International Journal of Nanomedicine最新文献

筛选
英文 中文
CD44-Receptors-Mediated Multiprong Targeting Strategy Against Breast Cancer and Tumor-Associated Macrophages: Design, Optimization, Characterization, and Cytologic Evaluation.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-25 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S480553
Zahid Hussain, Lama Abdulrahim Abdul Moti, Jayalakshmi Jagal, Hnin Ei Thu, Shahzeb Khan, Mohsin Kazi
{"title":"CD44-Receptors-Mediated Multiprong Targeting Strategy Against Breast Cancer and Tumor-Associated Macrophages: Design, Optimization, Characterization, and Cytologic Evaluation.","authors":"Zahid Hussain, Lama Abdulrahim Abdul Moti, Jayalakshmi Jagal, Hnin Ei Thu, Shahzeb Khan, Mohsin Kazi","doi":"10.2147/IJN.S480553","DOIUrl":"10.2147/IJN.S480553","url":null,"abstract":"<p><strong>Introduction: </strong>Owing to its high prevalence, colossal potential of chemoresistance, metastasis, and relapse, breast cancer (BC) is the second leading cause of cancer-related fatalities in women. Several treatments (eg, chemotherapy, surgery, radiations, hormonal therapy, etc.) are conventionally prescribed for the treatment of BC; however, these are associated with serious systemic aftermaths. In this research, we aimed to design a multiprong targeting strategy for concurrent action against different phenotypes of BC (MCF-7 and SK-BR-3) and tumor-associated macrophages (TAMs) for relapse-free treatment of BC.</p><p><strong>Methods: </strong>Paclitaxel (PTX) and tamoxifen (TMX) co-loaded chitosan (CS) nanoparticles (NPs) were prepared using the ionic-gelation method and optimized using the Design Expert<sup>®</sup> software by controlling different material attributes. For selective targeting through CD44-receptors that are heavily expressed on the BC cells and TAMs, the fabricated NPs (PTX-TMX-CS-NPs) were functionalized with hyaluronic acid (HA) as a targeting ligand.</p><p><strong>Results: </strong>The optimized HA-PTX-TMX-CS-NPs exhibited desired physicochemical properties (PS ~230 nm, PDI 0.30, zeta potential ~21.5 mV), smooth spherical morphology, high encapsulation efficiency (PTX ~72% and TMX ~97%), good colloidal stability, and biphasic release kinetics. Moreover, the lowest cell viability depicted in MCF-7 (~25%), SK-BR-3 (~20%), and RAW 264.7 cells (~20%), induction of apoptosis, cell cycle arrest, enhanced cell internalization, and alleviation of MCF-7 and SK-BR-3 migration proved the superior anticancer potential of HA-PTX-TMX-CS-NPs compared to unfunctionalized NPs and other control medicines.</p><p><strong>Conclusion: </strong>HA-functionalization of NPs is a promising multiprong strategy for CD44-receptors-mediated targeting of BC cells and TAMs to mitigate the progression, metastasis, and relapse in the BC.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"991-1020"},"PeriodicalIF":6.6,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11776558/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143065455","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Solid Self-Microemulsifying Drug Delivery System for Improved Oral Bioavailability of Relugolix: Preparation and Evaluation.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-25 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S497099
Zi-Lin Li, Guo-Xing Deng, Chuan-Zhou Fang, Yue-Qi Zhao, Jing Yuan, Liang Chen, Hai-Jun Zhong, Feng Guo
{"title":"Solid Self-Microemulsifying Drug Delivery System for Improved Oral Bioavailability of Relugolix: Preparation and Evaluation.","authors":"Zi-Lin Li, Guo-Xing Deng, Chuan-Zhou Fang, Yue-Qi Zhao, Jing Yuan, Liang Chen, Hai-Jun Zhong, Feng Guo","doi":"10.2147/IJN.S497099","DOIUrl":"10.2147/IJN.S497099","url":null,"abstract":"<p><strong>Purpose: </strong>To improve the oral absorption of relugolix (RLGL), which has low oral bioavailability due to its low solubility and being a substrate of P-glycoprotein (P-gp). A solid self-microemulsifying drug delivery system of relugolix (RLGL-S-SMEDDS) was prepared and evaluated in vitro and in vivo.