scholarly journals Two novel anticancer compounds with minimum cardiotoxic property

2020 ◽  
Author(s):  
Tayebeh Afsharirad ◽  
Raheleh Tahmasvand ◽  
Mohsen Amini ◽  
Bahram Daraei ◽  
Mona Salimi

Abstract Background: Although two novel synthesized compounds with tri-aryl structures (A and B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. Methods: Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. Results: MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 µM) at 24 and 48 hours. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. Conclusions: Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.

2020 ◽  
Author(s):  
Tayebeh Afsharirad ◽  
Raheleh Tahmasvand ◽  
Mohsen Amini ◽  
Bahram Daraei ◽  
Mona Salimi

Abstract Background: Although two novel synthesized compounds with tri-aryl structures; 3-(4-chlorophenyl)-5-(4-fluorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (A) and 3,5-bis-(4-chlorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. Methods: Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. Results: MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 µM) at 24 and 48 hours. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. Conclusions: Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.


2020 ◽  
Author(s):  
Tayebeh Afsharirad ◽  
Raheleh Tahmasvand ◽  
Mohsen Amini ◽  
Bahram Daraei ◽  
Mona Salimi

Abstract Background: Although two novel synthesized compounds with tri-aryl structures; 3-(4-chlorophenyl)-5-(4-fluorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (A) and 3,5-bis-(4-chlorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. Methods: Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. Results: MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 µM) at 24 and 48 hours. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. Conclusions: Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.


2020 ◽  
Vol 21 (1) ◽  
Author(s):  
Tayebeh Afsharirad ◽  
Raheleh Tahmasvand ◽  
Mohsen Amini ◽  
Bahram Daraei ◽  
Mona Salimi

Abstract Background Although two novel synthesized compounds with tri-aryl structures; 3-(4-chlorophenyl)-5-(4-fluorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (A) and 3,5-bis-(4-chlorophenyl)-4-phenyl-4,5-dihydro-1,2,4-oxadiazole (B) have been previously demonstrated to possess remarkable anti-breast cancer activity, their cardiotoxicity remains a major concern due to their mechanism of action. To address this concern, we assessed the ability of these compounds to cause toxicity towards H9c2 cardiomyocytes as an in vitro model of cardiotoxicity. Methods Cytotoxic activity of both compounds was explored in vitro on H9c2 cells using MTT assay. Annexin V/PI method, intracellular ROS determination and mitochondrial membrane potential assay were applied to elucidate the mechanism of action of the cell death. Results MTT assay revealed a concentration- and time-dependent cardiotoxicity. Findings of apoptosis by double staining with annexin V and propidium iodide divulged no cell death including apoptosis and necrosis at the concentration that were effective to inhibit cancer cells proliferation (10 μM) at 24 and 48 h. Furthermore, flow cytometric measurement of membrane potential and ROS determination using DCFH-DA verified the safe concentration of the compounds against H9c2 cells with no cardiotoxic effect. However, the higher concentration of the compounds could induce cell death through ROS-mediated mitochondrial dysfunction. Conclusions Altogether, the results represented two novel chemical molecules possessing anti-breast cancer activity with minimum cardiac side effect.


