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

Journals

Article Types

Countries / Regions

Search Results (154)

Search Parameters:
Keywords = microtubule-targeting agent

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 801 KB  
Review
Tau-Targeted Therapeutic Strategies: Mechanistic Targets, Clinical Pipelines, and Analysis of Failures
by Xinai Shen, Huan Li, Beiyu Zhang, Yunan Li and Zheying Zhu
Cells 2025, 14(19), 1506; https://doi.org/10.3390/cells14191506 - 26 Sep 2025
Viewed by 2088
Abstract
Tau protein, a neuron-enriched microtubule-associated protein encoded by the MAPT gene, plays pivotal roles in microtubule stabilisation, axonal transport, and synaptic plasticity. Aberrant post-translational modifications (PTMs), hyperphosphorylation, acetylation, ubiquitination, oxidative stress and neuroinflammation disrupt tau’s normal functions, drive its mislocalization, and promote aggregation [...] Read more.
Tau protein, a neuron-enriched microtubule-associated protein encoded by the MAPT gene, plays pivotal roles in microtubule stabilisation, axonal transport, and synaptic plasticity. Aberrant post-translational modifications (PTMs), hyperphosphorylation, acetylation, ubiquitination, oxidative stress and neuroinflammation disrupt tau’s normal functions, drive its mislocalization, and promote aggregation into neurofibrillary tangles, a hallmark of Alzheimer’s disease (AD) and related tauopathies. Over the past two decades, tau-targeted therapies have advanced into clinical development, yet most have failed to demonstrate efficacy in human trials. This review synthesises mechanistic insights into tau biology and pathology, highlighting phosphorylation and acetylation pathways, aggregation-prone motifs, and immune-mediated propagation. We analyse the current therapeutic landscape, including kinase and phosphatase modulators, O-GlcNAcase inhibitors, aggregation blockers, immunotherapies, and microtubule-stabilising agents, while examining representative clinical programs and the reasons underlying their limited success. By combining mechanistic understanding with clinical experience, this review outlines emerging opportunities for rational treatment development, aiming to inform future tau-targeted strategies for AD and other tauopathies. Full article
(This article belongs to the Special Issue Recent Advances in the Study of Tau Protein)
Show Figures

Figure 1

39 pages, 13134 KB  
Article
Mitotic Disruption and Cytoskeletal Alterations Induced by Acorus calamus Essential Oil: Implications for Bioherbicidal Potential
by Mateusz Wróblewski, Natalia Gocek, Aneta Żabka and Justyna T. Polit
Int. J. Mol. Sci. 2025, 26(18), 8933; https://doi.org/10.3390/ijms26188933 - 13 Sep 2025
Viewed by 483
Abstract
Essential oils are increasingly recognized as promising agents for sustainable weed control due to their selectivity and complex modes of action. This study evaluated the effects of Acorus calamus essential oil (SEO) on mitosis in two Fabaceae species (Vicia faba, Lupinus [...] Read more.
Essential oils are increasingly recognized as promising agents for sustainable weed control due to their selectivity and complex modes of action. This study evaluated the effects of Acorus calamus essential oil (SEO) on mitosis in two Fabaceae species (Vicia faba, Lupinus luteus) and two Brassicaceae species (Brassica napus, Arabidopsis thaliana) treated with species-specific IC50 concentrations (0.03%, 0.025%, 0.01%, and 0.005%, respectively). Previous research showed that SEO induces oxidative stress and S-phase delay via genome instability. Here, SEO consistently disrupted mitosis across all species, reducing mitotic index by 50–60%, decreasing Cdc2 (CDKA homolog) levels, and causing chromosomal aberrations, including uneven chromatin condensation, sticky chromosomes, bridges, and micronuclei. Cells accumulated in metaphase and exhibited abnormal karyokinetic and cytokinetic spindles. Immunolabeling revealed thick, tightly packed microtubules and actin filaments, indicating excessive stabilization and impaired reorganization. Epigenetic regulation was also affected: H3T3 phosphorylation was abnormally strong, widely distributed, and persistent into anaphase/telophase, while H3S10Ph intensity was weakened. These results suggest that SEO targets multiple components of mitotic machinery and epigenetic control, regardless of species. The observed selectivity depends on dosage, not mechanism. This multi-targeted action may limit the development of plant resistance, supporting the potential of SEO as a bioherbicide in sustainable agriculture. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

