Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry - Anti-Cancer Agents) - Online First
Description text for Online First listing goes here...
46 results
-
-
Evaluation of Anticancer Potential in Human Colorectal Carcinoma HCT-116 Cells by Fungal-Mediated Zinc Oxide Nanoparticles
Available online: 27 August 2025More LessIntroductionChemotherapy faces limitations such as toxicity and resistance, necessitating novel cancer treatments. Green-synthesized zinc oxide nanoparticles (ZnO-NPs) have attracted attention for their safety, biocompatibility, and therapeutic potential. This study investigates the anticancer efficacy of ZnO-NPs synthesized using the extracellular matrix of Aspergillus biplanus against colorectal cancer cell lines (HCT-116).
MethodsZnO-NPs were synthesized extracellularly using A. biplanus fungal extract. The nanoparticles were characterized through UV-Vis spectrophotometry, showing an absorbance peak at 375 nm, and scanning electron microscopy (SEM), which determined their morphology and size. The anticancer activity was evaluated in vitro using HCT-116 cells. Reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP) were assessed to understand the mechanism of cytotoxicity. In vivo studies were proposed for further validation.
ResultsThe synthesized ZnO-NPs appeared pale white and exhibited a characteristic absorbance at 375 nm. SEM revealed spherical particles ranging from 35–150 nm. The ZnO-NPs showed strong anticancer activity with an IC50 value of 40.6 µg/mL. ROS levels increased significantly in treated cells, while the MMP decreased to 77.25% compared to 100% in controls.
DiscussionZnO-NPs exerted cytotoxic effects via ROS generation and mitochondrial dysfunction. These results underscore the nanoparticles’ ability to induce apoptosis in cancer cells through oxidative stress pathways.
ConclusionBiogenically synthesized ZnO-NPs from A. biplanus show promise as eco-friendly anticancer agents. Further in vivo studies are recommended to confirm their therapeutic potential.
-
-
-
The Hematological Variations and Effect of Cadmium Induced Toxicity on Mammary Tumors Development in Albino Mice. A Comparative Model Study on the Effect of Heavy Metals in Human Breast Cancer
Authors: Saba Munir, Yasir Nawaz, Fouzia Tanvir and Khalid Mahmood AnjumAvailable online: 08 August 2025More LessIntroductionBreast cancer develops in breast tissues, in ducts and lobules. It affects both genders, though it is uncommon in men. Hematological variations are important considerations and deficiencies in metals can negatively impact human health. Cadmium is highly toxic and plays role in breast cancer progression. This study was designed for hematological variations and cadmium induced toxicity in mice and humans causing breast cancer.
MethodsMice, obtained from local supplier, housed at university laboratory for 11 weeks, exposed to cadmium. Following dissection, blood and organs were harvested for examination. Histological analysis of liver and mammary gland tissues was conducted.
ResultsAffected mice had higher Hb, RBC, HCT, MCV, and MCH, while humans showed lower Hb, HCT, and MCV but similar RBC and MCH. Other blood values also show changes. Histopathology revealed changes in mammary glands (higher cadmium led to increased fat deposition, degeneration of alveolar epithelial cells, and a reduction in alveolar milk lumen size, indicating compromised glandular function) and Liver damage (vacuolation, lipid accumulation, fibrosis, and collagen deposition, was noticeable with prolonged cadmium). These changes causes liver fibrosis and impaired mammary gland function.
DiscussionThe cadmium exposure induces distinct hematological alterations and severe tissues damage, reflecting species-specific responses. The observed liver fibrosis and mammary gland dysfunction emphasize cadmium’s potential to compromise critical organ functions over time.
ConclusionSignificant effects of cadmium exposure in mice were observed. Histological damage was seen in mammary glands and liver. Further research on protective measures and dose-response relationships for cadmium exposure is needed.
-
-
-
Anticancer Efficacy and Metabolomic Profiling of Punica granatum Leaf Extracts:
Available online: 06 August 2025More LessIntroductionCurrent research focuses on identifying and analyzing bioactive metabolites with significant therapeutic properties derived from Punic granatum L. (Pomegranate) leaves. Methods: The biological potential of these metabolites was evaluated through anticancer activity. In contrast, LC-QTOF-MS and GC-QTOF-MS methods were used to profile the metabolites. In silico molecular docking was performed using various online and offline tools to validate the active metabolites.
ResultsPAC exhibited significant anticancer activity. The identified metabolites were screened, and 40 compounds from different categories were chosen for further in silico interaction studies.
DiscussionThe molecular docking analysis discovered lead molecules that exhibited promising binding energy scores, efficiency, and stable modulation with specific protein domains. However, clinical trials are required for the applications of the lead molecules in the design of anticancer drugs.
ConclusionThe findings from both in vitro and in silico analyses support the notion that the P. granatum Acetone Extract (PAC) is an excellent source of potential metabolites with therapeutic properties. According to the findings, this research enhances the treatment of human breast cancer and validates several plant traditions for their numerous benefits.
-
-
-
Bioinformatics And Experimental Insights Into Sotorasib Resistance Mechanisms in Non-small-cell Lung Cancer
Authors: Dongbing Li and Guizhen LyuAvailable online: 06 August 2025More LessIntroductionThis study aims to identify the key genes and pathways associated with sotorasib resistance in Non-Small Cell Lung Cancer (NSCLC) using bioinformatics analyses and experimental validation, with a focus on uncovering the potential mechanisms underlying resistance.
MethodsWe compared gene expression profiles between sotorasib-resistant (SR) and non-resistant NSCLC cell lines using the GSE229070 dataset and between NSCLC tissues and adjacent normal tissues using the GSE18842 dataset. Differentially expressed genes (DEGs) were identified and intersected across datasets using the Venn diagram package. Functional enrichment analysis was performed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). The transcriptional activity and prognostic impact of key genes were evaluated using the UALCAN portal and Kaplan-Meier Plotter, respectively. The correlation between gene expression and immune cell infiltration was analyzed using the TIMER database, and co-expressed genes were explored using LinkedOmics. qRT-PCR and Western blot were used to validate the expression of AREG in parental and SR cell lines.
ResultsWe identified 33 overlapping DEGs, including TENM2, COL12A1, COL5A2, and LRRC15 (upregulated) and AREG (downregulated). AREG expression was significantly lower in NSCLC patients and associated with worse survival outcomes. AREG expression was also correlated with the levels of immune cell infiltration. Functional enrichment analysis revealed that AREG was associated with pathways including the NOD-like receptor signaling pathway, focal adhesion, DNA replication, and homologous recombination. Experimental validation confirmed that AREG mRNA and protein levels were significantly reduced in HCC78-SR cells compared to parental HCC78 cells.
DiscussionThe downregulation of AREG is closely associated with sotorasib resistance in NSCLC, potentially contributing to resistance through alterations in signaling pathways and the tumor immune microenvironment. This finding aligns with previous studies on AREG's role in drug resistance, highlighting its potential as a therapeutic target. However, limitations include reliance on publicly available datasets and the need for further validation in clinical cohorts.
ConclusionThe study identifies AREG as a key gene associated with sotorasib resistance in NSCLC, suggesting its potential as a biomarker and therapeutic target. Further research is needed to elucidate the mechanisms underlying AREG's role in resistance and to explore its clinical significance.
-
-
-
dHG-5 Exhibits Dual Efficacy of Anti-Metastatic and Anti-hypercoagulability in Mice by Inhibiting Heparanase and Intrinsic Coagulation Pathway
Authors: Ziheng Tong, Zhipeng Xu, Wen Yang, Huaizheng Song, Shuguo Zheng and Lutan ZhouAvailable online: 06 August 2025More LessIntroductionCancer metastasis and associated thrombosis are significant contributors to cancer-related mortality, necessitating therapeutic strategies that simultaneously address both issues. This study aimed to evaluate the dual anti-metastatic and anti-hypercoagulability properties of dHG-5, a low-molecular-weight fucosylated glycosaminoglycan derived from the sea cucumber Holothuria fuscopunctata.
MethodsThe heparanase-inhibitory and anticoagulant effects of dHG-5 were assessed in vitro using biochemical assays. The impact of dHG-5 on 4T1 cell migration and invasion was evaluated using Transwell assays. The anti-metastatic and anti-hypercoagulability efficacy of dHG-5 was further tested in a 4T1 mammary carcinoma mouse model, with enoxaparin (LMWH) used as a control.
ResultsdHG-5 exhibited potent heparanase inhibition (IC50 = 91.0 nM) and significantly reduced 4T1 cell migration and invasion at 4.0 µmol/L. In vivo, dHG-5 reduced lung metastasis without affecting tumor growth or proliferation. At a dose of 20 mg/kg, dHG-5 prolonged activated partial thromboplastin time (APTT) from 23.5 ± 1.85 s to 30.4 ± 3.36 s, effectively reversing hypercoagulability in tumor-bearing mice. Compared to low-molecular-weight heparin, dHG-5 selectively prolonged APTT with negligible effects on prothrombin time and thrombin time.
DiscussionThe findings highlighted the dual-action mechanism of dHG-5, namely inhibiting heparanase and selectively targeting the intrinsic coagulation pathway. This selective action minimized bleeding risk, a common issue with traditional anticoagulants. However, this study focused on a single cancer type and the use of a mouse model, which may not fully represent human pathophysiology. We would explore dHG-5's effects across different cancer types and investigate its potential synergistic effects with existing cancer therapies in the future.
ConclusiondHG-5 suppressed metastasis and hypercoagulability through heparanase inhibition and selective action on the intrinsic coagulation pathway. These findings highlight dHG-5 as a promising dual-action therapeutic candidate for managing metastasis and cancer-associated thrombosis, offering a safer alternative to traditional anticoagulants.
