Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (9,162)

Search Parameters:
Keywords = anticancer potentiality

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 6946 KB  
Article
Phlorotannins from Ecklonia cava Regulate Dual Signaling Pathways, IL-17RA/Act1 and ERK1/2, to Suppress Ovarian Cancer Progression and Tumor-Associated Macrophage Activation
by Eun-Hye Kim, Hwi-Ho Lee, Jung-Hye Choi and Ji-Hye Ahn
Mar. Drugs 2026, 24(1), 12; https://doi.org/10.3390/md24010012 - 24 Dec 2025
Abstract
Background: Marine-derived secondary metabolites such as phlorotannins from the edible brown alga Ecklonia cava exhibit diverse bioactivities. However, their mechanisms in inflammation-associated cancer remain insufficiently understood. Methods: This study explored the anticancer potential of three major phlorotannins (dieckol, 7-phloroeckol, and 8,8′-bieckol) through network [...] Read more.
Background: Marine-derived secondary metabolites such as phlorotannins from the edible brown alga Ecklonia cava exhibit diverse bioactivities. However, their mechanisms in inflammation-associated cancer remain insufficiently understood. Methods: This study explored the anticancer potential of three major phlorotannins (dieckol, 7-phloroeckol, and 8,8′-bieckol) through network pharmacology, molecular docking, molecular dynamics simulations, and in vitro validation in SKOV3 ovarian cancer cells and tumor-associated macrophages (TAMs). Results: Computational analyses revealed stable binding of phlorotannins to IL-17RA, with 7-phloroeckol and 8,8′-bieckol preferentially engaging loop-proximal regions of the receptor, while dieckol interacted with spatially distinct residues. In SKOV3 ovarian cancer cells, phlorotannins suppressed migration and invasion by approximately 40 to 60%, accompanied by reduced MMP expression linked to IL-17RA–Act1 signaling attenuation and by increased TIMP1 expression in association with transient ERK1/2 activation. In TAMs, phlorotannins attenuated pro-tumorigenic cytokine production and polarization marker expression, indicating suppression of tumor-supportive immune activity. Conclusions: Collectively, these findings demonstrate that E. cava-derived phlorotannins exert anti-metastatic effects through dual regulation of IL-17RA/Act1 and ERK1/2 signaling pathways, offering mechanistic insight into their therapeutic potential against inflammation-driven malignancies. Full article
20 pages, 2303 KB  
Article
Repurposing Rosiglitazone Induces Apoptosis Accompanied by Impaired Antioxidant Defense: Findings from Proteomic and Functional Analyses
by Benjaporn Buranrat, Prakasit Sa-Ngiamwibool, Auemduan Prawan, Sarinya Kongpetch, Piman Pocasap and Laddawan Senggunprai
Pharmaceuticals 2026, 19(1), 44; https://doi.org/10.3390/ph19010044 - 24 Dec 2025
Abstract
Objectives: The repurposing of existing drugs as anticancer agents has attracted attention in cancer drug discovery. This study aimed to examine the anticancer efficacy of rosiglitazone (RSG) against cholangiocarcinoma (CCA) and its underlying mechanisms. Methods: The effect of RSG on the [...] Read more.
Objectives: The repurposing of existing drugs as anticancer agents has attracted attention in cancer drug discovery. This study aimed to examine the anticancer efficacy of rosiglitazone (RSG) against cholangiocarcinoma (CCA) and its underlying mechanisms. Methods: The effect of RSG on the viability of KKU-100 CCA cells was examined. The possible molecular targets were identified using proteomic analysis and verified by a series of cell-based assays. Furthermore, the expression of PPARγ protein in CCA tissues was also assessed. Results: RSG exhibited a cytotoxic effect against KKU-100 cells. Proteomic analysis demonstrated a significant different expression protein pattern of the 100 μM RSG-treated group compared to the control group. Significant alteration of several proteins was found, including the up-regulation of calcium-binding, cytoskeletal, and metabolic proteins, concomitant with the down-regulation of antioxidant enzymes. Detailed analyses revealed that RSG induced apoptosis in CCA cells, accompanied by increased caspase 3/7 activities, reactive oxygen species (ROS) generation, and disruption of mitochondrial function. RSG altered the expressions of annexin A1 and antioxidant enzymes, according to Western blot analysis. GW9662, a PPARγ antagonist, did not affect the viability and apoptosis of KKU-100 cells caused by RSG. Immunohistochemistry analysis revealed that PPARγ expression in CCA patients was associated with sex, but not with other common clinicopathological parameters. Its expression did not correlate with patients’ overall survival time. Conclusions: RSG induced apoptotic cell death in CCA cells, which was accompanied by increased ROS levels and impaired antioxidant defense. Its apoptosis-inducing effect is independent of PPARγ activation. These findings underscore the therapeutic potential of RSG for CCA treatment. Full article
(This article belongs to the Section Medicinal Chemistry)
25 pages, 2906 KB  
Article
Design and Synthesis of New Coumarin Hybrids Active Against Drug-Sensitive and Drug-Resistant Neuroblastoma Cells
by Carola Grondona, Barbara Marengo, Giulia Elda Valenti, Sara Tirendi, Eleonora Russo, Cinzia Domenicotti and Bruno Tasso
Antioxidants 2026, 15(1), 31; https://doi.org/10.3390/antiox15010031 - 24 Dec 2025
Abstract
High-risk neuroblastoma (NB) is an aggressive pediatric tumor characterized by pronounced biological heterogeneity and frequent development of chemoresistance, which critically limits therapeutic efficacy. Identifying novel anti-NB agents remains an urgent unmet need. To address this, we designed and synthesized 17 hybrid molecules by [...] Read more.
High-risk neuroblastoma (NB) is an aggressive pediatric tumor characterized by pronounced biological heterogeneity and frequent development of chemoresistance, which critically limits therapeutic efficacy. Identifying novel anti-NB agents remains an urgent unmet need. To address this, we designed and synthesized 17 hybrid molecules by combining natural antioxidant scaffolds (coumarin, vanillin, and isovanillin) through an acyl-hydrazone linker. Several derivatives significantly reduced the viability of MYCN-amplified NB cells (HTLA-230) and their multi-drug resistant counterpart (ER) while not affecting human keratinocytes (HaCat). Among them, compounds 5, 9 and 12 selectively inhibited HTLA and ER growth (10–25%) without affecting HaCat, accompanied by robust ROS overproduction, particularly by 9 and 12 (up to 40%). None of these compounds induced apoptosis or ferroptosis. Instead, their antiproliferative effects were associated with senescence induction and, only for compound 5, with a decrease in clonogenic potential. Moreover, to further characterize compounds 5, 9, and 12, the analysis was extended across other human neuroblastoma cell lines. In parallel, the effects of the compounds on non-malignant cell lines were assessed to obtain an indication of their selectivity toward tumor cells. Compound 17, a structural analog lacking the second aromatic ring in the ex-aldehyde portion, displayed a distinct profile with a limited anticancer activity, underscoring the importance of this structural fragment for antiproliferative efficacy. Full article
(This article belongs to the Section Natural and Synthetic Antioxidants)
Show Figures

