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

Journals

Article Types

Countries / Regions

Search Results (158)

Search Parameters:
Keywords = toxic payload

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 3930 KB  
Review
Targeted Hepatic Delivery of Bioactive Molecules via Nanovesicles: Recent Developments and Emerging Directions
by Alessia Rita Canestrale, Sharad Kholia, Veronica Dimuccio and Maria Beatriz Herrera Sanchez
J. Pers. Med. 2026, 16(1), 1; https://doi.org/10.3390/jpm16010001 - 19 Dec 2025
Viewed by 344
Abstract
Liver diseases, including fibrosis, viral hepatitis, hepatocellular carcinoma, and monogenic genetic disorders, represent a major global health burden with limited therapeutic options and frequent systemic toxicity from conventional treatments. Nanovesicle-based drug and gene delivery systems offer targeted approaches that may improve therapeutic precision [...] Read more.
Liver diseases, including fibrosis, viral hepatitis, hepatocellular carcinoma, and monogenic genetic disorders, represent a major global health burden with limited therapeutic options and frequent systemic toxicity from conventional treatments. Nanovesicle-based drug and gene delivery systems offer targeted approaches that may improve therapeutic precision and reduce off-target effects. This review aims to evaluate the promise and comparative potential of three key nanovesicle platforms—lipid nanoparticles (LNPs), extracellular vesicles (EVs) and liposomes—for drug and gene delivery in liver disease therapy. A systematic search of peer-reviewed studies published in electronic databases was performed, focusing on preclinical and clinical research investigating the use of LNPs, EVs and liposomes for hepatic drug or gene delivery. Studies were analyzed for vesicle composition, targeting efficiency, payload capacity, therapeutic outcomes, and reported limitations. The analysis indicates that LNPs demonstrate strong efficiency in nucleic acid encapsulation and delivery, supported by growing clinical translation. EVs show promising biocompatibility and innate targeting to hepatic cells but face challenges in large-scale production and standardization. Liposomes remain versatile and well-characterized platforms capable of carrying diverse therapeutic molecules, though rapid clearance can limit their efficacy. Together, these nanovesicle systems hold considerable potential for advancing targeted drug and gene therapies in liver disease. Future work should focus on improving stability, manufacturing scalability, and cell-specific targeting to support clinical translation. Full article
(This article belongs to the Section Omics/Informatics)
Show Figures

Graphical abstract

38 pages, 2128 KB  
Review
Antibody–Drug Conjugates and Beyond: Next-Generation Targeted Therapies for Breast Cancer
by Adil Farooq Wali, Mohamed El-Tanani, Sirajunisa Talath, Syed Arman Rabbani, Imran Rashid Rangraze, Shakta Mani Satyam, Ashot Avagimyan, Karolina Hoffmann, Ioannis Ilias, Sorina Ispas, Maggio Viviana, Anna Paczkowska and Manfredi Rizzo
Cancers 2025, 17(24), 3943; https://doi.org/10.3390/cancers17243943 - 10 Dec 2025
Viewed by 1165
Abstract
Breast cancer is the most common cancer and the most important cause of cancer-related death in females worldwide. Antibody–drug conjugates (ADCs) represent a novel class of targeted therapies that combine the precision of monoclonal antibodies with the potent cell-killing activity of cytotoxic drugs. [...] Read more.
Breast cancer is the most common cancer and the most important cause of cancer-related death in females worldwide. Antibody–drug conjugates (ADCs) represent a novel class of targeted therapies that combine the precision of monoclonal antibodies with the potent cell-killing activity of cytotoxic drugs. This review highlights recent mechanistic, technological, and clinical developments of ADCs in breast cancer, including next-generation ADCs beyond those that target HER2 (human epidermal growth factor receptor 2). Authors performed a systematic literature study for ADCs and their structural features, including their components (antibody, linker, and payload) and their therapeutic efficacy. A frame of preclinical research findings and clinical evidence integration of HER2-targeted therapy outcomes in HER2-positive, HER2-low, and triple-negative breast cancer (TNBC) subtypes were presented. Clinical studies of antibody–drug conjugates such as trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and sacituzumab govitecan have demonstrated significant improvements in progression-free survival and overall survival across diverse breast cancer patient populations. ADCs offer unique advantages in breast cancer therapy by combining the precision of targeted antibodies with the potency of chemotherapy drugs. This allows them to selectively kill cancer cells, overcome resistance, reduce toxicity to healthy tissues, and expand treatment options for difficult subtypes like HER2-low and triple-negative breast cancer. Unlike previous reviews focusing on HER2-targeted ADCs, herein we review exciting ADCs targeting HER3 HER3 (human epidermal growth factor receptor 3) and Nectin-4, as well as the implications of bispecific and immune-stimulatory ADCs in the clinic. Additionally, it features mechanism-based innovations and novel trial data that revolutionize ADC applications in the HER2-low as well as the triple-negative breast cancer subtypes. The advent of ADC is changing precision oncology in breast cancer. With a new design and indications evolving, they are an attractive avenue for bypassing resistance and reducing toxicity and ultimately improving patient outcomes in the molecular subtypes. The present review summarizes recent advancements in antibody–drug conjugates (ADCs) and emerging targeted therapeutic strategies for breast cancer. It covers mechanistic insights, linker–payload innovations, receptor-based targeting approaches, clinical trial progress, and next-generation modalities that extend beyond HER2-directed ADCs. Current challenges, safety profiles, and future opportunities in engineering more selective and effective ADC platforms are also discussed. Full article
(This article belongs to the Special Issue Breast Cancer Research and Treatment)
Show Figures

