1
|
Wang Y, Ding Q, Ma G, Zhang Z, Wang J, Lu C, Xiang C, Qian K, Zheng J, Shan Y, Zhang P, Cheng Z, Gong P, Zhao Q. Mucus-Penetrable Biomimetic Nanoantibiotics for Pathogen-Induced Pneumonia Treatment. ACS NANO 2024. [PMID: 39485232 DOI: 10.1021/acsnano.4c10837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Bacterial pneumonia has garnered significant attention in the realm of infectious diseases owing to a surge in the incidence of severe infections coupled with the growing scarcity of efficacious therapeutic modalities. Antibiotic treatment is still an irreplaceable method for bacterial pneumonia because of its strong bactericidal activity and good clinical efficacy. However, the mucus layer forming after a bacterial infection in the lungs has been considered as the "Achilles' heels" facing the clinical application of such treatment. Herein, traceable biomimetic nanoantibiotics (BioNanoCFPs) were developed by loading indacenodithieno[3,2-b]thiophene (ITIC) and cefoperazone (CFP) in nanoplatforms coated with natural killer (NK) cell membranes. The BioNanoCFP exhibited excellent demonstrated mucus-penetrating abilities, facilitating their arrival at the infection site. The presence of Toll-like receptors in the NK cell membrane rendered the BioNanoCFP with the capability to recognize pathogen-associated molecular patterns within bacteria, allowing precise targeting of bacterial colonization sites and achieving substantial therapeutic efficacy. Overall, our findings demonstrate the viability and desirability of using NK cell membrane-mediated drug delivery as a promising strategy for precision treatment.
Collapse
Affiliation(s)
- Yue Wang
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Gongcheng Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhiwei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiaqi Wang
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chang Lu
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Chunbai Xiang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jun Zheng
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro Center of Biomedicine and Health, Luohu, Shenzhen 518024, China
| | - Qi Zhao
- Cancer Center, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| |
Collapse
|
2
|
de Lima JS, Leão AD, de Jesus Oliveira AC, Chaves LL, Ramos RKLG, Rodrigues CFC, Soares-Sobrinho JL, Soares MFDLR. Potential of plant-based polysaccharides as therapeutic agents in ulcerogenic diseases of the gastrointestinal tract: A review. Int J Biol Macromol 2024; 281:136399. [PMID: 39395521 DOI: 10.1016/j.ijbiomac.2024.136399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
In recent years, natural polysaccharides (PSs) have attracted increasing interest because of their remarkable biological properties and potential in various areas, such as medicine, and food. This study aimed to present a detailed review of the evidence on the therapeutic potential of PSs for the treatment of gastrointestinal diseases. The main evidence was correlated with their chemical composition, mechanism of action and therapeutic effect. The main results showed that the action can be attributed to their ability to suppress excessive inflammatory responses, regulating the expression of cytokines and interleukins, reducing intestinal inflammation and promoting wound healing. Furthermore, we discussed how PSs help in the repair of the intestinal mucosa and related these effects with the composition of monosaccharides. A detailed analysis was performed on the ability of PSs to modulate the intestinal microbiota, promoting the growth of beneficial bacteria and suppressing inflammatory bacteria, in addition to its probiotic action with production of short-chain fatty acids. All this evidence was also taken into a broader context, in which the main challenges in processing PSs were considered and strategies to circumvent them were pointed out. Therefore, this review sought to demonstrate the great potential and viability of PSs as innovative and effective therapeutic agents.
Collapse
Affiliation(s)
- Jucielma Silva de Lima
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Amanda Damaceno Leão
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Antônia Carla de Jesus Oliveira
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Luíse Lopes Chaves
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Renata Kelly Luna Gomes Ramos
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Carla Fernanda Couto Rodrigues
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - José Lamartine Soares-Sobrinho
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| | - Mônica Felts de La Roca Soares
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
3
|
Faurschou KL, Clasky AJ, Watchorn J, Tram Su J, Li NT, McGuigan AP, Gu FX. Lateral Assessment of Mucomimetic Hydrogels to Evaluate Correlation between Microscopic and Macroscopic Properties. Macromol Biosci 2024:e2400146. [PMID: 39374341 DOI: 10.1002/mabi.202400146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/06/2024] [Indexed: 10/09/2024]
Abstract
A major limitation in the development of mucosal drug delivery systems is the design of in vitro models that accurately reflect in vivo conditions. Traditionally, models seek to mimic characteristics of physiological mucus, often focusing on property-specific trial metrics such as rheological behavior or diffusion of a nanoparticle of interest. Despite the success of these models, translation from in vitro results to in vivo trials is limited. As a result, several authors have called for work to develop standardized testing methodologies and characterize the influence of model properties on drug delivery performance. To this end, a series of trials is performed on 12 mucomimetic hydrogels reproduced from literature. Experiments show that there is no consistent correlation between barrier performance and rheological or microstructural properties of the tested mucomimetic hydrogels. In addition, the permeability of both mucopenetrating and mucoadhesive nanoparticles is assessed, revealing non-obvious variations in barrier properties such as the relative contributions of electrostatic and hydrophobic interactions in different models. These results demonstrate the limitations of predicting mucomimetic behavior with common characterization techniques and highlight the importance of testing barrier performance with multiple nanoparticle formulations.
Collapse
Affiliation(s)
- Kristina L Faurschou
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Jeffrey Watchorn
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Jennifer Tram Su
- Department of Chemical Engineering, McGill University, Montreal, Quebec, H3A 0C5, Canada
| | - Nancy T Li
- Computational Biology, Ontario Institute for Cancer Research, Toronto, Ontario, M5G 0A3, Canada
| | - Alison P McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Frank X Gu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| |
Collapse
|
4
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2024; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
5
|
van Dinteren S, Araya-Cloutier C, Bastiaan-Net S, Boudewijn A, van Heek T, Vincken JP, Witkamp R, Meijerink J. Biotransformation and Epithelial Toxicity of Prenylated Phenolics from Licorice Roots ( Glycyrrhiza spp.) in 3D Apical-Out Mucus-Producing Human Enteroids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20396-20409. [PMID: 39240776 PMCID: PMC11421016 DOI: 10.1021/acs.jafc.4c03120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 09/08/2024]
Abstract
Apical-out enteroids mimic the in vivo environment well due to their accessible apical surface and mucus layer, making them an ideal model for studying the impact of (bioactive) food compounds. Generated human ileal apical-out enteroids showed a fucose-containing mucus layer surrounding the apical brush border on their exposure side, indicating their physiological relevance. Effects on the mucosal epithelium of antibacterial prenylated phenolics (glabridin, licochalcone A, and glycycoumarin) from licorice roots were investigated for cytotoxicity, cell viability, barrier integrity, and biotransformation. At concentrations up to 500 μg mL-1, licochalcone A and glycycoumarin did not significantly affect apical-out enteroids, with cytotoxicities of -6 ± 2 and -2 ± 2% and cell viabilities of 77 ± 22 and 77 ± 13%, respectively (p > 0.05). Conversely, 500 μg mL-1 glabridin induced significant cytotoxicity (31 ± 25%, p < 0.05) and reduced cell viability (21 ± 14%, p < 0.01). Apical-out enteroids revealed differential sensitivities to prenylated phenolics not observed in apical-in enteroids and Caco-2 cells. Both enteroid models showed phase II biotransformation but differed in the extent of glucuronide conversion. The apical mucus layer of apical-out enteroids likely contributed to these differential interactions, potentially due to differences in electrostatic repulsion. This study underscores the relevance of 3D apical-out enteroid models and highlights the promise of prenylated phenolics for antimicrobial applications.
Collapse
Affiliation(s)
- Sarah van Dinteren
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Carla Araya-Cloutier
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen
Food & Biobased Research, Wageningen
University & Research, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Anouk Boudewijn
- Wageningen
Food & Biobased Research, Wageningen
University & Research, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Tjarda van Heek
- Department
of Abdominal Surgery, Hospital Gelderse
Vallei, Willy Brandtlaan 10, Ede 6716 RP, The Netherlands
| | - Jean-Paul Vincken
- Laboratory
of Food Chemistry, Wageningen University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Renger Witkamp
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| | - Jocelijn Meijerink
- Division
of Human Nutrition and Health, Wageningen
University, P.O. Box 17, Wageningen 6700 AA, The Netherlands
| |
Collapse
|
6
|
Zheng Y, Luo S, Xu M, He Q, Xie J, Wu J, Huang Y. Transepithelial transport of nanoparticles in oral drug delivery: From the perspective of surface and holistic property modulation. Acta Pharm Sin B 2024; 14:3876-3900. [PMID: 39309496 PMCID: PMC11413706 DOI: 10.1016/j.apsb.2024.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 09/25/2024] Open
Abstract
Despite the promising prospects of nanoparticles in oral drug delivery, the process of oral administration involves a complex transportation pathway that includes cellular uptake, intracellular trafficking, and exocytosis by intestinal epithelial cells, which are necessary steps for nanoparticles to enter the bloodstream and exert therapeutic effects. Current researchers have identified several crucial factors that regulate the interaction between nanoparticles and intestinal epithelial cells, including surface properties such as ligand modification, surface charge, hydrophilicity/hydrophobicity, intestinal protein corona formation, as well as holistic properties like particle size, shape, and rigidity. Understanding these properties is essential for enhancing transepithelial transport efficiency and designing effective oral drug delivery systems. Therefore, this review provides a comprehensive overview of the surface and holistic properties that influence the transepithelial transport of nanoparticles, elucidating the underlying principles governing their impact on transepithelial transport. The review also outlines the chosen of parameters to be considered for the subsequent design of oral drug delivery systems.
