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

Article Types

Countries / Regions

Search Results (148)

Search Parameters:
Keywords = antimitotics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
41 pages, 8942 KB  
Article
Synthesis, Computational Studies, and Structural Analysis of 1-(3,5-Dimethoxyphenyl)azetidin-2-ones with Antiproliferative Activity in Breast Cancer and Chemoresistant Colon Cancer
by Azizah M. Malebari, Shubhangi Kandwal, Abdirahman Ali, Darren Fayne, Brendan Twamley, Daniela M. Zisterer and Mary J. Meegan
Pharmaceuticals 2025, 18(9), 1330; https://doi.org/10.3390/ph18091330 - 5 Sep 2025
Abstract
Background/Objectives: A series of 1-(3,5-dimethoxyphenyl)azetidine-2-ones were synthesised to evaluate their antiproliferative activity in MCF-7 breast cancer cells and HT-29 chemoresistant colon cancer cells. The 1,4-diarylazetidin-2-ones were designed by replacing the characteristic 3,4,5-trimethoxyphenyl Ring A of the antimitotic stilbene combretastatin CA-4 with a [...] Read more.
Background/Objectives: A series of 1-(3,5-dimethoxyphenyl)azetidine-2-ones were synthesised to evaluate their antiproliferative activity in MCF-7 breast cancer cells and HT-29 chemoresistant colon cancer cells. The 1,4-diarylazetidin-2-ones were designed by replacing the characteristic 3,4,5-trimethoxyphenyl Ring A of the antimitotic stilbene combretastatin CA-4 with a 3,5-dimethoxyphenyl substituent at N-1, together with phenyl, hydroxyl, and phenoxy substituents at C-3 of the four-membered ring. Methods: A panel of 12 novel compounds was synthesized and evaluated in estrogen receptor (ER)- and progesterone receptor (PR)-positive MCF-7 breast cancer cells followed with the more potent compounds further evaluated in HT-29 chemoresistant colon cancer cells. Cytotoxicity was determined by LDH assay. The structures of the 1-(3,5-dimethoxyphenyl)azetidine-2-ones 12i, 12k, 12o, 12p together with the 1-(3,5-dimethoxyphenyl)azetidine-2-one 12s were determined by X-ray crystallography. The trans configuration of the C-3 and C-4 substituents of the β-lactam ring was confirmed for compounds 12k and 12u. Molecular modelling and molecular dynamics studies examined the molecular interactions of the compounds with the colchicine binding site of tubulin. Results: The 1-(3,5-Dimethoxyphenyl)-4-(4-ethoxyphenyl)-3-hydroxyazetidin-2-one 12l was identified as the most potent antiproliferative compound in the series (with an IC50 value of 10 nM in MCF-7 breast cancer cells and 3 nM in HT-29 colon cancer cells) and with greater potency than CA-4 in the chemoresistant HT-29 cells. Computational docking studies predicted binding conformations for 12l and the related series of compounds in the colchicine binding site of tubulin and rationalised the impact of the 3,5-dimethoxyphenyl substituent at N-1 of the azetidine-2-one on activity. Conclusions: These findings indicate that the novel 1-(3,5-dimethoxyphenyl)-2-azetidinone 12l is a suitable candidate for further investigation as a potential antiproliferative microtubule-targeting agent for breast and chemoresistant colon cancers. Full article
(This article belongs to the Special Issue Advances in Medicinal Chemistry: 2nd Edition)
Show Figures

