Iosif Mikhel, Elena Bakhrushina, Olga Stepanova, Sofiya Prilepskaya, Dmitriy Kosenkov, Anastasia Belyatskaya, Grigory Evzikov, Natalia Demina, Ivan Krasnyuk, Ivan Krasnyuk
{"title":"Ribavirin in Modern Antitumor Therapy: Prospects for Intranasal Administration.","authors":"Iosif Mikhel, Elena Bakhrushina, Olga Stepanova, Sofiya Prilepskaya, Dmitriy Kosenkov, Anastasia Belyatskaya, Grigory Evzikov, Natalia Demina, Ivan Krasnyuk, Ivan Krasnyuk","doi":"10.2174/0115672018305548240614113451","DOIUrl":"https://doi.org/10.2174/0115672018305548240614113451","url":null,"abstract":"<p><p>Ribavirin has been used as an antiviral agent to treat a variety of viral infections since the 1970s. Over the past few decades, studies have been conducted on the pharmacology of ribavirin, and the possibility of its use in new indications has been explored. According to the results of a number of studies, ribavirin efficacy in the therapy of malignant neoplasms of various genesis has been proven. Furthermore, due to the complexity of brain tumor therapy using surgical methods, targeted delivery of ribavirin to the brain becomes a promising alternative to existing treatment methods. Targeting of active pharmaceutical ingredient (API) to the brain tumor is achieved by intranasal drug delivery via a Nose-to-Brain mechanism. In addition, using this delivery mechanism, it is possible to reach the brain while bypassing the blood-brain barrier (BBB), thus avoiding the effects of the first passage through the liver. Despite the significant advantages of the method, there are limiting factors to its application - mucociliary clearance, which aims to remove foreign bodies from the surface of the nasal mucosa. In situ, systems are able to reduce the intensity of interfering factors on API and allow the achievement of maximum bioavailability during intranasal administration.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142083027","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiangtao Su, Xue Chen, Fan Ye, Chuchu Liu, Jiahao Liang, Xuejun Zhang, Xiaoxia Guo
{"title":"Celastrol Derivative/DOX Co-Assembled Nanodrug for Enhanced Antitumor Therapy.","authors":"Jiangtao Su, Xue Chen, Fan Ye, Chuchu Liu, Jiahao Liang, Xuejun Zhang, Xiaoxia Guo","doi":"10.2174/0115672018298512240819101159","DOIUrl":"https://doi.org/10.2174/0115672018298512240819101159","url":null,"abstract":"<p><strong>Background: </strong>Multidrug resistance (MDR) is a key challenge in clinical chemotherapy. The combination of drugs can effectively reverse multi-drug resistance.</p><p><strong>Objective: </strong>In this study, doxorubicin (DOX) was capsulated into nanoparticles formed by an amphiphilic PEGylated-poly (α-lipoic acid)-methanamide analogue of celastrol (mPEG-PαLA-CEN) prodrug polymer. CEN was linked to the branched chain of poly (α-lipoic acid) by forming ester bonds. DOX was physically trapped inside the nanoparticles via electrostatic interaction. Both drugs can be simultaneously released in response to low pH and high GSH in order to overcome DOX resistance.</p><p><strong>Methods: </strong>The chemical structure of the mPEG-PαLA-CEN-DOX NPs was confirmed through 1H NMR, FT-IR spectroscopy, UV-Vis spectrum, DLS, and TEM. Drug-loading content, efficacy, and drug release were measured using HPLC. Cell toxicity was examined using an MTT assay.</p><p><strong>Results: </strong>CEN/DOX-loaded nanoparticles were found to have spherical shapes with diameters of around 229.7 nm. The NPs exhibited high biocompatibility and released 92% DOX and 71.8% CEN in response to low pH and high GSH of tumor microenvironments. As dual drug-loaded nanoparticles, the efficacy of mPEG-PαLA-CEN-DOX NPs against tumor cell lines in vitro was enhanced for both MCF-7 and MCF-7/ADR compared to free DOX. Compared to free DOX, the IC50 of mPEG-PαLA-CEN-DOX NPs reduced from 46.10 μM to 8.36 μM for the MCF-7/ADR cell line.</p><p><strong>Conclusion: </strong>In conclusion, this study demonstrated that PEGylated poly (α-lipoic acid)-CEN copolymers can be used not only as biocompatible, stimulation-responsive anticancer drug nanocarriers but also as chemosensitizers to overcome multidrug resistance, which provide a theoretical base for clinical application of CEN/DOX nanodrug.