scholarly journals Environmentally Relevant Concentrations of Bisphenol A Interact with Doxorubicin Transcriptional Effects in Human Cell Lines

Toxics ◽  
2019 ◽  
Vol 7 (3) ◽  
pp. 43 ◽  
Author(s):  
Edna Ribeiro ◽  
Mariana Delgadinho ◽  
Miguel Brito

The worldwide production of synthetic chemicals, including endocrine disruptor chemicals (EDCs), such as Bisphenol A (BPA) has increased significantly in the last two decades. Human exposure to BPA, particularly through ingestion, is continuous and ubiquitous. Although, considered a weak environmental estrogen, BPA can induce divergent biological responses through several signaling pathways, including carcinogenesis in hormone-responsive organs. However, and despite the continuous increase of tumor cell-resistance to therapeutic drugs, such as doxorubicin (DOX), information regarding BPA drug interactions is still scarce, although its potential role in chemo-resistance has been suggested. This study aims to assess the potential interactions between environmentally relevant levels of BPA and DOX at a therapeutic dosage on Hep-2 and MRC-5 cell lines transciptome. Transcriptional effects in key-player genes for cancer biology, namely c-fos, p21, and bcl-xl, were evaluated through qRT-PCR. The cellular response was analyzed after exposure to BPA, DOX, or co-exposure to both chemicals. Transcriptional analysis showed that BPA exposure induces upregulation of bcl-xl and endorses an antagonistic non-monotonic response on DOX transcriptional effects. Moreover, the BPA interaction with DOX on c-fos and p21 expression emphasize its cellular specificity and divergent effects. Overall, Hep-2 was more susceptible to BPA effects in a dose-dependent manner while MRC-5 transcriptional levels endorsed a non-monotonic response. Our data indicate that BPA environmental exposure may influence chemotherapy outcomes, which emphasize the urgency for a better understanding of BPA interactions with chemotherapeutic agents, in the context of risk assessment.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4783-4783
Author(s):  
Hirokazu Miki ◽  
Shuji Ozaki ◽  
Osamu Tanaka ◽  
Shiro Fujii ◽  
Shingen Nakamura ◽  
...  

Abstract Multiple myeloma (MM) is a plasma cell malignancy characterized by the accumulation of neoplastic plasma cells in the bone marrow. Although new classes of agents such as thalidomide, lenalidomide, and bortezomib have shown marked anti-MM activity in clinical settings, MM remains an incurable disease due to increased resistance to these agents. Therefore, alternative approaches are necessary to overcome drug resistance in MM. KRN5500 is a new derivative of spicamycin produced by Streptomyces alanosinicus (Kirin Pharma, Tokyo, Japan). This drug potently decreases protein synthesis and inhibits cell growth in human tumor cell lines both in vitro and in vivo. Several phase I studies of KRN5500 were conducted in patients with solid tumors, which showed Cmax values of 1000–3000 nM at the maximum tolerated doses. However, no objective anti-tumor response to KRN5500 alone was observed in these patients. In this study, we examined the anti-tumor activity of KRN5500 against MM cells and evaluated its therapeutic potential in combination with other anti-MM agents. MM cell lines and freshly-isolated MM cells were incubated with various concentrations of KRN5500 for 24 hours. Cell proliferation assay showed marked inhibition of cell growth in MM cells such as RPMI 8226, KMS12-BM, and UTMC-2 (IC50 = 10–40 nM), and U266, MM.1S, and primary MM cells (IC50 = 500–1000 nM). Importantly, a chemotherapy-resistant subclone of RPMI 8226 had a similar sensitivity to KRN5500. Annexin V/propidium iodide staining confirmed that KRN5500 induced apoptosis of MM cells in a dose- and time-dependent manner. Moreover, cleavage of poly (ADP-ribose) polymerase (PARP) was detected after 24 hours with only modest activation of caspase-8, -9, and -3 by immunoblotting. Flow cytometric analysis of anti-apoptotic proteins revealed that apoptosis induced by KRN5500 was associated with down-regulation of Mcl-1 and Bcl-2 expression. To determine the effect of KRN5500 on the unfolded protein response (UPR), splicing of XBP-1 mRNA was analyzed by reverse transcription-polymerase chain reaction. In response to stimulation with KRN5500, splicing of XBP-1 mRNA occurred after 24 hours in RPMI 8226 cells, suggesting that KRN5500-induced apoptosis is mediated in part by the inhibition of UPR. Furthermore, synergistic effects on MM cells were observed when KRN5500 was combined with anti-MM agents including melphalan, dexamethasone, and bortezomib. These results suggest that KRN5500 induces apoptosis in MM cells mainly by the caspase-independent pathway and that its unique mechanism of action provides a valuable therapeutic option to overcome drug resistance in patients with MM.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 523-523
Author(s):  
Leandro Cerchietti ◽  
Maria E. Figueroa ◽  
David Meyers ◽  
Philip A. Cole ◽  
Kapil Bhalla ◽  
...  