Figure 1

12 pages, 6471 KB  
Article
The Effects of Co-Culturing ND7/23 Sensory Neuron-like Cells and IFRS1 Schwann Cells on Myelination: A Single-Arm Nonrandomized Study
by Shizuka Takaku and Kazunori Sango
Neurol. Int. 2025, 17(9), 138; https://doi.org/10.3390/neurolint17090138 - 1 Sep 2025
Viewed by 243
Abstract
Background/Objectives: Co-culture models of neurons and Schwann cells have been used to explore the mechanisms of myelination during development, axonal regeneration after injury, and the pathogenesis of various demyelinating neuropathies. A spontaneously immortalized Fischer rat Schwann cell line 1 (IFRS1), established from [...] Read more.
Background/Objectives: Co-culture models of neurons and Schwann cells have been used to explore the mechanisms of myelination during development, axonal regeneration after injury, and the pathogenesis of various demyelinating neuropathies. A spontaneously immortalized Fischer rat Schwann cell line 1 (IFRS1), established from the primary culture of adult Fischer344 rat peripheral nerves, can myelinate neurites in co-cultures with primary cultured dorsal root ganglion neurons and neuronal cell lines, such as nerve growth factor (NGF)-primed PC12 cells and NSC-34 motor neuron-like cells. In this study, we aimed to establish a stable co-culture system using IFRS1 cells and ND7/23 sensory neuron-like cells. Methods: ND7/23 cells were seeded at a low density (2 × 103/cm2) and maintained for 7 days in serum-containing medium supplemented with NGF (10 ng/mL) and the Rho kinase inhibitor Y27632 (5 μM) to promote neurite elongation. The cells were then treated with the anti-mitotic agent mitomycin C (1 μg/mL) for 12–16 h to suppress proliferative activity. Following this, the cells were co-cultured with IFRS1 cells (2 × 104/cm2) and maintained at 37 °C in serum-containing medium supplemented with ascorbic acid (50 μg/mL), NGF (10 ng/mL), and ciliary neurotrophic factor (10 ng/mL). Results: Double-immunofluorescence staining performed on day 21 of the co-culture revealed myelin protein 22- or myelin basic protein-immunoreactive IFRS1 cells surrounding βIII tubulin-immunoreactive neurites emerging from ND7/23 cells. Myelin formation was further confirmed via Sudan Black B staining and electron microscopy. Conclusions: This co-culture system may provide a valuable tool for studying the processes of myelination in the peripheral nervous system, as well as the pathogenesis of various sensory neuropathies and potential novel therapeutic approaches for these conditions. Full article
Show Figures

Figure 1

2 pages, 446 KB  
Correction
Correction: Malebari et al. Synthesis and Antiproliferative Evaluation of 3-Chloroazetidin-2-ones with Antimitotic Activity: Heterocyclic Bridged Analogues of Combretastatin A-4. Pharmaceuticals 2021, 14, 1119
by Azizah M. Malebari, Shu Wang, Thomas F. Greene, Niamh M. O’Boyle, Darren Fayne, Mohemmed Faraz Khan, Seema M. Nathwani, Brendan Twamley, Thomas McCabe, Daniela M. Zisterer and Mary J. Meegan
Pharmaceuticals 2025, 18(8), 1219; https://doi.org/10.3390/ph18081219 - 19 Aug 2025
Viewed by 258
Abstract
In the original publication [...] Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 8

27 pages, 1680 KB  
Review
Microtubule-Targeting Agents: Advances in Tubulin Binding and Small Molecule Therapy for Gliomas and Neurodegenerative Diseases
by Maya Ezzo and Sandrine Etienne-Manneville
Int. J. Mol. Sci. 2025, 26(15), 7652; https://doi.org/10.3390/ijms26157652 - 7 Aug 2025
Viewed by 1121
Abstract
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central [...] Read more.
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central nervous system (CNS) applications, including brain malignancies such as gliomas and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Microtubule-stabilizing agents, such as taxanes and epothilones, promote microtubule assembly and have shown efficacy in both tumour suppression and neuronal repair, though their CNS use is hindered by blood–brain barrier (BBB) permeability and neurotoxicity. Destabilizing agents, including colchicine-site and vinca domain binders, offer potent anticancer effects but pose greater risks for neuronal toxicity. This review highlights the mapping of nine distinct tubulin binding pockets—including classical (taxane, vinca, colchicine) and emerging (tumabulin, pironetin) sites—that offer new pharmacological entry points. We summarize the recent advances in structural biology and drug design, enabling MTAs to move beyond anti-mitotic roles, unlocking applications in both cancer and neurodegeneration for next-generation MTAs with enhanced specificity and BBB penetration. We further discuss the therapeutic potential of combination strategies, including MTAs with radiation, histone deacetylase (HDAC) inhibitors, or antibody–drug conjugates, that show synergistic effects in glioblastoma models. Furthermore, innovative delivery systems like nanoparticles and liposomes are enhancing CNS drug delivery. Overall, MTAs continue to evolve as multifunctional tools with expanding applications across oncology and neurology, with future therapies focusing on optimizing efficacy, reducing toxicity, and overcoming therapeutic resistance in brain-related diseases. Full article
(This article belongs to the Special Issue New Drugs Regulating Cytoskeletons in Human Health and Diseases)
Show Figures

