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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (184)

Search Parameters:
Keywords = anticancer prodrug

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
36 pages, 3847 KB  
Review
Lysosome as a Chemical Reactor
by Mahendiran Dharmasivam and Busra Kaya
Int. J. Mol. Sci. 2025, 26(23), 11581; https://doi.org/10.3390/ijms262311581 - 29 Nov 2025
Viewed by 1167
Abstract
The lysosome is no longer viewed as a simple degradative “trash can” of the cell. The lysosome is not only degradative; its acidic, redox-active lumen also serves as a chemical “microreactor” that can modulate anticancer drug disposition and activation. This review examines how [...] Read more.
The lysosome is no longer viewed as a simple degradative “trash can” of the cell. The lysosome is not only degradative; its acidic, redox-active lumen also serves as a chemical “microreactor” that can modulate anticancer drug disposition and activation. This review examines how the distinctive chemical features of the lysosome, including its acidic pH (~4.5–5), strong redox gradients, limited thiol-reducing capacity, generation of reactive oxygen (ROS), diverse acid hydrolases, and reservoirs of metal ions, converge to influence the fate and activity of anticancer drugs. The acidic lumen promotes sequestration of weak-base drugs, which can reduce efficacy by trapping agents within a protective “safe house,” yet can also be harnessed for pH-responsive drug release. Lysosomal redox chemistry, driven by intralysosomal iron and copper, catalyzes Fenton-type ROS generation that contributes to oxidative damage and ferroptosis. The lysosome’s broad enzyme repertoire enables selective prodrug activation, such as through protease-cleavable linkers in antibody–drug conjugates, while its membrane transporters, particularly P-glycoprotein (Pgp), can sequester chemotherapies and promote multidrug resistance. Emerging therapeutic strategies exploit these processes by designing lysosomotropic drug conjugates, pH- and redox-sensitive delivery systems, and combinations that trigger lysosomal membrane permeabilization (LMP) to release trapped drugs. Acridine–thiosemicarbazone hybrids exemplify this approach by combining lysosomal accumulation with metal-based redox activity to overcome Pgp-mediated resistance. Advances in chemical biology, including fluorescent probes for pH, redox state, metals, and enzymes, are providing new insights into lysosomal function. Reframing the lysosome as a chemical reactor rather than a passive recycling compartment opens new opportunities to manipulate subcellular pharmacokinetics, improve drug targeting, and overcome therapeutic resistance in cancer. Overall, this review translates the chemical principles of the lysosome into design rules for next-generation, more selective anticancer strategies. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Graphical abstract

16 pages, 3941 KB  
Article
Bis-Oxadiazole Assemblies as NO-Releasing Anticancer Agents
by Egor M. Matnurov, Irina A. Stebletsova, Alexander A. Larin, Jemma Arakelyan, Ivan V. Ananyev, Artem L. Gushchin, Leonid L. Fershtat and Maria V. Babak
Pharmaceutics 2025, 17(11), 1494; https://doi.org/10.3390/pharmaceutics17111494 - 19 Nov 2025
Viewed by 924
Abstract
Background: Malignant pleural mesothelioma (MPM) is an aggressive, asbestos-associated cancer characterized by dysregulated nitric oxide (NO) signaling and increased NO levels that facilitate tumor progression. Paradoxically, this aberrant NO environment creates a therapeutic vulnerability that can be exploited by NO-donor prodrugs, which [...] Read more.
Background: Malignant pleural mesothelioma (MPM) is an aggressive, asbestos-associated cancer characterized by dysregulated nitric oxide (NO) signaling and increased NO levels that facilitate tumor progression. Paradoxically, this aberrant NO environment creates a therapeutic vulnerability that can be exploited by NO-donor prodrugs, which overwhelm cellular defenses with cytotoxic concentrations of NO, inducing nitrosative stress and apoptosis. Within this framework, oxadiazole-based scaffolds have emerged as a promising platform for prodrug development owing to their versatile chemistry and potential as novel NO donors or synergistic agents. In our previous studies, we developed several series of hybrid architectures incorporating 1,2,5-oxadiazole 2-oxide (furoxan) and 1,2,4-oxadiazole scaffolds, producing compounds with diverse and tunable NO-donor activities. We further observed that the cytotoxicity of these hybrids was significantly influenced by the substituents introduced at position 3 of the furoxan ring. Methods: We designed and synthesized a series of bis(1,2,4-oxadiazolyl)furoxans to systematically investigate their NO-donating capacity, cytotoxicity against MPM cell lines, selectivity over healthy lung fibroblasts, and underlying anticancer mechanisms. Results: The bis(1,2,4-oxadiazolyl)furoxans exhibited lower overall cytotoxicity but significantly higher selectivity compared with previously studied 3-cyano-4-(1,2,4-oxadiazolyl)furoxans. Their NO-releasing properties showed a strong correlation with their ability to induce mitochondrial damage, as evidenced by membrane depolarization. Moreover, the incorporation of specific substituents, such as a furan ring, on the 1,2,4-oxadiazole moiety introduced an additional mechanism of action through the induction of reactive oxygen species. Conclusions: Analysis of cancer cell death confirmed that these compounds acted through a multimodal mechanism dependent on both NO release and the specific substituents on the 1,2,4-oxadiazole moiety. Full article
(This article belongs to the Special Issue Prodrug Applications for Targeted Cancer Therapy)
Show Figures

