  
- 积分
- 0
- 威望
- 0
- 包包
- 483
|
作者:Bertrand W. Parcellsa, Alan K. Ikedaa, Tiffany Simms-Waldripa, Theodore B. Moorea, Kathleen M. Sakamotoa,b,c作者单位:a Division of Hematology/Oncology, Department of Pediatrics, Gwynne Hazen Cherry Memorial Laboratories, Mattel Childrens Hospital, Jonsson Comprehensive Cancer Center, Los Angeles, California, USA;b Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of Calif
3 t/ {3 x& Y* _0 T3 i Q' W / y p: Q) ^ J& m
# M: v" [7 B+ L! z8 O
+ q& R" l; e: R* c/ E. T " O% l t/ p% t
& x8 j* B# ?- G3 n# B! t
0 G2 t0 n4 I. f* P" s! A
0 r% f: Q9 M! ?& l! H4 Q1 p
9 J9 \( |( A* ^% H' X) L! h% i" W
9 _% c5 l e7 J6 I) g # j; f( X2 ]3 b& o) K4 l
9 _1 K W4 f- F4 S . W1 K# i5 w. s0 j ~* k; w# _* _
【摘要】
3 B, w5 {( V, {0 [ Ligand-mediated activation of the FMS-like tyrosine kinase 3 (FLT3) receptor is important for normal proliferation of primitive hematopoietic cells. However, activating mutations in FLT3 induce ligand-independent downstream signaling that promotes oncogenesis through pathways involved in proliferation, differentiation, and survival. FLT3 mutations are identified as the most frequent genetic abnormality in acute myeloid leukemia and are also observed in other leukemias. Multiple small-molecule inhibitors are under development to target aberrant FLT3 activity that confers a poor prognosis in patients. 5 q4 t1 H3 f, _8 U2 H+ s8 u
【关键词】 FMS-like tyrosine kinase Hematopoiesis Acute myeloid leukemia Internal tandem duplication Small molecule inhibitor
/ j: R# g; u8 u% m$ e INTRODUCTION
& W2 g1 @: Q! k5 K; X m$ e: \# D* @
The hematopoietic system is organized into a strict hierarchy that sustains a steady-state production of more than one million blood cells per second . The hematopoietic system has evolved to offset the cellular processes of differentiation and proliferation to limit opportunities for oncogenic mutations. However, in spite of the molecular and systemic checks and balances, malignancies arise in the hematopoietic system as various forms of leukemia.
: { n8 N! w/ K) Z5 [. G- G" @+ F6 I* |- s) D- f1 g
Acute myeloid leukemia (AML) is a heterogeneous malignant disorder of myeloid precursor cells with clinical features of increased blasts in the blood and bone marrow . Mutations leading to the constitutive activation of the FMS-like tyrosine kinase 3 (FLT3) receptor, also known as fetal liver kinase 2 and human stem cell kinase 1, have been identified as the most frequent genetic disorder in AML and also confer a poor clinical prognosis. This review examines the function of normal FLT3 signaling and the aberrant downstream signaling caused by FLT3 mutations that evade normal control mechanisms. Subsequently, the clinical implications associated with this oncogenic pathway and the potential for targeted therapy are considered. b' y0 [+ `. Q9 E ~7 b0 p& `
2 Y$ g. i( k# e, ^( d9 Y j
FLT3 IN NORMAL HEMATOPOIESIS
5 j& w& p' _7 Z) d/ Y
5 f* v$ F; X2 w V2 [! Z* t9 GFLT3 Ligand. T1 |$ d% L' p H9 }$ S
; V3 [' L+ ?# {
Several cytokines in the bone marrow act through stem cell-specific or progenitor cell-specific receptors to regulate the capacity of immature hematopoietic cells to potentiate downstream multilineage expansion. The FLT3 ligand (FL) regulates early hematopoiesis by stimulating the FLT3 signal transduction pathway. FL is a type I transmembrane protein belonging to a small family of cytokines including stem cell factor and macrophage colony-stimulating factor (CSF) .
8 N/ r# k; o8 m6 X5 l' n5 ]2 W& O" B0 M$ J
FLT3 Receptor& V% @9 G7 }1 b% ~
1 ]! r' |. s' Y4 E) qFL associates with the transmembrane receptor FLT3, a member of the type III receptor tyrosine kinase (RTK) subfamily that includes c-KIT, c-FMS, and platelet-derived growth factor (PDGF) /ß (Fig. 1).
1 C$ n8 A& Q# \3 K2 U9 a5 H) N6 b$ P: ~& n# S, C& c$ v, @
Figure 1. Structure of the FLT3 receptor. Abbreviations: FLT, FMS-like tyrosine kinase 3; ITD, internal tandem duplication.
& h4 u& `0 p# j1 | e- c
4 u( G$ I1 j/ j7 wFLT3 Receptor Expression
) ^6 [# ~' F8 P5 \1 ~/ V% E( I2 |1 r& F, I. x) e1 C" L
The narrow range of cells expressing the FLT3 receptor primarily determines the specificity of FL signaling. FLT3 expression in the bone marrow is restricted to CD34 cells and the subset of dendritic precursor cells. FLT3 expression is correlated with "short-term" reconstituting HSCs, the Lin Sca-1 c-Kit FLT3 compartment .7 i% |0 H3 E$ ?
q1 V/ H% P9 ?( iSynergy of FL with Other Cytokines. P0 h' f. a+ B! R/ d- x2 T$ l
+ i! M- c7 k* Y
Experimental evidence indicates that FL signals synergistically with other growth factors to promote proliferation in early progenitor cells of the myeloid and lymphoid lineages. FL-mediated signaling is highly dependent on related growth factors such as interleukin 3 (IL-3), granulocyte colony-stimulating factor (G-CSF), colony-stimulating factor-1 (CSF-1), and granulocyte macrophage colony-stimulating factor (CM-CSF), because FL inefficiently promotes proliferation as a single cytokine in vitro ., V9 g, U" Z1 y
* v+ \" P1 z$ ?# s' @& `; c2 ?Normal FL signaling is also associated with proliferation of the lymphoid lineage. In fact, targeted disruption of FL in mice led to a particular reduction of lymphoid precursor cells within the general reduction of primitive hematopoietic cells. FL¨C/¨Ccaused impairment of the immune system by reducing numbers of pro-B cells, dendritic cells (DCs), and natural killer cells .
8 _* B6 o+ Z2 y% x) o' g% m; \/ V- v7 \) W- N& d
FLT3 Receptor Signaling8 g/ Y6 ?* c# l5 Z+ e0 ^0 d5 ~
' g7 M0 Y# p4 MFL binds to the FLT3 receptor to induce formation of a homodimer in the plasma membrane. The dimer couples cytoplasmic domains thereby enabling transphosphorylation of specific tyrosine residues, likely Tyr-589 and Tyr-591, on the JM domain . The FLT3 signal-transduction pathways are not conclusively mapped, yet current understanding of downstream pathways provides molecular support for its vital role in proliferation and apoptosis of primitive hematopoietic cells.
' X9 x2 u6 x* M _/ Y' |5 y. `
7 E; s( C: H5 O" ^# tDespite the observed molecular effects of the FLT3 pathway, knockout mice have relatively stable hematopoiesis . Although these results indicate the presence of functional redundancies between hematopoietic growth factors, they do not detract from the role of FLT3 in differentiation and proliferation of early progenitor cells of the myeloid and lymphoid lineages.- B7 n- D8 d/ G& r. _
8 p* @; F" {9 M- Y2 Y2 t4 h2 I
FLT3 IN LEUKEMIA' [+ V) ~3 m: a8 Z
0 n, Q4 Y. y4 i, B
Self-renewal and growth factor-independent proliferation are two important traits for oncogenesis. The involvement of FLT3 in proliferation of highly undifferentiated hematopoietic cells suggests the oncogenic potential of this signaling pathway. Clinical and experimental evidence both indicate that FLT3 is a proto-oncogene with the capacity to enhance survival and proliferation of leukemia blast cells. Wild type FLT3 is expressed in a wide range of hematopoietic malignancies, including acute lymphoid leukemia and mixed lineage leukemia; most notably, it is expressed in 70%¨C100% of AML . Two types of mutations have been attributed to the deregulated FLT3 receptor.
