Glutathione-Depletion Potently Enhances the Cytotoxic Activity of Arsenic Trioxide in Imatinib Sensitive and Resistant Bcr-Abl Positive Cell Lines.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2098-2098
Author(s):  
Heiko Koenig ◽  
Erika M. Moseson ◽  
Christian Lorentz ◽  
Peter Paschka ◽  
Junia V. Melo ◽  
...  

Abstract Arsenic trioxide (ATO, Trisenox™) is currently being investigated as a potential drug to optimize imatinib based treatment of chronic myelogenous leukemia (CML). ATO has antileukemic activity against imatinib sensitive and resistant CML cell lines and primary cells derived from patients with CML. However, preliminary clinical data reveal only moderate activity of ATO in CML (imatinib sensitive or resistant). ATO-activity in acute myelogenous leukemia or multiple myeloma cells is inversely related to the intracellular Glutathione- (GSH-) content with cells bearing low GSH-levels being the most sensitive. To investigate whether this relationship plays a role in CML cell lines sensitive or resistant to the Bcr-Abl inhibitor imatinib, we determined the antiproliferative activity of ATO with regard to the cellular GSH-levels. Furthermore, we examined the ability of GSH-depletion as a therapeutic tool to sensitize CML cells to ATO. MTS-proliferation assays were performed to determine the concentration of ATO needed to induce 50% cellular growth inhibition (IC50). Cell lines used were the imatinib sensitive CML blast crisis lines AR230-s, KCL22-s, LAMA-s as well as their imatinib resistant derivatives AR230-r1, KCL22-r1, LAMA-r1. Known ATO-sensitive AML cell lines (NB4, HL60) with low GSH-content were also included in this study. Intracellular GSH-levels were measured biochemically using a commercially available kit. Protein content was analyzed using the Bradford method. ATO toxicity was analyzed using trypan blue exclusion and flow cytometric analysis of Annexin/PI-stained cells. MTS-proliferation assays indicate a cell type dependent activity of ATO with IC50-values ranging from 0.37±0.03 μM (NB4) up to 6.9±1.43 μM (AR230-r1). The most sensitive NB4 cells express low GSH-levels (8.74±2.9 nmol/mg), whereas highly resistant AR230-r1 cells reveal a threefold increased GSH-content (26.7±8nmol/mg). Imatinib resistance in AR230-r1 and LAMA-r1 cells is not associated with significant GSH-content modulation when compared to the imatinib naïve counterparts (23.6±5.4nmol/mg [AR230-s] vs 26.7±8nmol/mg [AR230-r1]; 8.3±1.8nmol/mg [LAMA-s] vs 6.9±0.2 [LAMA-r1]). Treatment of AR230-s cells with 100 μM of the GSH-depleting agent L-Buthionine-Sulfoximine (BSO) for 12 h leads to significant downregulation of cellular GSH (23.6±5.4 nmol/mg [control] vs 4,25±0,53 nmol/mg [100 μM BSO]). Treatment with BSO alone does not affect cellular viability nor induces apoptosis. Subsequent cotreatment of AR230-s cells with ATO (1 μM) and BSO (100 μM) for 24 h reduces viability to 31.6 % compared to untreated cells. In contrast, treatment with 1 μM ATO alone does not affect viability. Flow cytometric analysis of apoptosis reflects viability data with a 5.5 fold increase of apoptotic cells in the combined treated fraction. Similar data were generated using the KCL22-s cell line. Experiments using primary patient cells are currently in progress. Our data indicate that high intracellular GSH-content confers relative resistance to ATO in Bcr-Abl positive cell lines regardless whether they are imatinib sensitive or resistant. A promising tool to increase the antileukemic activity of ATO is the application of GSH-depleting agents. Therefore, GSH-dependent response to ATO treatment needs further investigation in individual CML patients.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4246-4246
Author(s):  
Xuemei Sun ◽  
Junhao Chen ◽  
Pengjun Jiang ◽  
Xuejun Zhu