Figure 1

28 pages, 3580 KB  
Review
Vanillin Beyond Flavor: Therapeutic Potentials and Emerging Applications in Hydrogel-Based Biomaterials
by Lei Cui, Dong Uk Yang, Jing Liu, Ramya Mathiyalagan, Jong-Hoon Kim, Sathiyamoorthy Subramaniyam, Changbao Chen, Deok-Chun Yang and Ling Li
Gels 2026, 12(1), 16; https://doi.org/10.3390/gels12010016 - 24 Dec 2025
Abstract
Vanillin (4-hydroxy-3-methoxybenzaldehyde) is widely recognized for its aromatic flavor and established pharmacological properties, including antioxidant, antimicrobial, anti-inflammatory, and anticancer effects. While these biological activities underpin its therapeutic potential, recent advances have expanded the application of vanillin into the field of biomaterials. In particular, [...] Read more.
Vanillin (4-hydroxy-3-methoxybenzaldehyde) is widely recognized for its aromatic flavor and established pharmacological properties, including antioxidant, antimicrobial, anti-inflammatory, and anticancer effects. While these biological activities underpin its therapeutic potential, recent advances have expanded the application of vanillin into the field of biomaterials. In particular, vanillin’s unique chemical structure enables its use as a multifunctional building block for the development of innovative hydrogels with dynamic covalent bonding, injectability, and self-healing capabilities. Vanillin-based hydrogels have demonstrated promising applications in wound healing, drug delivery, tissue engineering, and antimicrobial platforms, combining structural support with intrinsic bioactivity. These hydrogels benefit from vanillin’s biocompatibility and functional versatility, enhancing mechanical properties and therapeutic efficacy. This review provides an overview of vanillin’s pharmacological effects, with a primary focus on the synthesis, properties, and biomedical applications of vanillin-derived hydrogels. By highlighting recent material innovations and their translational potential, we aim to position vanillin as a valuable natural compound bridging bioactivity and biomaterial science for future clinical and therapeutic advancements. Full article
(This article belongs to the Section Gel Applications)
Show Figures