& M: a$ w# e( v, i2 ?! m
; m% q0 @# _& JFLT3 Internal Tandem Duplication Mutation
, a) a: l* g% \& X x' ?- F8 F2 ?8 s
An in-frame internal tandem duplication (ITD) mutation in the JM domain of the FLT3 receptor correlates with the highest frequency of FLT3 related AML cases. Clinical studies identify the FLT3-ITD mutation in 17%¨C26% of AML cases .
' z# s' [, h" V& ^! \6 z, P+ b, b4 X& L4 t7 k5 u% `/ f- j( ^$ _
The ITD mutation causes constitutive activation of the FLT3 receptor. The JM region of many RTKs acts through various mechanisms to regulate the kinase domain. The crystal structure of the normal FLT3 receptor offers direct insight into the mechanism used by the JM domain to regulate catalytic activity of the kinase domain and suggests how the ITD disrupts this mechanism. FLT3 activity occurs when normal phosphorylation of specific tyrosine residues prevents the JM domain from folding properly to induce autoinhibition . The ITD insertion generally occurs near the JM hinge region and may offset the JM orientation thereby causing a "leaky" autoinhibition of catalytic activity.# a/ ^* K& }, M
4 e8 L N+ |: u5 D. ~2 RThere is no correlation between the size of the insertional mutation and the degree of autophosphorylation . Such conflicting results may be reconciled by further analysis into the ratio of wild type to mutant receptors on the plasma membrane. This ratio may affect the degree of autophosphorylation because the FLT3-ITD receptor can homodimerize with mutant receptors or heterodimerize with wild-type receptors independent of the ligand." I, V* ^. |$ L1 E
7 L/ g/ Q! E( \1 ` Y: p
Single-Amino Acid Mutations in FLT3
. H9 i6 R$ M ^
. X- x3 w0 x' P" ]) N4 [% BMissense point mutations in the kinase domain can also confer constitutive activation of the FLT3 receptor. The most common activating point mutation is the substitution of tyrosine for aspartic acid at position 835 within the activation loop of the kinase domain. Point mutations at other positions, such as 836 or 841, have also been associated with FL-independent activation .4 e) d' U' ?; q! {+ \; F6 w5 {
& l* { ~: @: _9 ]# F/ u- N6 w
FLT3 Mutant Signaling5 p9 Z0 P% U* M
5 }1 c- X9 _0 R
Parallels between the FLT3 receptor and related RTKs suggest that activating mutations disrupt an autoinhibitory mechanism that is critical for maintaining the inactive conformation of the kinase domain. The mutant receptor activates effector proteins to mediate proliferation and survival and blocks differentiation through ligand-independent autophosphorylation. Identifying molecules of the mutant signaling pathway offers insight into its role in the pathogenesis of leukemia (Fig. 2).' `. g( q( l: P: A7 J- Q: o
1 U- i4 b$ i# h) _
Figure 2. Signal transduction pathways downstream of FMS-like tyrosine kinase-3 receptor activation. Abbreviations: C/EBP, CCAAT/ enhancer-binding protein ; ERK, extracellular-signal regulated kinase; MAP, mitogen-activated protein; PI-3 kinase, phosphatidylinositol 3-kinase.: A v) z( ?9 [' S0 ~; H
9 @$ S* V$ ^& u- gAberrant stimulation of proteins that confer a proliferative advantage was first identified in mutant-FLT3 myeloid leukemia cells. In vitro, the 32D mouse myeloid cell line harboring the ITD-mutation showed factor-independent proliferation and resistance to radiation-induced apoptosis .
) i( k1 J2 h. h( `8 z7 T9 m% v
. E7 ]/ b6 T5 UConstitutively phosphorylated STAT5 has been observed in primary AML blasts and in immortalized cell lines Ba/F3 and 32D that express the mutated FLT3 receptor . It remains unclear whether mutant FLT3 receptors promote leukemogenesis by increasing quantitative signaling of normal effector proteins or by causing qualitative differences in downstream targets.
4 T. p3 D+ L; u: D! V! l5 P1 r5 x4 h1 E) m. k( r% ^- S
STAT5 is known to activate PIM-1, and RNA microarray data identified a consistent increase in PIM-1 expression in FLT3-ITD cells . Quantitative PCR further implicated PIM-1 as a target of aberrant STAT5 expression by correlating a 10-fold decrease in its expression with FLT3 inhibition. PIM-1 is normally induced by a number of FL-related cytokines, including G-CSF and IL-3, to increase cell mitogenesis and survival. The effect of PIM-1 activity occurs through phosphorylation of a diversity of substrates including Cdc25A, which is key to cell cycle progression, and Bad, which blocks proapoptotic signaling. PIM-1 has also been reported to cooperate within antiapoptotic pathways in chronic myeloid leukemia (CML) cell lines.
4 q) I, h+ G. B* I! W4 }( }7 G! w# @* V' A- Y
FLT3 and Differentiation4 A) T' \2 j$ }
7 p, w2 s0 U' b7 [9 Q5 t' LFLT3 activity may also regulate differentiation pathways. Differentiation is tightly associated with the loss of proliferative capacity and increased propensity toward apoptosis. Normal myeloid progenitor cells proceed along a differentiation gradient to arrive at a terminally differentiated cell type with a limited lifespan. Blocking differentiation has been identified as an important mechanism for cancer progression. The preferential expression of FLT3 in primitive hematopoietic cells suggests its role in regulating differentiation. FL activation of 32D cells transfected with wild-type FLT3 mitigated their progression toward differentiated neutrophils but could not induce a complete block .! t V# d. d+ g
6 [- O6 b. B( _/ Q' q
However, RGS2 mRNA, a regulator of G-protein signaling, was observed to be significantly repressed in the majority of primary AML bone marrow samples that harbor the FLT3-ITD mutation compared with AML samples lacking the mutation . Several myeloid cell lines have been shown to induce RGS2 during granulocytic differentiation, and RGS2 overexpression in 32D cells expressing FLT3-ITD overcomes the block to differentiation. The study suggests that FLT3-ITD signaling represses differentiation to some degree in AML cases, depending on the cellular context.
) N& }7 V' f5 H8 R1 A0 X
9 W! \( @5 I. k$ O& }FLT3 Phosphatases3 z6 R/ W) G5 ^" P6 Z
; ?& y2 S: z* {) n5 R
The constitutive activation of FLT3 may be further promoted by suppression of protein-tyrosine phosphatases (PTPs). The SH2 domain-containing PTPs SHP-1 and SHP-2 are commonly involved in regulating cytokine- and growth factor-mediated signaling pathways . SHP-1 loss of function led to factor-independent growth. Increased phosphatase activity normally occurs in response to kinase activation, yet the observed SHP-1 suppression common to RTK-mediated malignancies suggests the importance of overcoming PTP activity to shift the phosphorylation balance toward a proliferative advantage. SHP-1 also regulates signaling of related hematopoietic receptors, and its suppression by FLT3 may induce synergistic effects by sensitizing cells to multiple growth factors.
* e# Z$ `2 n; W8 w( w) c
# l& g" [9 z+ t8 f: _' v- E" ^FLT3 and In Vivo Models of Leukemia% l. X' f5 l( F, Y, ~; u9 ~- h
/ h% m+ O- i9 A& H& h2 [3 g6 E5 bAt the molecular level, FLT3 appears to recruit many proteins involved in proliferation, self-renewal, and anti-apoptosis. The expression profiles of mutant FLT3 cell lines support its association with leukemogenesis by identifying a number of genes involved with growth and survival. However, the diversity of protein function and the redundancies in cellular processes prevent clear correlations between proteins and their function.
