Acute myeloid leukemia with a FLT3 internal tandem duplication (FLT3/ITD) mutation is an aggressive hematologic malignancy with a generally poor prognosis. It can be successfully treated into remission with intensive chemotherapy, but it routinely relapses. At relapse, the blasts tend to have higher mutant allelic ratios and, in vitro, are more addicted to the aberrant signaling from the FLT3/ITD oncoprotein. They remain highly responsive to FLT3 ligand, the levels of which rise several-fold during the course of chemotherapy. The question now arises as to whether these high levels of FLT3 ligand are actually promoting relapse, and, if so, how we can use this information to adjust our therapeutic approach and improve the cure rate for acute myeloid leukemia with FLT3/ITD.

The law of unintended consequences teaches us that, when we intervene in a complex system, we invariably create unanticipated and sometimes undesirable outcomes. Our attempts to treat the disease known as acute myeloid leukemia with a FLT3 internal tandem duplication (FLT3/ITD AML) mutation certainly could be characterized as an intervention in a complex system, and the question can now be raised as to whether we are creating an unintended consequence with our therapeutic approach.

FLT3 is a cytokine receptor that is expressed on the leukemic blasts in most cases of acute leukemia.1,,,5  On binding FLT3 ligand (FL), FLT3 dimerizes and undergoes a conformational change, causing its activation loop to assume an open conformation and to allow ATP access to the ATP-binding pocket. Ligand-activated FLT3 undergoes autophosphorylation and, through a series of kinase cascades, transduces signals promoting cell growth and inhibiting apoptosis through proteins such as Ras-GTPase activating protein, phospholipase C β, STAT5, and ERK1/2.6,,,,,12  The ligand, FL, is expressed in virtually all cell types thus far examined, including leukemia cells.13,15  In contrast, the receptor, FLT3, has a fairly narrow range of cell expression, being localized primarily to hematopoietic and neural tissues, which presumably confines its functions to these cell types.16  FL acts in synergy with other cytokines to promote hematopoietic precursor expansion, and targeted disruption of either FLT3 or FL in mice, although not embryonically lethal, leads to a reduction in hematopoietic precursors.17,,,,,,24 

FLT3/ITDs were first described in patients with AML in 1996 by Nakao et al.25  These mutations, which disrupt the autoinhibitory function of the receptor's juxtamembrane domain, result in constitutive autophosphorylation of FLT3 within the blasts that harbor them.26,27  Fifteen years after this initial discovery, FLT3/ITD AML now stands as a distinct clinical entity, an often lethal subtype of AML that has been a considerable challenge to those of us who treat it.28  Some recent clinical and laboratory findings about this disease may provide insight into why these patients relapse so quickly, and how we might improve their outcomes.

FLT3/ITD mutations are present in roughly a quarter of adult AML cases.29  In a minority of cases they represent a presumably late mutation in AML, evolving out of an antecedent myelodysplastic syndrome.30  However, the more characteristic presentation is that of de novo disease, presenting with a high leukocyte count and normal cytogenetics. Numerous retrospective analyses of clinical trial results have established that patients with FLT3/ITD AML achieve complete remission at or near the rate for patients with AML lacking these mutations.31,,34  However, equally well established is the fact that patients with FLT3/ITD are far more likely to relapse and do so more rapidly than their FLT3 wild-type counterparts. The median survival of FLT3 mutant AML after first relapse has been reported to be < 5 months.35,36 

Coincident with the recognition of FLT3/ITD AML as a disease entity was the emergence of an important new anticancer therapy: tyrosine kinase inhibitors (TKIs). The remarkable clinical activity of imatinib mesylate for blast crisis chronic myelogenous leukemia and Philadelphia-positive acute lymphocytic leukemia (2 diseases with a number of similarities to FLT3/ITD AML) spurred the development of FLT3 TKIs.37,38  Currently, older, multitargeted FLT3 inhibitors are in advanced clinical trials, whereas newer, more FLT3-selective inhibitors are entering development.39 