Collapse
Affiliation(s)
- Yaxian Zheng
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Shiqin Luo
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Min Xu
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin He
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiang Xie
- Department of Pharmacy, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiawei Wu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Bohley M, Leroux J. Gastrointestinal Permeation Enhancers Beyond Sodium Caprate and SNAC - What is Coming Next? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400843. [PMID: 38884149 PMCID: PMC11434117 DOI: 10.1002/advs.202400843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/13/2024] [Indexed: 06/18/2024]
Abstract
Oral peptide delivery is trending again. Among the possible reasons are the recent approvals of two oral peptide formulations, which represent a huge stride in the field. For the first time, gastrointestinal (GI) permeation enhancers (PEs) are leveraged to overcome the main limitation of oral peptide delivery-low permeability through the intestinal epithelium. Despite some success, the application of current PEs, such as salcaprozate sodium (SNAC), sodium caprylate (C8), and sodium caprate (C10), is generally resulting in relatively low oral bioavailabilities (BAs)-even for carefully selected therapeutics. With several hundred peptide-based drugs presently in the pipeline, there is a huge unmet need for more effective PEs. Aiming to provide useful insights for the development of novel PEs, this review summarizes the biological hurdles to oral peptide delivery with special emphasis on the epithelial barrier. It describes the concepts and action modes of PEs and mentions possible new targets. It further states the benchmark that is set by current PEs, while critically assessing and evaluating emerging PEs regarding translatability, safety, and efficacy. Additionally, examples of novel PEs under preclinical and clinical evaluation and future directions are discussed.
Collapse
Affiliation(s)
- Marilena Bohley
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
8
|
Gong T, Liu X, Wang X, Lu Y, Wang X. Applications of polysaccharides in enzyme-triggered oral colon-specific drug delivery systems: A review. Int J Biol Macromol 2024; 275:133623. [PMID: 38969037 DOI: 10.1016/j.ijbiomac.2024.133623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Enzyme-triggered oral colon-specific drug delivery system (EtOCDDS1) can withstand the harsh stomach and small intestine environments, releasing encapsulated drugs selectively in the colon in response to colonic microflora, exerting local or systematic therapeutic effects. EtOCDDS boasts high colon targetability, enhanced drug bioavailability, and reduced systemic side effects. Polysaccharides are extensively used in enzyme-triggered oral colon-specific drug delivery systems, and its colon targetability has been widely confirmed, as their properties meet the demand of EtOCDDS. Polysaccharides, known for their high safety and excellent biocompatibility, feature modifiable structures. Some remain undigested in the stomach and small intestine, whether in their natural state or after modifications, and are exclusively broken down by colon-resident microbiota. Such characteristics make them ideal materials for EtOCDDS. This article reviews the design principles of EtOCDDS as well as commonly used polysaccharides and their characteristics, modifications, applications and specific mechanism for colon targeting. The article concludes by summarizing the limitations and potential of ETOCDDS to stimulate the development of innovative design approaches.
Collapse
Affiliation(s)
- Tingting Gong
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xinxin Liu
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xi Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Yunqian Lu
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Peking Union Medical College, No.151, Malianwa North Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|
9
|
Liu Y, Long M, Wang Y, Liang Z, Dong Y, Qu M, Ge X, Nan Y, Chen Y, Zhou X. Chitosan-alginate/R8 ternary polyelectrolyte complex as an oral protein-based vaccine candidate induce effective mucosal immune responses. Int J Biol Macromol 2024; 275:133671. [PMID: 38971274 DOI: 10.1016/j.ijbiomac.2024.133671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Vaccination is the most effective method for preventing infectious diseases. Oral vaccinations have attracted much attention due to the ability to boost intestinal and systemic immunity. The focus of this study was to develop a poly (lactide-co-glycolide) acid (PLGA)-based ternary polyelectrolyte complex (PEC) with chitosan, sodium alginate, and transmembrane peptides R8 for the delivery of antigen proteins. In this study, the antigen protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis (MAP) antigens HBHA, Ag85B, and Bfra, was combined with R8 to generate self-assembled conjugates. The results showed that PEC presented a cross-linked reticular structure to protect the encapsulated proteins in the simulated gastric fluid. Then, the nanocomposite separated into individual nanoparticles after entering the simulated intestinal fluid. The ternary PEC with R8 promoted the in vivo uptake of antigens by intestinal lymphoid tissue. Moreover, the ternary PEC administered orally to mice promoted the secretion of specific antibodies and intestinal mucosal IgA. In addition, in the mouse models of MAP infection, the ternary PEC enhanced splenic T cell responses, thus reducing bacterial load and liver pathology score. These results suggested that this ternary electrolyte complex could be a promising delivery platform for oral subunit vaccine candidates, not limited to MAP infection.
Collapse
Affiliation(s)
- Yiduo Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Meizhen Long
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yuanzhi Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Zhengmin Liang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yuhui Dong
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Mengjin Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Xin Ge
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yue Nan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yulan Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Xiangmei Zhou
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China.
| |
Collapse
|
10
|
Caturano A, Nilo R, Nilo D, Russo V, Santonastaso E, Galiero R, Rinaldi L, Monda M, Sardu C, Marfella R, Sasso FC. Advances in Nanomedicine for Precision Insulin Delivery. Pharmaceuticals (Basel) 2024; 17:945. [PMID: 39065795 PMCID: PMC11279564 DOI: 10.3390/ph17070945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes mellitus, which comprises a group of metabolic disorders affecting carbohydrate metabolism, is characterized by improper glucose utilization and excessive production, leading to hyperglycemia. The global prevalence of diabetes is rising, with projections indicating it will affect 783.2 million people by 2045. Insulin treatment is crucial, especially for type 1 diabetes, due to the lack of β-cell function. Intensive insulin therapy, involving multiple daily injections or continuous subcutaneous insulin infusion, has proven effective in reducing microvascular complications but poses a higher risk of severe hypoglycemia. Recent advancements in insulin formulations and delivery methods, such as ultra-rapid-acting analogs and inhaled insulin, offer potential benefits in terms of reducing hypoglycemia and improving glycemic control. However, the traditional subcutaneous injection method has drawbacks, including patient compliance issues and associated complications. Nanomedicine presents innovative solutions to these challenges, offering promising avenues for overcoming current drug limitations, enhancing cellular uptake, and improving pharmacokinetics and pharmacodynamics. Various nanocarriers, including liposomes, chitosan, and PLGA, provide protection against enzymatic degradation, improving drug stability and controlled release. These nanocarriers offer unique advantages, ranging from enhanced bioavailability and sustained release to specific targeting capabilities. While oral insulin delivery is being explored for better patient adherence and cost-effectiveness, other nanomedicine-based methods also show promise in improving delivery efficiency and patient outcomes. Safety concerns, including potential toxicity and immunogenicity issues, must be addressed, with the FDA providing guidance for the safe development of nanotechnology-based products. Future directions in nanomedicine will focus on creating next-generation nanocarriers with precise targeting, real-time monitoring, and stimuli-responsive features to optimize diabetes treatment outcomes and patient safety. This review delves into the current state of nanomedicine for insulin delivery, examining various types of nanocarriers and their mechanisms of action, and discussing the challenges and future directions in developing safe and effective nanomedicine-based therapies for diabetes management.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Roberto Nilo
- Data Collection G-STeP Research Core Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Davide Nilo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | | | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
11
|
Serra-Casablancas M, Di Carlo V, Esporrín-Ubieto D, Prado-Morales C, Bakenecker AC, Sánchez S. Catalase-Powered Nanobots for Overcoming the Mucus Barrier. ACS NANO 2024; 18:16701-16714. [PMID: 38885185 PMCID: PMC11223492 DOI: 10.1021/acsnano.4c01760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Biological barriers present a significant obstacle to treatment, especially when drugs are administered locally to increase their concentrations at the target site while minimizing unintended off-target effects. Among these barriers, mucus presents a challenge, as it serves as a protective layer in the respiratory, urogenital, and gastrointestinal tracts. Its role is to shield the underlying epithelial cells from pathogens and toxic compounds but also impedes the efficient delivery of drugs. Despite the exploration of mucolytic agents to improve drug delivery, overcoming this protective barrier remains a significant hurdle. In our study, we investigate an alternative approach involving the use of catalase-powered nanobots. We use an in vitro model that simulates intestinal mucus secretion to demonstrate the dual functionality of our nanobots. This includes their ability to disrupt mucus, which we confirmed through in vitro and ex vivo validation, as well as their self-propulsion to overcome the mucus barrier, resulting in a 60-fold increase compared with passive nanoparticles. Therefore, our findings highlight the potential utility of catalase-powered nanobots as carriers for therapeutic agents since they could enhance drug delivery efficiency by penetrating the mucus barrier.