% j/ k& [% i) Z- `9 J
9 o6 x' Q/ s1 ? L [& p0 \/ WFLT3 mutations are insufficient to induce leukemia blasts in vivo . The association of the PML/ RAR mutation with block to differentiation suggests that although mutant FLT3 may effect differentiation, transformation depends on a more potent mechanism. The block to differentiation may be a critical limitation for leukemogenesis through aberrant FLT3 signaling.. } U% u" }+ j+ x! l, ^
: X3 V! O# p( W: a2 @Nevertheless, these studies are critical to implicating the mutated FLT3 receptor in leukemogenesis. As a single mutation, FLT3 induces a myeloproliferative phenotype, and primary AML cells expressing the FLT3-ITD receptor engraft into NOD/SCID mice with greater efficiency that those lacking the mutation .4 g; u/ F, c5 d; r
+ X1 n# U" ~9 M7 x# E
Clinical Implications of FLT3 Mutations in AML/ F' J7 } E4 C. J/ |
, t' J; i) u9 n, @3 a' U- D) c* Z
Substitution mutations in the tyrosine kinase domain (TKD) and insertional mutations in the JM domain of FLT3 represent the most frequent genetic abnormality associated with AML. Both mutations lead to constitutively active FLT3, although studies indicate that each mutation presents unique clinical features.2 o; d$ ]* g0 o7 f; I9 m
. m) p$ T! D: V( O; J! s& b1 ]. I
The FLT3-ITD mutation is recognized at diagnosis by leukocytosis, with the mean white blood cell (WBC) count typically elevated to levels significantly higher than that of patients without the mutation .
1 [9 G S+ ?) a+ _ ^/ u+ [# D1 b1 X" U8 ^
Multiple studies indicate that FLT3-ITD mutations confer a poor prognosis in AML patients less than 60 years of age .! I( e% J* n1 A( ?" e
5 d4 M1 [+ X* Y' E9 k4 K& y$ j
Multiple studies used PCR to correlate high ratios of mutant to wild-type FLT3 with decreased overall survival . This provides evidence that FLT3 expression levels significantly affect prognosis and relapse. This suggests that the pathway exerts a strong influence on leukemia progression. However, the heterogeneity in AML blast gene expression and the notable fraction of AML patients that lose the FLT3-ITD mutation in relapse indicate that multiple factors determine disease progression and that FLT3-ITD should not be exclusively used as a marker for MRD.
R* q9 W" C$ m$ ]( e
9 _% e+ l8 I* v+ IIn pediatric cases, the ITD mutation provides a more negative and independent prediction of prognosis. Data from six combined pediatric studies, which examined 461 pediatric AML patients, identified a 19% survival rate for cases associated with FLT-ITD compared with a 58% survival rate for patients with wild-type FLT3 receptors .- ^6 A/ L- F3 ~ Z! w0 {
+ s. B7 a- s9 g3 _5 m
The clinical pathogenesis of FLT3-TKD expressing AML is less understood than the FLT3-ITD due to its lower frequency in patients, which limits the number of statistically significant conclusions . Studies have also identified a higher frequency of FLT3-TKD mutations in patients with MLL duplications or a double-stranded break in the MLL gene. Future clinical studies must examine larger AML populations to achieve significant insight into potential effects of FLT3-TKD mutations.: Q: b* [- @- K9 E1 |
- i' h" T: u. ~FLT3 INHIBITORS( Q- v1 Z2 I8 I# }9 i/ l& Z
- n* W- Q. I6 A* a/ U
The fundamental principle of small-molecule therapy is to inhibit specific RTKs involved with tumorigenesis while minimizing the inhibition of normal cellular signaling to limit toxicity. The recognition of small-molecule inhibitors as therapy for deregulated RTKs emerged in the wake of the clinical success of imatinib mesylate in CML. CML progresses almost exclusively through the constitutively active bcr-abl fusion protein tyrosine receptor. Its robust response to imatinib provided proof of principle for the potential efficacy of targeted therapy . Preliminary clinical data from therapeutic targeting of the FLT3 mutations in AML suggests that small-molecule inhibitors may effectively complement traditional therapy regimens to treat a broader range of malignancies.
6 _ C/ m7 I- G! |. c
+ ]! t& K! ?8 c! K _, O7 @FLT3 is an appropriate candidate for targeted therapy because it is expressed in many hematopoietic malignancies, it is the most frequent molecular abnormality in AML, it confers a poor prognosis, and its signaling cascade has been implicated in multiple tumorigenic pathways. Currently, there are several small-molecule therapies at various stages of development that target mutant forms of the FLT3 receptor (Table 1). All of the FLT3 inhibitors being studied are heterocyclic compounds with a purine ring-like subunit that competitively inhibits ATP binding to FLT3 . Data on related receptors suggests that the molecules inhibit activity by entering into the ATP binding pocket through an induced fit or lock-and-key manner, although the specific mechanism may vary between inhibitors. A brief summary of the molecules is given below.
* L/ `0 R, d: U( b, M
. t4 P# D& b& b7 d4 ~7 X5 b! vTable 1. Small-molecule tyrosine-kinase inhibitors
& S6 w7 N5 k: z& ?% g
% p+ C# _' L' hThe compounds AG1295 and AG1296 were the first to be identified as inhibitors of cells harboring the FLT3-ITD mutation through a similar screening process used to identify imatinib .7 l" f+ F. H; v n) j+ Y
9 @9 B, i2 I: w) q& g; @# c( u* s( \SU5416 was the first indolinone compound to be identified as a potential inhibitor of FLT3 activity. It has also been shown to inhibit vascular endothelial growth factor 2 (VEGF2) and c-KIT .1 M, d! `' C3 i/ o
5 G5 L, z2 e7 \. L8 p8 ?: H1 }/ B9 m
The indolinone derivative PKC412 (4'-N-benzoylstaurosporine) was originally identified as an inhibitor of protein kinase C . The drug showed measurable activity, evidenced by a 50% reduction in peripheral blast count in 14 patients, a greater than 2-log reduction in peripheral blast count in seven of these patients, and a 50% reduction in bone marrow blast counts in six patients. PKC412 was generally well tolerated.
. U0 s$ E, e d! c; A
# G; s' J; v7 S3 nThe compound SU11248 is another indolinone derivative shown to inhibit FLT3, as well as VEGF2, PDGFß, and fibroblast growth factor 1 (FGF1) . One-third of patients exhibited a significant decrease in FLT3 phosphorylation, although a minority of patients experienced gastrointestinal or cardiac symptoms of toxicity.
$ O& V, w& o; X7 l ]+ |# Z
" D- j' O5 g# t( c, E1 f6 I5 WMLN518 (CT53518) is a piperazinyl quinazoline that inhibits growth of FLT3-ITD-transformed cells in vitro and in vivo .
, Y2 d$ B) b2 }( m) q; ]' ]% m
) o. h' p5 o4 f9 t) Y0 a% eThe indolocarbazole derivative CEP-701 was originally identified as an inhibitor of TrkA tyrosine kinase . Most responses were observed by reduced peripheral blast counts; however, one patient showed a 95% reduction of bone marrow blasts before relapse 3 months later. Minimal toxicity was observed.
: e- R2 y9 {$ |1 }$ B, s
- }& w) J" ^) w m" ?. YKomeno et al. recently identified Ki23819 as a potent inhibitor of mutant FLT3. The compound suppressed proliferation of MV4¨C11 cells at IC50 of 1 nM . These preliminary in vitro results suggest promising translation into clinical inhibition of mutant FLT3, but the compound requires further study in mouse models. To summarize, most clinical studies with FLT3 inhibitors have demonstrated a moderate response, more evident in the peripheral blood than in bone marrow.
; {1 Y8 K8 Q. r% ^% B9 k, F* K3 b4 E4 b& o4 Y
Resistance, C6 t1 g* g( p# X/ i( f( {* H
4 q& m0 i+ F8 d4 r
The development of small-molecule inhibitors is widely encouraged within the medical community because it offers an approach to treat cancer with relatively modest toxicity. Yet the targeting specificity that bestows a clinical advantage to this therapy may also be a critical flaw. The sensitivity of the compounds to the FLT3 structure confers not only specificity but also susceptibility to minor structural changes that are induced by mutation. The high mutational load that most tumors support during progression indicates a high probability of drug-resistant subclones. A mathematical model that closely recapitulates clinical findings from CML predicts that most leukemias will contain cells resistant to multiple molecular inhibitors at the time of diagnosis .
' k1 L+ s1 V7 b% y/ G2 I. b
* C$ n5 ?7 e. z3 l& }- V; UAcquired resistance to small-molecule inhibition has been identified in AML blasts harboring constitutively active FLT3 receptors . These mutations recovered sensitivity to PKC412, SUS5614, and a CEP-701-like compound at higher concentrations in vitro, yet increased drug concentrations may not translate into a successful method for circumventing resistance in clinical AML. Many FLT3 inhibitors were originally identified as inhibitors of other RTKs and became directed specifically against FLT3 because the receptor showed inhibition at the lowest concentration threshold. If mutations can raise the concentration threshold to a level at which other RTKs respond, then normal cells may experience significant cytotoxicity, and clinical side effects may become severe. Evidence of acquired resistance does not invalidate the clinical potential of targeted therapy but suggests that curative treatment must extend beyond a single agent.