Graphical abstract

41 pages, 7586 KB  
Review
Unlocking the Therapeutic Potential of Ellagitannins: A Comprehensive Review of Key Representatives
by Rositsa Mihaylova, Viktoria Elincheva, Georgi Momekov and Rumyana Simeonova
Molecules 2025, 30(22), 4328; https://doi.org/10.3390/molecules30224328 - 7 Nov 2025
Viewed by 1767
Abstract
The present review offers a comprehensive synthesis of the structural diversity, natural occurrence, and therapeutic promise of key ellagitannins (punicalagin, sanguiin H-6, corilagin, geraniin, oenothein B, chebulagic, and chebulinic acids) within the hydrolyzable ellagitannin pool. Distributed in medicinal and dietary plants long used [...] Read more.
The present review offers a comprehensive synthesis of the structural diversity, natural occurrence, and therapeutic promise of key ellagitannins (punicalagin, sanguiin H-6, corilagin, geraniin, oenothein B, chebulagic, and chebulinic acids) within the hydrolyzable ellagitannin pool. Distributed in medicinal and dietary plants long used in traditional medicine, ellagitannin-rich species serve as sources of both complex polyphenolic scaffolds and their bioactive metabolites, urolithins, which mediate many of their health-promoting effects. Special emphasis is placed on the multifaceted mechanisms that contribute to their potent antioxidant, anti-inflammatory, antimicrobial, and anticancer effects, extending to both non-communicable and communicable diseases. Despite their broad therapeutic spectrum, clinical translation is limited by challenges such as poor bioavailability, host-gut microbiota variability, and a lack of robust in vivo evidence. The review highlights future directions aimed at unlocking ellagitannins’ potential, including microbiota-targeted strategies for urolithin production, the design of stable prodrugs and analogs, and innovative delivery platforms. By integrating phytochemical, mechanistic and translational insights, this article positions ellagitannins as promising candidates for the development of novel polyphenol-based interventions. Full article
Show Figures

