Troglitazone Inhibits Cell Growth and Induces Apoptosis of B-Cell Acute Lymphoblastic Leukemia Cells with t(14;18)

2006 ◽  
Vol 116 (1) ◽  
pp. 30-40 ◽  
Author(s):  
M. Takenokuchi ◽  
K. Saigo ◽  
Y. Nakamachi ◽  
S. Kawano ◽  
M. Hashimoto ◽  
...  
Blood ◽  
1981 ◽  
Vol 58 (2) ◽  
pp. 330-332 ◽  
Author(s):  
GH Reaman ◽  
J Blatt ◽  
DG Poplack

Abstract Activities of enzymes of the purine metabolic pathway, adenosine deaminase (ADA), purine nucleoside phosphorylase (PNP), and 5′- nucleotidase (5′-N), were investigated in the lymphoblasts of a patient with B-cell acute lymphoblastic leukemia. These lymphoblasts exhibited increased ADA activity and diminished activities of both PNP and 5′N' as compared to normal lymphocytes as well as non-T, non-B leukemia cells. This enzymatic pattern is identical to that which has been described in T-cell leukemic lymphoblasts and differs from that which has been observed in the malignant cells of undifferentiated B-cell lymphomas. These data suggest that there is biochemical heterogeneity within the spectrum of B-cell malignancies. Furthermore, inhibitors of ADA may be of use in those B-cell lymphoid neoplasms that exhibit increased ADA activity.


2015 ◽  
Vol 112 (27) ◽  
pp. E3455-E3455 ◽  
Author(s):  
Karel Fišer ◽  
Lucie Slámová ◽  
Jean-Pierre Bourquin ◽  
Jan Trka ◽  
Jan Starý ◽  
...  

Blood ◽  
1981 ◽  
Vol 58 (2) ◽  
pp. 330-332
Author(s):  
GH Reaman ◽  
J Blatt ◽  
DG Poplack

Activities of enzymes of the purine metabolic pathway, adenosine deaminase (ADA), purine nucleoside phosphorylase (PNP), and 5′- nucleotidase (5′-N), were investigated in the lymphoblasts of a patient with B-cell acute lymphoblastic leukemia. These lymphoblasts exhibited increased ADA activity and diminished activities of both PNP and 5′N' as compared to normal lymphocytes as well as non-T, non-B leukemia cells. This enzymatic pattern is identical to that which has been described in T-cell leukemic lymphoblasts and differs from that which has been observed in the malignant cells of undifferentiated B-cell lymphomas. These data suggest that there is biochemical heterogeneity within the spectrum of B-cell malignancies. Furthermore, inhibitors of ADA may be of use in those B-cell lymphoid neoplasms that exhibit increased ADA activity.


Life Sciences ◽  
2019 ◽  
Vol 227 ◽  
pp. 153-165 ◽  
Author(s):  
Xiaoguang Xu ◽  
Lei Huang ◽  
Zilu Zhang ◽  
Jia Tong ◽  
Jianqing Mi ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 971-971
Author(s):  
Sakiko Yoshida ◽  
Emily Tuscano ◽  
Connie Duong ◽  
Joseph Tuscano ◽  
Noriko Satake

