Induction of caspase-dependent programmed cell death in B-cell chronic lymphocytic leukemia by anti-CD22 immunotoxins

Blood ◽  
2004 ◽  
Vol 103 (7) ◽  
pp. 2718-2726 ◽  
Author(s):  
Thomas Decker ◽  
Madlene Oelsner ◽  
Robert J. Kreitman ◽  
Giuliana Salvatore ◽  
Qing-cheng Wang ◽  
...  

Abstract B cells of chronic lymphocytic leukemia (CLL) are long-lived in vivo, possibly because of defects in apoptosis. We investigated BL22, an immunotoxin composed of the Fv portion of an anti-CD22 antibody fused to a 38-kDa Pseudomonas exotoxin-A fragment. B cells from 22 patients with CLL were immunomagnetically enriched (96% purity) and were cultured with BL22 or an immunotoxin that does not recognize hematopoietic cells. The antileukemic activity of BL22 was correlated with CD22 expression, as determined by flow cytometry. BL22 induced caspase-9 and caspase-3 activation, poly(adenosine diphosphate [ADP]-ribose)polymerase (PARP) cleavage, DNA fragmentation, and membrane flipping. Cell death was associated with the loss of mitochondrial membrane potential and the down-regulation of Mcl-1 and X-chromosomal inhibitor of apoptosis protein (XIAP). Furthermore, BL22 induced a proapoptotic 18-kDa Bax protein and conformational changes of Bax. Z-VAD.fmk abrogated apoptosis, confirming that cell death was executed by caspases. Conversely, interleukin-4, a survival factor, inhibited spontaneous death in culture but failed to prevent immunotoxin-induced apoptosis. BL22 cytotoxicity was markedly enhanced when combined with anticancer drugs including vincristine. We also investigated HA22, a newly engineered immunotoxin, in which BL22 residues are mutated to improve target binding. HA22 was more active than BL22. In conclusion, these immunotoxins induce caspase-mediated apoptosis involving mitochondrial damage. Combination with chemotherapy is expected to improve the efficacy of immunotoxin treatment. (Blood. 2004;103:2718-2726)

Blood ◽  
2004 ◽  
Vol 104 (3) ◽  
pp. 788-794 ◽  
Author(s):  
Yean K. Lee ◽  
Nancy D. Bone ◽  
Ann K. Strege ◽  
Tait D. Shanafelt ◽  
Diane F. Jelinek ◽  
...  

AbstractWe recently reported that chronic lymphocytic leukemia (CLL) cells synthesize and release vascular endothelial growth factor (VEGF) under normoxic and hypoxic conditions. CLL B cells also express VEGF membrane receptors (VEGF-R1 and VEGF-R2), suggesting that they use VEGF as a survival factor. To assess the mechanism of apoptosis resistance related to VEGF, we determined the impact of VEGF on CLL B cells, and we studied the impact of epigallocatechin-3-gallate (EGCG), a known receptor tyrosine kinase (RTK) inhibitor, on VEGF receptor status and viability of CLL B cells. VEGF165 significantly increased apoptotic resistance of CLL B cells, and immunoblotting revealed that VEGF-R1 and VEGF-R2 are spontaneously phosphorylated on CLL B cells. EGCG significantly increased apoptosis/cell death in 8 of 10 CLL samples measured by annexin V/propidium iodide (PI) staining. The increase in annexin V/PI staining was accompanied by caspase-3 activation and poly–adenosine diphosphate ribose polymerase (PARP) cleavage at low concentrations of EGCG (3 μg/mL). Moreover, EGCG suppressed the proteins B-cell leukemia/lymphoma-2 protein (Bcl-2), X-linked inhibitor of apoptosis protein (XIAP), and myeloid cell leukemia-1 (Mcl-1) in CLL B cells. Finally, EGCG (3-25 μg/mL) suppressed VEGF-R1 and VEGF-R2 phosphorylation, albeit incompletely. Thus, these results suggest that VEGF signaling regulates survival signals in CLL cells and that interruption of this autocrine pathway results in caspase activation and subsequent leukemic cell death.