-
-
-
The Potential of Next-generation Multi-functional Nanoplatforms for Breast Cancer
Authors: Shreya Gupta, Tanmay J Urs, Navya Aggarwal, Shinjini Sen and Banashree BondhopadhyayAvailable online: 24 July 2025More LessThe next-generation nanoparticles overcome the drawbacks of early nanoplatforms by integrating multiple functions, such as drug delivery, controlled drug release, and combination therapy, into a single system. This study examines the biomedical applications of quantum dots, carbon nanotubes, superparamagnetic iron oxide nanoparticles, and layered double hydroxides for the delivery of breast cancer drugs. They are termed as “next-generation” nanoparticles, as they are advanced nanocarriers that offer a comprehensive and alternative approach towards breast cancer treatment, providing enhanced specificity and efficacy compared to their predecessors. The development of these nanoplatforms has significantly enhanced drug bioavailability and reduced toxicity. A comprehensive analysis of a nanotechnology-based drug delivery system was conducted. The keywords used for this review were “Breast Cancer”, “Targeted Drug Delivery”, “Quantum Dots”, “Carbon Nanotubes”, “Layer Double Hydroxides”, and “Superparamagnetic Iron Oxide Nanoparticles”. The inclusion criteria consisted of studies focusing on breast cancer, targeted drug delivery, and therapeutic applications of these nanocarriers. In contrast, exclusion criteria included studies focusing on the synthesis of nanocarriers and the diagnostic applications of these nanostructures. The study underscores their mechanisms, limitations, and future development directions. Additionally, the study tracks the evolution of the nanocarriers since their early discovery. Next-generation nanocarriers (QDs, CNTs, SPIONs, and LDHs) have strong therapeutic potential owing to their precisely engineered properties, such as size, shape, morphology, and surface modifications. Their trigger-initiated drug release mechanisms enable targeted delivery with a better rate of tumor penetration, while their ability to co-deliver multiple therapeutic agents addresses drug resistance issues and provides synergistic effects. Comparative analyses have revealed that these advanced nanoplatforms significantly outperform early-generation carriers in terms of bioavailability, reduced toxicity, and treatment efficacy across various breast cancer types. Next-generation nanoplatforms offer unprecedented opportunities for targeted and efficient cancer treatment. Continued research and innovation are necessary to address existing challenges and to optimize their therapeutic potential for clinical applications.
-
-
-
The Role of Kinase Inhibitors in Cancer Neuroscience: Mechanisms, Therapeutic Potential, and Future Directions
Available online: 21 July 2025More LessIntroductionCancer progression is increasingly understood to be influenced by neural mechanisms, including neurotransmitter signaling, neurotrophic factor activity, neuroinflammation, and neurogenic inflammation. These neurobiological interactions contribute to tumor proliferation, angiogenesis, and metastasis. Kinase inhibitors, a class of targeted therapies that block dysregulated kinase activity, have demonstrated promise not only in direct tumor suppression but also in modulating neural pathways associated with cancer progression.
MethodsThis review examines the role of kinase inhibitors in modulating cancer-associated neural mechanisms. A comprehensive literature search was conducted to identify studies exploring the effects of kinase inhibition on: (1) neurotransmitter signaling pathways; (2) neurotrophic factors such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF); (3) neuroinflammation through glial cell modulation; and (4) neurogenic inflammation. Additionally, we assessed the impact of kinase inhibitors on tumor-induced axonogenesis and stress-related signaling. Clinical relevance was evaluated through analysis of preclinical models, human case studies, and outcomes from relevant clinical trials.
ResultsKinase inhibitors were found to significantly modulate neural factors that facilitate tumor growth. Specifically, they can suppress neurotrophic signaling (e.g., NGF/TrkA, BDNF/TrkB), inhibit glial activation, reduce pro-inflammatory cytokine production, and block neurotransmitter-induced proliferation. Inhibition of stress-responsive kinases such as p38 MAPK and JNK also disrupted tumor-associated axonogenesis and inflammation. Clinical trials demonstrate improved outcomes in cancers such as glioblastoma, breast cancer, and pancreatic cancer when kinase inhibitors are employed with consideration of neural mechanisms.
DiscussionThese findings support the emerging concept of targeting the neural tumor microenvironment as a therapeutic strategy. Kinase inhibitors represent a dual-action approach, suppressing both cancer cell intrinsic growth pathways and the neural factors that sustain them. However, several challenges persist, including resistance mechanisms, variability in patient neural profiles, and off-target effects. Future research should focus on the development of neural-specific kinase inhibitors, the use of neural biomarkers for therapy selection, and the integration of neuro-oncology into personalized treatment plans.
ConclusionKinase inhibitors offer a promising frontier in cancer treatment by targeting neural mechanisms that contribute to tumor progression. While current evidence is encouraging, further investigation is required to optimize their use within neuro-oncology. Personalized approaches and novel targets within the neural-cancer axis will be essential for translating this strategy into clinical practice and improving long-term patient outcomes.
-
-
-
Innovative Nanocarriers: Magnetosomes in the Fight against Cancer
Authors: Shivani Yadav and Manoj Kumar MishraAvailable online: 17 July 2025More LessRecent advancements in medication formulations and drug delivery systems over the past two decades have improved patient adherence and pharmacological responses. Efficient, target-specific medication delivery remains challenging, with many current systems designed to minimize drug loss and degradation. Magnetosomes, as nanocarriers, show promise for delivering antibodies, vaccine DNA, and siRNA, enhancing the stability of chemotherapeutics, and enabling targeted delivery to malignant tumors. Targeted drug delivery is crucial in cancer treatment, as anticancer drugs often cannot differentiate between healthy and malignant cells, causing side effects and systemic toxicity. Magnetosome-based drug delivery offers a potential solution, minimizing adverse effects and promoting drug accumulation at the target site. This review covers the design, development, and advancements in magnetosome-based drug delivery for cancer therapy.
-
-
-
Polyamines in Cancer: Mechanisms, Metabolic Targets, and Therapeutic Opportunities
Available online: 17 July 2025More LessIntroductionPolyamine metabolism is essential for cancer cell growth, with enzymes like ornithine decarboxylase (ODC) and S-adenosylmethionine decarboxylase (AdoMetDC) playing key roles in polyamine (PA) biosynthesis. These polyamines (putrescine, spermidine, and spermine) regulate vital cellular processes, including DNA replication, protein synthesis, and cell cycle progression. Dysregulated polyamine metabolism is common in cancer, making ODC and AdoMetDC attractive therapeutic targets. This review highlights polyamines’ role in cancer and explores combination therapies targeting polyamine metabolism and critical signaling pathways for improved clinical outcomes.
MethodsA comprehensive analysis of both historical and recent literature on polyamine metabolism in cancer was performed using PubMed, which provides access to over 37 million citations from biomedical literature. Expression data for key polyamine biosynthetic enzymes, ODC and AdoMetDC, were obtained from the UALCAN portal - an interactive web resource for the analysis of cancer OMICS data. The IUPAC names of drugs and inhibitors targeting the polyamine pathway were retrieved from the PubChem database and used to generate molecular structures using the BIOVIA Draw 2025 program. Additionally, the ClinicalTrials.gov database was explored to identify ongoing and completed clinical research studies, as well as to gather detailed information on therapeutic agents targeting polyamine metabolism.
ResultsAberrant polyamine metabolism in cancer is driven by oncogenic pathways like MYC, Akt, and mTOR. MYC upregulates ODC1, promoting polyamine dysregulation. Defects in enzymes such as MTA phosphorylase (MTAP) enhance cancer cell sensitivity to inhibitors of purine/pyrimidine synthesis and the ubiquitin-proteasome pathway, suggesting alternative therapeutic strategies.
DiscussionTherapeutic strategies combining polyamine biosynthesis inhibition with targeting nucleotide synthesis or proteasome function have shown synergistic potential. However, the dual nature of polyamines - supporting both, tumor growth and ferroptotic cell death - poses a therapeutic challenge. Balancing these effects is key to designing effective interventions. Advancing this field requires not only selective inhibitors but also a deeper understanding of context-dependent polyamine functions in tumor biology.
ConclusionDeveloping more potent inhibitors with improved drug-like properties is crucial for advancing polyamine-targeted therapies and positioning this field at the forefront of cancer research.
-
-
-
Design, Synthesis and Biological Evaluation of New 4-(4-(Methylsulfonyl)Phenyl)-6-Phenylpyrimidin-2-Amine Derivatives as Selective Cyclooxygenase (COX-2) Inhibitors
Available online: 16 July 2025More LessIntroductionCyclooxygenase, an enzyme that occurs in at least two distinct variants (COX-1 and COX-2), is the target of classical inhibitors, which lack selectivity and inhibit both types of COX. However, a recent approach focuses explicitly on inhibiting COX-2, commonly found in inflamed tissue, resulting in fewer adverse effects than COX-1 inhibitors.
MethodsA series of 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine derivatives were synthesized through a two-step process. First, 4-substituted acetophenones underwent base-catalyzed Claisen-Schmidt condensation with 4-(methylsulfonyl)benzaldehyde to yield chalcones, which were then cyclized with guanidine hydrochloride under basic reflux conditions. Molecular docking was performed using AutoDock Vina software. The inhibitory activities of COX-1 and COX-2 were evaluated using enzymatic assays. Antiplatelet aggregation was measured via a turbidimetric method, and antiproliferative activity was assessed using the MTT assay.
ResultsThe in vitro experiments on COX inhibition revealed that a substantial number of the synthesized compounds presented a strong suppressive effect against COX-2. The assessment of antiplatelet aggregation activity indicated that most of the derivatives effectively inhibited ADP-induced platelet aggregation. Compound 4i exhibited the most potent antiproliferative activity, comparable to cisplatin. The docking studies and molecular modeling results demonstrated that the designed compounds, except for 4b, exhibited a binding behavior comparable to that of celecoxib. In addition, the insertion of the SO2Me moiety within the secondary binding site of COX-2 was observed.
DiscussionThese findings suggest that the structural modifications introduced in the synthesized derivatives contribute significantly to their selective COX-2 inhibition and antiplatelet properties. The correlation between docking results and biological assays supports the rationale behind the design of the compound.
ConclusionThe 4-(4-(methylsulfonyl)phenyl)-6-phenylpyrimidin-2-amine exhibits unique properties as a COX-2 inhibitor, displaying effective inhibition of COX-2 while showing minimal interaction with the COX-1 enzyme. Furthermore, our study revealed that most of these compounds exhibited inhibitory effects on ADP-induced platelet aggregation.
-
-
-
Investigating the Therapeutic Potential of Cisplatin- and Rutin-Loaded Nanoliposomes against Colorectal Cancer Cells
Available online: 16 July 2025More LessIntroductionColorectal cancer is an important cause of cancer-related mortality, necessitating innovative therapies to improve efficacy and reduce side effects. This study explores the potential of Cisplatin and Rutin-loaded nanoliposomes (Cis-NLs and Rut-NLs) for anti-colorectal cancer activity.
MethodsCis-NLs and Rut-NLs were prepared using thin-film hydration, achieving encapsulation efficiencies of 95.5% and 62.5%, respectively. Drug release studies revealed controlled profiles, with Cis-NLs showing a complete release (100%) and Rut-NLs reaching 23.48% over 48 hours. Stability assessments demonstrated minimal changes in size, polydispersity index (PDI), and zeta potential over three months. Encapsulation efficiency decreased slightly for Cis-NLs (92.87%) and significantly for Rut-NLs (26.55%). Several tests were performed to evaluate the biological activity of this combination on colorectal cancer cells and HDF cells to check its selectivity.