Abstract Assay of phosphotyrosine levels using flow cytometry has been used to identify patients with chronic myelogenous leukemia (CML) positive for the Bcr-Abl fusion gene. We hypothesized that clinical monitoring could identify treatment response through reductions in intra-granulocyte phosphotyrosine. Initially, we studied cell lines FDC-P1 and HL60 (Bcr-Abl–negative) and FDrv210 and K562 (Bcr-Abl–positive) with our technique. A fluorescein isothiocyanate-conjugated monoclonal antibody was used along with fluorescence-conjugated microspheres for reference (ratio of sample fluorescence: bead fluorescence=relative fluorescence unit [RFU]). Samples from 20 controls and 32 patients undergoing treatment were analyzed using the same method. Bcr-Abl status for each patient was confirmed using fluorescent in situ hybridization (FISH) or polymerase chain reaction gene amplification (PCR). Testing of cell lines consistently produced expected results. Patient values were found to be significantly higher than control values (P<0.001) and values for patients with advanced disease were significantly higher than for patients with chronic-phase disease (P< 0.05). Results of clinical monitoring were consistent with results from PCR. Two patients who received allogeneic stem cell transplantation had molecular remission confirmed by PCR and had a marked decrease in RFU value (from 62 to 5 and from 131 to 23). No such fluorescence change was observed in patients who achieved clinical remission. Flow cytometric analysis of phosphotyrosine levels is a reliable and convenient adjuvant technique for diagnosis of Bcr-Abl–positive leukemias and shows promise for serial evaluation of patients undergoing treatment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4233-4233
Author(s):  
Hua Yan ◽  
Dao Li ◽  
Li Chen ◽  
Ying-Li Wu ◽  
Jun-Ming Li ◽  
...  

Abstract Chronic Myelogenous Leukemia (CML) is one of myeloproliferative disorders characterized by the chromosome translocation (9; 22), which causes the generation of the Bcr-Abl fusion protein. Latter activates a number of signal transducers and transcription factors, thus leading to multi-resistance to chemotherapeutic agents. Introduction of imatinib mesylate (Gleevec) significantly increased CML patients’ response but still some of patients got disease progression. Therefore, it still remains a high priority to develop new approaches to treat Bcr-Abl+ leukemia. Aim: We aim to investigate the synergy of bortezomib and arsenic trioxide on inducing the apoptosis on Bcr-Abl+ K562-s as well as K562-r cells, which was sensitive or resistant to imatinib treatment respectively. Material and methods: Bcr-Abl+ K562-s and K562-r cells were chosen to evaluate the drugs’ effect. We observed the inhibition of cell growth and cell viability after bortezomib and/or arsenic treatment. Flow cytometric evaluation and western blot analysis were performed to detect the development of apoptosis, the changeable expression of Bcr-Abl protein and the apoptosis-related proteins. Results: K562-s and K562-r cells had different response to imatinib treatment. As expected, 0.25μM of imatinib can inhibit half of K562-s cell growth whereas 10–15 more times of concentration of imatinib were necessary to reach the same inhibitory effect in K562-r cells. Combined bortezomib at 12nM with 1μM of ATO, the concentration inducing 50% of K562-s growth arrest, synergistically induced apoptosis in K562-s cells after 48hrs of cotreatment, confirmed by notable elevation of Annexin V+ cells through flow cytometric analysis, when compared to those in the control or single treatment (p<0.01), and immunoblotting detection of caspase-3 cleavage, degradation of poly-adenosine diphosphate-ribose polymerase (PARP), and decreased expression of Bcr-Abl protein. More surprisingly, 24nM concentration of bortezomib and 2μM of ATO, 2 times of concentration needed for 50% of K562-s cell growth inhibition respectively, inhibited 50% of K562-r cells growth, far less than the increased proportion needed during imatinib treatment. And more of important, although single treatment of bortezomib or ATO had no or little effect on inducing apoptosis, combined treatment significantly induced the apoptosis in K562-r cells (p<0.05), associated with activation of apoptosis-relevant proteins and obvious degradation of Bcr-Abl protein. Conclusion: Taken together, these findings indicate that bortezomib combined with ATO could be developed into a novel therapeutic strategy for CML. More study will be needed to further explore its potent mechanism involved during cotreatment.


Blood ◽  
1985 ◽  
Vol 66 (1) ◽  
pp. 39-46
Author(s):  
HM Kantarjian ◽  
B Barlogie ◽  
M Pershouse ◽  
D Swartzendruber ◽  
MJ Keating ◽  
...  