Figure 1

16 pages, 1875 KB  
Article
Structural Characterization of Linker Shielding in ADC Site-Specific Conjugates
by Maru Jaime-Garza, Andrew Waight, Manish Hudlikar, Michael J. Eddins, Elnaz S. Rasti, Jan Paulo T. Zaragoza, Laurence Fayadat-Dilman, Jill E. Chrencik, Sandra B. Gabelli, Yun-Ting Chen and Cameron L. Noland
Pharmaceutics 2025, 17(12), 1568; https://doi.org/10.3390/pharmaceutics17121568 - 5 Dec 2025
Viewed by 1399
Abstract
Background/Objectives: Antibody–Drug Conjugates (ADCs) have rapidly evolved from early, rudimentary conjugates to highly targeted and precisely engineered molecules. Despite notable clinical successes, ADCs continue to face significant challenges, including aggregation and high hydrophobicity driven by high drug-to-antibody ratios (DARs), premature payload release, [...] Read more.
Background/Objectives: Antibody–Drug Conjugates (ADCs) have rapidly evolved from early, rudimentary conjugates to highly targeted and precisely engineered molecules. Despite notable clinical successes, ADCs continue to face significant challenges, including aggregation and high hydrophobicity driven by high drug-to-antibody ratios (DARs), premature payload release, dose-limiting toxicities, and suboptimal pharmacokinetics. While site-specific linker–payload conjugation has improved ADC homogeneity and stability, the structural basis of antibody–linker interactions at specific sites remains underexplored. Methods: In this work, we present the crystal structures of trastuzumab Fab and Fc domains site-specifically conjugated with a cleavable linker–payload. Results: Our findings suggest that pockets within both Fab and Fc regions may interact with and shield the linker portion of the conjugate. Conclusions: These insights highlight the previously underappreciated potential of structure-based design to drive the optimization of ADC linker chemistry and facilitate the co-design of bespoke linker–payloads tailored to individual antibody conjugation sites. Full article
(This article belongs to the Special Issue Antibody–Drug Conjugates Therapeutics)
Show Figures

Figure 1

33 pages, 3055 KB  
Review
CAR Therapies: Ex Vivo and In Vivo Potential of Exosomes and Biomimetic Nanoparticles
by Ekaterina Tkachenko, Natalia Ponomareva, Konstantin Evmenov, Artyom Kachanov, Sergey Brezgin, Anastasiya Kostyusheva, Vladimir Chulanov, Elena Volchkova, Alexander Lukashev, Dmitry Kostyushev and Peter Timashev
Cancers 2025, 17(23), 3766; https://doi.org/10.3390/cancers17233766 - 25 Nov 2025
Viewed by 705
Abstract
Chimeric antigen receptor (CAR) therapy represents a promising modality for treating cancer and autoimmune diseases, employing genetically engineered immune cells. Despite remarkable clinical outcomes, its broad implementation is constrained by significant challenges, including toxicity, limited specificity, and complexities associated with genetic material delivery. [...] Read more.
Chimeric antigen receptor (CAR) therapy represents a promising modality for treating cancer and autoimmune diseases, employing genetically engineered immune cells. Despite remarkable clinical outcomes, its broad implementation is constrained by significant challenges, including toxicity, limited specificity, and complexities associated with genetic material delivery. Biological nanoparticles, such as exosomes, virus-like particles, and biomimetic nanostructures, possess unique properties that can address these limitations. These nanoplatforms enable targeted delivery of genetic constructs, mitigate the risk of cytokine release syndrome, modulate CAR cell activity, and can function as biosensors. Furthermore, they facilitate non-viral, in vivo CAR cell engineering, streamlining the process compared to conventional ex vivo methods. The advancement of in vivo strategies underscores the critical need to overcome toxicity hurdles inherent to current CAR-T platforms. In this context, exosomes and biomimetic nanoparticles offer considerable potential due to their innate biocompatibility, programmability, and versatile cargo capacity for payloads like mRNA and circular RNA. This review comprehensively outlines contemporary genetic platforms for CAR expression and examines the opportunities presented by biological delivery vehicles. It focuses on recent achievements and revisits fundamental CAR principles through the lens of emerging technologies aimed at confronting persistent challenges in the field. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