Blood ◽  
1996 ◽  
Vol 88 (6) ◽  
pp. 2172-2182 ◽  
Author(s):  
F Mentz ◽  
MD Mossalayi ◽  
F Ouaaz ◽  
S Baudet ◽  
F Issaly ◽  
...  

We tested the effects of theophylline, a phosphodiesterase inhibitor inducing intracellular accumulation of cyclic adenosine monophosphate (cAMP), on malignant B cells from 15 patients with B-chronic lymphocytic leukemia (B-CLL). We observed a large increase in apoptotic cell numbers (mean, 90% v 20% in medium alone) in the presence of theophylline (100 micrograms/mL) or chlorambucil (10 mumol/L) after 72 hours of incubation. Maximal apoptosis (90%) was reached after 36 hours when the two drugs were used together at fourfold lower concentrations, indicating a synergistic effect; no effect was observed with normal B cells, suggesting that the combination might have therapeutic interest. Chlorambucil induced intracellular Ca+2 influx, pointing to the involvement of two signaling pathways that might explain its synergy with theophylline through their effects on oncogenes. The expression of bcl-2 protein, a proto-oncogene inhibiting apoptosis, decreased after incubation with the drugs, while c-myc, recently described as having a potent role in apoptosis, was overexpressed. For p53 we observed an overexpression in the presence of chlorambucil or both theophylline- chlorambucil and a decrease after theophylline incubation. Chlorambucil- and theophylline-induced apoptosis was partially inhibited by interleukin-4 (IL-4), which also abrogated the effects on oncogene expression. These results provide insight into the mechanisms underlying B-CLL apoptosis and suggest that the theophylline- chlorambucil combination may be of therapeutic value in this setting.


Blood ◽  
1997 ◽  
Vol 89 (9) ◽  
pp. 3378-3384 ◽  
Author(s):  
Beatriz Bellosillo ◽  
Mireia Dalmau ◽  
Dolors Colomer ◽  
Joan Gil

Abstract B-chronic lymphocytic leukemia (B-CLL) is characterized by the accumulation of long-lived B lymphocytes that express high levels of Bcl-2. We examined the involvement of CED-3/ICE-like proteases in the apoptosis of B-CLL cells. One of the substrates of these proteases is poly(ADP [adenosine 5′-diphosphate]-ribose) polymerase (PARP). The effect of different factors that induce the apoptosis of B-CLL cells on the proteolytic cleavage of PARP has been studied. Treatment of B-CLL cells with different concentrations of dexamethasone (1 to 1,000 μmol/L) induced in a dose-dependent manner the cleavage of PARP. Dexamethasone induced PARP cleavage after 12 hours of incubation, which was almost complete at 48 hours. PARP cleavage during apoptosis of B-CLL cells was studied in cells from eight patients and a correlation was found between cell viability and the degree of PARP cleavage. Incubation in vitro of B-CLL cells with fludarabine for 48 hours induced PARP cleavage in all the cases studied. Protein kinase C (PKC) activation with 100 nmol/L TPA (12-O-tetradecanoylphorbol 13-acetate) or incubation with interleukin-4 (10 ng/mL) prevented either dexamethasone- or fludarabine-induced proteolysis of PARP. Incubation of B-CLL cells with the CED-3/ICE–like protease inhibitor Z-VAD.fmk inhibited spontaneous and dexamethasone-induced PARP cleavage and DNA fragmentation in a dose-dependent manner. Furthermore, Z-VAD.fmk prevented the cytotoxic effect of dexamethasone. These results indicate that CED-3/ICE–like proteases play an important role in the apoptosis of B-CLL cells.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3778-3778
Author(s):  
Li-Yuan Bai ◽  
Yihui Ma ◽  
Samuel K Kulp ◽  
Shu-Huei Wang ◽  
Chang-Fang Chiu ◽  
...  

