FTY720治疗伴C-KIT激酶区突变的急性髓系白血病的机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分C-KIT/D816V突变在急性髓系性白血病中的发生率调查
     目的:分析C-KIT/D816V突变在中国人AML患者中的发生率。
     方法:收集2008年至2011年送至干细胞中心行C-KIT/D816V检测的AML外周血
     标本286例,提取者外周血单个核细胞,采用Real-time Quantitative PCR SYBR green染料法检测C-KIT D816V突变。
     结果:286例AML患者中,有8例(2.8%,其中6例男性,2例女性)患者发生了C-KIT/D816V突变,其在男性患者和女性患者中的发生率分别为3.9%和1.5%,在男性患者中的发生率高于女性,差异有统计学意义(P<0.05)。8例C-KIT突变患者中有5例M2,1例M4,另外2例患者FAB亚型诊断未明。在AML各亚型中的发生率分别为:M26.8%,M44.3%。
     结论:C-KIT/D816V突变最常发生在M2中,其次为M4。在男性患者中的发生率高于女性。本研究从流行病学的角度为C-KIT/D816V突变在中国人AML及其亚型中的发生率提供了重要依据。
     第二部分关于携带C-KIT/D816V突变及C-KIT/WT的急性髓系白血病PP2A蛋白活性变化的研究
     目的:从流行病学的角度探讨携带C-KIT/D816V[位于C-KIT激酶区(C-KIT kinase domain, C-KIT/TKD)]突变的急性髓细胞性白血病患者及携带野生型C-KIT (C-KIT/WT)的急性髓细胞性白血病患者单个核细胞中蛋白磷酸酯酶2A(protein phosphotase 2A, PP2A)的活性变化,并对携带C-KIT/N822K(位于C-KIT/TKD)突变的AML细胞株Kasumi-1及携带野生型C-KIT的AML细胞株HL60中PP2A活性进行了比较。
     方法:选取8例C-KIT/D816V突变的AML患者,12例C-KIT/WT AML患者及20例健康对照,提取单个核细胞,使用PP2A免疫沉淀磷酸酶检测试剂盒检测PP2A活性。体外培养HL60及Kasumi-1细胞株,使用PP2A免疫沉淀磷酸酶检测试剂盒检测PP2A活性。使用SPSS16.0对数据进行分析。
     结果:C-KIT/WT AML组患者单个核细胞PP2A的活性(565.69±122.56pmol)与正常人(706.02±154.22pmol)比较有所降低,而C-KIT/D816V AML组(419.01±96.97pmol)活性最低,各组之间差异有统计学意义(P<0.05)。Kasumi-1细胞PP2A活性(886.420±152.187 pmol)与HL60细胞(1196.442±219.483pmol)比较有所降低,差异有统计学意义(P<0.05)。
     结论:AML患者PP2A活性有所降低,且携带C-KIT/TKD的AML患者及细胞株中PP2A活性与携带C-KIT/WT的AML患者或细胞株比较有所降低,PP2A蛋白活性与AML的发生、发展密切相关,对AML的诊断及治疗可能有参考价值。
     第三部分FTY720对携带C-KIT激酶区突变的AML体外抑制作用的机制研究
     目的:本研究探讨FTY720对携带C-KIT/TKD突变的AML细胞系Kasumi-1及未携带C-KIT/TKD突变的AML细胞系HL-60细胞增殖、凋亡的影响,并对FTY720对两种细胞作用的进行了比较了,探讨其可能机制。
     方法:体外培养AML细胞株HL60及Kasumi-1,使用FTY720(2.5,5,10和20μM)处理HL-60、Kasumi-1细胞24小时,应用CCK8试剂盒检测细胞的增殖状况。使用FTY720(10μM)处理细胞36h,应用流式细胞术检测细胞的凋亡率。使用FTY720(5μM,10μM)处理细胞5h,应用PP2A免疫沉淀磷酸酶活性检测试剂盒测定细胞PP2A活性。用FTY720(5μM)处理细胞5h,应用Western Blot检测PP2A的表达水平。
     结果:FTY720能有效抑制HL60和Kasumi-1细胞的增殖,随药物作用浓度的增加,抑制作用越强,作用24h后FTY720对HL60和Kasumi-1的IC50分别为13.5μM和20μM,P<0.05,两者差异有统计学意义。FTY720作用于HL60和Kasumi-1后,两种细胞的凋亡率均增加,且Kasumi-1的凋亡率增加更显著。FTY720能增强HL60和Kasumi-1细胞的PP2A活性,但不改变PP2A的表达水平。PP2A抑制剂OA能抑制FTY720所致的PP2A活性增强,同时也能减轻FTY720所致的细胞毒性作用。FTY720不改变PP2A的表达水平。
     结论:FTY720能通过活化PP2A抑制HL60和Kasumi-1细胞的增殖并诱导两种细胞发生凋亡,且对后者的作用强于前者。FTY720对AML,尤其是难治性AML可能有治疗价值。
PART I Detection of C-KIT/D816V mutation in Acute Myeloid Leukemia with Different Subtypes in China
     Objective To investigate and analyze the incidence of C-KIT/D816V mutation in AML patients in China.
     Methods Blood samples from 286 AML patients with various French-American-British (FAB) classifications were collected from Center for Stem Cell Research and Application between 2008 and 2011. The diagnosis of AML and the assignment of FAB classification were based on morphology and immunophenotype. Then the quantitative realtime-PCR was used to examine the C-KIT/D816V mutation.