31 pages, 1047 KB  
Review
Translational Advances in Lipid Nanoparticle Drug Delivery Systems for Cancer Therapy: Current Status and Future Horizons
by Hari Krishnareddy Rachamala
Pharmaceutics 2025, 17(10), 1315; https://doi.org/10.3390/pharmaceutics17101315 - 10 Oct 2025
Cited by 1 | Viewed by 2778
Abstract
Lipid nanoparticles/liposomes (LNPs) represent a highly adaptable nanocarrier system that has gained significant traction in oncology for both therapeutic and diagnostic (theranostic) purposes. Their structural flexibility, biocompatibility, and capacity to encapsulate diverse therapeutic agents ranging from chemotherapeutics to nucleic acids and imaging tracers [...] Read more.
Lipid nanoparticles/liposomes (LNPs) represent a highly adaptable nanocarrier system that has gained significant traction in oncology for both therapeutic and diagnostic (theranostic) purposes. Their structural flexibility, biocompatibility, and capacity to encapsulate diverse therapeutic agents ranging from chemotherapeutics to nucleic acids and imaging tracers have enabled targeted cancer treatment with improved efficacy and reduced systemic toxicity. This review critically examines liposome-based platforms across a broad spectrum of cancers, including melanoma, lung, colorectal, liver, breast, ovarian, pancreatic, brain tumors, sarcoma, neuroblastoma, and leukemia. It outlines recent advances in ligand-mediated targeting, pH- and temperature-responsive release systems, and multifunctional LNPs capable of delivering combined therapeutic and imaging payloads. Moreover, the review discusses preclinical outcomes, current clinical trial status, and the challenges hindering clinical translation. By integrating recent innovations and emphasizing translational potential, this work highlights the pivotal role of LNPs in advancing precision cancer therapeutics and diagnostics. Full article
(This article belongs to the Special Issue Advanced Liposomes for Drug Delivery, 2nd Edition)
Show Figures

Graphical abstract

23 pages, 5760 KB  
Article
Effect of Crosslinking Using Heat on the Physicochemical Features of Bsa–Capsaicin Nanoparticles
by Lino Sánchez-Segura, Silvio Zaina, Angela F. Kú-González, José Alfredo Guzmán-López, Laura E. Zavala-García and Mercedes G. López
Pharmaceutics 2025, 17(10), 1306; https://doi.org/10.3390/pharmaceutics17101306 - 8 Oct 2025
Viewed by 782
Abstract
Background/Objectives: The synthesis of protein nanoparticles (NPs) using the coacervation method is influenced by critical parameters. The use of glutaraldehyde limits the pharmacological applications of NPs in humans due to the potential toxicity of residual aldehydes that remain after the purification of [...] Read more.
Background/Objectives: The synthesis of protein nanoparticles (NPs) using the coacervation method is influenced by critical parameters. The use of glutaraldehyde limits the pharmacological applications of NPs in humans due to the potential toxicity of residual aldehydes that remain after the purification of the nanoparticles. The aim was to assess heat effect as a crosslinking agent for the synthesis of bovine serum albumin (BSA)–capsaicin nanoparticles and its effect on the physicochemical characteristics of nanoparticles. Results: The initial concentrations of BSA and capsaicin in the formulation were directly correlated with the amount of BSA that was transformed into nanoparticles and the loaded capsaicin (r = 0.97, p = 0.0003 and r = 0.95, p = 0.0003), respectively. Furthermore, the morphometric parameters of nanoparticles were affected by the increase in capsaicin concentration, but not by temperature. The nanoparticles increased in dimensions and showed a loss of shape due to coalescence between nanoparticles. The ζ-potential decreased with the increase in the concentration of capsaicin added. This effect compromised the stability of the nanoparticles; on the other hand, molecular interactions were observed between hydrophobic residues of phenylalanine and tyrosine in BSA and the hydrophobic moiety of capsaicin. At the same time, BSA nanoparticles showed a potential for disassembling and delivering the payload capsaicin, which caused an antisteatotic effect in the liver of a murine model. Conclusions: heat (70 °C) can replace crosslinking agents, such as glutaraldehyde. This property is particularly useful when an aldehyde-free synthesis of BSA nanoparticles is needed. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