Abstract Abstract 3778 Poster Board III-714 Introduction Chronic lymphocytic leukemia (CLL) is a most common form of adult leukemia with minimal treatment options. Drug resistance and associated immune deregulation limit use of current therapies, thus warranting need for alternative therapy development. Our laboratories recently identified a novel D-tyrosinol-derived compound targeting p38 MAPK pathway, OSU-DY7 [(R)-2-amino-3-(4-heptyloxy-phenyl)-propan-1-ol]. Here we demonstrate the efficacy of OSU-DY7 for lymphocytic cell lines and primary B cells from CLL patients. Materials and Methods We tested the pre-clinical efficacy of OSU-DY7 in primary CLL B cells and B cell lines representing CLL (MEC-1), ALL (697), and lymphoblastic lymphoma (Raji and Ramos). Results The cytotoxicity of OSU-DY7 was both dose- and time-dependent. The IC50 of OSU-DY7 at 24 hrs for MEC-1, 697, Raji, Ramos and primary B-CLL cells were 2.40 μM, 10.39 μM, 6.04 μM, 3.75 μM and 3.58 μM respectively. OSU-DY7 induced activation of caspase-3 and poly (adenocine diphosphate-ribose) polymerase (PARP) cleavage. Pancaspase inhibitor Z-VAD-FMK partially rescued OSU-DY7-induced cytotoxicity in CLL B cells and cell lines. Interestingly, OSU-DY7 mediated-cytotoxicity was associated with increased phosphorylation of p38 MAPK and its downstream target protein MAPKAPK2 in both lymphocytic cell lines and primary B-CLL cells. Compared with control group, the ratio of p-p38MAPK (Thr180Tyr182) versus p38MAPK in Raji cells treated with 2 μM, 4 μM and 8 μM of OSU-DY7 increased 2.2, 4.7 and 11 fold respectively (p<0.0001). Similar increase in p38MAPK phosphorylation was also observed in six CLL patient samples after treatment with OSU-DY7 2 μM, 4 μM and 8 μM for 24 hours (p=0.0004). Moreover, SB202190, a specific p38 MAPK inhibitor, decreased MAPKAPK2 protein level with concomitant rescue of the cells from the OSU-DY7 mediated-cytotoxicity. Thus pretreatment of Raji and primary CLL B cells with SB202190 reduced the OSU-DY7 induced cytotoxicity by 36.7% and 24.3% respectively by 24hrs (Raji:p<0.0001; primary CLL B cells: p<0.005 ). Furthermore, SB202190 rescued OSU-DY7 down regulated survivin protein by ∼2 fold (p<0.0001, n=3) and mRNA levels by 2.4 fold (95% CI: 1.56∼3.84 fold, p=0.0026, n=3) in Raji cells indicating a role for p38MAPK dependent regulation of survivin by OSU-DY7. Conclusions This study provides an evidence for a role of OSU-DY7 in p38 MAPK dependent activation and survivin down regulation associated with apoptosis in lymphocytic cells, thus warranting further development of this novel agent for alternative therapy for lymphocytic malignancies. [This work was supported by D. Warren Brown Foundation and Leukemia and Lymphoma Society] Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 377-377 ◽  
Author(s):  
Peng Liu ◽  
Bei Xu ◽  
Jianyong Li