     Results Of the 286 AML samples studied,8 were shown to have a point mutation at D816V, including 6 male cases and 2 female cases. Its incidence in male and female was 3.9% and 1.5% respectively, thus it was more inclined to occur in male than female (P< 0.05). This mutation was typically located within the M2 (5 cases) and M4 (1 cases) subclasses, with the other 2 mutation AML cases not definitely diagnosed. In the present work, the D816V mutation occurred in 6.8% of M2,4.3% of M4 subclasses, and the overall incidence of C-KIT/D816V was 2.8% in AML.
     Conclution C-KIT/D816V most frequently occurs in M2 subutype, then M4 in AML, and it is more inclined to occur in male than female. Our research provided significant evidence for the incidence of C-KIT/D816V mutation in AML and its subtypes in China.
     partⅡAnalysis of Changes of the Activity of PP2A in Acute Myeloid Leukemia
     Objective To analyze the changes of PP2A activity in AML patients with C-KIT/WT and C-KIT/D816V and compare the PP2A activity in HL60 cell harboring C-KIT/WT and kasumi-1 cell harboring C-KIT/TKD.
     Methods Eight AML patients harboring C-KIT/D816V mutation, twelve AML patients harboring wide-type C-KIT (C-KIT/WT) and twenty healthy controls were recruited and informed consent at Union hospital of Wuhan in this study. Real-time quantitative PCR was used to identify C-KIT/WT and C-KIT/D816V. Peripheral blood mononuclear cells (PBMCs) of patients with AML and normal controls were collected. In addition, HL60 and kasumi-1 cells were cultivated in vitro. The activity of PP2A in the PBMCs of patients and the cell lines was assayed with a PP2A Immunoprecipitation Phosphatase Assay Kit, the data was analyzed by SPSS 16.0.
     Results We detected PP2A activity of all samples with PP2A Immunoprecipitation Phosphatase Assay Kit. The results indicated that the PP2A activity in AML patients harboring C-KIT/WT(565.69±122.56 pmol) was obviously lower than the normal controls(706.02±154.22 pmol), and the PP2A activity in AML patients harboring C-KIT/D816V(419.01±96.97 pmol) was the lowest. There was significant difference between the three groups (P<0.05). In addition, PP2A activity in Kasumi-1 cell(886.420±152.187 pmol) was obviously lower than HL60 cell(1196.442±219.483 pmol). There was significant difference between them (P<0.05).
     Conclution It is concluded that from the standpoint of prevalence, PP2A activity is decreased in patients with C-KIT/TKD AML. In addition, compared with AML cells lines harboring C-KIT/WT, PP2A activity of AML cells harboring C-KIT/TKD is obviously lower. PP2A protein plays a key role in the genesis and development of AML, and may imply a diagnosis and treatment of AML, especially the refractory C-KIT/TKD AML.
     PARTⅢCytotoxic Effect of FTY720 on AML Cell Lines harboring C-KIT/TKD and C-KIT/WT and Its Mechanism
     Objective To observe the cytotoxicity of FTY720 in C-KIT/TKD AML and develop FTY720 as a novel therapy for C-KIT/TKD AML
     Methods Cell lines were treated with FTY720 alone or combined with specific PP2A inhibitor okadaic acid (OA) in culture. Cell growth was assessed using the CCK8 Kit. Cell apoptosis was analyzed by flow cytometry after dual staining with annexinⅤ/ propidium iodide. PP2A activity was assayed with a PP2A Immunoprecipitation Phosphatase Assay Kit. Western blotting was used to analyze the protein changes. All experiments were repeated three times. Statistical analysis was performed using SPSS 16.0.
     Results FTY720 induced toxicity in all AML cells, including Kasumi-1 and HL60 in a time-and dose-dependent manner. PP2A inhibitor OA rescued these cells from FTY720-induced apoptosis. Furthermore, Increased PP2A activity was detected after FTY720 treatment. When cells were pretreated with OA, PP2A activity was partially rescued. Finally, PP2A expression remained unchanged after FTY720 treatment. Our results strongly suggested that FTY720-induced cytotoxicity was mediated by PP2A activation without altering its expression. Interestingly, it was observed that cells harboring C-KIT/TKD mutation were more sensitive to this agent
     Conclusions Our study provides evidence for the PP2A-dependent toxicity of FTY720 in AML cells, and these effects of FTY720 appear to be most marked in C-KIT/TKD AML where conventional chemotherapeutic approaches are most likely to fail. The significant preclinical efficacy of FTY720 indicates that it may be valuable therapeutic agents for refractory C-KIT/TKD AML.
引文
[1]De La Rubia J, Regadera A, Martin G, et al. FLAG-IDA regimen (fludarabine, cytarabine, idarubicin and G-CSF) in the treatment of patients with high-risk myeloid malignancies.[J]. Leuk Res.2002,26(8):725-730.
    [2]Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML).[J]. Leukemia.2006,20(6):965-970.
    [3]Muller-Tidow C, Steffen B, Cauvet T, et al. Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells.[J]. Mol Cell Biol. 2004,24(7):2890-2904.
    [4]Renneville A, Roumier C, Biggio V, et al. Cooperating gene mutations in acute myeloid leukemia:a review of the literature. [J]. Leukemia.2008,22(5):915-931.
    [5]Shih L Y, Liang D C, Huang C F, et al. Cooperating mutations of receptor tyrosine kinases and Ras genes in childhood core-binding factor acute myeloid leukemia and a comparative analysis on paired diagnosis and relapse samples.[J]. Leukemia.2008, 22(2):303-307.