32 pages, 1169 KB  
Review
Actinium-225/Bismuth-213 as Potential Leaders for Targeted Alpha Therapy: Current Supply, Application Barriers, and Future Prospects
by Mohamed F. Nawar, Adli A. Selim, Basma M. Essa, Alaa F. El-Daoushy, Mohamed M. Swidan, Claudia G. Chambers, Mohammed H. Al Qahtani, Charles J. Smith and Tamer M. Sakr
Cancers 2025, 17(18), 3055; https://doi.org/10.3390/cancers17183055 - 18 Sep 2025
Viewed by 3409
Abstract
Alpha therapy (TAT) relies on combining alpha-emitting radionuclides with specific cell-targeting vectors to deliver a high payload of cytotoxic radiation capable of destroying tumor tissues. TAT efficacy comes from the tissue selectivity of the targeting vector, the high linear energy transfer (LET) of [...] Read more.
Alpha therapy (TAT) relies on combining alpha-emitting radionuclides with specific cell-targeting vectors to deliver a high payload of cytotoxic radiation capable of destroying tumor tissues. TAT efficacy comes from the tissue selectivity of the targeting vector, the high linear energy transfer (LET) of the radionuclide, and the short range of alpha particles in tissues. Recent research studies have been directed to evaluate TAT on a preclinical and clinical scale, including evaluating damage to tumor tissues with minimal toxic radiation effects on surrounding healthy tissues. This review highlights the use of Actinium-225/Bismuth-213 radionuclides as promising candidates for TAT. Herein, we begin with a discussion on the production and supply of [225Ac]Ac/[213Bi]Bi followed by the formulation of [225Ac]Ac/[213Bi]Bi-radiopharmaceuticals using different radiolabeling techniques. Finally, we have summarized the preclinical and clinical evaluation of these potential radiotheranostic agents. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

33 pages, 1320 KB  
Review
Antibody–Drug Conjugates in Breast Cancer: Navigating Innovations, Overcoming Resistance, and Shaping Future Therapies
by Hussein Sabit, Salma Abbas, Moataz T. El-Safoury, Engy M. Madkour, Sahar Mahmoud, Shaimaa Abdel-Ghany, Yasser Albrahim, Ibtesam S. Al-Dhuayan, Sanaa Rashwan, Ahmed El-Hashash and Borros Arneth
Biomedicines 2025, 13(9), 2227; https://doi.org/10.3390/biomedicines13092227 - 10 Sep 2025
Cited by 3 | Viewed by 5097
Abstract
Antibody–drug conjugates (ADCs) have revolutionized breast cancer (BC) therapy by combining targeted antibody specificity with potent cytotoxic payloads, thereby enhancing efficacy while minimizing systemic toxicity. This review highlights significant innovations driving ADC development alongside persistent challenges. Recent advancements include novel antibody–drug conjugate (ADC) [...] Read more.
Antibody–drug conjugates (ADCs) have revolutionized breast cancer (BC) therapy by combining targeted antibody specificity with potent cytotoxic payloads, thereby enhancing efficacy while minimizing systemic toxicity. This review highlights significant innovations driving ADC development alongside persistent challenges. Recent advancements include novel antibody–drug conjugate (ADC) designs targeting diverse antigens, such as HER2, HER3, and CD276, demonstrating potent anti-tumor activity and improved strategies for drug delivery. For instance, dual-payload ADCs and those leveraging extracellular vesicles offer new dimensions in precision oncology. The integration of ADCs into sequential therapy, such as sacituzumab govitecan with TOP1/PARP inhibitors, further underscores their synergistic potential. Despite these innovations, critical challenges remain, including tumor heterogeneity and acquired drug resistance, which often involve complex molecular alterations. Moreover, optimizing ADC components, including linker chemistry and payload characteristics, is essential for ensuring stability and minimizing off-target toxicity. The burgeoning role of artificial intelligence and machine learning is pivotal in accelerating the design of ADCs, target identification, and personalized patient stratification. This review aims to comprehensively explore the cutting-edge innovations and inherent challenges in ADC development for BC, providing a holistic perspective on their current impact and future trajectory. Full article
(This article belongs to the Special Issue New Insights into the Diagnosis and Treatment of Breast Cancer)
Show Figures

