B, Otevrelova P, Salek C, Fuchs O, Gasova Z, Kuzelova K. High PD-
L1 Expression Predicts for Worse Outcome of Leukemia Patients with
Concomitant NPM1 and FLT3 Mutations. Int J Mol S ci (2019) 20
(11):2823. doi: 10.3390/ijms20112823
112. Smith CC, Lasater EA, Zhu X, Lin KC, Stewart WK, Damon LE, et al. Activity of
ponatinib against clinically-relevant AC220-resistant kinase domain mutants of
FLT3-ITD. Blood (2013) 121(16):316571. doi: 10.1182/blood-2012-07-442871
113. Shah NP, Talpaz M, Deininger MW, Mauro MJ, Flinn IW, Bixby D, et al.
Ponatinib in patients with refractory acute myeloid leukaemia: ndings from
a phase 1 study. Br J Haematol (2013) 162(4):54852. doi: 10.1111/bjh.12382
114. Lu JW, Wang AN, Liao HA, Chen CY, Hou HA, Hu CY, et al. Cabozantinib
is selectively cytotoxic in acute myeloid leukemia cells with FLT3-internal
tandem duplication ( FLT3-ITD). Cancer Lett (2016) 376(2) :21825.
doi: 10.1016/j.canlet.2016.04.004
115. Fathi AT, Blonquist TM, Hernandez D, Amrein PC, Ballen KK, McMasters
M, et al. Cabozantinib is well tolerated in acute myeloid leukemia and
effectively inhibits the resistance-conferring FLT3/tyrosine kinase domain/
F691 mutation. Cancer (2018) 124(2):30614. doi: 10.1002/cncr.31038
116. Smith CC, Levis MJ, Frankfurt O, Pagel JM, Roboz GJ, Stone RM, et al. A
phase 1/2 study of the oral FLT3 inhibitor pexidartinib in relapsed/refractory
FLT3-ITD-mutant acute myeloid leukemia. Blood Adv (2020) 4(8):171121.
doi: 10.1182/bloodadvances.2020001449
117. Wu H, Hu C, Wang A, Weisberg EL, Wang W, Chen C, et al. Ibrutinib
selectively targets FLT3-ITD in mutant FLT3-positive AML. Leukemia
(2016) 30(3):7547. doi: 10.1038/leu.2015.175
118. Yamaura T, Nakatani T, Uda K, Ogura H, Shin W, Kurokawa N, et al. A
novel irreversible FLT3 inhibitor, FF-10101, shows excellent efcacy against
AML cells with FLT3 mutations. Blood (2018) 131(4):42638. doi: 10.1182/
blood-2017-05-786657
119. Ma HS, Nguyen B, Dufeld AS, Li L, Galanis A, Williams AB, et al. FLT3
kinase inhibitor TTT-3002 overcomes both activating and drug resistance
mutations in FLT3 in acute myeloid leukemia. Cancer Res (2014) 74
(18):520617. doi: 10.1158/0008-5472.CAN-14-1028
120. Lee HK, Kim HW, Lee IY, Lee J, Lee J, Jung DS, et al. G-749, a novel FLT3
kinase inhibitor, can overcome drug resistance for the treatment of acute
myeloid leukemia. Blood (2014) 123(14):220919. doi: 10.1182/blood-2013-
04-493916
121. Xu B, Zhao Y, Wang X, Gong P, Ge W. MZH29 is a novel potent inhibitor
that overcomes drug resistance FLT3 mutations in acute myeloid leukemia.
Leukemia (2017) 31(4):91321. doi: 10.1038/leu.2016.297
122. Daver NG LK, Yoon S, Jung CW, Kang HJ, Jung S, Seo S, et al. HM43239, a
Novel Potent Small Molecule FLT3 Inhibitor, in Acute Myeloid Leukemia
(AML) with FMS-like Tyrosine Kinase 3 (FLT3) Mutations: Phase 1 /2 Study.
