The Mll PTD and Flt3 ITD Double Knock-In Mouse Develops Acute Leukemia and Recapitulates Phenotypic, Molecular and Epigenetic Characteristics of the Counterpart Human Acute Myeloid Leukemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 150-150 ◽  
Author(s):  
Nicholas Zorko ◽  
Susan P. Whitman ◽  
Kelsie Bernot ◽  
Myntee T. Ngangana ◽  
Ronald Siebenaler ◽  
...  

Abstract Abstract 150 Background. The Mll PTD and Flt3 ITD are co-present in a subset of adult patients (pts) with cytogenetically normal (CN) acute myeloid leukemia (AML) and poor clinical outcomes. While the single mutant knock-in (KI) mice (Mll PTD or Flt3 ITD) exhibit enhanced myeloid progenitor self-renewal or reduced apoptosis, respectively, neither model develops acute leukemia. We hypothesized that with mutant expression driven via the endogenous promoters, the two mutations may cooperate in vivo to induce an acute leukemia that mimics the human counterpart. Methods. Single mutant heterozygous KI mice were crossed to produce the PTD/ITD double KI. PTD/ITD mice were bred with the homozygous Flt3 ITD to generate the PTD/ITD2 genotype. An AML diagnosis was based on blood differentials, immunophenotyping, tissue pathology and transplantability. Real time RT-PCR and 5'-methylcytosine LC/MS assays measured gene expression and global DNA methylation levels, respectively. Results. PTD/ITD and PTD/ITD2 mice developed transplantable, CN-AML/undifferentiated leukemia exhibiting expansion of monocytic/myelomonocytic Gr1±/Mac1+ and/or immature CD3−/CD19−/CD117+/Mac1−/B220lo cell populations, splenomegaly, leukocytosis, anemia and thrombocytopenia. PTD/ITD mice had significantly reduced lifespans compared to mice with single mutant PTD and ITD KIs and wild-type (Wt) controls (medians: 50, 99, 88, 94 weeks, respectively; P<0.001) (Figure 1). Increased ITD gene dosage (PTD/ITD2) was associated with an even shorter lifespan (median: 16 weeks) (Figure 1). This is consistent with the poor prognosis conferred by high FLT3 ITD-to-FLT3 wild-type (WT) gene ratio in diagnostic leukemia blasts from AML pts treated with intensive chemotherapy. As in human MLL PTD AML, the Mll WT allele was downregulated in the murine model. Mll WT expression was >2-fold lower in bone marrow (BM) of leukemic PTD/ITD mice compared to age-matched single mutant KIs or Wt controls. HoxA9 and its cofactor Meis1 were upregulated 15- and 5-fold, respectively, in PTD/ITD mice with leukemia versus Wt BM. Yet, compared to Wt BM, single PTD KI exhibited increased HoxA9 (∼6-fold) but not Meis1, implicating an expression threshold for HoxA9 and a crucial role for Meis1 for the development of acute leukemia in the double KI. Consistent with Flt3 being a downstream transcriptional target of Meis1, total Flt3 mRNA (WT and ITD) levels increased 3-fold in the leukemic PTD/ITD mice relative to either single mutant KIs or Wt controls. Furthermore, one consequence of constitutive Flt3 ITD kinase activity is the upregulation of the anti-apoptotic kinase, Pim1, in human AML. Compared to Wt BM, a 2-fold increase in Pim-1 expression was observed in single ITD KI and a 6-fold increase was observed in leukemic PTD/ITD BM, while expression was unchanged in the single PTD KI BM. Finally, MLL PTD presence in human AML associates with increased global DNA methylation and silencing of tumor suppressor genes. We observed 3-fold higher transcript levels of a de novo methyltransferase, DNA methyltransferase 3b (DNMT3b), increased global DNA methylation and ≥2-fold decrease in the expression of tumor suppressors Id4, Shp1 and Cdkn1b in BM of leukemia PTD/ITD mice compared to age-matched single mutant KIs and Wt controls. Conclusion. The Flt3 ITD and Mll PTD, expressed via their endogenous promoters, cooperate in vivo to give rise to AML and acute undifferentiated leukemia. Elevations of Meis1 and DNMT3b solely in PTD/ITD animals appear to be critical points of dysregulation leading to development of acute leukemia. This novel murine model phenotypically, molecularly, and epigenetically mimics the human AML counterpart, thus making it highly relevant for examining critical pathways in acute myeloid leukemogenesis, investigating leukemia stem/initiating cell biology and microenvironment contributions, and testing novel targeting therapeutics. Disclosures: No relevant conflicts of interest to declare.