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142083026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Nanocrystals: Versatile Platform for Traditional Chinese Medicine Delivery.","authors":"Chuanyu Ren, Yue Gao, Yeqi Huang, Siyuan Peng, Xuan Zhang, Weijie Wang, Chuanbin Wu, Xin Pan, Zhengwei Huang","doi":"10.2174/0115672018322054240813112111","DOIUrl":"https://doi.org/10.2174/0115672018322054240813112111","url":null,"abstract":"<p><p>The medicinal value of Chinese medicines has been recognized since ancient times, and they have also been used to treat various diseases. However, in-depth studies on the active ingredients of Chinese medicines have shown that many of them suffer from poor water-solubility, stability, and bioavailability, which has severely limited their further development. The advent of nanomedicine represents a novel direction and paradigm for addressing these challenges. Particularly, within the framework of nanocrystal technology, enhancements in the water solubility, stability, and bioavailability of Chinese medicines are expected to significantly improve the therapeutic efficiency. This advancement also holds promise for unlocking new therapeutic capabilities. Nanocrystals offer significant advantages in oral, intravenous, intranasal and targeted delivery. The drug loading principle is \"all in one\", with hydrophobic-drug-in and hydrophilic-drug-out and stabilization by amphiphilic agents. Nanocrystal technology in traditional Chinese medicine (TCM) holds extensive application potential. Continuous refinement of preparation techniques, sound safety assessments, and the promotion of large-scale production are anticipated to augment its pivotal role in TCM formulations, thereby creating novel opportunities for clinical drug therapy.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142020080","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Conjugated Linoleic Acid in Cancer Therapy.","authors":"Jeneesha George, Asit Ranjan Ghosh","doi":"10.2174/0115672018325362240811164655","DOIUrl":"https://doi.org/10.2174/0115672018325362240811164655","url":null,"abstract":"<p><p>Conjugated Linoleic Acid (CLA) is a polyunsaturated dietary fatty acid. Probiotics can biohydrogenate CLA with multiple health benefits, especially in cancer treatment. In vitro, in vivo, and clinical studies have confirmed CLA isomers to possess anti-cancer activity. CLA has demonstrated its potential as an alternative treatment for cancer and also used as an adjuvant to reduce the side effects of existing treatment methods. The mechanism of the anticancer activity of CLA is still not clear; however, it may involve intervention with the cell cycle and modulation of gene expression. A greater potential of CLA for cancer treatment has been supported by more and more clinical trials to evaluate its potential. Some advanced technologies are in progress to overcome the flaws of current methods and enhance the microbial production of CLA. In conclusion, nutritional enrichment as a functional food and direct consumption of CLA may contribute to cancer management.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141989875","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Myristic Acid Solid Lipid Nanoparticles Enhance the Oral Bioavailability and Therapeutic Efficacy of Rifaximin against MRSA Pneumonia.","authors":"Yumin Zhang, Aoxue Zhang, Dongmei Chen, Shuyu Xie","doi":"10.2174/0115672018276382231207103955","DOIUrl":"https://doi.org/10.2174/0115672018276382231207103955","url":null,"abstract":"<p><strong>Background: </strong>Methicillin-resistant Staphylococcus aureus (MRSA) pneumonia is one of the leading causes of death and an immense financial burden on healthcare systems. Rifaximin (RFX) has good antibacterial activity against MRSA, but its clinical application is limited due to its poor oral absorption. Solid lipid nanoparticles have good biocompatibility, high drug loading, sustained release performance, and the inertia of lipids in gastric acid, which facilitates oral drug delivery.</p><p><strong>Objective: </strong>In order to improve the oral bioavailability of rifaximin and expand the clinical application of RFX for MRSA pneumonia, this study developed RFX-loaded myristic acid solid lipid nanoparticles (RFX-SLNs).