AML-367: IMGN632, a CD123-Targeting ADC Bearing a DNA-Alkylating IGN Payload, Combines Effectively as a Triplet Regimen with Azacitidine and Venetoclax In Vivo, Prolonging Survival in Preclinical Models of Human Acute Myeloid Leukemia (AML)

2020 ◽  
Vol 20 ◽  
pp. S208-S209
Author(s):  
Vinitha Kuruvilla ◽  
Rebecca McCarthy ◽  
Qi Zhang ◽  
Krystal Watkins ◽  
Sharlene Adams ◽  
...  
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.


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

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 ◽  
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.


2010 ◽  
Vol 9 (10) ◽  
pp. 2737-2751 ◽  
Author(s):  
Deepika Lal ◽  
Jennifer A. Park ◽  
Kellie Demock ◽  
Joseph Marinaro ◽  
Amanda M. Perez ◽  
...  

2005 ◽  
Vol 79 (20) ◽  
pp. 13190-13194 ◽  
Author(s):  
Lucas Chan ◽  
Darren Nesbeth ◽  
Taylor MacKey ◽  
Joanna Galea-Lauri ◽  
Joop Gäken ◽  
...  

ABSTRACT Nonviral producer cell proteins incorporated into retroviral vector surfaces profoundly influence infectivity and in vivo half-life. We report the purification and concentration of lentiviral vectors using these surface proteins as an efficient gene transduction strategy. Biotinylation of these proteins and streptavidin paramagnetic particle concentration enhances titer 400- to 2,500-fold (to 109 CFU/ml for vesicular stomatitis virus G protein and 5 × 108 for amphotropic murine leukemia virus envelope). This method also uses newly introduced membrane proteins (B7.1 and ΔLNGFR) directed to lentiviral surfaces, allowing up to 17,000-fold concentrations. Particle conjugation of lentivirus allows facile manipulation in vitro, resulting in the transduction of 48 to 94% of human acute myeloid leukemia blasts.


Sign in / Sign up

Export Citation Format

Share Document