There are important differences between the FLT3/ITD and BCR-ABL oncoproteins however similar the diseases they cause might seem to be. Unlike BCR-ABL, FLT3 is a transmembrane protein primarily localized to the plasma membrane, where it binds its cognate ligand, the cytokine FL. Why would the ligand have any effect on a receptor tyrosine kinase that is supposed to be constitutively activated? Recent evidence suggests that the ITD-mutated receptor is, in fact, heavily influenced by FL. FL is coexpressed with FLT3 in leukemia cells and is up-regulated in response to FLT3 inhibition. When the FLT3/ITD receptor is expressed in cells that completely lack FL (derived from an FL−/− mouse), it shows only weak autophosphorylation.40  This finding casts the FLT3/ITD oncoprotein in a wholly different light: not as an autonomously activated receptor but rather as one that is simply hyperresponsive to its ligand. To complicate matters further, FL markedly interferes with the ability of TKIs to inhibit FLT3 signaling.41  Indeed, the surge of FL after chemotherapy may have been responsible for the generally poor level of in vivo FLT3 inhibition observed in the recent trial of lestaurtinib after chemotherapy.36  The influence of FL on FLT3/ITD signaling will probably be quite problematic to efforts aimed at incorporating FLT3 inhibition into conventional AML chemotherapy regimens. Aplasia-inducing radiation or chemotherapy has been well established to induce significant increases in FL levels,42,44  which in turn would block the effects of FLT3 inhibitors.

There is another important difference between the FLT3/ITD and BCR-ABL oncoproteins. Inhibition of the downstream signaling of BCR-ABL by an inhibitor such as imatinib mesylate results in rapid apoptosis of the bulk population of leukemia cells.45  This addiction of the leukemia cells to the oncoprotein is a signature feature of the Philadelphia-positive diseases that can be exploited for clinical benefit. In contrast to this, addiction to FLT3/ITD signaling is not necessarily a consistent feature of FLT3/ITD AML, at least according to in vitro studies. Inhibition of FLT3 alone is insufficient to induce apoptosis in a significant fraction of FLT3/ITD AML primary samples collected at initial diagnosis.46  However, samples collected at the inevitable relapse and tested in vitro are much more likely to undergo apoptosis in response to FLT3 inhibition in comparison to the diagnostic samples.46  The best predictor for an AML sample to have a cytotoxic response to FLT3 inhibition is a high FLT3/ITD mutant allelic burden. Perhaps not coincidentally, the FLT3/ITD mutant allelic ratio tends to be increased at relapse compared with diagnosis (although in a minority of cases the mutation can be lost altogether).47,49  Relapse, a higher mutant allelic ratio, and addiction to FLT3/ITD signaling appear inextricably linked in FLT3/ITD AML.

To summarize, then, FLT3/ITD AML is a disease that appears to evolve between diagnosis and relapse, with the leukemia cells becoming more addicted to FLT3 signaling after recurrence after chemotherapy. Treatment of a patient with chemotherapy leads to high levels of FL in the plasma throughout the period of recovery and during consolidation. FL is a cytokine that acts directly on the mutant FLT3/ITD receptor, maximizing its activity and promoting the survival of blasts. Although these findings have practical implications, in that these properties could be used to predict clinical response and to design treatment regimens, they uncover a potentially larger issue.

Are we promoting relapse of FLT3/ITD AML with successive rounds of chemotherapy? If induction regimens de-bulk the BM of blasts, leaving a residual leukemia stem cell population, do the recurrent waves of FL that follow select for the emergence of FLT3-addicted subclones? Patients with FLT3/ITD AML often relapse during consolidation. It is conceivable, given the above-mentioned findings, that we could be doing more harm than good by administering repeat cycles of high-dose cytarabine, or whichever consolidation regimen is being used. Indeed, in a recent randomized trial of induction chemotherapy using more intensive anthracycline use, patients with FLT3/ITD AML did not appear to benefit from intensifying therapy, in contrast to patients with FLT3 wild type.50 