</p><p><strong>Methods: </strong>This study first prepared RFX-SLNs through hot melt emulsification and ultrasonic methods and selected the optimal formula of RFX-SLNs through single-factor screening. Afterward, the particle size, zeta potential, and polydispersity index (PDI) of the RFX-SLNs were measured, the morphology of RFX-SLNs was observed by transmission electron microscopy, and the encapsulation efficiency (EE) and drug loading capacity (LC) of RFX-SLNs were detected by high-performance liquid chromatography. Then, the sustained release ability and oral bioavailability of RFX-SLNs were studied through in vitro release and pharmacokinetics. Finally, the therapeutic effect of RFX-SLNs on MRSA pneumonia infection was studied by using a mouse MRSA pneumonia infection model.</p><p><strong>Results: </strong>The optimal formulation of RFX-SLNs was 1% RFX with water (3% PVA) and oil (myristic acid) ratio of 1:19. RFX-SLNs were spherical in shape with a smooth surface and uniform size. The EE and LC of three different batches of RFX-SLNs were 89.35±2.47%, 90.45±3.69%, 88.72±1.18%, and 9.50 ± 0.01%, 10.09±0.01%, and 9.68±0.00%, respectively. In vitro release and pharmacokinetic studies showed that the myristic acid solid lipid nanoparticles showed excellent sustained release as expected and increased the oral bioavailability of RFX by 2.18 times. This indicates that RFX-SLNs can be used for the oral treatment of bacterial infections. Compared to RFX, RFX-SLNs showed good therapeutic effects in a mouse MRSA pneumonia infection model.</p><p><strong>Conclusion: </strong>This study indicates that the myristic acid solid lipid nanoparticles might be an effective way to enhance the oral absorption and therapy effects of RFX and other insoluble drugs. This not only opens up avenues for the clinical application of RFX but also provides a way for the development of new dosage forms of water-soluble drugs and the expansion of their clinical application scope.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141984240","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A Comprehensive Analysis of Liposomal-Based Nanocarriers for Treating Skin and Soft Tissue Infection.","authors":"Dyala M Khasawneh, Rami J Oweis, Mo'tasem Alsmadi","doi":"10.2174/0115672018328954240801110200","DOIUrl":"https://doi.org/10.2174/0115672018328954240801110200","url":null,"abstract":"<p><p>Bacterial skin and soft tissue infections (SSTIs) are widespread microbic invasions of the skin and deeper tissues. Topical drug delivery systems are the most favored administration pathway when treating SSTIs. This is down to their minimal risk of inducing systemic adverse events, reduced development of bacterial resistance, and ease of application. However, they have several drawbacks, including the lack of control over the drug release profile, skin irritations, and the limited permeability of certain compounds through the skin. To address these limitations, several nanocarrier systems were developed, with nanoliposomes standing out as the leading delivery system for the topical management of SSTIs. Despite considerable research into liposomes over the past decade, there remains a gap in detailed knowledge about designing these carriers specifically for SSTIs. Consequently, there is a pressing need for comprehensive research that focuses on the use of nanoliposomes for SSTIs and offers an extensive understanding of both SSTIs and liposomal formulations. This review explores bacterial SSTIs, covering their epidemiology, classification, microbiology, and management. It emphasizes the contribution of liposome-based nanovesicles in enhancing the local administration of antibiotics and natural antibacterial compounds for SSTI management. It also delves into the effects of liposomal formulation changes on the disease therapeutic outcomes. Additionally, it provides a guide for aligning the characteristics of the liposomes with the infection types, depths, properties, and causative agents. This signifies a substantial leap forward in the domains of drug design, development, and delivery.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141972519","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Muhammad Hadi Sultan, Yosif Almoshari, Syam Mohan, Mohamed Ahmed Al-Kasim, Hamad S Alyami, Mohammad Azam Ansari, Mohammad Intakhab Alam