21 pages, 2186 KB  
Review
Knocking on Cells’ Door: Strategic Approaches for miRNA and siRNA in Anticancer Therapy
by Massimo Serra, Alessia Buccellini and Mayra Paolillo
Int. J. Mol. Sci. 2025, 26(17), 8703; https://doi.org/10.3390/ijms26178703 - 6 Sep 2025
Viewed by 1665
Abstract
Metastasis is the main cause of failure in anticancer therapies, and is frequently related to poor prognosis for patients. The true challenge in extending cancer patient life expectancy, eventually managing cancer as a chronic disease with periodic but controllable relapses, relies on the [...] Read more.
Metastasis is the main cause of failure in anticancer therapies, and is frequently related to poor prognosis for patients. The true challenge in extending cancer patient life expectancy, eventually managing cancer as a chronic disease with periodic but controllable relapses, relies on the development of effective therapeutic strategies specifically targeting key mechanisms involved in the metastatic cascade. Traditional chemotherapy with alkylating agents, microtubule inhibitors, and antimetabolites has shown limited efficacy against metastatic cells, largely due to the emergence of chemoresistant populations that undergo epithelial-to-mesenchymal transition (EMT), promoting the colonization of distant organs and sustaining metastatic progression. This scenario has spurred significant efforts to identify small molecules and biologics capable of interfering with specific steps in the metastatic process. In this review, we provide an overview of recent advances involving small interfering RNAs (siRNAs) and microRNAs (miRNAs) in cancer therapy. Although most of these agents are still under investigation and have not yet been approved for clinical use, insights into their development stage offer valuable information to identify new targets in the ongoing fight against metastasis. Particular emphasis is placed on the role of chemical modifications applied to siRNAs, such as backbone, sugar, terminal, base, and conjugation changes, and how these factors influence their stability, immunogenicity, and targeting precision. By integrating these aspects into the discussion, this review provides a focused and up-to-date resource for researchers in medicinal chemistry, drug delivery, and pharmaceutical formulation, where molecular design plays a critical role in therapeutic success. Full article
(This article belongs to the Collection Latest Review Papers in Molecular Oncology)
Show Figures

Graphical abstract

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
Viewed by 1087
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

38 pages, 3745 KB  
Review
Recent Advances in Microtubule Targeting Agents for Cancer Therapy
by Henrique C. Assunção, Patrícia M. A. Silva, Hassan Bousbaa and Honorina Cidade
Molecules 2025, 30(16), 3314; https://doi.org/10.3390/molecules30163314 - 8 Aug 2025
Cited by 1 | Viewed by 2337
Abstract
Cancer mortality and the development of cancer resistance present significant challenges that must be addressed to ensure global health. Among anticancer agents, microtubule-targeting agents (MTAs) represent a well-recognized therapeutic approach that disrupts microtubule dynamics, thereby inhibiting cell division, and has been widely used [...] Read more.
Cancer mortality and the development of cancer resistance present significant challenges that must be addressed to ensure global health. Among anticancer agents, microtubule-targeting agents (MTAs) represent a well-recognized therapeutic approach that disrupts microtubule dynamics, thereby inhibiting cell division, and has been widely used to treat several types of cancers. However, even though MTAs are widely regarded as effective, their potential is limited primarily due to cancer resistance and toxicity. Consequently, in the last years, the exploration of new MTAs with the aim of identifying agents with improved cytotoxicity, selectivity, and adequate pharmacokinetic profile, as well as the ability to evade drug resistance mechanisms, has remained a major concern in the development of anticancer treatment. This review highlights the discovery of new MTAs since 2020, with the goal of understanding the advancements made in this field and its future directions. Special attention is given to structure–activity relationship (SAR) studies that could be important for the discovery of more effective MTAs in the future. Full article
(This article belongs to the Special Issue Nature-Inspired Antitumor Agents, 2nd Edition)
Show Figures