2013 ◽  
Vol 37 (2) ◽  
pp. 190-196 ◽  
Author(s):  
Rainer Claus ◽  
Dietmar Pfeifer ◽  
Maika Almstedt ◽  
Manuela Zucknick ◽  
Björn Hackanson ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1375-1375
Author(s):  
Marina Díaz-Beyá ◽  
Alfons Navarro ◽  
Rut Tejero ◽  
Marta Pratcorona ◽  
Tania Díaz ◽  
...  

Abstract Abstract 1375 Introduction: Acute myeloid leukemia (AML) is a highly heterogeneous disease, with diverse genetic and epigenetic variables determining sensitivity to current standard therapy. A disruption of the normal DNA methylation pattern, which can result in altered gene and microRNA (miRNAs) expression, has been observed in different AML subtypes. Hydroxymethylation of 5-methylcytosine (5-mC) has recently been described as an intermediate key step in the process of DNA demethylation. Nonetheless, the correlation of DNA methylation and hydroxymethylation levels with clinical and biological characteristics and clinical outcome in AML is mostly unknown. Aim: To investigate the prognostic impact of overall methylation and hydroxymethylation levels in patients with intermediate-cytogenetic risk AML (IR-AML) and to identify miRNAs correlated with methylation and hydroxymethylation in these patients. Patients and Methods: We have analyzed 86 IR-AML patients (median age, 53 [range, 17–74]; 52% males) who received intensive therapy from 1994 to 2009 in a single institution. The level of overall methylation and hydroxymethylation in total DNA was estimated after determining the percentage of 5-mC and hydroxymethylcytosine (5-hmC), using anti-5-mC and anti-5-hmC monoclonal antibodies (MethylFlash Methylated or Hydroxymethylated DNA Quantification Kit, Epigentek). The expression of 670 mature miRNAs was analyzed using TaqMan Human MicroRNA Arrays (Applied Biosystems). The statistical analysis was performed with SPSS version 15.0.1 and R software version 2.9.0. MaxStat package of R was used to determine the optimal cutoffs. Results: The univariate analysis for overall methylation showed that patients with lower levels of methylation (cutoff < percentile 75) had shorter overall survival (OS) than those with higher 5-mC levels (5-year OS: 30±6% vs 52±11%; p=0.03) and a trend for shorter leukemia-free survival (LFS)(p=0.06). Overall methylation levels did not show any correlation with clinical features at diagnosis or with gene mutations, including DNMT3A. Concerning hydroxymethylation, patients with lower 5-hmC levels had a worse prognosis than those with higher 5-hmC levels, with a lower complete response rate (79% vs. 96%; p=0.04), shorter OS (5-yr OS: 21± 6% vs. 55± 6%; p=0.008), and shorter LFS (5-yr LFS: 24±8% vs. 52 ±10%; p=0.03). Interestingly, when analyzed as a continuous variable, 5-hmC levels retained their prognostic value as a marker of response rate (T-test, p=0.007), OS (Cox, p=0.015), and LFS (Cox, p=0.041). Moreover, 5-hmC levels were inversely correlated with FLT3-ITD (p=0.001) and the FLT3-ITD/FLT3wild-type ratio (Pearson correlation:-0.6; p=0.01). The multivariate analyses, including the main clinical and biological variables, identified older age, wild-type NPM1, FLT3-ITD, and lower 5-hmC levels (HR=3.072; 95% CI: 1.096–3.917; p=0.025) as independent prognostic markers of shorter OS and wild-type NPM1, FLT3-ITD, and lower 5-hmC levels (HR=2.002, 95% CI: 1.032–3.881, p=0.040) as independent prognostic markers of shorter LFS. Of note, lower 5-hmC levels retained their value as a marker of worse prognosis in the subgroup of IR-AML patients with unfavorable molecular markers (wild-type NPM1 and CEBPA and/or FLT3-ITD; p= 0.037). Finally, we have identified a 3-miRNA signature (miR-378*, p=0.02; miR-493, p=0.02; and miR-181, p=0.02) associated with global methylation levels, and a 12-miRNA signature associated with hydroxymethylation, including miR-183* (p=0.001), miR-125a-3p (p= 0.01), miR-586 (p=0.02), and miR-513–3p (p=0.02). Conclusions: Hydroxymethylation levels appear as an independent prognostic factor in IR-AML and maintain their prognostic value in the subset of patients with unfavorable molecular markers. Moreover, methylation and hydroxymethylation are associated with specific miRNA profiles. Further studies are warranted to confirm the clinical impact of these findings and to clarify the underlying molecular mechanisms. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A474-A474
Author(s):  
Yifan Zhai ◽  
Tapan Kadia