{"title":"Capecitabine-loaded NLC for Breast Cancer Treatment: Preparation, Characterization, and In vitro Evaluation.","authors":"Muhammad Hadi Sultan, Yosif Almoshari, Syam Mohan, Mohamed Ahmed Al-Kasim, Hamad S Alyami, Mohammad Azam Ansari, Mohammad Intakhab Alam","doi":"10.2174/0115672018309370240708113038","DOIUrl":"10.2174/0115672018309370240708113038","url":null,"abstract":"<p><strong>Background: </strong>Cancer treatment often involves the use of potent antineoplastic drugs like Capecitabine [CAP], which can lead to serious toxicities. There is a need for dosage forms to manage these toxicities that can deliver the medication effectively to the target site while maintaining therapeutic efficacy at lower doses. To achieve the aforesaid objective, NLC containing capecitabine [NANOBIN] was prepared and evaluated. Different formulations of NANOBIN, denoted as CaTS, CaT1S, CaT2S, CaTS1, and CaTS2, were designed and evaluated to improve drug delivery and therapeutic outcomes.</p><p><strong>Methods: </strong>The NANOBIN formulations were prepared using the hot homogenization method. The characterization of these formulations was conducted based on various parameters such as particle size, Polydispersity Index [PDI], Zeta Potential [ZP], Transmission Electron Microscopy [TEM] imaging, and Encapsulation Efficiency [EE]. In vitro evaluations included stability testing, release studies to assess drug release kinetics, and a cytotoxicity assay [MTT assay] to evaluate the efficacy of these formulations against human breast cancer cells [MCF-7].</p><p><strong>Results: </strong>The characterization results revealed that all NANOBIN formulations exhibited particle sizes ranging from 65 to 193 nm, PDI values within the range of 0.26-0.37, ZP values between 46.47 to 61.87 mV [-ve], and high EE percentages ranging from 94.121% to 96.64%. Furthermore, all NANOBIN formulations demonstrated sustained and slow-release profiles of CAP. The MTT assay showed that the NANOBINs exhibited significantly enhanced cytotoxic efficacy, approximately 10 times greater than free CAP when tested on MCF-7 cells. These findings indicate the potential of NANOBINs to deliver CAP effectively to the target site, enabling prolonged drug availability and enhanced therapeutic effects at lower doses.</p><p><strong>Conclusion: </strong>The study demonstrates that NANOBINs can effectively deliver CAP to target sites, prolonging drug exposure and enhancing therapeutic efficacy while reducing the required dose. Further studies are necessary to validate these findings and establish NANOBINs as a preferred treatment option for cancer therapy.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141794474","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advances in Iron Deficiency Anaemia Management: Exploring Novel Drug Delivery Systems and Future Perspectives.","authors":"Muskan Saini, Karan Trehan, Shubham Thakur, Anuj Modi, Subheet Kumar Jain","doi":"10.2174/0115672018300804240426070552","DOIUrl":"https://doi.org/10.2174/0115672018300804240426070552","url":null,"abstract":"<p><p>Iron Deficiency Anaemia (IDA) is a prevalent global health issue characterized by inadequate iron levels in the body, leading to impaired red blood cell production and subsequent anaemia. Traditional treatment approaches for IDA, such as oral iron supplementation, often encounter challenges related to poor compliance, gastrointestinal side effects, and variable absorption rates. As a result, there is a growing interest in exploring novel drug delivery systems to enhance iron therapy efficacy and patient outcomes. This review discusses recent advances in IDA management, focusing on developing and utilizing innovative drug delivery systems for iron supplementation. Various strategies, including nanoformulations, microparticles, liposomes, and hydrogels, are explored for their potential to improve iron bioavailability, reduce adverse effects, and optimize therapeutic outcomes. Furthermore, promising strategies for the future management of IDA are explored, including the utilization of advanced technologies such as targeted drug delivery systems, controlled release mechanisms, and combination therapies. The integration of these novel drug delivery systems with advancements in diagnostics, personalized medicine, and patient-centered care holds great potential to revolutionize the management of IDA and improve the quality of life for individuals affected by this condition.