Figure 1

20 pages, 3217 KB  
Article
Design and In Vitro Evaluation of Cross-Linked Poly(HEMA)-Pectin Nano-Composites for Targeted Delivery of Potassium Channel Blockers in Cancer Therapy
by Gizem Ozkurnaz Civir, Fatemeh Bahadori, Ozgur Ozay, Gamze Ergin Kızılçay, Seyma Atesoglu, Ebru Haciosmanoglu Aldogan and Burak Celik
Gels 2026, 12(1), 13; https://doi.org/10.3390/gels12010013 - 24 Dec 2025
Abstract
Potassium (K+) channel blockers are promising anticancer agents but suffer from off-target toxicities. We designed cross-linked poly-2-Hydroxyethyl methacrylate (HEMA)–pectin nanogels (HPN) to deliver two model blockers—dofetilide (Dof) and azimilide (Azi)—and evaluated their physicochemical properties, release behavior, and in vitro anticancer activity. [...] Read more.
Potassium (K+) channel blockers are promising anticancer agents but suffer from off-target toxicities. We designed cross-linked poly-2-Hydroxyethyl methacrylate (HEMA)–pectin nanogels (HPN) to deliver two model blockers—dofetilide (Dof) and azimilide (Azi)—and evaluated their physicochemical properties, release behavior, and in vitro anticancer activity. HPN was synthesized by surfactant-assisted aqueous nanogel polymerization and comprehensively characterized (FTIR, DLS, TEM/SEM, XRD, BET). The particles were monodispersed with a mean diameter ~230 nm, compatible with tumor accumulation via the Enhanced Permeability and Retention (EPR) effect, and exhibited a microporous matrix suitable for controlled release. Drug loading was higher for Dof than for Azi, with DL% values of 82.30 ± 3.1% and 17.84 ± 2.9%, respectively. Release kinetics diverged: Azi-HPN followed primarily first-order diffusion with a rapid burst, whereas Dof-HPN showed mixed zero/first-order behavior. Cytotoxicity was assessed in A549 lung cancer and BEAS-2B bronchial epithelial cells. Both free and nano-formulated blockers were selectively toxic to A549 with minimal effects on BEAS-2B. Notably, a hormesis-like pattern (low-dose stimulation/high-dose inhibition in MTT) was evident for free Dof and Azi; encapsulation attenuated this effect for Dof but not for Azi. Co-administration with paclitaxel (Ptx) potentiated Dof-HPN cytotoxicity in A549 but did not enhance Azi-HPN, suggesting mechanism-dependent drug-drug interactions. Overall, HPN provides a biocompatible platform that improves K+ blocker delivery. Full article
Show Figures

Graphical abstract

29 pages, 4286 KB  
Review
Mechanistic Insights into the Metabolic Pathways and Neuroprotective Potential of Pentacyclic Triterpenoids: In-Depth Analysis of Betulin, Betulinic, and Ursolic Acids
by Mihai Adrian Socaciu, Zorita Diaconeasa, Dumitrita Rugina, Carmen Socaciu, Remus Moldovan and Simona Clichici
Biomolecules 2026, 16(1), 25; https://doi.org/10.3390/biom16010025 - 24 Dec 2025
Abstract
Due to their complexity, both genotypic and phenotypic, neurodegenerative diseases are one of the main causes of death globally nowadays. Among phytochemicals of high scientific interest, based on experimental studies, pentacyclic triterpenoids (TTs), including mainly betulin, betulinic, and ursolic acid, became targets of [...] Read more.
Due to their complexity, both genotypic and phenotypic, neurodegenerative diseases are one of the main causes of death globally nowadays. Among phytochemicals of high scientific interest, based on experimental studies, pentacyclic triterpenoids (TTs), including mainly betulin, betulinic, and ursolic acid, became targets of scientific research in recent years, especially in terms of their biological activity and pharmacological potential. Due to their anti-inflammatory and antioxidant properties, as well as their involvement in cellular signal transductions, they have been observed to act as anticancer, chemopreventive, and neuroprotective agents. The aim of this review is to update the reader on the diversity, bioavailability, pharmacological properties, and neuroprotective effects of TTs, as biomolecules that can interfere with metabolic mechanisms related to neurodegeneration and restoring of neuronal integrity. Recent data were analyzed, with a particular focus on mechanistic insights related to their neuroprotective effects. Starting with their biosynthetic pathways, bioavailability, and involvement in specific metabolic pathways, their impact on neurological pathology and benefits as natural neuroprotection agents through specific signaling pathways are presented. Furthermore, emphasis will also be put on current challenges and future strategies that could develop TTs into effective compounds for neuroprotection and personalized medicine. Full article
(This article belongs to the Special Issue Advances in Metabolomics in Health and Disease)
Show Figures