The clonal evolution of FLT3/ITD AML almost certainly occurs through > 1 mechanism. The lower allelic burden often seen at diagnosis may represent simple heterozygosity of the mutation in an otherwise uniform population of blasts. At relapse, deletion or point mutations of the wild-type allele, gene conversion, or outright loss of one copy of chromosome 13 (the chromosome on which the FLT3 gene is localized) could lead to a hemizygous or homozygous state.51,52  Increased expression of the mutant allele (or loss of the wild-type allele) presumably provides a selective advantage, resulting in expansion of these particular clones. Alternately, the blast population at diagnosis can be nonuniform, with some cells completely lacking the FLT3/ITD mutation and others harboring heterozygous or homozygous mutants.53  The leukemia stem cells harboring the FLT3/ITD mutation could have a survival advantage over their wild-type counterparts and emerge at relapse as the dominant clone. In any of these scenarios, however, the FLT3/ITD clones are still highly responsive to FL, which could contribute significantly to their ability to survive successive rounds of chemotherapy.

The patients who present with low mutant allelic ratio at diagnosis present an interesting counter-argument to the concept that FL promotes or influences relapse. Those relatively few patients with FLT3/ITD AML who have low allelic ratios at presentation often (but not always) lose the mutation altogether at relapse.4849  FL clearly does not select for the expansion of these apparent subclones. However, patients with low allelic burden seem to have a prognosis that is similar to that of patients with AML with wild-type FLT3.34  It is possible that in these cases the ITD mutation occurred relatively late in leukemogenesis, perhaps in a leukemia stem cell with lower long-term renewal potential.

The hypothesis that FL promotes relapse of FLT3/ITD AML is, in at least some ways, a testable one. In patients with newly diagnosed FLT3/ITD AML undergoing induction and consolidation there is significant interpatient variability in the degree to which FL rises from baseline.41  We might predict that relapses will be more likely to occur, and will occur earlier, in patients with high FL levels compared with low FL levels. The hypothesis could be refuted by finding no effect of FL on relapse risk or by finding that high levels actually predict for better outcomes. In fact, high FL levels after chemotherapy could be a surrogate for the intensity of aplasia, which from a traditional perspective is thought to be beneficial. We are prospectively examining this issue by serial measurements of FL levels during induction and consolidation in patients with FLT3/ITD AML, both at our own institution and in collaboration with ongoing cooperative group trials.

Many advocate the use of allogeneic transplantation as the most effective consolidation for FLT3/ITD AML.54,55  This is an issue that remains quite controversial.56,,,60  Certainly, there are numerous variables that could have influenced the outcomes of trials involving allogeneic transplantation (any one of which could cloud the interpretation of the results), including time to transplantation, preparative regimens, transplantation-related mortality, and graft-versus-host prophylaxis. However, if FL levels do contribute to relapse, and if the “graft-versus-leukemia” effect is a real one, then the best approach for the patient would be to proceed as rapidly as possible to allogeneic transplantation once remission is achieved. Allogeneic transplantation, of course, also involves chemotherapy, usually more intensive than a single course of consolidation. However, in substitution for 4 cycles of high-dose cytarabine, it probably results in a less prolonged overall elevation of FL (although this should be confirmed prospectively), and it also introduces a different type of therapeutic effect, that of immunotherapy. At our institution, where we have aggressively pursued allogeneic transplantation for these patients in first remission, the survival for patients with FLT3/ITD AML is equivalent to non–FLT3-mutated AML.55  Intriguingly, in a recent report from the AML Study Group in Ulm, Germany, where a similar strategy has been used since 2006, it was noted that patients with FLT3/ITD who received a transplant sooner rather than later after achieving remission had better outcomes.61  Normally, in landmark analyses of AML transplantation studies, longer time to transplantation is associated with better outcomes, presumably because patients who are well enough for transplantation after several months represent a generally favorable risk group. However, longer time to transplantation, which would necessitate extra courses of consolidation, was associated with worse overall survival in their analysis. This is exactly what would be predicted if those recurrent courses of consolidation were actually promoting relapse.

