scholarly journals The Effect of Iodine-131 Beta-particles in Combination With A-966492 and Topotecan on Radio-sensitization of Glioblastoma

Author(s):  
Fereshteh Koosha ◽  
Samira Eynali ◽  
Nazila Eyvazzadeh

Abstract Glioblastoma tumors are resistant to radiotherapy, and the need for drugs to induce radio-sensitization in tumor cells has always been a challenge. Besides, radiotherapy using targeted radionuclide is effective even for resistant tumors. Poly (ADP-ribose) polymerase (PARP) and topoisomerase I enzymes have critical roles in the repairmen of DNA damage in cells. Thus, the inhibition of the activity of these enzymes can prevent the process of DNA repair and lead to the accumulation of damaged DNA in cells, resulting in the induction of cell death in tumors. In the current study, we investigated the effect of beta-particles of iodine-131 in combination with Topotecan (TPT), as the inhibitor of topoisomerase I, and A-966492, as the inhibitor of the PARP enzyme to increase radio-sensitivity of glioblastoma cells.The U87MG cell line (a human glioblastoma cell line) were cultured in Poly-Hema-coated flasks to reach 300μm-diameter spheroids. Then, the cells were treated with non-toxic concentrations of A-966492 and TPT. The viability of the cells treated with iodine131 in combination with A-966492 and TPT was determined by the clonogenic assay. The expression level of the gamma-H2AX protein, as a biomarker of DNA double-strand breaks, was measured by the immunofluorescence staining method to examine the impact of A-966492 (1μM), TPT, and radiation on the induction cell death.The combination of A-966492 and TPT with radiation resulted in the enhanced cell death, and sensitizer enhancement ratios at 50% survival (SER50) were 1.25 and 1.45, respectively. Radio- and chemo-sensitization were promoted when iodine-131 was combined with A-966492 and TPT, with the SER50 of 1.68. Also, the expression of γ-H2AX was significantly increased in cells treated with A-966492 and TPT combined with radiation.The results demonstrated that iodine-131, in combination with A-966492 and TPT, had marked effects on radio-sensitizing and can be used as a targeted radionuclide for targeting radiotherapy in combination with PARP and topoisomerase I inhibitors to improve radiotherapy in clinics.

Circulation ◽  
2007 ◽  
Vol 116 (suppl_16) ◽  
Author(s):  
James Shaw ◽  
Natalia Yurkova ◽  
Kelly Regula ◽  
Tong Zhang ◽  
Floribeth Aguilar ◽  
...  

The hypoxia-inducible death factor Bnip3 is known to provoke mitochondrial perturbations and cell death of ventricular myocytes. The transcriptional control processes that govern Bnip3 gene expression under basal and inducible conditions remain cryptic. Sequence analysis of the Bnip3 promoter revealed the presence of distinct but overlapping DNA binding elements for the cell cycle factor E2F-1 and cellular factor NF-κB. Previously, we reported a survival role for NF-κB in ventricular myocytes. As a step toward elucidating the regulation of Bnip3 gene expression in ventricular myocytes, we tested the impact of E2F-1 and NF-κB on basal and inducible expression of Bnip3. A 2.0 fold increase in Bnip3 gene transcription was observed in cells expression wild type E2F-1 but not in cells expressing an E2F-1 mutant defective for DNA binding. Interestingly, basal Bnip3 gene transcription was increased by 2.5 fold in myocytes rendered defective for NF-κB activation with a non-phosphorylatable form of IκBα. Importantly, genetic ablation of E2F-1 inhibited basal and inducible Bnip3 transcription in NF-κB defective cells. Expression of the p65 subunit of NF-κB in NF-κB defective cells inhibited E2F-1 mediated Bnip3 transcription. Western blot analysis of cardiac cell lysate revealed that p65 NF-κB immunoprecipitated with E2F-1. ChIP analysis of the Bnip3 promoter indicated that the p65 NF-κB bound DNA under normoxic conditions. During hypoxia E2F-1 activity increased where as p65 NF-κB protein levels were decreased. ChIP analysis revealed increased binding of E2F-1 to the Bnip3 promoter during hypoxia which coincided with a 3.5 fold increase in Bnip3 gene transcription. IKKβ mediated activation of NF-κB activation abrogated hypoxia-induced E2F-1 binding to the Bnip3 promoter and Bnip3 gene transcription. To our knowledge our data provide the first direct evidence that a novel relationship exists between p65 NF-κB and E2F-1 for basal and hypoxia-inducible regulation of the Bnip3 promoter. Furthermore, our data highlight a novel survival pathway by which NF-κB averts hypoxia - induced cell death by antagonizing the E2F-1 dependent transcription of Bnip3 in ventricular myocytes.