% |; y( N: i4 Q% V4 t8 X2 s
2 H8 ^1 \( `. B6 JData from in vitro and in vivo experiments offers promising strategies for combination therapy. The synergistic effects of SU11657 and all-trans retinoic acid induced rapid regression in an APL mouse model . The treatment regimen examined in this study is currently being applied to a clinical trial. The diversity of potential combination therapies and the significance of sequence in drug administration indicates that substantial optimization studies must be conducted before treatment regimens are designed for specific genetic abnormalities.1 E8 [: f# Q8 J
6 g$ h- a$ q7 _8 w3 w
Many of the aforementioned compounds may translate into effective treatments for cancer, yet it is possible that they will only provide a selective force for highly resistant clones. One particular point mutation in the FLT3 receptor, Gly-697, poses a significant challenge because its structure confers resistance to multiple compounds, including PKC412, SU5614, and a molecule similar to CEP-701 . Similar to the CML blast crisis, AML blast cells that harbor FLT3 mutations are likely to have additional mutations that may drive tumor progression while showing no response to FLT3 inhibition. Ideally, the response rate for FLT3 inhibition will compare to the 30% response to imatinib during CML blast crisis. Identification of new molecules will be key to preventing resistance in the future. Combining compounds that inhibit various proteins within the signaling pathway may also improve the efficacy of treatment.
- Q: U! M7 W3 h: A
7 m, i8 z+ t; i& w6 Y( FACKNOWLEDGMENTS
0 d1 I3 i8 e2 l/ u, E( _
* k; k: W& T8 l0 q5 N9 @8 K. lB.W.P. is supported by the Skirball Foundation (awarded to T.B.M.). K.M.S. is a Scholar of the Lymphoma and Leukemia Society and is supported by the NIH (CA108545 , HL 75826, RHL083077A), American Cancer Society (RSG-99-081-04-LIB), Department of Defense (CM050077), and the Diamond-Blackfan Anemia Foundation. Both T.B.M. and K.M.S. are funded by the UCLA Jonsson Comprehensive Cancer Center.
8 w' H. G* \* ~7 M
2 E/ Z5 V0 H2 w/ K$ w, ]3 NDISCLOSURES
% J2 N* |& P& G
) W y+ M% C6 C4 U% J QThe authors indicate no potential conflicts of interest. a6 e+ M: v0 Y6 T" c
【参考文献】
- a& u, N9 e# u J* Q7 Y / Q, w9 s! ~; i; e
1 L" v6 x' v; X
Ogawa M. Differentiation and proliferation of hematopoietic stem cells. Blood 1993;81:2844¨C2853.
' ^0 v- d) h5 @ S+ \/ B% E8 w4 W4 s( W! x) ^/ Q
Metcalf D. Hematopoietic regulators: Redundancy or subtlety? Blood 1993;82:3515¨C3523.; [ N0 ~% q& J9 s1 C
) O8 y* n: Q9 i. ~( P
Shivdasani RA, Orkin SH. The transcriptional control of hematopoiesis. Blood 1996;87:4025¨C4039.
$ e4 C) m/ X+ I4 S }; H" F2 n" N/ }! o6 A, y8 k$ H
Zhu J, Emerson SG. Hematopoietic cytokines, transcription factors and lineage commitment. Oncogene 2002;21:3295¨C3313.
" \1 C! J. A! @: n1 Q1 Y( l! }/ {* ^7 Z7 I+ ]" O K
Lyman SD, Jacobsen SE. c-kit ligand and Flt3 ligand: Stem/progenitor cell factors with overlapping yet distinct activities. Blood 1998;91: 1101¨C1134.) n+ ?$ S, b" E' ]
1 m7 J5 `+ q- F- } m: d8 ?+ e
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;368:643¨C648.
8 m/ w1 s) |0 K* B5 J% f" n6 d! u# Z
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;83:2795¨C2801.
9 m1 v" k& ~ N6 ?; F' p' d
( b! T' s% [2 x1 _( v, bLyman SD, James L, Vanden Bos T et al. Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor: A proliferative factor for primitive hematopoietic cells. Cell 1993;75:1157¨C1167.
+ j3 ?2 z A! O$ ^6 J; h) s4 o) O. @/ {; K) o' B9 Z$ s$ t H
Zheng R, Levis M, Piloto O et al. FLT3 ligand causes autocrine signaling in acute myeloid leukemia cells. Blood 2004;103:267¨C274.. H( n8 S! ^# c- e- Y- Y% r5 k8 E
& c; E! B- n& ~. l
Chklovskaia E, Jansen W, Nissen C et al. Mechanism of flt3 ligand expression in bone marrow failure: Translocation from intracellular stores to the surface of T lymphocytes after chemotherapy-induced suppression of hematopoiesis. Blood 1999;93:2595¨C2604." l' L7 S, C/ e! o
: G9 R9 @8 R- I! i2 C; Y6 t9 TWodnar-Filipowicz A. Flt3 ligand: Role in control of hematopoietic and immune functions of the bone marrow. News Physiol Sci 2003;18:247¨C251.( Z! F0 i. o1 A5 X M& E
/ p+ V8 C$ I* c A; IMathews LS, Vale WW. Expression cloning of an activin receptor, a predicted transmembrane serine kinase. Cell 1991;65:973¨C982./ [' X& b; r* s: P
% ^1 R, F2 H: j& K6 tRosnet O, Mattei MG, Marchetto S et al. Isolation and chromosomal localization of a novel FMS-like tyrosine kinase gene. Genomics 1991; 9:380¨C385.6 r" `6 [& z& W: A* Z, I0 N
0 M0 l9 M" k, ]* m `& k$ dRosnet O, Schiff C, Pebusque MJ et al. Human FLT3/FLK2 gene: CDNA cloning and expression in hematopoietic cells. Blood 1993;82: 1110¨C1119., l8 ?( G7 s' _" A0 a
7 ?/ ?" X+ c2 v+ N$ k, ^& Y0 T: CSmall 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;91:459¨C463.. C4 r g: k' |9 W1 V0 |8 t' ]
# c7 f- T9 w1 H. `9 ]Abu-Duhier FM, Goodeve AC, Wilson GA et al. Genomic structure of human FLT3: Implications for mutational analysis. Br J Haematol 2001;113:1076¨C1077.5 O* g( ~& L5 ^* i- K4 n. \
4 W% p" y+ K. f" c
Agnes F, Shamoon B, Dina C et al. Genomic structure of the downstream part of the human FLT3 gene: Exon/intron structure conservation among genes encoding receptor tyrosine kinases (RTK) of subclass III. Gene 1994;145:283¨C288.# d% B9 G7 |2 N- W* y6 Y
( T& T! T& j' q4 R$ D
Carow CE, Kim E, Hawkins AL et al. Localization of the human stem cell tyrosine kinase-1 gene (FLT3) to 13q12¨C>q13. Cytogenet Cell Genet 1995;70:255¨C257.
) Y! s( L4 a8 g, {/ Q8 u
3 ^, J# P+ }+ A5 {Lyman SD, James L, Zappone J et al. Characterization of the protein encoded by the flt3 (flk2) receptor-like tyrosine kinase gene. Oncogene 1993;8:815¨C822.2 t a7 U) G4 b: D K: E, S
3 {2 G! C2 R: G$ e7 L+ A
Carow CE, Levenstein M, Kaufmann SH et al. Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood 1996;87:1089¨C1096.
7 d3 B9 b% ~9 U0 \
% L( Z% W2 d, [, iRosnet O, Marchetto S, deLapeyriere O et al. Murine Flt3, a gene encoding a novel tyrosine kinase receptor of the PDGFR/CSF1R family. Oncogene 1991;6:1641¨C1650.7 f s. q7 Y" T' `* X) S4 T
. D$ v+ a/ E4 ^. N% yAdolfsson J, Mansson R, Buza-Vidas N et al. Identification of Flt3 lympho-myeloid stem cells lacking erythro-megakaryocytic potential: A revised road map for adult blood lineage commitment. Cell 2005;121: 295¨C306.9 v. W& X* X3 ~
0 n( B$ m8 N) G: R; V1 }
Adolfsson J, Borge OJ, Bryder D et al. Upregulation of Flt3 expression within the bone marrow Lin(¨C)Sca1( )c-kit( ) stem cell compartment is accompanied by loss of self-renewal capacity. Immunity 2001;15: 659¨C669.* _: k2 ^8 S5 y5 l* y8 K1 J
9 O$ {) l: v1 L3 y& r, {Christensen JL, Weissman IL. Flk-2 is a marker in hematopoietic stem cell differentiation: A simple method to isolate long-term stem cells. Proc Natl Acad Sci U S A 2001;98:14541¨C14546.