Graphical abstract

24 pages, 2998 KB  
Article
Drug-Integrating Amphiphilic Nano-Assemblies: 3. PEG-PPS/Palmitate Nanomicelles for Sustained and Localized Delivery of Dexamethasone in Cell and Tissue Transplantations
by Giulio Palummieri, Saeida Saadat, Sung-Ting Chuang, Peter Buchwald and Diana Velluto
Pharmaceutics 2025, 17(10), 1337; https://doi.org/10.3390/pharmaceutics17101337 - 16 Oct 2025
Viewed by 1178
Abstract
Background: Glucocorticoids are an important class of therapeutics used in a variety of applications, including allotransplantations. Dexamethasone (Dexa) is well-known for its strong anti-inflammatory, immunosuppressive, and anticancer properties. However, its clinical use is often limited by its poor water solubility, poor pharmacokinetics, and [...] Read more.
Background: Glucocorticoids are an important class of therapeutics used in a variety of applications, including allotransplantations. Dexamethasone (Dexa) is well-known for its strong anti-inflammatory, immunosuppressive, and anticancer properties. However, its clinical use is often limited by its poor water solubility, poor pharmacokinetics, and high likelihood of systemic side effects. Methods: To address the issues, we tested a combined strategy where our original Drug-Integrating Amphiphilic Nano-Assemblies (DIANAs), a class of self-assembling polymeric nanoparticles designed for controlled drug release, were used to solubilize and deliver dexamethasone palmitate (DexP), a hydrophobic prodrug of dexamethasone. Results: The palmitate chains of the prodrug can form strong van der Waals interactions with the hydrophobic moieties of the PEG-PPS block copolymer used here. In water, this resulted in the self-assembling of stable dexamethasone palmitate–PEG–PPS nanomicelles, termed DexP-nMICs, with a 25 nm average diameter that slowly released Dexa over more than two weeks. Conclusions: Here we demonstrated that DexP-nMICs can carry elevated amounts of Dexa—increasing its solubility in water—prolong circulation in its pharmacologically active form in vivo and provide passive targeting to inflammation sites. The anti-inflammatory efficacy of DexP-nMICs was first confirmed in vitro on stimulated macrophages, demonstrating a significant reduction in cytokine secretion. An allogeneic mouse skin transplant model, used to assess the therapeutic potential of DexP-nMICs in vivo, confirmed its ability to provide graft-targeted delivery and prolong graft survival as compared to the unformulated parent drug. Therefore, DexP-nMICs are a promising candidate for sustained and localized use of anti-inflammatory drugs in cell and tissue transplantations. Full article
(This article belongs to the Special Issue Applications of Nanomaterials in Immunotherapies)
Show Figures

Figure 1

25 pages, 4125 KB  
Article
Enhanced Killing of Colon Cancer Cells by Mesoporous Silica Nanoparticles Loaded with Ellagic Acid
by Khaled AbouAitah, Amr Nassrallah, Ahmed A. F. Soliman, Anna Swiderska-Sroda, Tadeusz Chudoba, Julita Smalc-Koziorowska, Beom Soo Kim and Witold Łojkowski
Nanomaterials 2025, 15(20), 1547; https://doi.org/10.3390/nano15201547 - 10 Oct 2025
Cited by 1 | Viewed by 828
Abstract
Background: Natural compounds, including ellagic acid (ELG), are promising anticancer agents with low adverse effects. In this paper, we test in vitro the effectiveness of mesoporous silica nanoparticles (MSN) as an ELG carrier against colon cancer. Methods: We produced MSNs functionalized with triptycene [...] Read more.
Background: Natural compounds, including ellagic acid (ELG), are promising anticancer agents with low adverse effects. In this paper, we test in vitro the effectiveness of mesoporous silica nanoparticles (MSN) as an ELG carrier against colon cancer. Methods: We produced MSNs functionalized with triptycene (TRP) and loaded with ELG, further called MSNTRPELG nanoformulation. The nanoformulation contained over 11 wt.% TRP and approximately 25 wt.% ELG in the mesoporous structure and on the surface of particles. It was assessed for anticancer effects against two colon cancer cells: HCT-116 and HT-29 for treatment with up to 200 µM. Results: Comparing to free ELG, we have shown a three times higher cancer inhibition. The lowest IC50 values were for HCT-116 (88.1 ± 0.1 µM) and HT-29 (77.6 ± 0.1 µM). When treated with free ELG, the values were 187.1 ± 0.1 µM and 300.0 ± 0.1 µM, respectively. MSNTRPELG enhanced apoptosis primarily by activating caspase-3, p53, and Bax while downregulating Bcl-2 in HCT-116 and HT-29 cells. It also inhibited receptor tyrosine kinases (HER2 and VEGFR2). Preliminary Western blot observations suggest suppression of B-RAF, C-RAF, and K-RAS oncogenes, with stronger inhibition by the nanoformulation than by free ELG. Conclusions: This work highlights the potential of MSNs to enhance the efficacy of natural prodrugs, particularly ELG, in cancer therapy. Full article
(This article belongs to the Special Issue Metal Nanostructures in Biological Applications)
Show Figures

