Anti-Cancer Agents in Medicinal Chemistry - Online First
Description text for Online First listing goes here...
1 - 20 of 68 results
-
-
ALG3 as a PanCancer Oncogene: Bioinformatics Analysis and Identification of Small-Molecule Inhibitors
Available online: 03 November 2025More LessIntroductionGlycosylation plays a crucial role in cellular processes such as recognition and signaling, and its dysregulation is associated with tumor progression. Alpha-1,3-mannosyltransferase (ALG3) is a key enzyme in N-glycosylation, and its aberrant expression has been implicated in various malignancies. However, the mechanisms underlying ALG3-driven oncogenesis and the identification of potential ALG3 inhibitors remain largely unexplored. This study aims to comprehensively investigate the oncogenic role of ALG3 across different cancer types and identify potential inhibitors through bioinformatics analysis and molecular docking-coupled dynamics simulations.
MethodsMultiple cancer-related databases were analyzed to elucidate the oncogenic role of ALG and to assess its expression patterns, genetic alterations, and epigenetic regulation. Furthermore, molecular docking and dynamics simulations were employed to identify small-molecule inhibitors targeting the human ALG3.
ResultsOur findings demonstrated a significant upregulation of ALG3 at both transcript and protein levels in cancerous tissues compared to normal ones. High ALG3 expression correlated positively with tumor stage, grade, and metastasis while negatively influencing patient survival. Genetic analysis revealed that amplification was the most common alteration in ALG3, whereas DNA methylation played a key role in its upregulation. Molecular docking and dynamics simulation identified two mannosyltransferase inhibitors, Opn and Clo, as potential inhibitors of ALG3, suggesting their therapeutic potential.
DiscussionThis study highlights the oncogenic role of ALG3 in a pan-cancer model and identifies its potential inhibitors. Our findings provide valuable insights into ALG3-driven tumorigenesis and suggest that targeting ALG3 could be a promising strategy for cancer therapy.
ConclusionThe study first reported potential inhibitors of human ALG3 based on a molecular modelling approach. This opens the way for future experimental investigations of the testing of these lead compounds in ALG3-high cancer models.
-
-
-
Jianpi Yiwei Granules for Chemotherapy-Induced Gastrointestinal Reactions: A Randomized Double-Blind Placebo-Controlled Trial
Authors: Kerun Cai, Jiayang Chen, Nuo Li and Li FengAvailable online: 16 October 2025More LessIntroductionChemotherapy-induced gastrointestinal reactions are common in non-small cell lung cancer (NSCLC) patients undergoing carboplatin-based chemotherapy. Jianpi Yiwei granules (JPYW), a traditional Chinese medicine (TCM) formula, can alleviate these symptoms.
Materials and MethodsThis multi-center, randomized, double-blind, placebo-controlled trial enrolled 136 NSCLC patients scheduled for carboplatin-based chemotherapy. Participants were randomly assigned to the treatment group (JPYW with standard antiemetic drugs) and the control group (placebo with standard antiemetic drugs). The complete control rate of nausea and vomiting was assessed using the Visual Analog Scale (VAS) and patient diaries. Control of anorexia, bloating, constipation, and quality of life was measured using the Functional Living Index-Emesis scale and the Brief Fatigue Inventory (BFI).
ResultsThe primary objective of this study was to assess the efficacy of JPYW in alleviating non-vomiting digestive symptoms, such as nausea and anorexia, in NSCLC patients receiving carboplatin-based chemotherapy. The secondary objective was to evaluate its effect on improving bloating, constipation, quality of life, and safety.
DiscussionPrevious studies have shown that Chinese herbs, such as ginger, are effective in treating chemotherapy-induced nausea and vomiting (CINV). JPYW, a multi-component TCM formula, contains active compounds from Codonopsis pilosula and Atractylodes macrocephala. JPYW exerts anti-inflammatory and prokinetic effects that can synergistically regulate gastrointestinal functions. Preliminary observations confirmed the safety of JPYW combined with standard chemotherapy.
ConclusionThe current findings contribute to the treatment of adverse reactions to tumor chemotherapy and are expected to improve the quality of life for chemotherapy patients.
-
-
-
The EP300-Targeting Drug CCS1477 Inhibits the Growth and Development ofDiffuse Large B-Cell Lymphoma by Promoting Apoptosis and Mitophagy to Reduce Drug Resistance
Authors: Rujia Si, Yihan Zhang, Bowen Hu, Yuxin Du, Dan Zou, Shaodi Wen, Xiaoyue Du, Xin Chen, Chen Peng, Shulei Fu, Shiying Zhu, Fan Du, Xiaofeng Sha, Ning Ding, Cong Xu and Bo ShenAvailable online: 10 October 2025More LessIntroductionApproximately 30% of patients with diffuse large B-cell lymphoma (DLBCL) develop primary resistance or relapse, owing to the high heterogeneity and aggressive nature of the disease. Consequently, novel drugs are urgently needed to improve outcomes in patients who are resistant.