</p><p><strong>Methods: </strong>The composition of the solid self-microemulsifying drug delivery system (S-SMEDDS) was selected by solubility study and pseudo-ternary phase diagram, and further optimized by Design-Expert optimization design. The optimized RLGL-S-SMEDDS were evaluated in terms of particle size, zeta potential, morphology analysis, thermodynamic stability, drug release, flow properties, transporter pathways in Caco-2 cells, the influence of excipients on the intestinal transporters, transport within Caco-2 cell monolayers and transport in lymphocyte. In vivo pharmacokinetic study and toxicological study were also conducted.</p><p><strong>Results: </strong>The optimum formulation for self-microemulsifying drug delivery system (SMEDDS) consists of Ethyl Oleate (26% of the weight), Solutol HS15 (49% of the weight), Transcutol HP (25% of the weight) and loaded relugolix (4.8 mg/g). The S-SMEDDS was then formed by adsorbing 2.4 g of SMEDDS onto 1 g of hydrophilic-200 silica. In phosphate buffered saline (PBS) (pH 6.8) release medium containing 1% tween 80, the vitro release studies showed 86% cumulative drug release for RLGL-S-SMEDDS and 3.6% cumulative drug release for RLGL suspensions. In vitro cellular uptake experiments revealed that the uptake of RLGL-S-SMEDDS by Caco-2 cells was three times higher than that of free RLGL, and that S-SMEDDS can enhance the drug absorption through lymphatic absorption and inhibition of intestinal transporter. In vivo pharmacokinetic evaluation demonstrated that the oral bioavailability of RLGL-S-SMEDDS was 1.9 times higher than that of RLGL-suspensions. There was no apparent cardiac, hepatic, splenic, pulmonary or renal toxicity on the surface discovered by pathological analysis after oral administration.</p><p><strong>Conclusion: </strong>It is evident that S-SMEDDS may be a safe and effective method to improve oral absorption of drugs with low oral bioavailability.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"1065-1082"},"PeriodicalIF":6.6,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11780666/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143065456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LBP-CD155 Liposome Nanovaccine Efficiently Resist Colorectal Cancer and Enhance ICB Therapy.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-25 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S492734
Yajuan Yan, Ting Duan, Xiaonan Xue, Xiaojuan Yang, Miao Liu, Bin Ma, Xiangguo Duan, Chunxia Su
{"title":"LBP-CD155 Liposome Nanovaccine Efficiently Resist Colorectal Cancer and Enhance ICB Therapy.","authors":"Yajuan Yan, Ting Duan, Xiaonan Xue, Xiaojuan Yang, Miao Liu, Bin Ma, Xiangguo Duan, Chunxia Su","doi":"10.2147/IJN.S492734","DOIUrl":"10.2147/IJN.S492734","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) is a highly malignant and aggressive gastrointestinal tumor. Due to its weak immunogenicity and limited immune, cell infiltration lead to ineffective clinical outcomes. Therefore, to improve the current prophylaxis and treatment scheme, offering a favorable strategy efficient against CRC is urgently needed.</p><p><strong>Methods: </strong>Here, we developed a nanovaccine (LBP-CD155L NVs) loaded with CD155 gene in liposome, which was modified by Lycium barbarum polysaccharides (LBP) through electrostatic interaction. The nanovaccine was characterized by transmission electron microscopy and Zetasizer. It was evaluated in vitro, where NVs facilitated the endocytosis and maturation of DCs, and in vivo, where NVs improved the efficacy of prophylaxis and therapy. In addition, further confirmed the mechanisms by how TLR4 and MGL synergistic pathway endow the nanovaccines towards dendritic cells (DCs). Finally, the safety and tumor immunosuppressive microenvironment were evaluated in the CRC tumor-bearing mouse model.</p><p><strong>Results: </strong>We successfully developed a nanovaccine that facilitates the endocytosis and maturation of DCs via a synergistic pathway involving TLR4 and MGL, which endow the nanovaccines towards dendritic cells (DCs) and promote the differentiation, thereby enhancing the cytotoxicity of CD8<sup>+</sup>T cells. Consequently, LBP-CD155L NVs can potentiate the efficacy of prophylactic and therapeutic administration in a mouse CRC model, as evidenced by decreased infiltration of myeloid-derived suppressor cells (MDSCs) and Tregs, reprogrammed the macrophage phenotypes, which promoted polarization from M2-like macrophages to M1-like macrophages, increased infiltration of effector T cells. Prophylactic and therapeutic combination regimens with anti-PD-1 treatment demonstrate synergism that stimulates T-cell infiltration into tumors and counteracts immunosuppression, leading to remarkable tumor remission and enhancing the efficacy of immune checkpoint therapy in solid tumors.