Figure 1

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 2865
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

21 pages, 1133 KB  
Review
Beyond Docetaxel: Targeting Resistance Pathways in Prostate Cancer Treatment
by Tayo Alex Adekiya
BioChem 2025, 5(3), 24; https://doi.org/10.3390/biochem5030024 - 1 Aug 2025
Cited by 1 | Viewed by 1356
Abstract
Prostate cancer continues to be the most common cause of cancer-related disease and mortality among men worldwide, especially in the advanced stages, notably metastatic castration-resistant prostate cancer (mCRPC), which poses significant treatment challenges. Docetaxel, a widely used chemotherapeutic agent, has long served as [...] Read more.
Prostate cancer continues to be the most common cause of cancer-related disease and mortality among men worldwide, especially in the advanced stages, notably metastatic castration-resistant prostate cancer (mCRPC), which poses significant treatment challenges. Docetaxel, a widely used chemotherapeutic agent, has long served as the standard treatment, offering survival benefits and mitigation. However, its clinical impact is frequently undermined by the development of chemoresistance, which is a formidable challenge that leads to treatment failure and disease progression. The mechanisms driving docetaxel resistance are diverse and complex, encompassing modifications in androgen receptor signaling, drug efflux transporters, epithelial-mesenchymal transition (EMT), microtubule alterations, apoptotic pathway deregulation, and tumor microenvironmental influences. Recent evidence suggests that extracellular RNAs influence drug responses, further complicating the resistance landscape. This review offers a broad discussion on the mechanisms of resistance and explores novel therapeutic approaches to address them. These include next-generation taxanes, targeted molecular inhibitors, immunotherapies, and combination regimens that can be designed to counteract specific resistance pathways. By broadening our understanding of docetaxel resistance, this review highlights potential strategies to improve therapeutic efficacy and the potential to enhance outcomes in patients with advanced treatment-resistant prostate cancer. Full article
Show Figures

Figure 1

34 pages, 6455 KB  
Article
IBCar: Potent Orally Bioavailable Methyl N-[5-(3′-Iodobenzoyl)-1H-Benzimidazol-2-yl]Carbamate for Breast Cancer Therapy
by Janina Baranowska-Kortylewicz and Ying Yan
Cancers 2025, 17(15), 2526; https://doi.org/10.3390/cancers17152526 - 30 Jul 2025
Viewed by 788
Abstract
Objectives: To investigate the efficacy and underlying mechanisms of IBCar’s biological activity in breast cancer models, both in cell culture and in mice, and to compare its effects on cancer versus normal cells. Methods: The cytotoxicity of IBCar was evaluated using [...] Read more.
Objectives: To investigate the efficacy and underlying mechanisms of IBCar’s biological activity in breast cancer models, both in cell culture and in mice, and to compare its effects on cancer versus normal cells. Methods: The cytotoxicity of IBCar was evaluated using the MTS assay to assess metabolic activity and the clonogenic assay to determine reproductive integrity. The impact of IBCar on microtubule integrity, mitochondrial function, and multiple signaling pathways was analyzed using Western blotting, microarray analysis, and live cell imaging. The therapeutic effectiveness of orally administered IBCar was assessed in a transgenic mouse model of Luminal B breast cancer and in mice implanted with subcutaneous triple-negative breast cancer xenografts. Results: IBCar demonstrated potent cytotoxicity across a diverse panel of breast cancer cell lines, including those with mutant or wild-type TP53, and cell lines with short and long doubling times. Comparative analysis revealed distinct responses between normal and cancer cells, including differences in IBCar’s effects on the mitochondrial membrane potential, endoplasmic reticulum stress and activation of cell death pathways. In breast cancer cells, IBCar was cytotoxic at nanomolar concentrations, caused irreversible microtubule depolymerization leading to sustained mitochondrial dysfunction, endoplasmic reticulum stress, and induced apoptosis. In normal cells, protective mechanisms included reversible microtubule depolymerization and activation of pro-survival signaling via the caspase-8 and riptosome pathways. The therapeutic potential of IBCar was confirmed in mouse models of Luminal B and triple negative BC, where it exhibited strong antitumor activity without detectable toxicity. Conclusions: These findings collectively support IBCar as a promising, effective, and safe therapeutic candidate for breast cancer treatment. Full article
Show Figures

