FLT3 expression and IL10 promoter polymorphism in acute myeloid leukemia with RUNX1-RUNX1T1

2014 ◽  
Vol 42 (2) ◽  
pp. 451-456 ◽  
Author(s):  
Myungshin Kim ◽  
Jiyeon Kim ◽  
Jung Rok Kim ◽  
Eunhee Han ◽  
Joonhong Park ◽  
...  
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1427-1427 ◽  
Author(s):  
Tristan Knight ◽  
Xinan Qiao ◽  
Holly Edwards ◽  
Hai Lin ◽  
Jeffrey W. Taub ◽  
...  

Abstract Introduction: FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase, and is mutated in approximately one third of acute myeloid leukemia (AML) patients; this mutation confers a poor prognosis. Two FLT3 mutations are commonly seen in AML: internal tandem duplications (ITD) in the juxtamembrane domain (~25% of AML), and point mutations in the receptor tyrosine kinase at codon 835 (D835) (~7% of AML). Both mutations result in constitutive FLT3 activation, causing downstream activation of multiple pathways, in particular, those involved in cell survival including the RAS-RAF-MEK-ERK, JAK-STAT5, and PI3K/AKT pathways. PI3K-AKT may also be activated by AXL, also a tyrosine kinase, via its targets PLC, Grb2, and PI3K. Logically, then, inhibition of FLT3 is a promising pharmacological approach for treating this subtype of AML. Gilteritinib (ASP-2215) is a novel dual inhibitor of FLT3 and AXL, exposure to which results in upregulation of FLT3 as a resistance mechanism. Previously, we found that the novel dual PI3K/histone deacetylase (HDAC) inhibitor CUDC-907 downregulates FLT3 expression in AML cells (Figure 1A). Additionally, inhibition of FLT3 and AXL by gilteritinib may not result in robust inactivation of both the PI3K-Akt and MEK/ERK pathways due to crosstalk between the two pathways. Thus, our hypothesis was that CUDC-907 would sensitize AML cells to gilteritinib, resulting in concurrent inhibition of all the downstream signaling pathways of FLT3 and AXL, leading to synergistic antileukemic activities again FLT3-mutated AML (Figure 1B). Methods: FLT3-ITD AML cell lines (MV4-11 and MOLM-13) and primary patient samples were treated with CUDC-907, gilteritinib, both, or neither for 24 hours, at clinically achievable concentrations. Annexin V/Propidium Iodide (PI) staining and flow cytometry analyses was performed, and combination indexes (CI) calculated; CI<1, CI=1, and CI>1 indicating synergistic, additive, or antagonistic effects, respectively. Western blots were performed after treatment for 0-24 hours to determine protein expression of relevant targets. Results: CUDC-907 and gilteritinib demonstrated potent synergistic antileukemic effects in FLT3-ITD AML cell lines and FLT3-ITD patient samples (AML#171, AML#180), the combination exceeding either in isolation (Figure 1C). These findings were confirmed via western blot, which showed accentuated upregulation of cleaved caspase3 with combination therapy, in both cell lines and one patient sample, demonstrating drug-induced apoptosis. We confirmed that CUDC-907 abolishes gilteritinib-induced expression of FLT3 in a time-dependent fashion in cell lines MV4-11 and MOLM-13 (Figure 1D). Gilteritinib treatment decreased p-AKT, p-S6, and p-STAT5, while inhibition of the ERK pathway, as assessed by p-ERK expression, varied amongst the samples (Figure 1E). CUDC-907 treatment decreased both p-AKT and p-ERK. MOLM-13 cells showed increased p-ERK following gilteritinib treatment and increased p-STAT5 after CUDC-907 treatment. In all samples, combination of gilteritinib with CUDC-907 resulted in decrease of p-STAT5 and p-S6, similar to gilteritinib treatment alone, and further reduction of p-AKT and p-ERK compared to single drug treatments. Gilteritinib treatment also reduced expression of anti-apoptotic protein Mcl-1, which was further decreased in combination treated cells. Subsequently, time-course analysis was performed in both cell lines; findings were consistent with prior observations, and confirmed that protein expression changed over time, in relation to gilteritinib/CUDC-907/combined treatment exposure. Conclusion: We confirmed that CUDC-907 and Gilteritinib synergistically induce apoptosis in both cell lines and primary patient samples derived from patients with FLT3-ITD AML, and that CUDC-907 abolishes Gilteritinib-induced FLT3 expression. Additionally, the combination cooperatively inhibits the PI3K-AKT, JAK-STAT, and RAS-RAF pathways, while preventing escape via alternative pathways. Our results provide a strong foundation for subsequent in vivo murine studies, and eventual clinical evaluation of the combination of gilteritinib and CUDC-907 for the treatment of AML. Figure 1. Figure 1. Disclosures Ge: MEI Pharma: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2609-2609 ◽  
Author(s):  
Katherine Tarlock ◽  
Todd A. Alonzo ◽  
Michael R. Loken ◽  
Robert B. Gerbing ◽  
Richard Aplenc ◽  
...  