MethodsThis study quantified the anti-proliferative effects of CCS1477 in vitro using the Cell Counting Kit-8 assay, 5‐ethynyl‐2′‐deoxyuridine staining, and lactate dehydrogenase measurement. Flow cytometry and Western blot analyses were performed concurrently to investigate the induction of apoptosis and the activation of mitophagy. The efficacy and safety of CCS1477 were evaluated in in vivo models. To elucidate the mechanism, cell lines with EP300 knockdown and overexpression were established. Functional assays and Western blot analyses revealed that EP300 regulates apoptosis, mitophagy, and c-MYC-mediated drug-resistant phenotypes.
ResultsThis study demonstrated that CCS1477, a highly selective EP300/CBP bromodomain inhibitor, significantly suppressed the progression of diffuse large B-cell lymphoma. The study revealed that CCS1477 dose-dependently inhibited the proliferation of diffuse large B-cell lymphoma cells and induced apoptosis and mitophagy. Mechanistically, EP300 downregulation promoted apoptosis and activated the PINK1-dependent mitophagy pathway while suppressing c-MYC-mediated drug resistance genes, ultimately inhibiting DLBCL cell proliferation. In animal models, CCS1477 significantly reduced tumor volume and extended doubling time, providing the first evidence of its in vivo antitumor activity against DLBCL.
DiscussionThrough systematic in vitro and in vivo investigations, this study validated the significant therapeutic promise of EP300/CBP inhibitor CCS1477 for diffuse large B-cell lymphoma. However, the mechanistic basis for differential sensitivity across DLBCL subtypes, along with long-term efficacy and potential adverse effects, requires comprehensive investigation. Notably, EP300 has been verified as a novel prognostic biomarker and therapeutic target; this work establishes an innovative epigenetic-targeted strategy for relapsed/refractory diffuse large B-cell lymphoma.
ConclusionBy selectively targeting EP300, CCS1477 orchestrates a dual pro-death mechanism involving both intrinsic apoptosis execution and PINK1-driven mitochondrial clearance, resulting in significant inhibition of diffuse large B-cell lymphoma pathogenesis.
-
-
-
GPR65 as a Laryngeal Cancer Risk Gene Identified through Single-Cell Transcriptomics, Mendelian Randomization Analysis, and Experimental Validation
Authors: Qing-Jie Mao, Ya-Quan Zhou, Bing-Shuang Zhao, Hao Wu, Shu-Zheng Wang, Zhen-Xin Zhang and Hao-Sheng NiAvailable online: 09 October 2025More LessIntroductionLaryngeal cancer is a common malignant tumor of the head and neck worldwide. This study aimed to identify potential risk genes, with a particular focus on GPR65, and to investigate its functional mechanism in pathogenesis of laryngeal cancer.
Materials and MethodsComprehensive analyses, including scRNA-seq analysis, genome-wide association study (GWAS), eQTL, and TCGA data, were conducted to identify risk genes for laryngeal cancer and characterize the function of these risk genes. Next, qRT-PCR, immunohistochemistry, cell proliferation, cell migration, and invasion assays were employed to verify the expression of GPR65 and its function in laryngeal squamous cell carcinoma (LSCC) in vitro.
ResultsSingle-cell analysis screened 416 highly expressed genes in CD8+ central memory T cells (CD8_CM). Mendelian randomization (MR) analysis identified GPR65 as a crucial gene in the development of laryngeal cancer. GPR65 expression was significantly elevated in the tumor tissues compared to normal tissues, with particularly high levels observed in stage IV HNSCC. In vitro, LSCC cell lines (TU686 and Hep-2) exhibited marked upregulation of GPR65 relative to normal epithelial cells, and siRNA-mediated silencing of GPR65 suppressed the proliferation, migration, and invasion of LSCC cells. Furthermore, GPR65 expression showed a positive correlation with immune cell infiltration, particularly CD8+ T cells and M1 macrophages.
DiscussionThis study identified GPR65 as a potential risk gene for laryngeal cancer through single-cell transcriptomics and MR analyses and provided novel evidence of its involvement in the development of the cancer.
ConclusionThe present findings showed that highly expressed GPR65 was a tumor-promoting gene in laryngeal cancer, showing its clinical value as a potential therapeutic target.
-
-
-
Novel PD-L1 Small-Molecule Inhibitors Advancing Cancer Immunotherapy
Authors: Annoor Awadasseid, Mengda Wu, Feng Zhang, Yanhua Song, Yanling Wu and Wen ZhangAvailable online: 03 October 2025More LessIntroductionThe emergence of immune checkpoint inhibitors has revolutionized the treatment of cancer. Among these, the programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) axis remains a critical target. However, resistance to current biologics necessitates the development of novel Small-Molecule Inhibitors (SMIs) with distinct mechanisms and improved pharmacological profiles. This review provides a comprehensive analysis of recent progress in PD-L1-targeting SMIs, including original compounds from our laboratory.