Collapse
Affiliation(s)
- Meritxell Serra-Casablancas
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
- Universitat
de Barcelona, Facultat de Farmàcia i Ciències de l’Alimentació, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Valerio Di Carlo
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
| | - David Esporrín-Ubieto
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
| | - Carles Prado-Morales
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
- Universitat
de Barcelona, Facultat de Farmàcia i Ciències de l’Alimentació, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Anna C. Bakenecker
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
| | - Samuel Sánchez
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
- Institució
Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys,
23, 08010 Barcelona, Spain
| |
Collapse
|
12
|
Alwani S, Wasan EK, Badea I. Solid Lipid Nanoparticles for Pulmonary Delivery of Biopharmaceuticals: A Review of Opportunities, Challenges, and Delivery Applications. Mol Pharm 2024; 21:3084-3102. [PMID: 38828798 DOI: 10.1021/acs.molpharmaceut.4c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Biopharmaceuticals such as nucleic acids, proteins, and peptides constitute a new array of treatment modalities for chronic ailments. Invasive routes remain the mainstay of administering biopharmaceuticals due to their labile nature in the biological environment. However, it is not preferred for long-term therapy due to the lack of patient adherence and clinical suitability. Therefore, alternative routes of administration are sought to utilize novel biopharmaceutical therapies to their utmost potential. Nanoparticle-mediated pulmonary delivery of biologics can facilitate both local and systemic disorders. Solid lipid nanoparticles (SLNs) afford many opportunities as pulmonary carriers due to their physicochemical stability and ability to incorporate both hydrophilic and hydrophobic moieties, thus allowing novel combinatorial drug/gene therapies. These applications include pulmonary infections, lung cancer, and cystic fibrosis, while systemic delivery of biomolecules, like insulin, is also attractive for the treatment of chronic ailments. This Review explores physiological and particle-associated factors affecting pulmonary delivery of biopharmaceuticals. It compares the advantages and limitations of SLNs as pulmonary nanocarriers along with design improvements underway to overcome these limitations. Current research illustrating various SLN designs to deliver proteins, peptides, plasmids, oligonucleotides, siRNA, and mRNA is also summarized.
Collapse
Affiliation(s)
- Saniya Alwani
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ellen K Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| |
Collapse
|
13
|
Kishimoto H, Ridley C, Inoue K, Thornton DJ. Assessment of polymeric mucin-drug interactions. PLoS One 2024; 19:e0306058. [PMID: 38935605 PMCID: PMC11210812 DOI: 10.1371/journal.pone.0306058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Mucosal-delivered drugs have to pass through the mucus layer before absorption through the epithelial cell membrane. Although there has been increasing interest in polymeric mucins, a major structural component of mucus, potentially acting as important physiological regulators of mucosal drug absorption, there are no reports that have systematically evaluated the interaction between mucins and drugs. In this study, we assessed the potential interaction between human polymeric mucins (MUC2, MUC5B, and MUC5AC) and various drugs with different chemical profiles by simple centrifugal method and fluorescence analysis. We found that paclitaxel, rifampicin, and theophylline likely induce the aggregation of MUC5B and/or MUC2. In addition, we showed that the binding affinity of drugs for polymeric mucins varied, not only between individual drugs but also among mucin subtypes. Furthermore, we demonstrated that deletion of MUC5AC and MUC5B in A549 cells increased the cytotoxic effects of cyclosporin A and paclitaxel, likely due to loss of mucin-drug interaction. In conclusion, our results indicate the necessity to determine the binding of drugs to mucins and their potential impact on the mucin network property.
Collapse
Affiliation(s)
- Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Caroline Ridley
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - David J. Thornton
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Zhong Q, Zeng J, Jia X. Self-Assembled Aggregated Structures of Natural Products for Oral Drug Delivery. Int J Nanomedicine 2024; 19:5931-5949. [PMID: 38887690 PMCID: PMC11182358 DOI: 10.2147/ijn.s467354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
The self-assembling aggregated structures of natural products have gained significant interest due to their simple synthesis, lack of carrier-related toxicity, and excellent biological efficacy. However, the mechanisms of their assembly and their ability to traverse the gastrointestinal (GI) barrier remain unclear. This review summarizes various intermolecular non-covalent interactions and aggregated structures, drawing on research indexed in Web of Science from 2010 to 2024. Cheminformatics analysis of the self-assembly behaviors of natural small molecules and their supramolecular aggregates reveals assembly-favorable conditions, aiding drug formulation. Additionally, the review explores the self-assembly properties of macromolecules like polysaccharides, proteins, and exosomes, highlighting their role in drug delivery. Strategies to overcome gastrointestinal barriers and enhance drug bioavailability are also discussed. This work underscores the potential of natural products in oral drug delivery and offers insights for designing more effective drug delivery systems.
Collapse
Affiliation(s)
- Qiyuan Zhong
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Jingqi Zeng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, People’s Republic of China
| |
Collapse
|
15
|
Dubashynskaya NV, Petrova VA, Skorik YA. Biopolymer Drug Delivery Systems for Oromucosal Application: Recent Trends in Pharmaceutical R&D. Int J Mol Sci 2024; 25:5359. [PMID: 38791397 PMCID: PMC11120705 DOI: 10.3390/ijms25105359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Oromucosal drug delivery, both local and transmucosal (buccal), is an effective alternative to traditional oral and parenteral dosage forms because it increases drug bioavailability and reduces systemic drug toxicity. The oral mucosa has a good blood supply, which ensures that drug molecules enter the systemic circulation directly, avoiding drug metabolism during the first passage through the liver. At the same time, the mucosa has a number of barriers, including mucus, epithelium, enzymes, and immunocompetent cells, that are designed to prevent the entry of foreign substances into the body, which also complicates the absorption of drugs. The development of oromucosal drug delivery systems based on mucoadhesive biopolymers and their derivatives (especially thiolated and catecholated derivatives) is a promising strategy for the pharmaceutical development of safe and effective dosage forms. Solid, semi-solid and liquid pharmaceutical formulations based on biopolymers have several advantageous properties, such as prolonged residence time on the mucosa due to high mucoadhesion, unidirectional and modified drug release capabilities, and enhanced drug permeability. Biopolymers are non-toxic, biocompatible, biodegradable and may possess intrinsic bioactivity. A rational approach to the design of oromucosal delivery systems requires an understanding of both the anatomy/physiology of the oral mucosa and the physicochemical and biopharmaceutical properties of the drug molecule/biopolymer, as presented in this review. This review summarizes the advances in the pharmaceutical development of mucoadhesive oromucosal dosage forms (e.g., patches, buccal tablets, and hydrogel systems), including nanotechnology-based biopolymer nanoparticle delivery systems (e.g., solid lipid particles, liposomes, biopolymer polyelectrolyte particles, hybrid nanoparticles, etc.).
Collapse
Affiliation(s)
| | | | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
| |
Collapse
|
16
|
Chen Y, Ye Z, Chen H, Li Z. Breaking Barriers: Nanomedicine-Based Drug Delivery for Cataract Treatment. Int J Nanomedicine 2024; 19:4021-4040. [PMID: 38736657 PMCID: PMC11086653 DOI: 10.2147/ijn.s463679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/27/2024] [Indexed: 05/14/2024] Open
Abstract
Cataract is a leading cause of blindness globally, and its surgical treatment poses a significant burden on global healthcare. Pharmacologic therapies, including antioxidants and protein aggregation reversal agents, have attracted great attention in the treatment of cataracts in recent years. Due to the anatomical and physiological barriers of the eye, the effectiveness of traditional eye drops for delivering drugs topically to the lens is hindered. The advancements in nanomedicine present novel and promising strategies for addressing challenges in drug delivery to the lens, including the development of nanoparticle formulations that can improve drug penetration into the anterior segment and enable sustained release of medications. This review introduces various cutting-edge drug delivery systems for cataract treatment, highlighting their physicochemical properties and surface engineering for optimal design, thus providing impetus for further innovative research and potential clinical applications of anti-cataract drugs.
Collapse
Affiliation(s)
- Yilin Chen
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zi Ye
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Haixu Chen
- Institute of Geriatrics, National Clinical Research Center for Geriatrics Diseases, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhaohui Li
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
- Senior Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
17
|
Ding B, Zhu Z, Guo C, Li J, Gan Y, Yu M. Oral peptide therapeutics for diabetes treatment: State-of-the-art and future perspectives. Acta Pharm Sin B 2024; 14:2006-2025. [PMID: 38799624 PMCID: PMC11120284 DOI: 10.1016/j.apsb.2024.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/04/2023] [Accepted: 12/26/2023] [Indexed: 05/29/2024] Open
Abstract
Diabetes, characterized by hyperglycemia, is a major cause of death and disability worldwide. Peptides, such as insulin and glucagon-like peptide-1 (GLP-1) analogs, have shown promise as treatments for diabetes due to their ability to mimic or enhance insulin's actions in the body. Compared to subcutaneous injection, oral administration of anti-diabetic peptides is a preferred approach. However, biological barriers significantly reduce the efficacy of oral peptide therapeutics. Recent advancements in drug delivery systems and formulation techniques have greatly improved the oral delivery of peptide therapeutics and their efficacy in treating diabetes. This review will highlight (1) the benefits of oral anti-diabetic peptide therapeutics; (2) the biological barriers for oral peptide delivery, including pH and enzyme degradation, intestinal mucosa barrier, and biodistribution barrier; (3) the delivery platforms to overcome these biological barriers. Additionally, the review will discuss the prospects in this field. The information provided in this review will serve as a valuable guide for future developments in oral anti-diabetic peptide therapeutics.