BackgroundAcute myeloid leukemia (AML) is the most common form of acute leukemia in adults, with an incidence that increases with age and a generally poor prognosis. This aggressive blood and bone-marrow malignancy is characterized by rapid and uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Patients with R/R AML have very few approved effective treatment options, especially in the absence of a targetable mutation. Alrizomadlin is a novel, orally active, potent, small-molecule selective inhibitor that destabilizes the p53-MDM2 complex and activates p53-mediated apoptosis in tumor cells with wild-type TP53 and/or MDM2 amplification. In acute leukemia human wild-type TP53 AML cell lines and xenograft models, alrizomadlin potently inhibited tumor cell growth when administered alone or with concomitant chemotherapy.MethodsThis US open-label study is evaluating the safety and tolerability of alrizomadlin, with or without 5-azacitidine, in adults with histologically confirmed R/R AML and adequate organ function. Eligible candidates will have AML with no known available therapies that are either indicated or expected to confer a durable response. In Part 1 of this trial, the safety and tolerability of alrizomadlin monotherapy are being assessed by evaluating the dose-limiting toxicity rate during the first 4 weeks of treatment, using a standard 3+3 design. The starting once-daily oral dose of alrizomadlin administered on Day 1 to 5 of every 28-day cycle is 100 mg, increasing to 150, 200, and 250 mg in each subsequent cohort. The severity of adverse events is being assessed using NCI CTCAE v5.0. Once the recommended phase 2 dose (RP2D) has been determined, 3 to 6 additional patients will be enrolled in the dose-expansion phase. In Part 2, alrizomadlin will be administered in combination with 5-azacitidine 75 mg/m2/day on Days 1–7 of a 28-day cycles. Alternatively, a 5-days-on, 2-days-off, 2-days-on schedule is allowed if consecutive day infusion is not available. A standard 3+3 design will also be implemented to determine the maximum tolerated dose/RP2D in the dose-escalation phase. Once the RP2D has been determined, there will be an expansion cohort of up to 15 patients. As of July 13, 2021, 2 patients have been enrolled in the alrizomadlin monotherapy dose-escalation phase. The overall estimated enrollment will be 69 study participants. Internal study identifier APG115AU101. ClinicalTrials.gov identifier: NCT04358393.Trial RegistrationClinicalTrials.gov identifier: NCT04358393


2008 ◽  
Vol 32 (7) ◽  
pp. 1091-1100 ◽  
Author(s):  
Jianbiao Zhou ◽  
Jiaying Khng ◽  
Viraj J. Jasinghe ◽  
Chonglei Bi ◽  
Chiew Hoon Serene Neo ◽  
...  