Abstract Precursor B cell acute lymphoblastic leukemia (preB ALL) is the most common childhood cancer, as well as the leading cause of childhood cancer-related mortality. Despite overall progress in treatment, certain types of patients with preB ALL have a dismal prognosis with an overall survival of 30%. In addition, current approaches predispose these young patients to late effects, including secondary malignancies. Therefore, more efficacious and less toxic approaches are needed. Targeted therapy for leukemia has the potential to reduce off-target effects, thus minimizing toxicity and late effects and improving efficacy. Monoclonal antibodies (mAbs) have proven utility in leukemia therapy based on their ability to specifically target the leukemic clone and minimize off target effects. However, mAbs are generally not adequate as single agents because of limited efficacy. Antibody drug conjugates (ADCs) provide a method to deliver a potent toxin to the interior of antigen-positive tumor cells. CD22 is an ideal target for ADC-mediated therapeutics for B-cell malignancies because 1) there is high CD22 expression (more than 90%) in B-cell type ALL and 2) CD22 undergoes rapid internalization upon mAb binding. In this study, we evaluated the anti-CD22 (aCD22) mAb as a vehicle for the targeted delivery of Monomethyl Auristatin E (mMAE), a derivative of the cytotoxic tubulin modifier auristatin E. Figure 1 Treatments and outcome of the animals Figure 1. Treatments and outcome of the animals First, we assessed the in vitro cytotoxicities of the aCD22 mAb-mMAE in preB ALL cell lines Reh and JM1. MTS assay showed that IC50 doses of aCD22 mAb-mMAE were 0.7nM and 1nM in Reh and JM1, respectively. Next, we assessed in vivo therapeutic efficacy of the aCD22 mAb-mMAE in a pre-clinical xenograft animal model of preB ALL, using a primary leukemia sample which was confirmed to be CD22 positive. Age matched female NOD/SCID/IL2Rg-/- (NSG) mice were randomly assigned to 4 treatment groups (n=8 per group): 1) PBS, 2) free mMAE (0.165mg/kg), 3) free aCD22 mAb (7.335mg/kg), and 4) aCD22 mAb-mMAE conjugate (7.5mg/kg). The dose of free aCD22 mAb and free mMAE was equivalent to those of each component in the aCD22 mAb-mMAE conjugate. Five million leukemia cells were inoculated per mouse via intra-bone marrow injection. Twenty four hours after leukemia inoculation, animals started receiving weekly iv treatments for 3 weeks. When compared to controls (PBS, free aCD22 mAb or mMAE treatments), the treatment with the aCD22 mAb-mMAE conjugate increased the median survival time of the mice by two fold (Figure. PBS vs. aCD22 mAb-mMAE p<0.005, free mMAE vs. aCD22 mAb-mMAE p<0.05, free aCD22 mAb vs. aCD22 mAb-mMAE p<0.05, by Gehan-Breslow-Wilcoxon test). Leukemia-related death was confirmed by necropsy. Harvested leukemia cells were assessed by flow cytometry and found to be HLA and CD22 positive. During treatment, the mice in all the treatment groups remained healthy and active, and did not lose weight. Toxicity was assessed with weekly CBC and chemistry panels which revealed no significant toxicity. In conclusion, we demonstrated that aCD22 mAb-mMAE is efficacious in a preclinical preB ALL xenograft mouse model. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 7 (4) ◽  
pp. e2177-e2177 ◽  
Author(s):  
K Korfi ◽  
M Smith ◽  
J Swan ◽  
T C P Somervaille ◽  
N Dhomen ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 515-515
Author(s):  
Claire E. Pillsbury ◽  
Jairo A. Fonseca ◽  
Jodi Dougan ◽  
Hasan Abukharma ◽  
Gloria Gonzalez-Flamenco ◽  
...  

Abstract Immunotherapies have recently shown efficacy in treatment of aggressive, refractory pediatric B cell acute lymphoblastic leukemia (B-ALL), which remains one of the leading causes of cancer-related death in children. The immune evasion mechanisms of B-ALL are still being explored to discover new therapeutic targets and improve patient outcomes. Recent reports have implicated a role for the molecule Siglec-15 (Sig15) in regulating immune response in solid tumor-infiltrating macrophages. Our lab has found higher expression of SIGLEC15 at the RNA level in primary pediatric B-ALL as compared to healthy donor controls, as well as at the RNA and protein levels across a panel of B-ALL, T cell acute lymphoblastic leukemia (T-ALL), and diffuse large B cell lymphoma (DLBCL) cell lines compared to healthy donor PBMCs. Higher expression of SIGLEC15 in pediatric B-ALL samples from the TARGET database correlates with markers of PKC and NFκB activation known to drive B-ALL leukemogenesis, which we have demonstrated to regulate Sig15 RNA and protein expression in vitro. Knockout of Siglec15 expression in a BCR-ABL1 + murine model of B-ALL engrafted in immunocompetent and Rag1 -/- immunodeficient recipients resulted in leukemia clearance in immunocompetent, but not immunodeficient, recipients and 100% survival (Figure A, p=0.01 Sig15 KO into WT vs. Rag1 -/-). Further study indicates that Siglec15 expression on these leukemia cells suppresses T cell effector and memory population expansion at 7 days post-engraftment (Figure B) and correlates with higher levels of IL-10 and lower levels of CCL17 present in the bone marrow, representing a more immunosuppressive bone marrow milieu. These data suggest a prominent role for Sig15 in the suppression of adaptive immune response to B-ALL as well as other hematological malignancies. We have also reported for the first time the release of a soluble form of Sig15 (sSig15), which we have demonstrated to circulate at higher levels in the plasma of pediatric B-ALL patients compared to healthy donors (Figure C, ****P≤0.0001). Detection of this sSig15 negatively correlated with circulating levels of IL-12 and IL-1α/β (Figure D, depicting correlations of cytokines using Pearson's r), suggesting sSig15 levels correspond to a systemically immunosuppressive phenotype. Flow cytometry of fresh pediatric B ALL cells demonstrates expression of surface Sig15 in a subset of cases. Thus, Sig15 has the capacity to promote immunosuppressive effects at both marrow-localized and systemic levels. Together, these results suggest Siglec-15 is a novel, potent immunosuppressive molecule active in leukemia progression that may be targeted therapeutically to activate T lymphocytes against leukemia cells. Figure 1 Figure 1. Disclosures Abukharma: NextCure Inc.: Current Employment. Liu: NextCure: Current Employment, Current holder of stock options in a privately-held company.


Sign in / Sign up

Export Citation Format

Share Document