Figure 1

24 pages, 2374 KB  
Review
The Role of Colchicine in Plant Breeding
by Baljinder Singh, Sunyoung Yun, Yeji Gil and Myoung-Hwan Park
Int. J. Mol. Sci. 2025, 26(14), 6743; https://doi.org/10.3390/ijms26146743 - 14 Jul 2025
Cited by 1 | Viewed by 1064
Abstract
Colchicine, a strong antimitotic drug produced by the crocus Colchicum autumnale, induces polyploidy by interfering with spindle formation during mitosis, making it a crucial tool in plant breeding. In this review, we give a comprehensive overview of the function of colchicine in [...] Read more.
Colchicine, a strong antimitotic drug produced by the crocus Colchicum autumnale, induces polyploidy by interfering with spindle formation during mitosis, making it a crucial tool in plant breeding. In this review, we give a comprehensive overview of the function of colchicine in plant enhancement, emphasizing its modes of action, application techniques, and effects on phytochemistry, physiology, and plant morphology. A wide variety of plant species, especially medicinal plants, have been studied in this context, utilizing in vitro, ex vitro, and in vivo methods for applying colchicine. In addition, we discuss the safety and effectiveness of colchicine in comparison to other polyploidy-inducing drugs, including oryzalin, trifluralin, and mutagens such as ethyl methanesulfonate and methyl methanesulfonate. Furthermore, the effects of colchicine on genetic stability and secondary metabolite production are discussed, with a focus on its usefulness in boosting the medicinal and economic potential of the target species. This synthesis highlights the ongoing use of colchicine in plant breeding and provides useful information and suggestions for future advancements in crop development via induced polyploidy. Full article
(This article belongs to the Special Issue New Insights in Plant Cell Biology)
Show Figures

Figure 1

15 pages, 259 KB  
Review
Predictive Factors of Response to Neoadjuvant Chemotherapy (NACT) and Immune Checkpoint Inhibitors in Early-Stage Triple-Negative Breast Cancer Patients (TNBC)
by Khashayar Yazdanpanah Ardakani, Francesca Fulvia Pepe, Serena Capici, Thoma Dario Clementi and Marina Elena Cazzaniga
Curr. Oncol. 2025, 32(7), 387; https://doi.org/10.3390/curroncol32070387 - 4 Jul 2025
Viewed by 982
Abstract
Triple-negative breast cancer (TNBC) is a heterogenous group of breast tumors. This type of breast tumor is relatively difficult to manage, due to the lack of expression of Hormone Receptors (HR) and human epidermal growth factor receptor (HER2). Efforts have been made to [...] Read more.
Triple-negative breast cancer (TNBC) is a heterogenous group of breast tumors. This type of breast tumor is relatively difficult to manage, due to the lack of expression of Hormone Receptors (HR) and human epidermal growth factor receptor (HER2). Efforts have been made to understand the factors involved in determining how a triple-negative breast tumor responds to therapy. The standard of treatment in most cases today is a combined modality of immune checkpoint inhibitors (ICIs) and chemotherapy with agents such as anti-mitotic (taxanes) or DNA-damaging agents (alkylating agents, cyclophosphamides, platin salts). In this study, we investigated the predictive and prognostic factors for TNBC, in the neoadjuvant setting; understanding each patient’s response before treatment initiation is crucial to guiding the subsequent approach and finally improving patient outcomes. We focused on tumor-infiltrating lymphocytes at the site of the primary tumor (TILs), circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), the mutational status of protein 53 (p53), and Ki-67, investigating the potential roles of these factors in predicting responses to anti-cancer agents. Full article
(This article belongs to the Special Issue Advances in Immunotherapy for Breast Cancer)
23 pages, 2553 KB  
Article
A Green Integrated Approach to Multifunctional Silver Nanoparticles Derived from Aronia melanocarpa
by Andreia Corciova, Cornelia Mircea, Adrian Fifere, Ioana-Andreea Turin Moleavin, Ana Flavia Burlec, Bianca Ivanescu, Ana-Maria Vlase, Monica Hancianu and Irina Macovei
Pharmaceutics 2025, 17(5), 669; https://doi.org/10.3390/pharmaceutics17050669 - 20 May 2025
Viewed by 742
Abstract
Background/Objectives: This study reports the green synthesis, optimization, characterization, and multifunctional evaluation of silver nanoparticles (AgNPs) using an ethanolic Aronia melanocarpa berry extract. The objective was to establish optimal synthesis conditions; assess the in vitro stability; and evaluate the antioxidant, photocatalytic, and photoprotective [...] Read more.
Background/Objectives: This study reports the green synthesis, optimization, characterization, and multifunctional evaluation of silver nanoparticles (AgNPs) using an ethanolic Aronia melanocarpa berry extract. The objective was to establish optimal synthesis conditions; assess the in vitro stability; and evaluate the antioxidant, photocatalytic, and photoprotective activities. Methods: The cytogenotoxic effects of the AgNPs were evaluated on Triticum aestivum roots. The AgNPs were synthesized via bioreduction using an ethanolic extract of A. melanocarpa under varied pH, AgNO3 concentration, extract/AgNO3 ratio, temperature, and stirring time, with optimization guided by UV–Vis spectral analysis. The AgNPs were further characterized by FTIR, DLS, TEM, and EDX. In vitro stability was evaluated over six months in different dispersion media (ultrapure water; 5% NaCl; and PBS at pH 6, 7, and 8). Biological assessments included antioxidant assays (lipoxygenase inhibition, DPPH radical scavenging, metal chelation, and hydroxyl radical scavenging), photocatalytic dye degradation, and SPF determination. Results: Optimal synthesis was achieved at pH 8, 3 mM AgNO3, extract/AgNO3 ratio of 1:9, 40 °C, and 240 min stirring. The AgNPs were spherical (TEM), well dispersed (PDI = 0.32), and highly stable (zeta potential = −40.71 mV). PBS pH 6 and 7 ensured the best long-term colloidal stability. The AgNPs displayed strong dose-dependent antioxidant activity, with superior lipoxygenase inhibition (EC50 = 18.29 µg/mL) and the effective photocatalytic degradation of dyes under sunlight. Photoprotective properties were confirmed through UV absorption analysis. The AgNPs showed a strong antimitotic effect on wheat root cells. Conclusions: The study demonstrates that A. melanocarpa-mediated AgNPs are stable, biologically active, and suitable for potential biomedical, cosmetic, and environmental applications, reinforcing the relevance of plant-based nanotechnology. Full article
Show Figures