Abstract Abstract 377 Impaired cell death program has been noted as one of the hallmarks of Chronic lymphocytic leukemia (CLL) and contributes to its accumulation of malignant monoclonal B cells as well as to chemotherapy resistance. A cell can die through apoptosis or necrosis pathway. While apoptosis is known as a regulated cellular program, necrosis is known as an accidental event caused by overwhelming stress. However, accumulating evidence suggests that necrosis can also be executed by regulated mechanisms, especially in apoptotic-deficient conditions. Recently, the term necroptosis has been used to designate one particular form of programmed necrosis induced by stimulating death receptors with agonists such as TNFα, FasL, and TRAIL. Apoptosis suppression by caspase inhibitors such as zVAD may switch apoptotic response to necroptosis or enhance necroptosis. In contrast to well-characterized apoptotic pathway, the detailed molecular mechanisms underlying necroptosis are still not fully understood. A genome wide siRNA screen revealed two members of the receptor interacting protein (RIP) kinase family, RIP1 and RIP3P, to be essential for necroptosis. Upon stimulation of death receptors, RIP3 is recruited to RIP1 to form a necroptosis-inducing complex which is essential for cell death execution. The deubiquitinase cylindromatosis (CYLD) is recruited to TNFα receptor upon its activation and directly regulates RIP1 ubiquitination. In addition, by activating key enzymes of metabolic pathways, RIP3 regulates TNFα-inducing mitochondrial reactive oxygen species (ROS) production, which partly accounts for its ability to potentiate necroptosis. Until now, much less is known about the significance of necroptosis in malignant disease. Here we demonstrate that primary CLL cells failed to undergo necroptosis upon stimulation of TNFα combined with pan-caspase inhibitor zVAD. Upon TNFα+zVAD stimulation, normal CD19+ B cells increased ROS production > 8 fold, while same treatment only resulted in ∼ 2 fold induction in ROS generation in CLL samples. Two core components of necroptotic machine, RIP3 and CYLD, are markedly down-regulated in CLL compared with normal B cells, at both protein and transcription levels. Moreover, we identified LEF1, a downstream effector of Wnt/β-catenin pathway, as a transcription repressor of CYLD in CLL. LEF1 is highly expressed in CLL cells, whereas normal B cells have very low levels of LEF1 expression. Attenuation of LEF1 expression through RNAi technology resulted in a dramatic increase in CYLD levels in CLL cells, as determined by western blot and real time RT-PCR analysis. Dual-luciferase assays showed that forced expression of LEF1 markedly decreased CYLD promoter activity compared with controls. Mutation of LEF1 responsive elements (LERs) on CYLD promoter significantly abolished transcriptional repression of CYLD by LEF1. Chromatin immunoprecipitation assays showed that LEF1 is recruited to LER region within the CYLD promoter in CLL cells. Additionally, Knocking down LEF1 sensitizes CLL cells to TNFα-induced necroptosis. The present investigation provides the first evidence that CLL cells have defects not only in apoptotic program but also in necroptotic signaling. Targeting the key regulators of necroptotic machine such as LEF1 to restore this pathway may represent a novel approach for CLL treatment. Disclosures: No relevant conflicts of interest to declare.


PLoS Medicine ◽  
2015 ◽  
Vol 12 (3) ◽  
pp. e1001796 ◽  
Author(s):  
Ana-Carolina Martinez-Torres ◽  
Claire Quiney ◽  
Tarik Attout ◽  
Heloïse Boullet ◽  
Linda Herbi ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3510-3510
Author(s):  
Sneha V. Gupta ◽  
Erin K Hertlein ◽  
Jennifer A. Woyach ◽  
Ellen J. Sass ◽  
Melanie E. Davis ◽  
...  