Abstract FLT3 is a highly expressed cell surface receptor in a majority of acute leukemias with near universal expression in acute myeloid leukemia (AML). High FLT3 expression level in conjunction with 11q23/MLL-rearrangement has been associated with poor prognosis in pre-B acute lymphoblastic leukemia (ALL) and is considered a potential therapeutic target through FLT3 inhibition. In vitro studies have demonstrated enhanced sensitivity of ALL and AML cells with high FLT3 expression to the cytotoxic effects of FLT3 inhibition. Although the clinical impact of high allelic ratio FLT3/ITD has been demonstrated in AML, the significance of FLT3 expression in those without the mutation has not been well studied, and all previous studies have been limited to evaluation of FLT3 transcript levels for correlation with outcome. Here we present the results of a prospective evaluation of FLT3 cell surface protein expression (CD135) on the diagnostic leukemic blast population as determined by multi-dimensional flow cytometry (MDF) in specimens from patients treated on COG AAML0531. Of the 495 patients enrolled after September 25, 2008, 366 patients enrolled on the accompanying biology study and had diagnostic specimens for evaluation of central CD135 expression by MDF. All diagnostic specimens underwent central MDF analysis for CD135 expression. FLT3 mutation data was available on all 366 patients, 57 patients were FLT3/ITD (15.6%), 21 were FLT3/ALM (5.7%) and the remaining 290 were FLT3wild type (FLT3/WT). There was significant variability of CD135 expression across the population with a mean fluorescence intensity (MFI) ranging from 3-232 (median 18). Median CD135 expression for those with FLT3/ITD, FLT3/ALM, FLT3/WT was 22 vs. 19 vs. 17 respectively (p=NS). Patients were divided into quartiles based on CD135 expression and clinical characteristics and outcome were correlated with CD135 expression across the four quartiles. There was not a significant difference in CD135 expression by age, gender, race, or ethnicity across the four quartiles. Those with high CD135 expression (Q4) had similar median diagnostic WBC counts as those with lower CD135 (Q1-3), although median diagnostic blast % in those in Q4 was significantly elevated (p=0.003). Nearly half of those in Q4 were FAB M5 compared to 13% in Q1-3 (p<0.001) and all those with FAB M7 had low CD135 expression (p=0.004). Evaluation of the diagnostic cytogenetics and CD135 expression demonstrated significant correlation of CD135 expression with MLL translocations, as 22% of patients in Q4 had MLLtranslocations vs. 11% in Q1-3 (p=0.005). This was primarily driven by patients with t(9;11), p=0.001. There was an inverse correlation between inv(16) and CD135 expression as only 2% of those in Q4 had inv(16) compared to 15% in Q1-3 (p<0.001). CD135 was then correlated with response to induction and post-induction outcome. Complete remission (CR) rate was comparable between those with high or lower CD135 expression (p=0.76). Those with high or lower CD135 expression had similar 3-yr overall survival (OS) from diagnosis of 70% and 66% respectively (p=0.9) and relapse risk from CR of 42% and 36% respectively (p=0.35). Given the known prognostic and therapeutic significance of FLT3/ITD, we evaluated any prognostic implications of FLT3 expression in patients without FLT3/ITD, most of whom lack other cytogenetic risk features. Similar to the entire cohort, in the FLT3/ITD-negative cohort CD135 expression correlated with FAB M5 (p<0.001), t(9;11), (p<0.001) and inversely correlated with inv(16), p=0.001. 3-yr OS for those in Q4 was 71% vs. 67% in Q1-3 (p=0.955). Given the association of MLL and CD135 expression, we evaluated the association of CD135 expression with outcome in patients with MLLtranslocations (N=73); in these patients the 3-yr event free survival (EFS) was similar between those with high or lower CD135 expression (p=0.621). In summary, we found that CD135 surface expression did not correlate with FLT3 mutations or clinical outcomes. Although FLT3 expression in FLT3/ITD-negative patients does not appear to offer additional prognostic information with current therapy, it may provide a therapeutic target in a subset of high expressing patients. This study also demonstrates that elevated FLT3 expression is associated with MLL rearrangements and warrants further study in this population with regards to potential prognostic and therapeutic implications. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 42 (5) ◽  
pp. 1973-1984 ◽  
Author(s):  
Gang-Zhi Zhu ◽  
Yong-Long Yang ◽  
Yan-Jiao Zhang ◽  
Wei Liu ◽  
Mu-Peng Li ◽  
...  