% P% c- b/ S( R# k+ s
6 _- H/ f! M. P- e( J/ ZEbihara Y, Tsuji K, Lyman SD et al. Synergistic action of Flt3 and gp130 signalings in human hematopoiesis. Blood 1997;90:4363¨C4368.9 Z- N* o2 Z! R! X! D
' V' R/ t0 y9 F( ^ `Lyman SD. Biology of flt3 ligand and receptor. Int J Hematol 1995; 62:63¨C73.
6 f9 s! S6 c2 u) z1 D$ ~+ Z
3 e! y, ~" y; B9 ALyman SD, Brasel K, Rousseau AM et al. The flt3 ligand: A hematopoietic stem cell factor whose activities are distinct from steel factor. STEM CELLS 1994;12(suppl 1):99¨C107; discussion 108¨C110.# _& |/ r# y' v6 r
5 R- I2 e7 l! W& o: R2 h8 f$ q% [Ray RJ, Paige CJ, Furlonger C et al. Flt3 ligand supports the differentiation of early B cell progenitors in the presence of interleukin-11 and interleukin-7. Eur J Immunol 1996;26:1504¨C1510.& \! H) o) D6 ]
8 `. G0 d8 }0 }: }) t9 f/ D. E
Rusten LS, Lyman SD, Veiby OP et al. The FLT3 ligand is a direct and potent stimulator of the growth of primitive and committed human CD34 bone marrow progenitor cells in vitro. Blood 1996;87:1317¨C1325.
6 N9 n' Q$ o' Z" q" W& `5 }" G7 `0 I; g
Brasel K, McKenna HJ, Morrissey PJ et al. Hematologic effects of flt3 ligand in vivo in mice. Blood 1996;88:2004¨C2012.$ v* ]) D9 _6 S, s7 K- R. P
3 e9 y x/ n! r$ V: T( D# `- m
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;95:3489¨C3497.
" O; k+ k# S* r* C" K |& Y
* A: p' u2 A' I+ \& K& DMaraskovsky E, Brasel K, Teepe M et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: Multiple dendritic cell subpopulations identified. J Exp Med 1996;184:1953¨C1962.( c" a* f* r5 K0 m3 a) Y
! P9 V1 w& Y3 HSaunders D, Lucas K, Ismaili J et al. Dendritic cell development in culture from thymic precursor cells in the absence of granulocyte/ macrophage colony-stimulating factor. J Exp Med 1996;184:2185¨C2196.! X0 c0 K0 b) E, S! c
) Z; |0 c0 h- @& E$ H. i0 ~Maraskovsky E, Daro E, Roux E et al. In vivo generation of human dendritic cell subsets by Flt3 ligand. Blood 2000;96:878¨C884.
3 {& d! Q* v J6 V; h, K& {$ Q2 u1 T7 Y$ U% L$ s. \
Chakravarty PK, Alfieri A, Thomas EK et al. Flt3-ligand administration after radiation therapy prolongs survival in a murine model of metastatic lung cancer. Cancer Res 1999;59:6028¨C6032.# L0 s H2 n# C* m7 y/ P9 B: P
* E1 o9 J* z5 n
Chen K, Braun S, Lyman S et al. Antitumor activity and immunotherapeutic properties of Flt3-ligand in a murine breast cancer model. Cancer Res 1997;57:3511¨C3516.
5 |9 H( _6 x3 H- j
1 D$ ]9 G& I7 `) H9 @5 RLynch DH, Andreasen A, Maraskovsky E et al. Flt3 ligand induces tumor regression and antitumor immune responses in vivo. Nat Med 1997;3:625¨C631.
1 X# u& B4 D" i: u# P6 l
4 t% M( g+ ^& ~1 x" ?Pawlowska AB, Hashino S, McKenna H et al. In vitro tumor-pulsed or in vivo Flt3 ligand-generated dendritic cells provide protection against acute myelogenous leukemia in nontransplanted or syngeneic bone marrow-transplanted mice. Blood 2001;97:1474¨C1482.. H; t, s" J# |+ p6 P
. R! s+ n) _) P, q: _
Griffith J, Black J, Faerman C et al. The structural basis for autoinhibition of FLT3 by the juxtamembrane domain. Mol Cell 2004;13:169¨C178.6 T3 W8 O% P! z( \ T
# T/ h' u2 _$ ~- r5 QDosil 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: 6572¨C6585.
' P, u4 l/ S- X7 Y4 _/ a7 u, W a, [, Y9 ~! @) @/ W v
Rottapel R, Turck CW, Casteran N et al. Substrate specificities and identification of a putative binding site for PI3K in the carboxy tail of the murine Flt3 receptor tyrosine kinase. Oncogene 1994;9:1755¨C1765.
s6 ]: B) @# J- Z
" }# r+ A9 N- W3 J3 p! ~+ t% F3 @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;192:719¨C728.( q& a9 D1 y6 d
+ {* O, m1 h! F. D/ d6 Z: V7 vMarchetto S, Fournier E, Beslu N et al. SHC and SHIP phosphorylation and interaction in response to activation of the FLT3 receptor. Leukemia 1999;13:1374¨C1382.
$ B C/ B5 T" u; K4 ~6 j$ z) M8 T. _ y# y9 o+ w- I& [
Mackarehtschian K, Hardin JD, Moore KA et al. Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors. Immunity 1995;3:147¨C161.* ]# D6 N+ Y- s/ f' U5 a% E
1 H3 N, w6 ~0 n3 nBirg 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;80:2584¨C2593.
% ~' L0 Z1 }1 Z. r z g! ]1 ?9 Q. c3 @- H" u9 y
Birg F, Rosnet O, Carbuccia N, Birnbaum D. The expression of FMS, KIT and FLT3 in hematopoietic malignancies. Leuk Lymphoma 1994; 13:223¨C227.4 u/ |- G: v' S- L6 n* U
" W/ g2 m1 B2 G* yLevis M, Small D. FLT3: ITDoes matter in leukemia. Leukemia 2003; 17:1738¨C1752.( H7 w) }0 ?. Q* j$ h2 N- w
) T: {% ~; x9 y1 K6 L& P3 j
Kottaridis PD, Gale RE, Frew ME et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: Analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 2001;98:1752¨C1759., O( J: N8 C" a' R
% }# K. p4 q' G2 M( gMoreno I, Martin G, Bolufer P et al. Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia. Haematologica 2003;88:19¨C24.
4 D/ T# g# u1 f! r. e) Q2 J1 A, L% h$ ~9 w8 c0 D" j
Nakao M, Yokota S, Iwai T et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 1996;10:1911¨C1918.
- L3 s) s8 X8 [, f: a' |; i6 i$ d% f5 a# y# Z, C$ l
Schnittger S, Schoch C, Dugas M et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: Correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood 2002;100:59¨C66.( g/ _- r; |% ~3 {# @8 [7 x4 o( w
1 ?8 X9 }- k: T* C4 e
Chia W, Savakis C, Karp R et al. Mutation of the Adh gene of Drosophila melanogaster containing an internal tandem duplication. J Mol Biol 1985;186:679¨C688.* G; }$ [' o! S9 P) b# D
5 e2 {% k: Q3 O* r5 }* V% QKiyoi 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;12:1333¨C1337.