Abstract DLBCL is the most common form of non-Hodgkin’s lymphoma. Combinations of untargeted chemotherapeutic agents cure between 40–60% of DLBCL patients. We are interested in the rational design of targeted combinatorial therapy for DLBCL using non-chemotherapy agents. Towards this goal we developed an inhibitor of the BCL6 transcriptional repressor, the most commonly involved oncogene in DLBCL. This BCL6 peptide inhibitor (BPI) causes de-repression of BCL6 target genes and kills DLBCL cells. Since single agent targeted therapy is unlikely to cure tumors, we hypothesized that identification of survival pathways triggered by BPI would facilitate rational design of combinatorial biological therapy for DLBCL. In order to identify such pathways we performed gene expression (GE) microarray studies in ten DLCBL cell lines treated with BPI vs. control. Six cell lines were BCL6 positive and four were BCL6 negative. Only the BCL6 positive cells yielded differences in gene expression. Among BPI induced genes was the p300 histone acetyl-transferase. The overlapping genes among the six cell lines were used to generate a BPI response signature. We used this signature to query the Broad Institute Connectivity Map, which contains the GE signature of 164 distinct small-molecule perturbagens. The top scoring classes of drugs were the histone deacetylase inhibitors (HDIs) and HSP90 inhibitors. Considering that BPI is chemically un-related to HDIs or HSP90 inhibitors and that BPI induces p300, we hypothesized that a major biological effect of BPI is to cause the acetylation of HSP90 (which inhibits Hsp90 pro-survival activity) and p53, (which enhances its pro-apoptotic activity). We verified that p300 is a direct BCL6 target gene by ChIP assays, that BPI induces p300 mRNA and protein by QPCR and western blot, and that p300 is silenced in most primary DLBCLs at both the mRNA and protein levels. Accordingly, BPI induced acetylation of Hsp90 and inhibited its function, as demonstrated by the decrease in the abundance of Hsp90 client proteins (AKT/PKB and c-raf). BPI also induced acetylation and functional activity of p53 in a p300-dependent manner (and also induces p53 expression). The importance of p300 was confirmed since a p300-dominant negative construct and the specific p300(HAT) inhibitor Lys-CoA-TAT could block BPI antilymphoma activity. Remarkably, we observed a dose-sequence dependent synergistic effect of BPI followed by Hsp90 inhibitors in killing DLBCL cells. Hsp90 is a relevant target in DLBCL since HSP90?/? protein was expressed in ∼90% of DLBCL patients (n=70). HDIs also increase acetylation of Hsp90 and p53. The HDI drugs SAHA, valproic acid and TSA all profoundly synergized with BPI to specifically eradicate BCL6 positive DLBCL cell lines. In conclusion, we discovered an unexpected mechanistic link between BCL6 and suppression of protein acetylation in lymphomagenesis. This information was harnessed for the rational design of synergistic targeted therapy with BCL6 inhibitors followed by Hsp90 or HDAC inhibitors to target cellular pathways induced by BPI. We anticipate that these drug combinations will result in more potent and less toxic therapeutic treatment of DLBCL, possibly with less or no added chemotherapy.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4794-4794
Author(s):  
Renate Burger ◽  
Hanna Czekalla ◽  
Tanja Ahrens ◽  
Kathrin Richter ◽  
Andreas Guenther ◽  
...  

