Midostaurin was recently approved by the US Food and Drug Administration for the treatment of FLT3-mutant acute myeloid leukemia (AML). This is the first drug to receive regulatory approval for AML in the United States since the year 2000. Midostaurin is a small-molecule kinase inhibitor with activity against the receptor tyrosine kinase FLT3, and its approval will hopefully mark the beginning of an era of targeted agents for the treatment of molecularly defined subtypes of AML.

On April 28, 2017, the US Food and Drug Administration (FDA) approved midostaurin (Rydapt; Novartis Pharmaceuticals, Inc) for the treatment of adult patients with newly diagnosed FLT3-mutated acute myeloid leukemia (AML).1  A companion diagnostic test for the detection of FLT3 mutations (LeukoStrat CDx FLT3 Mutation Assay; Invivoscribe Technologies, Inc) was also approved. According to the FDA label, the recommended dose of midostaurin (available in 25-mg capsules) is 50 mg twice daily on days 8 to 21 of each cycle of induction with cytarabine and daunorubicin and days 8 to 21 of each cycle of consolidation with high-dose cytarabine. The label notes that the drug is not indicated for single-agent treatment of AML. Midostaurin, as an inhibitor of both wild-type and D816V-mutated KIT,2  was simultaneously approved for the treatment of aggressive systemic mastocytosis, although as a single agent and at a higher dose (100 mg twice daily).

Activating mutations of the receptor tyrosine kinase FLT3 are among the most common genetic lesions found in AML and represent an often difficult to treat subtype.3  The most common mutation (∼23% of AML cases), an internal tandem duplication (ITD; FLT3-ITD), is associated with leukocytosis (often life-threatening) and a high relapse rate, all leading to reduced overall survival (OS).4-9  The less common mutations (7% of cases) are those found in the tyrosine kinase domain (TKD; FLT3-TKD).9  Most published reports suggest that they confer a negative prognosis, although to a lesser degree than the FLT3-ITD mutations, and there are some reports suggesting that FLT3-TKD mutations have no prognostic impact or even have a favorable impact.7,10-13  Regardless, FLT3 mutations are a common feature of AML, as anyone who treats the disease will readily attest. There is a general consensus that allogeneic transplant, when feasible, is the preferred consolidation treatment of patients with FLT3-ITD mutations, although there is ongoing debate regarding the impact of NPM1 mutations and FLT3-ITD allelic burden on the benefit of transplant.14-24 

Midostaurin (N-benzoyl staurosporine, previously referred to as CGP41251 and PKC412) is an indolocarbazole and a direct derivative of staurosporine, the original “pan-kinase” inhibitor.25  By no means a pan-kinase inhibitor, midostaurin can certainly be referred to as a multitargeted kinase inhibitor, at least in comparison with some other compounds (Figure 1).26  The drug has quite literally been under investigation for decades, originally as an inhibitor of protein kinase C.27  Around the same time that FLT3 mutations were being recognized as important prognostic factors in AML, midostaurin was characterized as a FLT3 inhibitor using in vitro and animal models.28  In a study of 20 relapsed/refractory patients with FLT3-mutated AML treated with 75 mg of midostaurin 3 times daily, no patient achieved a complete remission (CR), but peripheral blood blasts were reduced in 70% (14 of 20), and 6 patients achieved a greater reduction in bone marrow blasts of at least 50%.29  A larger study examined the effects of 50 mg or 100 mg twice daily in both FLT3-mutated and nonmutated AML patients and noted blast reductions in about half of the FLT3-mutated patients and, interestingly, blast reductions in about a third of the nonmutated patients, but still no CRs.30  Next, a phase 1B study examined the safety and tolerability of combining midostaurin with induction chemotherapy in newly diagnosed AML patients, using different doses and schedules.31  Gastrointestinal adverse events were common, as was the withdrawal rate from this trial, and there were concerns that midostaurin administered concomitantly with daunorubicin resulted in elevated mean levels of the anthracycline.31  Of 6 cohorts of patients, the most tolerable regimen to emerge was midostaurin 50 mg twice daily on days 8 to 21 following a conventional induction with cytarabine and daunorubicin, which was the regimen carried forward into the phase 3 study.

Figure 1.

Kinase interaction maps. Shown here are the maps for 5 small-molecule kinase inhibitors ranging from very nonselective (staurosporine) to highly selective (quizartinib). The black arrow on each dendrogram denotes the approximate location for the FLT3 receptor. Lestaurtinib, midostaurin, sorafenib, and quizartinib have all been studied as FLT3 inhibitors. Adapted from Davis et al26  with permission.