MethodsWe conducted a structured literature review using electronic databases such as PubMed, Scopus, and Web of Science. Articles published between 2015 and 2025 were included based on relevance to small-molecule PD-L1 inhibitors in cancer immunotherapy. Key data were extracted and synthesized regarding molecular design strategies, mechanisms of action, pharmacokinetics, and therapeutic efficacy. Compounds synthesized in our laboratory (Compounds 5–10 [A56]) were evaluated using in vitro assays, including PD-L1/PD-1 binding inhibition, cancer cell viability assays, and gene expression profiling.
ResultsRecent SMIs exhibit diverse functional profiles: direct blockade of PD-1/PD-L1 interaction, intracellular PD-L1 modulation, and transcriptional downregulation. Notably, Compound 7 demonstrated significant suppression of PD-L1 mRNA expression, while Compounds 9 and 10 (A56) achieved nanomolar-level binding affinity. These findings reflect innovative approaches to overcoming immune resistance and enhancing antitumor responses.
DiscussionsOur findings underscore a trend toward multifunctional PD-L1-targeting SMIs that operate through both extracellular and intracellular mechanisms. Compounds from our laboratory represent potential leads for further optimization and clinical translation. However, challenges remain regarding oral bioavailability, metabolic stability, and immune-related adverse events.
ConclusionSmall-molecule PD-L1 inhibitors offer a promising avenue for expanding cancer immunotherapy. Our review highlights key advances and introduces novel small-molecule PD-L1 inhibitors with strong potential for future development, particularly in combination regimens.
-
-
-
Induction of Apoptosis and Activation of Endoplasmic Reticulum Stress by SJ6986 in Diffuse Large B-cell Lymphoma
Authors: Chenxing Zhang, Bangxue Jiang, Xiaomei Liang, Yinting Chen, Zhaozheng Li, Minyi Zhao and Dongjun LinAvailable online: 29 September 2025More LessIntroductionDiffuse large B-cell lymphoma (DLBCL) is one of the most prevalent hematological malignancies with high mortality. G1 to S phase transition 1 (GSPT1), a key translation termination factor involved in protein synthesis, has been implicated in tumor progression. This study aimed to investigate the effectiveness and underlying mechanisms of the GSPT1 degrader SJ6986 in DLBCL.
MethodsThe TCGA and GTEx datasets were utilized to assess the expression of GSPT1 in DLBCL. The viability and proliferation of DLBCL cells were detected using the Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis was detected via flow cytometry. The expression of GSPT1 was evaluated using qRT-PCR and Western blot. Xenograft mouse models were employed to explore the in vivo therapeutic potential of SJ6986. RNA sequencing was used to explore the potential mechanism of SJ6986 in DLBCL.
ResultsThis study first identified that GSPT1 is highly expressed in DLBCL and demonstrated that its genetic knockdown significantly suppressed the activity of DLBCL cells. Furthermore, it was found that SJ6986 effectively reduced the proliferation of DLBCL cells, induced cell apoptosis, and inhibited tumor growth in vivo without significant toxicity. Mechanistically, RNA sequencing analysis showed that the endoplasmic reticulum (ER) stress was significantly triggered following SJ6986 treatment, and SJ6986 was found to activate the ER stress-related apoptosis in DLBCL cells.
DiscussionOur findings suggested that SJ6986 exerts its anti-tumor effects in DLBCL and activates the ER stress-related apoptotic signaling. These results supported SJ6986 as a viable anticancer drug for treating DLBCL. Future studies should further investigate its mechanism and evaluate its clinical application value.
ConclusionsThis study validated the efficacy and safety of SJ6986 in treating DLBCL and discovered its role in inducing ER stress and subsequent apoptosis, offering a promising therapeutic option for DLBCL patients.
-
-
-
Apoptosis-Mediated Anticancer Activity of Zinc Oxide Nanoparticles Derived and Characterized from Halophila beccarii
Authors: Jayasheela Sarilla, Vani Mathakala and Uma Maheswari Devi PalempalliAvailable online: 26 September 2025More LessIntroductionRecent advancements in nanomedicine have drawn attention to the use of zinc oxide nanoparticles as apoptotic agents to address triple-negative breast cancer. Halophila beccarii-mediated zinc oxide nanoparticles (Hb-ZnONPs) were fabricated using zinc acetate dihydrate as the precursor.
MethodsThe fabricated nanoparticles were characterized based on morphological, structural, and elemental composition using SEM and XRD. The antiproliferative potential of Hb-ZnONPs was studied using the BT-549 cell line as an in vitro model, employing the MTT assay and Annexin V-FITC/PI-based flow cytometry analysis.