On the basis of these findings, it can be proposed that the optimal therapeutic approach for a patient with FLT3/ITD AML would be a single course of induction therapy followed as rapidly as possible by allogeneic transplantation, including the use of alternative donors (matched unrelated, haploidentical, or cord blood derived) if necessary. Novel therapeutic agents can be introduced into this paradigm. FLT3 inhibitors can be used early in therapy (before the rise in FL levels, before their effectiveness is limited) to improve remission rate and, if the FL levels return to baseline, to maintain the patient in remission until transplantation. FLT3 inhibition could also be used as maintenance therapy after allogeneic transplantation, as is commonly done with BCR-ABL inhibitors,62  particularly in light of the mounting anecdotal evidence of activity in this setting.63,64  Finally, consideration could be given to targeting FL with monoclonal antibodies.

Obviously, it would be far more preferable to avoid allogeneic transplantation, with its concomitant short- and long-term risks of morbidity and mortality,65  by curing our patients with a combination of chemotherapy and FLT3 inhibition. Indeed, this approach continues to look promising, because preliminary results from large combination trials appear to show a survival improvement from this approach (compared with historic controls).66,67  The final results of these trials, however, may be difficult to interpret in light of the reported high rates of allogeneic transplantations occurring in first remission in the enrolled patients.66,67 

Great strides have been made in improving the survival of patients with AML over the past several decades.68  It has been increasingly recognized that AML is not a single disease, and the improvements in survival have been brought about as a result of tailoring therapy according to the molecular features of the disease. In this case, we may need to tailor the therapy to try to account for the law of unintended consequences. Even recent medical history is full of examples of the best intentions gone awry: estrogen therapy that prevents osteoporosis but causes breast cancer,69  erythropoietin therapy that decreases transfusion requirements but kills patients with cardiovascular and thromboembolic events,70  and intensive blood glucose control that results in hypoglycemic deaths.71  More is not always better, and this may well apply to chemotherapy and FLT3/ITD AML.

This work was supported by grants from the National Institutes of Health (NCI Leukemia SPORE P50 CA100632-06, R01 CA128864) and the ASCO Foundation.

M.L. is a Clinical Scholar of the Leukemia and Lymphoma Society.

National Institutes of Health

Contribution: M.L. conceived all of the ideas expressed in this work and wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Mark Levis, Kimmel Cancer Center at Johns Hopkins, 1650 Orleans St, Rm 243, Baltimore, MD 21231; e-mail: levisma@jhmi.edu.