Graphical abstract

13 pages, 2820 KB  
Communication
NIR Responsive Polymeric Prodrug Micelles ZnPc@P(PEG-CMA-TKGEM) for Combating Gemcitabine Drug Delivery in Anticancer Chemotherapy
by Heng Zhang, Yiping Yang, Shengchao Yang, Yuchang Qin, Xuan Lv, Lin Cui, Wei Jia and Zhiyong Liu
Materials 2025, 18(17), 4165; https://doi.org/10.3390/ma18174165 - 5 Sep 2025
Viewed by 9509
Abstract
The impact of encapsulating gemcitabine (GEM) into nanoparticles on its delivery remains underexplored, with the potential benefits of targeted drug delivery and stimuli-responsive release yet to be fully clarified. Herein, we designed a near-infrared (NIR) light-responsive polymeric nanoparticle, ZnPc@P(PEG-CMA-TKGEM), which integrates reactive oxygen [...] Read more.
The impact of encapsulating gemcitabine (GEM) into nanoparticles on its delivery remains underexplored, with the potential benefits of targeted drug delivery and stimuli-responsive release yet to be fully clarified. Herein, we designed a near-infrared (NIR) light-responsive polymeric nanoparticle, ZnPc@P(PEG-CMA-TKGEM), which integrates reactive oxygen species (ROS) generation and cell imaging capabilities. The self-assembled polymeric micelles exhibit a hydrodynamic size of ~134 nm. Under NIR irradiation, the cumulative drug release rate reaches 51% within 48 h, which is three times higher than that of the non-irradiated control group. In cytotoxicity assays, the cell viability of the NIR-irradiated drug-loaded group is approximately 17%, while that of the NIR-irradiated blank group (without drug loading) remains above 80%. These results confirm that the nanocarriers successfully deliver GEM to target cells and achieve controlled drug release via NIR stimulation. Full article
(This article belongs to the Section Biomaterials)
Show Figures

Figure 1

2 pages, 143 KB  
Correction
Correction: Anderson, R.F.; Qi, W. Coenzyme Q10 as an Inhibitor of Effector Release from One-Electron-Reduced Bioreductive Anticancer Prodrugs. Molecules 2025, 30, 760
by Robert F. Anderson and Wen Qi
Molecules 2025, 30(17), 3515; https://doi.org/10.3390/molecules30173515 - 28 Aug 2025
Viewed by 545
Abstract
In the original publication [...] Full article
25 pages, 3526 KB  
Article
Valine–Niclosamide for Treatment of Androgen Receptor Splice Variant-Positive Hepatocellular Carcinoma
by Emma J. Hoelzen, Hanna S. Radomska, Samuel K. Kulp, Adeoluwa A. Adeluola, Lauren A. Granchie, Jeffrey Cheng, Anees M. Dauki, Moray J. Campbell, Shabber Mohammed, Enming Xing, Min Hai, Mayu Fukuda, Xiaolin Cheng, Mitch A. Phelps, Pui-Kai Li and Christopher C. Coss
Cancers 2025, 17(15), 2535; https://doi.org/10.3390/cancers17152535 - 31 Jul 2025
Cited by 1 | Viewed by 1426
Abstract
Background/Objectives: Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and currently is the second-leading cause of cancer-related mortality globally. Current front-line systemic therapies for advanced HCC offer only modest improvements in patient overall survival. HCC is a sexually dimorphic disease, and [...] Read more.
Background/Objectives: Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and currently is the second-leading cause of cancer-related mortality globally. Current front-line systemic therapies for advanced HCC offer only modest improvements in patient overall survival. HCC is a sexually dimorphic disease, and cancer progression is driven in part by AR activity. Here, we present novel niclosamide pro-drugs for use in advanced HCC based upon niclosamide’s known anti-AR activity and additional anti-cancer pathway efficacy. Methods: Niclosamide analogs were evaluated for their impacts on the AR protein in two HCC cell lines with different AR phenotypes. Amino acid conjugates of niclosamide were developed, and pharmacokinetic (PK) analyses were conducted to determine improvements in clearance and oral exposure. Finally, niclosamide analogs and amino acid conjugates were evaluated in an in vivo model of HCC. Results: Niclosamide analogs maintained anti-AR properties in HCC. Valine-conjugated niclosamide showed improved oral exposure, positioning it as a potential therapeutic in advanced HCC. Conclusions: Valine–niclosamide improves upon niclosamide’s poor solubility and oral bioavailability, increasing its utility for a variety of therapeutic uses. Further study of valine–niclosamide in advanced HCC and in other cancers or diseases is warranted. Full article
(This article belongs to the Special Issue Drug Repurposing and Reformulation for Cancer Treatment: 2nd Edition)
Show Figures