ResultsIn vitro cytotoxicity studies on HT29 colorectal cancer cells revealed IC50 values of 1.72 µg/mL for free Cisplatin, 2.35 µg/mL for Cis-NLs, >100 µg/mL for free Rutin, and 63.33 µg/mL for Rut-NLs. A combination of Cis-NLs and Rut-NLs reduced the IC50 to 2.2 µg/mL. Selective toxicity evaluation using human dermal fibroblasts showed an IC50 of 79.24 µM for cisplatin, reduced to 63.3 µM in Cis-NLs, with Rut-NLs demonstrating negligible toxicity.
DiscussionWound healing assays confirmed significant inhibition of cell migration, with wound closure reduced from 62.41% in controls to 34.35% in treated groups. Utilizing nanotechnology, liposomal formulations were synthesized to enhance drug delivery and therapeutic synergy.
ConclusionThese results highlight the potential of Cisplatin and Rutin-loaded nanoliposomes as a combination therapy for colorectal cancer.
-
-
-
The Safety and Efficacy of Anti-LAG-3 for Patients with Melanoma: A Systematic Review and Meta-analysis Study
Available online: 11 July 2025More LessIntroductionMelanoma, an aggressive skin cancer, has seen treatment advancements with immune checkpoint inhibitors (ICIs) like ipilimumab and nivolumab. Despite improved survival rates, resistance remains a challenge. The recent focus on lymphocyte activation gene-3 (LAG-3) inhibitors, such as relatlimab, shows promise in combination therapies, potentially improving outcomes with fewer adverse effects. This review evaluates the safety and efficacy of anti-LAG-3 antibodies in melanoma treatment.
MethodsThis systematic review and meta-analysis, following the PRISMA guidelines and registered in PROSPERO (CRD42024565756), assessed anti-LAG-3 antibodies in melanoma treatment. A thorough search across PubMed, Embase, Scopus, and Web of Science up to January 2024 yielded relevant studies. Data on study characteristics, patient demographics, disease characteristics, treatment details, and clinical outcomes were extracted. Quality assessment was performed using the MINOR criteria. The meta-analysis, using STATA and random-effects models, addressed heterogeneity to determine safety and efficacy outcomes.
ResultsWe examined the clinical benefit of this combination therapeutic approach by measuring several primary endpoints and running a meta-analysis to determine the pooled estimate of 6-month progression-free survival (PFS), 1-year PFS, 6-month duration of response (DoR), 1-year DoR, 1-year overall survival (OS), 2-year OS, partial response (PR), complete response (CR), objective response rate (ORR), disease control rate (DCR), stable disease (SD), and progressive disease (PD) for patients diagnosed with melanoma. Our analysis showed 66% of any grade treatment-related adverse events (trAEs) (95% CI: 51%-81%), 19% of grade ≥ 3 trAEs (95% CI: 11%-27%), 12% of any grade AEs leading to discontinuation (95% CI: 9%-14%), and 8% of grade ≥ 3 AEs leading to discontinuation (95% CI: 6%-10%). 76% of any grade overall AEs (95% CI: 34%-100%), and 33% of grade ≥ 3 overall AEs (95% CI: 15%-50%). The most common AEs were fatigue, pneumonitis, rash, pruritus, colitis, hepatitis, diarrhea, hypothyroidism, thyroiditis, and adrenal insufficiency.
DiscussionThis systematic review and meta-analysis provide comprehensive evidence regarding the safety and efficacy of anti-LAG-3 antibodies in melanoma therapy. Pooled data reveals encouraging outcomes across several key endpoints, including PFS, OS, and ORR. While trAEs were common (66% for any grade and 19% for grade ≥3), most were manageable.
ConclusionAnti-LAG-3 therapy is an active and safe treatment that shows promising results in melanoma treatment.
-
-
-
Lifileucel Therapy for Metastatic Melanoma: Advancements in Tumor-infiltrating Lymphocyte-based Immunotherapy
Available online: 04 July 2025More LessMetastatic melanoma is an aggressive malignancy with limited treatment options at advanced stages. Lifileucel, an FDA-approved autologous Tumor-Infiltrating Lymphocyte (TIL) therapy, marks a major advancement in immunotherapy, particularly for patients who fail conventional treatments like immune checkpoint inhibitors and targeted therapies. The mechanism of lifileucel involves the ex vivo expansion of patient-derived TILs to boost immune responses against melanoma cells. These expanded TILs are re-infused into patients, enhancing tumor-specific cytotoxicity and modulating the tumor microenvironment for sustained immune activation. Clinical trials have demonstrated its efficacy, with the overall response rate (ORR) reaching up to 36% in heavily pretreated populations, offering durable responses and improved progression-free survival compared to traditional therapies. The personalized approach of lifileucel, leveraging the patient’s own T-cell repertoire, highlights its potential for precision oncology by targeting individual tumor profiles. Its integration with combination therapies, particularly immune checkpoint inhibitors, shows promising synergistic effects, broadening its clinical applicability. In addition to clinical success, the role of lifileucel in influencing the melanogenesis pathway offers insights into optimizing therapeutic strategies for melanoma. Ongoing research focuses on enhancing TIL functionality, overcoming challenges like tumor-induced immune suppression, and extending the applicability of lifileucel to other solid tumors. This breakthrough therapy not only addresses a critical unmet need in melanoma treatment but also represents a paradigm shift toward personalized medicine in oncology. Lifileucel underscores the potential of TIL-based approaches to revolutionize cancer care, setting the stage for future advancements in immunotherapy.
-
-
-
Anticancer Compounds from Myxobacteria: Current Scenario and Future Perspectives
Authors: Swati Sihag, Shweta Sinha and Ramandeep KaurAvailable online: 04 July 2025More LessNatural products and their derivatives have played a dominant role in the development of therapeutic agents. Traditionally, most of the natural products developed for the effective treatment of different diseases have been sourced from plants. Natural product discovery has seen a shift of focus towards microorganisms due to the chemical diversity of bioactive products they synthesize. Myxobacteria produce a large variety of novel chemical entities with diverse structures and varied bioactivities. In the last few decades, secondary metabolites from different genera of myxobacteria have been recognized as harbouring potent anticancer activity. Several analogs of these anticancer compounds have been prepared to address the limitations such as, poor solubility, high toxicity and low production yield, in order to obtain the compounds in higher quantities with better pharmacological properties and target selectivity. For example, a semi-synthetic derivative of epothilone obtained from a strain of myxobacterium has been approved for clinical use against taxane-resistant breast cancer. The anticancer compounds from myxobacteria target microtubules, the cytoskeleton, vacuolar ATPase, methionine aminopeptidase, exportin, the proteasome or translation elongation factor to exert anticancer activity. The focus of this review is on the promising anticancer compounds produced by myxobacteria, their targets and their mechanisms of action in cancer cells.
-
-
-
CD98 Light Chain LAT1 Tracers in PET-CT Diagnosis of Cancer Patients
By Pu XiaAvailable online: 02 July 2025More LessAmino acid-based PET tracers have become vital tools for non-invasive tumor imaging, offering greater specificity and sensitivity than conventional 18F-FDG. These tracers target amino acid transporters, particularly L-type Amino Acid Transporter 1 (LAT1), which is overexpressed in rapidly proliferating tumor cells. Various
18F-labeled amino acid tracers have been explored for imaging different malignancies, including gliomas, neuroendocrine tumors, and lung cancers. This review summarizes the performance of LAT1-specific radiotracers, comparing their uptake ratios, sensitivity, and specificity in cancer diagnosis. These tracers have led to significant advancements in tumor imaging, providing better diagnostic accuracy, enhanced tumor delineation, and reduced interference from inflammatory tissue. Although promising, the clinical utility of these tracers requires further research and clinical trials to refine their applications and optimize their role in routine clinical practice. Continued development will be crucial in making these tracers more effective and widely applicable for cancer diagnosis and treatment planning.
-
-
-
Secondary Malignancies of Chimeric Antigen Receptor T-cell Therapy: A Multidimensional Analysis of Mechanisms, Risk Factors, and Treatment Strategies
Authors: Ye Kang, Da-Sheng Dang, Xue Sun and Xiao ZhangAvailable online: 26 June 2025More LessChimeric Antigen Receptor T-cell (CAR-T) therapy represents a pioneering advancement in immunotherapy, demonstrating substantial clinical success in the treatment of hematologic malignancies, particularly in B-cell hematologic malignancies. This therapeutic approach involves the genetic modification of a patient's T-cells to express receptors specific to tumor antigens, thereby enabling the CAR T-cells to identify and eradicate tumor cells, which significantly enhances the patient's treatment prognosis. Despite the remarkable efficacy of CAR-T therapy, concerns regarding its safety have emerged during clinical implementation. Notably, research has indicated that CAR T-cell therapy may be associated with the development of secondary primary malignancies, prompting considerable apprehension within the clinical community regarding the long-term adverse effects of this treatment modality. This article aims to investigate the potential mechanisms responsible for the induction of secondary primary malignancies by CAR T-cells, evaluate the associated risk factors, and discuss therapeutic strategies to mitigate this issue. Furthermore, the article will explore future research directions focused on optimizing the safety profile of CAR-T therapy, thereby providing a theoretical foundation for the development of safer and more effective therapeutic interventions.
-
-
-
Screening of Bioactive Fractions from Balanites aegyptiaca and Pterocarpus marsupium for Anticancer Effects in HepG2 and U87MG Cells
Authors: Divya Vashishth, Mansi Yadav, Ajay Kumar, Gulshan Rohilla, Minakshi Vashist and Sudhir Kumar KatariaAvailable online: 23 June 2025More LessIntroductionCancer is a group of diseases caused by uncontrollable cell growth. Herbal medicines, derived from plants, have been used for centuries across cultures for their therapeutic benefits, effectively treating conditions like cancer. This study represents the anticancer effects of fractions of some medicinal plant extracts along with their apoptotic studies and their induction through p53-mediated Bax and Bcl-2 mRNA expression in HepG2 and U87MG cells.
MethodsThe fractionation of crude methanolic extracts was done using Column Chromatography and Thin Layer Chromatography. The fractions were analysed for cytotoxicity against both the cell lines by MTT assay. Cancer cells were treated with 2 most active fractions and their mechanism of apoptosis induction was assessed by Flow Cytometry studies and the mRNA expression levels of p53, Bax, and Bcl-2 were determined by Reverse Transcriptase PCR. The presence of phytoconstituents in the active fractions was analysed by GC-MS.