3 @9 t, R$ r# x* w. N) p' ^
, q- Q- Z u+ R& v! A8 u3 eAbu-Duhier FM, Goodeve AC, Wilson GA et al. FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group. Br J Haematol 2000;111:190¨C195.- @# k; \/ o( C
8 n, x1 }+ J% J$ a P+ [Iwai T, Yokota S, Nakao M et al. Internal tandem duplication of the FLT3 gene and clinical evaluation in childhood acute myeloid leukemia. The Children¡¯s Cancer and Leukemia Study Group, Japan. Leukemia 1999;13:38¨C43.3 c6 Q S1 R. T5 n1 z
0 I2 `4 |5 t6 h3 Z
Kiyoi H, Naoe T, Nakano Y et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia. Blood 1999;93: 3074¨C3080.6 [- Z1 ~2 P, y
5 x# O6 f V$ L3 S" w0 |: Q, w3 y' yKiyoi H, Naoe T, Yokota S et al. Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia. Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho). Leukemia 1997;11:1447¨C1452.
( T( X8 N' I8 _4 H" T
, t. A- J# n0 qKondo M, Horibe K, Takahashi Y et al. Prognostic value of internal tandem duplication of the FLT3 gene in childhood acute myelogenous leukemia. Med Pediatr Oncol 1999;33:525¨C529.' T( f. b+ X U$ v; b" D" V
}: S1 ?% t6 e& b0 l
Yokota S, Kiyoi H, Nakao M et al. Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines. Leukemia 1997;11: 1605¨C1609." U& {! d3 O' o& A: U m
3 Z# }* F2 x* Y \
Wybenga-Groot LE, Baskin B, Ong SH et al. Structural basis for autoinhibition of the Ephb2 receptor tyrosine kinase by the unphosphorylated juxtamembrane region. Cell 2001;106:745¨C757.
6 T6 {3 F- p1 l1 Z0 I+ c3 ?
% K4 X! m, B2 D# {1 sChan PM, Ilangumaran S, La Rose J et al. Autoinhibition of the kit receptor tyrosine kinase by the cytosolic juxtamembrane region. Mol Cell Biol 2003;23:3067¨C3078.
- ~- s4 E( x3 v* T+ Q( G5 s: V% o0 N. h$ H: W
Irusta PM, Luo Y, Bakht O et al. Definition of an inhibitory juxtamembrane WW-like domain in the platelet-derived growth factor beta receptor. J Biol Chem 2002;277:38627¨C38634.
+ s' k. Y/ d L' B# S3 X1 d4 B& Q" F- t7 q( ~$ M% m
Ma Y, Longley BJ, Wang X et al. Clustering of activating mutations in c-KIT¡¯s juxtamembrane coding region in canine mast cell neoplasms. J Invest Dermatol 1999;112:165¨C170.
; W1 w8 G2 k! w* _3 ?" Y! ?0 m8 Q* M$ [9 I$ b& K1 l: m' X; I6 {
Birkenkamp KU, Geugien M, Lemmink HH et al. Regulation of constitutive STAT5 phosphorylation in acute myeloid leukemia blasts. Leukemia 2001;15:1923¨C1931.
; W1 E. T' w0 q3 D9 h' \9 b$ @; y, h& E' ]- v/ L
Fenski R, Flesch K, Serve S et al. Constitutive activation of FLT3 in acute myeloid leukaemia and its consequences for growth of 32D cells. Br J Haematol 2000;108:322¨C330.
& P$ R+ D7 {/ _5 o5 E% @: B: Q4 R: P0 {, {: M
Tse KF, Allebach J, Levis M et al. Inhibition of the transforming activity of FLT3 internal tandem duplication mutants from AML patients by a tyrosine kinase inhibitor. Leukemia 2002;16:2027¨C2036.
' _# ]+ V+ F( N: S6 P7 R& Q9 @( Q" t) \' Z' l' E( s x( N
Jiang J, Paez JG, Lee JC et al. Identifying and characterizing a novel activating mutation of the FLT3 tyrosine kinase in AML. Blood 2004; 104:1855¨C1858.4 v$ o* s! c1 t# v& k. C. q
1 U1 J( R- ~+ `2 MThiede C, Steudel C, Mohr B et al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: Association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002;99:4326¨C4335.
% B6 g8 }+ [: {& [+ C+ Q4 H% e. o1 x0 _' G. r# D+ t0 U
Yamamoto Y, Kiyoi H, Nakano Y et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 2001;97:2434¨C2439.6 o n( C+ B3 I6 D' Y/ u
; M, _+ K( I; E4 H
Furitsu T, Tsujimura T, Tono T et al. Identification of mutations in the coding sequence of the proto-oncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product. J Clin Invest 1993;92:1736¨C1744.1 R( t9 P+ u' }( B' D7 E
8 @* p, H4 X0 @0 V( z9 [( v6 `
Santoro M, Carlomagno F, Romano A et al. Activation of RET as a dominant transforming gene by germline mutations of MEN2A and MEN2B. Science 1995;267:381¨C383.
3 o6 n; v6 {: Q R4 A
! t) S/ S; g0 M: |3 P+ S7 |Till JH, Ablooglu AJ, Frankel M et al. Crystallographic and solution studies of an activation loop mutant of the insulin receptor tyrosine kinase: Insights into kinase mechanism. J Biol Chem 2001;276:10049¨C10055.7 P8 n$ e. b( }
/ H: i! S2 L6 X4 L
Hayakawa F, Towatari M, Kiyoi H et al. Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines. Oncogene 2000;19: 624¨C631.2 z0 M, b' P. o$ [
7 v4 a, ]0 i& ~2 H8 kMizuki M, Fenski R, Halfter H et al. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood 2000;96:3907¨C3914.. P4 o7 w4 @0 { Z/ C
! F5 T/ s5 A9 v7 |: ?: D+ s
Scheijen B, Ngo HT, Kang H et al. FLT3 receptors with internal tandem duplications promote cell viability and proliferation by signaling through Foxo proteins. Oncogene 2004;23:3338¨C3349.5 d4 d: h- H! ]4 a. y# I
+ M$ Y: [, w; m$ ?- V# NBrandts CH, Sargin B, Rode M et al. Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation. Cancer Res 2005;65:9643¨C9650./ x0 \+ `2 |" d# l+ u$ n+ u
: C& {) Z+ v5 d: \+ t
Skorski T, Bellacosa A, Nieborowska-Skorska M et al. Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway. EMBO J 1997;16:6151¨C6161.
! N9 b% v/ c! f+ s* V2 Z: T/ b/ S5 ? W- X e4 H1 y% I! N
Levis M, Allebach J, Tse KF et al. A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo. Blood 2002;99:3885¨C3891. C) I' p8 C$ f' S0 g. B2 L% @4 i( a
' P' a0 l& Y) O; l: vTse KF, Mukherjee G, Small D. Constitutive activation of FLT3 stimulates multiple intracellular signal transducers and results in transformation. Leukemia 2000;14:1766¨C1776.( O b0 D, ?2 h3 k3 r$ x7 W8 K
0 Y3 `) | b# o: r7 E0 o
Kim KT, Baird K, Ahn JY et al. Pim-1 is up-regulated by constitutively activated FLT3 and plays a role in FLT3-mediated cell survival. Blood 2005;105:1759¨C1767.9 U$ w1 S9 R& O9 q3 |$ F4 U2 T
4 |; b1 J+ `/ M. V( L
Kelly LM, Liu Q, Kutok JL et al. FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model. Blood 2002;99:310¨C318.
" s0 t8 N4 ~' ~" M/ n' o3 z6 r4 I' P9 A* u
Zheng R, Friedman AD, Levis M et al. Internal tandem duplication mutation of FLT3 blocks myeloid differentiation through suppression of C/EBPalpha expression. Blood 2004;103:1883¨C1890.* L0 q; ?8 N( i3 f
, j7 C, Z2 M' W
Schwable J, Choudhary C, Thiede C et al. RGS2 is an important target gene of Flt3-ITD mutations in AML and functions in myeloid differentiation and leukemic transformation. Blood 2005;105:2107¨C2114.
# G# g# W2 M3 R
: I- {) S4 c6 t) h+ uChen P, Levis M, Brown P et al. FLT3/ITD mutation signaling includes suppression of SHP-1. J Biol Chem 2005;280:5361¨C5369.
0 J G/ m+ F; {& C
0 C: u5 q' G% f0 k6 zKelly LM, Kutok JL, Williams IR et al. PML/RARalpha and FLT3-ITD induce an APL-like disease in a mouse model. Proc Natl Acad Sci U S A 2002;99:8283¨C8288.
8 Q0 _) k) Q. Z9 F! v
/ p1 ^; J+ O( yLumkul R, Gorin NC, Malehorn MT et al. Human AML cells in NOD/SCID mice: Engraftment potential and gene expression. Leukemia 2002;16:1818¨C1826.