Figure 1

20 pages, 3147 KB  
Article
Network Pharmacology and Molecular Docking Identify Medicarpin as a Potent CASP3 and ESR1 Binder Driving Apoptotic and Hormone-Dependent Anticancer Activity
by Yanisa Rattanapan, Sirinya Sitthirak, Aman Tedasen, Thitinat Duangchan, Hasaya Dokduang, Nawanwat C. Pattaranggoon, Krittamate Saisuwan and Takol Chareonsirisuthigul
Int. J. Mol. Sci. 2026, 27(1), 174; https://doi.org/10.3390/ijms27010174 - 23 Dec 2025
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecologic malignancies due to late diagnosis, rapid progression, and frequent chemoresistance. Despite advances in targeted therapy, durable responses are uncommon, underscoring the need for novel multitarget agents capable of modulating key oncogenic networks. Medicarpin, [...] Read more.
Ovarian cancer (OC) remains one of the most lethal gynecologic malignancies due to late diagnosis, rapid progression, and frequent chemoresistance. Despite advances in targeted therapy, durable responses are uncommon, underscoring the need for novel multitarget agents capable of modulating key oncogenic networks. Medicarpin, a natural pterocarpan phytoalexin, exhibits diverse pharmacological activities; however, its molecular mechanisms in OC are poorly defined. This study employed an integrative in silico framework combining network pharmacology, pathway enrichment, molecular docking, and survival analysis to elucidate medicarpin’s therapeutic landscape in OC. A total of 107 overlapping targets were identified, resulting in a dense protein–protein interaction network enriched in kinase-mediated and apoptotic signaling pathways. Ten hub genes were emphasized: CASP3, ESR1, mTOR, PIK3CA, CCND1, GSK3B, CDK4, PARP1, CHEK1, and ABL1. Gene Ontology and KEGG analyses demonstrated substantial enrichment in the PI3K–Akt/mTOR and prolactin signaling pathways. Docking revealed the stable binding of medicarpin to CASP3 (−6.13 kcal/mol) and ESR1 (−7.68 kcal/mol), supporting its dual regulation of hormonal and apoptotic processes. Although CASP3 and ESR1 expression alone lacked prognostic significance, their network interplay suggests synergistic relevance. Medicarpin exhibits multitarget anticancer potential in OC by modulating kinase-driven and hormone-dependent pathways, warranting further experimental validation. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

13 pages, 3394 KB  
Article
Resveratrol Prevents Breast Cancer Metastasis by Inhibiting Wnt/β-Catenin Pathway-Mediated Epithelial–Mesenchymal Transition
by Xue Fang, En Ma, Runshu Wang, Jingwei Zhang, Yirong Tang, Jinxiao Chen, Weidong Zhu, Da Wo and Dan-ni Ren
Pharmaceuticals 2026, 19(1), 41; https://doi.org/10.3390/ph19010041 - 23 Dec 2025
Abstract
Background: Breast cancer is the most prevalent cancer in women, and metastatic breast cancer remains a major cause of cancer-related deaths. Resveratrol (RSV) is a natural compound found in various plants and is known to exhibit various anti-cancer effects. The present study [...] Read more.
Background: Breast cancer is the most prevalent cancer in women, and metastatic breast cancer remains a major cause of cancer-related deaths. Resveratrol (RSV) is a natural compound found in various plants and is known to exhibit various anti-cancer effects. The present study aims to investigate the therapeutic effects and mechanisms of RSV in inhibiting breast cancer metastasis in a murine model of 4T1 breast tumor that shares close molecular features with human triple negative breast cancer. Methods: Murine breast cancer 4T1 cells were used to examine the effects of RSV on breast cancer metastasis and epithelial–mesenchymal transition (EMT). In vitro cell proliferation and Transwell migration assays and in vivo 4T1 tumor transplantation models were established in female Balb/c mice to determine the anti-metastatic effects of RSV and its mechanism of action. Results: RSV significantly inhibited 4T1 tumor cell migration and significantly decreased expression levels of EMT markers Snail and Vimentin, as well as the nuclear translocation of β-catenin both in vitro and in vivo. Knockdown of β-catenin similarly reduced the expression levels of EMT markers. RSV significantly decreased the number of lung metastases in 4T1-implanted mice by inhibiting Wnt/β-catenin signaling pathway activation. RSV (150 mg/kg/day) reduced the number of visible tumor metastatic nodules and the histological count of metastatic lung carcinomas by 51.82% and 62.58%, respectively, compared to vehicle administration. Conclusions: Our study provides important new mechanistic insight into the strong anti-cancer effects of RSV in inhibiting 4T1 breast cancer metastasis by preventing Wnt/β-catenin signaling pathway-mediated epithelial–mesenchymal transition. These findings suggest the therapeutic potential of RSV as a promising drug in the treatment of metastatic breast cancer. Full article
(This article belongs to the Section Medicinal Chemistry)
33 pages, 1901 KB  
Review
Ligustilide: A Phytochemical with Potential in Combating Cancer Development and Progression—A Comprehensive and Critical Review
by Victória Dogani Rodrigues, Mayara Longui Cabrini, Eliana de Souza Bastos Mazuqueli Pereira, Manuela dos Santos Bueno, Virgínia Maria Cavallari Strozze Catharin, Jesselina Francisco dos Santos Haber, Rachel Gomes Eleutério, Lidiane Indiani, Vitor Cavallari Strozze Catharin, Raquel Cristina Ferraroni Sanches, Flávia Cristina Castilho Carácio, Tereza Lais Menegucci Zutin, Vitor Engrácia Valenti, Sandra Maria Barbalho and Lucas Fornari Laurindo
Pharmaceuticals 2026, 19(1), 36; https://doi.org/10.3390/ph19010036 - 23 Dec 2025
Abstract
Cancer remains one of the leading global health challenges, with increasing resistance to conventional therapies hindering treatment efficacy. Ligustilide, a bioactive compound derived from Ligusticum chuanxiong, has garnered attention for its multifaceted pharmacological properties, including anti-inflammatory, neuroprotective, and anticancer effects. This review [...] Read more.
Cancer remains one of the leading global health challenges, with increasing resistance to conventional therapies hindering treatment efficacy. Ligustilide, a bioactive compound derived from Ligusticum chuanxiong, has garnered attention for its multifaceted pharmacological properties, including anti-inflammatory, neuroprotective, and anticancer effects. This review comprehensively examines Ligustilide and its isomer, (Z)-Ligustilide, focusing on their anticancer potential across various cancer types. Ligustilide exerts its therapeutic effects through multiple mechanisms, including inhibition of cell proliferation, induction of apoptosis, and modulation of autophagy. Additionally, (Z)-Ligustilide has been shown to enhance drug sensitivity and modulate epigenetic regulation, providing a novel approach to overcoming chemoresistance. Despite promising preclinical results, the precise molecular mechanisms, pharmacokinetics, and bioavailability of Ligustilide remain under investigation. Future research should focus on optimizing its therapeutic applications, exploring its synergy with other chemotherapeutic agents, and assessing its potential in personalized cancer therapies. This review offers an in-depth analysis of Ligustilide’s anticancer mechanisms, its role in overcoming drug resistance, and its potential as a novel therapeutic strategy in cancer treatment. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