Collapse
Affiliation(s)
- Bingwen Ding
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhu Zhu
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Cong Guo
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxin Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong Gan
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China
| | - Miaorong Yu
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Yang D, Feng Y, Yuan Y, Zhang L, Zhou Y, Midgley AC, Wang Y, Liu N, Li G, Yao X, Liu D. Protein Coronas Derived from Mucus Act as Both Spear and Shield to Regulate Transferrin Functionalized Nanoparticle Transcellular Transport in Enterocytes. ACS NANO 2024; 18:7455-7472. [PMID: 38417159 DOI: 10.1021/acsnano.3c11315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2024]
Abstract
The epithelial mucosa is a key biological barrier faced by gastrointestinal, intraoral, intranasal, ocular, and vaginal drug delivery. Ligand-modified nanoparticles demonstrate excellent ability on this process, but their efficacy is diminished by the formation of protein coronas (PCs) when they interact with biological matrices. PCs are broadly implicated in affecting the fate of NPs in vivo and in vitro, yet few studies have investigated PCs formed during interactions of NPs with the epithelial mucosa, especially mucus. In this study, we constructed transferrin modified NPs (Tf-NPs) as a model and explored the mechanisms and effects that epithelial mucosa had on PCs formation and the subsequent impact on the transcellular transport of Tf-NPs. In mucus-secreting cells, Tf-NPs adsorbed more proteins from the mucus layers, which masked, displaced, and dampened the active targeting effects of Tf-NPs, thereby weakening endocytosis and transcellular transport efficiencies. In mucus-free cells, Tf-NPs adsorbed more proteins during intracellular trafficking, which enhanced transcytosis related functions. Inspired by soft coronas and artificial biomimetic membranes, we used mucin as an "active PC" to precoat Tf-NPs (M@Tf-NPs), which limited the negative impacts of "passive PCs" formed during interface with the epithelial mucosa and improved favorable routes of endocytosis. M@Tf-NPs adsorbed more proteins associated with endoplasmic reticulum-Golgi functions, prompting enhanced intracellular transport and exocytosis. In summary, mucus shielded against the absorption of Tf-NPs, but also could be employed as a spear to break through the epithelial mucosa barrier. These findings offer a theoretical foundation and design platform to enhance the efficiency of oral-administered nanomedicines.
Collapse
Affiliation(s)
- Dan Yang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
- Xi'an Key Laboratory of Antiviral and Antimicrobial Resistant Bacteria Therapeutics Research, Xi'an, 710021, China
| | - Yuqi Feng
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Ying Yuan
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Linxuan Zhang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Yao Zhou
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Adam C Midgley
- Key Laboratory of Bioactive Materials (MoE), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yanrong Wang
- Key Laboratory of Bioactive Materials (MoE), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ning Liu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Guoliang Li
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Xiaolin Yao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Dechun Liu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| |
Collapse
|
19
|
Deng B, Liu S, Wang Y, Ali B, Kong N, Xie T, Koo S, Ouyang J, Tao W. Oral Nanomedicine: Challenges and Opportunities. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306081. [PMID: 37724825 DOI: 10.1002/adma.202306081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/03/2023] [Indexed: 09/21/2023]
Abstract
Compared to injection administration, oral administration is free of discomfort, wound infection, and complications and has a higher compliance rate for patients with diverse diseases. However, oral administration reduces the bioavailability of medicines, especially biologics (e.g., peptides, proteins, and antibodies), due to harsh gastrointestinal biological barriers. In this context, the development and prosperity of nanotechnology have helped improve the bioactivity and oral availability of oral medicines. On this basis, first, the biological barriers to oral administration are discussed, and then oral nanomedicine based on organic and inorganic nanomaterials and their biomedical applications in diverse diseases are reviewed. Finally, the challenges and potential opportunities in the future development of oral nanomedicine, which may provide a vital reference for the eventual clinical transformation and standardized production of oral nanomedicine, are put forward.
Collapse
Affiliation(s)
- Bo Deng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
- Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, 710049, China
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Shaomin Liu
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Ying Wang
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Barkat Ali
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jiang Ouyang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
- Department of Oncology of the First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
20
|
Azadi M, David AE. Enhancing Ocular Drug Delivery: The Effect of Physicochemical Properties of Nanoparticles on the Mechanism of Their Uptake by Human Cornea Epithelial Cells. ACS Biomater Sci Eng 2024; 10:429-441. [PMID: 38055935 DOI: 10.1021/acsbiomaterials.3c01144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
This study investigates the effect of nanoparticle size and surface chemistry on interactions of the nanoparticles with human cornea epithelial cells (HCECs). Poly(lactic-co-glycolic) acid (PLGA) nanoparticles were synthesized using the emulsion-solvent evaporation method and surface modified with mucoadhesive (alginate [ALG] and chitosan [CHS]) and mucopenetrative (polyethylene glycol [PEG]) polymers. Particles were found to be monodisperse (polydispersity index (PDI) below 0.2), spherical, and with size and zeta potential ranging from 100 to 250 nm and from -25 to +15 mV, respectively. Evaluation of cytotoxicity with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay indicated that incubating cells with nanoparticles for 24 h at concentrations up to 100 μg/mL caused only mild toxicity (70-100% cell viability). Cellular uptake studies were conducted using an in vitro model developed with a monolayer of HCECs integrated with simulated mucosal solution. Evaluation of nanoparticle uptake revealed that energy-dependent endocytosis is the primary uptake mechanism. Among the different nanoparticles studied, 100 nm PLGA NPs and PEG-PLGA-150 NPs showed the highest levels of uptake by HCECs. Additionally, uptake studies in the presence of various inhibitors suggested that macropinocytosis and caveolae-mediated endocytosis are the dominant pathways. While clathrin-mediated endocytosis was found to also be partially responsible for nanoparticle uptake, phagocytosis did not play a role within the studied ranges of size and surface chemistries. These important findings could lead to improved nanoparticle-based formulations that could improve therapies for ocular diseases.
Collapse
Affiliation(s)
- Marjan Azadi
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Allan E David
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
21
|
Fey C, Truschel T, Nehlsen K, Damigos S, Horstmann J, Stradal T, May T, Metzger M, Zdzieblo D. Enhancing pre-clinical research with simplified intestinal cell line models. J Tissue Eng 2024; 15:20417314241228949. [PMID: 38449469 PMCID: PMC10916479 DOI: 10.1177/20417314241228949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/12/2024] [Indexed: 03/08/2024] Open
Abstract
Two-dimensional culture remains widely employed to determine the bioavailability of orally delivered drugs. To gain more knowledge about drug uptake mechanisms and risk assessment for the patient after oral drug admission, intestinal in vitro models demonstrating a closer similarity to the in vivo situation are needed. In particular, Caco-2 cell-based Transwell® models show advantages as they are reproducible, cost-efficient, and standardized. However, cellular complexity is impaired and cell function is strongly modified as important transporters in the apical membrane are missing. To overcome these limitations, primary organoid-based human small intestinal tissue models were developed recently but the application of these cultures in pre-clinical research still represents an enormous challenge, as culture setup is complex as well as time- and cost-intensive. To overcome these hurdles, we demonstrate the establishment of primary organoid-derived intestinal cell lines by immortalization. Besides exhibiting cellular diversity of the organoid, these immortalized cell lines enable a standardized and more cost-efficient culture. Further, our cell line-based Transwell®-like models display an organ-specific epithelial barrier integrity, ultrastructural features and representative transport functions. Altogether, our novel model systems are cost-efficient with close similarity to the in vivo situation, therefore favoring their use in bioavailability studies in the context of pre-clinical screenings.
Collapse
Affiliation(s)
- Christina Fey
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| | | | | | - Spyridon Damigos
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Julia Horstmann
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Marco Metzger
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Daniela Zdzieblo
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Project Center for Stem Cell Process Engineering (PZ-SPT), Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| |
Collapse
|
22
|
Chen X, Yang Y, Mai Q, Ye G, Liu Y, Liu J. Pillar arene Se nanozyme therapeutic systems with dual drive power effectively penetrated mucus layer combined therapy acute lung injury. Biomaterials 2024; 304:122384. [PMID: 38016334 DOI: 10.1016/j.biomaterials.2023.122384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 11/30/2023]
Abstract
siRNA has demonstrated a promising paradigm for therapy of acute lung injury(ALI). However, the pulmonary mucus layer barrier powerfully hinders the therapeutic efficacy. Herein, we proposed to use dual drive power to enhance the mucus permeation of siRNA by constructing the neutral and targeted selenium nanozymes therapeutic system. The multifunctional selenium nanozymes (CWP-Se@Man) were synthesized by modifying with cationic water-soluble pillar arene (CWP) and mannose (Man). After loading CCR2-siRNA, the CWP-Se@Man reached electroneutrality that co-driven by electroneutrality and targeting, the mucus permeation capacity of CWP-Se@Man enhanced by ∼15 fold, thus effectively penetrate pulmonary mucus layer and deliver CCR2-siRNA into macrophages. Moreover, with optimizing the composition of CWP-Se@Man made of CWP (Slutsky, 2013) [5] or CWP (Ichikado et al., 2012) [6], the therapeutic system CWP (Ichikado et al., 2012) [6]-Se@Man showed better biological activities due to smaller size. In inflamed modes, the CWP-Se@Man nanotherapeutic systems loading CCR2-siRNA not only exerted pronounced anti-inflammatory effect through combining inhibit the chemotactic effect and ROS, but also effectively against ALI after blocking the circulatory effect of ROS and inflammatory cytokines. Therefore, this strategy of dual-driving force penetration mucus renders a unique approach for mediating trans-mucus nucleic acid delivery in lungs, and provide a promising treatment for the acute lung injury therapy.
