Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry - Anti-Cancer Agents) - Volume 25, Issue 13, 2025
Volume 25, Issue 13, 2025
-
-
Cucurbitacin E Glucoside as an Apoptosis Inducer in Melanoma Cancer Cells by Modulating AMPK/PGK1/PKM2 Pathway
BackgroundCucurbitacin E glucoside (CEG), a prominent constituent of Cucurbitaceae plants, exhibits notable effects on cancer cell behavior, including inhibition of invasion and migration, achieved through mechanisms such as apoptosis induction, autophagy, cell cycle arrest, and disruption of the actin cytoskeleton.
ObjectiveMelanoma, the fastest-growing malignancy among young individuals in the United States and the predominant cancer among young adults aged 25 to 29, poses a significant health threat.
AimThis study aims to elucidate the apoptotic mechanism of CEG against the melanoma cancer cell line (A375).
MethodsThe study estimated the IC50 of CEG against the A375 cell line and assessed cell viability, apoptosis, and necrosis upon CEG treatment. Additionally, IC50 values of CEG against Phosphoglycerate kinase1 (PGK1) and Pyruvate Kinase M2 (PKM2) were determined at various levels of concentrations. The impact of CEG on intracellular glutathione (GSH) levels and the activity of key enzymes (GR, SOD, GPx, CAT), as well as markers of apoptosis (p53), and cell cycle regulation (cyclin D1, cyclin E2, cdk2, cdk4), were estimated. Finally, the level of AMP-activated protein kinase (AMPK), PGK1, and PKM2 gene expression levels in A375 cells were also evaluated.
ResultsThe IC50 value of CEG against A375 cells was determined to be 41.87 ± 2.47 µg/mL. A375 cells treated with CEG showed a significant increase in the G0/G1 phase and a decrease in the S and G2/M phases, indicating cell cycle arrest and reduced proliferation. Additionally, there was an increase in the sub-G1 peak, suggesting enhanced apoptosis. Additionally, the pharmacological analysis revealed potent inhibitory activity of CEG against both PGK1 and PKM2 gene expression, with IC50 values 27.89, 11.70, 7.43 and 2.74 µg/mL after incubation periods interval of 30, 60, 90 and 120 minutes, respectively. In In-Silico study, computational simulations showed a strong binding affinity of CEG towards AMPK, PGK1, and PKM2 activities, with estimated binding energy (∆G) values of -6.5, -7.9, and -8.3 kcal/mol, respectively. Furthermore, incubation of A375 cells with CEG (at concentrations of 20.9, 41.87, and 83.74 µg/mL) led to a significant decrease in GSH levels and the activity of GR, SOD, GPx, CAT, cyclin D1, cyclin E2, cdk2, and cdk4. Notably, CEG treatment upregulated AMPK levels while downregulating PGK1 and PKM2 gene expression significantly.
ConclusionCEG induces apoptosis in melanoma cancer cells (A375) through various mechanisms, including enhanced production of p53 and MDA, inhibition of key enzymes (GR, SOD, GPx, CAT) involved in oxidative stress defense and production of cell cycle regulating enzymes (cyclin D1, cyclin E2, cdk2, cdk4, and upregulation of AMPK and downregulation PGK1, and PKM2 in A375 tumor cells pathways. The downregulation of PKM2 in CEG-treated A375 cells inhibits ATP generation via aerobic glycolysis, a metabolic preference of cancer cells.
-
-
-
Anticancer Properties of Phenylboronic Acid in Androgen-Dependent (LNCaP) and Androgen-Independent (PC3) Prostate Cancer Cells via MAP Kinases by 2D and 3D Culture Methods
ObjectiveThis study utilized three cell lines: normal prostate epithelial RWPE-1, androgen-dependent LNCaP, and androgen-independent PC3. We investigated the inhibitory effects of phenylboronic acid (PBA)’s inhibitory effect on cellular proliferation due to its ability to disrupt microtubule formation in prostate cancer cell lines. Additionally, this study aimed to assess the cytotoxic effects of PBA on prostate cancer cells using two-dimensional (2D) and three-dimensional (3D) cell culture models.
MethodsThe IC50 values of PBA and colchicine were determined through viability assays in 2D and 3D models. Colony formation, proliferation, and migration assays were conducted. Immunofluorescence intensity analysis of MAPKKK proteins (ERK, JNK, p38) was performed to explore the mechanism of cellular response to PBA.
ResultsThe IC50 values were determined for each treatment group. After 48-hour of PBA treatment, migration was inhibited more effectively than with colchicine in both cancer cell lines. After 24-hour, PBA reduced colony formation and proliferation. PBA treatment for 24-hour decreased JNK expression in PC3 and LNCaP cells in 2D models. Both PBA and colchicine increased p38 expression in PC3 spheroids. PBA’s effects on cell deformation were visualized in semi-thin sections, marking the first ultrastructural observation of PBA-induced morphological defects in cancer cells.
ConclusionPBA exerts antimitotic effects by inhibiting proliferation and migration and triggers diverse metabolic responses across different cell lines. Furthermore, PBA’s low toxicity on RWPE-1 cells suggests its potential as a promising chemotherapeutic agent for future studies.