21 pages, 3020 KB  
Article
Potential Anticancer Effect of Cannabis sativa L. Dichloromethane Extract Through Oxidative Stress-Related Pathways and the Inhibition of the Migration and Invasiveness of Human Breast Cancer Cells (MDA-MB-231 and MCF-7)
by Corinne Raïssa Ngnameko, Jacqueline Njikam Manjia and Motlalepula Gilbert Matsabisa
Int. J. Mol. Sci. 2026, 27(1), 152; https://doi.org/10.3390/ijms27010152 - 23 Dec 2025
Abstract
Breast cancer remains a leading cause of cancer-related morbidity and mortality globally, highlighting the urgent need for novel therapeutic strategies. This study investigates the molecular mechanisms underlying the anti-proliferative potential of Cannabis sativa dichloromethane extract (C. sativa DCM) on oxidative stress, apoptosis, [...] Read more.
Breast cancer remains a leading cause of cancer-related morbidity and mortality globally, highlighting the urgent need for novel therapeutic strategies. This study investigates the molecular mechanisms underlying the anti-proliferative potential of Cannabis sativa dichloromethane extract (C. sativa DCM) on oxidative stress, apoptosis, and invasion in human breast cancer cells. Key biomarkers, such as antioxidant enzymes (Superoxide Dismutase (SOD) and Glutathione (GSH)), the transcription factor Nrf2, apoptotic proteins (p53, caspase-8 and 9), metalloproteinase (MMP-1 and MMP-9), and Transforming Growth Factor Beta (TGF-β) were examined. Cytotoxicity was assessed using an MTT assay in the MDA-MB-231 and MCF-7 breast cancer cell lines, with comparisons to normal skin fibroblasts (HS27). Oxidative stress biomarkers were quantified using enzymatic assays and ELISA kits, while apoptotic and anti-metastatic factors were determined by Western blotting. Results demonstrated that C. sativa DCM extract induced significant cell death in a concentration-dependent manner, with IC50 values of 75.46 ± 0.132 μg/mL for MDA-MB-231 and 78.68 ± 0.50 μg/mL for MCF-7 cells. The extract decreased SOD and GSH levels while increasing p53 and caspase activity, confirming apoptosis activation. Additionally, C. sativa DCM inhibited migration and invasion by downregulating MMP-1, MMP-9, and TGF-β. The anti-proliferative potential of C. sativa DCM in breast cancer cells is mediated through a continuous biological pathway involving oxidative stress modulation, apoptotic signaling, and anti-invasive effects. Phytochemical analysis revealed terpenoids and steroids, including compounds like cannabidiol and tetrahydrocannabinol acid. These findings suggest that C. sativa DCM extract holds potential as an anti-breast cancer therapeutic and warrants further preclinical and clinical investigations. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