Abstract Dietary plant polyphenols are known to have antitumor, antiinflammatory, and antioxidant activity and as such may sensitize tumor cells to chemotherapeutic agents. We have evaluated the effects of the green tea polyphenol epigallocatechin gallate (EGCG) alone and in combination with other drugs on human myeloma cells. The compound is currently under investigation in several phase I/II clinical trials including for treatment of patients with early stage chronic lymphocytic leukemia. EGCG inhibited the in vitro growth of human myeloma cell lines in a time and dose-dependent manner. IC50 concentrations were between 12.5 μM and 50 μM as measured in a colorimetric tetrazolium (MTS) based assay and by trypan blue exclusion. Excess amounts of IL-6, bone marrow stromal cells, or overexpression of Mcl-1 and Bcl-xL could not protect from EGCG induced cytotoxicity. Pretreatment of INA-6 cells with EGCG resulted in a dose-dependent inhibition of IL-6 induced STAT3 tyrosine phosphorylation. In accordance with the essential role of STAT3 for INA-6 cell survival, EGCG induced apoptosis as determined by flow cytometry upon 7-amino-actinomycin D/annexin-V staining. In cell lines not dependent on exogenous IL-6, EGCG induced growth inhibition was abolished by pretreating the cells with 200 U/ml catalase, an enzyme which reduces reactive oxygen species (ROS). The combination of EGCG with doxorubicin, dexamethason, or rapamycin did not result in increased growth inhibition. In contrast, growth inhibition by bortezomib was antagonized with EGCG at concentrations that were not inhibitory when used alone (1–10 μM). In conclusion, EGCG exerts its effects on myeloma cells through several mechanisms including inhibition of IL-6 mediated signalling pathways via STAT3 and induction of oxidative stress. Notably, at pharmacologically achievable concentrations, EGCG antagonized bortezomib activity. Thus, the intake of natural polyphenols (high consumption of green tea or taking green tea extracts) may be critical during therapy with bortezomib.


Pharmacia ◽  
2021 ◽  
Vol 68 (4) ◽  
pp. 779-787
Author(s):  
Sahar M. AlMotwaa

The anticancer effects of chemotherapeutic agents may be accentuated, and their side effects minimized by combining them with essential oils in nanocarrier systems. This study aimed to incorporate ifosfamide (IF) into nanoemulsion-based clove oil (IF-CLV). The nano-emulsion (NE) formulas were characterized with Zetasizer. The cytotoxicity of the formulated NEs against cervical (HeLa) and breast (MCF-7) cancer cell lines was determined using MTT assay, light microscopy, and DAPI staining. The z – average diameters of NE-CLV and IF-CLV were 63.1±1.00 and 89.4±2.64 nm, while Zeta potential values were – 4.39±0.4 and – 11.65±1.1mV, respectively. Cytotoxicity studies revealed that relative to free IF, NE-CLV and IF-CLV were highly toxic on HeLa and MCF-7cells, in a dose-dependent manner. The half-maximal inhibitory concentration (IC50) values of NE-CLV against HeLa and MCF-7 have decreased 38 and 27 folds, while the corresponding IC50 values of IF-CLV have decreased 57 and 35 folds, respectively. These results suggest that the incorporation of IF into NE-based clove oil produces potent therapeutic effects against cancer cell lines.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4225-4225
Author(s):  
Benet Pera ◽  
Michel Jordi ◽  
Ruben Hoya-Arias ◽  
José Fernando Díaz ◽  
Weishuo Fang ◽  
...  