Collapse
Affiliation(s)
- Xu Chen
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Yonglan Yang
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Qiongmei Mai
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Gang Ye
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yanan Liu
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, 518110, China.
| | - Jie Liu
- College of Chemistry and Materials Science, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
23
|
Baldassi D, Ngo TMH, Merkel OM. Optimization of Lung Surfactant Coating of siRNA Polyplexes for Pulmonary Delivery. Pharm Res 2024; 41:77-91. [PMID: 36447020 PMCID: PMC9708138 DOI: 10.1007/s11095-022-03443-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
PURPOSE The aim of this study was to understand how coating with a pulmonary surfactant, namely Alveofact, affects the physicochemical parameters as well as in vitro behavior of polyethylenimine (PEI) polyplexes for pulmonary siRNA delivery. METHODS Alveofact-coated polyplexes were prepared at different Alveofact:PEI coating ratios and analyzed in terms of size, PDI and zeta potential as well as morphology by transmission electron microscopy. The biological behavior was evaluated in a lung epithelial cell line regarding cell viability, cellular uptake via flow cytometry and gene downregulation by qRT-PCR. Furthermore, a 3D ALI culture model was established to test the mucus diffusion and cellular uptake by confocal microscopy as well as gene silencing activity by qRT-PCR. RESULTS After optimizing the coating process by testing different Alveofact:PEI coating ratios, a formulation with suitable parameters for lung delivery was obtained. In lung epithelial cells, Alveofact-coated polyplexes were well tolerated and internalized. Furthermore, the coating improved the siRNA-mediated gene silencing efficiency. Alveofact-coated polyplexes were then tested on a 3D air-liquid interface (ALI) culture model that, by expressing tight junctions and secreting mucus, resembles important traits of the lung epithelium. Here, we identified the optimal Alveofact:PEI coating ratio to achieve diffusion through the mucus layer while retaining gene silencing activity. Interestingly, the latter underlined the importance of establishing appropriate in vitro models to achieve more consistent results that better predict the in vivo activity. CONCLUSION The addition of a coating with pulmonary surfactant to polymeric cationic polyplexes represents a valuable formulation strategy to improve local delivery of siRNA to the lungs.
Collapse
Affiliation(s)
- Domizia Baldassi
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians University of Munich, Butenandtstraße 5, 81377, Munich, Germany
| | - Thi My Hanh Ngo
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians University of Munich, Butenandtstraße 5, 81377, Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians University of Munich, Butenandtstraße 5, 81377, Munich, Germany.
| |
Collapse
|
24
|
Islam MM, Raikwar S. Enhancement of Oral Bioavailability of Protein and Peptide by Polysaccharide-based Nanoparticles. Protein Pept Lett 2024; 31:209-228. [PMID: 38509673 DOI: 10.2174/0109298665292469240228064739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/22/2024]
Abstract
Oral drug delivery is a prevalent and cost-effective method due to its advantages, such as increased drug absorption surface area and improved patient compliance. However, delivering proteins and peptides orally remains a challenge due to their vulnerability to degradation by digestive enzymes, stomach acids, and limited intestinal membrane permeability, resulting in poor bioavailability. The use of nanotechnology has emerged as a promising solution to enhance the bioavailability of these vital therapeutic agents. Polymeric NPs, made from natural or synthetic polymers, are commonly used. Natural polysaccharides, such as alginate, chitosan, dextran, starch, pectin, etc., have gained preference due to their biodegradability, biocompatibility, and versatility in encapsulating various drug types. Their hydrophobic-hydrophilic properties can be tailored to suit different drug molecules.
Collapse
Affiliation(s)
- Md Moidul Islam
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| |
Collapse
|
25
|
Naik DA, Matonis S, Balakrishnan G, Bettinger CJ. Intestinal retentive systems - recent advances and emerging approaches. J Mater Chem B 2023; 12:64-78. [PMID: 38047746 DOI: 10.1039/d3tb01842c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Intestinal retentive devices (IRDs) are devices designed to anchor within the lumen of the intestines for long-term residence in the gastrointestinal tract. IRDs can enable impactful medical device technologies including sustained oral drug delivery systems, indwelling sensors, or real-time diagnostics. The design and testing of IRDs present a myriad of challenges, including precise deployment of the device at desired intestinal locations, secure anchoring within the gastrointestinal tract to allow for natural function, and safe removal of the IRD at user-defined times. Advancing the state-of-the-art of IRD is an interdisciplinary effort that requires innovations such as new materials, novel anchoring mechanisms, and medical device design with consistent input from clinical practitioners and end-users. This perspective briefly reviews the current state-of-the-art for IRDs and charts a path forward to inform the design of future concepts. Specifically, this article will highlight materials, retention mechanisms, and test beds to measure the efficacy of IRDs and their mechanisms. Finally, potential synergies between IRD and other medical device technologies are presented to identify future opportunities.
Collapse
Affiliation(s)
- Durva A Naik
- Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Wean Hall 3325, Pittsburgh, PA 15213, USA.
| | - Spencer Matonis
- Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Wean Hall 3325, Pittsburgh, PA 15213, USA.
| | - Gaurav Balakrishnan
- Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Wean Hall 3325, Pittsburgh, PA 15213, USA.
| | - Christopher J Bettinger
- Materials Science and Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Wean Hall 3325, Pittsburgh, PA 15213, USA.
- Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Scott Hall 4N201, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Meziu E, Shehu K, Koch M, Schneider M, Kraegeloh A. Impact of mucus modulation by N-acetylcysteine on nanoparticle toxicity. Int J Pharm X 2023; 6:100212. [PMID: 37771516 PMCID: PMC10522980 DOI: 10.1016/j.ijpx.2023.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023] Open
Abstract
Human respiratory mucus is a biological hydrogel that forms a protective barrier for the underlying epithelium. Modulation of the mucus layer has been employed as a strategy to enhance transmucosal drug carrier transport. However, a drawback of this strategy is a potential reduction of the mucus barrier properties, in particular in situations with an increased exposure to particles. In this study, we investigated the impact of mucus modulation on its protective role. In vitro mucus was produced by Calu-3 cells, cultivated at the air-liquid interface for 21 days and used for further testing as formed on top of the cells. Analysis of confocal 3D imaging data revealed that after 21 days Calu-3 cells secrete a mucus layer with a thickness of 24 ± 6 μm. Mucus appeared to restrict penetration of 500 nm carboxyl-modified polystyrene particles to the upper 5-10 μm of the layer. Furthermore, a mucus modulation protocol using aerosolized N-acetylcysteine (NAC) was developed. This treatment enhanced the penetration of particles through the mucus down to deeper layers by means of the mucolytic action of NAC. These findings were supported by cytotoxicity data, indicating that intact mucus protects the underlying epithelium from particle-induced effects on membrane integrity. The impact of NAC treatment on the protective properties of mucus was probed by using 50 and 100 nm amine-modified and 50 nm carboxyl-modified polystyrene nanoparticles, respectively. Cytotoxicity was only induced by the amine-modified particles in combination with NAC treatment, implying a reduced protective function of modulated mucus. Overall, our data emphasize the importance of integrating an assessment of the protective function of mucus into the development of therapy approaches involving mucus modulation.
Collapse
Affiliation(s)
- Enkeleda Meziu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Kristela Shehu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Annette Kraegeloh
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| |
Collapse
|
27
|
Hanio S, Möllmert S, Möckel C, Choudhury S, Höpfel AI, Zorn T, Endres S, Schlauersbach J, Scheller L, Keßler C, Scherf-Clavel O, Bellstedt P, Schubert US, Pöppler AC, Heinze KG, Guck J, Meinel L. Bile Is a Selective Elevator for Mucosal Mechanics and Transport. Mol Pharm 2023; 20:6151-6161. [PMID: 37906224 DOI: 10.1021/acs.molpharmaceut.3c00550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Mucus mechanically protects the intestinal epithelium and impacts the absorption of drugs, with a largely unknown role for bile. We explored the impacts of bile on mucosal biomechanics and drug transport within mucus. Bile diffused with square-root-of-time kinetics and interplayed with mucus, leading to transient stiffening captured in Brillouin images and a concentration-dependent change from subdiffusive to Brownian-like diffusion kinetics within the mucus demonstrated by differential dynamic microscopy. Bile-interacting drugs, Fluphenazine and Perphenazine, diffused faster through mucus in the presence of bile, while Metoprolol, a drug with no bile interaction, displayed consistent diffusion. Our findings were corroborated by rat studies, where co-dosing of a bile acid sequestrant substantially reduced the bioavailability of Perphenazine but not Metoprolol. We clustered over 50 drugs based on their interactions with bile and mucin. Drugs that interacted with bile also interacted with mucin but not vice versa. This study detailed the dynamics of mucus biomechanics under bile exposure and linked the ability of a drug to interact with bile to its abbility to interact with mucus.