Figure 1.

Kinase interaction maps. Shown here are the maps for 5 small-molecule kinase inhibitors ranging from very nonselective (staurosporine) to highly selective (quizartinib). The black arrow on each dendrogram denotes the approximate location for the FLT3 receptor. Lestaurtinib, midostaurin, sorafenib, and quizartinib have all been studied as FLT3 inhibitors. Adapted from Davis et al26  with permission.

Close modal

From these preclinical and early-phase clinical studies, we can draw the following conclusions about midostaurin: (1) the drug is a multitargeted kinase inhibitor with inhibitory activity against both the FLT3-ITD and FLT3-TKD mutants; (2) as a single agent, it has little or no clinical utility in AML (ie, it is unlikely to induce remissions or allow for a bridge to transplant), but clearly has biologic activity as manifest by reductions in blood and marrow blasts; (3) it probably has activity in nonmutated FLT3 AML, although the specific subtypes will hopefully be better defined using molecular techniques; (4) midostaurin is associated with an increase in gastrointestinal side effects (nausea, vomiting, diarrhea) both as monotherapy and when the drug is incorporated into chemotherapy regimens, which may limit our ability to incorporate it into different AML treatment scenarios; (5) the pharmacokinetics are complex,29,30,32  with 2 active metabolites, and the highest levels of active drug are seen during the first few weeks of treatment (either as monotherapy or when given sequentially following chemotherapy); and (6) it can be safely combined with induction and consolidation chemotherapy.

CALGB10603 (“RATIFY”) was a global, randomized, placebo-controlled phase 3 trial carried out in 225 sites across 17 countries to determine whether the addition of midostaurin to induction and consolidation, followed by 1 year of maintenance, would improve the OS of patients with FLT3-mutated AML aged 18 to 59 years.33  Patients were screened for the presence of FLT3 mutations at the start of the trial. As noted, this trial implemented the regimen deemed to be most tolerable of the ones tested in the phase 1B study. After mutation status was confirmed, patients were randomized to receive either placebo or 50 mg of midostaurin twice daily, administered on days 8 to 21 following a 7+3 regimen (cytarabine 200 mg per meter-square per day on days 1-7 by continuous IV infusion and daunorubicin 60 mg per meter-square per day on days 1-3) and on days 8 to 21 of consolidation with high-dose cytarabine (cytarabine 3000 mg per meter-square IV over 3 hours twice daily on days 1, 3, and 5). On completion of consolidation, patients were treated with midostaurin as a single agent 50 mg twice daily for 12 months. There were 3 strata: FLT3-TKD alone, FLT3-ITD allelic ratio <0.7, and FLT3-ITD allelic ratio ≥0.7. Enrollment started in May 2008 and was completed in October 2011. The original design was to conduct the final analysis after 509 OS events had occurred. However, as of April 2015 there were still only 357 events, and the event rate appeared to have reached a plateau. The reason for this was likely because of a higher than expected transplant rate and a higher than expected incidence of FLT3-TKD mutations (23%). Given that there was sufficient follow-up to assess OS, the trial was amended to conduct the primary analysis in May of 2015. The results (Figure 2) indicated that patients randomized to receive midostaurin had a higher median OS, with a hazard ratio of 0.78 compared with those receiving placebo. The 4-year OS rates were 51.4% in the midostaurin arm vs 44.3% in the placebo arm, and the rate of protocol-defined CR (CR within 60 days of initiation of protocol therapy) was 59% vs 53%, respectively.

Figure 2.

Kaplan-Meier curve for OS for patients on CALGB10603. This was presented at the 2015 annual meeting of the American Society of Hematology, Orlando, FL, 5-8 December 2015. Reprinted from Stone et al.33 

Figure 2.

