Potent Effects Of The Vascular Disrupting Agent, ASA404 (DMXAA) On The Marrow Microenvironment In Preclinical Human Leukemia and Lymphoma Models

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.

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.


Blood ◽  
2011 ◽  
Vol 117 (12) ◽  
pp. 3294-3301 ◽  
Author(s):  
Mark Levis ◽  
Farhad Ravandi ◽  
Eunice S. Wang ◽  
Maria R. Baer ◽  
Alexander Perl ◽  
...  

AbstractIn a randomized trial of therapy for FMS-like tyrosine kinase-3 (FLT3) mutant acute myeloid leukemia in first relapse, 224 patients received chemotherapy alone or followed by 80 mg of the FLT3 inhibitor lestaurtinib twice daily. Endpoints included complete remission or complete remission with incomplete platelet recovery (CR/CRp), overall survival, safety, and tolerability. Correlative studies included pharmacokinetics and analysis of in vivo FLT3 inhibition. There were 29 patients with CR/CRp in the lestaurtinib arm and 23 in the control arm (26% vs 21%; P = .35), and no difference in overall survival between the 2 arms. There was evidence of toxicity in the lestaurtinib-treated patients, particularly those with plasma levels in excess of 20μM. In the lestaurtinib arm, FLT3 inhibition was highly correlated with remission rate, but target inhibition on day 15 was achieved in only 58% of patients receiving lestaurtinib. Given that such a small proportion of patients on this trial achieved sustained FLT3 inhibition in vivo, any conclusions regarding the efficacy of combining FLT3 inhibition with chemotherapy are limited. Overall, lestaurtinib treatment after chemotherapy did not increase response rates or prolong survival of patients with FLT3 mutant acute myeloid leukemia in first relapse. This study is registered at www.clinicaltrials.gov as #NCT00079482.


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

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3611-3611
Author(s):  
Scott Portwood ◽  
Deepika Lal ◽  
Yung-Chun Hsu ◽  
Rodrigo Vargas ◽  
Meir Wetzler ◽  
...  

Abstract Abstract 3611 Recent evidence has demonstrated the bone marrow (BM) microenvironment, the principal site of acute myeloid leukemia (AML) initiation and expansion, is characterized by intrinsically low oxygen tension. Theoretically, such microenvironmental changes may lead to the selective outgrowth of AML clones which are “better adapted” to survive within a severely hypoxic microenvironment and/or may confer resistance to chemotherapeutic agents, similar to solid tumor cells. We report here that human AML cells (HL60, ML-2) cultured under chronic hypoxic conditions mimicking the marrow microenvironment (1% O2, 72 hours) exhibited reduced sensitivity to cytarabine-induced apoptosis as compared with normoxic cells, as determined by flow cytometric analysis, western blot analysis, and cell viability assays. Similar results were noted in primary AML samples treated with cytarabine under normoxic and hypoxic conditions in colony formation assays (n=3 samples, p=0.01). In order to improve upon chemotherapy outcomes, we investigated the effects of TH-302, a hypoxia-activated bromo-isophosphoramidate mustard prodrug, which is currently undergoing clinical trial evaluation in multiple tumor types. Treatment of AML cell lines (HL60, HEL) and primary AML samples with TH-302 (at doses ranging from 0.1– 5 mM, p values ranging from <0.05–0.0001) resulted in dose- and hypoxic-dependent inhibition of AML proliferation and apoptosis. In vivo TH-302 treatment significantly decreased disease burden, as measured by total animal bioluminescence, and prolonged overall survival in two systemic human AML xenograft models (HEL-luciferase, HL60-luciferase) (Figure 1). Immunohistochemical studies demonstrated that TH-302 treatment reduced numbers of hypoxic (pimonidazole-positive) cells within the leukemic marrow microenvironment. Because prior data in animal models has shown that AML progression within the marrow is associated with expansion of hypoxic BM areas, we examined the effects of TH-302 treatment on systemic AML growth when initiated early (prior to AML inoculation) or late (several days following AML engraftment) in the disease process. TH-302 was equally effective at both time points. Although anti-vascular therapy has been shown to enhance tumor hypoxia in other cancer types, we noted no synergistic or additive in vivo effects when TH-302 therapy was combined with sorafenib, an inhibitor of vascular endothelial growth factor receptors (VEGFR), in our models. TH-302 therapy administered for two weeks in non-leukemic and leukemia-engrafted mice was not associated with hematologic toxicities. In summary, our results demonstrate the anti-leukemic activity of TH-302 in preclinical AML models and suggest that the efficacy of this and other drugs for AML therapy may be uniquely affected by the BM microenvironment. Further clinical development of TH-302 and other hypoxia-targeted drugs for AML therapy are warranted. Based on our data, higher TH-302 doses and/or chronic drug administration may be needed for optimal in vivo anti-leukemic activity. Figure 1. Effects of TH-302 treatment on systemic AML growth and overall survival in HL60-luciferase engrafted SCID mice. Figure 1. Effects of TH-302 treatment on systemic AML growth and overall survival in HL60-luciferase engrafted SCID mice. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 8 ◽  
Author(s):  
Huiqing Qu ◽  
Ye Zhu