Abstract Poor outcomes in patients with acute myeloid leukemia (AML) are related to the high proportion of patients with chemorefractory disease or relapse after an initial response due to the development of resistance. This treatment failure is frequently due to the persistence of a cell population that is inherently resistant to classical chemotherapeutic agents through different mechanisms, in which an increased efflux of these agents via transmembrane proteins of the ATP-binding cassette (ABC) family is one of the best recognized. Classically, the main approach to overcome the transport effect has involved the co-administration of competitive inhibitors of these transporters. However, the large number of failed clinical trials involving these inhibitors has demonstrated the necessity to adopt different strategies. We therefore hypothesized that molecules with covalent binding and/or high binding affinities well above their affinities for ABC-transporters will overcome the transport efflux, allowing AML cells to recover drug-sensitivity. To test this we have evaluated the anti-neoplastic activity of a series of fourteen synthetic paclitaxel analogues called chitaxes (CTXs), as well as the marine compound with tubulin-covalent binding ability zampanolide (ZMP) and seven novel synthetic derivatives. The CTXs compounds were designed combining the most favorable substitutions that increased the binding free energies up to 500-fold the paclitaxel binding affinity. For the ZMP derivatives, the overall rationale was to produce structurally simplified analogs that would be easier to synthesize and also be chemically more stable than ZMP. First we determined the GI50,72h of the studied molecules in MV4-11 and HL-60 AML-cell lines using alamarBlue, resulting all of them active in the subnanomolar to low-micromolar range. ZMP and CTX number 40 (CTX-40) presented the highest activities with GI50,72h values of 0.16±0.03 nM in MV4-11 and 0.38±0.2 nM in HL-60, and 0.22±0.03 nM in MV4-11 and 0.27±0.04 nM in HL-60, respectively. With the exception of CTXs number 6, 30, 34 and 35, which shown GI50,72h values around 10 nM, the remaining paclitaxel derivatives shown values around 1 nM. On the other hand, the novel ZMP analogues presented activities from 30 nM to 2 μM. ZMP and CTX-40 were the more active compounds of the studied series, and they also shown activities around 8, 40 and 80 times higher than paclitaxel, daunorubicin and cytarabine, respectively. We then performed an early-apoptosis evaluation, treating the cells with ZMP or CTX-40 at their GI50,72h and staining with 7-AAD/Annexin-V, and we determined that more than 75 % of the cells entered apoptosis after 48 h of drug-incubation. Cell cycle analysis revealed that the addition of ZMP or CTX-40 induced a shift from G1 to G2/M as early as 12 h after treatment in a concentration dependent manner. In order to validate our working hypothesis, we evaluated the activity of ZMP and CTX-40 in resistant cells with P-glycoprotein overexpression, and we obtained low resistance indexes (GI50,72h resistant / GI50,72h sensitive) that revealed both compounds to be active in such resistant cells. To investigate the effect of these molecules on normal human hematopoietic progenitors and stem cells, we performed colony forming unit (CFU) assays and cobblestone area-forming (CAFC) assays, respectively. ZMP seems to be more active than CTX-40 in inhibiting progenitor differentiation as well as the growth of hematopoietic stem cells, however both compounds allowed the survival of these normal cells in the presence of drug concentrations up to 2 fold of their GI50,72h values. We are now evaluating the efficacy of these molecules on leukemic cells and their toxic effects on the healthy hematopoietic cells at the same time, by co-culturing HL-60 GFP-transduced cells with cord blood-CD34+ cells and distinguishing the leukemic colonies from the healthy ones under fluorescence microscopy. We are also exploring different combinations of ZMP or CTX-40 with daunorubicin or cytarabine, with the aim of identifying a combination that could allow reduction of the dose of the approved drug and increase the therapeutic window. Our data presents ZMP and CTX-40 as candidates to be considered for therapeutic use in AML, not only due their promising in vitro activity but also because of their ability to retain such activity in resistant cell lines associated with P-glycoprotein overexpression. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e17035-e17035
Author(s):  
Jason Yap ◽  
Raji Ganesan ◽  
Richard Fox ◽  
Christopher Dawson ◽  
Sean Kehoe

e17035 Background: Vulval squamous cell carcinoma (VSCC) is predominantly a disease of the elderly; with three-quarters of cases occurring in those aged over 60 years. Currently, chemotherapy is only used as an adjunct to surgery in advanced disease or for palliation. Unfortunately, the treatment response of VSCC to chemotherapies is variable, and toxicity remains a major problem, rendering it unsuitable for most patients. Dysregulation of the Hedgehog (HH) pathway has been described in many cancers, and HH inhibitors are being employed for the treatment of many cancers displaying aberrant pathway activation. To this end, we set out to evaluate 1) whether the HH pathway is dysregulated in VSCC and 2) the therapeutic utility of using HH inhibitors in the treatment of VSCC. Methods: Immunohistochemical staining of HH pathway components was performed on formalin-fixed paraffin-embedded sections of a well-characterized cohort of 201 patients with primary VSCC. Expression of HH pathway proteins was then correlated with clinico-pathological data, disease recurrence and survival. A panel of VSCC-derived cell lines was examined for HH pathway activity using an HH-GLI luciferase reporter assay. The effects of HH inhibitors on the growth and viability of VSCC cell lines, alone and in combination with cisplatin, was examined by BrdU label incorporation and XTT viability assays. Results: The HH signaling pathway was found to be over-expressed in approximately 80% of VSCC cases. Moreover, patients whose tumours displayed Sonic HH (SHH) ligand overexpression were more likely to develop a local recurrence within 3 years. Two HH inhibitors, Itraconazole and LDE225, not only inhibited the growth of VSCC cell lines in a dose-dependent manner, but also sensitized cells to the genotoxic affects of Cisplatin. Conclusions: We have shown for the first time that HH pathway is overexpressed in VSCC and that SHH overexpression may serve as a useful biomarker to predict local disease recurrence. In addition, results from our laboratory studies have shown that HH inhibitors may be used as a neo-adjuvant therapy to augment the actions of chemotherapeutic agents in this disease.