Abstract Abstract 3510 The reversible proteasome inhibitor bortezomib is effective in the treatment of multiple myeloma and mantle cell lymphoma, but failed to produce objective responses in chronic lymphocytic leukemia (CLL). Carfilzomib (CFZ) is a tetrapeptide ketoepoxide that belongs to a new class of irreversible proteasome inhibitors that specifically target the chymotrypsin-like subunit of the 26S proteasome. Based on preclinical data demonstrating potent cytotoxicity in primary CLL cells, CFZ is currently in a phase I clinical trial at The Ohio State University in patients with relapsed or refractory CLL. However, the mechanism of action of CFZ in CLL is unknown. We have therefore investigated the effects of CFZ on CLL patient cells ex vivo. Here we demonstrate that a short (1 hr) exposure of 100 nM CFZ is sufficient to inhibit the chymotrypsin-like proteasome subunit in CLL cells. This exposure is also rapidly cytotoxic, inducing apoptosis in approximately 50% of cells by 24 hr (annexin+ and/or PI+). Unlike bortezomib, the cytotoxicity of carfilzomib is not diminished in media with human serum compared to fetal bovine serum. Additionally, CFZ is more cytotoxic to normal CD19+ B cells than normal CD3+ T cells at clinically relevant concentrations of 33 to 300 nM, suggesting that this agent could potentially avoid immune-suppressing T-cell depletion that is commonly noted with chemotherapy. CFZ causes CLL cell death ex vivo by a caspase-dependent apoptotic pathway, indicated by PARP cleavage and rescue by the broad caspase inhibitor Boc-D-fmk. Importantly, our studies indicate that CFZ causes cytotoxicity in primary CLL cells irrespective of p53 status. This tumor suppressor, which is functional in most CLL patients at the time of diagnosis, is mutated or deleted in at least one allele in approximately 40% of patients with advanced CLL, and p53 pathway dysfunction is associated with resistance to standard therapies and poor overall survival. Therefore, the ability of CFZ to induce apoptosis irrespective of p53 function is of important clinical significance. Additionally, the pro-apoptotic protein Noxa is increased following CFZ treatment despite a lack of induction in mRNA, suggesting accumulation of protein via inhibition of proteasome-mediated degradation. The NF-kB signaling pathway is broadly implicated in CLL cell survival and resistance to therapy, and proteasome inhibitors have been reported to block this pathway via inhibition of IkB degradation. We therefore investigated the impact of CFZ on NF-kB-mediated transcription in CLL patient cells. Paradoxically, our results indicate that CFZ leads to activation of NF-kB, as evidenced by increased nuclear accumulation of the p50 and p65 subunits of NF-kB, as well as phosphorylated IkBα. This correlates with enhanced binding of the p50/p65 heterodimer to an NF-kB probe in an electrophoretic mobility shift assay. However, despite this apparent NF-kB activation, no transcriptional increases were observed in NF-kB targets genes including Mcl-1, p53, Bcl-2, Bcl2A1 or XIAP. In addition, inhibition of NF-kB activity using a dominant negative (non-phosphorylatable) IkBα did not impair CFZ-induced cytotoxicity. This is the first study suggesting that treatment with a proteasome inhibitor induces a defective NF-κB response in CLL cells. The mechanism and relevance of this effect, as well as the pathway by which CFZ causes CLL cell death, continues to be investigated. Collectively, our data indicate that proteasome inhibition is a relevant therapeutic target in CLL and supports the development of carfilzomib for the treatment of this currently incurable disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2978-2978
Author(s):  
Carlos E. Prada ◽  
Januario E. Castro ◽  
Dayong Zhai ◽  
Shinichi Kitada ◽  
John C. Reed ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of monoclonal B cells in the blood, secondary lymphoid tissues, and marrow. The leukemia cells primarily are arrested in the G0/G1 phase of the cell cycle and appear resistant to programmed cell death. Several anti-apoptotic proteins are over expressed in CLL and this correlates with resistance to treatment, disease progression and overall poor prognosis. Proteins in the Bcl-2 family are central regulators of programmed cell death, and members that inhibit apoptosis, such as Bcl-X(L), Bcl-2, and Mcl-1, are overexpressed in many cancers including CLL and contribute to tumour initiation, progression and resistance to therapy. Mcl-1 is of particular interest because this molecule appears to be regulated by Nurse-like Cells and other stromal cells that promote survival of CLL cells in vitro and very likely also in vivo. These proteins enhance the resistance of CLL cells to spontaneous and/or drug-induced apoptosis primarily by interacting with, and antagonizing the activity of mitochondria membrane pro-apoptotic proteins such as Bax and Bak. The protein-protein interaction of Bcl-2 family members is critical for their activity, and these interactions are governed by binding to the BH3 domain. Racemic gossypol is found in cotton seeds and has been studied as a cytotoxic agent in cancer cell lines and in clinical studies in patients with a large variety of cancers. The antitumor activity of racemic gossypol appears to reside principally in the R-(−)-enantiomer (AT101), with reduced activity observed for the S-(+)-enantiomer. AT101 is an antagonist of the BH3-binding groove of the Bcl-2 family of proteins that can inhibit the interactions of these proteins with pro-apoptotic molecules. We examined whether AT101 could induce apoptosis in Chronic Lymphocytic Leukemia (CLL) and its ability to bind in vitro anti-apoptotic molecules from the Bcl-2 family. Using a Fluorescence Polarization Assay (FPA) we studied the competitive binding affinity of AT101 to Bcl-2 family member proteins. We observed that both, racemic gossypol and AT101 had comparable affinity for Bck-2, Bcl-B, Bfl-1 with EC50=0.6 to 10 μM range. AT101 had a stronger binding affinity to Bcl-X(L) (EC50=0.998 μM vs. 3.084 μM for racemic gossypol), and to Mcl-1 (EC50= 0.52μM vs. 1.07μM for racemic gossypol). CLL cells were incubated with AT101 for 48 hrs at different concentrations. We observed that leukemia cells were induced to undergo apoptosis in a time and dose dependent manner and that this effect was independent of ZAP-70 expression or IgVH gene mutational status (IC50= 2μM). Cells undergoing apoptosis showed PARP-1 cleavage and upregulation of pro-apoptotic molecules such as Bid, p53, as well as downregulation of Mcl-1. These results indicate that AT101 has stronger pan-specific binding affinity for Bcl-2 family proteins than racemic gossypol, in particular to Mcl-1 and Bcl-X(L), and that this compound induces apoptosis in CLL B cells independently of ZAP-70 expression or IgVH gene mutational status. Because of these encouraging results a clinical trial using AT101 in CLL patients with high-risk features is currently open at our institution.