Collapse
Affiliation(s)
- Simon Hanio
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Stephanie Möllmert
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Conrad Möckel
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Susobhan Choudhury
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Andreas I Höpfel
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Theresa Zorn
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Sebastian Endres
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Jonas Schlauersbach
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Lena Scheller
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Christoph Keßler
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Peter Bellstedt
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Institute for Clinical Chemistry, University of Zürich,Rämistrasse 100, 8091 Zurich, Switzerland
| | - Ulrich S Schubert
- Institute of Organic Chemistry, University of Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter (JCSM), University of Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Ann-Christin Pöppler
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Staudtstrasse 2, 91058 Erlangen, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Josef-Schneider-Strasse 2, 97080 Wuerzburg, Germany
| |
Collapse
|
28
|
Krishna SS, Sudheesh MS, Viswanad V. Liposomal drug delivery to the lungs: a post covid-19 scenario. J Liposome Res 2023; 33:410-424. [PMID: 37074963 DOI: 10.1080/08982104.2023.2199068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/10/2023] [Indexed: 04/20/2023]
Abstract
High local delivery of anti-infectives to the lungs is required for activity against infections of the lungs. The present pandemic has highlighted the potential of pulmonary delivery of anti-infective agents as a viable option for infections like Covid-19, which specifically causes lung infections and mortality. To prevent infections of such type and scale in the future, target-specific delivery of drugs to the pulmonary region is a high-priority area in the field of drug delivery. The suboptimal effect of oral delivery of anti-infective drugs to the lungs due to the poor biopharmaceutical property of the drugs makes this delivery route very promising for respiratory infections. Liposomes have been used as an effective delivery system for drugs due to their biocompatible and biodegradable nature, which can be used effectively for target-specific drug delivery to the lungs. In the present review, we focus on the use of liposomal drug delivery of anti-infectives for the acute management of respiratory infections in the wake of Covid-19 infection.
Collapse
Affiliation(s)
- S Swathi Krishna
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Kochi, India
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Kochi, India
| | - Vidya Viswanad
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Kochi, India
| |
Collapse
|
29
|
Zeng X, Yang M, Liu H, Zhang Z, Hu Y, Shi J, Wang ZH. Light-driven micro/nanomotors in biomedical applications. NANOSCALE 2023; 15:18550-18570. [PMID: 37962424 DOI: 10.1039/d3nr03760f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Nanotechnology brings hope for targeted drug delivery. However, most current drug delivery systems use passive delivery strategies with limited therapeutic efficiency. Over the past two decades, research on micro/nanomotors (MNMs) has flourished in the biomedical field. Compared with other driven methods, light-driven MNMs have the advantages of being reversible, simple to control, clean, and efficient. Under light irradiation, the MNMs can overcome several barriers in the body and show great potential in the treatment of various diseases, such as tumors, and gastrointestinal, cardiovascular and cerebrovascular diseases. Herein, the classification and mechanism of light-driven MNMs are introduced briefly. Subsequently, the applications of light-driven MNMs in overcoming physiological and pathological barriers in the past five years are highlighted. Finally, the future prospects and challenges of light-driven MNMs are discussed as well. This review will provide inspiration and direction for light-driven MNMs to overcome biological barriers in vivo and promote the clinical application of light-driven MNMs in the biomedical field.
Collapse
Affiliation(s)
- Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| |
Collapse
|
30
|
Schmid R, Volcic M, Fischer S, Qu Z, Barth H, Popat A, Kirchhoff F, Lindén M. Surface functionalization affects the retention and bio-distribution of orally administered mesoporous silica nanoparticles in a colitis mouse model. Sci Rep 2023; 13:20175. [PMID: 37978264 PMCID: PMC10656483 DOI: 10.1038/s41598-023-47445-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Besides the many advantages of oral drug administration, challenges like premature drug degradation and limited bioavailability in the gastro-intestinal tract (GIT) remain. A prolonged residence time in the GIT is beneficial for enhancing the therapeutic outcome when treating diseases associated with an increased intestinal clearance rate, like inflammatory bowel disease (IBD). In this study, we synthesized rod-shaped mesoporous silica nanoparticles (MSNs) functionalized with polyethylene glycol (PEG) or hyaluronic acid (HA) and investigated their bio-distribution upon oral administration in vivo. The negatively charged, non-toxic particles showed different accumulation behavior over time in healthy mice and in mice with dextran sulfate sodium (DSS)-induced intestinal inflammation. PEGylated particles were shown to accumulate in the lower intestinal tract of healthy animals, whereas inflammation promoted retention of HA-functionalized particles in this area. Overall systemic absorption was low. However, some particles were detected in organs of mice with DSS-induced colitis, especially in the case of MSN-PEG. The in vivo findings were connected to surface chemistry-related differences in particle adhesion on Caco-2/Raji and mucus-producing Caco-2/Raji/HT29 cell co-culture epithelial models in vitro. While the particle adhesion behavior in vivo was mirrored in the in vitro results, this was not the case for the resorption results, suggesting that the in vitro model does not fully reflect the erosion of the inflamed epithelial tissue. Overall, our study demonstrates the possibility to modulate accumulation and retention of MSNs in the GIT of mice with and without inflammation through surface functionalization, which has important implications for the formulation of nanoparticle-based delivery systems for oral delivery applications.
Collapse
Affiliation(s)
- Roman Schmid
- Inorganic Chemistry II, Ulm University, 89081, Ulm, Germany
| | - Meta Volcic
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Stephan Fischer
- Institute of Experimental and Clinical Pharmacology, and Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Zhi Qu
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology, and Toxicology and Pharmacology of Natural Products, Ulm University Medical Center, Ulm, Germany
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Mika Lindén
- Inorganic Chemistry II, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
31
|
Xu M, Qi Y, Liu G, Song Y, Jiang X, Du B. Size-Dependent In Vivo Transport of Nanoparticles: Implications for Delivery, Targeting, and Clearance. ACS NANO 2023; 17:20825-20849. [PMID: 37921488 DOI: 10.1021/acsnano.3c05853] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Understanding the in vivo transport of nanoparticles provides guidelines for designing nanomedicines with higher efficacy and fewer side effects. Among many factors, the size of nanoparticles plays a key role in controlling their in vivo transport behaviors due to the existence of various physiological size thresholds within the body and size-dependent nano-bio interactions. Encouraged by the evolving discoveries of nanoparticle-size-dependent biological effects, we believe that it is necessary to systematically summarize the size-scaling laws of nanoparticle transport in vivo. In this review, we summarized the size effect of nanoparticles on their in vivo transport along their journey in the body: begin with the administration of nanoparticles via different delivery routes, followed by the targeting of nanoparticles to intended tissues including tumors and other organs, and eventually clearance of nanoparticles through the liver or kidneys. We outlined the tools for investigating the in vivo transport of nanoparticles as well. Finally, we discussed how we may leverage the size-dependent transport to tackle some of the key challenges in nanomedicine translation and also raised important size-related questions that remain to be answered in the future.
Collapse
Affiliation(s)
- Mingze Xu
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Yuming Qi
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Gaoshuo Liu
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Yuanqing Song
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| | - Xingya Jiang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Bujie Du
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, P.R. China
| |
Collapse
|
32
|
Ishiki AK, Neeves KB, Marr DWM. Reversible Microwheel Translation Induced by Polymer Depletion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:15547-15552. [PMID: 37877804 DOI: 10.1021/acs.langmuir.3c01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
For in vivo applications, microbots (μbots) must move, which is a need that has led to designs, such as helical swimmers, that translate through the bulk fluid. We have previously demonstrated that, upon application of a rotating magnetic field, colloidal particles in aqueous systems can be reversibly assembled from superparamagnetic particles into μbots that translate along surfaces using wet friction. Here, we show that high-molecular-weight polymers of a size that approaches the length scale of the gap between the μbot and surface can be excluded, impacting μbot transport. Using xanthan gum as a convenient high-molecular-weight model, we determine that polymer depletion imparts only a weak effect on colloid-surface interactions but has a significant influence on local viscosity, which is an effect great enough to induce a reversal in the μbot translation direction.
Collapse
Affiliation(s)
- Aaron K Ishiki
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Keith B Neeves
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - David W M Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| |
Collapse
|
33
|
Guo Y, Mao Z, Ran F, Sun J, Zhang J, Chai G, Wang J. Nanotechnology-Based Drug Delivery Systems to Control Bacterial-Biofilm-Associated Lung Infections. Pharmaceutics 2023; 15:2582. [PMID: 38004561 PMCID: PMC10674810 DOI: 10.3390/pharmaceutics15112582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.
Collapse
Affiliation(s)
- Yutong Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Ran
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jingfeng Zhang
- The Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315000, China
| | - Guihong Chai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| |
Collapse
|
34
|
Watchorn J, Stuart S, Clasky AJ, Oliveira MH, Burns DC, Gu FX. Transfer-based nuclear magnetic resonance uncovers unique mechanisms for protein-polymer and protein-nanoparticle binding behavior. J Mater Chem B 2023; 11:10121-10130. [PMID: 37824091 DOI: 10.1039/d3tb01668d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Nanoparticle-based drug delivery systems have shown increasing popularity as a means to improve patient outcomes by improving the effectiveness of active pharmaceutical ingredients (APIs). Similarly, nanoparticles have shown success in targeting alternative routes of API administration, such as applying mucoadhesion or mucopenetration to mucosal drug delivery to enhance uptake. While there are many promising examples of mucoadhesive nanomedicines in literature, there are also many examples of contradictory mucoadhesive binding behavior, most prominently in cases using the same nanoparticle materials. We have uncovered mechanistic insights in polymer-protein binding systems using nOe transfer-based NMR and sought to leverage them to explore nanoparticle-protein interactions. We tested several polymer-coated nanoparticles and micellar polymer nanoparticles and evaluated their binding with mucin proteins. We uncovered that the composition and interaction intimacy of polymer moieties that promote mucin binding change when the polymers are incorporated onto nanoparticle surfaces compared to polymer in solution. This change from solution state to nanoparticle coating can enable switching of behavior of these materials from inert to binding, as we observed in polyvinyl pyrrolidone. We also found the nanoparticle core was influential in determining the binding fate of polymer materials, whereas the nanoparticle size did not possess a clear correlation in the ranges we tested (60-270 nm). These experiments demonstrate that identical polymers may switch their binding behavior to mucin as a function of conformational changes that are induced by incorporating the polymers onto the surface of nanoparticles. These NMR-derived insights could be further leveraged to optimize nanoparticle formulations and guide polymer-mediated mucoadhesion.