1
Meierhoff
G
Dehmel
U
Gruss
HJ
et al
Expression of FLT3 receptor and FLT3-ligand in human leukemia-lymphoma cell lines.
Leukemia
1995
9
8
1368
1372
2
Drexler
HG
Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells.
Leukemia
1996
10
4
588
599
3
Carow
CE
Levenstein
M
Kaufmann
SH
et al
Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias.
Blood
1996
87
3
1089
1096
4
Birg
F
Courcoul
M
Rosnet
O
et al
Expression of the FMS/KIT-like gene FLT3 in human acute leukemias of the myeloid and lymphoid lineages.
Blood
1992
80
10
2584
2593
5
Rosnet
O
Buhring
HJ
Marchetto
S
et al
Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells.
Leukemia
1996
10
2
238
248
6
Dosil
M
Wang
S
Lemischka
IR
Mitogenic signalling and substrate specificity of the Flk2/Flt3 receptor tyrosine kinase in fibroblasts and interleukin 3-dependent hematopoietic cells.
Mol Cell Biol
1993
13
10
6572
6585
7
Rosnet
O
Buhring
HJ
deLapeyriere
O
et al
Expression and signal transduction of the FLT3 tyrosine kinase receptor.
Acta Haematol
1996
95
3–4
218
223
8
Lavagna-Sevenier
C
Marchetto
S
Birnbaum
D
Rosnet
O
FLT3 signaling in hematopoietic cells involves CBL, SHC and an unknown P115 as prominent tyrosine-phosphorylated substrates.
Leukemia
1998
12
3
301
310
9
Lavagna-Sevenier
C
Marchetto
S
Birnbaum
D
Rosnet
O
The CBL-related protein CBLB participates in FLT3 and interleukin-7 receptor signal transduction in pro-B cells.
J Biol Chem
1998
273
24
14962
14967
10
Zhang
S
Mantel
C
Broxmeyer
HE
Flt3 signaling involves tyrosyl-phosphorylation of SHP-2 and SHIP and their association with Grb2 and Shc in Baf3/Flt3 cells.
J Leukoc Biol
1999
65
3
372
380
11
Marchetto
S
Fournier
E
Beslu
N
et al
SHC and SHIP phosphorylation and interaction in response to activation of the FLT3 receptor.
Leukemia
1999
13
9
1374
1382
12
Zhang
S
Fukuda
S
Lee
Y
et al
Essential role of signal transducer and activator of transcription (Stat)5a but not Stat5b for Flt3-dependent signaling.
J Exp Med
2000
192
5
719
728
13
Lyman
SD
Jacobsen
SE
c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities.
Blood
1998
91
4
1101
1134
14
Hannum
C
Culpepper
J
Campbell
D
et al
Ligand for FLT3/FLK2 receptor tyrosine kinase regulates growth of haematopoietic stem cells and is encoded by variant RNAs.
Nature
1994
368
6472
643
648
15
Lyman
SD
James
L
Johnson
L
et al
Cloning of the human homologue of the murine flt3 ligand: a growth factor for early hematopoietic progenitor cells.
Blood
1994
83
10
2795
2801
16
Small
D
Levenstein
M
Kim
E
et al
STK-1, the human homolog of Flk-2/Flt-3, is selectively expressed in CD34+ human bone marrow cells and is involved in the proliferation of early progenitor/stem cells.
Proc Natl Acad Sci U S A
1994
91
2
459
463
17
Mackarehtschian
K
Hardin
JD
Moore
KA
Boast
S
Goff
SP
Lemischka
IR
Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors.
Immunity
1995
3
1
147
161
18
McKenna
HJ
Stocking
KL
Miller
RE
et al
Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells.
Blood
2000
95
11
3489
3497
19
Ray
RJ
Paige
CJ
Furlonger
C
Lyman
SD
Rottapel
R
Flt3 ligand supports the differentiation of early B cell progenitors in the presence of interleukin-11 and interleukin-7.
Eur J Immunol
1996
26
7
1504
1510
20
Veiby
OP
Jacobsen
FW
Cui
L
Lyman
SD
Jacobsen
SE
The flt3 ligand promotes the survival of primitive hemopoietic progenitor cells with myeloid as well as B lymphoid potential. Suppression of apoptosis and counteraction by TNF-alpha and TGF-beta.
J Immunol
1996
157
7
2953
2960
21
Broxmeyer
HE
Lu
L
Cooper
S
Ruggieri
L
Li
ZH
Lyman
SD
Flt3 ligand stimulates/costimulates the growth of myeloid stem/progenitor cells.
Exp Hematol
1995
23
10
1121
1129
22
Hirayama
F
Lyman
SD
Clark
SC
Ogawa
M
The flt3 ligand supports proliferation of lymphohematopoietic progenitors and early B-lymphoid progenitors.
Blood
1995
85
7
1762
1768
23
Nicholls
SE
Winter
S
Mottram
R
Miyan
JA
Whetton
AD
Flt3 ligand can promote survival and macrophage development without proliferation in myeloid progenitor cells.