21 pages, 43352 KB  
Article
Green Synthesis of Copper-Doped ZrO2 Nanoparticles Using Calendula officinalis Flower Extract: Comprehensive Characterization, Biocompatibility, and Anticancer Evaluation
by Dhruv Suraneni, Balasubramanian Deepika, Kavinithi Jaganathan Mahadevan, Sanjana Raghupathy, Shangavy Pandiarajan, Devadass Jessy Mercy, Agnishwar Girigoswami, Sanjay Kisan Metkar, Surajit Hansda and Koyeli Girigoswami
Physchem 2026, 6(1), 1; https://doi.org/10.3390/physchem6010001 - 23 Dec 2025
Abstract
The field of nanotechnology has witnessed a paradigm shift towards eco-friendly and sustainable synthesis methods for nanoparticles due to increasing concerns over environmental toxicity and resource sustainability. Among various metal oxide nanoparticles, zirconium dioxide (ZrO2) nanoparticles have garnered significant attention owing [...] Read more.
The field of nanotechnology has witnessed a paradigm shift towards eco-friendly and sustainable synthesis methods for nanoparticles due to increasing concerns over environmental toxicity and resource sustainability. Among various metal oxide nanoparticles, zirconium dioxide (ZrO2) nanoparticles have garnered significant attention owing to their exceptional thermal stability, biocompatibility, mechanical strength, and catalytic properties. Doping ZrO2 with transition metals such as copper (Cu) further enhances its physicochemical attributes, including antibacterial activity, redox behaviour, and electronic properties, rendering it suitable for a diverse range of biomedical and industrial applications. In the present study, we report the green synthesis of copper-doped ZrO2 nanoparticles (Cu-ZrO2-CO NPs) using an aqueous extract of Calendula officinalis (marigold) flowers as a natural reducing and stabilizing agent. The complete characterization was performed using UV–vis spectrophotometry, dynamic light scattering (DLS), zeta potential, FTIR, SEM, EDAX, and XRD, revealing its size to be around 20–40 nm and zeta potential as −20 mV, indicating nano size and stability. The biocompatibility of the as-synthesized nanoparticle was analyzed in vitro using fibroblast cell viability and haemolysis assay, and in vivo using brine shrimp assay. The nanoparticles were safe up to a dose of 50 μg/mL, showing more than 95% cell viability and less than 2% haemolysis, which is within an acceptable range. Finally, the anticancer activity was explored for A549 cells by MTT assay and live-dead assay, with an IC50 value of 38.63 μg/mL. The chorioallantoic membrane (CAM) model was used to assess the anti-angiogenesis potential of the Cu-ZrO2-CO NPs. The results showed that the nanoparticles could kill the cancer cells via apoptosis, and one of the reasons for the anticancer effect was angiogenesis inhibition. Further research is needed using other cancer cell lines and animal tumour models. Full article
(This article belongs to the Section Biophysical Chemistry)
Show Figures