Blood (2019) 134(Supplement_1):1331. doi: 10.1182/blood-2019-129670
123. Kayser S HJ, Dörfel D, Thol F, Heuser M, Märklin M, Müller-Tidow C, et al.
Interim Results of a First in Man Study with the Fc-Optimized FLT3
Antibody Flysyn for Treatment of Acute Myeloid Leukemia with Minimal
Residual Disease. Blood (2019) 134(Supplement_1):3928. doi: 10.1182/
blood-2019-125327
124. Rudra-Ganguly N LC, Virata C, Leavitt M, Jin L, Mendelsohn B, Snyder J,
et al. AGS62P1, a Novel Anti-FLT3 Antibody Drug Conjugate, Employing
Site Specic Conjugation, Demonstrates Preclinical Anti-Tumor Efcacy in
AML Tumor and Patient Derived Xenografts. Blood
(2015) 12623):3806. doi:
10.1182/blood.V126.23.3806.3806
125. Yeung YA, Krishnamoorthy V, Dettling D, Sommer C, Poulsen K, Ni I, et al.
An Optimized Full-Length FLT3/CD3 Bispecic Antibody Demonstrates
Potent Anti-leukemia Activity and Reversible Hematological Toxicity. Mol
Ther (2020) 28(3):889900. doi: 10.1016/j.ymthe.2019.12.014
126. Wang Y, Xu Y, Li S, Liu J, Xing Y, Xing H, et al. Targeting FLT3 in acute
myeloid leukemia using ligand-based chimeric antigen receptor-engineered
T cells. J Hematol Oncol (2018) 11(1):60. doi: 10.1186/s13045-018-0603-7
127. Jetani H, Garcia-Cadenas I, Nerreter T, Thomas S, Rydzek J, Meijide JB, et al.
CAR T-cells targeting FLT3 have potent activity against FLT3(-)ITD(+)
AML and act synergistically with the FLT3-inhibitor crenolanib. Leukemia
(2018) 32(5):116879. doi: 10.1038/s41375-018-0009-0
128. Larson R, Mandrekar S, Sanford BL, Laumann K, Geyer SM, Bloomeld CD,
et al. An Analysis of Maintenance Therapy and Post-Midostaurin Outcomes
in the International Prospective Randomized, Placebo-Controlled, Double-
Blind Trial (CALGB 10603/RATIFY [Alliance]) for Newly Diagnosed Acute
Myeloid Leukemia (AML) Patients with FLT3 Mutations. Blood (2017)
2017130(Supplement 1):145. doi: 10.1182/blood.V130.Suppl_1.145.145
129. Schlenk RF, Weber D, Fiedler W, Salih HR, Wulf G, Salwender H, et al.
Midostaurin added to chemo therapy and continued single-agent
maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood
(2019) 133(8):84051. doi: 10.1182/blood-2018-08-869453
130. Maziarz RTT PM, Scott BL, Mohan SR, Deol A, Rowley SD, Kim D, et al.
Radius: A Phase 2 Randomized Trial Investigating Standard of Care ±
Midostaurin after Allogeneic Stem Cell Transplant in FLT3-ITD-Mutated
AML. Blood (2018) 132(Supplement 1):662. doi: 10.1182/b lood-2018-99-113582
131.BrunnerAM,LiS,FathiAT,WadleighM,HoVT,CollierK,etal.
Haematopoietic cell transplantation with and without sorafenib
maintenance for patients with FLT3-ITD acute myeloid leukaemia in rst
complete remission. Br J Haematol (2016) 175(3):496504. doi: 10.1111/
bjh.14260
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288019
132. Chen YB, Li S, Lane AA, Connolly C, Del Rio C, Valles B, 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(12):20428. doi: 10.1016/
j.bbmt.2014.09.007
133. Burchert A, Bug G, Fritz LV, Finke J, Stelljes M, Rollig C, et al. Sorafenib
Maintenance After Allogeneic Hematopoietic Stem Cell Transplantation for
Acute Myeloid Leukemia With FLT3-Internal Tandem Duplication Mutation
(SORMAIN). JClinOncol(2020) 38(26):29933002. doi: 10.1200/
JCO.19.03345
134. Sandmaier BM, Khaled S, Oran B, Gammon G, Trone D, Frankfurt O. 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):22231. doi: 10.1002/ajh.24959
