André F Moreira, Hugo A L Filipe, Sónia P Miguel, Maximiano J Ribeiro, Paula Coutinho
{"title":"Recent advances in smart gold nanoparticles for photothermal therapy.","authors":"André F Moreira, Hugo A L Filipe, Sónia P Miguel, Maximiano J Ribeiro, Paula Coutinho","doi":"10.1080/17435889.2025.2500912","DOIUrl":"10.1080/17435889.2025.2500912","url":null,"abstract":"<p><p>Gold nanoparticles (AuNPs) possess unique properties, including low toxicity and excellent optical characteristics, making them highly appealing for biomedical applications. The plasmonic photothermal effect of AuNPs has been explored to trigger localized hyperthermia. Four commonly explored gold nanoparticles (spheres, rods, stars, and cages) are produced and optimized to present the localized surface plasmon resonance effect in the near-infrared region, exploiting the increased penetration in the human body. Additionally, the production of hybrid AuNPs, combining them with other materials, such as silica, graphene, zinc oxide, polymers, and small molecules has been explored to amplify the photothermal effect (<i>T</i> ≥ 45ºC). This review provides an overview of AuNPs' application in photothermal therapy, describing the general synthesis processes and the main particle parameters that affect their application in photothermal therapy, including the hybrid nanomaterials. Associated with this rapid progress, surface functionalization can also improve colloidal stability, safety, and therapeutic outcomes. In this regard, we also highlight the emerging trend of applying cell-derived vesicles as biomimetic coatings, capable of evading immune recognition, increasing blood circulation, and targeting specific tissues. In addition, the challenges and future developments of AuNPs for accelerating the clinical translations are discussed in light of their therapeutic and theragnostic potential.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1339-1353"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12140457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143999868","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Next generation gold nanomaterials for photoacoustic imaging.","authors":"Brendan R Barlow, Jinhwan Kim","doi":"10.1080/17435889.2025.2504330","DOIUrl":"10.1080/17435889.2025.2504330","url":null,"abstract":"<p><p>Photoacoustic (PA) imaging integrates ultrasound with the molecular contrast afforded by optical imaging, enabling noninvasive, real-time visualization of tissue structures and contrasts. Gold nanoparticles (GNPs) have been extensively studied as contrast agents for PA imaging due to their strong optical absorption derived from localized surface plasmon resonance, particularly when engineered to absorb in the near-infrared (NIR) region to enhance tissue penetration. However, the use of conventional anisotropic nanoparticles that absorb the NIR wavelengths is limited by their poor photostability under pulsed lasing conditions, which restricts their applicability in longitudinal <i>in vivo</i> imaging studies. This review first outlines the fundamental principles of PA imaging and introduces conventional GNP-based contrast agents, emphasizing their applications and inherent limitations. Subsequently, recent advances in GNP engineering are discussed, with particular focus on strategies to improve photostability, and a future perspective on the development of GNP-based PA contrast agents is provided.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1479-1493"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12143679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144044420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chandra Sekhar Kathera, Zehra Cobandede, Kaylea Titus, Ibrahim Mohammad, Mustafa Culha