</p><p><strong>Conclusion: </strong>Our work provided that LBP-CD155L NVs may serve as a promising tool for reversing tumor immunosuppressive microenvironment and enhancing ICB therapy in CRC.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"1047-1063"},"PeriodicalIF":6.6,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11773180/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059041","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhanced Antitumor Efficacy and Reduced Toxicity in Colorectal Cancer Using a Novel Multifunctional Rg3- Targeting Nanosystem Encapsulated with Oxaliplatin and Calcium Peroxide.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-24 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S502076
Yizhuo Xie, Ming Zhu, Han Bao, Kejia Chen, Shanshan Wang, Jingwen Dai, Hongzhu Chen, He Li, Qi Song, Xinlu Wang, Liangping Yu, Jin Pei
{"title":"Enhanced Antitumor Efficacy and Reduced Toxicity in Colorectal Cancer Using a Novel Multifunctional Rg3- Targeting Nanosystem Encapsulated with Oxaliplatin and Calcium Peroxide.","authors":"Yizhuo Xie, Ming Zhu, Han Bao, Kejia Chen, Shanshan Wang, Jingwen Dai, Hongzhu Chen, He Li, Qi Song, Xinlu Wang, Liangping Yu, Jin Pei","doi":"10.2147/IJN.S502076","DOIUrl":"10.2147/IJN.S502076","url":null,"abstract":"<p><strong>Purpose: </strong>Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Oxaliplatin (OXA) is currently the primary chemotherapeutic agent for CRC, but its efficacy is limited by the tumor microenvironment (TME). Here, we present a combined approach of chemotherapy and TME modulation for CRC treatment. A multifunctional nanosystem (Rg3-Lip-OXA/CaO<sub>2</sub>) was established using Ginsenoside Rg3 liposomes targeting glucose transporter 1 overexpressed on the surface of CRC cells to co-deliver OXA and calcium peroxide (CaO<sub>2</sub>).</p><p><strong>Methods: </strong>The CaO<sub>2</sub> nanoparticles were synthesized via the CaCl<sub>2</sub>-H<sub>2</sub>O<sub>2</sub> reaction under alkaline conditions and characterized using X-ray diffraction (XRD) and X-ray photoelectron spectroscopy (XPS). Rg3-Lip-OXA/CaO<sub>2</sub> was prepared through a thin-film hydration approach and characterized; additionally, its stability and release behavior were studied. The O<sub>2</sub>, H<sub>2</sub>O<sub>2</sub>, and Ca<sup>2+</sup> generation ability of Rg3-Lip-OXA/CaO<sub>2</sub> in solution and HCT116 cells were measured. The in vitro cellular uptake was observed via fluorescence microscope and flow cytometry. In vitro cytotoxicity was evaluated using the CCK-8 assay, flow cytometry, and live/dead cell staining. The in vivo targeting effect as well as antitumor efficacy were determined in HCT116 tumor-bearing mice. Finally, the acute toxicity of Rg3-Lip-OXA/CaO<sub>2</sub> was investigated in ICR mice to explore its safety.</p><p><strong>Results: </strong>The XRD and XPS analyses confirmed the successful synthesis of CaO<sub>2</sub> nanoparticles. The Rg3-Lip-OXA/CaO<sub>2</sub> exhibited an average particle size of approximately 92.98 nm with good stability and sustained release behavior. In vitro and in vivo studies confirmed optimal targeting by Rg3-Lip and demonstrated that the nanosystem effectively produced O<sub>2</sub>, H<sub>2</sub>O<sub>2</sub> and Ca<sup>2+</sup>, resulting in significant cytotoxicity. Additionally, in vivo studies revealed substantial tumor growth suppression and reduced tumor-associated fibroblasts (TAFs) and collagen. Acute toxicity studies indicated that Rg3-Lip-OXA/CaO<sub>2</sub> markedly reduced the toxicity of chemotherapeutic drugs.</p><p><strong>Conclusion: </strong>This multifunctional nanosystem enhances chemotherapy efficacy and reduces toxicity, offering a promising approach for optimizing CRC treatment and potential clinical application.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"1021-1046"},"PeriodicalIF":6.6,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11774109/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nanoparticle Therapies: Targeted Treatment for Bladder Cancer With Reduced Side Effects [Letter].