Results & DiscussionThe Hb-ZnONPs exhibited characteristic absorption maxima at 367 nm with a particle size of 35 nm and −44.7 mV stability. XRD confirmed the hexagonal wurtzite structure with an elemental composition of 62.3% Zn and 25.79% Oxygen. The Hb-ZnONPs demonstrated significant cytotoxicity against BT-549 cells, with 35.26% apoptosis at 5 µg/ml and 38.25% apoptosis at 10 µg/ml. However, cells in the late apoptosis stage increased from 14.48% at 5 µg/ml to 28.16% at 10 µg/ml, indicating a nearly twofold increase with the higher concentration.
ConclusionHb-ZnONPs may act as promising apoptotic inducers in the chemotherapy of breast cancer.
-
-
-
PRR22: A Novel Prognostic Indicator and Therapeutic Target for Prostate Cancer
Authors: Wenxia Chen, Guodong Ding, Yuantang Zhong, Meiting Lao, Qing Zhang, Dongbing Li, Wangdong Deng and Yiwen ChenAvailable online: 26 September 2025More LessIntroductionProstate cancer (PRAD) remains a leading malignancy with limited prognostic biomarkers and therapeutic targets. PRR22, a proline-rich protein-coding gene, has a role in PRAD that remains undefined. This study is the first to systematically investigate the clinical relevance and mechanistic implications of PRR22 in PRAD.
MethodsPRR22 expression was analyzed in TCGA-PRAD (n = 501), GSE55945, and the Human Protein Atlas datasets. Prognostic value was assessed via Kaplan-Meier and multivariate Cox analyses. Mechanistic insights were derived from GSEA, immune infiltration profiling, MSI/mRNA-si correlations, and drug sensitivity analysis. Experimental validation was performed via qRT-PCR in PRAD cell lines.
ResultsPRR22 was significantly upregulated in PRAD tissues compared to normal tissues (p < 0.001) and independently predicted shorter progression-free survival (HR = 1.82, p = 0.009). Novel associations were identified between PRR22 and TGF-β signaling, immune evasion (e.g., LAG3 upregulation), microsatellite instability (MSI), and stemness (mRNA-si). High PRR22 correlated with resistance to multiple drugs (e.g., bicalutamide, vorinostat).
DiscussionPRR22 overexpression in PRAD is linked to poor prognosis and immune regulation, suggesting its potential as a prognostic biomarker and therapeutic target. Future research should focus on clinical validation and on exploring the molecular mechanisms underlying PRR22's role in PRAD.
ConclusionPRR22 is a novel, independent prognostic biomarker and actionable therapeutic target in PRAD, linking tumor aggressiveness to immune microenvironment remodeling and drug resistance. These findings establish PRR22 as a candidate for clinical implementation in risk stratification and targeted therapy.
-
-
-
A Review of the Anticancer Properties of Cedrol and its Molecular Mechanisms
Available online: 24 September 2025More LessDespite decades of research on promising new therapies, cancer remains a leading cause of morbidity and mortality. Over the years, extensive research has been conducted on the potential anticancer effects of various medicinal plants. One extremely promising agent or adjuvant that may be utilized for the prevention/treatment of several malignancies is cedrol, a naturally occurring sesquiterpene. Cedrol modulates multiple molecular pathways involved in the protracted carcinogenesis process, including the generation of reactive oxygen species, activation of pro-death autophagy, inhibition of survival signals, promotion of apoptosis, and inhibition of minichromosome maintenance proteins. This review suggests that cedrol might be a unique medication for the treatment of glioblastoma, lung cancer, and colorectal cancers. Further in-depth investigations of cedrol's anticancer mechanisms are needed.
-
-
-
Rosmarinic Acid as a Potential Therapeutic Agent against Neuroblastoma: Anticancer Activity and Molecular Docking Insights
Authors: Pınar Yumrutaş, Demet Taşdemir and Önder YumrutaşAvailable online: 24 September 2025More LessIntroductionRosmarinic acid (RA) is a phenolic acid known for its important biological activities. Although it has been shown to inhibit various cancer cell types, its effects on the suppression and induction of apoptosis in neuroblastoma cells remain unclear. In this study, the antiproliferation and apoptosis-inducing effects of various concentrations of rosmarinic acid on neuroblastoma cells (SH-SY5Y) were investigated. Additionally, molecular docking analysis was conducted to examine the interaction between rosmarinic acid and the antiapoptotic protein BCL2.
MethodsSH-SY5Y cells were treated with rosmarinic acid at concentrations of 50, 100, 150, and 200 µg/ml for 24 hours. The percentages of apoptotic and necrotic cells in cultures treated with the lowest and highest concentrations were assessed using the Annexin V/PI staining method. Furthermore, the interaction between rosmarinic acid and BCL2 protein was analyzed using molecular docking techniques.