Kaplan-Meier curve for OS for patients on CALGB10603. This was presented at the 2015 annual meeting of the American Society of Hematology, Orlando, FL, 5-8 December 2015. Reprinted from Stone et al.33 

Close modal

While we await the final peer-reviewed publication, a closer look at the data from the FDA label and from the meeting abstract reveals a number of interesting findings.1,33  First, patients in all 3 strata (FLT3-TKD, FLT3-ITD low, or FLT3-ITD high ratio) benefitted from midostaurin, and those in the TKD strata actually had the lowest hazard ratio (0.648) compared with placebo. The fact that these 3 groups behave very differently with respect to their clinical outcomes in response to standard therapy, coupled with the biologic activity of midostaurin against nonmutant FLT3 AML, may indicate that a significant component of midostaurin’s efficacy is derived from its multitargeted nature. Second, there was a higher than expected incidence of FLT3-TKD patients (23% vs the expected rate of 7%). We can speculate that this is because patients were required to undergo molecular screening for FLT3 mutations prior to enrollment. Patients with FLT3-TKD–mutated AML have lower white blood cell counts and generally less proliferative disease than FLT3-ITD patients, and this likely biased the enrollment. FLT3-TKD patients were more likely than FLT3-ITD patients to be stable enough to allow for treatment delay until the results of molecular testing were known. Third, the survival curves separate quickly and remain roughly parallel thereafter (Figure 1), suggesting that the primary benefit from midostaurin occurs early on. This would be consistent with the drug’s pharmacokinetic profile, in which plasma levels are highest during the first few weeks of treatment.27,30,32  A 2-week course of treatment following chemotherapy, therefore, may be the best way to adapt this drug into a standard regimen. A fourth interesting finding is the remarkable difference in survival of midostaurin-treated patients who underwent allogeneic transplant in first CR compared with those on the placebo arm. Given the well-described impact of minimal residual disease on outcomes after allogeneic transplant for AML,34,35  this finding may represent evidence that midostaurin truly augments induction chemotherapy and leads to deeper remissions. To address this issue, future comparative studies could focus on minimal residual disease levels following midostaurin treatment.

Now that midostaurin is approved, how shall we use it to treat FLT3-mutated AML? Of course, we are awaiting the final publication, and any ancillary studies, and therefore important additional details may emerge to guide us. However, to start with, we should use it exactly as the label states. During the roughly 10 years that elapsed from trial conception to drug approval, 2 important concepts regarding FLT3-mutated AML emerged from studies around the globe. First, allogeneic transplant is an effective consolidation treatment of FLT3-ITD AML.14-16,19,21,23,24,36  Second, the disease is genetically polyclonal at diagnosis, with subclones defined by different mixtures of driver mutations.37,38  Midostaurin, as a multitargeted kinase inhibitor, can fit very neatly into that paradigm, particularly in light of the impressive survival results achieved in the midostaurin patients who underwent transplant in CR1. The data we have before us suggest that a newly diagnosed patient with FLT3-ITD AML should be given chemotherapy in combination with midostaurin, and, once remission is achieved, proceed as soon as possible to an allogeneic transplant. Transplant in first CR may, of course, not be necessary for those patients with FLT3-TKD AML, as the role of allogeneic transplant for these patients is less obvious.

What about maintenance therapy? Even though CALGB10603 included a maintenance phase of treatment, from what has been presented thus far, there is little to support the use of midostaurin as a maintenance drug. More than half of midostaurin-treated patients (59%) underwent allogeneic transplant, and, per protocol, therefore did not receive maintenance therapy. The lack of further separation of the survival curves after the first few months of therapy attests to the fact that either midostaurin does not work as maintenance or that few patients were taking midostaurin, either because they had been transplanted or because of some other reason. Regardless, midostaurin seems better suited to use during induction and consolidation, given that drug levels are highest early on, and given that its side-effect profile (mainly gastrointestinal) does not lend itself readily to patient compliance. Midostaurin administered as maintenance therapy after allogeneic transplant is currently under active study, although it is too early to draw any conclusions about tolerability or efficacy in this setting.39,40 

There are, of course, several other kinase inhibitors under investigation for the treatment of FLT3-mutated AML.41  Midostaurin is the “oldest,” and so it is not surprising that it is the first across the finish line. The inhibitors vary in their selectivity (Figure 1), and it is interesting to note that lestaurtinib, a drug even less selective than midostaurin, failed to lead to improvement in survival in a trial relatively similar in design to CALGB10603.42  Certainly it makes sense to move in the direction of more selective, more potent FLT3 inhibitors, and to this end there are several interesting drug candidates43-45 ; a number of phase 3 trials of these newer FLT3 inhibitors are under way. QuANTUM-First (NCT02668653), for example, will test quizartinib in combination with chemotherapy administered during induction, consolidation, and maintenance (including posttransplant) against placebo for newly diagnosed FLT3-ITD AML patients; the Blood and Marrow Transplant Clinical Trials Network (BMT-CTN) 1506/Morpho trial (NCT02997202) will examine the effect of gilteritinib vs placebo as posttransplant maintenance therapy. Crenolanib, another new FLT3 inhibitor,44  is being tested in combination with salvage chemotherapy in a phase 3 trial for relapsed or refractory FLT3-mutated AML patients (NCT02298166).