    [6]Wang Y Y, Zhou G B, Yin T, et al. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia:implication in stepwise leukemogenesis and response to Gleevec.[J]. Proc Natl Acad Sci U S A.2005,102(4):1104-1109.
    [7]Corbacioglu S, Kilic M, Westhoff M A, et al. Newly identified c-KIT receptor tyrosine kinase ITD in childhood AML induces ligand-independent growth and is responsive to a synergistic effect of imatinib and rapamycin.[J]. Blood.2006,108(10):3504-3513.
    [8]Pedersen M, Ronnstrand L, Sun J. The c-K.it/D816V mutation eliminates the differences in signal transduction and biological responses between two isoforms of c-Kit.[J]. Cell Signal.2009,21(3):413-418.
    [9]Roberts K G, Smith A M, Mcdougall F, et al. Essential requirement for PP2A inhibition by the oncogenic receptor c-KIT suggests PP2A reactivation as a strategy to treat c-KIT+ cancers.[J]. Cancer Res.2010,70(13):5438-5447.
    [10]Sritana N, Auewarakul C U. KIT and FLT3 receptor tyrosine kinase mutations in acute myeloid leukemia with favorable cytogenetics:two novel mutations and selective occurrence in leukemia subtypes and age groups.[J]. Exp Mol Pathol.2008,85(3): 227-231.
    [11]Xiang Z, Kreisel F, Cain J, et al. Neoplasia driven by mutant c-KIT is mediated by intracellular, not plasma membrane, receptor signaling.[J]. Mol Cell Biol.2007,27(1): 267-282.
    [12]Advani A S, Rodriguez C, Jin T, et al. Increased C-kit intensity is a poor prognostic factor for progression-free and overall survival in patients with newly diagnosed AML.[J]. Leuk Res.2008,32(6):913-918.
    [13]Li W Y, Sun A N, Wu D P. [Analysis of c-kit and JAK2 gene mutations in t(8;21) acute myeloid leukemia].[J]. Zhonghua Xue Ye Xue Za Zhi.2008,29(12):797-801.
    [14]Luck S C, Russ A C, Du J, et al. KIT mutations confer a distinct gene expression signature in core binding factor leukaemia.[J]. Br J Haematol.2010,148(6):925-937.
    [15]Malaise M, Steinbach D, Corbacioglu S. Clinical implications of c-Kit mutations in acute myelogenous leukemia.[J]. Curr Hematol Malig Rep.2009,4(2):77-82.
    [16]Potenza L, Luppi M, Riva G, et al. May the correlation between Kit-D816 mutation and AML1-ETO level change the use of prognostic factors in t(8;21) AML?[J]. Leuk Res.2007,31(2):269-271.
    [17]Roberts K G, Odell A F, Byrnes E M, et al. Resistance to c-KIT kinase inhibitors conferred by V654A mutation.[J]. Mol Cancer Ther.2007,6(3):1159-1166.
    [18]Schnittger S, Kohl T M, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival.[J]. Blood.2006,107(5):1791-1799.
    [19]Sun J, Pedersen M, Ronnstrand L. The D816V mutation of c-Kit circumvents a requirement for Src family kinases in c-Kit signal transduction.[J]. J Biol Chem.2009, 284(17):11039-11047.
    [20]Ustun C, Corless C L, Savage N, et al. Chemotherapy and dasatinib induce long-term hematologic and molecular remission in systemic mastocytosis with acute myeloid leukemia with KIT D816V.[J]. Leuk Res.2009,33(5):735-741.
    [21]Eichhorn P J, Creyghton M P, Bernards R. Protein phosphatase 2A regulatory subunits and cancer.[J]. Biochim Biophys Acta.2009,1795(1):1-15.
    [22]Epie N, Ammosova T, Turner W, et al. Inhibition of PP2A by LIS1 increases HIV-1 gene expression.[J]. Retrovirology.2006,3:65.
    [23]Guenin S, Schwartz L, Morvan D, et al. PP2A activity is controlled by methylation and regulates oncoprotein expression in melanoma cells:a mechanism which participates in growth inhibition induced by chloroethylnitrosourea treatment.[J]. Int J Oncol.2008, 32(1):49-57.
    [24]Kolupaeva V, Laplantine E, Basilico C. PP2A-mediated dephosphorylation of p107 plays a critical role in chondrocyte cell cycle arrest by FGF.[J]. PLoS One.2008,3(10): e3447.
    [25]Lu J, Kovach J S, Johnson F, et al. Inhibition of serine/threonine phosphatase PP2A enhances cancer chemotherapy by blocking DNA damage induced defense mechanisms. [J]. Proc Natl Acad Sci U S A.2009,106(28):11697-11702.
    [26]Silverstein A M, Barrow C A, Davis A J, et al. Actions of PP2A on the MAP kinase pathway and apoptosis are mediated by distinct regulatory subunits.[J]. Proc Natl Acad Sci U S A.2002,99(7):4221-4226.
    [27]Chatfield K, Eastman A. Inhibitors of protein phosphatases 1 and 2A differentially prevent intrinsic and extrinsic apoptosis pathways.[J]. Biochem Biophys Res Commun. 2004,323(4):1313-1320.
    [28]Gallay N, Dos S C, Cuzin L, et al. The level of AKT phosphorylation on threonine 308 but not on serine 473 is associated with high-risk cytogenetics and predicts poor overall survival in acute myeloid leukaemia.[J]. Leukemia.2009,23(6):1029-1038.