ResultsThe viability of rosmarinic acid-treated SH-SY5Y cells decreased. In SH-SY5Y cells, the percentage of late apoptotic cells increased to 40%. Molecular docking results showed that the benzene ring of rosmarinic acid formed pi-alkyl interactions with PHE71 and van der Waals interactions with SER64, ALA72, SER75, and VAL115 of BCL2. The lowest binding energy was calculated as -7.2 kcal/mol.
DiscussionRA demonstrated a suppressive effect on SH-SY5Y cells by targeting the antiapoptotic protein BCL2, suggesting a potential mechanism of action through the induction of apoptosis.
ConclusionRA inhibited neuroblastoma SH-SY5Y cell proliferation and induced apoptotic cell death. It inhibited the proliferation of neuroblastoma SH-SY5Y cells and promoted apoptotic cell death, potentially through interaction with the BCL2 protein.
-
-
-
Nanosomal-Mediated Lipid Suspension Delivery of Docetaxel as a Promising Landscape to Enhance the Therapeutic Potential in Triple-Negative Breast Cancer
Authors: Pritya Jha, Varisha Anjum, Rabia Choudhary, Ammar Kadi, Faraat Ali and Irina PotorokoAvailable online: 18 September 2025More LessThe challenging subtype of breast cancer known as Triple-Negative Breast Cancer (TNBC) is characterized by the absence of HER2 expression, progesterone receptors, and estrogen receptors. TNBC is linked to a harsh treatment trajectory, elevated rates of recurrence, and restricted therapeutic alternatives. The mainstay of treatment for TNBC has historically been conventional chemotherapy, especially taxanes like Docetaxel. However, the effectiveness of these drugs is frequently compromised by systemic toxicity and resistance mechanisms. The development of Nanosomal Docetaxel Lipid Suspension (NDLS) offers a promising alternative, designed to enhance Docetaxel's therapeutic index by improving solubility, reducing side effects, and optimizing tumor-targeted drug delivery. NDLS has potential as a delivery system for additional chemotherapy drugs or combination treatments. This study addresses the cellular and molecular causes of TNBC, emphasizes the drawbacks of traditional treatments, and offers a thorough examination of NDLS in preclinical and clinical settings. This review provides a thorough analysis of NDLS in TNBC, laying the groundwork for further studies and therapeutic applications.
-
-
-
Targeting the Lin28/let-7 Axis with Compounds to Regulate Transcriptional Control in Cancer
Authors: Xingpeng Wang, Pham Kim Thuong Van, Bo Liu, Tingxiu Zhao and Yun-shan WuAvailable online: 17 September 2025More LessLin28 is a pivotal RNA-binding protein that regulates the biogenesis of let-7 microRNAs, which play a crucial role in the post-transcriptional regulation of oncogenes in cancer. The Lin28/let-7 axis is integral to the regulation of key cellular processes such as proliferation, differentiation, and apoptosis. Lin28 promotes the upregulation of oncogenes, including MYC, RAS, and HMGA2, by inhibiting the maturation of let-7, thereby facilitating tumor initiation, progression, and metastasis. Consequently, targeting the Lin28/let-7 interaction has emerged as a promising therapeutic strategy, particularly for malignancies that lack specific molecular targets. This approach holds potential for downregulating oncogene expression and inhibiting tumor progression. Through a comprehensive review of the literature, this article classifies Lin28/let-7 inhibitors into three categories: CSD/ZKD inhibitors, non-CSD/ZKD inhibitors, and let-7 restorers. CSD/ZKD inhibitors, such as TPEN and KCB3602, function by binding to the CSD or ZKD domains of Lin28, thereby inhibiting its activity. Non-CSD/ZKD inhibitors, including compounds like C1632 and Simvastatin, have been identified as molecules that can reduce Lin28 activity, though their binding sites remain unknown. Let-7 restorers, on the other hand, do not directly target Lin28 but instead work indirectly by modulating the activity of associated molecules, such as Zcchc11 and Zcchc6, thereby promoting the restoration of let-7 expression levels. Notable examples of these include IPA-3 and FPA124. This review summarizes recent advances in the development of Lin28/let-7 inhibitors and their therapeutic potential, providing an important reference for ongoing research on Lin28 inhibitors in cancer therapy.