135. Maziarz RT, Fernandez H, Patnaik MM, Scott BL, Mohan S, Deol A, et al.
Radius: Midostaurin (mido) Plus Standard of Care (SOC) after Allogeneic
Stem Cell Transplant (alloSCT) in Patients (pts) with FLT3-Internal Tandem
Duplication (ITD)Mutated Acute Myeloid Leukemia (AML). Biol Blood
Marrow Transplant (2019) 25(3):S112. doi: 10.1016/j.bbmt.2018.12.077
136. Bornhauser M, Illmer T, Schaich M, Soucek S, Ehninger G, Thiede C, et al.
Improved outcome after stem-cell transplantation in FLT3/ITD-positive
AML. Blood (2007) 109(5):22645. doi: 10.1182/blood-2006-09-047225
137. DeZern AE, Sung A, Kim S, Smith BD, Karp JE, Gore SD, 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(9):14049. doi: 10.1016/j.bbmt.2011.02.003
138. Dohner K, Thiede C, Jahn N, Panina E, Gambietz A, Larson RA, et al. Impact
of NPM1/FLT3-ITD genotypes dened by the 2017 European LeukemiaNet
in patients with acute m yeloid leukemia. Blood (2020) 135(5):37180.
doi: 10.1182/blood.2019002697
139. Sakaguchi M, Yamaguchi H, Najima Y, Usuki K, Ueki T, Oh I, et al.
Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in
acute myeloid leukemia. Blood Adv (2018) 2(20):274454. doi: 10.1182/
bloodadvances.2018020305
140. Oran B, Cortes J, Beitinjaneh A, Chen HC, de Lima M, Patel K, et al.
Allogeneic Transplantation in First Remission Improves Outcomes
Irrespective of FLT3-ITD A llelic Ratio in FLT3-ITD-Positive Acute
Myelogenous Leukemia. Biol Blood Marrow Transplant (2016) 22(7):1218
26. doi: 10.1016/j.bbmt.2016.03.027
141. Bazarbachi A, Bug G, Baron F, Brissot E, Ciceri F, Dalle IA, et al. Clinical
practice recommendation on hematopoietic stem cell transplantation for
acute myeloid leukemia patients with FLT3-internal tandem duplication: a
position statement from the Acute Leukemia Working Party of the European
Society for Blood and Marrow Transplantation. Haematologica (2020) 105
(6):150716. doi: 10.3324/haematol.2019.243410
142. Kayser S, Schlenk RF, Londono MC, Breitenbuecher F, Wittke K, Du J, et al.
Insertion of FLT3 internal tandem duplication in the tyrosine kinase
domain-1 is associated with resistance to chemot herapy and inferior
outcome. Blood (2009) 114(12):238692. doi: 10.1182/blood-2009-03-
209999
143. Herold T, Rothenberg-Thurley M, Grunwald VV, Janke H, Goerlich D,
Sauerland MC, et al. Validation and renement of the revised 2017 European
LeukemiaNet genetic risk strati
cation of acute myeloid leukemia. Leukemia
(2020) 34:316172. doi: 10.1038/s41375-020-0806-0
144. Frohling S, Scholl C, Levine RL, Loriaux M, Boggon TJ, Bernard OA, et al.
Identication of driver and passenger mutations of FLT3 by high-throughput
DNA sequence analysis and functional assessment of candidate alleles. Cancer
Cell (2007) 12(6):50113. doi: 10.1016/j.ccr.2007.11.005
145. Tarlock K HM, Hylkema T, Ries R, Farrar JE, Auvil JG, Gerhard DS, et al.
Discovery and Functional Validation of Novel Pediatric Specic FLT3
Activating Mutations in Acute Myeloid Leukemia: Results from the COG/
NCI Target Initiative. Blood (2015) 126(23):87. doi: 10.1182/
blood.V126.23.87.87
146. Daver N, Price A, Benton CB, Patel K, Zhang W, Konopleva M, et al. First
Report of Sorafenib in Patients With Acute Myeloid Leukemia Harboring