Figure 1

11 pages, 2454 KB  
Communication
Effect of a Novel Antidepressant and Anticancer Nuc01 on Depression in Cancer Survivors
by Changchun Yuan, Xudong Shi, Zhiqiang Wang, Yuqiang Li, Wenbing Ma and Kai Fu
Curr. Issues Mol. Biol. 2025, 47(8), 587; https://doi.org/10.3390/cimb47080587 - 24 Jul 2025
Viewed by 1311
Abstract
Depression in cancer survivors is commonly treated with serotonin and norepinephrine reuptake inhibitors (SNRIs), such as venlafaxine. These drugs alleviate depressive symptoms by inhibiting the reuptake of serotonin and norepinephrine. However, a novel approach has emerged with the development of trans-2-phenylcyclopropylamine (PCPA)–drug [...] Read more.
Depression in cancer survivors is commonly treated with serotonin and norepinephrine reuptake inhibitors (SNRIs), such as venlafaxine. These drugs alleviate depressive symptoms by inhibiting the reuptake of serotonin and norepinephrine. However, a novel approach has emerged with the development of trans-2-phenylcyclopropylamine (PCPA)–drug conjugates that inhibit lysine-specific demethylase 1 (LSD1), which is a biomarker and molecular target for cancer therapy. LSD1 inhibition can effectively suppress cancer cell proliferation. Nuc01 is a novel PCPA–drug conjugate designed as a prodrug of venlafaxine. In vivo studies showed that Nuc01 dose-dependently reduced immobility time in the tail suspension test in mice, outperforming desmethylvenlafaxine. This suggests that Nuc01 may act as a potent triple reuptake inhibitor, potentially offering enhanced efficacy in the treatment of depression. Additionally, in vitro studies demonstrated that Nuc01 effectively occupies the PCPA binding site within LSD1 (IC50 = 530 nm) and inhibits the proliferation of MDA-MB-231 cancer cells (IC50 = 1130 nm). These findings suggest that Nuc01 may function as an LSD1 inhibitor with potential anticancer properties. Collectively, the data indicate that Nuc01 appears to exhibit dual functional characteristics: acting as a triple reuptake inhibitor potentially applicable for depression treatment and as an LSD1 inhibitor demonstrating anticancer potential. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

29 pages, 3081 KB  
Review
“Non-Classical” Platinum Complexes: A Concise Review
by Adriana Bakalova, Nina Ruseva and Emiliya Cherneva
Int. J. Mol. Sci. 2025, 26(13), 6270; https://doi.org/10.3390/ijms26136270 - 28 Jun 2025
Cited by 1 | Viewed by 2294
Abstract
The utilization of platinum complexes in medicine continues to be a prevalent treatment modality for diverse tumour types. However, it should be noted that certain platinum complexes are characterized by a high degree of toxicity. In recent years, there has been a focus [...] Read more.
The utilization of platinum complexes in medicine continues to be a prevalent treatment modality for diverse tumour types. However, it should be noted that certain platinum complexes are characterized by a high degree of toxicity. In recent years, there has been a focus among scientists on synthesizing “non-classic” platinum complexes, such as those with a trans-configuration, Pt(IV) complexes, and mixed ammine/amine platinum complexes, with the aim of reducing the toxic side effects of certain platinum complexes, including cisplatin. For instance, newly synthesized platinum complexes with a trans-configuration exhibited substantial cytotoxic activity which was comparable to that of the corresponding cis-isomers and cisplatin. This finding challenged the prevailing cis-geometry paradigm and prompted a re-evaluation of the structural activity relationships (SARs) of antitumour platinum complexes. It is widely accepted that Pt(IV) complexes act as prodrugs and release the active Pt(II) species. This property renders them promising candidates as anticancer drugs. Furthermore, it has been established that mixed ammine/amine platinum complexes are less toxic than cisplatin. In addition, compared to cisplatin, they have been observed to have equivalent or greater cytotoxic activity. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