2021 ◽  
Vol 11 (5) ◽  
Author(s):  
Maria H. Gilleece ◽  
Avichai Shimoni ◽  
Myriam Labopin ◽  
Stephen Robinson ◽  
Dietrich Beelen ◽  
...  

AbstractMeasurable residual disease (MRD) prior to hematopoietic cell transplant (HCT) for acute myeloid leukemia (AML) in first complete morphological remission (CR1) is an independent predictor of outcome, but few studies address CR2. This analysis by the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation registry assessed HCT outcomes by declared MRD status in a cohort of 1042 adult patients with AML CR2 at HCT. Patients were transplanted 2006–2016 from human leukocyte antigen (HLA) matched siblings (n = 719) or HLA 10/10 matched unrelated donors (n = 293). Conditioning was myeloablative (n = 610) or reduced-intensity (n = 432) and 566 patients (54%) had in-vivo T cell depletion. At HCT, 749 patients (72%) were MRD negative (MRD NEG) and 293 (28%) were MRD positive (MRD POS). Time from diagnosis to HCT was longer in MRD NEG than MRD POS patients (18 vs. 16 months (P < 0.001). Two-year relapse rates were 24% (95% CI, 21–28) and 40% (95% CI, 34–46) in MRD NEG and MRD POS groups (P < 0.001), respectively. Leukemia-free survival (LFS) was 57% (53–61) and 46% (40–52%), respectively (P = 0.001), but there was no difference in terms of overall survival. Prognostic factors for relapse and LFS were MRD NEG status, good risk cytogenetics, and longer time from diagnosis to HCT. In-vivo T cell depletion predicted relapse.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2981-2981
Author(s):  
Emmet McCormack ◽  
Ingvild Haaland ◽  
Gurid Venås ◽  
Rakel Brendsdal Forthun ◽  
Øystein Bruserud ◽  
...  