Figure 1

13 pages, 3427 KB  
Article
Carrot (Daucus carota L.) Haploid Embryo Genome Doubling with Colchicine and Trifluralin
by Maria Fomicheva, Elena Kozar and Elena Domblides
Horticulturae 2025, 11(5), 505; https://doi.org/10.3390/horticulturae11050505 - 8 May 2025
Viewed by 662
Abstract
The production of carrot (D. carota L.) doubled haploids (DH) for the acceleration of this important vegetable crop breeding requires genome doubling of haploid regenerants. If spontaneous doubling does not occur, artificial chromosome doubling can be complicated by the lack of efficient [...] Read more.
The production of carrot (D. carota L.) doubled haploids (DH) for the acceleration of this important vegetable crop breeding requires genome doubling of haploid regenerants. If spontaneous doubling does not occur, artificial chromosome doubling can be complicated by the lack of efficient genome-doubling protocols. We tested an antimitotic agent treatment of carrot at the embryo stage. It allowed us to produce and treat a large number of clonal carrot embryos (at least 30 embryos per treatment condition) in small volumes with minimal reagent amounts. We showed that 0.01–1 g/L colchicine did not perturb carrot development. Trifluralin showed no signs of toxicity at 0.001 and 0.01 g/L concentrations, but 0.1 g/L trifluralin reduced survival by 40% and delayed plantlet regeneration. We showed via DNA content flow cytometry that 0.01–1 g/L colchicine and 0.001–0.1 g/L trifluralin could double the carrot genome. The highest diploid percent was observed at 1 g/L colchicine (34%) and 0.1 g/L trifluralin (28%). The highest percent of diploids together with mixoploids (partial diploids) was at 0.01 and 0.1 g/L trifluralin (over 70%), followed by 1 g/L colchicine (56%). To our knowledge, this is the first report on trifluralin application for genome doubling in Apiaceae. In our study, we determined colchicine and trifluralin toxicity and doubling efficiency at different concentrations that can be used for carrot DH-line production and further improvement of genome doubling methods. Full article
(This article belongs to the Special Issue A Decade of Research on Vegetable Crops: From Omics to Biotechnology)
Show Figures