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-22 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S513952
Jingguo Sun
{"title":"Nanoparticle Therapies: Targeted Treatment for Bladder Cancer With Reduced Side Effects [Letter].","authors":"Jingguo Sun","doi":"10.2147/IJN.S513952","DOIUrl":"10.2147/IJN.S513952","url":null,"abstract":"","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"989-990"},"PeriodicalIF":6.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11766204/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046620","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chondroitin Sulphate-Chitosan Based Nanogels Loaded with Naringenin-β-Cyclodextrin Complex as Potential Tool for the Treatment of Diabetic Retinopathy: A Formulation Study.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-22 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S488507
Gaia Zucca, Barbara Vigani, Caterina Valentino, Marco Ruggeri, Nicoletta Marchesi, Alessia Pascale, Giulia Giovilli, Lorenzo Malavasi, Giuseppina Sandri, Silvia Rossi
{"title":"Chondroitin Sulphate-Chitosan Based Nanogels Loaded with Naringenin-β-Cyclodextrin Complex as Potential Tool for the Treatment of Diabetic Retinopathy: A Formulation Study.","authors":"Gaia Zucca, Barbara Vigani, Caterina Valentino, Marco Ruggeri, Nicoletta Marchesi, Alessia Pascale, Giulia Giovilli, Lorenzo Malavasi, Giuseppina Sandri, Silvia Rossi","doi":"10.2147/IJN.S488507","DOIUrl":"10.2147/IJN.S488507","url":null,"abstract":"<p><strong>Purpose: </strong>The main purpose of the study was the formulation development of nanogels (NHs) composed of chondroitin sulfate (CS) and low molecular weight chitosan (lCH), loaded with a naringenin-β-cyclodextrin complex (NAR/β-CD), as a potential treatment for early-stage diabetic retinopathy.</p><p><strong>Methods: </strong>Different formulations of NHs were prepared by varying polymer concentration, lCH ratio, and pH and, then, characterized for particle size, zeta potential, particle concentration (particles/mL) and morphology. Cytotoxicity and internalization were assessed in vitro using Human Umbilical Vein Endothelial Cells (HUVEC). The NAR/β-CD complex was prepared and evaluated for morphology, complexation efficiency, and solubility. Finally, the most promising NH prototype was loaded with NAR/β-CD (NH@NAR/β-CD) and further characterized for encapsulation efficiency, loading capacity, opacity and cytotoxicity on HUVEC; in vitro release test and DPPH assay were performed to investigate NH capability to sustain NAR release and NH@NAR/β-CD antioxidant properties, respectively.</p><p><strong>Results: </strong>NH properties were influenced by polymer concentration, lCH ratio, and pH. N3 (0.5 mg/mL; lCH=1.5:1; pH = 5) and N9 (0.5 mg/mL; lCH=1:1; pH = 5) showed optimal characteristics, including small size (<350 nm) and positive zeta potential, facilitating cellular uptake. The NAR/β-CD complex showed 71% complexation efficiency and enhanced NAR solubility. Since characterized by superior properties and better in vitro biocompatibility, N3 was loaded with NAR/β-CD. N3@NAR/β-CD capability to sustain in vitro NAR release, radical scavenging activity and in vitro biocompatibility were finally demonstrated.</p><p><strong>Conclusion: </strong>The physico-chemical properties of N3@NAR/β-CD were responsible for their cell uptake, suggesting their potential to target retinal endothelial cells. The high NAR/β-CD complexation efficiency and the sustained NAR release over 72 hours could guarantee the maintenance of an effective drug concentration at the damage site while reducing the injection number. Further studies about the safety and the effectiveness of the intravitreal injection of NHs@NAR/β-CD will be performed on a diabetic animal model.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"907-932"},"PeriodicalIF":6.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11766310/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted Delivery of Nanovaccine to Dendritic Cells via DC-Binding Peptides Induces Potent Antiviral Immunity in vivo [Expression of Concern].