9 pages, 757 KB  
Communication
Sugar-Linked Diethyldithiocarbamate Derivatives: A Novel Class of Anticancer Agents
by Mohammad Najlah, Niamh McCallum, Ana Maria Pereira, Dan Alves, Niusha Ansari-Fard, Sahrish Rehmani and Ayşe Kaya
Int. J. Mol. Sci. 2025, 26(12), 5589; https://doi.org/10.3390/ijms26125589 - 11 Jun 2025
Cited by 1 | Viewed by 1021
Abstract
Disulfiram (DSF), a well-known anti-alcoholism drug, exhibits potent anticancer activity via its metabolite, diethyldithiocarbamate (DDC), which forms a cytotoxic copper complex that selectively targets cancer stem cells. However, its clinical utility is limited by poor solubility and rapid plasma metabolism. This study explores [...] Read more.
Disulfiram (DSF), a well-known anti-alcoholism drug, exhibits potent anticancer activity via its metabolite, diethyldithiocarbamate (DDC), which forms a cytotoxic copper complex that selectively targets cancer stem cells. However, its clinical utility is limited by poor solubility and rapid plasma metabolism. This study explores saccharide-linked DDCs as novel prodrugs designed to enhance stability, solubility, and tumour-selective activation. These compounds feature thioglycosidic bonds that shield the DDC moiety from premature degradation while retaining its metal-chelating function to form the active copper(II)bis(N,N-diethyldithiocarbamate) (Cu(DDC)2) complex. The synthesised derivatives were characterised and evaluated for serum stability and in vitro cytotoxicity across several cancer cell lines, including colorectal, breast, lung, and brain cancers. Copper-complexed saccharide-DDC prodrugs demonstrated remarkable cytotoxicity, with improved biostability and solubility profiles. These findings highlight the potential of saccharide-linked DDCs as stable, copper-activated prodrugs for cancer therapy. Further in vivo studies are warranted to validate their pharmacokinetics and clinical relevance. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

15 pages, 1915 KB  
Communication
Performance of Imidazoquinoline Glycoconjugate BAIT628 as a TLR7 Agonist Prodrug for Prostate Cancer
by Seyedeh A. Najibi, S. M. Al Muied Pranto, Muhammad Haroon, Amy E. Nielsen and Rock J. Mancini
Pharmaceuticals 2025, 18(6), 804; https://doi.org/10.3390/ph18060804 - 27 May 2025
Cited by 1 | Viewed by 2036
Abstract
Despite broad anti-cancer efficacy as Toll-Like Receptor (TLR) 7/8 agonists, imidazoquinolines remain limited in use via systemic administration or in situ vaccination therapies due to inflammatory toxicity. One approach to address this challenge involves better targeting the action of imidazoquinolines by caging them [...] Read more.
Despite broad anti-cancer efficacy as Toll-Like Receptor (TLR) 7/8 agonists, imidazoquinolines remain limited in use via systemic administration or in situ vaccination therapies due to inflammatory toxicity. One approach to address this challenge involves better targeting the action of imidazoquinolines by caging them as glycoconjugate prodrugs. Within cancer cells, imidazoquinoline glycoconjugates are activated by hydrolases prior to efflux by ABC transport proteins, where they then elicit tumoricidal effects from the assistance of bystander immune cells, such as tumor-infiltrating lymphocytes and associated macrophages, in local proximity. While this concept of Bystander-Assisted ImmunoTherapy (BAIT) has been established at a molecular level in vitro, tolerability or efficacy of BAIT has not been reported in vivo. Here, we evaluate the MTD and tumor growth delay efficacy of a lead BAIT prodrug (BAIT628) in a male C57BL/6 mouse TRAMP-C2 prostate cancer model to further establish this methodology. Overall, we find that systemic BAIT628 is well tolerated at over 5-fold the dose-limiting inflammatory toxicity of the parent imidazoquinoline (up to 5 mg/mouse/day I.P. for 10 days). Analyzing serum cytokines reveals that IL-10 production, elicited by the mannoside caging group, likely contributes to the enhanced MTD. Using BAIT628 as an in situ vaccination immunotherapy (seven times over 3 weeks) resulted in significant tumor growth delay and increased survival, both alone and in combination with a murinized α-PD-L1 checkpoint blockade. The tumor histology of tumor-infiltrating immune cell subsets (CD4+, CD8+, CD11c+) reveals significant increases in CD11c+ populations, consistent with TLR7/8 agonism. Overall, BAIT628 is well tolerated and exhibits significant efficacy in the TRAMP-C2 model. These results demonstrate how the BAIT approach can optimize imidazoquinolines for in vivo tolerability and subsequent efficacy as cancer immunotherapeutics. Full article
Show Figures