ResultsThe active fractions revealed the apoptosis induction in both the cell lines and the RT-PCR studies suggested the mechanism of apoptosis induction through upregulation of p53 and Bax and downregulation of Bcl-2 mRNA. The GC-MS analysis of active fractions from Balanites aegyptiaca and Pterocarpus marsupium revealed the presence of phytochemicals such as 4-O-Methylmannose, Oleic acid, Erucic acid, etc. which might have contributed to the anti-proliferative and apoptotic effects of these fractions.
Discussion4-O-Methylmannose was the major component identified with the highest peak area of 59%. The fractions from all the 4 plant extracts demonstrated significant cytotoxic effects on the liver (HepG2) and brain (U87MG) cancer cell lines, with particular emphasis on the active fractions BA FII, PM FII, and PM FIII. Additionally, the mechanisms of apoptosis induction through the modulation of p53, Bax, and Bcl-2 pathways, along with the presence of bioactive compounds further support the anticancer efficacy of these plant extracts. Also, to the best of our knowledge, this is the first study on fractions of Balanites aegyptiaca and Pterocarpus marsupium against U87MG cells.
ConclusionThe results highlight the promising potential of plant-derived natural products as anticancer agents. These findings provide valuable insight into the potential of herbal medicines and encourage further exploration of plant-based therapies for cancer treatment.
-
-
-
Advances in Metal-based Nanotechnology-based Optical Therapy for the Targeted Treatment of Colorectal Cancer
Authors: Huiling Zuo, Yuhang Jiao, Fengyu Wang, Junzi Wu and Wenling ChenAvailable online: 19 June 2025More LessColorectal cancer (CRC) is one of the most prevalent gastrointestinal malignancies in the world. To overcome clinical challenges, such as high postoperative recurrence rates and prominent resistance to chemotherapy, new therapeutic strategies are urgently needed. Phototherapy, particularly Photodynamic Therapy (PDT) and Photothermal Therapy (PTT), has unique advantages in selectively killing tumor cells. However, traditional Photosensitizers (PSs) and Photothermal Agents (PTAs) have inherent defects, such as limited tissue penetration depth, poor optical stability, and insufficient targeting ability, which severely restrict phototherapy in clinical applications. Significant advancements have been made in enhancing the phototherapeutic effects of metal nanomaterials in recent years. This progress can be attributed to their tunable optical properties, exceptional Photothermal Conversion Efficiency (PCE), and unique Surface Plasmon Resonance (SPR) effects. In this review, we systematically summarized the latest progress in research on the use of metal nanomaterials for the optical diagnosis and treatment of colorectal cancer. We focused on the mechanism by which typical nanomaterials such as gold, silver, and platinum enhance the therapeutic effect of PDT/PTT. Additionally, a comprehensive analysis was conducted to evaluate the application and potential of nano-optical sensitizers incorporating metallic cores such as gold, silver, iridium, platinum, iron, zinc, copper, ruthenium, and titanium for the diagnosis and treatment of Colorectal Cancer (CRC). This review may provide theoretical guidance for developing new-generation optical diagnostic and therapeutic strategies for treating colorectal cancer.
-
-
-
Unveiling the Vital Role of ACTA2-AS1 in Human Cancers: Molecular Mechanisms and Clinical Applications
Authors: Haodong He, Lumei Xiang, Baoqin Pi, Jingjie Yang, Wenjin Peng, Moyu Li, Haoran Liu, Xinyan Zheng, Haoyi Liu, Yuxiang Peng, Pengbo Zhang, Jiahe Zhang, Xin Chen, Yanlin Zhang, Meiyan Shuai, Feng Xu, Yan Cai and Chengfu YuanAvailable online: 19 June 2025More LessBackgroundThe smooth muscle α-actin 2-antisense 1 (ACTA2-AS1), also known as ZXF1, is an emerging cancer-associated long non-coding RNA (lncRNA) that has garnered significant attention in recent years. ACTA2-AS1 is situated on human chromosome 10 at location 10q23.31, comprising five exons and a single transcript. The aberrant expression of ACTA2-AS1 has been noted in 10 malignant tumors, correlating significantly with unfavorable clinicopathological characteristics and poor patient prognosis.
ObjectiveThis review encapsulates recent progress in ACTA2-AS1 research, examining its expression profile, biological functions, molecular mechanisms, and anticipated influence on cancer diagnosis, treatment, and prognosis, emphasizing its potential as a novel therapeutic target based on lncRNA and its prognostic utility as a biomarker.
MethodsBased on a comprehensive search of the PubMed database for the biological function of lncRNA ACTA2-AS1 in malignant tumors, the current research is systematically summarized and critically analyzed.
ResultsACTA2-AS1 plays a complex role in various biological processes in tumor cells, encompassing proliferation, apoptosis, and cell cycle arrest. It also contributes to migration, invasion, epithelial-mesenchymal transition (EMT), and drug resistance. Mechanistically, ACTA2-AS1 influences oncogenic or tumor-suppressive effects via a complex regulatory network. It can adsorb specific 5 miRNAs as competitive endogenous RNAs (ceRNAs), thereby mitigating the suppression of downstream mRNA targets implicated in tumorigenesis (e.g., SOX7, KLF9, CXCL2, BCL2L11, etc.) and modulating their downstream signaling pathways (e.g., Wnt5a/PKC, SMAD3, mTOR, etc.), demonstrating a broad spectrum of dual roles in carcinogenesis and tumor suppression.
ConclusionACTA2-AS1 is a promising biomarker and molecular target for the treatment of cancer.
-
-
-
Exploring Natural Coumarins: Breakthroughs in Anticancer Therapeutics
Authors: Emine Terzi, Beyza Ecem Oz-Bedir and Jean-Yves WinumAvailable online: 19 June 2025More LessNatural coumarins, a class of compounds found abundantly in various plants, are emerging as promising candidates in fight against cancer. Their ability to target multiple cancer-related processes has drawn significant interest from researchers. Natural coumarins exhibit anticancer effects through mechanisms such as inducing apoptosis, which is the programmed death of cancer cells, inhibiting cell proliferation, and disrupting angiogenesis, the process by which tumors develop their own blood supply to sustain growth. What makes coumarins particularly intriguing is their broad-spectrum activity against various types of cancer cells, from breast to lung to colon cancers. They interact with key molecular pathways that drive tumor progression, making them versatile agents in cancer therapy. Additionally, unlike many conventional chemotherapy drugs, natural coumarins generally have lower toxicity, which could translate to fewer side effects for patients. This characteristic makes them attractive as potential standalone treatments or as complementary therapies that enhance the efficacy of existing drugs while minimizing harm to normal cells. Ongoing research continues to explore the therapeutic potential of natural coumarins to better understand their full therapeutic potential and how they might work in combination with other anticancer agents. As the body of evidence grows, these natural compounds could become integral components of more effective and less harmful cancer treatment regimens, offering new hope for patients facing this challenging disease. This review was conducted by systematically analyzing the existing literature on natural coumarins and their anticancer potential.
-
-
-
Determination of PD-L1 Expression in Circulating Tumor Cells of Hypopharyngeal and Laryngeal Cancers and Correlation with Tissue Detection
Authors: Chen Li, Hongyu Zhu, Qin Lin, Wei Chen, Xiaoting Huang and Desheng WangAvailable online: 12 June 2025More LessBackgroundPD-L1 plays a pivotal role as an immunoregulatory checkpoint within the immune system, exerting a critical influence on the internal functioning and survival mechanisms of cancer cells. Our study aimed to elucidate the clinical significance of PD-L1 expression in circulating tumor cells (CTCs) derived from individuals afflicted with Hypopharyngeal and Laryngeal Cancers (HLC).
ObjectiveTo verify the relationship between the expression of PD-L1 in CTCs in HLC and the consistency in tissue and the preliminary clinical application.
MethodsA laboratory-based experimental study was carried out at Fujian Medical University Union Hospital. CTCs were identified using an immunomagnetic positive sorting methodology. Simultaneous detection was conducted on the CTC levels among PD-L1 positive patients, aiming to ascertain the dynamic relationship between real-time CTC fluctuations and the clinicopathological indices of the patients. This investigation encompassed a cohort of 38 individuals, wherein PD-L1 expression analysis was executed to delineate CTC variations in PD-L1-positive patients.
ResultsThe constructed immunolipid magnetic nano-beads demonstrated pronounced efficacy in capturing CTCs, and the lipid nanoparticles exhibited noteworthy capture efficiency coupled with minimal cytotoxic effects. The assessment of PD-L1 expression consistency between CTCs and tissue specimens revealed a substantial agreement surpassing 70%. Furthermore, inhibition of PD-L1 yielded a significant elevation in the cytokine TNF-α levels, accompanied by a concomitant reduction in IL-10 levels.
ConclusionThe CTC sorting system devised in this investigation boasts attributes of remarkable specificity and sensitivity. By virtue of PD-L1 expression analysis, it holds the potential to offer instructive implications for tailoring individualized treatments in clinical scenarios.
-
-
-
HSP Inhibitor Sensitize Resistant MCF-7 Cells to Doxorubicin through Suppressing HSP90AB4P Pseudogene and HSPB1 Expression
Available online: 11 June 2025More LessIntroductionDoxorubicin, a first-line chemotherapeutic agent, often faces resistance in breast cancer subtypes, leading to treatment failure. HSPs (Heat shock proteins), especially HSP90, and their pseudogenes like HSP90AB4P have been implicated in fostering resistance mechanisms by regulating apoptotic and survival pathways in cancer cells. The aim of this study is to investigate how inhibiting HSPs using a novel pyro-salicylic acid derivative (7A) can sensitize doxorubicin-resistant breast cancer cells (MCF-7/ADR) to chemotherapy.
MethodsThe potential role of HSP inhibitor with doxorubicin at different concentrations was tested to reveal synergetic and additive effects by combination index (CI) analysis. Cell cycle analysis, apoptosis assays, and gene expression profiling via PCR arrays supported the impact of 7A over MCF-7/ADR cells' molecular pathways.
ResultsHSP inhibitor efficiently suppressed doxorubicin resistance over invasive breast ductal carcinoma and has a synergetic effect. The inhibitor decreases HSP90AB4P and small HSPB1 expression efficiently.
ConclusionOur findings demonstrate that 7A suppresses doxorubicin resistance in MCF-7/ADR cells by reducing the expression of HSP90AB4P and small HSPB1, leading to an increase in apoptosis and cell cycle arrest. The combination of 7A and doxorubicin exhibits a synergistic effect (CI < 1), enhancing cytotoxicity and overcoming resistance mechanisms. The cells are driven to apoptosis and the inhibitor significantly decreases doxorubicin resistance.