Figure 1

33 pages, 1878 KB  
Review
Strategic and Chemical Advances in Antibody–Drug Conjugates
by Ibrahim A. Alradwan, Meshal K. Alnefaie, Nojoud AL Fayez, Alhassan H. Aodah, Majed A. Majrashi, Meshael Alturki, Mohannad M. Fallatah, Fahad A. Almughem, Essam A. Tawfik and Abdullah A. Alshehri
Pharmaceutics 2025, 17(9), 1164; https://doi.org/10.3390/pharmaceutics17091164 - 5 Sep 2025
Cited by 2 | Viewed by 5152
Abstract
Antibody–drug conjugates (ADCs) are a rapidly advancing class of targeted cancer therapeutics that couple the antigen specificity of monoclonal antibodies (mAbs) with the potent cytotoxicity of small-molecule drugs. In their core design, a tumor-targeting antibody is covalently linked to a cytotoxic payload via [...] Read more.
Antibody–drug conjugates (ADCs) are a rapidly advancing class of targeted cancer therapeutics that couple the antigen specificity of monoclonal antibodies (mAbs) with the potent cytotoxicity of small-molecule drugs. In their core design, a tumor-targeting antibody is covalently linked to a cytotoxic payload via a chemical linker, enabling the selective delivery of highly potent agents to malignant cells while sparing normal tissues, thereby improving the therapeutic index. Humanized and fully human immunoglobulin G1(IgG1) antibodies are the most common ADC backbones due to their stability in systemic circulation, robust Fcγ receptor engagement for immune effector functions, and reduced immunogenicity. Antibody selection requires balancing tumor specificity, internalization rate, and binding affinity to avoid barriers to tissue penetration, such as the binding-site barrier effect, while emerging designs exploit tumor-specific antigen variants or unique post-translational modifications to further enhance selectivity. Advances in antibody engineering, linker chemistry, and payload innovation have reinforced the clinical success of ADCs, with more than a dozen agents FDA approved for hematologic malignancies and solid tumors and over 200 in active clinical trials. This review critically examines established and emerging conjugation strategies, including lysine- and cysteine-based chemistries, enzymatic tagging, glycan remodeling, non-canonical amino acid incorporation, and affinity peptide-mediated methods, and discusses how conjugation site, drug-to-antibody ratio (DAR) control, and linker stability influence pharmacokinetics, efficacy, and safety. Innovations in site-specific conjugation have improved ADC homogeneity, stability, and clinical predictability, though challenges in large-scale manufacturing and regulatory harmonization remain. Furthermore, novel ADC architectures such as bispecific ADCs, conditionally active (probody) ADCs, immune-stimulating ADCs, protein-degrader ADCs, and dual-payload designs are being developed to address tumor heterogeneity, drug resistance, and off-target toxicity. By integrating mechanistic insights, preclinical and clinical data, and recent technological advances, this work highlights current progress and future directions for next-generation ADCs aimed at achieving superior efficacy, safety, and patient outcomes, especially in treating refractory cancers. Full article
(This article belongs to the Section Biologics and Biosimilars)
Show Figures