{"title":"Nanomaterial-based scaffolds for bone regeneration with piezoelectric properties.","authors":"Chandra Sekhar Kathera, Zehra Cobandede, Kaylea Titus, Ibrahim Mohammad, Mustafa Culha","doi":"10.1080/17435889.2025.2504320","DOIUrl":"10.1080/17435889.2025.2504320","url":null,"abstract":"<p><p>For proper cellular growth, to prepare tissue scaffold mimicking the tissue properties is a significant challenge. Bone is a vital organ supporting the whole human body for its function. The efficiencies in its structure for a variety of reasons should properly be remedied. Bone tissue engineering (BTE) is an emerging field addressing to develop or repair bone tissue for its proper function. The bone is naturally a piezoelectric material and generates electrical stimuli because of mechanical stress. Thus, the use of piezoelectric materials to build bone tissue is of great interest in BTE. Both piezoelectric polymers and nanomaterials (NMs) are investigated for this goal. In this review, we give an overview of the recent advances in piezoelectric NMs to construct piezoelectric scaffolds in BTE.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1461-1477"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12143683/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144082605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Preparation of surface-modified PLGA nanoparticles containing carbon quantum dots: insights from C6 cell line assays.","authors":"Maral Motamedi, Fatemeh Madani, Masood Khosravani, Mahdi Adabi","doi":"10.1080/17435889.2025.2504322","DOIUrl":"10.1080/17435889.2025.2504322","url":null,"abstract":"<p><strong>Aims: </strong>In this study, carbon quantum dot (CQD)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were prepared, and their surfaces were modified with tween 20, tween 80, poloxamer 188, or poloxamer 407. The physicochemical properties, cytotoxicity, and cellular uptake of the NPs were subsequently evaluated using the C6 glioblastoma cell line.</p><p><strong>Materials & methods: </strong>The NPs were prepared via the nanoprecipitation method, and surface modifications were achieved through physical adsorption. The physicochemical characteristics of the NPs including mean diameter, zeta potential, and in vitro release of CQDs, were assessed. Hemolysis and in vitro studies, including MTT assay, apoptosis/necrosis assay, and cellular uptake, were conducted.</p><p><strong>Results: </strong>Poloxamer-coated NPs demonstrated a more sustained release of CQDs compared to tween-coated NPs. Hemolysis and cytotoxicity were concentration-dependent, with poloxamer-coated NPs exhibiting lower hemolysis at 2 mg/mL and reduced cytotoxicity at higher concentrations. All formulations were effectively internalized by C6 cells, and poloxamer 407-coated NPs showed the lowest rate of necrosis.</p><p><strong>Conclusions: </strong>Poloxamer-coated NPs exhibited favorable characteristics, including sustained release, lower toxicity, and enhanced cellular uptake. These findings support their potential as effective surface modifiers in the development of multifunctional nanocarriers for brain-targeted drug delivery.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1403-1416"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12143696/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144112795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rabia Liaqat, Fatima Rasool, Sobia Noreen, Nadia Rai, Ayesha Naseem, Muhammad Hamza Shoaib, Hassan Mahmood, Muhammad Azeem Ashraf
{"title":"Fabrication, characterization, and docking studies of furosemide-loaded nanosponges using the emulsion solvent diffusion method.","authors":"Rabia Liaqat, Fatima Rasool, Sobia Noreen, Nadia Rai, Ayesha Naseem, Muhammad Hamza Shoaib, Hassan Mahmood, Muhammad Azeem Ashraf","doi":"10.1080/17435889.2025.2501518","DOIUrl":"10.1080/17435889.2025.2501518","url":null,"abstract":"<p><strong>Aims: </strong>This study aimed to fabricate, characterize, and perform molecular docking of furosemide-loaded nanosponges (NSs) using the emulsion solvent diffusion method.</p><p><strong>Material and methods: </strong>Sustained-release NS formulations of furosemide were developed using ethylcellulose, polyvinyl alcohol (PVA), and dichloromethane (DCM) via the emulsion solvent diffusion technique. The formulations were evaluated for production yield, actual drug content, entrapment efficiency, drug-polymer compatibility, surface morphology, docking study, and in vitro drug release.