Targeting HSPB1 and its pseudogene HSP90AB4P with 7A offers a promising therapeutic strategy to overcome doxorubicin resistance in breast cancer.
-
-
-
Exploring the Therapeutic Potential of Ocimum sanctum and Phanera variegata in Breast Cancer Treatment: A Promising Natural Approach
Authors: Tohfa Siddiqui, Md. Nasar Mallick and Vikram SharmaAvailable online: 11 June 2025More LessBreast cancer is one of the most common malignancies affecting women worldwide. It is a complex, heterogeneous disease, classified into several subtypes, including hormone receptor-positive and triple-negative breast cancer (TNBC), each with distinct therapeutic challenges. TNBC, in particular, is characterized by its aggressive nature and lack of targeted therapies due to the absence of estrogen, progesterone, and HER2 receptors. This review explores the potential of natural plant-based compounds, especially focusing on Clove Basil (Ocimum sanctum) and Phanera variegata, in combating breast cancer. These plants have been traditionally used for their medicinal properties and are now being studied for their anticancer effects. Ocimum sanctum has demonstrated significant antiproliferative and pro-apoptotic effects against breast cancer cells, particularly the MCF-7 line, through mitochondrial pathway activation and gene regulation. Similarly, Phanera variegata exhibits potential through its rich content of flavonoids and other bioactive compounds, which have been shown to induce apoptosis, reduce tumor growth, and offer antioxidant benefits. The review highlights how these plant extracts, with their multiple mechanisms, including immune modulation and direct cytotoxic effects, hold promise as adjunctive or alternative therapies in breast cancer treatment, particularly for hard-to-treat subtypes like TNBC. Continued research into their molecular pathways and therapeutic efficacy could lead to new, less toxic treatment options.
-
-
-
Novel Heterocyclic Compounds Exhibit Potent Antileukemic Activity through Selective Induction of Apoptosis and HDAC8 Interaction in AML Cells
Available online: 05 June 2025More LessIntroductionHeterocyclic compounds serve as the structural framework for many commercially available drugs and are well known for their antitumor properties.
AimThis study aimed to evaluate the cytotoxic effects, apoptosis induction, changes in cell cycle progression, and gene expression alterations of new heterocyclic compounds and their precursors against the acute monocytic leukemia cell line THP-1 through in vitro experimentation and computational approaches.
MethodsThe study employed cytotoxicity assays, flow cytometry analyses, gene expression evaluations, oral bioavailability studies, and molecular modeling. Among the compounds tested, 6, 25, and 26 demonstrated the greatest potency and selectivity, exhibiting substantially increased cytotoxicity (1.18 μM < IC50 < 7.66 μM) against the THP-1 cell line. Investigations into apoptosis induction and cell cycle changes revealed that these compounds primarily caused an increase in the number of THP-1 cells undergoing apoptosis after 48 hours of treatment. Additionally, compounds 6 and 25 induced an accumulation of cells in the G0/G1 phase in the same cell line.
ResultsRegarding gene expression, a shift in the expression profile of genes associated with apoptotic mechanisms was observed. Furthermore, in silico analysis revealed that these three active compounds potentially interact with histone deacetylase 8 (HDAC8), a protein known to be associated with cancer.
ConclusionThese findings underscore the potential of these compounds as candidates for the development of novel therapeutic approaches in oncology.
-
-
-
Innovative Therapies for Oncogenic KRAS Mutations: Precision Strategies with PROTACs in Cancer Treatment
Available online: 04 June 2025More LessThe KRAS (Kirsten rat sarcoma viral oncogene homolog) gene mutation is commonly found in colorectal, lung, and pancreatic carcinomas. Unfortunately, blocking KRAS straight away has proven to be challenging. PROTACs (Proteolysis Targeting Chimeras), a class of bifunctional molecules, are designed to break down proteins, offering a unique strategy to target KRAS and overcome the limitations of traditional inhibition. This review discusses PROTACs targeting KRAS mutations in cancer, highlighting major findings, current limitations, and future perspectives. The review was performed using the databases, namely, Medline, Embase, Science Direct, and Scopus, using the keywords “PROTACs, protein degradation, anti-tumor action, cancer treatment, KRAS mutation”. Additional information was gathered from related textbooks, reviews, and documents. PROTAC treatment results in the suppression of downstream signalling pathways associated with KRAS, such as the MAPK and PI3K/AKT pathways. Animal studies demonstrate the ability of the PROTAC to effectively target KRAS-mutant tumors, inhibiting tumour growth without significant toxicities. New advances in this field can lead to cancer treatments that specifically target KRAS-mutant tumors.
-
-
-
Analytical Techniques as Indicators of Biomarkers in Proteomics Cancer Diagnosis
Authors: Pawan Kumar Goswami, Ranjeet Kumar, Dharmendra Kumar and Shubham DhimanAvailable online: 29 May 2025More LessBackgroundCancer is a complex disease marked by changes in the levels and functions of key cellular proteins, including oncogenes and tumor suppressors. Proteomics technology enables the identification of crucial protein targets and signaling pathways involved in cancer cell proliferation and metastasis. Various proteomics techniques have been employed to investigate the molecular mechanisms of cancer, aiding in the confirmation and characterization of heritable disorders.
MethodsA comprehensive literature search was conducted using PubMed, ScienceDirect, and Google Scholar with search terms like “Cancer and proteomics” and “Mass spectrometry in oncology,” utilizing Boolean operators for refinement. Selection criteria included peer-reviewed articles in English on MS-based biomarker detection, tumor-specific proteins, and drug resistance markers, excluding non-peer-reviewed works and pre-2000 publications unless foundational. Extracted data focused on MS methodologies, biomarker sensitivity, and clinical applications, particularly advances in detecting low-abundance biomarkers and monitoring treatment response. Methodological quality was assessed using PRISMA, evaluating study design, sample size, reproducibility, and statistical analysis. Ethical approval was not required, but adherence to systematic review guidelines and proper citation were ensured.
ResultsIn this review, we highlighted the advanced analytical technique for cancer diagnosis and management of cancer, and described the objective of novel cancer biomarkers. Mass spectrometry (MS) is transforming cancer diagnostics and personalized medicine by enabling precise biomarker detection and monitoring. Unlike traditional antibody-based methods, MS provides high-throughput, quantitative analysis of tumor-specific proteins in clinical samples like blood and tissue. Advanced MS techniques improve sensitivity, allowing for the identification of low-abundance biomarkers and tumor-associated proteoforms, including post-translational modifications and drug resistance markers. In research, MS-based proteomics supports multi-center biomarker validation studies with standardized protocols, enhancing reproducibility. The integration of proteomic data with genomic and transcriptomic datasets through proteogenomics is refining precision oncology strategies. These advancements are bridging the gap between research and clinical application, making MS a critical tool for early cancer detection, prognosis, and therapy selection.
ConclusionAdvancements in technology and analytical techniques have helped to produce more accurate and sensitive cancer-specific biomarkers. These methods are advancing rapidly, and developing high-throughput platforms has yielded great results. However, the substantial variation in protein concentrations makes cancer protein profiling extremely complicated. This shows that more technical developments are required in the future to improve proteome broad screening of cancer cells.
-
-
-
Maternal Embryonic Leucine Zipper Kinase (MELK) as a Promising Therapeutic Target in Triple Negative Breast Cancer
Authors: Amiya Das, Ajmer Singh Grewal, Pallavi Agarwal, Deepti Pandita and Viney LatherAvailable online: 23 May 2025More LessIntroductionMaternal Embryonic Leucine Zipper Kinase (MELK) is a serine/threonine protein kinase involved in regulating key cellular processes, including cell cycle progression, apoptosis, embryonic development, spliceosome assembly, and gene expression. Notably, MELK is overexpressed in Triple-Negative Breast Cancer (TNBC), an aggressive malignancy associated with poor prognosis, high drug resistance, and limited treatment options. Given its critical role in TNBC pathogenesis, MELK has emerged as a potential biomarker and therapeutic target. This review explores the molecular functions of MELK, its involvement in oncogenic signaling pathways, and the development of MELK-targeting small-molecule inhibitors.
MethodsA comprehensive literature review was conducted to evaluate current knowledge on MELK, including its molecular functions, interactions within signaling pathways, role in TNBC progression, and potential as a therapeutic target. Relevant databases, including PubMed, Web of Science, Embase, and Scopus, were searched for studies related to MELK expression, signaling mechanisms, and experimental therapeutic approaches.
ResultsMELK plays a central role in oncogenic signaling pathways that drive TNBC proliferation and survival. Preclinical studies have demonstrated that MELK inhibition can suppress TNBC cell growth and enhance chemotherapy efficacy. Several small-molecule inhibitors targeting MELK have shown promising anti-tumor activity in preclinical models. However, challenges remain in translating these findings into clinical applications due to drug specificity limitations and resistance mechanisms.
ConclusionMELK is a promising biomarker and therapeutic target in TNBC. However, further research is required to refine MELK inhibitors, enhance clinical efficacy, and overcome drug resistance mechanisms. Targeting MELK could offer a novel therapeutic strategy to improve TNBC treatment outcomes.
-
-
-
Computational Optimization and In silico Analysis for the Discovery of New HER2 and CDK4/6 Drug Candidates for Breast Cancer
Available online: 13 May 2025More LessBackgroundBreast cancer is an abnormal cell growth that develops in the breast and spreads throughout the body. Despite cancer being the second leading cause of death, survival rates are increasing as a result of progress in cancer screening and therapy. Breast cancer is the most frequently diagnosed cancer type among women, but in most cases, there are no obvious symptoms. Screening mammograms can be used for early detection of cancer. The size of the tumor and the extent of cancer spread determine the type of needed treatment. There are different forms of treatment, where targeted therapy is generally the least harmful. It targets specific characteristics of cancer cells, such as human epidermal growth factor receptor 2 (HER2). Tyrosine kinase inhibitors are effective targeted treatment of HER2 positive breast cancer. A newer class has emerged, cyclin dependent kinase (CDK4/6), which is used to treat metastatic breast cancer.
ObjectivesAlthough CDK4/6 inhibitors class of therapy has revolutionized the treatment of metastatic breast cancer, some patients showed resistance and decreased efficacy. This study is the first to propose innovative computational strategies to improve the effectiveness and pharmacokinetic properties of existing HER2/CDK4/6 inhibitors anti-cancer agents. Through computer-aided drug design, the activity of existing breast cancer drug candidates has been tested. Structural modifications have been applied for in-silico optimization of their biological activity.