Molecules ◽  
2020 ◽  
Vol 25 (21) ◽  
pp. 4940
Author(s):  
Zviadi Aburjania ◽  
Jason D. Whitt ◽  
Samuel Jang ◽  
Dwayaja H. Nadkarni ◽  
Herbert Chen ◽  
...  

In an effort to discover viable systemic chemotherapeutic agents for neuroendocrine tumors (NETs), we screened a small library of 18 drug-like compounds obtained from the Velu lab against pulmonary (H727) and thyroid (MZ-CRC-1 and TT) neuroendocrine tumor-derived cell lines. Two potent lead compounds (DHN-II-84 and DHN-III-14) identified from this screening were found to be analogs of the natural product makaluvamine. We further characterized the antitumor activities of these two compounds using pulmonary (H727), thyroid (MZ-CRC-1) and pancreatic (BON) neuroendocrine tumor cell lines. Flow cytometry showed a dose-dependent increase in apoptosis in all cell lines. Induction of apoptosis with these compounds was also supported by the decrease in myeloid cell leukemia-1 (MCL-1) and X-chromosome linked inhibitor of apoptosis (XIAP) detected by Western blot. Compound treatment decreased NET markers chromogranin A (CgA) and achaete-scute homolog 1 (ASCL1) in a dose-dependent manner. Moreover, the gene expression analysis showed that the compound treatment reduced c-Kit proto-oncogene expression in the NET cell lines. Induction of apoptosis could also have been caused by the inhibition of c-Kit expression, in addition to the known mechanisms such as damage of DNA by topoisomerase II inhibition for this class of compounds. In summary, makaluvamine analogs DHN-II-84 and DHN-III-14 induced apoptosis, decreased neuroendocrine tumor markers, and showed promising antitumor activity in pulmonary, thyroid, and pancreatic NET cell lines, and hold potential to be developed as an effective treatment to combat neuroendocrine tumors.


2018 ◽  
Author(s):  
Michel Owusu ◽  
Peter Bannauer ◽  
Athanasios Mourikis ◽  
Alistair Jones ◽  
Joana Ferreira da Silva ◽  
...  

SummaryWe provide a catalog for the effects of the human kinome on cell survival in response to DNA damaging agents, selected to cover all major DNA repair pathways. By treating 313 kinase-deficient cell lines with ten diverse DNA damaging agents, including seven commonly used chemotherapeutics, we were able to identify kinase specific vulnerabilities and resistances. In order to identify novel synthetic lethal interactions, we investigate the cellular response to carmustine for 25 cell lines, by establishing a phenotypic FACS assay designed to mechanistically investigate and validate gene-drug interactions. We show apoptosis, cell cycle, DNA damage and proliferation after alkylation or crosslink-induced damage for selected cell lines and rescue the cellular sensitivity of DYRK4, EPHB6, MARK3, PNCK as a proof of principle for our study. Our data suggest that some cancers with inactivated DYRK4, EPHB6, MARK3 or PNCK gene could be particularly vulnerable to treatment by alkylating chemotherapeutic agents carmustine or temozolomide.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3342-3342
Author(s):  
Hye-Ran Kim ◽  
Myung-Geun Shin ◽  
Sun-Woo Lee ◽  
Hee-Nam Kim ◽  
Il-Kwon Lee ◽  
...  