Abstract Nutlin-3 is a small-molecule antagonist of MDM2 that induces non-genotoxic stabilization and activation of the tumor suppressor protein p53, resulting in therapeutic effects in tumor models comprising wild type TP53. Valproic acid (VPA) is an anti-convulsive drug with histone deacetylase (HDAC) inhibitor activity that induces differentiation and apoptosis in acute myeloid leukemia (AML) cells. Both therapeutic compounds indirectly affect the regulation of p53; nutlin-3 by inhibiting MDM2, the main negative regulator of p53, and VPA by inhibiting HDACs that participate in p53 deacetylation and destabilization. While mutations in TP53 occur in less than 10% of AML, over-expression of MDM2 is frequently observed. Furthermore, aberrant recruitment of histone deacetylases (HDACs) is seen in AML, leading to block of myeloid differentiation. We therefore hypothesized that concomitant inhibition of MDM2 and HDACs would synergistically induce p53-mediated apoptosis and inhibit tumor growth. We examined the anti-leukemic effects of nutlin-3 in combination with low doses valproic acid in AML cell lines, primary AML cells, and in an in vivo xenograft model using optical imaging. To evaluate the efficacy of the combination of nutlin-3 and VPA in AML cells expressing wild type TP53, the AML cell line MOLM-13 was treated with various concentrations of the two drugs both alone and in combinations (nutlin-3; 0.5–10 μM, VPA; 50–1000 μM, nutlin-3:VPA; 1:100) over different time periods (24–72h, nutlin-3 only for the 24 last hours). Synergistic or additive effects were detected in three distinct viability assays; 3H-thymidine incorporation was used to examine effect on proliferation, WST-1 was used to determine number of metabolic active cells in culture, and DNA specific staining with Hoechst 33342 was used to determine apoptosis after drug-treatment. In addition, MOLM-13 cells treated with the combination showed super additive induction of p53 and target genes. The optimal combination and time period found in MOLM-13 cells were tested in 40 different primary AML samples using the three different viability assays. Toxicity of the combination treatment was tested in normal peripheral blood lymphocytes, while preliminary toxicity of nutlin-3 and VPA alone and in combination on healthy NOD/SCID IL2γnull mice permitted determination of treatment regime. We developed an optical imagable model for in vivo evaluation of the combinational therapy by injecting NOD/SCID IL2γnull mice with MOLM-13 cells transfected with a tetracycline activated luciferase expressing construct (termed L192). Bioluminescent imaging was performed using a TD-SAMI (Time-domain small animal molecular imager). The efficacy of the combinational therapy was tested in 20 NOD/SCID IL2γnull mice injected with MOLM-13 L192 cells and divided into four groups; control, VPA (50 mg/kg b.i.d), nutlin-3 (200 mg/kg b.i.d) and nutlin-3 + VPA (200 mg/kg + 50 mg/kg) b.i.d. Treatment was scheduled for three weeks. The combination of nutlin-3 and VPA significantly inhibited disease development after one week, as monitored by in vivo imaging. Limitations due to nutlin related toxicity prevented further evaluation of continuous combinational therapy after 14 days. However, all treatment groups showed a significant increase in survival compared to the control group, with the combination group demonstrating decreased leukaemic burden as visualized by optical imaging and longer mean average survival time. VPA effect on survival was also tested in a BNML rat leukemia model, in which VPA-treatment (170 mg/kg) resulted in significant longer mean survival compared to the control. Together, the results suggest combined targeting of MDM2 and HDACs as a promising therapeutic approach in AML. Future studies will apply the established bioluminescent MOLM-13 AML xenograft model for further evaluation of the combinational therapy, using a different dosing regimen and scheduling. In addition, we will evaluate combinations of differing classes of HDAC inhibitors and MDM2 antagonists.


Blood ◽  
2012 ◽  
Vol 120 (9) ◽  
pp. 1765-1773 ◽  
Author(s):  
Parvathi Ranganathan ◽  
Xueyan Yu ◽  
Caroline Na ◽  
Ramasamy Santhanam ◽  
Sharon Shacham ◽  
...  

AbstractChromosome maintenance protein 1 (CRM1) is a nuclear export receptor involved in the active transport of tumor suppressors (eg, p53 and nucleophosmin) whose function is altered in cancer because of increased expression and overactive transport. Blocking CRM1-mediated nuclear export of such proteins is a novel therapeutic strategy to restore tumor suppressor function. Orally bioavailable selective inhibitors of nuclear export (SINE) that irreversibly bind to CRM1 and block the function of this protein have been recently developed. Here we investigated the antileukemic activity of KPT-SINE (KPT-185 and KPT-276) in vitro and in vivo in acute myeloid leukemia (AML). KPT-185 displayed potent antiproliferative properties at submicromolar concentrations (IC50 values; 100-500nM), induced apoptosis (average 5-fold increase), cell-cycle arrest, and myeloid differentiation in AML cell lines and patient blasts. A strong down-regulation of the oncogene FLT3 after KPT treatment in both FLT3-ITD and wild-type cell lines was observed. Finally, using the FLT3-ITD–positive MV4-11 xenograft murine model, we show that treatment of mice with oral KPT-276 (analog of KPT-185 for in vivo studies) significantly prolongs survival of leukemic mice (P < .01). In summary, KPT-SINE are highly potent in vitro and in vivo in AML. The preclinical results reported here support clinical trials of KPT-SINE in AML.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 226-226 ◽  
Author(s):  
Miguel Gallardo ◽  
Hun Ju Lee ◽  
Xiaorui Zhang ◽  
Aziz Nazha ◽  
Laura R. Pageon ◽  
...  