    [29]Neviani P, Santhanam R, Trotta R, et al. The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein.[J]. Cancer Cell.2005,8(5):355-368.
    [30]Chevallier P, Hunault-Berger M, Larosa F, et al. A phase II trial of high-dose imatinib mesylate for relapsed or refractory c-kit positive and Bcr-Abl negative acute myeloid leukaemia:the AFR-15 trial.[J]. Leuk Res.2009,33(8):1124-1126.
    [31]Corless C L, Harrell P, Lacouture M, et al. Allele-specific polymerase chain reaction for the imatinib-resistant KIT D816V and D816F mutations in mastocytosis and acute myelogenous leukemia.[J]. J Mol Diagn.2006,8(5):604-612.
    [32]Kosmider O, Denis N, Dubreuil P, et al. Semaxinib (SU5416) as a therapeutic agent targeting oncogenic Kit mutants resistant to imatinib mesylate.[J]. Oncogene.2007, 26(26):3904-3908.
    [33]Piccaluga P P, Malagola M, Rondoni M, et al. Imatinib mesylate in the treatment of ewly diagnosed or refractory/resistant c-KIT positive acute myeloid leukemia. Results of an Italian Multicentric Phase Ⅱ Study.[J]. Haematologica.2007,92(12):1721-1722.
    [34]Verstovsek S, Tefferi A, Cortes J, et al. Phase Ⅱ study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis.[J]. Clin Cancer Res.2008,14(12):3906-3915.
    [1]Beghini A, Peterlongo P, Ripamonti C B, et al. C-kit mutations in core binding factor 1 eukemias.[J]. Blood.2000,95(2):726-727.
    [2]Renneville A, Roumier C, Biggio V, et al. Cooperating gene mutations in acute myeloid leukemia:a review of the literature. [J]. Leukemia.2008,22(5):915-931.
    [3]Longley B J, Reguera M J, Ma Y. Classes of c-KIT activating mutations:proposed mechanisms of action and implications for disease classification and therapy.[J]. Leuk Res.2001,25(7):571-576.
    [4]Stirewalt D L, Radich J P. The role of FLT3 in haematopoietic malignancies.[J]. Nat Rev Cancer.2003,3(9):650-665.
    [5]Liu H, Chen X, Focia P J, et al. Structural basis for stem cell factor-KIT signaling and activation of class III receptor tyrosine kinases.[J]. EMBO J.2007,26(3):891-901.
    [6]Kahler C, Didlaukat S, Feller A C, et al. Sensitive and reliable detection of Kit point mutation Asp 816 to Val in pathological material.[J]. Diagn Pathol.2007,2:37.
    [7]Kelly L M, Yu J C, Boulton C L, et al. CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML).[J]. Cancer Cell.2002,1(5): 421-432.
    [8]Frost M J, Ferrao P T, Hughes T P, et al. Juxtamembrane mutant V560GKit is more sensitive to Imatinib (STI571) compared with wild-type c-kit whereas the kinase domain mutant D816VKit is resistant.[J]. Mol Cancer Ther. 2002,1(12):1115-1124.
    [9]Fabbro D, Ruetz S, Buchdunger E, et al. Protein kinases as targets for anticancer agents: from inhibitors to useful drugs.[J]. Pharmacol Ther.2002,93(2-3):79-98.
    [10]Rowe J M. Optimal induction and post-remission therapy for AML in first remission.[J]. Hematology Am Soc Hematol Educ Program.2009:396-405.
    [11]Rombouts W J, Blokland I, Lowenberg B, et al. Biological characteristics and prognosis of adult acute myeloid leukemia with internal tandem duplications in the Flt3 gene.[J]. Leukemia.2000,14(4):675-683.
    [12]Williams D E. In vivo effects of flt3 ligand.[J]. Blood.1997,90(12):5022.
    [13]Care R S, Valk P J, Goodeve A C, et al. Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias.[J]. Br J Haematol. 2003,121(5):775-777.
    [14]Wang Y Y, Zhou G B, Yin T, et al. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia:implication in stepwise leukemogenesis and response to Gleevec.[J]. Proc Natl Acad Sci U S A.2005,102(4):1104-1109.
    [15]Zaker F, Mohammadzadeh M, Mohammadi M. Detection of KIT and FLT3 mutations in acute myeloid leukemia with different subtypes.[J]. Arch Iran Med.2010,13(1): 21-25.
    [16]Jr Roskoski R. Structure and regulation of Kit protein-tyrosine kinase--the stem cell factor receptor.[J]. Biochem Biophys Res Commun.2005,338(3):1307-1315.
    [1]Mumby M. PP2A:unveiling a reluctant tumor suppressor.[J]. Cell.2007,130(1): 21-24.
    [2]Millward T A, Zolnierowicz S, Hemmings B A. Regulation of protein kinase cascades by protein phosphatase 2A.[J]. Trends Biochem Sci.1999,24(5):186-191.
    [3]Westermarck J, Hahn W C. Multiple pathways regulated by the tumor suppressor PP2A in transformation.[J]. Trends Mol Med.2008,14(4):152-160.
    [4]Neviani P, Santhanam R, Trotta R, et al. The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein.[J]. Cancer Cell.2005,8(5):355-368.
    [5]Yarden Y, Kuang W J, Yang-Feng T, et al. Human proto-oncogene c-kit:a new cell surface receptor tyrosine kinase for an unidentified ligand.[J]. EMBO J.1987,6(11): 3341-3351.