Exp Hematol
1999
27
4
663
672
24
Sitnicka
E
Buza-Vidas
N
Larsson
S
Nygren
JM
Liuba
K
Jacobsen
SE
Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells.
Blood
2003
102
3
881
886
25
Nakao
M
Yokota
S
Iwai
T
et al
Internal tandem duplication of the flt3 gene found in acute myeloid leukemia.
Leukemia
1996
10
12
1911
1918
26
Kiyoi
H
Towatari
M
Yokota
S
et al
Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product.
Leukemia
1998
12
9
1333
1337
27
Griffith
J
Black
J
Faerman
C
et al
the structural basis for autoinhibition of FLT3 by the juxtamembrane domain.
Mol Cell
2004
13
2
169
178
28
Kindler
T
Lipka
DB
Fischer
T
FLT3 as a therapeutic target in AML: still challenging after all these years.
Blood
2010
116
24
5089
5102
29
Levis
M
Small
D
FLT3: ITDoes matter in leukemia.
Leukemia
2003
17
9
1738
1752
30
Pinheiro
RF
de Sa Moreira
E
Silva
MR
Alberto
FL
Chauffaille Mde
L
FLT3 internal tandem duplication during myelodysplastic syndrome follow-up: a marker of transformation to acute myeloid leukemia.
Cancer Genet Cytogenet
2008
183
2
89
93
31
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
32
Frohling
S
Schlenk
RF
Breitruck
J
et al
Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm.
Blood
2002
100
13
4372
4380
33
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
34
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
35
Ravandi
F
Kantarjian
H
Faderl
S
et al
Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse.
Leuk Res
2010
34
6
752
756
36
Levis
M
Ravandi
F
Wang
ES
et al
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse.
Blood
2011
117
12
3294
3301
37
Druker
BJ
Sawyers
CL
Kantarjian
H
et al
Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome.
N Engl J Med
2001
344
14
1038
1042
38
Druker
BJ
Talpaz
M
Resta
DJ
et al
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med
2001
344
14
1031
1037
39
Levis
MJ
Will newer tyrosine kinase inhibitors have an impact in AML?
Best Pract Res Clin Haematol
2010
23
4
489
494
40
Zheng
R
Bailey
E
Nguyen
B
et al
Further activation of FLT3 mutants by FLT3 ligand [published ahead of print April 25, 2011].
Oncogene
41
Sato
T
Yang
X
Knapper
S
et al
FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo.
Blood
2011
117
12
3286
3293
42
Lyman
SD
Seaberg
M
Hanna
R
et al
Plasma/serum levels of flt3 ligand are low in normal individuals and highly elevated in patients with Fanconi anemia and acquired aplastic anemia.
Blood
1995
86
11
4091
4096
43
Wodnar-Filipowicz
A
Lyman
SD
Gratwohl
A
Tichelli
A
Speck
B
Nissen
C
Flt3 ligand level reflects hematopoietic progenitor cell function in aplastic anemia and chemotherapy-induced bone marrow aplasia.
Blood
1996
88
12
4493
4499
44
Bojko
P
Pawloski
D
Stellberg
W
Schroder
JK
Seeber
S
Flt3 ligand and thrombopoietin serum levels during peripheral blood stem cell mobilization with chemotherapy and recombinant human glycosylated granulocyte colony-stimulating factor (rhu-G-CSF, lenograstim) and after high-dose chemotherapy.
Ann Hematol
2002
81
9
522
528
45
Beran
M
Cao
X
Estrov
Z
et al
Selective inhibition of cell proliferation and BCR-ABL phosphorylation in acute lymphoblastic leukemia cells expressing Mr 190,000 BCR-ABL protein by a tyrosine kinase inhibitor (CGP-57148).
Clin Cancer Res
1998
4
7
1661
1672
46
Pratz
KW
Sato
T
Murphy
KM
Stine
A
Rajkhowa
T
Levis
M
FLT3-mutant allelic burden and clinical status are predictive of response to FLT3 inhibitors in AML.
Blood
2010
115
7
1425
1432
47
Nakano
Y
Kiyoi
H
Miyawaki
S
et al
Molecular evolution of acute myeloid leukaemia in relapse: unstable N-ras and FLT3 genes compared with p53 gene.
Br J Haematol
1999
104
4
659
664
48
Kottaridis
PD
Gale
RE
Langabeer
SE
Frew
ME
Bowen
DT
Linch
DC
Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors.
Blood
2002
100
7
2393
2398
49
Shih
LY
Huang
CF
Wu
JH
et al
Internal tandem duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis of bone marrow samples from 108 adult patients at diagnosis and relapse.