Graphical abstract

24 pages, 2711 KB  
Article
Evaluation of Cedrus atlantica Essential Oil: Chemical Composition, Anticancer Activity and Molecular Docking Studies
by Silvia Gruin, Octavian Crețu, Alexandra Mioc, Marius Mioc, Alexandra Prodea, Elisabeta Atyim, Alexandra Teodora Lukinich-Gruia, Maria-Alexandra Pricop, Armand Gogulescu and Codruța Șoica
Molecules 2026, 31(1), 46; https://doi.org/10.3390/molecules31010046 - 22 Dec 2025
Abstract
Due to their high content of bioactive compounds with anticancer properties, essential oils (EO) are increasingly viewed as valuable therapeutic strategies in oncology. The aim of this study was to evaluate the chemical composition and anticancer activity of Cedrus atlantica EO (CAEO) and [...] Read more.
Due to their high content of bioactive compounds with anticancer properties, essential oils (EO) are increasingly viewed as valuable therapeutic strategies in oncology. The aim of this study was to evaluate the chemical composition and anticancer activity of Cedrus atlantica EO (CAEO) and its PEG-400 and Tween 20 formulations. The gas-chromatography (GC) analysis revealed a sesquiterpene-rich profile, with β-himachalene (39.32%) as the major constituent, followed by α-Himachalene (16.76%) and γ-Himachalene (12.92%). The cytotoxicity studies, performed using Alamar Blue assay on normal HaCaT human keratinocytes and A375 human melanoma and HT-29 colorectal carcinoma cell lines, revealed that CAEO displayed minimal toxicity on HaCaT cells, while significantly reducing A735 and HT-29 cell viability, at any of the concentrations tested. The PEG- and Tween-based formulations of CAEO exhibited the same effect on cell viability as the simple water dispersion of CAEO. The immunofluorescence-based examination of cellular morphology suggested that CAEO induces apoptosis in both cancer cell lines: A375 and HT-29; this apoptosis-related mechanism was further supported by the caspase-3/7 assay, which revealed a significant increase in caspase-3/7 activity after CAEO treatment. To further investigate the underlying mechanism, the JC-1 staining and high-resolution respirometry assays demonstrated that CAEO induces mitochondrial membrane depolarization and reduced mitochondrial active respiration (OXPHOS). Molecular docking further indicated that isoledene and β-himachalene exhibit the highest predicted affinity for PI3Kγ, suggesting a potential involvement of PI3K-related signaling in the pro-apoptotic activity of CAEO. Together, these results suggest that CAEO induces apoptosis through a mitochondria-mediated mechanism. Full article
19 pages, 1625 KB  
Article
Enhanced Antitumor Effect of Oncolytic Virotherapy Combined with mRNA-Encoded Immunoadjuvants in Colorectal Carcinoma (CT26) Tumor Model
by Anastasia Ryapolova, Daniil Shevyrev, Angela Tsvetkova, Olesya Sokolova, Olga Vasileva, Vladimir Andriianov, Bogdan Krapivin, Alena Kozlova, Kristina Vorona, Sofia Kondratieva, Roman A. Ivanov, Alexander Karabelsky, Vasiliy Reshetnikov and Ekaterina Minskaia
Appl. Biosci. 2026, 5(1), 1; https://doi.org/10.3390/applbiosci5010001 - 22 Dec 2025
Viewed by 6
Abstract
The search for effective anti-cancer therapies is one of the most significant goals of modern medicine. The combination of oncolytic viruses (OV) and mRNA immunoadjuvants can significantly improve the outcome or even substitute traditional immunotherapy. In addition to the direct OV-mediated cytotoxic elimination [...] Read more.
The search for effective anti-cancer therapies is one of the most significant goals of modern medicine. The combination of oncolytic viruses (OV) and mRNA immunoadjuvants can significantly improve the outcome or even substitute traditional immunotherapy. In addition to the direct OV-mediated cytotoxic elimination of tumor cells, both OV and mRNA immunoadjuvants can significantly alter the immunosuppressive tumor microenvironment (TME) supporting cancer cells and unleash the immune response against the malignant cells. The present study is aimed at assessing the therapeutic effects of recombinant vesicular stomatitis virus (rVSV) and lipid nanoparticles (LNP) delivering mRNA coding for murine interleukin-12 (mIL12) and granulocyte-macrophage colony-stimulating factor (mGMCSF) (mRNA-LNP) in colorectal carcinoma CT26-induced tumors both as independent therapies and in combination with each other. The results of the in vivo experiment on BALB/c mice demonstrated that rVSV monotherapy did not have a significant effect, with the tumor growth inhibition index (TGII) ranging from 13.7 to 29.8% on days 6–10 after the therapy start. While monotherapy with mRNA-LNP was more effective (TGII of 48.6–53.7%), it was the therapy combining the two approaches (rVSV and mRNA-LNP) that resulted in the highest TGII of 66.7% on day 10. While these results can be further improved by optimizing the experimental design, they show the great potential of combination immunotherapy for the treatment of oncological diseases. Full article
Show Figures