Abstract In light of the recent studies that showed significant causal relationship between mitochondrial genetic mutation and apoptosis in pathology of various tumors and degenerative diseases, mitochondrial proteins have been interesting targets for the study of apoptosis in leukemia and other malignancies. This observation prompted us to analyze mitochondrial proteins and develop new anti-proliferative agents targeting them. Mitochondria were isolated from AML cells by density-gradient ultracentrifugation using swelling buffer and sucrose buffer. We identified 48 spots corresponding to 38 proteins in primary AML cells using 2-DE and mass spectrometry (MALDI-TOF/TOF technology), the expression of which were altered significantly compared to that of normal hematopoietic cells. Out of these deregulated proteins, 12 and 20 proteins were observed in up- or down-regulated spots, respectively. Interestingly, prohibitin (PHB) (gi4505773) was highly expressed in primary AML cells, which was confirmed by Western blot, immunohistochemistry and immunofluorecenct study in the primary AML bone marrow cells and sections. To assess the functional significance of aberrant prohibitin expression, we applied siRNA delivery for silencing of prohibitin. Transduction with a siRNA 11867 construct resulted in 75% decrease of AML cells as compared to the nonsilencing control construct after two days. Potent chemical substances that can alkylate PHB in AML cells, cyclohexylphenyl-chloroethyl urea (CCEU) and iodophenyl-chloroethyl urea (ICEU), were synthesized in our laboratory. Time and dose dependent manner of proliferation suppression when treated with CCEU and ICEU was observed in leukemic cell lines including THP-1, K-562 and Kasumi-1. Moreover, notable morphological transformation of leukemic cells was observed when treated with 50 – 200 umol of CCEU and ICEU for 24 hours. Cell cycle analysis of CCEU-and-ICEU-treated- THP-1 and Kasumi-1 cell lines showed a remarkable increase of the sub-G1 phase. Immunoblotting experiment revealed the change of cytoplasmic and nucleoplasmic PHB in K-562 cell line. Expression of cleaved caspase3 and poly ADP-ribose polymerases were also observed to have increased in primary AML cells and cell lines. By analyzing AML cell mitochondrial protein we discovered a new molecular marker, PHB, characteristically overexpressed in AML cells and developed new anti-cancer agents such as CCEU and ICEU that target against PHB in AML cells.


2020 ◽  
Vol 20 (3) ◽  
pp. 301-314 ◽  
Author(s):  
Leonard Barasa ◽  
Hari P. Vemana ◽  
Nirupama Surubhotla ◽  
Sin S. Ha ◽  
Jing Kong ◽  
...  

Background and Objective : Drug resistance and adverse effects are immense healthcare challenges in cancer therapy. Benzimidazole ring-based small molecules have been effective anticancer agents in drug development. In an effort to develop novel chemotherapeutics, we synthesized and assessed the anticancer and antibacterial activities of a small library of structurally unique benzimidazoles. Methods : The benzimidazoles were derived from indole, N-alkyl indole, fatty acid, and alpha-amino acid scaffolds providing a panel of diverse structures. The compounds were tested in three different cancer cell lines for cytotoxicity: HepG2 (human hepatocellular carcinoma), HeLa (human cervical carcinoma), and A549 (human lung carcinoma). Mechanism of cell death induced by benzimidazoles was evaluated using fluorescent dye-based apoptosis-necrosis assay, immunoblotting for active caspases, topoisomerase-II activity assay, and cell cycle assay. Results : Cell viability testing revealed that indole- and fatty acid-based benzimidazoles were most potent followed by the amino acid derivatives. Many compounds induced cytotoxicity in a concentration-dependent manner with cellular cytotoxicity (CC50) <20μM in the cell lines tested. Most compounds exhibited cytotoxicity via apoptosis through the intrinsic pathway. Inhibition of topoisomerase activity and cell cycle alterations were not the primary mechanisms of cytotoxicity. In addition, several compounds showed promising activity against S. aureus and S. epidermidis (Minimum Inhibitory Concentration (MIC) of as low as 0.04μmol/mL). Conclusion: The reported benzimidazole derivatives possess promising anticancer and antibacterial properties. Additionally, we discovered apoptosis to be the primary mechanism for cancer cell death induced by the tested benzimidazoles. Our findings suggest that further development of these scaffolds could provide drug leads towards new chemotherapeutics.


Sign in / Sign up

Export Citation Format

Share Document