Figure 1

34 pages, 1871 KB  
Review
Docetaxel Resistance in Breast Cancer: Current Insights and Future Directions
by Fátima Postigo-Corrales, Asunción Beltrán-Videla, Antonio David Lázaro-Sánchez, Ana María Hurtado, Pablo Conesa-Zamora, Ana Belén Arroyo and Ginés Luengo-Gil
Int. J. Mol. Sci. 2025, 26(15), 7119; https://doi.org/10.3390/ijms26157119 - 23 Jul 2025
Cited by 1 | Viewed by 2076
Abstract
Docetaxel is a chemotherapeutic agent widely used for breast cancer treatment; however, its efficacy is often limited by drug resistance and associated toxicity. This review examines the molecular mechanisms of docetaxel resistance in breast cancer and discusses research advances and future directions for [...] Read more.
Docetaxel is a chemotherapeutic agent widely used for breast cancer treatment; however, its efficacy is often limited by drug resistance and associated toxicity. This review examines the molecular mechanisms of docetaxel resistance in breast cancer and discusses research advances and future directions for overcoming this challenge. Key resistance mechanisms include alterations in drug targets (microtubules), increased drug efflux, suppression of apoptosis, activation of survival signalling pathways, epithelial-to-mesenchymal transition (EMT), and cancer stem cell enrichment. An evolutionary perspective distinguishes between intrinsic and acquired resistance, emphasising the need for adaptive therapeutic strategies. Recent advances in genomic profiling, non-coding RNA research, novel drug combinations, and biomarker-guided therapies have also been reviewed. Emerging approaches, such as targeting the tumour microenvironment, harnessing immunotherapy, and implementing adaptive dosing schedules, have been discussed. This review emphasises the understanding of resistance as a multifactorial phenomenon that requires multipronged interventions. Research has aimed to identify predictive biomarkers, develop targeted agents to reverse resistance, and design rational combination strategies to improve patient outcomes. Progress in deciphering and targeting docetaxel resistance mechanisms holds promise for enhancing treatment responses and extending survival in patients with breast cancer. Full article
(This article belongs to the Special Issue Molecular Research and Cellular Biology of Breast Cancer)
Show Figures

Figure 1

15 pages, 2357 KB  
Article
Development of a Novel, Highly Sensitive System for Evaluating Ebola Virus Particle Formation
by Wakako Furuyama, Miako Sakaguchi, Hanako Ariyoshi and Asuka Nanbo
Viruses 2025, 17(7), 1016; https://doi.org/10.3390/v17071016 - 19 Jul 2025
Viewed by 960
Abstract
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently [...] Read more.
Ebola virus (EBOV) causes severe hemorrhagic fevers in humans, and effective countermeasures remain limited. The EBOV-encoded major matrix protein VP40 is essential for viral assembly, budding, and particle release, making it a promising target for antiviral drug development. However, no approved drugs currently target the viral particle formation process. In this study, we established a simple and highly sensitive screening system to evaluate VP40-mediated virus-like particle (VLP) formation under biosafety level −2 conditions. The system uses the HiBiT luminescence-based reporter fused to VP40, allowing for the detection of VP40 release. Our results demonstrate that the HiBiT sequence fused at the N-terminus [HiBiT-VP40 (N)] retains VP40′s ability to form VLPs, supporting its use as a functional reporter. Furthermore, we validated the system by assessing the role of Rab11-dependent trafficking in VP40-mediated budding and by evaluating the effect of nocodazole, a microtubule depolymerizer, on VLP release. This novel screening system provides a convenient and reliable platform for screening potential inhibitors targeting the late stages of EBOV infection, including viral particle formation and release. Additionally, its potential adaptability to other filoviruses suggests wide applicability in the discovery and development of additional novel therapeutic agents. Full article
Show Figures