Non-Canonical FLT3 Mutations [Case Report]. Front Oncol (2020) 10:1538
(1538):1538. doi: 10.3389/fonc.2020.01538
147. Heidel F, Solem FK, Breitenbuecher F, Lipka DB, Kasper S, Thiede MH, 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(1):293300. doi: 10.1182/blood-2005-06-2469
148. Pauwels D, Sweron B, Cools J. The N676D and G697R mutations in the
kinase domain of FLT3 confer resis tance to the inhibitor AC220.
Haematologica (2012) 97(11):17734. doi: 10.3324/haematol.2012.069781
149. Tarver TC, Hill JE, Rahmat L, Perl AE, Bahceci E, Mori K, et al. Gilteritinib is
a clinically active FLT3 inhibitor with broad activity against FLT3 kinase
domain mutations. Blood Adv (2020) 4(3):5 1424. doi: 10.1182/
bloodadvances.2019000919
150. Malaise M, Steinbach D, Corbacioglu S. Clinical implications of c-Kit
mutations in acute myelogenous leukemia. Curr Hematol Malig Rep
(2009) 4(2):7782. doi: 10.1007/s11899-009-0011-8
151. Chen W, Xie H, Wang H, Chen L, Sun Y, Chen Z, et al. Prognostic
Signicance of KIT Mutations in Core-Binding Factor Acute Myeloid
Leukemia: A Systematic Review and Meta-Analysis. PloS One (2016) 11
(1):e0146614. doi: 10.1371/journal.pone.0146614
152. Levis MJ, Perl AE, Dombret H, Do
̈
hner H, Steffen B, Rousselot P, et al. Final
Results of a Phase 2 Open-Label, Monotherapy Efcacy and Safety Study of
Quizartinib (AC220) i n Patients with FLT3-ITD Positive or Negative
Relapsed/Refractor y Acute Myel oid Leukemia After Second-Line
Chemotherapy or Hematopoietic Stem Cell Transplantation. Blood (2012)
120(21):6733. doi: 10.1182/blood.V120.21.673.673
153. Wattad M, Weber D, Dohner K, Krauter J, Gaidzik VI, Paschka P, et al.
Impact of salvage regimens on response and overall survival in acute myeloid
leukemia with induction failure. Leukemia (2017) 31(6):130613 .
doi: 10.1038/leu.2017.23
154. Choe S, Wang H, DiNardo CD, Stein EM, de Botton S, Roboz GJ, et al.
Molecular mechanisms mediating relapse following ivosidenib monotherapy
in IDH1-mutant relapsed or refractory AML. Blood Adv (2020) 4(9):1894
905. doi: 10.1182/bloodadvances.2020001503
155. Quek L, David MD, Kennedy A, Metzner M, Amatangelo M, Shih A, et al.
Clonal heterogeneity of acute myeloid leukemia treated with the IDH2
inhibitor enasidenib. Nat Med (2018) 24(8):116777. doi: 10.1038/s41591-
018-0115-6
156. Abbas HA, Alfayez M, Kadia T, Ravandi-Kashani F, Daver N. Midostaurin
In Acute Myeloid Leukemia: An Evidence-Based Review And Patient
Selection. Cancer Manag Res ( 2019) 11:8817 28. doi: 10.2147/
CMAR.S177894.Citedin:Pubmed
157. Pratz KW, Levis M. How I treat FLT3-mutated AML. Blood (2017) 129
(5):56571. doi: 10.1182/blood-2016-09-693648
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Kennedy and Smith. This is an open-access article distributed under
the terms of the Creative Commons Attribution License (CC BY). The use, distribution
or reproduction in other forums is permitted, provided the original author(s) and the
copyright owner(s) are credited and that the original publication in this journal is
cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Kennedy and Smith Emerging Topics in FLT3 AML
Frontiers in Oncology | www.frontiersin.org December 2020 | Volume 10 | Article 61288020