Network-based analysis with primary cells reveals drug response landscape of acute myeloid leukemia

2020 ◽  
Vol 393 (1) ◽  
pp. 112054
Author(s):  
Cheng Chen ◽  
Li Wang ◽  
Lili Li ◽  
Aoli Wang ◽  
Tao Huang ◽  
...  
Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2024-2024
Author(s):  
Michael Heuser ◽  
Luzie U. Wingen ◽  
Doris Steinemann ◽  
Gunnar Cario ◽  
Nils von Neuhoff ◽  
...  

Abstract Resistance to induction chemotherapy is of independent prognostic value in acute myeloid leukemia (AML). DNA microarrays were used to determine the gene-expression profile of AML blasts in 38 patients with good or poor response to induction chemotherapy. We selected an 11-sample training set, applying prediction analysis of microarrays (PAM) to devise a drug-response predictor which was tested on the remaining 27 samples and an independent set of samples recently published (Bullinger et al. 2004). Our drug-response predictor with 46 clones divided the 27 samples into two prognostic subgroups, the poor response group having a significantly shorter overall survival (P= .021). A subset of these 46 clones was sufficient to divide the published 62-sample test-set with intermediate risk cytogenetics into prognostically relevant subgroups (P= .028), adding prognostic information to that of known risk factors in multivariate analysis (hazard ratio, 2.8; 95 percent confidence interval, 1.4 to 5.8; P= .005). Thirteen of 39 drug resistance-enriched genes are known to be upregulated in hematopoietic stem/progenitor cells, and the expression pattern in normal CD34+ cells is highly correlated to the drug-resistance signature. This suggests that drug resistant AMLs show molecular features of hematopoietic stem/progenitor cells and can be identified by a characteristic gene-expression profile.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3624-3624
Author(s):  
Weihsu Claire Chen ◽  
Andreea C. Popescu ◽  
Yan Xing ◽  
Gitte Gerhard ◽  
Julie S. Yuan ◽  
...  

Abstract Abstract 3624 Small molecule inhibitors targeting somatic mutations of Janus kinase 2 (JAK2) have demonstrated effectiveness in clinical trials for treatment of myeloproliferative disorders. Activated JAK2 signaling has been reported in some acute myeloid leukemia (AML) samples even though the frequency of JAK2 mutations is relatively rare in AML. Whether JAK2 inhibitors are effective in AML is not clear. Since many myeloid malignancies including AML are organized as cellular hierarchies driven by leukemia stem cells (LSC) at the apex, it is also unknown whether LSCs are sensitive to JAK2 inhibition. We report that SAR302503 (SAR503, Sanofi, Cambridge, MA), an orally administered small molecule inhibitor of JAK2, shows therapeutic efficacy in a xenograft model of human AML established by intrafemoral injection of primary human AML cells into anti-NK treated irradiated NOD.SCID mice. Drug pharmacokinetic studies confirm that SAR503 exhibits good bioavailability in NOD.SCID mice. Starting 2 weeks post transplantation to permit establishment of an AML graft, mice were orally gavaged twice a day with 60 mg/kg SAR503 or vehicle alone (0.5% methylcellulose) for 14 consecutive days. In 5 of 7 AML samples, treated mice exhibited significantly lower engraftment (3 to 18 fold; p < 0.05) in the injected femur compared to control mice. For 4 samples, there was also a significantly reduced level of engraftment (2 to 19 fold; p < 0.05) in non-injected bones, indicating that JAK2 inhibition affected the migratory ability of AML cells. The observed heterogeneous drug response (5 responders and 2 non-responders) correlated well with phosphoflow analysis showing that AML samples that responded to JAK2 inhibition in vivo had high basal and marked reduction in the STAT signaling pathway after in vitro treatment, whereas non-responding samples did not. Serial transplantation studies are ongoing to evaluate the effect of JAK2 inhibition on LSCs in treated primary mice. To evaluate whether AML samples that did not respond to JAK2 inhibition alone would respond to combination therapy, we treated engrafted mice with SAR503 plus cytarabine, a standard chemotherapeutic drug used in AML. Combination therapy of one non-responding sample resulted in a significantly reduced leukemic burden (2.3%) compared to mice treated with SAR503 alone (85.8%) or cytarabine alone (16.8%; p < 0.05 versus combination therapy). Our results demonstrate the potential of SAR503 to target AML cells and AML LSCs across a cross section of primary AML samples. Our pilot studies warrant a much larger scale evaluation of AML samples to identify responders and non-responders along with associated proteomic and genomic biomarkers of drug response. The approach we have taken, which focuses on large-scale analysis of primary samples using state-of-the-art xenograft assays, offers a new paradigm for preclinical drug development to identify both novel agents that effectively target LSCs and the patient populations most likely to benefit from targeted treatment. Disclosures: Off Label Use: We describe using SAR302503 to treat AML in a mouse model.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3612-3612 ◽  
Author(s):  
Kaikai Huang ◽  
Bingjie Ding ◽  
Qingxiu Zhong ◽  
Xuejie Jiang ◽  
Xiaodong Li ◽  
...  