Background/Aims: Acute myeloid leukemia (AML) is a heterogeneous clonal disease and patients with AML who harbor an FMS-like tyrosine kinase 3 (FLT3) mutation present several dilemmas for the clinician. This study aims to identify novel targets for explaining the dilemmas. Methods: We analyzed four microarray gene expression profiles to investigate changes in whole genome expression associated with FLT3-ITD mutation. Results: We identified 22 differentially expressed genes which are commonly expressed among all four profiles. Kaplan-Meier analysis of the dataset GSE12417 revealed that low expression of AHSP, EPB42, GYPC and HEMGN predicted poor prognosis (AHSP: P=0.0317, HR=1.894; EPB42: P=0.0382, HR=1.859; GYPC: P=0.0015, HR=2.051; HEMGN: P=0.0418, HR=1.838 in GSE12417 test cohort; AHSP: P=0.0279, HR=1.548; EPB42: P=0.0398, HR=1.505; GYPC: P=0.0408, HR=1.501; HEMGN: P=0.0143, HR=1.630 in GSE12417 validation cohort). When patients were FLT3-ITD positive, the expression of FLT3 was significantly increased (all P<0.05 in four profiles), and correleation analysis of four profiles revealed that the expression of the four candidate genes negatively correlated with FLT3 expression. Conclusions: Our findings suggest that AHSP, EPB42, GYPC and HEMGN may be suitable biomarkers for diagnostic or therapeutic strategies for FLT3-ITD-positive AML patients.


2008 ◽  
Vol 14 (23) ◽  
pp. 7896-7899 ◽  
Author(s):  
Parinda A. Mehta ◽  
Robert B. Gerbing ◽  
Todd A. Alonzo ◽  
James S. Elliott ◽  
Tiffany A. Zamzow ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2552-2552
Author(s):  
Lars Palmqvist ◽  
Nicolas Pineault ◽  
Bob Argiropoulos ◽  
Adrian Wan ◽  
Keith R. Humphries

Abstract The TALE family member and HOX cofactor MEIS1 is important in leukemic transformation. MEIS1 has, although non-leukemogenic on its own, been shown to strongly collaborate with several HOX genes and NUP98-HOX fusions to induce acute myeloid leukemia (AML). We have recently described a novel in vitro culture system of cell lines established from murine primary bone marrow cells transduced with the AML-associated fusion gene NUP98-HOXD13 or an engineered NUP98-HOXA10 fusion. These pre-leukemic NUP98-HOX cell lines are transplantable and can efficiently be converted into AML-inducing cells upon MEIS1 transduction. Conveniently, the MEIS1 transduced cells can be purified and preserve their leukemogenic potential even after extensive in vitro expansion. Thus, the availability of the NUP98-HOX cell lines system provided the opportunity to investigate and characterize the mechanism of MEIS1-mediated AML-conversion. Potentially interesting target or candidate genes were screened for expression changes between the parental pre-leukemic lines and AML-inducing MEIS1 transduced cell lines with quantitative RT-PCR and Western blotting. Aberrant expression or mutations of the receptor tyrosine kinase FLT3 gene is a common finding in human AML. Interestingly, Flt3 was found induced 5 to 10 fold in MEIS1 transduced cell lines compared to the parental cell lines. The observed increase in Flt3 expression provided the MEIS1 transduced cells with Flt3 ligand driven growth. This was not seen in the parental cell lines, which could not proliferate with Flt3 ligand as single cytokine or with a MEIS1-homeodomain mutant expressing cell line. Importantly, the Flt3 inhibitor AG1295 could block the proliferative effect of the Flt3 ligand in the MEIS1 transduced cell lines. To test whether Flt3 could substitute for MEIS1-mediated induction of AML in NUP98-HOX pre-leukemic cells, a NUP98-HOXA10 cell line was transduced with an MSCV-Flt3-IRES-YFP construct. The resulting Flt3-transduced cells were shown to express Flt3 at levels similar to that of MEIS1 transduced cells without any significant increase in endogenous Meis1 expression. Transplantation of these cells into mice led to lethal and transplantable AML with a median disease onset of 116 days (n=8) compared to 59 days for MEIS1 (n=4), whereas control transplants remained healthy (n=2). In conclusion, this study demonstrates that MEIS1 can induce Flt3 expression and that Flt3 can collaborate with NUP98-HOX fusion genes in the induction of acute myeloid leukemia. Furthermore, theses results suggest a model in which the leukemogenic synergism of MEIS1 on HOX-mediated leukemia might in part be mediated through FLT3-dependent pathways.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2187-2187
Author(s):  
Helmut R. Salih ◽  
Tina Baessler ◽  
Martin Hofmann ◽  
Ludger Grosse-Hovest ◽  
Maximilian Bamberg ◽  
...  

