In vivo efficacy of the recombinant anti-CD64 immunotoxin H22(scFv)-ETA′ in a human acute myeloid leukemia xenograft tumor model

2011 ◽  
Vol 129 (5) ◽  
pp. 1277-1282 ◽  
Author(s):  
Mehmet Kemal Tur ◽  
Michael Huhn ◽  
Edgar Jost ◽  
Theo Thepen ◽  
Tim H. Brümmendorf ◽  
...  
2018 ◽  
Vol 38 (20) ◽  
Author(s):  
Dong-Mei Wu ◽  
Xin Wen ◽  
Xin-Rui Han ◽  
Shan Wang ◽  
Yong-Jian Wang ◽  
...  

ABSTRACT In the current study, we were interested in exploring the molecular mechanism of circular RNA DLEU2 (circRNA-DLEU2) (hsa_circ_0000488) and microRNA 496 (miR-496), as well as PRKACB, in human acute myeloid leukemia (AML) cell activities. The RNA expression levels of circRNA-DLEU2, hsa-miR-496, and PRKACB were assessed by quantitative real-time PCR (qRT-PCR). The proliferation and apoptosis abilities of the cells were determined by CCK8 assay and flow cytometry analysis. Target relationships between circRNA-DLEU2 and miR-496, as well as PRKACB, were analyzed by luciferase reporter assay and probe assay. Immunoblotting assays were used to detect the protein expression level of PRKACB. We also did in vivo experiments to observe tumor formation after overexpression of circRNA-DLEU2. Our data showed that circRNA-DLEU2 was upregulated in AML tissues and cells, which promoted AML cell proliferation and inhibited cell apoptosis. circRNA-DLEU2 promoted AML tumor formation in vivo. miR-496 was inhibited by circRNA-DLEU2 and was downregulated in AML tissues. circRNA-DLEU2 inhibited miR-496 expression and promoted PRKACB expression. miR-496 antagonized the effects of PRKACB on MOLM-13 cell proliferation and apoptosis. Collectively, circRNA-DLEU2 accelerated human AML by suppressing miR-496 and promoting PRKACB expression.


2012 ◽  
Vol 160 (2) ◽  
pp. 290-298 ◽  
Author(s):  
Kuan-Boone Tan ◽  
Leong-Uung Ling ◽  
Ralph M. Bunte ◽  
Wee-Joo Chng ◽  
Gigi N.C. Chiu

PLoS ONE ◽  
2013 ◽  
Vol 8 (4) ◽  
pp. e60680 ◽  
Author(s):  
Malte von Bonin ◽  
Martin Wermke ◽  
Kadriye Nehir Cosgun ◽  
Christian Thiede ◽  
Martin Bornhauser ◽  
...  

2016 ◽  
Vol 15 (10) ◽  
pp. 2422-2431 ◽  
Author(s):  
Urs B. Hagemann ◽  
Katrine Wickstroem ◽  
Ellen Wang ◽  
Adam O. Shea ◽  
Kristine Sponheim ◽  
...  

Blood ◽  
1996 ◽  
Vol 87 (11) ◽  
pp. 4754-4761 ◽  
Author(s):  
HJ Sutherland ◽  
A Blair ◽  
RW Zapf

Despite the usual uniform and primitive appearance of cells derived from the leukemic clone in most patients with acute myeloid leukemia (AML), there is considerable heterogeneity among leukemic blasts, particularly with respect to their capacity to proliferate and/or self renew. We have assessed whether these differences in proliferative potential are correlated with the phenotypic changes that characterize normal hematopoiesis, which might suggest an analogous hierarchy of AML progenitors. We have used the ability of primitive AML cells to persist or produce blast colony forming cells (CFU-blast) detected after 2 to 8 weeks in the presence of growth factors in suspension cultures (SC) termed SC-initiating cells (IC), or with stroma in long-term cultures (LTC-IC) as a quantitative assay for a cell that may have primitive characteristics. This SC assay is linear, cell concentration independent, and the frequency of SC-IC by limiting dilution analysis is lower than primary CFU-blast. The average output of CFU-blast after 2 to 8 weeks by individual SC-IC varied between 2 and more than 100 in individual patients. Leukemic blasts were sorted based on their expression of antigens previously found useful to characterize normal progenitor differentiation, and analyzed for the percentage of CFU- blast SC-IC, and leukemic LTC-IC within each fraction. All of these progenitor types were heterogeneous in their expression of CD45RA and CD33, but expressed uniformly low levels of CD15 and differed from normal primitive progenitors in their high expression of HLA-DR. CFU- blast had a significantly higher expression of CD71 and CD38 as compared with SC-IC or leukemic LTC-IC. In patients with CD34+ blasts, the majority of their SC-IC at 4 weeks were CD34+/CD38-; however, patients with CD34- blasts had at least some CD34- progenitors. These results show that while heterogeneity exists between patients, it is possible to physically separate subpopulations of AML cells with different proliferative potentials. It also provides some support for the concept that quantitation of leukemic cells capable of producing CFU-blast for 4 weeks or more in vitro measures a less frequent leukemic progenitor with higher proliferative potential that may be the only relevant cell for maintaining the leukemic clone in vivo.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1511-1511
Author(s):  
Ander Estella-Hermoso de Mendoza ◽  
Remedios Castello-Cros ◽  
Cristina Cirauqui ◽  
Raffaella Pippa ◽  
Isabel Moreno ◽  
...  