Abstract Hh/IGF-1R/PI3K/Akt/MRP1 pathway induce refractory acute myeloid leukemia Background: Although a great progress has made in the treatment of acute myeloid leukemia(AML), chemoresistance and relapse due to multidrug resistance (MDR) remains a therapeutic challenge. We have proven that LBH589 combined with Bortezomib had synergistic effects on proliferation, apoptosis and sensitivity to cytotoxic drugs via AKT and NF-κB pathways in chemoresistant HL60/ADR cells and refractory AML primary cells. Using gene chip analysis, we found that among those disregulated pathways, Hedgehog(Hh) signaling pathway was probably the upstream paths which could regulate others. Aims: The purpose of the study was to investigate the expression of Hh pathway in AML including primary cells and cell lines, analyze the relationship between Hh expression and clinical prognosis. Also investigate the biological effect of Hh pathway inhibitor NVP-LDE225 in reversing drug-resistance in HL60/adriamycin-resistant(ADR) cells and refractory AML primary cells and the underlying mechanisms. Besides, animal experiments were done to verify the results in vitro. Methods: Western blot assay were used to determine the protein expression of Ptch–Smo–Gli-1–Shh in HL60 and HL60/ADR cell lines and refractory or non-refractory primary AML cells. Kaplan–Meier curves were used to estimate Relapse-free survival(RFS) and Over-all survival(OS). HL60/ADR cells and refractory primary cells were treated with adriamycin or combined with NVP-LDE225 concentration among 0-10µM). Proliferation were evaluated by 3-(4,5)-dimethylthiahiazo (-z-yl)-3,5-di-phenytetrazoliumromide (MTT) assay and cell apoptosis were analyzed by Annexin V-FITC/PI staining through flow-cytometry(FCM). Intercellular adriamycin accumulation(MFI) were analyzed by FCM. The changes in protein levels of Gli-1–IGF-1R–p-IGF-1R–IRS-1–Akt–p-Akt–MRP-1–Bcl-2 were detected by Western blot. Besides, we used a nude mouse xenograft model to verify the anti-proliferative effects in vivo. Results: We found that the refractory primary AML cells and HL60/ADR cells correlated with higher activation of the Hh pathway, however in non-refractory primary cells or chemosensitive cell lines HL60, such activation was less pronounced. Higher protein expression of this pathway was related to higher recurrence rate and associated with poor relapse free survival (RFS) and poor overall survival (OS)(P=0.002)(a). NVP-LDE225, a potent and selective Hh inhibitor, significantly reverted resistance of adriamycin(reversal fold was 3.75), induced cell apoptosis(P=0.007), increased the intracellular adriamycin accumulation(P=0.000), decreased protein expression of p-IGF-1R, IRS-1, p-Akt, Bcl-2, MRP1 also Gli-1(b). These effects were likely to be mediated via inhibition of IGF-1R/PI3K/Akt/MRP1 pathway. Besides, in a AML xenograft mouse model, NVP-LDE225 combined with ADM resulted in significant tumor regression(P=0.002) (c). Conclusions: These findings provided evidence that targeting the Hh pathway might be a therapeutic avenue for overcoming MDR resistance in myeloid leukemia. Keywords: Hedgehog signaling, acute myeloid leukemia, IGF-1R/PI3K/Akt signaling Disclosures No relevant conflicts of interest to declare.