+ H* T( m4 h8 B R& I9 `, a5 w5 F! o+ O5 T5 B4 }. Y7 Z& R3 i
Yanada M, Matsuo K, Suzuki T et al. Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: A meta-analysis. Leukemia 2005;19:1345¨C1349.$ D" j% ?# R) p4 _
" I7 N1 L6 g7 q+ {. KMeshinchi S, Woods WG, Stirewalt DL et al. Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia. Blood 2001;97:89¨C94.
. k# \7 }6 r9 v' z
" ^* g5 a4 n( |1 Q8 l8 i0 n) yXu F, Taki T, Eguchi M et al. Tandem duplication of the FLT3 gene is infrequent in infant acute leukemia. Japan Infant Leukemia Study Group. Leukemia 2000;14:945¨C947.- ?: \( u+ N U1 A$ s p. q
8 n, S/ H4 w$ v5 h
Xu F, Taki T, Yang HW et al. Tandem duplication of the FLT3 gene is found in acute lymphoblastic leukaemia as well as acute myeloid leukaemia but not in myelodysplastic syndrome or juvenile chronic myelogenous leukaemia in children. Br J Haematol 1999;105:155¨C162.
& N( |0 M$ E. u6 y7 s( |3 S! m- W: v5 M. x/ Y
Boissel N, Cayuela JM, Preudhomme C et al. Prognostic significance of FLT3 internal tandem repeat in patients with de novo acute myeloid leukemia treated with reinforced courses of chemotherapy. Leukemia 2002;16:1699¨C1704.! e1 @. l+ Z# V( b6 H3 b
9 D& ^' F/ Y% z; c, DFrohling S, Schlenk RF, Breitruck J et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: A study of the AML Study Group Ulm. Blood 2002;100:4372¨C4380.' d$ r" X% M- x* ?: O( E
( }7 d$ d+ C+ j& W9 ~, @& W0 ZJilani I, Estey E, Manshuri T et al. Better detection of FLT3 internal tandem duplication using peripheral blood plasma DNA. Leukemia 2003;17:114¨C119.
% k/ y% A, d* P; t: A
2 h% o, p6 {% Q+ QRombouts WJ, Blokland I, Lowenberg B et al. Biological characteristics and prognosis of adult acute myeloid leukemia with internal tandem duplications in the Flt3 gene. Leukemia 2000;14:675¨C683.
" z0 n/ p, r! o+ m9 E r
! B% }& A. Z5 g2 p K. ^Strout MP, Marcucci G, Caligiuri MA et al. Core-binding factor (CBF) and MLL-associated primary acute myeloid leukemia: Biology and clinical implications. Ann Hematol 1999;78:251¨C264.
, h% a! x: d) P
7 ~6 |3 K; t% z1 g/ R) {& _Olesen LH, Aggerholm A, Andersen BL et al. Molecular typing of adult acute myeloid leukaemia: Significance of translocations, tandem duplications, methylation, and selective gene expression profiling. Br J Haematol 2005;131:457¨C467.3 {/ m- Q2 L" `* V) r& k, ?% n7 V3 a: C
! D4 _$ `$ [- X: C# W$ y3 N( X2 k T
Taki T, Ida K, Bessho F et al. Frequency and clinical significance of the MLL gene rearrangements in infant acute leukemia. Leukemia 1996; 10:1303¨C1307.
# p0 y$ p* ~; m0 Y$ x2 ?4 e$ k- V! q# q
Marcucci G, Baldus CD, Ruppert AS et al. Overexpression of the ETS-related gene, ERG, predicts a worse outcome in acute myeloid leukemia with normal karyotype: A Cancer and Leukemia Group B study. J Clin Oncol 2005;23:9234¨C9242.
. L9 l4 U$ y1 G) _' ^: X" N, M2 n6 Q4 c
Plass C, Yu F, Yu L et al. Restriction landmark genome scanning for aberrant methylation in primary refractory and relapsed acute myeloid leukemia; involvement of the WIT-1 gene. Oncogene 1999;18:3159¨C3165.8 z, B/ P( G4 j2 X
6 l' R; y N) tReilly JT. Class III receptor tyrosine kinases: Role in leukaemogenesis. Br J Haematol 2002;116:744¨C757.
3 U$ @7 h) R* i4 ` g) D
) G& O8 {9 V4 s3 a0 S! L( sBeran M, Luthra R, Kantarjian H et al. FLT3 mutation and response to intensive chemotherapy in young adult and elderly patients with normal karyotype. Leuk Res 2004;28:547¨C550.; O1 V! O; B. O
' G) Z- h; |6 G9 B" {$ P6 AChillon MC, Fernandez C, Garcia-Sanz R et al. FLT3-activating mutations are associated with poor prognostic features in AML at diagnosis but they are not an independent prognostic factor. Hematol J 2004;5: 239¨C246.0 A2 ~; a! o9 N& h- E
6 u& S: H/ f) C2 [4 \- hSheikhha MH, Awan A, Tobal K et al. Prognostic significance of FLT3 ITD and D835 mutations in AML patients. Hematol J 2003;4:41¨C46.
% q* G4 B" @% g0 M" D6 k8 f/ V8 P Z# [
Libura M, Asnafi V, Tu A et al. FLT3 and MLL intragenic abnormalities in AML reflect a common category of genotoxic stress. Blood 2003;102:2198¨C2204.
) v3 c3 v+ E2 v- L' w/ S& H: _& b# X( [. B4 S7 w$ S, ^# Y
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;344:1031¨C1037.5 A4 J1 S F! \% [# `
7 ^2 g+ V- {) g- [2 N8 |2 d
Levis M, Small D. Small molecule FLT3 tyrosine kinase inhibitors. Curr Pharm Des 2004;10:1183¨C1193.
/ x- o p* j8 x: g v8 Q" E3 B" E
7 Y5 H) K! m% {' pLevis M, Tse KF, Smith BD et al. A FLT3 tyrosine kinase inhibitor is selectively cytotoxic to acute myeloid leukemia blasts harboring FLT3 internal tandem duplication mutations. Blood 2001;98:885¨C887.
$ ~7 F- j; {4 O# h: f& `# R' H2 E% b! o( s4 Q
Tse KF, Novelli E, Civin CI et al. Inhibition of FLT3-mediated transformation by use of a tyrosine kinase inhibitor. Leukemia 2001;15: 1001¨C1010. I* Y2 r: x0 [7 F- V0 h% u
# c; D _" Z8 \% s+ ~# X) N
Brown P, Small D. FLT3 inhibitors: A paradigm for the development of targeted therapeutics for paediatric cancer. Eur J Cancer 2004;40:707¨C721, discussion 722¨C704./ b$ m: J g/ c2 g# F/ q' x
# F \5 z5 F8 i. D9 E* l" L! sFong TA, Shawver LK, Sun L et al. SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types. Cancer Res 1999;59:99¨C106.. t% G8 \5 o/ p \: \4 Z
y- S$ L1 ?* ]2 }# L; i
Mendel DB, Schreck RE, West DC et al. The angiogenesis inhibitor SU5416 has long-lasting effects on vascular endothelial growth factor receptor phosphorylation and function. Clin Cancer Res 2000;6:4848¨C4858.1 b- G1 O% ?& g! T/ @
# l! j5 P" A5 A) d( ], @2 u6 s
Smolich BD, Yuen HA, West KA et al. The antiangiogenic protein kinase inhibitors SU5416 and SU6668 inhibit the SCF receptor (c-kit) in a human myeloid leukemia cell line and in acute myeloid leukemia blasts. Blood 2001;97:1413¨C1421.
# Y+ ~1 o& V% f. g2 Y% V7 u6 o4 `3 f' q3 N1 g) M# k
Sukbuntherng J, Cropp G, Hannah A et al. Pharmacokinetics and interspecies scaling of a novel VEGF receptor inhibitor, SU5416. J Pharm Pharmacol 2001;53:1629¨C1636.
4 S! F2 i, A* l. {6 ]
2 _1 }) U1 T# U, ^' zYee KW, O¡¯Farrell AM, Smolich BD et al. SU5416 and SU5614 inhibit kinase activity of wild-type and mutant FLT3 receptor tyrosine kinase. Blood 2002;100:2941¨C2949.