    [6]Shih L Y, Liang D C, Huang C F, et al. Cooperating mutations of receptor tyrosine kinases and Ras genes in childhood core-binding factor acute myeloid leukemia and a comparative analysis on paired diagnosis and relapse samples.[J]. Leukemia.2008, 22(2):303-307.
    [7]Sotlar K, Fridrich C, Mall A, et al. Detection of c-kit point mutation Asp-816--> Val in microdissected pooled single mast cells and leukemic cells in a patient with systemic mastocytosis and concomitant chronic myelomonocytic leukemia.[J]. Leuk Res.2002, 26(11):979-984.
    [8]Roberts K G, Smith A M, Mcdougall F, et al. Essential requirement for PP2A inhibition by the oncogenic receptor c-KIT suggests PP2A reactivation as a strategy to treat c-KIT+ cancers.[J]. Cancer Res.2010,70(13):5438-5447.
    [9]De La Rubia J, Regadera A, Martin G, et al. FLAG-IDA regimen (fludarabine, cytarabine, idarubicin and G-CSF) in the treatment of patients with high-risk myeloid malignancies.[J]. Leuk Res.2002,26(8):725-730.
    [10]Wang Y Y, Zhou G B, Yin T, et al. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia:implication in stepwise leukemogenesis and response to Gleevec.[J]. Proc Natl Acad Sci U S A.2005,102(4):1104-1109.
    [11]Schnittger S, Kohl T M, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival.[J]. Blood.2006,107(5):1791-1799.
    [12]Sun J, Pedersen M, Ronnstrand L. The D816V mutation of c-Kit circumvents a requirement for Src family kinases in c-Kit signal transduction.[J]. J Biol Chem.2009, 284(17):11039-11047.
    [13]Neviani P, Santhanam R, Oaks J J, et al. FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia.[J]. J Clin Invest.2007,117(9):2408-2421.
    [14]Adachi Y, Pavlakis G N, Copeland T D. Identification and characterization of SET, a nuclear phosphoprotein encoded by the translocation break point in acute undifferentiated leukemia.[J]. J Biol Chem.1994,269(3):2258-2262.
    [15]Adler H T, Nallaseth F S, Walter G, et al. HRX leukemic fusion proteins form a heterocomplex with the leukemia-associated protein SET and protein phosphatase 2A.[J]. J Biol Chem.1997,272(45):28407-28414.
    [16]Gallay N, Dos S C, Cuzin L, et al. The level of AKT phosphorylation on threonine 308 but not on serine 473 is associated with high-risk cytogenetics and predicts poor overall survival in acute myeloid leukaemia.[J]. Leukemia.2009,23(6):1029-1038.
    [17]Rodriguez-Viciana P, Collins C, Fried M. Polyoma and SV40 proteins differentially regulate PP2A to activate distinct cellular signaling pathways involved in growth control.[J]. Proc Natl Acad Sci U S A.2006,103(51):19290-19295.
    [18]Andrabi S, Gjoerup O V, Kean J A, et al. Protein phosphatase 2A regulates life and death decisions via Akt in a context-dependent manner. [J]. Proc Natl Acad Sci U S A. 2007,104(48):19011-19016.
    [19]Liu Q, Zhao X, Frissora F, et al. FTY720 demonstrates promising preclinical activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma.[J]. Blood. 2008,111(1):275-284.
    [1]Renneville A, Roumier C, Biggio V, et al. Cooperating gene mutations in acute myeloid leukemia:a review of the literature.[J]. Leukemia.2008,22(5):915-931.
    [2]Shih L Y, Liang D C, Huang C F, et al. Cooperating mutations of receptor tyrosine kinases and Ras genes in childhood core-binding factor acute myeloid leukemia and a comparative analysis on paired diagnosis and relapse samples.[J]. Leukemia.2008, 22(2):303-307.
    [3]Roberts K G, Smith A M, Mcdougall F, et al. Essential requirement for PP2A inhibition by the oncogenic receptor c-KIT suggests PP2A reactivation as a strategy to treat c-KIT+ cancers.[J]. Cancer Res.2010,70(13):5438-5447.
    [4]Wang Y Y, Zhou G B, Yin T, et al. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia:implication in stepwise leukemogenesis and response to Gleevec.[J]. Proc Natl Acad Sci U S A.2005,102(4):1104-1109.
    [5]Potenza L, Luppi M, Riva G, et al. May the correlation between Kit-D816 mutation and AML1-ETO level change the use of prognostic factors in t(8;21) AML?[J]. Leuk Res.2007,31(2):269-271.
    [6]Sun J, Pedersen M, Ronnstrand L. The D816V mutation of c-Kit circumvents a requirement for Src family kinases in c-Kit signal transduction.[J]. J Biol Chem.2009, 284(17):11039-11047.
    [7]Mrozek K, Marcucci G, Paschka P, et al. Advances in molecular genetics and treatment of core-binding factor acute myeloid leukemia.[J]. Curr Opin Oncol.2008,20(6): 711-718.
    [8]Epie N, Ammosova T, Turner W, et al. Inhibition of PP2A by LIS1 increases HIV-1 gene expression. [J]. Retro virology.2006,3:65.
    [9]Guenin S, Schwartz L, Morvan D, et al. PP2A activity is controlled by methylation and regulates oncoprotein expression in melanoma cells:a mechanism which participates in growth inhibition induced by chloroethylnitrosourea treatment.[J]. Int J Oncol.2008, 32(1):49-57.