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-22 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S518089
{"title":"Targeted Delivery of Nanovaccine to Dendritic Cells via DC-Binding Peptides Induces Potent Antiviral Immunity in vivo [Expression of Concern].","authors":"","doi":"10.2147/IJN.S518089","DOIUrl":"https://doi.org/10.2147/IJN.S518089","url":null,"abstract":"","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"967-968"},"PeriodicalIF":6.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11772996/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059043","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combined Graphene Oxide with 2-Methoxyestradiol for Effective Anticancer Therapy in-vitro Model.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-22 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S498947
Katarzyna Uzdrowska, Narcyz Knap, Lucyna Konieczna, Anna Kamm, Alicja Kuban-Jankowska, Joanna Gierałtowska, Mariusz Belka, Magdalena Baran, Adrian Chlanda, Krystian Michał Kowiorski, Aleksander Żołnierski, Jacek Gulczynski, Ludwika Lipińska, Tomasz Bączek, Ewa Izycka-Swieszewska, Magdalena Górska-Ponikowska
{"title":"Combined Graphene Oxide with 2-Methoxyestradiol for Effective Anticancer Therapy in-vitro Model.","authors":"Katarzyna Uzdrowska, Narcyz Knap, Lucyna Konieczna, Anna Kamm, Alicja Kuban-Jankowska, Joanna Gierałtowska, Mariusz Belka, Magdalena Baran, Adrian Chlanda, Krystian Michał Kowiorski, Aleksander Żołnierski, Jacek Gulczynski, Ludwika Lipińska, Tomasz Bączek, Ewa Izycka-Swieszewska, Magdalena Górska-Ponikowska","doi":"10.2147/IJN.S498947","DOIUrl":"10.2147/IJN.S498947","url":null,"abstract":"<p><strong>Introduction: </strong>This article describes the invention of graphene oxide (GO) or reduced graphene oxide (rGO) functionalised with 2-methoxy estradiol. The presence of polar hydroxyl groups enables the binding of 2-ME to GO/rGO through hydrogen bonds with epoxy and hydroxyl groups located on the surface and carbonyl and carboxyl groups located at the edges of graphene flake sheets.</p><p><strong>Methods: </strong>The patented method of producing the subject of the invention and the research results regarding its anticancer effectiveness via cytotoxicity in an in vivo model (against A375 melanoma and 143B osteosarcoma cells) are described.</p><p><strong>Results: </strong>It was shown that the inhibition of PTP1B phosphotyrosine phosphatase is one of the mechanisms of action of GO functionalised with 2-ME (GO-2-ME). This is a very important result, considering the fact that 2-ME itself has no inhibitory properties against this phosphatase.</p><p><strong>Discussion: </strong>Graphene oxide flakes embroidered with 2-methoxyestradiol molecules may be a promising solution, bringing a new and important effect in the form of improving the bioavailability of the therapeutic substance, ie 2-ME. An appropriate dosage of GO-2-ME/rGO-2-ME, in which GO/rGO is a carrier of 2-methoxyestradiol (2-ME), can ensure effective penetration of the active substance through biological boundaries/membranes and controlled modification of cell signalling, ultimately leading to the selective elimination of malignant cells.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"933-950"},"PeriodicalIF":6.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11756907/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143028776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Multifunctional MIL-101-NH2(Fe) Nanoplatform for Synergistic Melanoma Therapy.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-22 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S502089
Jinlu Shang, Yongjun Chen, Fangliang Wang, Jing Yang, Yi Li, Liuxuan Yang, Xiuqiong Liu, Zhirong Zhong, Chaochi Yue, Meiling Zhou
{"title":"A Multifunctional MIL-101-NH<sub>2</sub>(Fe) Nanoplatform for Synergistic Melanoma Therapy.","authors":"Jinlu Shang, Yongjun Chen, Fangliang Wang, Jing Yang, Yi Li, Liuxuan Yang, Xiuqiong Liu, Zhirong Zhong, Chaochi Yue, Meiling Zhou","doi":"10.2147/IJN.S502089","DOIUrl":"10.2147/IJN.S502089","url":null,"abstract":"<p><strong>Background: </strong>Melanoma is an aggressive form of skin cancer, and single-modality treatments often fail to prevent tumor recurrence and metastasis. Combination therapy has emerged as an effective approach to improve treatment outcomes.</p><p><strong>Methods: </strong>In this study, we developed a multifunctional nanoplatform, MIL@DOX@ICG, utilizing MIL-101-NH<sub>2</sub>(Fe) as a carrier to co-deliver the chemotherapeutic agent doxorubicin (DOX) and the photosensitizer indocyanine green (ICG). MIL-101-NH<sub>2</sub>(Fe) was synthesized via a hydrothermal method. Drug release was evaluated under different pH conditions, and the photothermal effect was tested under near-infrared (NIR) laser irradiation. Hydroxyl radical and reactive oxygen species generation capacities were quantified. Cellular studies using B16F10 cells assessed cytotoxicity, cellular uptake, migration inhibition, and colony formation suppression. In vivo experiments in melanoma-bearing mice evaluated antitumor efficacy and systemic safety through tumor growth inhibition, histological analyses, and toxicity assessments.</p><p><strong>Results: </strong>MIL@DOX@ICG exhibited a uniform octahedral structure with a particle size of approximately 139 nm and high drug loading efficiencies for DOX (33.70%) and ICG (30.59%). The nanoplatform demonstrated pH-responsive drug release and potent photothermal effects. The generation of hydroxyl radicals via the Fenton reaction and reactive oxygen species production under NIR laser irradiation by MIL@DOX@ICG were confirmed. In vitro assessments revealed significant cytotoxicity of MIL@DOX@ICG against B16F10 cells under NIR laser irradiation, with improved efficacy in inhibiting cell proliferation and migration. In vivo studies confirmed the superior antitumor efficacy of MIL@DOX@ICG + NIR treatment, synergistically harnessing chemotherapy, photothermal therapy, photodynamic therapy, and chemodynamic therapy effects while maintaining excellent biocompatibility.</p><p><strong>Conclusion: </strong>Our findings underscore the potential of MIL-101-NH<sub>2</sub>(Fe) nanoparticles as a versatile and effective platform for synergistic melanoma therapy, offering a promising strategy for overcoming the limitations of conventional treatment modalities.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"969-988"},"PeriodicalIF":6.6,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11766718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046601","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Conglomerated Imiquimod and Metronidazole Incorporated Biodegradable Nanofibrous Mats for Potential Therapy of Cervical Cancer.