Blood ◽  
2005 ◽  
Vol 105 (5) ◽  
pp. 2099-2106 ◽  
Author(s):  
Tait D. Shanafelt ◽  
Yean K. Lee ◽  
Nancy D. Bone ◽  
Ann K. Strege ◽  
Ven L. Narayanan ◽  
...  

AbstractB-cell chronic lymphocytic leukemia (CLL) is characterized by accumulation of clonal lymphocytes resistant to apoptosis. We evaluated the ability of the investigational antileukemic agent adaphostin to induce apoptosis in CLL B cells and synergize with fludarabine in vitro. Analysis by annexin V/propidium iodide (PI) staining revealed that the concentration of adaphostin required to induce 50% cell death (IC50) at 24 hours was 4.2 μM (range, 1.10-11.25 μM; median, 4.25 μM; n = 29) for CLL isolates and more than 10 μM for B and T cells from healthy donors. Immunoblots demonstrated adaphostin induced poly(adenosine diphosphate-ribose) polymerase (PARP) cleavage and cleavage of caspase-3 substrates, suggesting that adaphostin induces apoptosis. Adaphostin increased the level of reactive oxygen species (ROS) within CLL B cells, and the antioxidant N-acetylcysteine blocked both adaphostin-induced ROS generation and apoptosis. Adaphostin also caused a decrease in the level of the antiapoptotic protein Bcl-2. When adaphostin was combined with fludarabine (F-ARA-AMP), a synergistic effect on cell death was observed in all 10 CLL samples. These findings not only indicate that adaphostin induces apoptosis selectively in CLL B cells through a mechanism that involves ROS generation but also demonstrate its ability to augment the effects of fludarabine. Further preclinical development of adaphostin as a novel agent for the treatment of CLL appears warranted.


Sign in / Sign up

Export Citation Format

Share Document