Figure 1

21 pages, 9638 KB  
Article
Development of GSH-Stimuli-Responsive Micelles Using a Targeted Paclitaxel Prodrug for Enhanced Anticancer Effect
by Qian Ning, Guangping Yu, Wenkai Yi, Minhui Gu, Qianqian Xu, Zhiting Ye, Mengxia Zhang and Shengsong Tang
Pharmaceutics 2025, 17(4), 538; https://doi.org/10.3390/pharmaceutics17040538 - 21 Apr 2025
Cited by 1 | Viewed by 1116
Abstract
Background: Cancer ranks as a leading cause of death worldwide. It is urgent to develop intelligent co-delivery systems for cancer chemotherapy to achieve reduced side-effects and enhanced therapeutic efficacy. Methods: We chose oligo-hyaluronic acid (oHA, a low molecular weight of HA) as the [...] Read more.
Background: Cancer ranks as a leading cause of death worldwide. It is urgent to develop intelligent co-delivery systems for cancer chemotherapy to achieve reduced side-effects and enhanced therapeutic efficacy. Methods: We chose oligo-hyaluronic acid (oHA, a low molecular weight of HA) as the carrier, and adriamycin (ADM) and paclitaxel (PTX) as the co-delivered drugs. The oHA-ss-PTX macromolecular prodrug was synthesized by introducing glutathione-stimuli-responsive disulfide bonds through chemical reactions. Then, we constructed ADM-loading micelles (ADM/oHA-ss-PTX) in one step by microfluidic preparation. The delivery efficacy was evaluated comprehensively in vitro and in vivo. The biocompatibility of ADM/oHA-ss-PTX was assessed by hemolysis activity analysis, BSA adsorption testing, and cell viability assay in endothelial cells. Results: The resulting ADM/oHA-ss-PTX micelles possessed a dynamic size (127 ± 1.4 nm, zeta potential −9.0 mV), a high drug loading content of approximately 21.2% (PTX) and 7.6% (ADM). Compared with free ADM+PTX, ADM/oHA-ss-PTX showed enhanced blood stability and more efficiently inhibited cancer cell proliferation. Moreover, due to the CD44-mediated endocytosis pathway, a greater number of ADM/oHA-ss-PTX micelles were absorbed by A549 cells than by oHA-saturated A549 cells. In vivo experiments also showed that ADM/oHA-ss-PTX micelles had excellent therapeutic effects and targeting ability. These results show that ADM/oHA-ss-PTX micelles were a promising platform for co-delivery sequential therapy in CD44-positive cancer. Conclusions: In conclusion, these results convincingly demonstrate that ADM/oHA-ss-PTX micelles hold great promise as a novel platform for co-delivering multiple drugs. Their enhanced properties not only validate the potential of this approach for sequential cancer therapy in CD44-positive cancers but also pave the way for future clinical translation and further optimization in cancer treatment. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