Abstract In acute myeloid leukemia (AML), numerous genetic and epigenetic changes have been identified that result in loss of differentiation, apoptosis, and cell cycle arrest. In contrast, changes in protein expression are not as well characterized. To address these deficiencies, we performed RPPA analysis on 415 de novo AML patient samples and identified increased expression of heterogeneous nuclear ribonucleoprotein K (hnRNP K) as a predictor of poor outcome. These elevated hnRNP K levels were most predictive in AML patients who also harbored mutant NPM1 and wild type FLT3. While NPM1MUT/FLT3WT status typically confers favorable prognoses, increased hnRNP K expression negated this effect, as greater than 90% of NPM1MUT/FLT3WTindividuals with elevated hnRNP K expression died within 12 months. hnRNP K is a multifunctional protein that controls basic cellular functions through RNA, DNA, and protein-protein interactions (e.g.; p53) and whose expression is often altered in cancer. To examine the biological role of hnRNP K in vivo, we generated mouse models that either increase or decrease hnRNP K expression. Biallelic hnRNP K deletion results in embryonic lethality, while haploinsufficiency (hnRNP K+/-) results in a partial neonatal lethal phenotype. Surviving hnRNP K+/- mice have reduced survival and are more tumor prone than wild type mice (Fig. 1A and B). Analysis of hnRNP K+/- peripheral blood and bone marrow revealed significant hematologic neoplasms, including myeloid hyperplasias. The myeloid expansion appears to be a consequence of defects in proliferation (decreased p21) and differentiation (increased C/EBPβ and ε expression and activation of Stat3) pathways (Fig. 1C). In vitro studies using hnRNP K+/- hematopoietic stem cells (HSC) and mouse embryo fibroblast (MEFs) likewise revealed defects in differentiation and proliferation potential. HSC were used in burst formation unit erythroid colony assays (BFU-E). In these experiments, we observed a significant increase in the number of hnRNP K+/- cells and immature cells as compared to wild type BFU-E (Fig. 2A and B). RT-PCR analysis of BFU-Es revealed deregulation of p53/p21 and TGFβ- pathway genes (Fig. 2C). Similarly, hnRNP K+/- MEFs failed to properly activate the p53/p21 pathway following exposure to ionizing radiation (Fig. 2D). In contrast to diminished hnRNP K expression, overexpression results in activation of pro-growth and self-renewal pathway proteins in both humans and mice. RPPA analysis of AML patient samples that overexpress hnRNP K, as well as transient overexpression of hnRNP K in cell lines, results in increased expression of c-Myc. To directly examine the impact of hnRNP K overexpression in vivo, we generated transgenic mouse models. hnRNPK-transgenic mice express elevated levels of hnRNP K and are tumor prone. While it is tempting to classify hnRNP K as either an oncogene or a tumor suppressor, our haploinsufficiency and overexpression data seem to indicate that abnormal expression in either direction has a significant impact on tumor predisposition. Mechanistically, hnRNP Kappears to be an influential regulator involved in proliferation, self-renewal, and differential programs. The functional consequences reduced and overexpression of hnRNP K is currently under investigation. Given the clinical relationship between hnRNP K expression and NPM1 status, it is our next priority to evaluate the synergistic relationship between hnRNPK and NPM1 in regulating hematopoiesis and AML progression. To this end, we have taken two approaches. The first is to identify the biological relationship between these two proteins. To do this, we are attempting to generate double heterozygous NPM1+/- and hnRNP K+/- mice. However, thus far, double haploinsufficiency (NPM1+/-hnRNP K+/-) appears to result in a synthetic lethal phenotype. This surprising result highlights the unique in vivo relationship between these proteins. In the second approach, we have generated hnRNP K-transgenic (hnRNP KTG) mice on a mutant NPM1 (NPM1Mut) background. This hnRNPKTG: NPM1Mut mouse model specifically recapitulates the genetic alterations observed in the AML human patients who have reduction in survival. Together, these models will allow us to determine the relationship between these two proteins and examine how they impact leukemogenesis. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document