    [10]Bengtsson L, Schwappacher R, Roth M, et al. PP2A regulates BMP signalling by interacting with BMP receptor complexes and by dephosphorylating both the C-terminus and the linker region of Smadl.[J]. J Cell Sci.2009,122(Pt 8):1248-1257.
    [11]Chatfield K, Eastman A. Inhibitors of protein phosphatases 1 and 2A differentially prevent intrinsic and extrinsic apoptosis pathways.[J]. Biochem Biophys Res Commun. 2004,323(4):1313-1320.
    [12]Takagi Y, Futamura M, Yamaguchi K, et al. Alterations of the PPP2R1B gene located at 11q23 in human colorectal cancers.[J]. Gut.2000,47(2):268-271.
    [13]Calin G A, Di Iasio M G, Caprini E, et al. Low frequency of alterations of the alpha (PPP2R1A) and beta (PPP2R1B) isoforms of the subunit A of the serine-threonine phosphatase 2A in human neoplasms.[J]. Oncogene.2000,19(9):1191-1195.
    [14]Suzuki K, Takahashi K. Reduced expression of the regulatory A subunit of serine/threonine protein phosphatase 2A in human breast cancer MCF-7 cells.[J]. Int J Oncol.2003,23(5):1263-1268.
    [15]Neviani P, Santhanam R, Trotta R, et al. The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein.[J]. Cancer Cell.2005,8(5):355-368.
    [16]Piccaluga P P, Malagola M, Rondoni M, et al. Imatinib mesylate in the treatment of newly diagnosed or refractory/resistant c-KIT positive acute myeloid leukemia. Results of an Italian Multicentric Phase Ⅱ Study.[J]. Haematologica.2007,92(12):1721-1722.
    [17]Verstovsek S, Tefferi A, Cortes J, et al. Phase Ⅱ study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid diseases, including systemic mastocytosis.[J]. Clin Cancer Res.2008,14(12):3906-3915.
    [18]De La Rubia J, Regadera A, Martin G, et al. FLAG-IDA regimen (fludarabine, cytarabine, idarubicin and G-CSF) in the treatment of patients with high-risk myeloid malignancies.[J]. Leuk Res.2002,26(8):725-730.
    [19]Rowe J M. Optimal induction and post-remission therapy for AML in first remission.[J]. Hematology Am Soc Hematol Educ Program.2009:396-405.
    [20]Cristobal I, Garcia-Orti L, Cirauqui C, et al. PP2A impaired activity is a common event in acute myeloid leukemia and its activation by forskolin has a potent anti-leukemic effect.[J]. Leukemia.2011.
    [21]Liu Q, Zhao X, Frissora F, et al. FTY720 demonstrates promising preclinical activity for chronic lymphocytic leukemia and lymphoblastic leukemia/lymphoma.[J]. Blood. 2008,111(1):275-284.
    [22]Neviani P, Santhanam R, Oaks J J, et al. FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia.[J]. J Clin Invest.2007,117(9):2408-2421.
    [23]Payne S G, Oskeritzian C A, Griffiths R, et al. The immunosuppressant drug FTY720 inhibits cytosolic phospholipase A2 independently of sphingosine-1-phosphate receptors.[J]. Blood.2007,109(3):1077-1085.
    [1]Mrozek K, Heerema N A, Bloomfield C D. Cytogenetics in acute leukemia. [J]. Blood Rev.2004,18(2):115-136.
    [2]Bullinger L, Valk P J. Gene expression profiling in acute myeloid leukemia.[J]. J Clin Oncol.2005,23(26):6296-6305.
    [3]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.[J]. Blood.2002,100(1):59-66.
    [4]Shih L Y, Lin T L, Wang P N, et al. Internal tandem duplication of fins-like tyrosine kinase 3 is associated with poor outcome in patients with myelodysplastic syndrome.[J]. Cancer.2004,101(5):989-998.
    [5]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.[J]. Blood.2002,99(12):4326-4335.
    [6]Zwaan C M, Meshinchi S, Radich J P, et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia:prognostic significance and relation to cellular drug resistance.[J]. Blood.2003,102(7):2387-2394.
    [7]Wang Y Y, Zhou G B, Yin T, et al. AML1-ETO and C-KIT mutation/overexpression in t(8;21) leukemia:implication in stepwise leukemogenesis and response to Gleevec.[J]. Proc Natl Acad Sci U S A.2005,102(4):1104-1109.
    [8]Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias:an Italian retrospective study.[J]. Blood.2006,107(9): 3463-3468.
    [9]Schnittger S, Kohl T M, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival.[J]. Blood.2006,107(5):1791-1799.
    [10]Dash A, Gilliland D G. Molecular genetics of acute myeloid leukaemia.[J]. Best Pract Res Clin Haematol.2001,14(1):49-64.
    [11]Yarden Y, Kuang W J, Yang-Feng T, et al. Human proto-oncogene c-kit:a new cell surface receptor tyrosine kinase for an unidentified ligand.[J]. EMBO J.1987,6(11): 3341-3351.
    [12]Copeland N G, Gilbert D J, Cho B C, et al. Mast cell growth factor maps near the steel locus on mouse chromosome 10 and is deleted in a number of steel alleles.[J]. Cell. 1990,63(1):175-183.
    [13]Zsebo K M, Wypych J, Mcniece I K, et al. Identification, purification, and biological characterization of hematopoietic stem cell factor from buffalo rat liver--conditioned medium.[J]. Cell.1990,63(1):195-201.
    [14]Flanagan J G, Leder P. The kit ligand:a cell surface molecule altered in steel mutant fibroblasts.[J]. Cell.1990,63(1):185-194.