-
-
-
State of the Art of IDH Inhibitors: Emerging Questions and Perspectives
Authors: João A. L. de Lima and Lídia Moreira LimaAvailable online: 16 September 2025More LessIsocitrate Dehydrogenases (IDH) are ubiquitous enzymes essential for cellular metabolism, including the Krebs cycle, glutamine metabolism, lipogenesis, and redox balance. Mutations in IDH1 and IDH2 are implicated in several tumors-gliomas, Acute Myeloid Leukemia (AML), cholangiocarcinoma-altering enzyme activity and causing the overproduction of 2-hydroxyglutarate (2-HG). This oncometabolite disrupts α-KG-dependent proteins, impairing key processes such as differentiation, division, and DNA repair. Understanding these genetic, biochemical, and clinical aspects has made IDH enzymes promising therapeutic targets, prompting the development of targeted inhibitors for tumors harboring IDH1 or IDH2 point mutations. Selective inhibitors like ivosidenib (AG-120) and enasidenib (AG-221), targeting mutant IDH1 and IDH2 respectively, block 2-HG production and induce differentiation, achieving clinical success - particularly in AML. However, resistance due to secondary mutations, especially in the allosteric binding site, remains a major obstacle. In response, novel approaches have emerged, such as covalent inhibitors like LY3410738, which irreversibly bind mutant residues, and dual inhibitors like vorasidenib (AG-881), which act on both IDH1 and IDH2 mutations and penetrate the blood-brain barrier for treating solid tumors. Still, many clinical factors must be considered. This review explores the current landscape of IDH-targeted therapies, emphasizing the need for novel inhibitors and highlighting innovative strategies, including the design of smaller, more potent molecules with favorable pharmacokinetics and the potential of drug repositioning. We underscore that discovering new antitumor compounds targeting IDH requires a collaborative effort across biomedical fields. These advancements aim to overcome resistance, broaden therapeutic options, and improve the effectiveness of IDH-targeted treatments.
-
-
-
Targeting WWPI HECT Domain by Small Inhibitors for Restoring PTEN Tumor Suppressive Role in Glioblastoma Therapy
Available online: 09 September 2025More LessIntroductionPTEN (Phosphatase and tensin homolog) is a valuable regulator of the PI3K-AKT and mTOR pathways and is frequently mutated in cancer-like glioblastoma. The WWPI HECT domain has a group of enzymes called E3 ligases that ubiquitinate and inactivate PTEN by binding to it, which ultimately inhibits its lipid phosphatase function and promotes nuclear delocalization. This investigation seeks to restore the PTEN tumor suppressive activity by inhibiting the WWPI HECT domain in-silico.
MethodsWe virtually screened a library of ~960 compounds in the active pocket of the human WWPI HECT domain, and fifteen compounds were chosen based on their favorable binding affinities and highly negative docking scores.
ResultsAmong those hits, five compounds, C5, C6, C8, C9 and C11, properly fit the standard with favorable pharmacokinetic and drug-like quality. Their capacity to suppress cell propagation was evaluated in the U87 glioma cell line. The compounds (C5, C6, C8, C9 and C11) exhibited significant anti-proliferative capability with IC50 values of 6.98 ± 0.14 µM, 14.58 ± 1.49 µM, 11.12 ± 0.73 µM, 13.85 ± 1.63 µM and 18 ± 1.23 µM, respectively.
DiscussionStrong inhibitory action against glioma cells was shown by the discovered compounds, especially C5 and C8, suggesting that they may be able to restore PTEN tumor suppressive capabilities. A potential therapeutic intervention mechanism for glioblastoma is suggested by their interaction with the WWPI HECT domain.
ConclusionThis study has discovered novel inhibitors against the WWPI HECT domain, and a treatment option for glioblastoma.
-
-
-
Unveiling the Distinct Effects of a Two-Dimensional Copper/Sodium Complex: Oxidative Stress on Erythrocytes and Cytotoxicity, Apoptosis, Drug Resistance, and Inflammation in Lung Cancer Cells
Authors: Chenchen Li, Mostafa Heidari Majd, Ameneh Heidari, Zohreh Razmara and Dongdong GuoAvailable online: 03 September 2025More LessIntroductionCopper complexes, as endogenous metals, have potential in cancer therapy, addressing issues associated with cisplatin. Since cisplatin uses Copper Transporter 1 (CTR1) for cellular entry, copper complexes may utilize this pathway to enhance transport efficiency.
MethodsThe Cu/Na dipicolinic acid complex was synthesized to assess its cytotoxicity, induction of apoptosis, drug resistance, and inflammation in cancerous and normal lung cells. The effects of oxidative stress on erythrocytes were also examined.
ResultsCytotoxicity tests (MTT and SRB) showed superior inhibitory effects on A549 lung cancer cells compared to cisplatin, with no toxicity observed in MRC-5 normal lung fibroblast cells. Real-time PCR revealed increased caspase-3 expression (extrinsic apoptosis) for the complex compared to cisplatin, possibly due to CTR1-mediated entry. The complex did not induce drug resistance, as shown by AKT1 expression, and reduced TNF-α expression, preventing inflammation in normal cells. In contrast to cisplatin, the complex caused minimal oxidative stress in erythrocytes.