MethodsIn this research, twenty-two analogues of the tested compounds have been proposed. Their biological activity and pharmacokinetic properties (ADMET) have been tested using BIOVIA Discovery Studio software.
ResultsOut of the designed analogous compounds, seven proposed structures demonstrated superior efficacy compared to the original drugs. The research study docking studies revealed that modifications to lapatinib and tucatinib improved binding affinity to HER2 by 15-25%, with docking scores of -18.34 kcal/mol and -1.04 kcal/mol, respectively. Similarly, CDK4/6 inhibitors exhibited enhanced selectivity, with abemaciclib showing the highest binding energy of -13.2 kcal/mol. ADMET predictions suggested improved solubility and reduced toxicity risks compared to the original drugs.
ConclusionThe research study results demonstrate that the synthesis of more lipophilic analogues of lapatinib or tucatinib and, likewise designing of fluorinated derivatives of CDK4/6 inhibitors play a crucial role in improving the efficacy of these anti-cancer agents. These findings highlight the potential of the proposed modifications as promising candidates for further pharmacological and in vitro and in vivo clinical validation.
-
-
-
Precision-engineered Carrageenan Gels: Boosting the Efficacy, Selectivity, and Release of Celecoxib for Lung Cancer Therapy
Authors: Akanksha Bhatt, Priyank Purohit and Magda H. AbdellattifAvailable online: 12 May 2025More LessBackgroundLung cancer is one of the most widespread malignancies among all types of cancers. There is uncertainty in its treatment because of the selectivity. The investigation is aimed to enhance therapeutic efficacy through targeted improvements in drug selectivity and reduced toxicity by analyzing well-accepted cyclooxygenase (COX)-2, which is an enzyme target and a known therapeutic target for anti-inflammatory and antitumor agents.
ObjectiveThe objective of the present research was to identify the most suitable counterpart for celecoxib, which would produce synergistic effects and improve the selectivity index, safety, and efficacy of targeting cancer cells.
MethodsThe HOPE-62 cancer cell line and noncancerous LLC-MK2 cell line were used to analyze the activity of the prepared formulations. The effectiveness was compared by calculating the half-maximal inhibitory concentration (IC50) values of carrageenan, celecoxib, and celecoxib embedded with carrageenan. The release pattern of celecoxib from the carrageenan matrix was also determined by using a trans-diffusion cell; moreover, the binding sites of carrageenan and celecoxib were also evaluated through in silico molecular docking studies.
ResultsCarrageenan showed promising anticancer activity, with an IC50 value of 17.3±2 µM against the HOPE-62 cell line. When blended with celecoxib (15.6±2 µM), the combination achieved enhanced efficacy and improved selectivity over celecoxib alone (IC50 of 10.3±1.5 µM). In noncancerous LLC-MK2 cells, the IC50 values were observed to be significantly higher: 1484 ±6 µM in the combined formulation and with IC50 values of 559±3 µM and 878±4 µM, respectively, in celecoxib and carrageenan alone.
ConclusionThe carrageenan-embedded celecoxib exhibited a significant increase in the selectivity index from 32 to 144, which suggests enhanced anticancer activity with a favorable safety profile. Initially, sustained release of celecoxib from the blend was at a higher rate, but steadily maintained rates were. The In-silico docking studies also supported the synergistic activity of the combined form through separate interaction patterns without interfering with others. These findings underscore the therapeutic potential of excipient–drug blending strategies to achieve synergistic effects, excellent selectivity, and reduced toxicity in cancer treatments.
-
-
-
Talimogene Laherparepvec (T-VEC): Expanding Horizons in Oncolytic Viral
Authors: Run-Bin Tan and Yeannie Hui-Yeng YapAvailable online: 06 May 2025More LessTalimogene laherparepvec (T-VEC), the first FDA-approved oncolytic viral therapy, has transformed cancer immunotherapy since its 2015 approval for unresectable melanoma. Engineered from Herpes Simplex Virus type 1 (HSV-1) with deletions in ICP34.5 and ICP47 genes and GM-CSF insertion, T-VEC selectively replicates within the tumor cells, inducing lysis and releasing tumor-derived antigens while stimulating systemic antitumor immunity through dendritic cell activation. Although extensively studied for melanoma, its potential extends beyond this malignancy, with emerging applications in breast cancer, Head and Neck Squamous Cell Carcinoma (HNSCC), and other solid tumors. This review synthesizes T-VEC’s mechanism of action, leveraging dysregulated Ras signalling, impaired interferon pathways in cancer cells, its clinical outcomes, and safety profile across these indications. While prior literature emphasizes melanoma monotherapy and combinations with immune checkpoint inhibitors, less attention has been given to its efficacy in non-melanoma cancers and synergistic potential with chemotherapy or radiation therapy. By exploring recent trials, such as T-VEC with neoadjuvant chemotherapy in triple-negative breast cancer and pembrolizumab in HNSCC, highlighting its versatility. Comparative analysis with other oncolytic viruses like HF-10, oncorine (H101), and measles virus variants positions T-VEC within the virotherapy landscape. Key challenges—systemic delivery, immune clearance, and biomarker development for patient selection—are addressed alongside strategies to enhance immune modulation through novel combinations. This review underscores T-VEC’s expanding role in cancer treatment, offering clinicians’ and researchers’ insights to optimize its therapeutic horizons across diverse malignancies.
-
-
-
Unraveling the Resistance: Challenges and Advances in PARP Inhibitor Therapy for BRCA1/2 Breast Cancer
Authors: Hongjun Tang, Jingsheng Chen, Kangwei Jiang, Jiangtao He, Fangming Tang, Dongbing Li and Yuye WuAvailable online: 06 May 2025More LessBreast cancer is the most prevalent malignant tumor among women globally, with breast cancer susceptibility genes (BRCA1 and BRCA2, BRCA1/2) mutations significantly increasing the risk of developing aggressive forms of the disease. Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have shown promise in treating BRCA1/2-mutated breast cancer by exploiting deficiencies in homologous recombination (HR) repair. However, the emergence of acquired resistance poses a significant challenge. Our study examines the mechanisms of PARPi resistance in BRCA1/2-mutated breast cancer, synthesizing recent clinical advancements and identifying key resistance pathways, including HR recovery, DNA replication fork stability, and epigenetic modifications. We also highlight potential strategies to overcome these challenges to PARPi resistance, such as combination therapies and novel targets. Our comprehensive analysis aims to inform future clinical practices and guide the development of more effective treatment strategies.
-
-
-
Baicalin Inhibits Lung Cancer Cell Proliferation and Migration via ALOX12-Mediated Ferroptosis
Authors: Yishun Jin, JinYu Wen, Zhenbo Geng, Ling Wang, Wenzheng Fang, Hanqing Zhao, Xiaohua Yan, Biyin Chen, Hangju Hua, Wujin Chen and Jiumao LinAvailable online: 05 May 2025More LessBackgroundLung cancer remains a leading cause of cancer-related mortality worldwide, primarily due to late-stage diagnosis and resistance to conventional therapies. Recent studies have highlighted the potential of natural compounds in enhancing the efficacy and reducing the side effects of conventional cancer treatments. Baicalin, a bioactive compound from Scutellaria baicalensis, exhibits significant anticancer properties.
ObjectivesThis study aimed to investigate the role of baicalin in modulating lung cancer cell behavior through the arachidonate 12-lipoxygenase (ALOX12)-mediated ferroptosis pathway.
MethodsWe employed cyber pharmacology and molecular docking techniques to predict and validate the interaction between baicalin and ALOX12. In vitro experiments were conducted on A549 lung cancer cells to assess the effects of baicalin on cell proliferation, migration, and invasion. The expression levels of ALOX12, reactive oxygen species (ROS), and ferroptosis markers, such as Glutathione Peroxidase 4 (GPX4) and Acyl-CoA Synthetase Long-Chain Family Member 4 (ACSL4), were measured.
ResultsBaicalin treatment significantly upregulated ALOX12 expression in lung cancer cells, and this upregulation was associated with a reduction in cell proliferation, migration, and invasion. Furthermore, baicalin-induced ferroptosis was characterized by increased ROS levels, iron accumulation, and elevated expression of GPX4 and ACSL4. These findings suggest that baicalin enhances ferroptosis through ALOX12 activation, synergistically inhibiting cancer cell growth.
ConclusionBaicalin significantly upregulated ALOX12 expression, promoted ferroptosis, and inhibited the proliferation and migration of A549 lung cancer cells. This finding provides evidence for the potential use of baicalin as a therapeutic agent for lung cancer and highlights the importance of ALOX12 in lung cancer treatment strategies.
-
-
-
Phytochemical Profiling and Anticancer Potential of Fagonia cretica L. Extracts on Liver Cancer (HepG2) Cells using In vitro and In silico Approaches
Available online: 03 May 2025More LessBackgroundCancer is a complex multifactorial disease charcterized by the progression of genetic and epigenetic changes in human cells. Plant-based derivatives with antioxidant and anticancer properties have been of great interest in treating several human ailments.
ObjectiveThis study investigates the in-vitro antioxidative, cytotoxic, and apoptotic activities of different Fagonia cretica L. (F. cretica) leaf extracts.
MethodsIn-vitro DPPH, nitric oxide, superoxide anion, and hydrogen peroxide assays were used to evaluate the antioxidative potential of ethanolic extract of F. cretica (EFC) and hexane extract of F. cretica (HFC). The antiproliferative potential was determined using MTT, crystal violet, and annexin V/PI staining protocols on liver cancer (HepG2) and noncancerous (HEK-293) cell lines. Through in silico analysis, bioactive drug-like phytocompounds identified by GC-MS were evaluated.
ResultsHigher concentrations of total flavonoid contents (TFCs), total phenolic contents (TPCs), and tannins with strong antioxidant potential were observed in EFC extract as compared to HFC extract. Furthermore, the EFC extract proved to be more cytotoxic with a selective index (SI) of 12.92 than HFC (SI; 5.46) towards experimental cell lines. Moreover, EFC extract showed 82.31% apoptotic induction on HepG2 cells compared to hexane extract and cisplatin (standard drug). From the GC-MS analysis of F. cretica, 32 bioactive compounds were identified from the EFC extract and 21 from the HFC extract. In silico study revealed that 5-(4,5-Dihydro-3H-pyrrol-2-ylmethylene)-4,4-dimethylpyrrolidine-2-thione showed the highest docking score of -8.9 kcal/mol and -8.6 kcal/mol against TNF-α and TGF-β, respectively.