Abstract Abstract 2187 For more than a decade, chimeric/humanized second generation monoclonal antibodies (mAb) are used in cancer therapy. While their success e.g. in lymphoma therapy is undisputed, mAb are not established as treatment for acute myeloid leukemia (AML). Moreover, the therapeutic activity of available mAb leaves ample room for improvement. In the recent years, modifications of the human IgG1 Fc-part have enabled the development of third generation mAb with markedly improved capability to recruit Fc-receptor bearing immune effector cells. We here report on the development and evaluation of an Fc-optimized mAb directed to FLT3, an antigen expressed on leukemic cells of the majority of AML patients. In vitro, this antibody termed 4G8-SDIEM effectively induced antibody-dependent cellular cytotoxicity (ADCC) against FLT3-expressing AML cells at concentrations as low as 10ng/ml. Compared to the parental humanized antibody, the ADCC activity of 4G8-SDIEM was increased by a factor of 100. FLT3 expression on primary AML cells (range, 500–5000 molecules) was considerably higher as compared to healthy hematopoietic cells (several hundred molecules). 4G8-SDIEM did not induce relevant ADCC against healthy cells and did not decrease the CFU-forming capacity of bone marrow (BM) cells in vitro. The mAb was then produced in pharmaceutical quality and quantity at a university-owned production unit and used for compassionate need treatment of a 30 year old AML patient (FAB M0, complex kariotype with 45, XY, inv(3)(q21q26), -7, 9q-) with relapse after haploidentical and unrelated donor stem cell transplantation (SCT). Preclinical testing revealed that 4G8-SDIEM effectively induced ADCC of the patients peripheral blood (PB) mononuclear cells (PBMC) against NALM16 leukemia cells and autologous leukemic blasts. Directly before initiation of treatment, the percentage of leukemic blasts among the patients PBMC was 11% and 38% in PB and BM, respectively, and his AML cells displayed homogeneous FLT3 expression. The percentage of NK cells (CD56+CD3-) was 7% and 5% in PB and BM, respectively, with less than 2% displaying an activated phenotype (CD69+). 4G8-SDIEM was applied in escalating doses (d1: 10μg; d2: 100μg; d3: 1mg; d4: 2mg; d5, 7, 10: 10mg). Several hours after the first 10 mg-dose 5×108 CD3/CD19-depleted donor lymphocytes were infused. Besides elevated temperature (max. 38.2°C), no relevant side effects of treatment were observed. After the first first 10mg dose, BM blasts were nearly saturated with mAb as judged by crosscompetition assays. Serum concentration of 4G8-SDIEM reached 1.0μg/ml 1h after the first 10mg dose declining to 0.4 μg/ml 24h later. Upon treatment, serum levels of the index cytokines TNF and IL-6 (peak d4, 60 and 27pg/ml, respectively) and the percentage of activated NK cells (peak d6, 42%) in PB increased rapidly. Already at day 4, leukemic blasts were nearly completely cleared from the PB (<2%), while effects in BM were less pronounced (down to 15%) indicating that lower effector to target cell ratios in the BM limit the therapeutic activity of the mAb. In any case, the anti-leukemic effects of 4G8-SDIEM remained transient, as 10 days after initiation of therapy blast counts in PB and BM reached and later exceeded pre-treatment levels. Thus we conclude that although 4G8-SDIEM clearly exerts anti-leukemic effects in vivo, it may not be capable of inducing long-lasting responses of AML in proliferative phase. Rather, we envisage application of the mAb in settings with suitably high effector to target cell ratios such as minimal residual disease in morphological complete remission, possibly in combination with adoptive NK cell transfer. As of now, development of 4G8-SDIEM cost |CE2 million and required 2.5 years from bench to bedside. Thus, development and early clinical evaluation of novel mAb can be achieved at academic institutions at reasonable costs and time before initiation of larger clinical Phase II/III studies by the pharmaceutical industry. Such an approach may not only accelerate the developmental process for anti-tumor mAb, but rather for innovative biological drugs in general. Disclosures: Hofmann: University of Tuebingen, Germany: Patents & Royalties. Grosse-Hovest: University of Tuebingen, Germany: Patents & Royalties. Aulwurm: University of Tuebingen, Germany: Patents & Royalties. Buehring: University of Tuebingen, Germany: Patents & Royalties. Jung: University of Tuebingen, Germany: Patents & Royalties.


2019 ◽  
Vol 19 ◽  
pp. S239-S240
Author(s):  
Bartholomew Eisfelder ◽  
Joseph Wynne ◽  
Margert Colton ◽  
Jason Cheng ◽  
Olatoyosi Odenike ◽  
...  

2015 ◽  
Vol 16 (7) ◽  
pp. 2707-2712 ◽  
Author(s):  
Anuradha Cingeetham ◽  
Sugunakar Vuree ◽  
Sangeeta Jiwatani ◽  
Sailaja Kagita ◽  
Nageswara Rao Dunna ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document