2010 ◽  
Vol 2010 ◽  
pp. 1-7 ◽  
Author(s):  
Jeffrey Helgager ◽  
Jie Li ◽  
Irina A. Lubensky ◽  
Russell Lonser ◽  
Zhengping Zhuang

Despite resistance of most gliomas to chemotherapy, approximately 2/3 of oligodendrogliomas show sensitivity to such agents. This sensitivity has been associated with deletions on chromosome 1p alone or in combination with 19q. Higher expression of the enzyme glyoxalase I has been found in oligodendrogliomas with chromosome 1p intact compared to those with a deletion. Higher expression of this enzyme is also associated with tumor chemoresistance in other cancers. The present study tested whether the drug troglitazone would make a glioma cell line more sensitive to chemotherapeutic agents. This drug was chosen because it has been shown to decrease glyoxalase I enzyme activity in cells. Treatment with troglitazone decreased expression of glyoxalase I, and potentiated cell death when used in combination with chemotherapeutic agents. This decrease in glyoxalase I protein may be one mechanism by which this potentiation occurs, and troglitazone may be a candidate for use in glioma therapy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3723-3723
Author(s):  
Isere Kuiatse ◽  
Sheeba K. Thomas ◽  
Donna M. Weber ◽  
Adam M. Stein ◽  
Michael Wang ◽  
...  

Abstract Abstract 3723 Background: Waldenström macroglobulinemia (WM), a lymphoproliferative disorder with marrow infiltration by lymphoplasmacytic lymphoma cells and production of monoclonal IgM, is characterized clinically by good initial responses to standard therapeutic approaches. However, a minority of patients achieve a complete remission, and most inevitably relapse, indicating a need for validation of new, targeted therapies. Notably, B-cell receptor signaling has been linked to clonal evolution in WM, and Spleen tyrosine kinase (Syk) is over-expressed in primary patient cells. Also, a recent phase I/II study of the Syk inhibitor fostamatinib in patients with a variety of non-Hodgkin lymphomas showed evidence of activity in three patients with WM. These findings supported our central hypothesis, which proposed that Syk could be a rational new target for therapy of WM, which we pursued first with pre-clinical studies. Methods: Studies of fostamatinib were performed using the WM cell line MWCL-1, and the IgM-producing cell line BWCM.1, as well as in primary cells from patients with WM, and in a novel murine xenograft model utilizing MWCL-1 cells. Results: Fostamatinib induced a time- and dose-dependent reduction in viability of MWCL-1 and BWCM.1 cells, with median inhibitory concentrations (IC50) that were in the physiologically relevant range of approximately 0.25 and 1 μM, respectively, at 3 days. Cell cycle analysis showed a decrease in cells at S-phase and at G2/M, while an increase was seen in cells in G1, and in the sub-G1fraction, suggesting induction of apoptosis. Consistent with the possibility that there was activation of type I programmed cell death, increased staining was seen in fostamatinb-treated cells with an antibody to Annexin V. Interestingly, evidence was seen of the induction of type II programmed cell death, or autophagy, as measured by an increase in cells staining with the lysotropic dye acridine orange. At the molecular level, fostamatinib blocked signaling through p44/42 mitogen-activated protein kinase (MAPK), MAPK kinase (MEK), protein kinase B/Akt, and Zeta-chain-associated protein kinase (ZAP)-70/Syk, as judged by a reduction in the phosphorylated forms of these intermediates by Western blotting. Notably, the viability of primary cells isolated from patients with WM was also reduced in association with blockade of p44/42 MAPK. Finally, a novel murine xenograft model was generated by injection of MWCL-1 cells into immunodeficient mice, which developed measurable tumors. When these were treated with fostamatinib, a delay in tumor growth was demonstrable. Conclusions: Targeting Syk with fostamatinib is an attractive strategy against pre-clinical models of WM both in vitro and in vivo. These findings lend further support for translation of fostamatinib to the clinic for patients with relapsed, and possibly even newly diagnosed WM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 751-751
Author(s):  
Olga Zimmermannova ◽  
Eliska Doktorova ◽  
Jan Stuchly ◽  
Meritxell Alberich Jorda ◽  
Jiri Petrak ◽  
...  