DiscussionIt can be concluded that the Cu/Na dipicolinic acid complex may be easily transported by CTR1 to malignant tumors, particularly lung cancer. This complex has the ability to inhibit cancer cell growth and induce apoptosis in lung cancer cells. Therefore, copper complexes show promise as potential therapeutic options for treating this type of cancer.
ConclusionThe copper/sodium complex demonstrates enhanced therapeutic efficacy in lung cancer cells, requiring lower doses than cisplatin, while being safer for normal cells and erythrocytes.
-
-
-
Luteolin Enhances Anticancer Effects of PX-478 during Hypoxic Response in Metastatic Breast Cancer Cells
Authors: Muzaffer Dukel and Fatema ZarzourAvailable online: 03 September 2025More LessIntroductionThe presence of severe hypoxic stress can drive tumor growth, angiogenesis, and metastatic characteristics via up-regulated hypoxia-inducible factor 1-alpha (HIF-1a). Hence, targeting HIF-1α is considered a promising strategy, as increased HIF-1α activity is a key factor in the aggressive phenotype of malignancies. In this study, we aimed to investigate the anti-cancer effects of several flavonoids, both single and in combination with PX-478, in breast cancer cell lines.
MethodsWe tested the effects of luteolin and PX-478, both alone and in combination, on HIF-1a level in breast cancer cells under hypoxia using the cell viability assay. To determine the rationale for the cell growth inhibition induced by the luteolin+PX-478 combination, we conducted experiments to assess cell survival, apoptosis, cell cycle, invasion, and migration under both normoxic and hypoxic conditions. Furthermore, we evaluated the effect of this combination on DNA damage response under hypoxic stress via Comet assay and immunofluorescence staining.
ResultsOur findings revealed that the luteolin+PX-478 combination significantly suppressed the growth of MDA-MB-231 cells. In addition, we assessed time-dependent expression of HIF1a in MDA-MB-231 cells and observed that the combination of luteolin and PX-478 down-regulated the HIF-1a level. Finally, we found that the luteolin+PX-478 combination induced apoptosis and G2 cell cycle arrest and enhanced DNA damage response. This combination also sensitized breast cancer cells to ionizing radiation in hypoxic stress.
DiscussionThe findings suggested that targeting HIF-1α with a combination of luteolin and PX-478 may provide a synergistic approach to suppressing tumor growth and enhancing therapeutic response under hypoxic conditions. The observed effects on apoptosis, cell cycle arrest, and DNA damage response indicated that this combination could be a promising strategy for overcoming hypoxia-induced resistance in breast cancer therapy.
ConclusionCollectively, our results suggested the combination of luteolin and PX-478 to enhance the anti-cancer effects of PX-478 in breast carcinoma cells by impeding the cell growth and inducing DNA damage response under hypoxia.
-
-
-
A Comprehensive Review of the Anticancer Activity of Farnesiferol C and Umbelliferone
Available online: 02 September 2025More Lessancer remains a growing challenge in modern society, presenting a significant obstacle in both developed and developing countries. Conventional treatments are often costly and limited by issues such as drug resistance and undesirable side effects. Consequently, the exploration of natural compounds has emerged as a promising strategy for developing more effective and tolerable cancer therapies. Among these, Ferula plants have gained attention for their potential anticancer components. Notably, two coumarin compounds derived from these plants, farnesiferol C and umbelliferone, have demonstrated substantial anticancer activity, as supported by an increasing number of published studies. This review aims to consolidate existing evidence on the anticancer effects of farnesiferol C and umbelliferone while comparing their efficacy as potential therapeutic agents. To accomplish this, a comprehensive literature search was conducted using the terms “umbelliferone” and “farnesiferol C” paired with “anticancer” across databases such as ISI Web of Knowledge, PubMed, and Google Scholar. Relevant studies up to March 2024 were retrieved, summarized, and incorporated into this analysis. The findings indicate that both compounds exhibit significant anticancer properties, positioning them as viable candidates for future drug development. A comparative analysis of their IC50 values, the concentration required to inhibit 50% of cancer cell growth, reveals that farnesiferol C demonstrates greater cytotoxic potency against various cancer cell lines compared to umbelliferone. However, while these results are encouraging, further research is recommended, particularly in vivo studies to evaluate the compounds’ toxicity and therapeutic potential in living organisms.
-
-
-
Unlocking the Potential of Polysaccharides for the Treatment of Lung Cancer
Authors: Himanshu Singh, Rajnish Kumar and Avijit MazumderAvailable online: 01 September 2025More LessBackgroundLung cancer remains a leading cause of cancer-related deaths worldwide, with its incidence continuing to rise. Despite advancements in clinical treatments, their effectiveness is often restricted, emphasizing the need for novel therapeutic strategies. Natural products have long been explored for drug development, and among them, polysaccharides have gained significant attention due to their biocompatibility, biodegradability, and multiple biological functions.