ConclusionIn conclusion, EFC extract and its bioactive compounds have a scientifically proven role in liver cancer management, but further research is required to validate their therapeutics through clinical trials.
-
-
-
Enhanced Apoptosis in Pancreatic Cancer Cells through Thymoquinone-rich Nigella sativa L. Methanol Extract: Targeting NRF2/HO-1 and TNF-α Pathways
Available online: 29 April 2025More LessAimsThis study explores the therapeutic potential of Nigella sativa L. and its key bioactive compound, thymoquinone (TQ).
BackgroundPancreatic cancer presents a significant health challenge due to its aggressiveness and limited treatment options. N. sativa and its component TQ have demonstrated anticancer properties in other cancers, warranting exploration in pancreatic cancer models.
ObjectiveTo assess the antiproliferative, apoptotic, and anti-invasive effects of N. sativa extracts and TQ on pancreatic cancer cells, with a focus on modulating the NRF2/HO-1 and TNF-α signaling pathways.
MethodsMIA PaCa-2 and PANC-1 pancreatic cancer cell lines were treated with essential and fixed oils, methanol extracts (from Türkiye and Syria), and TQ. Cell viability, apoptosis, and invasiveness were assessed via XTT, Annexin V, and Matrigel assays, respectively. Gene expression and cytokine levels were evaluated using RT-qPCR and ELISA. HPLC was conducted to confirm TQ concentrations in extracts.
ResultsThe methanol extract of Türkiye-originated N. sativa seeds (TM) exhibited the highest cytotoxic effect, reducing cell viability in MIA PaCa-2 and PANC-1 at 0.05 mg/mL, while TQ significantly decreased viability at 20 µM. TM reduced MIA PaCa-2 and PANC-1 invasiveness (42 ± 1.23 and 35 ± 0.73, respectively) and contained a higher concentration of TQ (7.9168 ± 0.0561%) compared to the Syria-originated extract (SM).
ConclusionThe findings suggest that TM and TQ exhibit strong anticancer potential by modulating key signaling pathways in pancreatic cancer cells, supporting their potential for further development as therapeutic agents in pancreatic cancer treatment.
-
-
-
Synthesis and Biological Studies of Aurones Derivatives for its Anticancer Potential
Authors: Kulkarni P Yogesh and Pramod L. IngaleAvailable online: 25 April 2025More LessBackgroundAurone based compounds exhibited antioxidant and anti-inflammatory potential and documented for their anticancer potential. The anticancer potential of aurone derivatives AU3, AU4, AU5, AU7, and AU10 is yet to be studied against breast cancer.
ObjectiveThe present work was undertaken to evaluate the anticancer potential of aurone based test compounds AU3, AU4, AU5, AU7, and AU10 in breast cancer cell lines MCF-7.
MethodsThe azaindole based aurones were synthesized by the condensing 4,6-dimethoxybenzofuran-3(2H)-one derivative with various indole aldehydes in the presence of sodium hydroxide. The MCF-7 breast cancer cell line was used to assess the cytotoxic effects of these compounds. Molecular docking studies of the synthesized compounds against the Cyclin-dependent kinase 2 (CDK2)/Cyclin A complex were conducted.
ResultsOur experimental findings demonstrated that AU3, AU4, AU5, AU7, and AU10 elicited significant effects on MCF-7 by virtue of its minimum cell viability, with IC50 values of 70.14 µM, 87.85 µM, 133.21 µM, 52.79 µM, and 99.55 µM, respectively, thus, exhibits potential anticancer action. Further, to corroborate the anticancer potential, we investigated mechanisms of action through molecular docking studies with the CDK2/Cyclin A complex (PDB: 6GUC) and their findings demonstrated that test compounds showed robust binding through various interactions, including hydrogen bonds, Pi-interactions, and Alkyl bonds with key residues such as Lys129, Asp127, Gln131, and Asp145. Test compounds AU3 and AU7, exhibited better binding affinities and diverse interaction profiles, suggesting a potent disruption of CDK2/Cyclin A activity.
ConclusionThus, in conclusion, our findings revealed that AU3, AU4, AU5, AU7, and AU10 elicited anticancer action and their effects through CDK2/Cyclin A disruption.
-
-
-
Methyl (Z)-2-(Isothioureidomethyl)-2-pentenoate Hydrobromide Induces Cell Cycle Arrest and Disrupts Mitosis in a Melanoma Cell Line
Available online: 24 April 2025More LessIntroduction/ObjectiveCancer is a global health burden. Despite advances in early detection and therapeutics, cancer prevalence continues to increase, underscoring the need for innovative therapeutic strategies. Dysregulation of cell death mechanisms is a hallmark of cancer that can lead to apoptosis evasion, which strongly contributes to tumor progression and therapy resistance. Isothiouronium salts have attracted attention as promising antitumor agents. This study aimed to evaluate the in vitro antitumor effect of an isothiouronium salt (IS-MF08) on the B16F10 melanoma cell line.
MethodsThe antitumor properties of IS-MF08 were investigated by incubating B16F10 cells with the compound at different concentrations. Cytotoxicity was determined by the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay, cell cycle arrest and cell death mechanisms by flow cytometry, and morphological alterations by transmission electron microscopy. Physicochemical parameters related to drug-likeness were predicted in silico using the SwissADME tool.
ResultsIS-MF08 was cytotoxic to melanoma cells, triggering cell cycle arrest and disrupting mitosis. The mechanism of cell death was compatible with apoptosis, as indicated by annexin V-FITC experiments and the relevant morphological changes in cell structure observed by transmission electron microscopy. SwissADME predicted that IS-MF08 has good physicochemical properties related to absorption and permeation.
ConclusionThe numerous mechanisms of cell death triggered by IS-MF08 and its drug-likeness make it an interesting molecule in the search for new antitumor compounds, contributing to therapies targeting the dysregulation of cellular mechanisms such as apoptosis.
-
-
-
Advancements in Metal Complexation of Pyridine Derivatives (2022–2024): A Pathway to Enhanced Anticancer Potency
Available online: 24 April 2025More LessCancer remains a major global health challenge, necessitating innovative therapies that selectively target cancer cells while sparing healthy tissues. Pyridine and its derivatives have gained prominence in medicinal chemistry for their structural diversity and biological activity. However, their therapeutic potential is often hindered by low bioavailability, poor solubility, and rapid metabolism. Metal complexation has emerged as a promising solution, with pyridine nitrogen serving as an excellent coordination site for transition metals. These pyridine-metal complexes enhance stability, bioavailability, and anticancer properties, exhibiting potent cytotoxicity through mechanisms like ROS generation, DNA intercalation, and apoptosis induction. This review highlights the latest progress (2022-2024) in the field, emphasizing the structural modifications, and mechanistic insights that have propelled pyridine-metal complexes as potent anticancer agents. Special attention is given to the role of metal complexation in enhancing the anticancer potency of pyridine derivatives, with examples of preclinical studies showing their efficacy against various cancer types. The findings emphasize the potential of pyridine-metal complexes as a transformative approach in oncology, bridging the gap between innovative chemical design and impactful therapeutic applications.
-
-
-
Recent Approaches on Oncolytic Viruses, Aptamers, TTFields and Personalized Treatment used for the Management of Glioblastoma: A Comprehensive Review
Authors: Archna Singh, Anmol Kanda and Jyoti KandaAvailable online: 10 April 2025More LessGlioblastoma (GB) remains a formidable challenge in oncology, with current treatment approaches providing only marginal improvements in patient outcomes. Despite significant advances in understanding its molecular and genetic characteristics, median survival for untreated patients remains distressingly low, emphasizing the urgent need for novel therapeutic strategies. This review comprehensively examines the standard first-line treatments for GB, including surgery, concomitant radio-chemotherapy, and maintenance chemotherapy, while highlighting the limitations of these approaches. Consequently, we explore emerging novel therapeutic modalities such as Oncolytic Viral Therapy with genetically modified oncolytic viruses that enhance the capabilities of antigen-presenting cells. These cells migrate to lymph nodes to recruit cytotoxic CD8+ T lymphocytes, directing them to the site of infection where they eradicate cells that promote tumour growth. Aptamer-based therapies, such as GMT-3, AS1411, GS24, GMT8, and Gint4.T, which exhibit specificity for their biological targets and can act as drug transporters by facilitating receptor-mediated transcytosis within the endothelial cells of the blood-brain barrier, thus improving drug delivery. Tumour-treating fields (TTFields) that have shown increased overall survival rates in patients. Personalized genomic medicine, driven by biomarkers, which provokes immune responses tailored to the tumour’s specific antigens, thereby customizing patient-specific treatments to improve effectiveness. By synthesizing current evidence and recent breakthroughs, we underscore the potential use of advancing novel therapies to address the unmet clinical needs of GB patients and ultimately enhance their overall survival and quality of life.
-
-
-
Clinical Characteristics and Prognostic Factors Associated with Herpes Zoster in Patients with Malignant Tumors: A Systematic Review and Meta-analysis
Authors: Mingming Ding, Shantao Qiu and Guan JiangAvailable online: 07 April 2025More LessBackgroundHerpes zoster (HZ) is a common complication in patients with malignant tumors (MT), impacting prognosis. Immunocompromised states due to malignancy or treatment increase HZ risk. However, comprehensive assessments of HZ's clinical features and its impact on prognosis in these patients are limited, general conclusions are challenging, prompting a systematic review and meta-analysis to better understand the relative risk of HZ in malignancy.
ObjectiveTo assess the clinical features and prognostic factors of HZ in cancer patients through systematic review and meta-analysis. The study aimed to calculate the relative risk of HZ in malignancy and analyze factors affecting prognosis, such as age, gender, tumor type, and treatment.
MethodsA systematic search in PubMed (2016-2024) identified studies on HZ and malignancy. Two reviewers independently screened and selected studies, extracting data on study characteristics, population demographics, and outcomes. Statistical heterogeneity across the studies was addressed using random-effects models, while subgroup analyses were performed to identify potential sources of heterogeneity.
ResultsOut of the 633 records reviewed, 13 studies satisfied the eligibility criteria and were incorporated into the meta-analysis. The combined relative risk for any type of cancer was found to be 1.82(95% CI: 1.29,2.57). The combined relative risk for any solid tumors was 1.63(95% CI: 1.08,2.46). The combined relative risk for any haematological cancer was 3.43(95% CI: 1.33,8.86). The combined analysis of all treatment modalities (including Radiotherapy, Chemotherapy, Immunosuppression, HSCT) shows a significant overall effect with a risk ratio of 1.78(95%CI: 1.59,2.00).