    [15]Huizinga J D, Thuneberg L, Kluppel M, et al. W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity.[J]. Nature.1995,373(6512):347-349.
    [16]Williams D E, Eisenman J, Baird A, et al. Identification of a ligand for the c-kit proto-oncogene.[J]. Cell.1990,63(1):167-174.
    [17]Martin F H, Suggs S V, Langley K E, et al. Primary structure and functional expression of rat and human stem cell factor DNAs.[J]. Cell.1990,63(1):203-211.
    [18]Geissler E N, Ryan M A, Housman D E. The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene.[J]. Cell.1988,55(1):185-192.
    [19]Anderson D M, Lyman S D, Baird A, et al. Molecular cloning of mast cell growth factor, a hematopoietin that is active in both membrane bound and soluble forms.[J]. Cell.1990,63(1):235-243.
    [20]Qiu F H, Ray P, Brown K, et al. Primary structure of c-kit:relationship with the CSF-1/PDGF receptor kinase family--oncogenic activation of v-kit involves deletion of extracellular domain and C terminus.[J]. EMBO J.1988,7(4):1003-1011.
    [21]Longley B J, Tyrrell L, Ma Y, et al. Chymase cleavage of stem cell factor yields a bioactive, soluble product.[J]. Proc Natl Acad Sci U S A.1997,94(17):9017-9021.
    [22]Arakawa T, Yphantis D A, Lary J W, et al. Glycosylated and unglycosylated recombinant-derived human stem cell factors are dimeric and have extensive regular secondary structure.[J]. J Biol Chem.1991,266(28):18942-18948.
    [23]Lux M L, Rubin B P, Biase T L, et al. KIT extracellular and kinase domain mutations in gastrointestinal stromal tumors.[J]. Am J Pathol.2000,156(3):791-795.
    [24]Kissel H, Timokhina I, Hardy M P, et al. Point mutation in kit receptor tyrosine kinase reveals essential roles for kit signaling in spermatogenesis and oogenesis without affecting other kit responses.[J]. EMBO J.2000,19(6):1312-1326.
    [25]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]. J Clin Invest.1993,92(4): 1736-1744.
    [26]Kitayama H, Kanakura Y, Furitsu T, et al. Constitutively activating mutations of c-kit receptor tyrosine kinase confer factor-independent growth and tumorigenicity of factor-dependent hematopoietic cell lines.[J]. Blood.1995,85(3):790-798.
    [27]Hashimoto K, Tsujimura T, Moriyama Y, et al. Transforming and differentiation-inducing potential of constitutively activated c-kit mutant genes in the IC-2 murine interleukin-3-dependent mast cell line.[J]. Am J Pathol.1996,148(1): 189-200.
    [28]Tsujimura T, Hashimoto K, Kitayama H, et al. Activating mutation in the catalytic domain of c-kit elicits hematopoietic transformation by receptor self-association not at the ligand-induced dimerization site.[J]. Blood.1999,93(4):1319-1329.
    [29]Ma Y, Carter E, Wang X, et al. Indolinone derivatives inhibit constitutively activated KIT mutants and kill neoplastic mast cells.[J]. J Invest Dermatol.2000,114(2): 392-394.
    [30]Ma Y, Cunningham M E, Wang X, et al. Inhibition of spontaneous receptor phosphorylation by residues in a putative alpha-helix in the KIT intracellular juxtamembrane region.[J]. J Biol Chem.1999,274(19):13399-13402.
    [31]London C A, Galli S J, Yuuki T, et al. Spontaneous canine mast cell tumors express tandem duplications in the proto-oncogene c-kit.[J]. Exp Hematol.1999,27(4): 689-697.
    [32]Ma Y, Longley B J, Wang X, et al. Clustering of activating mutations in c-KIT's juxtamembrane coding region in canine mast cell neoplasms.[J]. J Invest Dermatol. 1999,112(2):165-170.
    [33]Taniguchi M, Nishida T, Hirota S, et al. Effect of c-kit mutation on prognosis of gastrointestinal stromal tumors.[J]. Cancer Res.1999,59(17):4297-4300.
    [34]Ning Z Q, Li J, Mcguinness M, et al. STAT3 activation is required for Asp(816) mutant c-Kit induced tumorigenicity.[J]. Oncogene.2001,20(33):4528-4536.
    [35]Chian R, Young S, Danilkovitch-Miagkova A, et al. Phosphatidylinositol 3 kinase contributes to the transformation of hematopoietic cells by the D816V c-Kit mutant.[J]. Blood.2001,98(5):1365-1373.
    [36]Gari M, Goodeve A, Wilson G, et al. c-kit proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia.[J]. Br J Haematol.1999,105(4): 894-900.
    [37]Beghini A, Peterlongo P, Ripamonti C B, et al. C-kit mutations in core binding factor leukemias.[J]. Blood.2000,95(2):726-727.
    [38]Care R S, Valk P J, Goodeve A C, et al. Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias.[J]. Br J Haematol. 2003,121(5):775-777.
    [39]Kohl T M, Schnittger S, Ellwart J W, et al. KIT exon 8 mutations associated with core-binding factor (CBF)-acute myeloid leukemia (AML) cause hyperactivation of the receptor in response to stem cell factor.[J]. Blood.2005,105(8):3319-3321.
    [40]Beghini A, Ripamonti C B, Cairoli R, et al. KIT activating mutations:incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication.[J]. Haematologica.2004,89(8):920-925.