Figure 1

35 pages, 2365 KB  
Review
Therapeutic Effect of Membrane Vesicle Drug Delivery Systems in Inflammatory Bowel Disease
by Zhe Zhao, Ziyun Li, Yihuang Gu and Renjun Gu
Pharmaceutics 2025, 17(9), 1127; https://doi.org/10.3390/pharmaceutics17091127 - 28 Aug 2025
Cited by 3 | Viewed by 1932
Abstract
Inflammatory bowel disease (IBD) is a chronic, heterogeneous condition characterized by recurrent intestinal inflammation and sustained mucosal barrier damage, profoundly impairing patients’ quality of life and imposing a considerable socioeconomic burden. Current therapeutic options are often constrained by low oral bioavailability, pronounced systemic [...] Read more.
Inflammatory bowel disease (IBD) is a chronic, heterogeneous condition characterized by recurrent intestinal inflammation and sustained mucosal barrier damage, profoundly impairing patients’ quality of life and imposing a considerable socioeconomic burden. Current therapeutic options are often constrained by low oral bioavailability, pronounced systemic toxicity, and inadequate tissue specificity, limiting their ability to achieve precise and durable efficacy. In recent years, membrane vesicle-based drug delivery systems (MV-DDSs) have shown considerable promise for precision IBD therapy owing to their excellent biocompatibility, mucosal barrier-penetrating capacity, and low immunogenicity. Building upon a systematic discussion of the roles of MV-DDSs in suppressing inflammatory signaling, modulating oxidative stress, preserving barrier integrity, reshaping the gut microbiota, and regulating programmed cell death, this review further compares the differences in key molecular targets and functional outcomes among vesicles of diverse origins and carrying distinct therapeutic payloads. These insights provide a comprehensive strategic reference and theoretical foundation for the rational design, mechanistic optimization, and clinical translation of MV-DDSs in IBD therapy. Full article
(This article belongs to the Special Issue Advances of Membrane Vesicles in Drug Delivery Systems, 2nd Edition)
Show Figures

Figure 1

33 pages, 2203 KB  
Review
Cyclodextrin-Based Nanotransporters as a Versatile Tool to Manage Oxidative Stress-Induced Lung Diseases
by Supandeep Singh Hallan, Francesca Ferrara, Maddalena Sguizzato and Rita Cortesi
Antioxidants 2025, 14(8), 1007; https://doi.org/10.3390/antiox14081007 - 17 Aug 2025
Cited by 2 | Viewed by 2126
Abstract
Oxidative stress is one of the key elements in lung-related complications such as cystic fibrosis, acute lung injury, pulmonary hypertension, bronchopulmonary dysplasia, chronic airway diseases, lung cancer, COVID-19, and many others. Antioxidant and anti-inflammatory therapy can be considered as supportive alternatives in their [...] Read more.
Oxidative stress is one of the key elements in lung-related complications such as cystic fibrosis, acute lung injury, pulmonary hypertension, bronchopulmonary dysplasia, chronic airway diseases, lung cancer, COVID-19, and many others. Antioxidant and anti-inflammatory therapy can be considered as supportive alternatives in their management. However, most naturally derived antioxidants face issues with poor aqueous solubility and stability, which hinder their clinical utility. Remarkably, local pulmonary delivery circumvents the severe limitations of oral delivery, including hepatic first-pass metabolism and organ toxicity, and enables a higher drug payload in the lungs. Here, in this review, we present cyclodextrin as a potential drug carrier for pulmonary administration, exploring the possibilities of its surface modification, complexation with other drug transporters, and loading of cannabidiols, siRNA, and antibodies as future trends. However, the lack of a robust physiological model for assessing the efficacy of lung-oriented drug targeting is a significant concern in its path to clinical and commercial success. Full article
(This article belongs to the Special Issue Applications of Antioxidant Nanoparticles, 2nd Edition)
Show Figures

Figure 1

11 pages, 2406 KB  
Article
Surfactant-Free Electrosprayed Alginate Beads for Oral Delivery of Hydrophobic Compounds
by Hye-Seon Jeong, Hyo-Jin Kim, Sung-Min Kang and Chang-Hyung Choi
Polymers 2025, 17(15), 2098; https://doi.org/10.3390/polym17152098 - 30 Jul 2025
Viewed by 963
Abstract
Oral delivery of hydrophobic compounds remains challenging due to their poor aqueous solubility and the potential toxicity associated with conventional surfactant-based emulsions. To address these issues, we present a surfactant-free encapsulation strategy using electrosprayed alginate hydrogel beads for the stable and controlled delivery [...] Read more.
Oral delivery of hydrophobic compounds remains challenging due to their poor aqueous solubility and the potential toxicity associated with conventional surfactant-based emulsions. To address these issues, we present a surfactant-free encapsulation strategy using electrosprayed alginate hydrogel beads for the stable and controlled delivery of hydrophobic oils. Hydrophobic compounds were dispersed in high-viscosity alginate solutions without surfactants via ultrasonication, forming kinetically stable oil-in-water dispersions. These mixtures were electrosprayed into calcium chloride baths, yielding monodisperse hydrogel beads. Higher alginate concentrations improved droplet sphericity and suppressed phase separation by enhancing matrix viscosity. The resulting beads exhibited stimuli-responsive degradation and controlled release behavior in response to physiological ionic strength. Dense alginate networks delayed ion exchange and prolonged structural integrity, while elevated external ionic conditions triggered rapid disintegration and immediate payload release. This simple and scalable system offers a biocompatible platform for the oral delivery of lipophilic active compounds without the need for surfactants or complex fabrication steps. Full article
Show Figures