Figure 1

31 pages, 2913 KB  
Review
Hinokiflavone as a Potential Antitumor Agent: From Pharmacology to Pharmaceutics
by Fengrui Liu, Ranyi Li, Xiaolei Zhou and Xiaoyu Li
Cells 2026, 15(1), 17; https://doi.org/10.3390/cells15010017 - 22 Dec 2025
Viewed by 29
Abstract
Hinokiflavone (HF), a natural C-O-C-linked biflavonoid originally isolated from Chamaecyparis obtusa, is a promising multifunctional antitumor agent. Despite challenges posed by multidrug resistance and tumor heterogeneity, HF demonstrates notable therapeutic potential through a multifaceted pharmacological profile. HF exerts broad-spectrum anticancer effects by [...] Read more.
Hinokiflavone (HF), a natural C-O-C-linked biflavonoid originally isolated from Chamaecyparis obtusa, is a promising multifunctional antitumor agent. Despite challenges posed by multidrug resistance and tumor heterogeneity, HF demonstrates notable therapeutic potential through a multifaceted pharmacological profile. HF exerts broad-spectrum anticancer effects by targeting multiple oncogenic pathways, including the MDM2-p53 axis, MAPK/JNK/NF-κB signaling, ROS/JNK-mediated apoptosis, and Bcl-2/Bax-regulated mitochondrial pathways. These actions are further complemented by inhibition of cell proliferation through cell cycle arrest and suppression of metastasis via downregulation of matrix metalloproteinases and reversal of epithelial–mesenchymal transition. Additionally, HF displays antioxidant, anti-inflammatory, and antimicrobial activities, enhancing treatment efficacy. However, its clinical translation remains limited by poor aqueous solubility, low oral bioavailability, and incomplete pharmacokinetic characterization. Recent advances in nanotechnology-based formulation strategies, such as polymeric micelles and metal–organic frameworks, have enhanced HF’s bioavailability and in vivo antitumor efficacy. This review comprehensively delineates HF’s molecular mechanisms of anticancer action, evaluates its pharmacokinetics and bioformulation developments, and highlights challenges and prospects for clinical application. Integration of tumor microenvironment-responsive delivery systems with synergistic therapeutic strategies is essential to fully realize HF’s therapeutic potential, positioning it as a valuable scaffold for novel anticancer drug development. Full article
Show Figures

Figure 1

17 pages, 9440 KB  
Article
Gedunin Impacts Pancreatic Cancer Stem Cells Through the Sonic Hedgehog Signaling Pathway
by Karla Perez, Sheryl Rodriguez, Jose Barragan, Poornimadevi Narayanan, Alberto Ruiseco, Preetha Rajkumar, Nallely Ramirez, Victor Vasquez, Rajkumar Lakshmanaswamy and Ramadevi Subramani
Pharmaceuticals 2026, 19(1), 19; https://doi.org/10.3390/ph19010019 - 22 Dec 2025
Viewed by 20
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with a high rate of recurrence and a dismal prognosis. Studies have shown that pancreatic cancer stem cells (PCSCs) are a subpopulation that contributes to tumor progression, resistance to therapeutics, and metastasis, making [...] Read more.
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with a high rate of recurrence and a dismal prognosis. Studies have shown that pancreatic cancer stem cells (PCSCs) are a subpopulation that contributes to tumor progression, resistance to therapeutics, and metastasis, making them a key subpopulation to target for treatment. Gedunin (GD), a natural compound derived from Azadirachta indica (neem), has shown anticancer properties in pancreatic cancer cells, but its effects on PCSCs remains unclear. This study evaluated the effects of GD in pancreatic cancer stem cells, highlighting its impacts on tumor growth and progression and focusing on its impact on the sonic hedgehog (Shh) signaling pathway. Methods: Functional assays were performed to assess the effect of GD on the sphere-forming ability, colony formation, and self-renewal of PCSCs. Athymic mice xenograft models were utilized to evaluate the tumor suppression effect of GD in vivo. Furthermore, the anticancer effect of GD on PCSCs was assessed using both in vitro and in vivo limiting dilution assay. GD-induced changes in Shh signaling and key stem cell marker expressions in PCSCs were evaluated. Results: GD effectively inhibited tumor growth in xenograft models and reduced the percentage of PCSCs. GD was effective in decreasing PCSCs’ proliferative, self-renewal, and colony-forming capacity. GD decreased the protein expression levels of key Shh signaling markers Gli1 and Shh, stem cell markers SOX2, Nanog, and Oct4, metastasis-related proteins MMP-2, MMP-3, and MMP-9, and EMT markers Tgf1, Slug, Snail, and Twist in both PDAC cells and PCSCs. We demonstrated a significant decrease in the spheroid formation and self-renewal capacity of the (ALDH+) PCSC population following GD treatment in HPAC cells, indicating its potential antagonistic effects on PCSCs. GD was highly effective in reducing tumor volume, stemness, and metastasis in both early and late chemotherapy. In vivo limiting dilution assay using CD133+/LGR5+ PCSC xenografts demonstrated that GD reduces tumor growth, metastasis, and stemness associated with PCSCs by downregulating the expression of Shh and Gli1. GD treatment also reduced micrometastatic lesions in the lung, liver, and brain, as identified using H&E staining. Conclusions: The findings highlight GD’s potential as a promising therapeutic candidate for PDAC, with the ability to target both bulk tumor cells and PCSCs. By simultaneously suppressing tumor growth, stemness, and metastatic spread, GD may contribute to more effective treatment strategies and improved patient outcomes. Full article
(This article belongs to the Special Issue Anticancer Compounds in Medicinal Plants—4th Edition)
Show Figures

Graphical abstract

Back to TopTop