Figure 1

35 pages, 5069 KB  
Review
Small-Molecule Mitotic Inhibitors as Anticancer Agents: Discovery, Classification, Mechanisms of Action, and Clinical Trials
by Yazmin Salinas, Subhash C. Chauhan and Debasish Bandyopadhyay
Int. J. Mol. Sci. 2025, 26(7), 3279; https://doi.org/10.3390/ijms26073279 - 1 Apr 2025
Cited by 1 | Viewed by 2302
Abstract
Despite decades of research, cancer continues to be a disease of great concern to millions of people around the world. It has been responsible for a total of 609,820 deaths in the U.S. alone in 2023. Over the years, many drugs have been [...] Read more.
Despite decades of research, cancer continues to be a disease of great concern to millions of people around the world. It has been responsible for a total of 609,820 deaths in the U.S. alone in 2023. Over the years, many drugs have been developed to remove or reduce the disease’s impact, all with varying mechanisms of action and side effects. One class of these drugs is small-molecule mitotic inhibitors. These drugs inhibit cancer cell mitosis or self-replication, impeding cell proliferation and eventually leading to cell death. In this paper, small-molecule mitotic inhibitors are discussed and classified through their discovery, underlying chemistry, and mechanism(s) of action. The binding/inhibition of microtubule-related proteins, DNA damage through the inhibition of Checkpoint Kinase 1 protein, and the inhibition of mitotic kinase proteins are discussed in terms of their anticancer activity to provide an overview of a variety of mitotic inhibitors currently commercially available or under investigation, including those in ongoing clinical trial. Clinical trials for anti-mitotic agents are discussed to track research progress, gauge current understanding, and identify possible future prospects. Additionally, antibody–drug conjugates that use mitotic inhibitors as cytotoxic payloads are discussed as possible ways of administering effective anticancer treatments with minimal toxicity. Full article
(This article belongs to the Collection Feature Papers in Molecular Oncology)
Show Figures

Figure 1

18 pages, 7179 KB  
Article
Machine Learning-Aided Optimization of In Vitro Tetraploid Induction in Cannabis
by Marzieh Jafari, Nathan Paul, Mohsen Hesami and Andrew Maxwell Phineas Jones
Int. J. Mol. Sci. 2025, 26(4), 1746; https://doi.org/10.3390/ijms26041746 - 18 Feb 2025
Cited by 3 | Viewed by 1606
Abstract
Polyploidy, characterized by an increase in the number of whole sets of chromosomes in an organism, offers a promising avenue for cannabis improvement. Polyploid cannabis plants often exhibit altered morphological, physiological, and biochemical characteristics with a number of potential benefits compared to their [...] Read more.
Polyploidy, characterized by an increase in the number of whole sets of chromosomes in an organism, offers a promising avenue for cannabis improvement. Polyploid cannabis plants often exhibit altered morphological, physiological, and biochemical characteristics with a number of potential benefits compared to their diploid counterparts. The optimization of polyploidy induction, such as the level of antimitotic agents and exposure duration, is essential for successful polyploidization to maximize survival and tetraploid rates while minimizing the number of chimeric mixoploids. In this study, three classification-based machine learning algorithms—probabilistic neural network (PNN), support vector classification (SVC), and k-nearest neighbors (KNNs)—were used to model ploidy levels based on oryzalin concentration and exposure time. The results indicated that PNN outperformed both KNNs and SVC. Subsequently, PNN was combined with a genetic algorithm (GA) to optimize oryzalin concentration and exposure time to maximize tetraploid induction rates. The PNN-GA results predicted that the optimal conditions were a concentration of 32.98 µM of oryzalin for 17.92 h. A validation study testing these conditions confirmed the accuracy of the PNN-GA model, resulting in 93.75% tetraploid induction, with the remaining 6.25% identified as mixoploids. Additionally, the evaluation of morphological traits showed that tetraploid plants were more vigorous and had larger leaf sizes compared to diploid or mixoploid plants in vitro. Full article
Show Figures