Figure 1

17 pages, 1513 KB  
Review
Rational Modulation of Liquid–Liquid Phase Separation Offers Novel Ways to Combat Tauopathies
by Xingxing Zhang, Lumiao Wang, Nixin Lin, Meng Gao and Yongqi Huang
Int. J. Mol. Sci. 2025, 26(14), 6709; https://doi.org/10.3390/ijms26146709 - 12 Jul 2025
Cited by 1 | Viewed by 1448
Abstract
The microtubule-associated protein tau plays an essential role in regulating the dynamic assembly of microtubules and is implicated in axonal elongation and maturation, axonal transport, synaptic plasticity regulation, and genetic stability maintenance. Nevertheless, the assembly of tau into neurofibrillary tangles in neurons is [...] Read more.
The microtubule-associated protein tau plays an essential role in regulating the dynamic assembly of microtubules and is implicated in axonal elongation and maturation, axonal transport, synaptic plasticity regulation, and genetic stability maintenance. Nevertheless, the assembly of tau into neurofibrillary tangles in neurons is a pathological hallmark of a group of neurodegenerative diseases known as tauopathies. Despite enormous efforts and rapid advancements in the field, effective treatment remains lacking for these diseases. In this review, we provide an overview of the structure and phase transition of tau protein. In particular, we focus on the involvement of liquid–liquid phase separation in the biology and pathology of tau. We then discuss several potential strategies for combating tauopathies in the context of phase separation: (i) modulating the formation of tau condensates, (ii) delaying the liquid-to-solid transition of tau condensates, (iii) reducing the enrichment of aggregation-prone species into tau condensates, and (iv) suppressing abnormal post-translational modifications on tau inside condensates. Deciphering the structure–activity relationship of tau phase transition modulators and uncovering the conformational changes in tau during phase transitions will aid in developing therapeutic agents targeting tau in the context of phase separation. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

28 pages, 831 KB  
Review
Cyanobacterial Peptides in Anticancer Therapy: A Comprehensive Review of Mechanisms, Clinical Advances, and Biotechnological Innovation
by Heayyean Lee, Khuld Nihan and Yale Ryan Kwon
Mar. Drugs 2025, 23(6), 233; https://doi.org/10.3390/md23060233 - 29 May 2025
Cited by 1 | Viewed by 1810
Abstract
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic [...] Read more.
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic payloads in antibody–drug conjugates (ADCs). However, challenges such as limited tumor selectivity, systemic toxicity, and production scalability remain barriers to broader application. Recent advances in targeted delivery technologies, combination therapy strategies, synthetic biology, and genome mining offer promising solutions. Emerging data from preclinical and clinical studies highlight their therapeutic potential, particularly in treatment-resistant cancers. In this review, we (i) summarize key cyanobacterial peptides and their molecular mechanisms of action, (ii) examine progress toward clinical translation, and (iii) explore biotechnological approaches enabling sustainable production and structural diversification. We also discuss future directions for enhancing specificity and the therapeutic index to fully exploit the potential of these marine-derived peptides in oncology. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Figure 1