Cancer Cell ◽  
2017 ◽  
Vol 32 (3) ◽  
pp. 324-341.e6 ◽  
Author(s):  
Diana Passaro ◽  
Alessandro Di Tullio ◽  
Ander Abarrategi ◽  
Kevin Rouault-Pierre ◽  
Katie Foster ◽  
...  

2020 ◽  
Author(s):  
Valentina Gaidano ◽  
Mohammad Houshmand ◽  
Nicoletta Vitale ◽  
Giovanna Carrà ◽  
Alessandro Morotti ◽  
...  

AbstractBackgroundDihydroorotate Dehydrogenase (DHODH) is a key enzyme of the de novo pyrimidine biosynthesis, whose inhibition was recently found to induce differentiation and apoptosis in acute myeloid leukemia (AML). DHODH inhibitors were previously investigated in solid tumors, where they showed promising antiproliferative activity, both in vitro and in vivo. However, their effectiveness was not confirmed in clinical trials, probably due to the pyrimidine salvage pathway that cancer cells could exploit to survive. In this study we investigated the pro-apoptotic activity of MEDS433, the DHODH inhibitor developed by our group, against AML. Learning from previous failures, we challenged our model mimicking in vivo conditions, and looked for synergic combination to boost apoptosis.MethodsWe evaluated the apoptotic rate of multiple AML cell lines and AML primary cells treated with MEDS433 or other DHODH inhibitors, alone and in combination with classical antileukemic drugs or with dipyridamole, a blocker of the pyrimidine salvage pathway. Experiments were also performed mimicking in vivo conditions, i.e., in the presence of physiological uridine plasma levels (5 μM).ResultsMEDS433 showed a strong apoptotic effect against multiple AML cell lines, which was at least partially independent from the differentiation process. Its combination with classical antileukemic agents resulted in a further increase of the apoptotic rate. However, when MEDS433 was tested in the presence of 5 μM uridine and/or in primary AML cells, results were less impressive. On the contrary, the combination of MEDS433 with dipyridamole resulted in an outstanding synergistic effect, with a dramatic increase of the apoptotic rate both in AML cell lines and AML primary cells, which was unaffected by physiological uridine concentrations. Preliminary analyses show that the toxicity of this treatment should be limited to proliferating cells.ConclusionsThe combination of a DHODH inhibitor and dipyridamole is characterized by differentiating and pro-apoptotic features and induces metabolic lethality on a wide variety of AMLs with different genetic backgrounds.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2662-2662 ◽  
Author(s):  
Diana Passaro ◽  
Alessandro Di Tullio ◽  
Ander Abarrategi ◽  
Kevin Rouault-Pierre ◽  
Katie Foster ◽  
...  