Finally, we must be grateful to the patients who participated in this study, and we must congratulate the investigators, the leadership of the international cooperative groups, and the industry sponsor for having the foresight to design the trial and the resolve to open it globally and to see it through to the end. Their efforts have brought us the first major breakthrough in AML therapy in many years.

Looking back on the years it took to accomplish this breakthrough, we should also take this opportunity to join others in questioning the wisdom of a rigid insistence on OS as the only acceptable end point in AML trials.46  Indeed, this rigidity resulted in the withdrawal from the market of gemtuzumab ozogamicin in 2010, a drug that we subsequently learned is probably quite useful in the treatment of select subsets of AML patients.47  As we learn more about this disease and as we subdivide it into smaller and smaller molecularly defined groups, we may find that, in using OS as our only definitive end point, we have truly made the perfect enemy of the good.

Contribution: M.L. wrote the manuscript.

Conflict-of-interest disclosure: M.L. receives research funding from Novartis and Astellas and serves as a consultant for Novartis, Daiichi-Sankyo, Astellas, and Arog.

Correspondence: Mark Levis, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans St, Room 2M44, Baltimore, MD 21287; e-mail: levisma@jhmi.edu.

1.
RYDAPT (midostaurin) for oral use. Silver Spring, MD: US Food and Drug Administration;
2017
. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/207997s000lbl.pdf. Accessed 16 May 2017.
2.
Gotlib
J
,
Kluin-Nelemans
HC
,
George
TI
, et al
.
Efficacy and safety of midostaurin in advanced systemic mastocytosis
.
N Engl J Med
.
2016
;
374
(
26
):
2530
-
2541
.
3.
Pratz
KW
,
Levis
M
.
How I treat FLT3-mutated AML
.
Blood
.
2017
;
129
(
5
):
565
-
571
.
4.
Kiyoi
H
,
Naoe
T
,
Nakano
Y
, et al
.
Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia
.
Blood
.
1999
;
93
(
9
):
3074
-
3080
.
5.
Kottaridis
PD
,
Gale
RE
,
Frew
ME
, et al
.
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials
.
Blood
.
2001
;
98
(
6
):
1752
-
1759
.
6.
Schnittger
S
,
Schoch
C
,
Dugas
M
, et al
.
Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease
.
Blood
.
2002
;
100
(
1
):
59
-
66
.
7.
Thiede
C
,
Steudel
C
,
Mohr
B
, et al
.
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis
.
Blood
.
2002
;
99
(
12
):
4326
-
4335
.
8.
Fröhling
S
,
Schlenk
RF
,
Breitruck
J
, et al
;
AML Study Group Ulm. Acute myeloid leukemia
.
Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm
.
Blood
.
2002
;
100
(
13
):
4372
-
4380
.
9.
Levis
M
,
Small
D
.
FLT3: ITDoes matter in leukemia
.
Leukemia
.
2003
;
17
(
9
):
1738
-
1752
.
10.
Moreno
I
,
Martín
G
,
Bolufer
P
, et al
.
Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia
.
Haematologica
.
2003
;
88
(
1
):
19
-
24
.
11.
Yanada
M
,
Matsuo
K
,
Suzuki
T
,
Kiyoi
H
,
Naoe
T
.
Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis
.
Leukemia
.
2005
;
19
(
8
):
1345
-
1349
.
12.
Mead
AJ
,
Linch
DC
,
Hills
RK
,
Wheatley
K
,
Burnett
AK
,
Gale
RE
.
FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia
.
Blood
.
2007
;
110
(
4
):
1262
-
1270
.
13.
Whitman
SP
,
Ruppert
AS
,
Radmacher
MD
, et al
.
FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications
.
Blood
.
2008
;
111
(
3
):
1552
-
1559
.
14.
Bornhäuser
M
,
Illmer
T
,
Schaich
M
,
Soucek
S
,
Ehninger
G
,
Thiede
C
;
AML SHG 96 Study Group
.
Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML
.
Blood
.
2007
;
109
(
5
):
2264
-
2265
.
15.
Schlenk
RF
,
Döhner
K
,
Krauter
J
, et al
;
German-Austrian Acute Myeloid Leukemia Study Group
.
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia
.
N Engl J Med
.
2008
;
358
(
18
):
1909
-
1918
.
16.
DeZern
AE
,
Sung
A
,
Kim
S
, et al
.
Role of allogeneic transplantation for FLT3/ITD acute myeloid leukemia: outcomes from 133 consecutive newly diagnosed patients from a single institution
.
Biol Blood Marrow Transplant
.
2011
;
17
(
9
):
1404
-
1409
.
17.
Pratcorona
M
,
Brunet
S
,
Nomdedéu
J
, et al
;
Grupo Cooperativo Para el Estudio y Tratamiento de las Leucemias Agudas Mieloblásticas
.
Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy
.
Blood
.
2013
;
121
(
14
):
2734
-
2738
.
18.
Lin
PH
,
Lin
CC
,
Yang
HI
, et al
.
Prognostic impact of allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia patients with internal tandem duplication of FLT3
.
Leuk Res
.
2013
;
37
(
3
):
287
-
292
.
19.
Brunet
S
,
Labopin
M
,
Esteve
J
, et al
.
Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis
.
J Clin Oncol
.
2012
;
30
(
7
):
735
-
741
.
20.
Levis
M
.
FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013?
Hematology Am Soc Hematol Educ Program
.
2013
;
2013
:220-226.
21.
Schlenk
RF
,
Kayser
S
,
Bullinger
L
, et al
;
German-Austrian AML Study Group
.
Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation
.
Blood
.
2014
;
124
(
23
):
3441
-
3449
.
22.
Schechter
T
,
Gassas
A
,
Chen
H
, et al
.
The outcome of allogeneic hematopoietic cell transplantation for children with FMS-like tyrosine kinase 3 internal tandem duplication-positive acute myelogenous leukemia
.
Biol Blood Marrow Transplant
.
2015
;
21
(
1
):
172
-
175
.
23.
Oran
B
,
Cortes
J
,
Beitinjaneh
A
, et al
.
Allogeneic transplantation in first remission improves outcomes irrespective of FLT3-ITD allelic ratio in FLT3-ITD-positive acute myelogenous leukemia
.
Biol Blood Marrow Transplant
.
2016
;
22
(
7
):
1218
-
1226
.
24.
Ho
AD
,
Schetelig
J
,
Bochtler
T
, et al
;
Study Alliance Leukemia
.
Allogeneic stem cell transplantation improves survival in patients with acute myeloid leukemia characterized by a high allelic ratio of mutant FLT3-ITD
.
Biol Blood Marrow Transplant
.
2016
;
22
(
3
):
462
-
469
.
25.
Tamaoki
T
,
Nomoto
H
,
Takahashi
I
,
Kato
Y
,
Morimoto
M
,
Tomita
F
.
Staurosporine, a potent inhibitor of phospholipid/Ca++ dependent protein kinase
.
Biochem Biophys Res Commun
.
1986
;
135
(
2
):
397
-
402
.
26.
Davis
MI
,
Hunt
JP
,
Herrgard
S
, et al
.
Comprehensive analysis of kinase inhibitor selectivity
.
Nat Biotechnol
.
2011
;
29
(
11
):
1046
-
1051
.
27.
Propper
DJ
,
McDonald
AC
,
Man
A
, et al
.
Phase I and pharmacokinetic study of PKC412, an inhibitor of protein kinase C
.
J Clin Oncol
.
2001
;
19
(
5
):
1485
-
1492
.
28.
Weisberg
E
,
Boulton
C
,
Kelly
LM
, et al
.
Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412
.
Cancer Cell
.
2002
;
1
(
5
):
433
-
443
.
29.
Stone
RM
,
DeAngelo
DJ
,
Klimek
V
, et al
.
Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412
.
Blood
.
2005
;
105
(
1
):
54
-
60
.
30.
Fischer
T
,
Stone
RM
,
Deangelo
DJ
, et al
.
Phase IIB trial of oral midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and high-risk myelodysplastic syndrome with either wild-type or mutated FLT3
.
J Clin Oncol
.
2010
;
28
(
28
):
4339
-
4345
.
31.
Stone
RM
,
Fischer
T
,
Paquette
R
, et al
.