</p><p><strong>Results: </strong>SEM images displayed the nanosized, spherical, porous, and spongy texture of furosemide NS. Fourier-transform infrared spectroscopy (FTIR) spectra showed no drug-polymer incompatibility. Powder X-ray diffraction (PXRD) analysis indicated an amorphous state of furosemide, while differential scanning calorimetry (DSC) suggested drug-polymer complexation. In vitro studies demonstrated sustained drug release for up to 10 h. Molecular docking supported stable interactions between furosemide and polymers. Molecular dynamics (MD) simulations further revealed adequate hydrogen bonding and diffusion behavior, confirming polymer composition-dependent release and structural stability.</p><p><strong>Conclusion: </strong>These findings indicate that furosemide-loaded NSs are a promising sustained-release delivery system capable of reducing dosing frequency and enhancing patient compliance.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1235-1247"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12140477/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144058353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rodolfo Villa, Ya-Ping Shiau, Sohaib Mahri, Kelsey Jane Racacho, Menghuan Tang, Qiufang Zong, Donovan Ruiz, Judy Kim, Yuanpei Li
{"title":"Immunomodulatory nanoplatforms with multiple mechanisms of action in cancer treatment.","authors":"Rodolfo Villa, Ya-Ping Shiau, Sohaib Mahri, Kelsey Jane Racacho, Menghuan Tang, Qiufang Zong, Donovan Ruiz, Judy Kim, Yuanpei Li","doi":"10.1080/17435889.2025.2500906","DOIUrl":"10.1080/17435889.2025.2500906","url":null,"abstract":"<p><p>Cancer immunotherapies have transformed oncology by utilizing the immune system to target malignancies; however, limitations in efficacy and potential side effects remain significant challenges. Nanoparticles have shown promise in enhancing drug delivery and improving immune activation, with the potential for numerous modifications to tailor them for specific environments or targets. Integrating nanoplatforms offers a promising avenue to overcome these hurdles, enhancing treatment outcomes and reducing adverse effects. By improving drug delivery, targeting, and immune modulation, nanoplatforms can unlock the full potential of cancer immunotherapy. This review explores the role of nanoplatforms in addressing these limitations and enhancing cancer immunotherapy outcomes, examining various types of nanoplatforms. Understanding the mechanisms of immunomodulation through nanoplatform deliveries is crucial. We discuss how these nanoplatforms interact with the tumor microenvironment, modulate tumor-associated macrophages and regulatory T cells, activate immune cells directly, enhance antigen presentation, and promote immunological memory. Further benefits include combination approaches integrating nanoplatforms with chemotherapy, radiotherapy, and phototherapy. Immunotherapy is a relatively new approach, but numerous clinical studies already utilize nanoplatform-based immunotherapies with promising results. This review aims to provide insights into the potential of nanoplatforms to enhance cancer immunotherapy and pave the way for more effective and personalized treatment strategies.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1321-1338"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12140459/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144063429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bo-Dou Zhang, Xi Chen, Jing-Yun Su, Shao-Hua Zhuo, Lang Zhao, Jun-Jun Wu, Wen-Hao Li, Tian-Yang Wang, Ling Liu, Tao Yang, Li-Jun Yang, Yu-Fen Zhao, Yan-Mei Li
{"title":"Rationally designed anti-autophagy nanosystems for reversing the immunosuppressive network in the tumor environment.","authors":"Bo-Dou Zhang, Xi Chen, Jing-Yun Su, Shao-Hua Zhuo, Lang Zhao, Jun-Jun Wu, Wen-Hao Li, Tian-Yang Wang, Ling Liu, Tao Yang, Li-Jun Yang, Yu-Fen Zhao, Yan-Mei Li","doi":"10.1080/17435889.2025.2508133","DOIUrl":"10.1080/17435889.2025.2508133","url":null,"abstract":"<p><strong>Aims: </strong>To develop a nano-immunotherapy system combining autophagy inhibition and innate immune activation to reverse the immunosuppressive tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC).</p><p><strong>Materials & methods: </strong>The pH-responsive polymer PC7A was utilized to co-deliver the autophagy inhibitor chloroquine (CQ) and the STING agonist cyclic diguanylate (CDG), forming the CQCP nanosystem. In vitro and in vivo experiments evaluated autophagy inhibition, MHC-I expression, dendritic cell activation, tumor infiltration of lymphocytes, and survival in PDAC-bearing mice.</p><p><strong>Results: </strong>CQCP enhanced MHC-I expression on PDAC cells by 2.1-fold (<i>p</i> < 0.001) and increased activated dendritic cells (CD86+/CD40+) by 3.5-fold (<i>p</i> < 0.01) in the TME. Tumor-infiltrating CD8+ T cells rose by 42.6% (<i>p</i> < 0.001), and systemic immune activation in peripheral lymphoid tissues was observed. CQCP achieved an 86% survival rate in tumor-bearing mice, significantly outperforming monotherapies or free drug combinations.</p><p><strong>Conclusions: </strong>The CQCP system synergistically reverses PDAC immunosuppression by restoring antigen presentation and activating innate immunity. This dual-targeted strategy demonstrates robust antitumor efficacy and offers a promising immunotherapy approach for PDAC.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1429-1440"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12143676/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144121516","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Anticancer efficacy of dual-loaded SLNs with Doxorubicin (DOX) and Pterostilbene (PTS): formulation, characterization, and evaluation for breast cancer.","authors":"Harneet Marwah, Janmejay Pant, Kamal Shah, Perwez Alam, Hitesh Kumar Dewangan","doi":"10.1080/17435889.2025.2501526","DOIUrl":"10.1080/17435889.2025.2501526","url":null,"abstract":"<p><strong>Aims: </strong>This study aimed to develop optimized doxorubicin (DOX) and pterostilbene (PTS) co-loaded solid lipid nanoparticles (SLNs) for synergistic triple-negative breast cancer (TNBC) therapy, enhancing drug stability, tumor targeting, and therapeutic efficacy.</p><p><strong>Materials & methods: </strong>Calibration curves for DOX and PTS were validated. Synergy was assessed in MDA-MB-231 cells via Combination Index (CI) and Loewe-HSA models. SLNs were optimized using Box-Behnken Design (BBD), evaluating lipid content, surfactant concentration, and sonication time. Formulations were characterized by Zetasizer, high-resolution transmission electron microscopy (HR-TEM), Fourier-transform infrared (FTIR), X-ray powder diffraction (XRD), and differential scanning calorimetry (DSC). In in-vitro cytotoxicity, Reactive Oxygen Species (ROS) generation, apoptosis, and mitochondrial depolarization were assessed. Pharmacokinetics and tumor regression were studied in rats.</p><p><strong>Results: </strong>The optimal 1:4 (DOX:PTS) ratio showed strong synergy (CI = 0.83). BBD-optimized SLNs had 97.92 nm size, high entrapment (DOX: 54.%; PTS: 77.5%), and pH-responsive release (78.78% DOX at pH 5.5). SLNs exhibited enhanced cytotoxicity (IC₅₀ = 0.833 µg/mL), elevated ROS (59.5%), and apoptosis induction. In in-vivo, SLNs prolonged circulation, increased tumor accumulation, and reduced tumor volume (701.50 ± 11.83 mm<sup>3</sup> vs. 3506.58 ± 17.06 mm<sup>3</sup> control).</p><p><strong>Conclusions: </strong>DOX-PTS SLNs demonstrated synergistic anticancer effects, improved stability, and targeted delivery, offering a promising strategy for TNBC treatment.</p>","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1249-1265"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12140452/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143999682","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Magalí Di Meglio, Daniela Maza Vega, Jorge Montanari
{"title":"How could the translation of nanotechnology-based therapies revolutionize cutaneous leishmaniasis treatments?","authors":"Magalí Di Meglio, Daniela Maza Vega, Jorge Montanari","doi":"10.1080/17435889.2025.2485024","DOIUrl":"10.1080/17435889.2025.2485024","url":null,"abstract":"","PeriodicalId":74240,"journal":{"name":"Nanomedicine (London, England)","volume":" ","pages":"1377-1380"},"PeriodicalIF":0.0,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12143689/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143733456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}