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141790735","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Reduction-Responsive Polyprodrug Nanoplatform Based on Curcumin for Tumor-Targeted Therapy.","authors":"Ziyi Zhang, Jinyuan Tian, Xiaoqing Xu, Wei Shi, Yajuan Qi, Zhanjun Liu","doi":"10.2174/0115672018314506240723080113","DOIUrl":"https://doi.org/10.2174/0115672018314506240723080113","url":null,"abstract":"<p><strong>Introduction: </strong>Polymer prodrug nanoparticles have become an emerging drug delivery system in cancer therapy due to their high drug loading. However, their poor drug release and lack of tumor cell targeting limit their clinical application.</p><p><strong>Objective: </strong>This study aimed to prepare targeted and reduction-reactive polyprodrug nanocarriers based on curcumin (CUR) for co-delivery of doxorubicin (DOX), labeled as DOX/HAPCS NPs, and to investigate their anticancer activity.</p><p><strong>Methods: </strong>The polymer was synthesized and characterized by chemical method. The drug loading and drug release behavior of DOX and CUR in polymer nanoparticles were determined. Moreover, the antitumor effects of polymer nanoparticles were evaluated using an MTT experiment and tumor inhibition experiment, and the synergistic effect of co-delivered DOX and CUR was explored.</p><p><strong>Results: </strong>The particle size of DOX/HAPCS NPs was 152.5nm, and the potential was about -26.74 mV. The drug-carrying capacity of DOX and CUR was about 7.56% and 34.75%, respectively, indicating high drug-carrying capacity and good stability. DOX and CUR released over 90% within 24 hours in the tumor environment. Compared with free DOX, DOX/HAPCS NPs demonstrated significantly enhanced cell and tumor inhibitory effects (P< 0.05) in vivo and in vitro and changed drug distribution to avoid toxic side effects on normal tissues. The combined index showed that DOX and CUR showed synergistic anticancer effects at a set ratio.</p><p><strong>Conclusion: </strong>The prepared reduction-responsive targeted polymer nanomedical DOX/HAPCS NPs exhibited a synergistic anti-cancer effect, with high drug loading capacity and the ability to release drugs in proportion, making it a promising polymer nanoparticle drug delivery system.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141763744","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Ginger Extract-Loaded Chitosan Nanoparticles: Investigating their Impact on Lipid Metabolism and Obesity in a Rat Model through In vitro and In vivo Analysis.","authors":"Rui Gao, Qunbin Fan, Saeed Rohani","doi":"10.2174/0115672018315676240715065750","DOIUrl":"https://doi.org/10.2174/0115672018315676240715065750","url":null,"abstract":"<p><strong>Introduction: </strong>Obesity has become a pressing global health crisis, reaching alarming proportions and bearing significant consequences for public health on a global scale.</p><p><strong>Aim: </strong>In this research, chitosan nanoparticles were employed to encapsulate ginger extract, and the impact of this formulation on lipid metabolism and obesity was investigated using a rat model.</p><p><strong>Methods: </strong>In vitro experiments, encompassing assessments of cell viability, microstructure, anti-inflammatory activity, and release dynamics, were conducted to comprehensively evaluate the nanoformulation. The study extended to examining the potential anti-obesity efficacy of the developed nanoformulation in rats induced with obesity through a high-fat diet.</p><p><strong>Results: </strong>In vitro findings affirmed the safety of the carriers and revealed their robust anti-inflammatory properties. The average particle size for ginger-loaded and ginger-free chitosan nanoparticles was measured to be 458.92 ± 139.35 nm and 466.29 ± 142.71 nm, respectively. The in vivo investigation demonstrated the dose-dependent effects of ginger extract-loaded chitosan nanoparticles, manifesting in a reduction of obesity and improvement in liver function.</p><p><strong>Conclusion: </strong>These promising results suggest that the developed nanoformulation could be considered a viable therapeutic option for individuals struggling with obesity.</p>","PeriodicalId":94287,"journal":{"name":"Current drug delivery","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}