Graphical abstract

13 pages, 5669 KB  
Article
Small Molecules Identified by an In Silico Docking Screen Targeting Anaphase-Promoting Complex/Cyclosome Subunit 1 (APC1) Potentiate Paclitaxel-Induced Breast Cancer Cell Death
by Scott C. Schuyler, Rythm Gupta, Tran Thi Bao Nguyen, Cheng-Ye Weng and Hsin-Yu Chen
Molecules 2025, 30(4), 895; https://doi.org/10.3390/molecules30040895 - 14 Feb 2025
Viewed by 1252
Abstract
Delaying mitotic cell cycle progression has been proposed as a strategy to potentiate the effects of anti-mitotic anti-cancer drugs that induce multipolar mitotic spindles. Toward this end, we have performed an in silico docking screen targeting anaphase-promoting complex/cyclosome subunit 1 (APC1) at a [...] Read more.
Delaying mitotic cell cycle progression has been proposed as a strategy to potentiate the effects of anti-mitotic anti-cancer drugs that induce multipolar mitotic spindles. Toward this end, we have performed an in silico docking screen targeting anaphase-promoting complex/cyclosome subunit 1 (APC1) at a conserved 10-amino acid surface site that was modeled to interact via a single hydrogen bond with the essential mitotic anaphase-promoting complex/cyclosome (APC/C) co-factor cell division cycle 20 (CDC20). Five molecules were identified after screening 15,000 small molecules. As a secondary in cellulo bioactivity screening, MDA-MB-231 genomically unstable aneuploid breast cancer cells were exposed to each compound in the absence and presence of 10 nM paclitaxel or 1 nM eribulin, the likely clinically relevant doses of these drugs in these cells. Two of the five compounds, which share a common 2-(trifluoromethyl)quinazolin-4-amine chemical structure, induced elevated levels of cell death in combination with paclitaxel, as observed by fluorescence-activated cell sorting (FACS). These two compounds will now serve as a starting point for further optimization and target validation experiments and for additional in silico screens in search of other chemically related small molecules that display more potent but specific anti-cancer cell effects. Full article
(This article belongs to the Section Computational and Theoretical Chemistry)
Show Figures

Figure 1

15 pages, 2919 KB  
Article
Toxicity, Half-Life and Antitumor Activity of Phenyl 4-(2-Oxo-3-alkylimidazolidin-1-yl)benzenesulfonates as Novel Antimitotic CYP1A1-Targeted Prodrugs in Female Mouse Models
by Atziri Corin Chavez Alvarez, Chahrazed Bouzriba, Vincent Ouellette, Mathieu Gagné-Boulet, Alexandre Patenaude, Sylvie Pilote, René C.-Gaudreault, Chantale Simard and Sébastien Fortin
Pharmaceutics 2025, 17(2), 233; https://doi.org/10.3390/pharmaceutics17020233 - 11 Feb 2025
Viewed by 1095
Abstract
Background/Objectives: Chemoresistance of breast cancers (BCs) is a major impediment to current chemotherapeutics that urges the development of new drugs and new therapeutic approaches. To that end, phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) were recently prepared to fulfill some of the unmet needs with classic [...] Read more.
Background/Objectives: Chemoresistance of breast cancers (BCs) is a major impediment to current chemotherapeutics that urges the development of new drugs and new therapeutic approaches. To that end, phenyl 4-(2-oxo-3-alkylimidazolidin-1-yl)benzenesulfonates (PAIB-SOs) were recently prepared to fulfill some of the unmet needs with classic chemotherapeutics. PAIB-SOs are prodrugs bioactivated into potent antimitotics by the cytochrome P450 1A1 (CYP1A1), which is a frequent enzyme in resistant BC cells, but mostly missing in normal cells. Our screening program studies of PAIB-SO chemolibraries selected three prototypical PAIB-SOs as antimitotic prodrugs amenable for studies using BC animal models. Methods: Healthy female CD1® IGS mice were treated with three prototypical PAIB-SOs, namely CEU-835, -934, and -938, for the determination of their toxicity and half-lives. Moreover, MCF7 tumor-bearing CD1-Foxn1nu Nude female mice were treated with the three prototypical PAIB-SOs for the determination of their antitumor activity. Results: Herein, we show that multi-intravenous administrations of CEU-835, -934, and -938 at their maximal solubilities are well tolerated in healthy female CD1® IGS mice, as depicted by the evaluation of distress behaviors, organ necropsy, total blood cell count, and histology. Moreover, the half-life of CEU-835, -934, and -938 administered intravenously in healthy CD1® IGS female mice were 8.1, 23.2, and 21.5 h, respectively. Finally, their intravenous administrations of CEU-934 and -938 decreased MCF7 tumor growth as efficiently as paclitaxel in MCF7 tumor-bearing CD1-Foxn1nu Nude mouse model. Conclusions: overall, our study demonstrated for the first time that pentyl-bearing PAIB-SOs are new CYP1A1-dependent prodrugs efficiently decrease breast cancer tumor growth, and show no side effects at their pharmacological concentration in mouse models. Full article
Show Figures

