Share this post on:

reatment Reduces NOTCH1Mutated T-ALL LIC Survival The relative leukemic regenerative potential of NOTCH1Mutated, and NOTCH1WT samples was determined in serial transplantation studies. FACS analysis of cells from bone marrow, spleen and thymus showed that while the levels of thymic engraftment were equivalent, NOTCH1Mutated T-ALL LIC gave rise to a significantly higher CD34+ leukemic burden in the marrow and spleen of primary transplant recipients than NOTCH1WT T-ALL samples. Hence, we sought to determine whether selective NOTCH1 inhibition could reduce LIC burden, NOTCH1Mutated T-ALL LIC survival is dependent on activated NOTCH1 receptor signaling, and selective NOTCH1 inhibition could spare NOTCH1WT or normal cord blood CD34+ progenitors in engrafted mice. For these purposes, NOTCH1Mutated T-ALL LIC-engrafted mice were treated with a selective NOTCH1-NRR/Fc mAb that specifically inhibits NOTCH1 receptor signaling. Animals were treated with hN1 mAb or a control mouse IgG1 mAb every 4 days for 3 weeks, and over this time period both antibodies were well-tolerated in treated animals NOTCH1 Inhibition in T-ALL Initiating Cells . As anticipated, treatment with the hN1 mAb had no detectable toxicity or deleterious effects on survival in mice, as this antibody does not bind to PubMed ID:http://www.ncbi.nlm.nih.gov/pubmed/22205030 endogenous murine NOTCH1 and is expected to only target activity of human NOTCH1 in transplanted human cells. Following hN1 mAb treatment of NOTCH1Mutated T-ALL LIC-transplanted mice, FACS analysis revealed a significant reduction in leukemic CD34+ cell burden in both the marrow and spleen of hN1 mAb-treated mice. Levels of CD34+ cell burden in the thymus were similar in both Gynostemma Extract biological activity groups, which is likely a result of relatively lower engraftment rates in this hematopoietic organ. Notably, LIC from one T-ALL NOTCH1Mutated patient sample, with a PTEN frame-shift mutation, retained sensitivity to hN1 inhibition. While survival of LIC from a sample with both PTEN and PIK3R1 mutations was not significantly inhibited in the bone marrow, LIC burden was significantly reduced in the spleen by hN1 mAb treatment, highlighting the influence of additional mutations and microenvironmental context in responses to selective NOTCH1 inhibitory strategies. Although engraftment rates of normal human cord blood progenitors were low, the survival of normal CD34+ hematopoietic progenitors was not significantly reduced by targeted NOTCH1 inhibition. These results suggest a greater functional dependence of NOTCH1Mutated T-ALL LIC on NOTCH1 signaling in selective hematopoietic niches compared to NOTCH1WT progenitors and normal hematopoietic stem cells. Following hN1 mAb treatment, immunohistochemical analyses revealed a marked increase in levels of activated caspase 3, and a concomitant reduction in levels of NOTCH1 in NOTCH1Mutated T-ALL LIC-engrafted bone marrow compared with control IgG1 mAb-treated control bone marrow. To assess whether hN1 mAb treatment could inhibit the generation of transcriptionally active NOTCH1, which may be involved in promoting therapeutic resistance through induction of self-renewal, ICN1 immunohistochemical analysis was performed on bone marrow derived from the NOTCH1Mutated LIC-engrafted mice after NOTCH1 Inhibition in T-ALL Initiating Cells treatment with hN1 mAb or IgG1 control mAb. Treatment with the hN1 mAb was associated with a reduction in bone marrow ICN1 levels. These data corroborate that the antibody’s mechanism of action involves both interference with ligand

Share this post on:

Author: HMTase- hmtase