Abstract Leukemias harboring ETV6-ABL1 fusion represent a rare prognostically unfavorable subgroup of hematological malignancies. To a great extent they share biological and clinical characteristics with more common BCR-ABL1+ leukemias. Both fusion genes encode constitutively activated aberrant tyrosine kinases which can be targeted by specific tyrosine kinase inhibitors (TKI). The efficiency of TKI to inhibit ETV6-ABL1 kinase activity was proven in vitro and substantiated their use in clinical practice. However, there is still a lack of knowledge on potential induction and mechanisms of TKI resistance in ETV6-ABL1+ malignancies. To address this issue we established a multi-TKI resistant ETV6-ABL1+ lymphoblastic cell line via a long term cultivation with TKI imatinib mesylate (IM) and investigated the mechanisms of the acquired TKI resistance. We excluded the most common mechanisms known from BCR-ABL1+ leukemias (kinase domain mutations, genomic amplification and enhanced kinase expression/activation) as causes of resistance in our model. Importantly, using shRNA mediated silencing we showed that unlike their sensitive counterparts, the resistant cells did not respond to the ETV6-ABL1 knockdown by growth suppression or cell death and, therefore, became independent on pro-survival signaling from the chimeric kinase. To further elucidate molecular mechanisms underlying the TKI resistance, we compared biological profiles of resistant and parental sensitive cells. Molecular changes potentially associated with the TKI resistance were analyzed via genomic (single nucleotide polymorphism array, whole exome sequencing), gene expression and proteomic profiling. We integrated data from all platforms and identified K89M mutation in GNB1 gene as the top candidate causative lesion. A mutation affecting the same GNB1 codon, K89E, was recently identified in a blastic plasmacytoid dendritic cell neoplasm by Yoda et al. (Nature Medicine, 2015) and was shown to result in the upregulation of PI3K/Akt/mTOR and MAPK pathways signaling. Interestingly, Yoda et al. also identified functionally similar mutations of GNB1 and GNB2 in several TKI-resistant primary malignancies including a single case with BCR-ABL1+ leukemia. In accordance with these findings we showed that unlike the parental sensitive cells, the resistant ETV6/ABL1+ cells were able to restore PI3K/Akt/mTOR and p44/MAPK signaling after IM treatment. To further confirm the key role of GNB1 K89M mutation in our TKI resistance model we transduced the parental sensitive ETV6/ABL1+ cell line with wild type (WT) GNB1, GNB1-K89M, GNB1-K89E or with empty vector, and studied the impact on TKI sensitivity and kinase signaling. While the cells transduced with WT-GNB1 or with empty vector remained sensitive to TKI, the cells transduced with mutated GNB1 (both K89E and K89M)became resistant to IM, nilotinib, dasatinib and ponatinib. Similarly, while the repeated application of IM resulted in cell growth arrest and cell death in the cells with WT-GNB1 and empty vector, the cells transduced with mutated GNB1 showed only subtle changes in proliferation and viability. Administration of IM resulted in the inhibition of ETV6-ABL1-mediated signaling in all cell lines as demonstrated by diminished phosphorylation of the ETV6-ABL1 direct substrate - CRKL adaptor protein. However, while the IM induced a prolonged inhibition of p44/MAPK and PI3K/Akt/mTOR pathway signaling in the cells with WT-GNB1 and with empty vector, the cells transduced by mutated GNB1 (both K89E and K89M) were able to restore activation of both pathways within 24 hours after IM treatment (similarly to the original multi-TKI resistant cell line). In summary, we showed that the long-term IM treatment of ETV6-ABL1+ leukemia resulted in a gain of TKI-resistance which was driven by the acquired GNB1 mutation activating pro-proliferative and pro-survival signaling, independent on the ETV6-ABL1 kinase activity. Importantly, leukemic cells with mutated GNB1 were resistant not only to IM, but also to the next generation TKI. The frequency of GNB1/GNB2 mutations in primary ETV6-ABL1+ and BCR-ABL1+ leukemias and its clinical relevance is to be determined in the study currently initiated within an international collaboration. Supported by grants GACR P302/12/G10 and GAUK 554214. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 16 (2) ◽  
pp. 106-112 ◽  
Author(s):  
Rossella Gratton ◽  
Paola Maura Tricarico ◽  
Rafael Lima Guimaraes ◽  
Fulvio Celsi ◽  
Sergio Crovella