Graphical abstract

18 pages, 2199 KB  
Article
The Co-Administration of Paclitaxel with Novel Pyridine and Benzofuran Derivatives that Inhibit Tubulin Polymerisation: A Promising Anticancer Strategy
by Magdalena Perużyńska, Radosław Birger, Patrycja Kłos, Halina Kwiecień, Łukasz Struk, Jacek G. Sośnicki, Laurence Lafanechère and Marek Droździk
Pharmaceutics 2025, 17(2), 223; https://doi.org/10.3390/pharmaceutics17020223 - 9 Feb 2025
Viewed by 1219
Abstract
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses [...] Read more.
Background: Paclitaxel (PTX), a crucial microtubule-stabilising agent in cancer treatment, is limited by its adverse effects and hydrophobic nature, which necessitate the use of toxic solvents. This study proposes a novel approach combining PTX with new microtubule-destabilising compounds at low, safe doses that are ineffective when used individually. Objective: The aim was to evaluate the therapeutic efficacy of combining PTX with previously described pyridine (S1, S22) and benzofuran derivatives (13b, 14), which have demonstrated promising anticancer properties by inhibiting microtubule polymerisation. Methods: The PrestoBlue assay was used to determine the optimal concentrations of each compound, enabling synergistic interactions with a low dose of PTX in HeLa cervical cancer cells. The combined effects of the compounds and PTX on apoptosis, cell cycle distribution, and mitotic spindle formation were then evaluated. Results: The results showed that compounds 13b (1 µM), 14 (0.1 µM), S1 (2 µM), and S22 (2 µM) enhanced the proapoptotic and antimitotic effects of 1 nM PTX, which was ineffective alone. Notably, live-cell imaging revealed that the concurrent use of S1 and PTX produced effects similar to those of a higher PTX concentration (5 nM). Conclusions: These findings suggest that these compounds enhance the anticancer efficacy of low-dose PTX, potentially paving the way for more effective and safer cancer therapies. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Graphical abstract

19 pages, 1839 KB  
Review
The Effects of Podophyllotoxin Derivatives on Noncancerous Diseases: A Systematic Review
by Piotr Strus, Karol Sadowski, Weronika Ploch, Adrianna Jazdzewska, Paulina Oknianska, Oliwia Raniszewska and Izabela Mlynarczuk-Bialy
Int. J. Mol. Sci. 2025, 26(3), 958; https://doi.org/10.3390/ijms26030958 - 23 Jan 2025
Cited by 3 | Viewed by 3003
Abstract
Podophyllotoxin (PPT) is commonly used for genital warts due to its antimitotic properties and relatively good accessibility since it can be extracted from plants in low-economy countries. However, due to relatively high toxicity, it cannot be used in a systematic way (intravenously). Thus, [...] Read more.
Podophyllotoxin (PPT) is commonly used for genital warts due to its antimitotic properties and relatively good accessibility since it can be extracted from plants in low-economy countries. However, due to relatively high toxicity, it cannot be used in a systematic way (intravenously). Thus, there is a need to find or create an equally effective derivative of PPT that will be less toxic. Natural PPT is a suitable and promising scaffold for the synthesis of its derivatives. Many of them have been studied in clinical and preclinical models. In this systematic review, we comprehensively assess the medical applications of PPT derivatives, focusing on their advantages and limitations in non-cancerous diseases. Most of the existing research focuses on their applications in cancerous diseases, leaving non-cancerous uses underexplored. To do that, we systematically reviewed the literature using PubMed, Embase, and Cochrane databases from January 2013 to January 2025. In total, 5333 unique references were identified in the initial search, of which 44 were included in the quantitative synthesis. The assessment of the quality of eligible studies was undertaken using the PRISMA criteria. The risk of bias was assessed using a predefined checklist based on PRISMA guidelines. Each study was independently reviewed by two researchers to evaluate bias in study design, reporting, and outcomes. Our analysis highlights the broad therapeutic potential of PPT derivatives, particularly in antiviral applications, including HPV, Dengue, and SARS-CoV-2 infections. Apart from their well-known anti-genital warts activity, these compounds exhibit significant anti-inflammatory, antimitotic, analgesic, and radioprotective properties. For instance, derivatives such as cyclolignan SAU-22.107 show promise in antiviral therapies, while compounds like G-003M demonstrate radioprotective effects by mitigating radiation-induced damage. To build on this, our review highlights that PPT derivatives, apart from anti-genital warts potential, exhibit four key properties—anti-inflammatory, antimitotic, analgesic, and radioprotective—making them promising candidates not only for treating viral infections such as HPV, Dengue, and SARS-CoV-2 but also for expanding their therapeutic potential beyond cancerous diseases. In conclusion, while PPT derivatives hold great potential across various medical domains, their applications in non-cancerous diseases remain limited by the scarcity of dedicated research. Continued exploration of these compounds is essential to unlock their full therapeutic value. Full article
(This article belongs to the Special Issue Current Research on Cancer Biology and Therapeutics: Third Edition)
Show Figures