Blood
2002
100
7
2387
2392
50
Fernandez
HF
Sun
Z
Yao
X
et al
Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med
2009
361
13
1249
1259
51
Griffiths
M
Mason
J
Rindl
M
et al
Acquired isodisomy for chromosome 13 is common in AML, and associated with FLT3-itd mutations.
Leukemia
2005
19
12
2355
2358
52
Raghavan
M
Lillington
DM
Skoulakis
S
et al
Genome-wide single nucleotide polymorphism analysis reveals frequent partial uniparental disomy due to somatic recombination in acute myeloid leukemias.
Cancer Res
2005
65
2
375
378
53
Green
C
Linch
DC
Gale
RE
Most acute myeloid leukaemia patients with intermediate mutant FLT3/ITD levels do not have detectable bi-allelic disease, indicating that heterozygous disease alone is associated with an adverse outcome.
Br J Haematol
2008
142
3
423
426
54
Schlenk
RF
Dohner
K
Krauter
J
et al
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
358
18
1909
1918
55
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 [published online ahead of print February 12, 2011].
Biol Blood Marrow Transplant
56
Gale
RE
Hills
R
Kottaridis
PD
et al
No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials.
Blood
2005
106
10
3658
3665
57
Bornhauser
M
Illmer
T
Schaich
M
Soucek
S
Ehninger
G
Thiede
C
Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML [letter].
Blood
2007
109
5
2264
2265
author reply 2265
58
Meshinchi
S
Arceci
RJ
Sanders
JE
et al
Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML [letter].
Blood
2006
108
1
400
author reply 400–401
59
Rowe
JM
Tallman
MS
How I treat acute myeloid leukemia.
Blood
2010
116
17
3147
3156
60
Gupta
V
Tallman
MS
Weisdorf
DJ
Allogeneic hematopoietic cell transplantation for adults with acute myeloid leukemia: myths, controversies, and unknowns.
Blood
2011
117
8
2307
2318
61
Kayser
S
Dohner
K
Krauter
J
et al
Impact of allogeneic transplantation from matched related and unrelated donors on clinical outcome in younger adult AML patients with FLT3 internal tandem duplications [abstract].
Blood
2010
116
21
Abstract 909
62
Bassan
R
Rossi
G
Pogliani
EM
et al
Chemotherapy-phased imatinib pulses improve long-term outcome of adult patients with Philadelphia chromosome-positive acute lymphoblastic leukemia: Northern Italy Leukemia Group protocol 09/00.
J Clin Oncol
2010
28
22
3644
3652
63
Metzelder
S
Wang
Y
Wollmer
E
et al
Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation.
Blood
2009
113
26
6567
6571
64
Sora
F
Chiusolo
P
Metafuni
E
et al
Sorafenib for refractory FMS-like tyrosine kinase receptor-3 (FLT3/ITD+) acute myeloid leukemia after allogenic stem cell transplantation.
Leuk Res
2011
35
3
422
423
65
Martin
PJ
Counts
GW
Jr.
Appelbaum
FR
et al
Life expectancy in patients surviving more than 5 years after hematopoietic cell transplantation.
J Clin Oncol
2010
28
6
1011
1016
66
Knapper
S
Burnett
A
Hills
RK
Small
D
Levis
M
Lestaurtinib FLT3 inhibitory activity is modulated by concomitant azole therapy and may influence relapse risk [abstract].
Blood
2009
114
22
Abstract 326
67
Stone
R
Fischer
JT
Paquette
R
et al
A phase 1B study of midostaurin (PKC412) in combination with daunorubicin and cytarabine induction and high-dose cytarabine consolidation in patients under age 61 with newly diagnosed de novo acute myeloid leukemia: overall survival of patients whose blasts have FLT3 mutations is similar to those with wild type FLT3 [abstract].
Blood
2009
114
Abstract 634
68
Burnett
A
Wetzler
M
Lowenberg
B
Therapeutic advances in acute myeloid leukemia.
J Clin Oncol
2011
29
5
487
494
69
Chlebowski
RT
Kuller
LH
Prentice
RL
et al
Breast cancer after use of estrogen plus progestin in postmenopausal women.
N Engl J Med
2009
360
6
573
587
70
Singh
AK
Szczech
L
Tang
KL
et al
Correction of anemia with epoetin alfa in chronic kidney disease.
N Engl J Med
2006
355
20
2085
2098
71
Gerstein
HC
Miller
ME
Byington
RP
et al
Effects of intensive glucose lowering in type 2 diabetes.
N Engl J Med
2008
358
24
2545
2559
Sign in via your Institution