Phase IB study of the FLT3 kinase inhibitor midostaurin with chemotherapy in younger newly diagnosed adult patients with acute myeloid leukemia
.
Leukemia
.
2012
;
26
(
9
):
2061
-
2068
.
32.
Levis
M
,
Brown
P
,
Smith
BD
, et al
.
Plasma inhibitory activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic response to FLT3 inhibitors
.
Blood
.
2006
;
108
(
10
):
3477
-
3483
.
33.
Stone
R
,
Mandrekar
S
,
Sanford
BL
, et al
.
The multi-kinase inhibitor midostaurin (M) prolongs survival compared with placebo (P) in combination with daunorubicin (D)/cytarabine (C) induction (ind), high-dose C consolidation (consol), and as maintenance (maint) therapy in newly diagnosed acute myeloid leukemia (AML) patients (pts) age 18-60 with FLT3 mutations (muts): an international prospective randomized (rand) P-controlled double-blind trial (CALGB 10603/RATIFY [Alliance])
[abstract].
Blood
.
2015
;
126
(
23
). Abstract 6.
34.
Bastos-Oreiro
M
,
Perez-Corral
A
,
Martínez-Laperche
C
, et al
.
Prognostic impact of minimal residual disease analysis by flow cytometry in patients with acute myeloid leukemia before and after allogeneic hemopoietic stem cell transplantation
.
Eur J Haematol
.
2014
;
93
(
3
):
239
-
246
.
35.
Walter
RB
,
Gyurkocza
B
,
Storer
BE
, et al
.
Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation
.
Leukemia
.
2015
;
29
(
1
):
137
-
144
.
36.
Labouré
G
,
Dulucq
S
,
Labopin
M
, et al
.
Potent graft-versus-leukemia effect after reduced-intensity allogeneic SCT for intermediate-risk AML with FLT3-ITD or wild-type NPM1 and CEBPA without FLT3-ITD
.
Biol Blood Marrow Transplant
.
2012
;
18
(
12
):
1845
-
1850
.
37.
Ding
L
,
Ley
TJ
,
Larson
DE
, et al
.
Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing
.
Nature
.
2012
;
481
(
7382
):
506
-
510
.
38.
Welch
JS
,
Ley
TJ
,
Link
DC
, et al
.
The origin and evolution of mutations in acute myeloid leukemia
.
Cell
.
2012
;
150
(
2
):
264
-
278
.
39.
Schlenk
R
,
Fiedler
W
,
Salih
H
, 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
[abstract].
Blood
.
2016
;
128
(
22
). Abstract 449.
40.
Maziarz
RT
,
Patnaik
M
,
Scott
B
, et al
.
Radius: a phase 2, randomized trial of standard of care (SOC) with or without midostaurin to prevent relapse following allogeneic hematopoietic stem cell transplant (alloHSCT) in patients (pts) with FLT3-Itd-mutated acute myeloid leukemia (AML)
[abstract].
Blood
.
2016
;
128
(
22
). Abstract 2248.
41.
Leick
MB
,
Levis
MJ
.
The future of targeting FLT3 activation in AML [published online ahead of print 18 April 2017]
.
Curr Hematol Malig Rep
.
doi:10.1007/s11899-017-0381-2
.
42.
Knapper
S
,
Russell
N
,
Gilkes
A
, et al
.
A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML
.
Blood
.
2017
;
129
(
9
):
1143
-
1154
.
43.
Zarrinkar
PP
,
Gunawardane
RN
,
Cramer
MD
, et al
.
AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML)
.
Blood
.
2009
;
114
(
14
):
2984
-
2992
.
44.
Galanis
A
,
Ma
H
,
Rajkhowa
T
, et al
.
Crenolanib is a potent inhibitor of FLT3 with activity against resistance-conferring point mutants
.
Blood
.
2014
;
123
(
1
):
94
-
100
.
45.
Lee
LY
,
Hernandez
D
,
Rajkhowa
T
, et al
.
Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor
.
Blood
.
2017
;
129
(
2
):
257
-
260
.
46.
Estey
E
.
Why is progress in acute myeloid leukemia so slow?
Semin Hematol
.
2015
;
52
(
3
):
243
-
248
.
47.
Hills
RK
,
Castaigne
S
,
Appelbaum
FR
, et al
.
Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials
.
Lancet Oncol
.
2014
;
15
(
9
):
986
-
996
.
Sign in via your Institution