2013 ◽  
Vol 2 (2) ◽  
pp. 80 ◽  
Author(s):  
Dessy Arisanty

AbstrakSuatu senyawa obat dapat menjadi kemoterapi kanker adalah dengan cara menskrining terlebih dahulu tumbuhan obat yang berpotensi sebagai obat antikanker. Salah satunya adalah tanaman obat daun nimba (Azadirachta indica L.Juss) yang terbukti secara significant menyebabkan apoptosis pada beberapa jenis sel line kanker. Dalam penelitian ini, ekstrak ethanol dari A. indica dipelajari untuk melihat efeknya pada pertumbuhan sel kanker payudara manusia jenis MDA-MB-231 dengan menggunakan tes untuk proliferasi yaitu MTT assai dan untuk mengetahui perubahan morphologi dari apoptosis selnya dengan menggunakan TUNEL assay Ekstrak daun nimba (A. indica) dapat menurunkan keberadaan jumlah sel kanker dengan cara menghambat perkembangan daripada sel tersebut dan menginduksi proses apoptosis pada sel kanker tersebut. Hasil pemeriksaan MTT assai didapatkan nilai IC50 nya adalah 55 ug / mL. Kematian MDA-MB231 sel yang disebabkan oleh ekstrak daun nimba (A.indica) ditemukan melalui mekanisme apoptosis yang secara morfologinya menunjukan ciri ciri dari kematian secara apoptosis seperti kondensasi dari nucleus, membrane nukleus yang melebur dan akhirnya terjadinya fragmentasi dari DNA. Analisis struktur dalaman sel juga mengungkapkan karakteristik apoptosis yaitu marginasi dari kromosom yang disertai dengan fragmentasi DNA dan selanjutnya akan terbentuk badan apoptotik pada sel kanker yang diinkubasi dengan ekstrak tersebut. Pada penelitian ini juga dijumpai peningkatan jumlah sel apoptosis dari hari 1 sampai hari 3 inkubasi oleh ekstrak nimba. Ekstrak ethanol A.indica mungkin mengandung senyawa bioaktif(s) yang menyebabkan kanker payudara MDA-MNB 231 mengalami kematian sel secara apoptosis. Penelitian lebih lanjut masih diperlukan untuk mengetahui mekanisme tumbuhan ini membunuh sel kanker MDA-MB 231.Kata kunci: Studi In vitro, Azadirachta indica, apoptosis, TUNEL assayAbstractA screening is conducted on plants that have potential as anticancer is a promising way for discovering novel chemotherapeutic compound. A medicinal plant neem leaf (Azadirachta indica L.Juss) intake has been shown to induce significant levels of apoptosis in various cancer cells. In this present study, ethanol extract of Azadirachta indica was studied for its effects on growth in MDA-MB 231 human breast cancer cells using assays for proliferation (MTT assay) and mechanisme of cell apoptosis using TUNEL assay. Neem leaf extract decreased cell viability, inhibited cell proliferation, and induced cell apoptosis. Result of MTT assay was 55 μg/mL of neem remarkably reduced cell viability of MDA-MB 231 cells. MDA-MB231 cell death elicited by the extract was found to be apoptotic in nature based the indication of nucleus condensation, shrinkage of nucleus membrane and also DNA fragmentation which are a hallmark of apoptosis. In addition, ultrastructural analysis also revealed apoptotic characteristics which are the presence of chromatin margination and apoptotic bodies in the extract-treated cells. There was an increase in the number of apoptotic cells from day 1 to day 3 post incubation with neem extract. Thus, the results from this study strongly suggest that the ethanol extract of A.indica may contain bioactive compound(s) that caused breast carcinoma, MDA-MNB 231 cell death by apoptosis. It’s needed to do advance research to know more deeply the mechanism this plant on breast cancer cell line MDA-MB.Keywords:In vitro study, Azadirachta indica, apoptosis, TUNEL assay


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3471-3471
Author(s):  
Amy Johnson ◽  
Lisa Smith ◽  
Jiuxiang Zhu ◽  
Nyla Heerema ◽  
Sara Guster ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) is an incurable adult leukemia characterized by disrupted apoptosis. While the majority of patients with CLL are asymptomatic at diagnosis, most progress and require therapy. Identification of new targets and therapeutic agents is therefore a high priority for the treatment of CLL. Synthetic chemistry yielded derivatives of the COX-2 inhibitor, celecoxib, with increased ability to induce apoptosis in the 1–10 μ M range in prostate cancer cells, a similar proposed mechanism of action, and increased in vivo activity in a murine prostate cancer xenograft model. Based upon these data, a Rapid Access to Intervention Development (RAID) proposal is underway to generate OSU03012 for clinical studies in prostate cancer. In addition, we are examining the biologic effects of these new agents in primary CLL cells and lymphoblastic cell lines, showing a novel mechanism of cell killing independent of caspase activation and bcl-2 over-expression. To determine the in vitro activity against CLL cells, 11 CLL patient PBMCs were incubated in various concentrations of OSU03012. The LC50 at 24 hrs was 7.12μM and decreased to 5.45μM at 72 hrs. We show both early (annexin-V positive) and late (both annexin-V/PI positive) apoptosis concurrent with loss of mitochondrial membrane potential typical of apoptosis. These data suggest OSU03012 is highly cytotoxic toward CLL cells in vitro at doses well below those attainable without toxicity in a murine model. Additionally, we show that OSU03012 mediates apoptosis by activation of the intrinsic, mitochondrial pathway of apoptosis but also activates alternative caspase independent cell death pathways. CLL cells from 8 patients were incubated in 10μM OSU03012 for 24 hrs and assessed for caspase-3 and PARP. Immunoblots reveal a dose dependent increase in active caspase-3 concurrent with a decrease in the pro-form. This occurred concurrently with the appearance of the 85 kD cleaved product of PARP that is a known downstream target of caspase-3. In the same 8 patient lysates we saw no change in the inactive pro-form of caspase-8, but consistent processing of caspase-9. These data suggest that OSU03012 in part utilizes the intrinsic pathway of apoptosis to promote CLL cell death. Incubation of CLL cells with z-VAD-fmk and OSU03012 did not abrogate cell death, but eliminated processing of caspase-9, caspase-3 and PARP, suggesting that this agent also activates caspase independent mechanisms of cell death. Given the caspase dependent and independent pathways utilized by OSU03012, we assessed the dependence of cell death on bcl-2 expression. Here we show that bcl-2 over-expression in the 697 lymphoblastic cell line greatly diminishes the apoptosis observed with fludarabine, but potent apoptosis is equally observed with OSU03012 compared to the empty vector cell line. Furthermore, in the bcl-2 over-expressing cell line, caspase-3 and PARP cleavage was not observed despite equivalent apoptosis supporting further multiple mechanisms of cell killing induced by OSU03012. In summary, OSU03012 is an oral bioavailable therapeutic agent that has potent in vitro activity against primary CLL cells. This cytotoxicity is mediated by both caspase dependent and independent pathways and can overcome bcl-2 over-expression. These data provide support for further investigation of the mechanism of action of OSU03012 in CLL cells and performance of early Phase I studies in CLL as part of the RAID process.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2098-2098
Author(s):  
David M. Lucas ◽  
Syed-Rehan A. Hussain ◽  
Amy J. Johnson ◽  
Lisa L. Smith ◽  
Amy J. Wagner ◽  
...  