-
- Medicine, Oncology, Drug Design, Discovery and Therapy, Drug Design & Discovery, Chemistry, Medicinal Chemistry, Pharmacology
-
-
-
Chlorogenic Acid Ameliorates CCl4-induced Liver Fibrosis by Modulating the PI3K/AKT/mTOR Autophagy Pathway
Authors: Amr Negm, Amira Afifi El-Neanaey, Abada El Sayed Khadr, Maher Abd El Naby Kamel, Abd El-Hamid Abdo Ismail, Ibrahim El Tantawy El Sayed, Wael Sobhy Darwish, Mabrouk Attia Abd Eldaim, Raghda Sobhy Okaz, Mohamed Hamdy Bahr, Nihal Almuraikhi, Nermine Beshara, Tamer Mohamed Shawky and Ezat A. MersalBackgroundLiver fibrosis represents a serious risk to global health by impairing quality of life and elevating the chances of hepatocellular carcinoma, while the intricate role of autophagy can either alleviate or worsen fibrosis depending on its functioning.
ObjectiveHerein, we aimed to investigate the therapeutic effect of chlorogenic acid in CCl4-induced hepatic fibrosis and explored the autophagy pathway as the possible molecular target of chlorogenic acid.
MethodsRats were injected with carbon tetrachloride (1ml/kg) to induce liver fibrosis for 10 weeks. In the current study, the liver fibrosis rats were treated daily with chlorogenic acid (20, 40, and 60 mg/kg) for 30 days. Liver function tests, renal function tests, lipid peroxidation, antioxidant enzyme, anti-inflammatory NF-κB level, and autophagy pathway parameters (PI3K, AKT, mTOR, LC3, and Beclin-1) were assessed.
ResultsCCl4 elevated serum AST and ALT activity, and hepatic malondialdehyde, PI3K, AKT, and mTOR expressions. It decreased LC3, Beclin-1 expression, and hepatic glutathione level. The results indicated that chlorogenic acid treatment ameliorated the hepatic functions. It declined serum AST and ALT activities, improved hepatic GSH concentration, decreased lipid peroxidation, and downregulated PI3K, AKT, and mTOR protein expressions in hepatic tissue. Moreover, chlorogenic acid increased the hepatic expression of LC3 and Beclin-1. It also significantly decreased NF-kB expression.
ConclusionChlorogenic acid showed promise in reducing liver damage in rats caused by CCl4 by influencing the autophagy process and adjusting levels of antioxidant and inflammatory markers.
-
-
-
-
Effects of Citrus-derived Diosmetin on Melanoma: Induction of Apoptosis and Autophagy Mediated by PI3K/Akt/mTOR Pathway Inhibition
Authors: Jie Li, Mingyuan Xu, Nanhui Wu, Fei Wu, Jiashe Chen, Xiaoxiang Xu, Fei Tan and Yeqiang LiuBackgroundDiosmetin (DIOS) is a naturally abundant flavonoid and possesses various biological activities that hold promise as an anti-cancer agent. However, the anti-cancer activities and underlying mechanism of DIOS on cutaneous melanoma remain unclear.
ObjectiveThis study seeks to explore the anti-tumor effect and mechanism of DIOS in cutaneous melanoma.
MethodsHere, a variety of in vitro and in vivo experiments, combined with RNA sequencing (RNA-seq), were employed to ascertain the potential anti-cutaneous melanoma capacity and mechanism of DIOS.
ResultsThe results demonstrated that DIOS considerably impeded cell proliferation and triggered cell apoptosis in a dose- and time-dependent manner. Concurrently, DIOS markedly elevated the expression of pro-apoptotic proteins (Cleaved caspase-3, Bax, Cleaved PARP, and Cleaved caspase-9) and downregulated the expression of Bcl-2. Additionally, DIOS markedly upregulated the protein expressions of LC3B-II and Atg5, while downregulating p62 protein expression. Notably, pre-treatment with an autophagy inhibitor significantly inhibited DIOS-induced cell apoptosis and autophagy. Mechanistically, DIOS was identified to repress the PI3K/Akt/mTOR signaling pathway by western blot analyses and RNA-seq. Finally, in vivo experiments using a syngeneic mouse model confirmed the anti-tumor effect of DIOS, which exhibited high levels of apoptosis and autophagy.
ConclusionThese findings propose that DIOS acts as a potential melanoma therapy that exerts its anti-tumor effects by triggering apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathway.
-
-
-
In-silico and In-vitro Molecular Analysis of Oleanolic Acid and Cisplatin on Pancreatic Cancer (Panc-1 Cell Line)
Authors: Srimathi Devi Jegannathan, Wishwaa Jayapal, Bindhu Jayaprakash and Teena PrabhuBackgroundCisplatin (CIS) is a standard chemotherapeutic drug currently used for various cancer treatments. Due to its chemo-resistance and toxic effects, a new combinatorial approach was preferred. Oleanolic acid is one such pentacyclic terpenoid compound that tends to have various anti-cancer properties against a wide range of human carcinoma models. Yet, the final mechanisms of individual and Combinational Treatment of OA and CIS on pancreatic carcinoma persist indescribable.