IF 6.6 2区 医学
International Journal of Nanomedicine Pub Date : 2025-01-21 eCollection Date: 2025-01-01 DOI: 10.2147/IJN.S482290
Yi-Pin Chen, Chiao-Fan Chiu, Chien-Neng Wang, Chu-Chi Lin, Chia-Rui Shen, Yi-Chen Yao, Yi-Hua Kuo, Shih-Jung Liu
{"title":"Conglomerated Imiquimod and Metronidazole Incorporated Biodegradable Nanofibrous Mats for Potential Therapy of Cervical Cancer.","authors":"Yi-Pin Chen, Chiao-Fan Chiu, Chien-Neng Wang, Chu-Chi Lin, Chia-Rui Shen, Yi-Chen Yao, Yi-Hua Kuo, Shih-Jung Liu","doi":"10.2147/IJN.S482290","DOIUrl":"10.2147/IJN.S482290","url":null,"abstract":"<p><strong>Background: </strong>In clinical practice, imiquimod is used to treat Human Papillomavirus (HPV)-related lesions, such as condyloma and Cervical Intraepithelial Neoplasia (CIN). Metronidazole is the most commonly prescribed antibiotic for bacterial vaginosis. The study developed biodegradable imiquimod- and metronidazole-loaded nanofibrous mats and assessed their effectiveness for the topical treatment of cervical cancer, a type of HPV-related lesion.</p><p><strong>Methods: </strong>Nanofibers of two distinct poly[(d,l)-lactide-<i>co</i>-glycolide] (PLGA)-to-drug ratios (6:1 and 4:1) were manufactured through the electrospinning technology. The in vitro release behavior of imiquimod and metronidazole was evaluated via an elution method, while the in vivo discharge behavior was evaluated on a mice model. Additionally, a model of cervical cancer was established using C57BL/6J mice, and it was utilized to evaluate the efficacy of drug-eluting nanofibers through in vivo testing. Mice afflicted with cervical cancer were separated into three distinct groups for the study: The mice in Group A served as the control and received no treatment. Group B received treatment with pure PLGA nanofibers (no drugs loaded), whereas Group C received treatment with nanofibers loaded with imiquimod and metronidazole. Post implantation, the variations in tumor sizes of rats receiving the implantation of drug-eluting nanofibers were monitored.</p><p><strong>Results: </strong>The experimental data show that drug-eluting nanofibers could discharge in vitro high concentrations of imiquimod and metronidazole for exceeding 30 days. In vivo, each membrane consistently released elevated concentrations of imiquimod/metronidazole at the intended site in mice over a four-week period, with minimal systemic drug concentration detected in the bloodstream. The mice treated with drug-loaded nanofibers displayed noticeably reduced tumor volumes compared to both the control group and the group treated with pristine nanofibers. Histological examination revealed the absence of any discernible tissue inflammation.</p><p><strong>Conclusion: </strong>Biodegradable nanofibers with a sustainable release of imiquimod and metronidazole demonstrated their effectiveness and lasting impact of treating mice with cervical cancer.</p>","PeriodicalId":14084,"journal":{"name":"International Journal of Nanomedicine","volume":"20 ","pages":"951-966"},"PeriodicalIF":6.6,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11762495/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信