Background:Lopinavir and Ritonavir (LPV/r) treatment is widely used to prevent HIV mother-to-child transmission. Nevertheless, studies related to the impact of these compounds on patients, in particular in the foetus and newborns, are strictly required due to the controversial findings reported in the literature concerning possible neurologic side effects following the administration of these drugs.Objectives:In our study, we evaluated the impact of LPV/r treatment on the human glioblastoma U- 87 MG cell line.Methods:In order to evaluate the influence of Lopinavir and Ritonavir in terms of oxidative stress (ROS production), mitochondrial morphology and apoptotic cell death, the latter either in the presence or in the absence of caspase-3 and -9 inhibitors, we treated U-87 MG with increasing doses (0.1-1-10-25-50 µM) of Lopinavir and Ritonavir for 24h, either in single formulation or in combination. ROS production was measured by flow cytometry using H2DCFDA dye, mitochondrial morphology was evaluated using MitoRed dye and apoptotic cell death was monitored by flow cytometry using Annexin V-FITC and Propidium Iodide.Results:We observed that co-treatment with Lopinavir and Ritonavir (25 and 50 µM) promoted a significant increase in ROS production, caused mitochondrial network damage and induced apoptosis in a caspase-independent manner.Conclusion:Based on our findings, concordant with others reported in the literature, we hypothesize that LPV/r treatment could not be entirely free from side effects, being aware of the need of validation in in vivo models, necessary to confirm our results.


2018 ◽  
Vol 16 (2) ◽  
pp. 127-137
Author(s):  
Paula Sofia Coutinho Medeiros ◽  
Ana Lúcia Marques Batista de Carvalho ◽  
Cristina Ruano ◽  
Juan Carlos Otero ◽  
Maria Paula Matos Marques

Background: The impact of the ubiquitous dietary phenolic compound p-coumaric acid on human breast cancer cells was assessed, through a multidisciplinary approach: Combined biological assays for cytotoxicity evaluation and biochemical profiling by Raman microspectroscopic analysis in cells. </P><P> Methods: Para-coumaric acid was shown to exert in vitro chemoprotective and antitumor activities, depending on the concentration and cell line probed: a significant anti-invasive ability was detected for the triple-negative MDA-MB-231 cells, while a high pro-oxidant effect was found for the estrogen- dependent MCF-7 cells. A striking cell selectivity was obtained, with a more noticeable outcome on the triple-negative MDA-MB-231 cell line. Results: The main impact on the cellular biochemical profile was verified to be on proteins and lipids, thus justifying the compound´s anti-invasive effect and chemoprotective ability. Conclusion: p-Coumaric acid was thus shown to be a promising chemoprotective/chemotherapeutic agent, particularly against the low prognosis triple-negative human breast adenocarcinoma.


2013 ◽  
Vol 13 (3) ◽  
pp. 414-421 ◽  
Author(s):  
Raquel T. Lima ◽  
Gemma A. Barron ◽  
Joanna A. Grabowska ◽  
Giovanna Bermano ◽  
Simranjeet Kaur ◽  
...  

2018 ◽  
Vol 18 (4) ◽  
pp. 573-582 ◽  
Author(s):  
Khaled R.A. Abdellatif ◽  
Mostafa M. Elbadawi ◽  
Mohammed T. Elsaady ◽  
Amer A. Abd El-Hafeez ◽  
Takashi Fujimura ◽  
...  

Background: Some 2-thioxoimidazolidinones have been reported as anti-prostate and anti-breast cancer agents through their inhibitory activity on topoisomerase I that is considered as a potential chemotherapeutic target. Objective: A new series of 3,5-disubstituted-2-thioxoimidazolidinone derivatives 10a-f and their S-methyl analogs 11a-f were designed, synthesized and evaluated for cytotoxicity against human prostate cancer cell line (PC-3), human breast cancer cell line (MCF-7) and non-cancerous human lung fibroblast cell line (WI-38). </P><P> Results and Method: While compounds 10a-f showed a broad range of activities against PC-3 and MCF-7 cell lines (IC50 = 34.0 – 186.9 and 24.6 – 147.5 µM respectively), the S-methyl analogs 11a-f showed (IC50 = 22.7 – 198.5 and 16.9 – 188.2 µM respectively) in comparison with 5-fluorouracil (IC50 = 60.7 and 40.7 µM respectively). 11c (IC50 = 22.7 and 29.2 µM) and 11f (IC50 = 28.7 and 16.9 µM) were the most potent among all compounds against both PC-3 and MCF-7 respectively with no cytotoxicity against WI-38. Conclusion: The newly synthesized compounds showed good activity against PC-3 and MCF-7 cell lines in comparison with 5-fluorouracil. Compounds 11c and 11f bound with human topoisomerase I similar to its known inhibitors and significantly inhibited its DNA relaxation activity in a dose dependent manner which may rationalize their molecular mechanism as cytotoxic agents.


Sign in / Sign up

Export Citation Format

Share Document