Figure 1

54 pages, 6031 KB  
Article
(E)-1-(3-(3-Hydroxy-4-Methoxyphenyl)-1-(3,4,5-Trimethoxyphenyl)allyl)-1H-1,2,4-Triazole and Related Compounds: Their Synthesis and Biological Evaluation as Novel Antimitotic Agents Targeting Breast Cancer
by Gloria Ana, Azizah M. Malebari, Sara Noorani, Darren Fayne, Niamh M. O’Boyle, Daniela M. Zisterer, Elisangela Flavia Pimentel, Denise Coutinho Endringer and Mary J. Meegan
Pharmaceuticals 2025, 18(1), 118; https://doi.org/10.3390/ph18010118 - 17 Jan 2025
Cited by 2 | Viewed by 4210
Abstract
Background/Objectives: The synthesis of (E)-1-(1,3-diphenylallyl)-1H-1,2,4-triazoles and related compounds as anti-mitotic agents with activity in breast cancer was investigated. These compounds were designed as hybrids of the microtubule-targeting chalcones, indanones, and the aromatase inhibitor letrozole. Methods: A panel of [...] Read more.
Background/Objectives: The synthesis of (E)-1-(1,3-diphenylallyl)-1H-1,2,4-triazoles and related compounds as anti-mitotic agents with activity in breast cancer was investigated. These compounds were designed as hybrids of the microtubule-targeting chalcones, indanones, and the aromatase inhibitor letrozole. Methods: A panel of 29 compounds was synthesized and examined by a preliminary screening in estrogen receptor (ER) and progesterone receptor (PR)-positive MCF-7 breast cancer cells together with cell cycle analysis and tubulin polymerization inhibition. Results: (E)-5-(3-(1H-1,2,4-triazol-1-yl)-3-(3,4,5-trimethoxyphenyl)prop-1-en-1-yl)-2-methoxyphenol 22b was identified as a potent antiproliferative compound with an IC50 value of 0.39 mM in MCF-7 breast cancer cells, 0.77 mM in triple-negative MDA-MB-231 breast cancer cells, and 0.37 mM in leukemia HL-60 cells. In addition, compound 22b demonstrated potent activity in the sub-micromolar range against the NCI 60 cancer cell line panel including prostate, melanoma, colon, leukemia, and non-small cell lung cancers. G2/M phase cell cycle arrest and the induction of apoptosis in MCF-7 cells together with inhibition of tubulin polymerization were demonstrated. Immunofluorescence studies confirmed that compound 22b targeted tubulin in MCF-7 cells, while computational docking studies predicted binding conformations for 22b in the colchicine binding site of tubulin. Compound 22b also selectively inhibited aromatase. Conclusions: Based on the results obtained, these novel compounds are suitable candidates for further investigation as antiproliferative microtubule-targeting agents for breast cancer. Full article
Show Figures

Graphical abstract

Back to TopTop