MethodsA comprehensive review examined contemporary research on the anticancer properties of natural polysaccharides, focusing specifically on their effects in lung cancer. The analysis included studies investigating their influence on cancer cell growth, immune system modulation, and therapeutic outcomes. Evidence from laboratory (in vitro), animal (in vivo), and clinical studies was evaluated to provide a comprehensive overview of their potential role in lung cancer management.
ResultsFindings from recent studies indicate that polysaccharides can effectively inhibit the proliferation of lung cancer cells, thereby slowing tumor development. These compounds also appear to enhance immune responses by activating various immune cells and regulating cytokine production. Furthermore, polysaccharides have been shown to positively affect the gut microbiota, which may contribute to improved drug efficacy and a reduction in resistance to chemotherapy.
DiscussionThe evidence suggests that natural polysaccharides exert multifaceted effects in the context of lung cancer treatment. Their ability to directly suppress tumor growth, modulate the immune system, and interact with the gut microbiome positions them as promising adjuncts to existing therapies. However, the precise molecular mechanisms underlying these effects are not yet fully understood, and variability in study designs warrants cautious interpretation of the results.
ConclusionNatural polysaccharides represent a promising complementary approach for lung cancer therapy, given their potential to inhibit tumor progression, enhance immune function, and improve the effectiveness of conventional drugs. Continued research is essential to fully elucidate their mechanisms of action and to translate these findings into effective clinical interventions.
-
-
-
Microbial-Derived Anti-Cancer Compounds: Advances in Drug Discovery, Bioengineering, and Therapeutic Applications
Authors: Ekta Tyagi, Divya Jain, Rajabrata Bhuyan and Anand PrakashAvailable online: 01 September 2025More LessIntroductionMicrobial metabolites represent a valuable source of bioactive compounds with promising anticancer properties. However, conventional drug discovery approaches are time-intensive and resource-demanding.
MethodsRecent developments in artificial intelligence (AI), machine learning (ML), molecular docking, and quantitative structure-activity relationship (QSAR) modeling have been examined for their role in the identification and optimization of microbial metabolites.
ResultsAI-driven approaches have significantly enhanced compound screening and prediction of therapeutic efficacy. Nanocarrier-based drug delivery systems have improved the bioavailability, specificity, and stability of microbial metabolites while minimizing systemic toxicity. Despite these advancements, challenges remain in clinical translation due to the lack of in vivo validation and comprehensive pharmacokinetic data.
DiscussionThis review highlights the integration of advanced computational tools and nanotechnology in accelerating the discovery and delivery of microbial-derived anticancer agents.
ConclusionFuture directions should focus on integrating AI with synthetic biology to engineer microbial strains capable of producing enhanced bioactive compounds. Additionally, leveraging nanotechnology could refine targeted delivery mechanisms. A deeper understanding of molecular pathways and drug resistance mechanisms is essential to support the development of combination therapies. Overall, microbial-derived compounds hold substantial potential in advancing precision oncology.
-
-
-
Long Non-Coding RNA VPS9D1-AS1 in Human Cancer: Functions, Mechanisms, and Clinical Utility
Authors: Jingjie Yang, Haodong He, Haoran Liu, Zhouya Xu, Li Li, Houdong Li and Chengfu YuanAvailable online: 01 September 2025More LessIntroductionVPS9 domain-containing 1 antisense RNA 1 (VPS9D1-AS1), also known as c-Myc-upregulated lncRNA (MYU) and FAK-interacting and stabilizing lncRNA (FAISL), is a novel long non-coding RNA (lncRNA) located at the human chromosome 16q24.3 locus. It has been reported to be highly expressed in various human cancers and associated with poor clinical pathological features and unfavorable prognosis in eight of the malignant tumors.
MethodsA comprehensive literature search was conducted using PubMed, Web of Science, and Google Scholar databases to identify relevant articles on “VPS9D1-AS1”, “MYU”, or “FAISL”. Only peer-reviewed publications were included, and articles related to oncology were specifically collected.
ResultsMechanistically, VPS9D1-AS1 serves as a key regulator in four molecular models: signal, scaffold, guide, and decoy. These functions allow it to regulate the expression of target genes and activation of signaling pathways, thereby influencing the malignant phenotype of tumors.
DiscussionThe diverse molecular mechanisms of VPS9D1-AS1 highlight its significant role in the development and progression of various cancers. Its ability to act as a signal, scaffold, guide, and decoy suggests that it can influence multiple aspects of tumor biology, including proliferation, invasion, and metastasis.
ConclusionVPS9D1-AS1 plays a significant role in the development and progression of various cancers through its diverse molecular mechanisms. Further research on VPS9D1-AS1 may provide valuable insights, which may facilitate the development of new diagnostic and therapeutic strategies for cancer.
-