Abstract We have noted impressive activity of the cyclin-dependent kinase inhibitor flavopiridol in advanced-stage CLL patients, including those with a deletion of chromosome 17p13. Using a novel, effective schedule of flavopiridol, substantial and sometimes dramatic evidence of tumor cell death is observed as early as 4–6 hours. This is accompanied by hyperkalemia, hyperphosphatemia, hypocalcemia, and dramatic elevation in LDH consistent with acute tumor lysis syndrome. Studies by several groups including our own have demonstrated that Mcl-1 protein and mRNA is down-regulated with flavopiridol. Mcl-1 is an important protein that contributes to mitochondrial membrane stability. We have therefore sought to determine the role of mitochondria disruption in the mechanism of action of flavopiridol. By flow cytometry using the voltage-sensitive dye JC-1, loss of mitochondrial membrane potential was detected in flavopiridol-treated whole blood as early as five hours, prior to the onset of annexin-V or propidium iodide staining. This is in contrast to in vitro studies using human serum, in which mitochondrial depolarization and annexin-V staining occurred simultaneously. In isolated CLL cells treated with flavopiridol in vitro, loss of mitochondrial membrane potential was not affected by inhibitors of caspases 8 or 9 or by the broad caspase inhibitor Z-VAD-fmk, although apoptosis was effectively blocked by Z-VAD-fmk and caspase-8 inhibitor, and to a lesser extent, caspase-9 inhibitor. Flavopiridol was also able to effectively induce apoptosis and mitochondrial membrane depolarization in Jurkat cell lines deficient in caspase-8 or its adapter protein FADD. Additionally, lymphoid cells overexpressing Bcl-2 are resistant to flavopiridol-mediated apoptosis relative to the vector control. This suggests there is not direct binding of flavopiridol or its metabolites to APAF-1 (cytosolic adapter protein) and apoptosome assembly, as this process is insensitive to Bcl-2 family proteins. Further mechanistic studies were undertaken using isolated liver mitochondria. While the electron transport system was not uncoupled in this system, potential mechanisms of mitochondrial injury in leukemic cells from CLL patients are currently under exploration. Taken together, these observations suggest that mitochondrial perturbation contributes significantly to the death process induced by flavopiridol. Further studies to identify the mechanism of mitochondrial perturbation will be essential to understanding flavopiridol’s mechanism of action and for predicting patients at risk for acute tumor lysis syndrome. (Support for this work was provided by the Samuel Waxman Foundation and the Leukemia & Lymphoma Society.)


2020 ◽  
Vol 20 (4) ◽  
pp. 437-449
Author(s):  
Jubie Selvaraj ◽  
Jameera B.A. John ◽  
Nanjan M. Joghee ◽  
Justin Antony ◽  
Ashish Wadhwani ◽  
...  