29 pages, 2914 KB  
Review
Interplay Between the Cytoskeleton and DNA Damage Response in Cancer Progression
by Clarissa Esmeralda Halim, Shuo Deng, Karen Carmelina Crasta and Celestial T. Yap
Cancers 2025, 17(8), 1378; https://doi.org/10.3390/cancers17081378 - 21 Apr 2025
Viewed by 1545
Abstract
DNA damage has emerged as a critical factor in fuelling the development and progression of cancer. DNA damage response (DDR) pathways lie at the crux of cell fate decisions following DNA damage induction, which can either trigger the repair of detrimental DNA lesions [...] Read more.
DNA damage has emerged as a critical factor in fuelling the development and progression of cancer. DNA damage response (DDR) pathways lie at the crux of cell fate decisions following DNA damage induction, which can either trigger the repair of detrimental DNA lesions to protect cancer cells or induce the cell death machinery to eliminate damaged cells. Cytoskeletal dynamics have a critical role to play and influence the proper function of DDR pathways. Microfilaments, intermediate filaments, microtubules, and their associated proteins are well involved in the DDR. For instance, they are not only implicated in the recruitment of specific DDR molecules to the sites of DNA damage but also in the regulation of the mobility of the damaged DNA to repair sites in the periphery of the nucleus. The exquisite roles that these cytoskeletal proteins play in different DDR pathways, such as non-homologous end joining (NHEJ), homologous recombination (HR), base excision repair (BER), and nucleotide excision repair (NER), in cancer cells are extensively discussed in this review. Many cancer treatments are reliant upon inducing DNA damage in cancer cells to eliminate them; thus, it is important to shed light on factors that could affect their efficacy. Although the cytoskeleton is intricately involved in the DDR process, this has often been overlooked in cancer research and has not been exploited in developing DDR-targeting cancer therapy. Understanding the interplay between the cytoskeleton and the DDR in cancer will then provide insights into improving the development of cancer therapies that can leverage the synergistic action of DDR inhibitors and cytoskeleton-targeting agents. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

28 pages, 15072 KB  
Article
Unravelling Paclitaxel Resistance in Gastric Cancer: The Role of Small Extracellular Vesicles in Epithelial Mesenchymal Transition and Extracellular Matrix Remodelling
by Giorgia Panzetta, Annalisa Schirizzi, Francesco Balestra, Maria De Luca, Nicoletta Depalo, Federica Rizzi, Angela Dalia Ricci, Giampiero De Leonardis, Claudio Lotesoriere, Gianluigi Giannelli, Rosalba D’Alessandro and Maria Principia Scavo
Cancers 2025, 17(8), 1360; https://doi.org/10.3390/cancers17081360 - 18 Apr 2025
Viewed by 1148
Abstract
Background: Gastric cancer (GC) is a highly aggressive disease often complicated by resistance to chemotherapy agents like paclitaxel (PTX), which targets microtubules to induce apoptosis. Resistance arises through complex molecular mechanisms, including the overexpression of pro-angiogenic factors (VEGFA, ANG-2), activation of survival pathways [...] Read more.
Background: Gastric cancer (GC) is a highly aggressive disease often complicated by resistance to chemotherapy agents like paclitaxel (PTX), which targets microtubules to induce apoptosis. Resistance arises through complex molecular mechanisms, including the overexpression of pro-angiogenic factors (VEGFA, ANG-2), activation of survival pathways (PDGFRβ, PPARγ), and epithelial-mesenchymal transition (EMT) driven by proteins such as VIM, E-CAD, N-CAD, and FLOT-1. The extracellular matrix (ECM), regulated by COL1A1 and influenced by PPARγ, acts as a physical barrier to drug penetration. Small extracellular vesicles (sEVs) have emerged as critical mediators of intercellular communication and may influence these resistance pathways. Methods: This study investigated the role of sEVs isolated from metastatic GC patients treated with Ramucirumab and PTX. Patients were stratified by progression-free survival (PFS) into rapidly progressing (RP) and controlled disease (CD) groups. sEVs from these patients were applied to HCEC-1CT and HEPA-RG cell lines. Cell viability assays, gene and protein expression analyses, and bioinformatic studies were conducted to assess the impact of sEVs on resistance-related markers. Results: Results showed that sEVs from CD patients reduced the expression of markers associated with drug resistance, while sEVs from RP patients increased these markers, promoting angiogenesis, EMT, and ECM remodeling. These changes correlated with enhanced cell survival and resistance phenotypes. Bioinformatic analyses confirmed that sEVs modulate inflammation, ECM dynamics, and EMT pathways. Conclusions: In conclusion, sEVs from metastatic GC patients significantly influence chemoresistance and tumor progression. Targeting sEV-mediated signaling may offer novel therapeutic strategies to overcome resistance and improve treatment outcomes in gastric cancer. Full article
(This article belongs to the Special Issue Extracellular Matrix Proteins in Cancer)
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 4 | Viewed by 4782
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