Collapse
Affiliation(s)
- Jeffrey Watchorn
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.
| | - Samantha Stuart
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.
| | - Matthew H Oliveira
- Division of Engineering Science, University of Toronto, Toronto, Ontario, M5S 2E4, Canada
| | - Darcy C Burns
- Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| | - Frank X Gu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada
- Acceleration Consortium, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
35
|
Jin Z, Gao Q, Wu K, Ouyang J, Guo W, Liang XJ. Harnessing inhaled nanoparticles to overcome the pulmonary barrier for respiratory disease therapy. Adv Drug Deliv Rev 2023; 202:115111. [PMID: 37820982 DOI: 10.1016/j.addr.2023.115111] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The lack of effective treatments for pulmonary diseases presents a significant global health burden, primarily due to the challenges posed by the pulmonary barrier that hinders drug delivery to the lungs. Inhaled nanomedicines, with their capacity for localized and precise drug delivery to specific pulmonary pathologies through the respiratory route, hold tremendous promise as a solution to these challenges. Nevertheless, the realization of efficient and safe pulmonary drug delivery remains fraught with multifaceted challenges. This review summarizes the delivery barriers associated with major pulmonary diseases, the physicochemical properties and drug formulations affecting these barriers, and emphasizes the design advantages and functional integration of nanomedicine in overcoming pulmonary barriers for efficient and safe local drug delivery. The review also deliberates on established nanocarriers and explores drug formulation strategies rooted in these nanocarriers, thereby furnishing essential guidance for the rational design and implementation of pulmonary nanotherapeutics. Finally, this review cast a forward-looking perspective, contemplating the clinical prospects and challenges inherent in the application of inhaled nanomedicines for respiratory diseases.
Collapse
Affiliation(s)
- Zhaokui Jin
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qi Gao
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Keke Wu
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Jiang Ouyang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Weisheng Guo
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xing-Jie Liang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, PR China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China.
| |
Collapse
|
36
|
Zhao S, Zhao Y, Yang X, Zhao T. Recent research advances on oral colon-specific delivery system of nature bioactive components: A review. Food Res Int 2023; 173:113403. [PMID: 37803751 DOI: 10.1016/j.foodres.2023.113403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 10/08/2023]
Abstract
Oral colon-specific delivery system (OCDS) is a targeted approach that aims to directly deliver bioactive compounds directly to the colon following oral administration, thereby enhancing the colonic release of bioactive substances and minimizing adverse reactions. The effectiveness of bioactive substances in the colon hinges on the degree of release, which are affected by various factors including pH, mucosal barrier, delivery time and so on. Therefore, this review provides a comprehensive overview of the key factors affecting oral colon-specific release of bioactive components firstly. Considering the oral safety, this review then mainly focuses on the types of carriers with edible OCDS and preparation strategies for OCDS. Finally, several preparation strategies for loading typical natural bioactive ingredients into oral safe OCDS are reviewed, along with future development prospects.
Collapse
Affiliation(s)
- Shuang Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Tong Zhao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
37
|
Drago SE, Cabibbo M, Craparo EF, Cavallaro G. TAT decorated siRNA polyplexes for inhalation delivery in anti-asthma therapy. Eur J Pharm Sci 2023; 190:106580. [PMID: 37717668 DOI: 10.1016/j.ejps.2023.106580] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/20/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
In this work, a novel protonable copolymer was designed to deliver siRNA through the inhalation route, as an innovative formulation for the management of asthma. This polycation was synthesized by derivatization of α,β-poly(N-2-hydroxyethyl)D,L-aspartamide (PHEA) first with 1,2-Bis(3-aminopropylamino)ethane (bAPAE) and then with a proper amount of maleimide terminated poly(ethylene glycol) (PEG-MLB), with the aim to increase the superficial hydrophilicity of the system, allowing the diffusion trough the mucus layer. Once the complexation ability of the copolymer has been evaluated, obtaining nanosized polyplexes, polyplexes were functionalized on the surface with a thiolated TAT peptide, a cell-penetrating peptide (CPP), exploiting a thiol-ene reaction. TAT decorated polyplexes result to be highly cytocompatible and able to retain the siRNA with a suitable complexation weight ratio during the diffusion process through the mucus. Despite polyplexes establish weak bonds with the mucin chains, these can diffuse efficiently through the mucin layer and therefore potentially able to reach the bronchial epithelium. Furthermore, through cellular uptake studies, it was possible to observe how the obtained polyplexes penetrate effectively in the cytoplasm of bronchial epithelial cells, where they can reduce IL-8 gene expression, after LPS exposure. In the end, in order to obtain a formulation administrable as an inhalable dry powder, polyplexes were encapsulated in mannitol-based microparticles, by spray freeze drying, obtaining highly porous particles with proper technological characteristics that make them potentially administrable by inhalation route.
Collapse
Affiliation(s)
- Salvatore Emanuele Drago
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Marta Cabibbo
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Emanuela Fabiola Craparo
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Gennara Cavallaro
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy; Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM) of Palermo, Palermo, Italy; Advanced Technology and Network Center (ATeN Center), Università di Palermo, Palermo 90133, Italy.
| |
Collapse
|
38
|
Bartlett BA, Feng Y, Fromen CA, Ford Versypt AN. Computational fluid dynamics modeling of aerosol particle transport through lung airway mucosa. Comput Chem Eng 2023; 179:108458. [PMID: 37946856 PMCID: PMC10634618 DOI: 10.1016/j.compchemeng.2023.108458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Delivery of aerosols to the lung can treat various lung diseases. However, the conducting airways are coated by a protective mucus layer with complex properties that make this form of delivery difficult. Mucus is a non-Newtonian fluid and is cleared from the lungs over time by ciliated cells. Further, its gel-like structure hinders the diffusion of particles through it. Any aerosolized treatment of lung diseases must penetrate the mucosal barrier. Using computational fluid dynamics, a model of the airway mucus and periciliary layer was constructed to simulate the transport of impacted aerosol particles. The model predicts the dosage fraction of particles of a certain size that penetrate the mucus and reach the underlying tissue, as well as the distance downstream of the dosage site where tissue concentration is maximized. Reactions that may occur in the mucus are also considered, with simulated data for the interaction of a model virus and an antibody.
Collapse
Affiliation(s)
- Blake A. Bartlett
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Yu Feng
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
| | - Catherine A. Fromen
- Department of Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Ashlee N. Ford Versypt
- School of Chemical Engineering, Oklahoma State University, Stillwater, OK 74078, USA
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| |
Collapse
|
39
|
Wang J, Viola M, Migliorini C, Paoletti L, Arpicco S, Di Meo C, Matricardi P. Polysaccharide-Based Nanogels to Overcome Mucus, Skin, Cornea, and Blood-Brain Barriers: A Review. Pharmaceutics 2023; 15:2508. [PMID: 37896268 PMCID: PMC10610445 DOI: 10.3390/pharmaceutics15102508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Nanocarriers have been extensively developed in the biomedical field to enhance the treatment of various diseases. However, to effectively deliver therapeutic agents to desired target tissues and enhance their pharmacological activity, these nanocarriers must overcome biological barriers, such as mucus gel, skin, cornea, and blood-brain barriers. Polysaccharides possess qualities such as excellent biocompatibility, biodegradability, unique biological properties, and good accessibility, making them ideal materials for constructing drug delivery carriers. Nanogels, as a novel drug delivery platform, consist of three-dimensional polymer networks at the nanoscale, offering a promising strategy for encapsulating different pharmaceutical agents, prolonging retention time, and enhancing penetration. These attractive properties offer great potential for the utilization of polysaccharide-based nanogels as drug delivery systems to overcome biological barriers. Hence, this review discusses the properties of various barriers and the associated constraints, followed by summarizing the most recent development of polysaccharide-based nanogels in drug delivery to overcome biological barriers. It is expected to provide inspiration and motivation for better design and development of polysaccharide-based drug delivery systems to enhance bioavailability and efficacy while minimizing side effects.
Collapse
Affiliation(s)
- Ju Wang
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Marco Viola
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Claudia Migliorini
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Luca Paoletti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy;
| | - Chiara Di Meo
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| | - Pietro Matricardi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Roma, Italy; (J.W.); (M.V.); (C.M.); (L.P.); (C.D.M.)
| |
Collapse
|
40
|
Yan X, Sha X. Nanoparticle-Mediated Strategies for Enhanced Drug Penetration and Retention in the Airway Mucosa. Pharmaceutics 2023; 15:2457. [PMID: 37896217 PMCID: PMC10610050 DOI: 10.3390/pharmaceutics15102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Airway mucus is a complex viscoelastic gel composed mainly of water, glycoproteins, lipids, enzymes, minerals, etc. Among them, glycoproteins are the main factors determining mucus's gel-like rheology. Airway mucus forms a protective barrier by secreting mucin, which represents a barrier for absorption, especially for more lipophilic drugs. It rapidly removes drugs from the airway through the physiological mucus clearance mechanism so drugs cannot remain in the lungs or reach the airway epithelial tissue for a long time. Significant progress has been made in enhancing drug lung deposition recently, but strategies are still needed to help drugs break through the lung mucosal barrier. Based on the physiopathological mechanisms of airway mucus, this paper reviews and summarizes strategies to enhance drug penetration and retention in the airway mucosa mediated by nano-delivery systems, including mucosal permeation systems, mucosal adhesion systems, and enzyme-modified delivery systems. On this basis, the potential and challenges of nano-delivery systems for improving airway mucus clearance are revealed. New ideas and approaches are provided for designing novel nano-delivery systems that effectively improve drug retention and penetration in the airway mucus layer.