Abstract Abstract 1511 Protein phosphatase 2A (PP2A) is a human tumor suppressor that inhibits cellular transformation by regulating the activity of several signaling proteins critical for malignant cell behavior. Our group has shown that PP2A activity is reduced in acute myeloid leukemia (AML), and that restoration of PP2A activity in AML cells blocks proliferation, induces caspase-dependent apoptosis, and affects AKT and ERK1/2 activity. It has been proposed that FTY720 suppress PP2A activity by directly interacting with PP2AA-C dimers, and possibly interfering with the SET-PP2A interplay. Reactivation of PP2A by FTY720 in vivo restricts leukemogenesis in different types of hematological malignancies; however, its antitumor effects in AML remain underexplored. Here, we developed lipid nanoparticles (LN) encapsulating FTY720, and evaluated its pharmacokinetic profile and biodistribution in mice, and its antileukemic effects in AML in vitro and in vivo. FTY720-LN show suitable size (around 210 nm) and encapsulation efficiency (nearly 75%) for the oral and intravenous administration of the compound. Blood concentration of FTY720 decreased rapidly 2 hours after the intravenous administration of FTY720-LN; however, orally administered LN provided both a sustained release of the drug over four days and a high distribution in tissues, significantly increasing the accumulation of FTY720 in spleen, compared to the administration of the same dose of FTY720 solution and intravenous administration of FTY720-LN. In vitro studies revealed that FTY720 and FTY720-LN restore PP2A phosphatase activity in AML cells, leading to cell growth arrest, induction of caspase-dependent apoptosis and decrease in phosphorylation of PP2A targets AKT and ERK. Moreover, in vivo daily intraperitoneal administration of FTY720 in RAG2−/− γc−/− mice bearing an AML xenograft tumor reduced significant tumor growth. Of note, oral administration of FTY720-LN every three days was as efficient in reducing AML xenograft tumor growth as a daily oral administration of FTY720. These results provide the first evidence for the potential use of lipid nanoparticles encapsulating FTY720 as a therapeutic agent in AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3953-3953
Author(s):  
Scott Portwood ◽  
Deepika Lal ◽  
Christopher W Schaefer ◽  
Odochi Uwazurike ◽  
Joseph A Spernyak ◽  
...  

Abstract Background Tumor vascular disrupting agents (VDAs) represent a novel class of anti-cancer therapeutics with the unique ability to rapidly destroy established tumor vasculature and induce ischemic necrosis. In contrast to most VDAs which are derivatives of anti-tubulin agents, ASA404 (5,6-dimethylxanthenone-4-acetic acid, DMXAA) is an unique non-steroidal anti-inflammatory VDA which induces potent host immune responses and disrupts tumor vasculature in solid tumor mouse models. Methods The overall aim of these studies was to examine the effects of ASA404 treatment on tumor growth in preclinical human acute myeloid leukemia (HEL, HL60) and lymphoma (Raji) xenograft models. We specifically investigated the effects of drug treatment on host immune and inflammatory responses, marrow vasculature, and initial tumor engraftment. Disease progression in vivo was measured using small animal magnetic resonance imaging and bioluminescent imaging of luciferase-transfected human tumor cells. Host cytokine and chemokine levels in the plasma and other tissues (marrow, spleen, liver) were determined by multiplex flow cytometric assays. Results In local acute myeloid leukemia (HEL-luciferase) xenografts, a single dose of ASA404 (25 mg/kg) markedly enhanced tumor vascular permeability, as determined by magnetic resonance imaging at 4 hours. This was followed by vascular disruption and central necrosis at 24 hours, as determined by immunohistochemistry. In systemic leukemia xenografts, a single dose of ASA404 enhanced marrow vascular permeability and induced several-fold elevations in host chemokine levels (G-CSF, IL-6, MCP-1, KC) in circulating plasma as well as bone marrow and spleen cells (MIG, G-CSF, MIP-1alpha, IP-10). Administration of ASA404 4-24 hours prior to inoculation of human acute myeloid leukemia (HEL) or lymphoma (Raji) cells in immunodeficient mice delayed disease progression and prolonged overall survival. Continuous ASA404 treatment (20 mg/kg twice weekly) for up to three weeks in established systemic leukemia and lymphoma xenografts resulted in marrow vascular disruption and significant inhibition of tumor growth. Prolonged ASA404 treatment was associated with elevations in circulating host TNF-a levels and increased numbers of macrophages within the leukemic marrow microenvironment. Combination ASA404 and doxorubicin therapy further improved the overall survival of HEL and Raji-engrafted mice as compared with AS404 or doxorubicin alone. Quantification of doxorubicin levels by HPLC in local tumor xenografts and leukemic marrow samples revealed several-fold higher doxorubicin levels following ASA404 vs. vehicle (PBS) treatment, consistent with ASA404-mediated improvements in leukemia drug delivery. Preliminary data suggest a therapeutic benefit of combination ASA404 and cytarabine therapy in the same xenograft models. Conclusions Our results demonstrate that ASA404 exerts anti-tumor activity in human acute myeloid leukemia and lymphoma xenografts via myriad unique effects on the marrow microenvironment. Single dose administration of ASA404 results in rapid and marked alterations in multiple host cytokine/chemokine levels in association with enhanced tumor vascular permeability and impaired in vivo leukemia cell engraftment. Prolonged drug administration leads to pronounced vascular disrupting effects and inhibition of tumor growth. Combination regimens of ASA404 with anthracycline or cytarabine lead to further improvements in anti-tumor activity and overall survival in leukemia-bearing mice. These data suggest that ASA404/DMXAA may be worthy of further clinical investigation for the treatment of aggressive hematological malignancies. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document