25 pages, 1732 KB  
Review
Pt(IV) Complexes as Anticancer Drugs and Their Relationship with Oxidative Stress
by Vlad Iova, Radu Ciprian Tincu, Ioana Scrobota and Mihail Silviu Tudosie
Biomedicines 2025, 13(4), 981; https://doi.org/10.3390/biomedicines13040981 - 17 Apr 2025
Cited by 3 | Viewed by 2225
Abstract
Despite continuous research, cancer is still a leading cause of death worldwide; therefore, new methods of cancer management improvement are emerging. It is well known that in the pathophysiology of cancer, oxidative stress (OS) is a significant factor. Nevertheless, there is currently no [...] Read more.
Despite continuous research, cancer is still a leading cause of death worldwide; therefore, new methods of cancer management improvement are emerging. It is well known that in the pathophysiology of cancer, oxidative stress (OS) is a significant factor. Nevertheless, there is currently no quick or easy way to identify OS in cancer patients using blood tests. Currently, in cancer treatments, Pt(IV) complexes are preferred to Pt(II) complexes in terms of adverse effects, drug resistance, and administration methods. Intracellular reductants convert Pt(IV) complexes to their Pt(II) analogs, which are Pt compounds with anti-carcinogenic effects. Our aim was to find out if Pt(IV) complexes could be used to assess blood oxidative stress indicators and, consequently, monitor the development of cancer. In this review, we analyzed previous research using the PubMed and Google Scholar public databases to verify the potential use of Pt(IV) complexes in cancer management. We found that two main serum antioxidants, glutathione and ascorbic acid, which are easily measured using conventional methods, react favorably with Pt(IV) complexes. Our research results suggest Pt(IV) complexes as therapeutic anticancer drugs and potential diagnosis agents. However, further research must be conducted to verify this hypothesis. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

17 pages, 1547 KB  
Article
Green Biocatalysis of Xylitol Monoferulate: Candida antarctica Lipase B-Mediated Synthesis and Characterization of Novel Bifunctional Prodrug
by Federico Zappaterra, Francesco Presini, Domenico Meola, Chaimae Chaibi, Simona Aprile, Lindomar Alberto Lerin and Pier Paolo Giovannini
BioTech 2025, 14(2), 25; https://doi.org/10.3390/biotech14020025 - 2 Apr 2025
Cited by 1 | Viewed by 1434
Abstract
Natural compounds with significant bioactive properties can be found in abundance within biomasses. Especially prominent for their anti-inflammatory, neuroprotective, antibacterial, and antioxidant activities are cinnamic acid derivatives (CAs). Ferulic acid (FA), a widely studied phenylpropanoid, exhibits a broad range of therapeutic and nutraceutical [...] Read more.
Natural compounds with significant bioactive properties can be found in abundance within biomasses. Especially prominent for their anti-inflammatory, neuroprotective, antibacterial, and antioxidant activities are cinnamic acid derivatives (CAs). Ferulic acid (FA), a widely studied phenylpropanoid, exhibits a broad range of therapeutic and nutraceutical applications, demonstrating antidiabetic, anticancer, antimicrobial, and hepato- and neuroprotective activities. This research investigates the green enzymatic synthesis of innovative and potentially bifunctional prodrug derivatives of FA, designed to enhance solubility and stability profiles. Selective esterification was employed to conjugate FA with xylitol, a biobased polyol recognized for its bioactive antioxidant properties and safety profile. Furthermore, by exploiting t-amyl alcohol as a green solvent, the enzymatic synthesis of the derivative was optimized for reaction parameters including temperature, reaction time, enzyme concentration, and molar ratio. The synthesized derivative, xylitol monoferulate (XMF), represents a novel contribution to the literature. The comprehensive characterization of this compound was achieved using advanced spectroscopic methods, including 1H-NMR, 13C-NMR, COSY, HSQC, and HMBC. This study represents a significant advancement in the enzymatic synthesis of high-value biobased derivatives, demonstrating increased biological activities and setting the stage for future applications in green chemistry and the sustainable production of bioactive compounds. Full article
(This article belongs to the Section Industry, Agriculture and Food Biotechnology)
Show Figures

Figure 1

Back to TopTop