Background: : Current drugs used for the treatment of hormone-dependent breast cancer function as anti-estrogens in the breast, in addition to Estrogen Receptor (ER) agonists in the uterus, thus elevate a woman’s risk of developing uterine cancer. This is due to the lack of selective binding and partial agonistic effect of these drugs towards estrogen receptors. In recent years, therefore, researchers have turned their attention towards antiestrogens devoid of these agonist properties and thus have a mechanism of action different from the existing drugs. Objective:: In this context, we report here the design, development and in vitro evaluation of some novel pharmacophores containing coumarin and fatty acid scaffolds for their anti-breast cancer activity. Methods: : A library of coumarin-fatty acid conjugates was designed using structure-based drug design approach. The conjugates which have shown good in silico results were then synthesized, characterized and evaluated for their anti-breast cancer activity by MTT assay, Apoptotic assay, Cell proliferation assay, Estrogen binding assay and Gene expression study. Results: Out of the fifteen compounds screened, two compounds, SAC-2 and LNAC-2, showed good activity with IC50 values 22µg/ml, 25μg/ml, respectively. These compounds suppressed the proliferation of ER overexpressed MCF-7 cells, increased ERα degradation and hence inactivate the ERα pathway. ER binding assay and gene expression RT-PCR study reveal that SAC-2 downregulated the expression of ERα receptor and AKT-1 gene. Conclusion:: Compound SAC-2 is a good antagonist to ER and hence has a potential for treating breast cancer and other cancers where AKT plays an important role.


2021 ◽  
Vol 22 (11) ◽  
pp. 5782
Author(s):  
Ashwini Makhale ◽  
Devathri Nanayakkara ◽  
Prahlad Raninga ◽  
Kum Kum Khanna ◽  
Murugan Kalimutho

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer lacking targeted therapy. Here, we evaluated the anti-cancer activity of APR-246, a P53 activator, and CX-5461, a RNA polymerase I inhibitor, in the treatment of TNBC cells. We tested the efficacy of individual and combination therapy of CX-5461 and APR-246 in vitro, using a panel of breast cancer cell lines. Using publicly available breast cancer datasets, we found that components of RNA Pol I are predominately upregulated in basal-like breast cancer, compared to other subtypes, and this upregulation is associated with poor overall and relapse-free survival. Notably, we found that the treatment of breast cancer cells lines with CX-5461 significantly hampered cell proliferation and synergistically enhanced the efficacy of APR-246. The combination treatment significantly induced apoptosis that is associated with cleaved PARP and Caspase 3 along with Annexin V positivity. Likewise, we also found that combination treatment significantly induced DNA damage and replication stress in these cells. Our data provide a novel combination strategy by utilizing APR-246 in combination CX-5461 in killing TNBC cells that can be further developed into more effective therapy in TNBC therapeutic armamentarium.


Author(s):  
I.V. Leshkova ◽  
◽  
O.V. Dolgih ◽  
O.YU. Ustinova

Abstract. Introduction. The protection of the reproductive health of the working-age population is the most important direction of State policy. In 5-15% of cases, the causes of reproductive dysfunction are immunological disorders. Benzene belongs to the group of industrial reprotoxicants, however, its effect of benzene on the reproductive system has not been sufficiently studied. Objective: to study the immunological aspects of the effect of benzene on the reproductive system. Methods. We examined 50 men exposed to benzene with reproductive disorders (26-49 years old), as well as 4 workers with normal sexual function aged 53-60 years. Spontaneous and induced changes in the cellular expression of apoptosis markers were studied. For the study, the ANNEXIN V-FITC/7-AAD kit was used for the detection of cells that have undergone apoptosis. The experiment was conducted in vitro using a biological medium (ejaculate). A factor of the chemical nature was benzene. Results. According to the results of the comparative analysis, there were no significant deviations of pathogenetic tests of immunological markers in comparison with the reference level in the spontaneous expression samples, but there was an excess of expression of the CD95 + cell death receptor (p<0.05) in 30% of the samples examined, and a decrease in the number of Annexin V-FITC+7AAD negative cells (without reaching the significance level) in samples with a load of (15%). There was a difference in the expression levels of CD95+ and CD25+ CD-reception indicators by 20% and 10% in relation to the spontaneous level (p<0.05). Representatives of the chemical group of aromatic hydrocarbons realize reprotoxicity, using the mechanism of excessive induction of the membrane signaling of the cell death receptor, accelerate the natural program of cell death by approximately 20% compared to the state of reproductive cells that were not stimulated. Conclusion. At the present stage, one of the tasks of occupational medicine is to study the effect of chemicals on the processes of reproduction, to develop new approaches to assessing the risk of their impact on the reproductive health of workers.


Sign in / Sign up

Export Citation Format

Share Document