Figure 1

22 pages, 1549 KB  
Review
Nanotechnology-Based Delivery of CRISPR/Cas9 for Cancer Treatment: A Comprehensive Review
by Mohd Ahmar Rauf, Afifa Rao, Siva Sankari Sivasoorian and Arun K. Iyer
Cells 2025, 14(15), 1136; https://doi.org/10.3390/cells14151136 - 23 Jul 2025
Cited by 5 | Viewed by 6678
Abstract
CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats-associated protein 9)-mediated genome editing has emerged as a transformative tool in medicine, offering significant potential for cancer therapy because of its capacity to precisely target and alter the genetic modifications associated with the disease. However, a [...] Read more.
CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats-associated protein 9)-mediated genome editing has emerged as a transformative tool in medicine, offering significant potential for cancer therapy because of its capacity to precisely target and alter the genetic modifications associated with the disease. However, a major challenge for its clinical translation is the safe and efficient in vivo delivery of CRISPR/Cas9 components to target cells. Nanotechnology is a promising solution to this problem. Nanocarriers, owing to their tunable physicochemical properties, can encapsulate and protect CRISPR/Cas9 components, enabling targeted delivery and enhanced cellular uptake. This review provides a comprehensive examination of the synergistic potential of CRISPR/Cas9 and nanotechnology in cancer therapy and explores their integrated therapeutic applications in gene editing and immunotherapy. A critical aspect of in vivo CRISPR/Cas9 application is to achieve effective localization at the tumor site while minimizing off-target effects. Nanocarriers can be engineered to overcome biological barriers, thereby augmenting tumor-specific delivery and facilitating intracellular uptake. Furthermore, their design allows for controlled release of the therapeutic payload, ensuring sustained efficacy and reduced systemic toxicity. The optimization of nanocarrier attributes, including size, shape, surface charge, and composition, is crucial for improving the cellular internalization, endosomal escape, and nuclear localization of CRISPR/Cas9. Moreover, surface functionalization with targeting ligands can enhance the specificity of cancer cells, leading to improved gene-editing accuracy. This review thoroughly discusses the challenges associated with in vivo CRISPR/Cas9 delivery and the innovative nanotechnological strategies employed to overcome them, highlighting their combined potential for advancing cancer treatment for clinical application. Full article
Show Figures