ConclusionCancer patients have increased HZ risk due to immunosuppression from the malignancy and its treatment, especially in hematological cancers and those undergoing stem cell transplantation.
-
-
-
A Review of Cryptotanshinone and its Nanoformulation in Cancer Therapy
Authors: Xin Liu, Yahan Gao, Fan Yang, Min Qian, Shuhui You, Xiaoxiao Wang, Fenju Qin, Min Xiang and Weiqiang GuoAvailable online: 28 March 2025More LessCancer, with a high incidence and mortality rate, has emerged as a major public health problem worldwide. Currently, new approaches, such as targeted therapy and immunotherapy, are giving hope to patients. However, drug resistance and adverse side effects are major barriers to cancer treatment. As a result, there is a greater focus on the development of cancer therapy strategies and medications with low toxicity and high efficacy. Cryptotanshinone (CTS), a diterpenoid quinone extracted from Salvia miltiorrhiza Bunge, exhibits a wide range of biological activities, including immunomodulatory, anti-inflammatory, and antitumor effects. In recent years, numerous studies have highlighted its significant antitumor properties, indicating potential clinical applications and development value. However, the clinical use of cryptotanshinone has been limited due to its poor water solubility and low bioavailability. To overcome these limitations, researchers are exploring new drug delivery systems, and novel formulation systems based on nanotechnology are being developed to improve the delivery and effectiveness of cryptotanshinone. In this review, we aim to consolidate the existing knowledge regarding the antitumor effects of cryptotanshinone and emphasize the latest advancements in its nanoformulation development. We hope to provide insights that will further improve the antitumor efficacy and clinical applicability of cryptotanshinone.
-
-
-
Recent Advances in Therapeutic Potential of Dual-Acting Aromatase/COX-2
Available online: 26 March 2025More LessAromatase, a crucial enzyme assigned for transforming androgen into estrogen, has a vital function in the advancement of drug-resistant breast cancers that respond to endocrine treatments. Aromatase (CYP19A1) is a monooxygenase from the cytochrome P450 family that is involved in the conversion of androgens to estrogens. Breast cancer cells express aromatase activity, indicating that the tumor cells may be able to produce local estrogen. By inhibiting aromatase, serum estrogen levels decrease, which, in turn, hinders estrogen-driven cancer cell growth in hormone receptor-positive breast cancer cases. In this sense, the introduction of novel aromatase inhibitors could be a significant step forward in the fight against cancer. This is especially true in hormone-dependent cancers. Many compounds have been introduced as aromatase inhibitors, classified as steroidal or nonsteroidal. However, it should be noted that these drugs have encountered resistance in numerous cases, particularly in recent years. Thus, the search for new aromatase inhibitor drugs has always been critical. Newly, there seems to be a surge of enthusiasm in the discovery and production of molecules with dual inhibitory effects, which can inhibit two or more enzymes simultaneously. This method enables a significant reduction in potential drug resistance. The design of these compounds has an opportunity to significantly boost the efficacy of anti-cancer treatments by causing synergistic effects. This article offers a review of newly developed aromatase inhibitors with potential anticancer effects.
-
-
-
Unraveling the Role of Tumor-infiltrating Immune Cells in Modulating Cancer Drug Resistance
Available online: 24 March 2025More LessTumor-infiltrating immune cells (TIICs) have been identified as critical components in the development of cancer drug resistance. This review aims to discuss the various types of TIICs, such as macrophages and T cells, that have been linked to cancer drug resistance. Furthermore, we explore the mechanisms by which TIICs contribute to drug resistance and how these mechanisms may differ across various tumor types. Additionally, we examine the potential of immune checkpoint inhibitors in combination with traditional cancer therapies as a strategy to overcome TIIC-mediated cancer drug resistance. In conclusion, this review provides an in-depth analysis of the current knowledge on the role of TIICs in cancer drug resistance and highlights potential avenues for future research to develop more effective treatment strategies. The findings presented in this review emphasize the importance of understanding the complex interactions between cancer cells and the immune system in order to develop novel therapeutic approaches that can overcome TIIC-mediated cancer drug resistance.
-
-
-
Synthesis and Evaluation of Optical Properties, SHP2 Inhibitory Activity, and Cellular Imaging for Novel 2-Quinolone Derivatives
Authors: Chun Zhang, Yuting Yang, Li-Xin Gao, Suya Gan, Jia Li, Xin Wang, Yu-Bo Zhou and Wen-Long WangAvailable online: 17 March 2025More LessIntroductionAlthough the development of SHP2 inhibitors has made striking progress, there is no inhibitor in clinical evaluation because of the potential side effects induced by poor drug distribution. Fluorescence imaging technology is widely used in the process of diagnosis and treatment of diseases because of the advantages of rapid imaging and non-destructive detection and might provide a new way to explore the mechanism of drug-target interactions in intact tissue.
MethodsA series of 2-quinolone derivatives as fluorescent inhibitors against SHP2 were designed and synthesized, and their spectral properties and biological activities were evaluated in this report. The representative compound 8A had excellent fluorescence properties (: 562 nm, Stokes shift: 170 nm, fluorescence quantum yield: 0.072) and optical stability.
ResultsMoreover, compound 8A emitted a blue signal in SHP2WT U2OS cells and inhibited the SHP2 enzyme abilities (IC50: 20.16 ± 0.95 μM) without the extra combination of suitable fluorophores, linker, or selective-activated molecules.
ConclusionTherefore, we hope that compound 8A could act as a lead to develop novel, convenient, and bifunctional chemical tools to explore the mechanism of drug-target interactions in intact tissue and promote the integrated research progress of diagnosis and treatment of SHP2 related diseases.
-
-
-
Recurrent Missense Driver STAT5B N642H Mutation in Children Transiting into Adolescence, with Acute Lymphoid Leukemia and its In silico Inhibition
Authors: Rehana Yasmin, Rashda Abbasi, Tajdar Jahangir Gohar, Hina, Nafees Ahmad and Sajid MalikAvailable online: 10 February 2025More LessBackgroundThe occurrence of gain of function mutations in STAT5B has been associated to survival, and drug resistance in Leukemia. In silico screening of compounds having inhibitory potential towards mutated proteins, can be helpful in the development of specific inhibitors.
ObjectiveThis study was designed to screen selected JAK-STAT mutations in leukemia patients and virtual exploration of molecular interaction of potential inhibitors with their mutated products.
MethodsIn total 276 patients were randomly recruited for this study. Demographic and clinical data were summarized. The genetic status of JAK1V623A, JAK2 S473 and STAT5BN642H were screened through allele specific PCR. In-silico analysis was performed on wild type and mutant protein sequences retrieved from Protein databank. The ligands and protein were prepared through standard protocols, and docking was performed through Auto Dock Vina 1.2.0.
ResultsAcute lymphoblastic leukemia comprises 70% of the total patients. Male to female ratio was 3:1. All the patients were homozygous for JAK1V623A, JAK2 S473 major allele. However, 6 patients (5 male, 1 female) with ALL were STAT5BN642H+. The molecular docking of the ligands to wild type and STAT5BN642H+revealed that AC-4-130, Pimozide, Indirubin and Stafib-2 have higher but differential docking affinities for SH2-domain of both normal and mutated STAT5B. However, AC-4-130 has a higher affinity for wild type and Stafib-2 has stable molecular interaction with STAT5BN642H+.
ConclusionThe aggressive form of pediatric leukemia, carrying STAT5BN642H+ mutation is identified in the studied population. It is predicted that AC-14-30 and stafib-2 have potential for inhibition of constitutively active STAT5B if optimized for use in combination therapy.
-
-
-
A Systematic Quantitative Approach to Rational Drug Design and the Discovery of Novel Human Antigen R (HuR) Inhibitors
Authors: Juhi Dey, Kumari Kaushiki, KM Abha Mishra, Paga Sudheer and Kalyan Kumar SethiAvailable online: 04 February 2025More LessBackground1,4-Naphthoquinone and its derivatives are recognized for their potent anticancer effects, establishing this pharmacophore as a key focus in cancer research. Their potential to modulate cellular pathways suggests they could be effective in developing new HuR inhibitors, targeting a protein crucial for regulating cancer-related gene expression. Compounds C1-C20 were designed by using Discovery Studio (DS) software.
MethodsIn this study, a systematic approach involves scaffold hopping followed by additional research such as molecular docking, ADMET, drug-likeness, toxicity prediction, molecular dynamic (MD) simulation, and binding free energy analysis was used to discover novel Human Antigen R (HuR) inhibitors.
ResultsIn molecular docking, 1,4-Naphthoquinone derivatives showed better interactions with the HuR protein compared to that of the conventional HuR inhibitor MS-444. Among twenty 1,4-Naphthoquinone derivatives, most of the compounds showed favorable pharmacokinetic characteristics. In the toxicity prediction model, most of the designed compounds were neither mutagenic nor carcinogenic. According to MD simulation, C5 is more stable than MS-444.
ConclusionThe designed 1,4-Naphthoquinone derivatives have been found to be crucial structural motifs for the discovery of novel HuR inhibitors, which was well supported by the in-silico screening and molecular modeling methods.
-
-
-
The Function of Poly (U) Binding Splicing Factor 60 (PUF60) in Disease Regulation
Authors: Huijuan Chen, Tian Guan, Jingfeng Song and Yihua ChenAvailable online: 03 January 2025More LessThe alternative splicing (AS) of pre-mRNA is an important process in controlling the expression of human genes, which can enrich the diversity of the proteome and regulate gene function. On the contrary, aberrant splicing contributes significantly to numerous human diseases progression, including tumors, neurological diseases, metabolic diseases, infections, and immune diseases. The PUF60, a protein related to RNA splicing, plays critical functions in RNA splicing and gene transcription regulation. In addition, it can achieve synergistic binding with U2AF65 on RNA through interactions in the pyrimidine region, promoting the splicing of introns with weak 3'- splice sites and pyrimidine bundles. Nevertheless, an increasing amount of evidence supports that it shows a significant overexpression pattern in the vast majority of cancer cells and is crucial for embryonic development, indicating that PUF60 may hold the post of a potential therapeutic target for such diseases. These studies have significantly increased our interest in PUF60. Thus, we briefly reviewed the structural domain characteristics of the PUF60, splicing mutants of PUF60, and the roles and functions in human diseases, including various cancers, infections of bacterium and viruses, myositis, and Verheij syndrome. Furthermore, the targeted PUF60 inhibitors and boundedness of the current research were elaborated on in the article. The article effectively communicates critical perception and insight, making it a precious resource for those interested in PUF60 research and treatment.
-