Background: Acute myeloid leukemia (AML), characterized by the low cure rate and high relapse, urgently needs novel diagnostic or prognostic biomarkers and potential therapeutic targets. Sphingomyelin Phosphodiesterase Acid Like 3B (SMPDL3B) is a negative regulator of Toll-like receptor signaling that plays important roles in the interface of membrane biology and innate immunity. However, the potential role of SMPDL3B in human cancer, especially in AML, is still unknown.Methods: The expression of SMPDL3B in AML samples was investigated through data collected from Gene Expression Omnibus (GEO). Association between SMPDL3B expression and clinicopathologic characteristics was analyzed with the chi-square test. Survival curves were calculated by the Kaplan–Meier method. Cox univariate and multivariate analyses were used to detect risk factors for overall survival. The biological functions of SMPDL3B in human AML were investigated both in vitro and in vivo.Results: Expression of SMPDL3B mRNA was significantly upregulated in human AML samples and closely correlated to cytogenetics risk and karyotypes. Elevated expression of SMPDL3B was associated with poor overall survival and emerged as an independent predictor for poor overall survival in human AML. Blocked SMPDL3B expression inhibited AML cells growth both in vitro and in vivo via promoting cell apoptosis.Conclusion: Taken together, our results demonstrate that SMPDL3B could be used as an efficient prognostic biomarker and represent a potential therapeutic target for human AML.


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. 1515-1515
Author(s):  
Hongyan Tong ◽  
Kongfei Li ◽  
Chen Mei ◽  
Chao Hu ◽  
Jie Jin

Abstract Abstract 1515 Background: Decitabine is a prototype for epigenetic therapy in cancer which targets DNA methyltransferase. While it was known that decitabine monotherapy has been associated with a relative low rate of complete remission rates in acute myeloid leukemia and myelodysplastic syndrome. Several groups have attempted to increase the response rate with decitabine-based therapy by developing combinations. In the preliminary experiments, we investigated the effect of anti-leukemia drugs (idarubicin, daunorubicin, cytarabine, thalidomide and homoharringtonine) in combination simultaneously or sequentially with decitabine of inhibiting proliferation of acute myeloid leukemia cell lines. The result showed that sequential combination of decitabine and idarubicin induced a significant synergistic effect in cell apoptosis, inhibition of cell proliferation. With the gene chip technology, we found that the gene expression of Wnt pathway changed obviously after the treatment of DAC combined with IDA. Further we confirm that sequential combination of DAC and IDA caused depression of wnt pathway in vitro. In this study, we evaluate the anti-leukemic effect and the combination mechanism of DAC in sequential combination with IDA in vivo. Methods: The AML cell line U937 (14×107 cells per animal) were injected subcutaneously into the right flank of 6 to 8-week old NOD-SCID mice. After tumor growth, decitabine(0.5mg/kg/day) were injected intraperitoneal of a five consecutive days followed by a three days of idarubicin(0.5 mg/kg/day). The tumor growth inhibition effect was evaluated by microPET to reveal the synergistic effect of decitabine and idarubicin. Using TUNEL method and electron microscopy, we detected the apoptosis of leukemic cells from tumor bearing mice. The expression of β-catenin which is the key gene of Wnt/β-catenin pathway was also examined by immunochemistry. We then detected the expression of protein levels of the other genes(cyclinD1, c-myc, SFRP1,HDPR1 and DKK3) of Wnt/β-catenin pathway in tumor cells of tumor-bearing mice by western blot. Results: In vivo, the effect of sequential combination of decitabine and idarubicin was initially examined in a subcutaneous AML mouse model. The AML cell line U937(1×107 cell per animal) was transplanted subcutaneously into the right flank of 2-week-old female NOD-SCID mouse. The cell line developed into a rapidly growing tumor. Treatment was initiated in 7th days after injection of the leukemic cells, included with 5 days of decitabine alone, 3 days of idarubicin alone and 5 days of decitabine followed by 3 days of idarubicin. A significant inhibition of tumor growth was demonstrated with microPET in animals treated with sequential combination of decitabine and idarubicin compared with the other treatment and control group animals. The apoptosis of tumor cells was also significantly increased in the sequential combination treatment group detected by TUNEL and electron microscope. We also found that the sequential treatment group induced synergistic effect in re-expression or up-expression of the Wnt inhibitor genes (SFRP1, HDPR1 and DKK3) in tumor cells. Furthermore, the down-steam genes of wnt pathway including β-catenin, c-myc and cyclinD1 were down-regulated which suggest depression of wnt/β-catenin pathway. These results suggest that sequential combination of decitabine and idarubicin has a powerful anti-leukemic effect not only in vitro but also in vivo. Conclusion: Our results demonstrate decitabine in sequential combination with idarubicin in mouse model of AML and suggest that this combination may be of clinical value in the treatment of patients with AML. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document