Abstract The biological and clinical behavior of hematological malignancies are not only determined by the properties of the leukemic cells themselves, but are also highly affected by interaction with the microenvironment, pointing to the existence of an active crosstalk between the two compartments. Previous studies showed that acute myeloid leukemia (AML) cells actively modify endothelial cells ex vivovia several pathways, mainly mediated by VEGF. However, as anti-VEGF therapies haven't produced successful results in clinical trials, an extensive study of the crosstalk between AML and the vascular niche in the bone marrow (BM) is required to provide new therapeutic strategies. In the present study we combined the use of mouse models of AML, human AML patient-derived xenografts (PDX) and direct analysis on patient-derived BM biopsies to provide a global, reliable picture of the bone marrow vasculature in AML disease. We found several abnormalities in the vascular architecture and function in PDX, such as increased number of endothelial cells, increased microvascular density (MVD), decreased vascular mean diameter and increased hypoxia. Furthermore, using two-photon confocal intravital imaging we witnessed increased vascular permeability upon AML engraftment, observed homogeneously among different PDX. Interestingly, induction chemotherapy failed to normalize the vascular permeability in the BM, despite significant reduction in AML engraftment. We identified increased nitric oxide (NO) as a major mediator of the AML-induced vascular leakiness in the BM. Increased levels of NO and activated NOS3 were found in PDX and in an independent cohort of patient-derived BM biopsies. Strikingly, inhibition of NO production using genetic and pharmacological approaches reduced the vascular permeability, potentiated the normal HSC function and significantly improved treatment response in PDX. These results strongly support the notion of a primary function of the vascular permeability in AML progression, drug response and in affecting normal stem cell function, and they call for clinical trials incorporating NOS inhibitors during the remission phase to target the abnormal vascular niche and improve the treatment response. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 35 (5) ◽  
Author(s):  
Iryna Kolosenko ◽  
Oksana Goroshchuk ◽  
Linda Vidarsdottir ◽  
Ann‐Charlotte Björklund ◽  
Steven F. Dowdy ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1616-1616
Author(s):  
Martin Carroll ◽  
Tae Kon Kim ◽  
Kenichi Higashino ◽  
Alan M. Gewirtz

Abstract Signal transducer and activator of transcription (STAT) family proteins play crucial roles in the cytokine signaling pathways which regulate survival and proliferation of normal hematopoietic cells. However, the role of STAT proteins in regulating survival in leukemia remains poorly defined. STAT3 and STAT5, for example, have been reported to be constitutively activated in acute myeloid leukemia (AML) cells, however the physiologic significance of this activation is unknown. In order to better understand the role of STAT3 and STAT5 in AML biology, we studied their expression, activation, and requirement for cell growth in several AML cell lines and primary AML cells collected from patients at the University of Pennsylvania Cancer Center. We first confirmed the activation of STAT3 and STAT5 in primary AML cells by western blotting. An analysis of AML patient samples revealed elevated levels of constitutive STAT3 phosphorylation in 6 of 7 patient samples and constitutive STAT5 phosphorylation in 8 of 9 patient samples. In addition, 6 AML cell lines (K562, HL-60, MOLM-14, U937, KG-1, NB4) displayed constitutive STAT3 and STAT5 activation. In order to evaluate the functional significance of constitutive activation of STAT3 and STAT5 in AML cells, we designed and synthesized short interfering RNAs (siRNAs) to silence the expression of these proteins. For initial characterization, the siRNAs were delivered to MOLM-14 cells using an AMAXA nucleofector device (AMAXA, Inc. Gaithersburg, MD)(Program O-17/Solution V). Nucleofected siRNA diminished STAT3 expression by 85% at 24 hour but had little effect on cell proliferation (13%±3% decrease at 24 hour, 8%±4% at 48 hour, 5%±1% at 72 hour) compared to control siRNA treated cells. In contrast, STAT5 siRNA decreased STAT5 expression 80% at 24 hour compared to control treated cells but inhibited cell proliferation by 19%±1% at 24 hour, 22%±1% at 48 hour, 16%±3% at 72 hour in comparison to control siRNA treated cells suggesting a more important role for STAT5 in regulating cell proliferation. To study the effect of these siRNA molecules in primary AML cells, we first determined our ability to nucleofect primary cells by examining delivery efficiency of fluorescein labeled siRNA. Four different patient samples were evaluated and the mean ± SD of cells successfully transfected was 52%±12. Transfection of multiple AML patient samples with STAT3 siRNA decreased STAT3 expression but led to only modest decrease (6–18% at 48 hour, 7–36% at 72 hour) in AML cell survival. However, transfection of cells with STAT5 siRNA, but not control siRNA, led to a consistent decrease (25–54% at 48 hour, 21–60% at 72 hour) in AML cell survival. The decrease in survival was proportional to the transfection efficiency in the different samples. These results provide the first evidence that STAT5 expression and activation is necessary for the survival of primary AML cells. In fact, the data suggests a greater role of STAT5 in survival of primary cells than in survival of AML cell lines. Accordingly, STAT5 appears to be a legitimate target for the treatment of AML.


Sign in / Sign up

Export Citation Format

Share Document