0 {9 t& _0 ]- c$ Q, m. e+ O
7 U P6 M/ X7 O. Q \Spiekermann K, Dirschinger RJ, Schwab R et al. The protein tyrosine kinase inhibitor SU5614 inhibits FLT3 and induces growth arrest and apoptosis in AML-derived cell lines expressing a constitutively activated FLT3. Blood 2003;101:1494¨C1504.
: `! @. }' x5 |4 o, {' J
" A) f. [7 s9 V" n0 yGiles FJ, Stopeck AT, Silverman LR et al. SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients with refractory acute myeloid leukemia or myelodysplastic syndromes. Blood 2003;102:795¨C801.
& }$ o0 o) {- N8 p: o) y8 ]3 W& ]8 ?$ R s; z, O3 t8 }
Fiedler W, Mesters R, Tinnefeld H et al. A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia. Blood 2003;102:2763¨C2767.6 i8 W6 L5 X) b1 Z) K
" q7 y7 b: t' w4 \5 oGiles FJ, Cooper MA, Silverman L et al. Phase II study of SU5416¡ªa small-molecule, vascular endothelial growth factor tyrosine-kinase receptor inhibitor¡ªin patients with refractory myeloproliferative diseases. Cancer 2003;97:1920¨C1928.$ X% v5 z8 ^5 z
- Q# X0 E# T j9 ~Levis M. Recent advances in the development of small-molecule inhibitors for the treatment of acute myeloid leukemia. Curr Opin Hematol 2005;12:55¨C61.
! V9 b' z8 A$ E4 f0 E" g2 T: A- i+ c! [0 C( b5 _( p, T. o
Weisberg E, Boulton C, Kelly LM et al. Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell 2002;1:433¨C443.
9 d9 D. a* P/ y+ q! {7 h
, t7 i, W; O E2 C& IStone 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;105:54¨C60. V" A0 s3 G$ \5 [) Y8 j$ k0 I6 H
4 Z( i; k' X% s0 r( W& E
Mendel DB, Laird AD, Xin X et al. In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: Determination of a pharmacokinetic/pharmacodynamic relationship. Clin Cancer Res 2003;9:327¨C337.% P" v% @+ v! o& Y$ A
( y# F& J/ X5 u( E9 @1 yO¡¯Farrell AM, Abrams TJ, Yuen HA et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003;101:3597¨C3605." K# I9 ]. H8 h' l4 q
: ?, A% J. }$ W7 d3 W# |8 PO¡¯Farrell AM, Foran JM, Fiedler W et al. An innovative phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients. Clin Cancer Res 2003;9:5465¨C5476.
5 t N5 X4 d" S0 Y/ U& R- [3 ]+ e3 i, i S
Kelly LM, Yu JC, Boulton CL et al. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML). Cancer Cell 2002;1:421¨C432.2 j5 @$ j! m$ g6 }( @3 D, [5 c
. S( c, G& M5 y6 L
Corbin AS, Griswold IJ, La Rosee P et al. Sensitivity of oncogenic KIT mutants to the kinase inhibitors MLN518 and PD180970. Blood 2004; 104:3754¨C3757.
) G, K: V/ W: X) s
( H4 s) ^; o4 I- P% uPandey A, Volkots DL, Seroogy JM et al. Identification of orally active, potent, and selective 4-piperazinylquinazolines as antagonists of the platelet-derived growth factor receptor tyrosine kinase family. J Med Chem 2002;45:3772¨C3793.: n3 ?: X; C# Y$ e5 ^. s: l
; g2 l; N8 j& ~9 ^' u: \ AYu JC, Lokker NA, Hollenbach S et al. Efficacy of the novel selective platelet-derived growth factor receptor antagonist CT52923 on cellular proliferation, migration, and suppression of neointima following vascular injury. J Pharmacol Exp Ther 2001;298:1172¨C1178." D' q5 y, z% e, N
5 L! H/ x! A% G6 S! QHeinrich MC, Griffith DJ, Druker BJ et al. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor. Blood 2000;96:925¨C932.
& |! n3 |( R, ~1 j( T4 g4 S' u" Z
George DJ, Dionne CA, Jani J et al. Sustained in vivo regression of Dunning H rat prostate cancers treated with combinations of androgen ablation and Trk tyrosine kinase inhibitors, CEP-751 (KT-6587) or CEP-701 (KT-5555). Cancer Res 1999;59:2395¨C2401.; y3 u4 `4 H7 I1 h
7 V' a8 m) T3 Q6 h* IZheng R, Friedman AD, Small D. Targeted inhibition of FLT3 overcomes the block to myeloid differentiation in 32Dcl3 cells caused by expression of FLT3/ITD mutations. Blood 2002;100:4154¨C4161.
0 K9 a5 Y% }- B( Q& z: Q" C1 K' s, S4 b# u; {7 k; j0 r
Smith BD, Levis M, Beran M et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 2004;103:3669¨C3676.6 F7 |& Y/ U5 z" T) r7 a W% m: e
2 v) d v- x: B% b, | f+ U0 KKomeno Y, Kurokawa M, Imai Y et al. Identification of Ki23819, a highly potent inhibitor of kinase activity of mutant FLT3 receptor tyrosine kinase. Leukemia 2005;19:930¨C935.
* \5 j) B4 N$ N' `0 {7 o+ ~- y0 ]. }& n+ l' i& i& p
Komarova NL, Wodarz D. Drug resistance in cancer: Principles of emergence and prevention. Proc Natl Acad Sci U S A 2005;102:9714¨C9719.! l8 D: w, o! A; w$ a9 ~4 p9 t
7 R O2 y* h2 G# r0 [; }% sGorre ME, Mohammed M, Ellwood K et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 2001;293:876¨C880.1 P9 k B' Q4 x B; t- ~5 s3 x
/ J- X7 X5 Y9 V x
Hochhaus A, Kreil S, Corbin AS et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia 2002; 16:2190¨C2196.
8 l& L$ B* N; c1 | U$ w4 H# o
) L. ]7 L6 z2 D- a; h& u, H. U; ZSchindler T, Bornmann W, Pellicena P et al. Structural mechanism for STI-571 inhibition of abelson tyrosine kinase. Science 2000;289:1938¨C1942.
- p) r. G$ h) p( s) w) j/ |3 _. S0 ` d# C& H9 n" O
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;344:1038¨C1042.
1 e& m) C( J8 x- M5 k$ b# p
9 T6 t) n" G. I" l1 C+ ?' `/ \' XClark JJ, Cools J, Curley DP et al. Variable sensitivity of FLT3 activation loop mutations to the small molecule tyrosine kinase inhibitor MLN518. Blood 2004;104:2867¨C2872.
* O g. G/ D( ]$ c( r
3 A7 O5 C7 K8 i) Q5 [3 RGrundler R, Thiede C, Miething C et al. Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor. Blood 2003;102:646¨C651.
% V7 e! r) I& c* }5 M% D) ~
% l. U; N4 S, a9 v oBagrintseva K, Schwab R, Kohl TM et al. Mutations in the tyrosine kinase domain of FLT3 define a new molecular mechanism of acquired drug resistance to PTK inhibitors in FLT3-ITD-transformed hematopoietic cells. Blood 2004;103:2266¨C2275.& M7 R: ^( I, W5 M
" t* ]/ I+ j. ~8 q
Cools J, Mentens N, Furet P et al. Prediction of resistance to small molecule FLT3 inhibitors: Implications for molecularly targeted therapy of acute leukemia. Cancer Res 2004;64:6385¨C6389.: n+ v0 U+ T5 Q. B& t/ m
6 `( Z, E/ Q5 E* p
Sohal J, Phan VT, Chan PV et al. A model of APL with FLT3 mutation is responsive to retinoic acid and a receptor tyrosine kinase inhibitor, SU11657. Blood 2003;101:3188¨C3197.
/ K- i% B8 U0 @" a9 V2 k' V$ H3 m7 t
Mohi MG, Boulton C, Gu TL et al. Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci U S A 2004;101:3130¨C3135.
; E& a; `+ S7 x7 l" {# p
) s% k- z* w$ \2 QLevis M, Pham R, Smith BD, Small D. In vitro studies of a FLT3 inhibitor combined with chemotherapy: Sequence of administration is important to achieve synergistic cytotoxic effects. Blood 2004;104: 1145¨C1150. |
|