In an effort to develop a new tumor marker suitable for flow cytometric analysis, we examined the value of double-stranded ribonucleic acid (ds- RNA) measurements using propidium iodide after DN'ase treatment. Cellular ds-RNA content was evaluated both in experimental cell lines and in clinical specimens. Higher levels of ds-RNA were present in tumor cells as compared with normal cells. In tumor cells, fluorescence was intensely localized in the nucleolus and was more diffuse in the cytoplasm. Change of less than 10% in the ds-RNA levels was observed in cell lines as a function of cytokinetic determinants such as cycle phase, culture age, and cycle traverse rate. Tumor differentiation by dimethylsulfoxide resulted in a significant decrease in cellular ds-RNA content. For quantitative comparison of clinical material, a ds-RNA excess was defined in relationship to normal peripheral blood lymphocytes. ds-RNA excess greater than 30% was observed in only one of 34 normal tissues (3%) as compared with 124 of 201 neoplastic tissue samples (62%). This incidence was higher in patients with acute leukemia (76%), high-grade and intermediate-grade lymphoma (75%), and high tumor stage myeloma (83%), as compared with chronic leukemia (20%), low-grade lymphoma (25%), and intermediate or low tumor mass myeloma (43%). Prognostically, a high pretreatment ds-RNA excess in myeloma was associated with a lower remission rate. The persistence of ds-RNA excess in the bone marrow of patients with acute myelogenous leukemia in remission predicted for a shorter remission duration (seven v 22 months; P = .05). We conclude that ds-RNA excess, as readily measured objectively and quantitatively by flow cytometry, may have important diagnostic and prognostic implications for the management of patients with malignant disease.


Leukemia ◽  
2003 ◽  
Vol 17 (5) ◽  
pp. 931-940 ◽  
Author(s):  
K Davison ◽  
S Côté ◽  
S Mader ◽  
W H Miller

2017 ◽  
Vol 44 (1) ◽  
pp. 99-109 ◽  
Author(s):  
Fang Yang ◽  
Lizhi Lv ◽  
Kun Zhang ◽  
Qiucheng Cai ◽  
Jianyong Liu ◽  
...  

Background/Aims: Increasing evidence has indicated that Forkhead box protein C2 (FOXC2) plays an important role in carcinogenesis. However, the expression and the role of FOXC2 in hepatocellular carcinoma (HCC) have not been extensively studied. Methods: FOXC2 expression was analyzed by quantitative real-time polymerase chain reaction, Western blot analysis and immunohistochemistry in HCC tissue and cells. The relationship between FOXC2 expression and patient clinical significance and survival were assessed by Pearson’s correlation and Kaplan-Meier analysis, respectively. Cell proliferation assays, colony formation assays, flow cytometric analysis and Transwell assays were employed to measure the effects of FOXC2 on HCC cells in vitro. Results: The expression of FOXC2 was increased in HCC tissue, and high FOXC2 expression was associated with worse patient survival. Knockdown of FOXC2 inhibited HCC cell growth, migration, and invasion in vitro, as well as tumor growth. Furthermore, we found that activation of AKT-mediated MMP-2 and MMP-9 was involved in FOXC2 promoting an aggressive phenotype. Conclusions: Taken together, these findings demonstrate that FOXC2 is upregulated in HCC tissue and is associated with tumor size, vascular invasion and advanced TNM stage. Further investigation suggested that FOXC2 may play a vital role in promoting proliferation and invasion in HCC and serves as a novel therapeutic target in HCC.


Blood ◽  
2003 ◽  
Vol 102 (13) ◽  
pp. 4512-4519 ◽  
Author(s):  
Joya Chandra ◽  
Jennifer Hackbarth ◽  
Son Le ◽  
David Loegering ◽  
Nancy Bone ◽  
...  

Abstract Adaphostin (NSC 680410), an analog of the tyrphostin AG957, was previously shown to induce Bcr/abl down-regulation followed by loss of clonogenic survival in chronic myelogenous leukemia (CML) cell lines and clinical samples. Adaphostin demonstrated selectivity for CML myeloid progenitors in vitro and remained active in K562 cells selected for imatinib mesylate resistance. In the present study, the mechanism of action of adaphostin was investigated in greater detail in vitro. Initial studies demonstrated that adaphostin induced apoptosis in a variety of Bcr/abl- cells, including acute myelogenous leukemia (AML) blasts and cell lines as well as chronic lymphocytic leukemia (CLL) samples. Further study demonstrated that adaphostin caused intracellular peroxide production followed by DNA strand breaks and, in cells containing wild-type p53, a typical DNA damage response consisting of p53 phosphorylation and up-regulation. Importantly, the antioxidant N-acetylcysteine (NAC) blunted these events, whereas glutathione depletion with buthionine sulfoximine (BSO) augmented them. Collectively, these results not only outline a mechanism by which adaphostin can damage both myeloid and lymphoid leukemia cells, but also indicate that this novel agent might have a broader spectrum of activity than originally envisioned. (Blood. 2003;102:4512-4519)


Sign in / Sign up

Export Citation Format

Share Document