Figure 1

35 pages, 3359 KB  
Article
GSH/pH-Responsive Chitosan–PLA Hybrid Nanosystems for Targeted Ledipasvir Delivery to HepG2 Cells: Controlled Release, Improved Selectivity, DNA Interaction, Electrochemical and Stopped-Flow Kinetics Analyses
by Ahmed M. Albasiony, Amr M. Beltagi, Mohamed M. Ibrahim, Shaban Y. Shaban and Rudi van Eldik
Int. J. Mol. Sci. 2025, 26(13), 6070; https://doi.org/10.3390/ijms26136070 - 24 Jun 2025
Cited by 3 | Viewed by 1633
Abstract
This study aimed to design dual-responsive chitosan–polylactic acid nanosystems (PLA@CS NPs) for controlled and targeted ledipasvir (LED) delivery to HepG2 liver cancer cells, thereby reducing the systemic toxicity and improving the therapeutic selectivity. Two formulations were developed utilizing ionotropic gelation and w/ [...] Read more.
This study aimed to design dual-responsive chitosan–polylactic acid nanosystems (PLA@CS NPs) for controlled and targeted ledipasvir (LED) delivery to HepG2 liver cancer cells, thereby reducing the systemic toxicity and improving the therapeutic selectivity. Two formulations were developed utilizing ionotropic gelation and w/o/w emulsion techniques: LED@CS NPs with a size of 143 nm, a zeta potential of +43.5 mV, and a loading capacity of 44.1%, and LED-PLA@CS NPs measuring 394 nm, with a zeta potential of +33.3 mV and a loading capacity of 89.3%, with the latter demonstrating significant drug payload capacity. Since most drugs work through interaction with DNA, the in vitro affinity of DNA to LED and its encapsulated forms was assessed using stopped-flow and other approaches. They bind through multi-modal electrostatic and intercalative modes via two reversible processes: a fast complexation followed by a slow isomerization. The overall binding activation parameters for LED (cordination affinity, Ka = 128.4 M−1, Kd = 7.8 × 10−3 M, ΔG = −12.02 kJ mol−1), LED@CS NPs (Ka = 2131 M−1, Kd = 0.47 × 10−3 M, ΔG = −18.98 kJ mol−1) and LED-PLA@CS NPs (Ka = 22026 M−1, Kd = 0.045 × 10−3 M, ΔG = −24.79 kJ mol−1) were obtained with a reactivity ratio of 1/16/170 (LED/LED@CS NPs/LED-PLA@CS NPs). This indicates that encapsulation enhanced the interaction between the DNA and the LED-loaded nanoparticle systems, without changing the mechanism, and formed thermodynamically stable complexes. The drug release kinetics were assessed under tumor-mimetic conditions (pH 5.5, 10 mM GSH) and physiological settings (pH 7.4, 2 μM GSH). The LED@CS NPs and LED-PLA@CS NPs exhibited drug release rates of 88.0% and 73%, respectively, under dual stimuli over 50 h, exceeding the release rates observed under physiological conditions, which were 58% and 54%, thereby indicating that the LED@CS NPs and LED-PLA@CS NPs systems specifically target malignant tissue. Release regulated by Fickian diffusion facilitates tumor-specific payload delivery. Although encapsulation did not enhance the immediate cytotoxicity compared to free LED, as demonstrated by an in vitro cytotoxicity in HepG2 cancer cell lines, it significantly enhanced the therapeutic index (2.1-fold for LED-PLA@CS NPs) by protecting non-cancerous cells. Additionally, the nanoparticles demonstrated broad-spectrum antibacterial effects, suggesting efficacy in the prevention of chemotherapy-related infections. The dual-responsive LED-PLA@CS NPs allowed controlled tumor-targeted LED delivery with better selectivity and lower off-target toxicity, making LED-PLA@CS NPs interesting candidates for repurposing HCV treatments into safer cancer nanomedicines. Furthermore, this thorough analysis offers useful reference information for comprehending the interaction between drugs and DNA. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Graphical abstract

22 pages, 3036 KB  
Article
Synthesis and Characterization of Transferrin Receptor-Targeted Peptide Combination SN-38 and Rucaparib Conjugate for the Treatment of Glioblastoma
by Perpetue Bataille Backer and Simeon Kolawole Adesina
Pharmaceutics 2025, 17(6), 732; https://doi.org/10.3390/pharmaceutics17060732 - 2 Jun 2025
Viewed by 1888
Abstract
Background/Objectives: Glioblastoma represents a particularly aggressive and fatal type of brain tumor. Peptide-drug conjugates, which offer the promise of traversing the blood-brain barrier to selectively accumulate in tumor tissues and precisely target cancer cells, are an active area of research. We present the [...] Read more.
Background/Objectives: Glioblastoma represents a particularly aggressive and fatal type of brain tumor. Peptide-drug conjugates, which offer the promise of traversing the blood-brain barrier to selectively accumulate in tumor tissues and precisely target cancer cells, are an active area of research. We present the synthesis and characterization of the T7 peptide (HAIYPRH) as a targeting ligand for the transferrin receptor, which is highly expressed on both the blood-brain barrier and glioma cells. Methods: Using the T7 peptide, the synthesis, characterization, and biological evaluation of a transferrin receptor-targeted, combination SN-38 and rucaparib peptide drug conjugate (T7-SN-38-rucaparib) are described. Results: The T7 peptide drug conjugate readily cleaved in the presence of exogenous cathepsin B, releasing the active drug payloads. In vitro experiments demonstrated potent cytotoxic effects of the T7 peptide drug conjugate on glioblastoma cells (IC50 = 22.27 nM), with reduced toxicity to non-cancerous HEK 293 cells (IC50 = 115.78 nM), indicating selective toxicity toward cancer cells. Further investigations revealed that blocking transferrin receptors with drug-free T7 peptide significantly reduced the conjugate’s cytotoxicity, an effect that could be reversed by introducing exogenous cathepsin B to the cells. Conclusions: These findings highlight the potential of glioblastoma-targeted delivery of SN-38 and rucaparib based on specific recognition of the transferrin receptor for transport across the blood-brain barrier, offering the prospect of reduced toxicity and selective killing of cancer cells. Additionally, since rucaparib does not cross the blood-brain barrier, this work is significant to facilitate the use of rucaparib for the treatment of brain tumors. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Figure 1

Back to TopTop