    [41]Goemans B F, Zwaan C M, Miller M, et al. Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia.[J]. Leukemia.2005, 19(9):1536-1542.
    [42]Shimada A, Taki T, Tabuchi K, et al. KIT mutations, and not FLT3 internal tandem duplication, are strongly associated with a poor prognosis in pediatric acute myeloid leukemia with t(8;21):a study of the Japanese Childhood AML Cooperative Study Group.[J]. Blood.2006,107(5):1806-1809.
    [43]Paschka P, Marcucci G, Ruppert A S, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21):a Cancer and Leukemia Group B Study.[J]. J Clin Oncol.2006,24(24):3904-3911.
    [44]Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML).[J]. Leukemia.2006,20(6):965-970.
    [45]Corbacioglu S, Kilic M, Westhoff M A, et al. Newly identified c-KIT receptor tyrosine kinase ITD in childhood AML induces ligand-independent growth and is responsive to a synergistic effect of imatinib and rapamycin.[J]. Blood.2006,108(10): 3504-3513.
    [46]Muroi K, Amemiya Y, Miura Y. Specificity of CD117 expression in the diagnosis of acute myeloid leukemia.[J]. Leukemia.1996,10(6):1048.
    [47]Di Noto R, Lo P C, Schiavone E M, et al. Stem cell factor receptor (c-kit, CD117) is expressed on blast cells from most immature types of acute myeloid mallignancies but is also a characteristic of a subset of acute promyelocytic leukaemia.[J]. Br J Haematol.1996,92(3):562-564.
    [48]Valverde L R, Matutes E, Farahat N, et al. C-kit receptor (CD117) expression in acute leukemia.[J]. Ann Hematol.1996,72(1):11-15.
    [49]Nomdedeu J F, Mateu R, Altes A, et al. Enhanced myeloid specificity of CD117 compared with CD13 and CD33.[J]. Leuk Res.1999,23(4):341-347.
    [50]Uckan D, Hicsonmez G, Yetgin S, et al. CD34/CD117 co-expression in childhood acute leukemia.[J]. Leuk Res.2000,24(3):201-206.
    [51]Nakata Y, Kimura A, Katoh O, et al. c-kit point mutation of extracellular domain in patients with myeloproliferative disorders.[J]. Br J Haematol.1995,91(3):661-663.
    [52]Hirakawa K, Kimura S, Chikayama S, et al. Primary acute myeloid leukemia cells negative for c-kit receptor protein expression do not proliferate in vitro in response to colony-stimulating factors.[J]. Leukemia.1996,10(2):279-287.
    [53]Heinrich M C, Blanke C D, Druker B J, et al. Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies.[J]. J Clin Oncol.2002,20(6):1692-1703.
    [54]Demetri G D. Targeting c-kit mutations in solid tumors:scientific rationale and novel therapeutic options.[J]. Semin Oncol.2001,28(5 Suppl 17):19-26.
    [55]Nanri T, Matsuno N, Kawakita T, et al. Imatinib mesylate for refractory acute myeloblastic leukemia harboring inv(16) and a C-KIT exon 8 mutation.[J]. Leukemia. 2005,19(9):1673-1675.
    [56]Gleixner K V, Mayerhofer M, Aichberger K J, et al. PKC412 inhibits in vitro growth of neoplastic human mast cells expressing the D816V-mutated variant of KIT: comparison with AMN107, imatinib, and cladribine (2CdA) and evaluation of cooperative drug effects.[J]. Blood.2006,107(2):752-759.
    [57]Growney J D, Clark J J, Adelsperger J, et al. Activation mutations of human c-KIT resistant to imatinib mesylate are sensitive to the tyrosine kinase inhibitor PKC412.[J]. Blood.2005,106(2):721-724.
    [58]Beghini A, Bellini M, Magnani I, et al. STI 571 inhibition effect on KITAsn822Lys-mediated signal transduction cascade.[J]. Exp Hematol.2005,33(6): 682-688.
    [59]Stone R M, O'Donnell M R, Sekeres M A. Acute myeloid leukemia.[J]. Hematology Am Soc Hematol Educ Program.2004:98-117.
    [60]O'Farrell A M, Yuen H A, Smolich B, et al. Effects of SU5416, a small molecule tyrosine kinase receptor inhibitor, on FLT3 expression and phosphorylation in patients with refractory acute myeloid leukemia.[J]. Leuk Res.2004,28(7):679-689.
    [61]Brown P, Levis M, Shurtleff S, et al. FLT3 inhibition selectively kills childhood acute lymphoblastic leukemia cells with high levels of FLT3 expression.[J]. Blood.2005, 105(2):812-820.
    [62]Stone R M, Deangelo D J, 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.[J]. Blood.2005,105(1):54-60.
    [63]Corbin A S, Griswold I J, La Rosee P, et al. Sensitivity of oncogenic KIT mutants to the kinase inhibitors MLN518 and PD180970.[J]. Blood.2004,104(12):3754-3757.
    [64]Boissel N, Leroy H, Brethon B, et al. Incidence and prognostic impact of c-Kit, FLT3, and Ras gene mutations in core binding factor acute myeloid leukemia (CBF-AML).[J]. Leukemia.2006,20(6):965-970.
    [65]Chevallier P, Hunault-Berger M, Larosa F, et al. A phase Ⅱ trial of high-dose imatinib mesylate for relapsed or refractory c-kit positive and Bcr-Abl negative acute myeloid leukaemia:the AFR-15 trial.[J]. Leuk Res.2009,33(8):1124-1126.