ObjectiveThe Current study analyses the in-silico and in-vitro Molecular efficacy of the combinational dose of OA and CIS in Pancreatic cancer using the Panc-1 cell line.
Methods and MaterialThe preliminary screening of the anti-cancer effect of OA and CIS was evaluated meticulously using docking score with Auto-Dock. For further in-vitro analysis of the ligand, OA was isolated from blueberry through ultrasonication extraction, followed by a comprehensive range of qualitative and quantitative analysis by chromatography techniques and GC-MS studies. Anti-proliferative and cytotoxicity activity of our combinational compounds were determined using the MTT assay and the LDH leakage assay. Cell membrane integrity was analyzed by measuring ROS generation and mitochondrial membrane potential in treated cells using fluorometric detection methods. Detection of the Anti-Apoptotic potential of our target compound was evaluated by DNA fragmentation assay and Caspase activity assay. Quantitative real-time PCR and Western Blotting were used to determine the genes and Protein expression intricated for apoptosis, angiogenesis, cell cycle regulation, and metastasis.
ResultsMolecular docking analysis suggests that OA and CIS possess a strong binding affinity for hydrogen bond interaction with the highest fitness score for various anti-cancer genes, leading to the drug's significant apoptotic and anti-angiogenic effects. Further preliminary analysis reports of UV spectra and GC-MS data suggested that the OA compound tends to exhibit a peak at 235-288 nm with a GC retention time of 15.45 min with m/z 240 and m/z 280 ratios. The output of In-vitro analysis of the anti-proliferative and cytotoxicity effect of OA and CIS tends to show the significant inhibition of cells in a dose-dependent manner with IC50 value of 5.75 µM OA and 2.95 µM of CIS with significant leakage in LDH was observed in combinational treated cells compare to individual treated cancer cells. The computational CI plot report of OA and CIS report revealed a synergistic dose effect with a CI value<1. Apoptotic effect of combinational dose revealed synergistic effects by down-regulation of angiogenic and metastatic genes and proteins (CDKN2A, SMAD4, VEG-F, MMP-9) stimulates the caspase cascade activation by intrinsic mediated apoptosis, which was further confirmed through DNA fragmentation assay by cleavage of fragments in treated cells compared to control.
ConclusionIn conclusion, the present study indicates that the co-treatment of OA with CIS in pancreatic cancer cells exerts strong interaction with synergistic effects on cell growth inhibition, apoptosis induction, and angiogenesis genes through regulating signal-target proteins applicable for pancreatic cancer.
-
- Medicine, Oncology, Drug Design, Discovery and Therapy, Drug Design & Discovery, Chemistry, Medicinal Chemistry, Pharmacology
-
-
-
Computational Modelling Strategies for Exploring Triazolopyridazine PIM1 Kinase Inhibitors as Anticancer Agents
BackgroundPIM (Proviral Integration site for Moloney Murine Leukemia virus) kinases are members of the class of kinase family serine/threonine kinases, which play a crucial role in cancer development. As there is no drug in the market against PIM-1, kinase has transpired as a budding and captivating target for discovering new anticancer agents targeting PIM-1 kinase.
AimThe current research pondered the development of new PIM-1 kinase inhibitors by applying a ligand-based and structure-based drug discovery approach involving 3D QSAR, molecular docking, and dynamics simulation.
MethodsIn this study, association allying the structural properties and biological activity was undertaken using 3D-QSAR analysis. The 3D-QSAR model was generated with the help of 35 compounds from which the best model manifested an appreciated cross-validation coefficient (q2) of 0.8866 and conventional correlation coefficient (r2) of 0.9298, respectively and the predicted correlation coefficient (r2 pred) was obtained as 0.7878.
ResultsThe molecular docking analysis demonstrated that the analogs under analysis occupied the active site of the PIM-1 kinase receptor and interactions with Lys67 in the catalytic region, Asp186 in the DFG motif, and Glu171 were noticed with numerous compounds.
DiscussionFurthermore, the molecular dynamics simulation study stated that the ligand portrayed strong conformational stability within the active site of PIM-1 kinase protein, forming two hydrogen bonds until 100 ns, respectively.
ConclusionOverall outcomes of the study revealed that applications of the ligand-based drug discovery approach and structure-based drug discovery strategy conceivably applied to discovering new PIM-1 kinase inhibitors as anticancer agents.
-
-
Volumes & issues
-
Volume 25 (2025)
-
Volume 24 (2024)
-
Volume 23 (2023)
-
Volume 22 (2022)
-
Volume 21 (2021)
-
Volume 20 (2020)
-
Volume 19 (2019)
-
Volume 18 (2018)
-
Volume 17 (2017)
-
Volume 16 (2016)
-
Volume 15 (2015)
-
Volume 14 (2014)
-
Volume 13 (2013)
-
Volume 12 (2012)
-
Volume 11 (2011)
-
Volume 10 (2010)
-
Volume 9 (2009)
-
Volume 8 (2008)
-
Volume 7 (2007)
-
Volume 6 (2006)
Most Read This Month