Collapse
Affiliation(s)
- Xin Yan
- Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Ministry of Education, Shanghai 201203, China;
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery, School of Pharmacy, Fudan University, Ministry of Education, Shanghai 201203, China;
- The Institutes of Integrative Medicine of Fudan University, 120 Urumqi Middle Road, Shanghai 200040, China
| |
Collapse
|
41
|
Jeong H, Park J, Kang JH, Sabaté del Río J, Kong S, Park T. Organoid-Based Human Stomach Micro-Physiological System to Recapitulate the Dynamic Mucosal Defense Mechanism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300164. [PMID: 37525340 PMCID: PMC10520631 DOI: 10.1002/advs.202300164] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 07/11/2023] [Indexed: 08/02/2023]
Abstract
Several stomach diseases are attributed to the dysregulation of physiological function of gastric mucosal barrier by pathogens. Gastric organoids are a promising tool to develop treatment strategies for gastric infections. However, their functional features of in vivo gastric mucosal barrier and host-microbe interactions are limited due to the lack of physiological stimuli. Herein, a human stomach micro-physiological system (hsMPS) with physiologically relevant gastric mucosal defense system is described based on the combination of organoid and MPS technology. A fluid flow enhanced epithelial-mesenchymal interaction in the hsMPS enables functional maturation of gastric epithelial cells, which allows for the recreation of mesh-like mucus layer containing high level of mucus protective peptides and well-developed epithelial junctional complexes. Furthermore, gastroprotection mechanisms against Helicobacter pylori (H. pylori) are successfully demonstrated in this system. Therefore, hsMPS represents a new in vitro tool for research where gastric mucosal defense mechanism is pivotal for developing therapeutic strategies.
Collapse
Affiliation(s)
- Hye‐Jin Jeong
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Ji‐Hyeon Park
- Department of SurgerySeoul National University HospitalSeoul National University College of MedicineSeoul03080Republic of Korea
- Department of SurgeryGachon University Gil Medical CenterIncheon21565Republic of Korea
| | - Joo H. Kang
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| | - Jonathan Sabaté del Río
- Center for Soft and Living MatterInstitute for Basic Science (IBS)Ulsan44919Republic of Korea
| | - Seong‐Ho Kong
- Department of SurgerySeoul National University HospitalSeoul National University College of MedicineSeoul03080Republic of Korea
| | - Tae‐Eun Park
- Department of Biomedical EngineeringUlsan National Institute of Science and TechnologyUlsan44919Republic of Korea
| |
Collapse
|
42
|
Xie G, Lin S, Wu F, Liu J. Nanomaterial-based ophthalmic drug delivery. Adv Drug Deliv Rev 2023; 200:115004. [PMID: 37433372 DOI: 10.1016/j.addr.2023.115004] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/13/2023]
Abstract
The low bioavailability and side effects of conventional drugs for eye disease necessitate the development of efficient drug delivery systems. Accompanying the developments of nanofabrication techniques, nanomaterials have been recognized as promising tools to overcome these challenges due to their flexible and programmable properties. Given the advances achieved in material science, a broad spectrum of functional nanomaterials capable of overcoming various ocular anterior and posterior segment barriers have been explored to satisfy the demands for ocular drug delivery. In this review, we first highlight the unique functions of nanomaterials suitable for carrying and transporting ocular drugs. Then, various functionalization strategies are emphasized to endow nanomaterials with superior performance in enhanced ophthalmic drug delivery. The rational design of several affecting factors is essential for ideal nanomaterial candidates and is depicted as well. Lastly, we introduce the current applications of nanomaterial-based delivery systems in the therapy of different ocular anterior and posterior segment diseases. The limitations of these delivery systems as well as potential solutions are also discussed. This work will inspire innovative design thinking for the development of nanotechnology-mediated strategies for advanced drug delivery and treatment toward ocular diseases.
Collapse
Affiliation(s)
- Guocheng Xie
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
43
|
Song Q, Yang J, Wu X, Li Y, Zhao H, Feng Q, Zhang Z, Zhang Y, Wang L. A multifunctional integrated biomimetic spore nanoplatform for successively overcoming oral biological barriers. J Nanobiotechnology 2023; 21:302. [PMID: 37641137 PMCID: PMC10463901 DOI: 10.1186/s12951-023-01995-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/10/2023] [Indexed: 08/31/2023] Open
Abstract
The biological barriers have seriously restricted the efficacious responses of oral delivery system in diseases treatment. Utilizing a carrier based on the single construction means is hard to overcome these obstacles simultaneously because the complex gastrointestinal tract environment requires carrier to have different or even contradictory properties. Interestingly, spore capsid (SC) integrates many unique biological characteristics, such as high resistance, good stability etc. This fact offers a boundless source of inspiration for the construction of multi-functional oral nanoplatform based on SC without further modification. Herein, we develop a type of biomimetic spore nanoplatform (SC@DS NPs) to successively overcome oral biological barriers. Firstly, doxorubicin (DOX) and sorafenib (SOR) are self-assembled to form carrier-free nanoparticles (DS NPs). Subsequently, SC is effectively separated from probiotic spores and served as a functional vehicle for delivering DS NPs. As expect, SC@DS NPs can efficaciously pass through the rugged stomach environment after oral administration and further be transported to the intestine. Surprisingly, we find that SC@DS NPs exhibit a significant improvement in the aspects of mucus penetration and transepithelial transport, which is related to the protein species of SC. This study demonstrates that SC@DS NPs can efficiently overcome multiple biological barriers and improve the therapeutic effect.
Collapse
Affiliation(s)
- Qingling Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Junfei Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Xiaocui Wu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Yao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
| | - Hongjuan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Qianhua Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| | - Yun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| | - Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, People's Republic of China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, People's Republic of China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, People's Republic of China.
| |
Collapse
|
44
|
Ma Y, Guo Y, Liu S, Hu Y, Yang C, Cheng G, Xue C, Zuo YY, Sun B. pH-Mediated Mucus Penetration of Zwitterionic Polydopamine-Modified Silica Nanoparticles. NANO LETTERS 2023; 23:7552-7560. [PMID: 37494635 DOI: 10.1021/acs.nanolett.3c02128] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Zwitterionic polymers have emerged as promising trans-mucus nanocarriers due to their superior antifouling properties. However, for pH-sensitive zwitterionic polymers, the effect of the pH microenvironment on their trans-mucus fate remains unclear. In this work, we prepared a library of zwitterionic polydopamine-modified silica nanoparticles (SiNPs-PDA) with an isoelectric point of 5.6. Multiple-particle tracking showed that diffusion of SiNPs-PDA in mucus with a pH value of 5.6 was 3 times faster than that in mucus with pH value 3.0 or 7.0. Biophysical analysis found that the trans-mucus behavior of SiNPs-PDA was mediated by hydrophobic and electrostatic interactions and hydrogen bonding between mucin and the particles. Furthermore, the particle distribution in the stomach, intestine, and lung demonstrated the pH-mediated mucus penetration behavior of the SiNPs-PDA. This study reveals the pH-mediated mucus penetration behavior of zwitterionic nanomaterials, which provides rational design strategies for zwitterionic polymers as nanocarriers in various mucus microenvironments.
Collapse
Affiliation(s)
- Yubin Ma
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shan Liu
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yu Hu
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Cheng Yang
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Gang Cheng
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
45
|
Zhang S, Zhu C, Huang W, Liu H, Yang M, Zeng X, Zhang Z, Liu J, Shi J, Hu Y, Shi X, Wang ZH. Recent progress of micro/nanomotors to overcome physiological barriers in the gastrointestinal tract. J Control Release 2023; 360:514-527. [PMID: 37429360 DOI: 10.1016/j.jconrel.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/12/2023]
Abstract
Oral administration is a convenient administration route for gastrointestinal disease therapy with good patient compliance. But the nonspecific distribution of the oral drugs may cause serious side effects. In recent years, oral drug delivery systems (ODDS) have been applied to deliver the drugs to the gastrointestinal disease sites with decreased side effects. However, the delivery efficiency of ODDS is tremendously limited by physiological barriers in the gastrointestinal sites, such as the long and complex gastrointestinal tract, mucus layer, and epithelial barrier. Micro/nanomotors (MNMs) are micro/nanoscale devices that transfer various energy sources into autonomous motion. The outstanding motion characteristics of MNMs inspired the development of targeted drug delivery, especially the oral drug delivery. However, a comprehensive review of oral MNMs for the gastrointestinal diseases therapy is still lacking. Herein, the physiological barriers of ODDS were comprehensively reviewed. Afterward, the applications of MNMs in ODDS for overcoming the physiological barriers in the past 5 years were highlighted. Finally, future perspectives and challenges of MNMs in ODDS are discussed as well. This review will provide inspiration and direction of MNMs for the therapy of gastrointestinal diseases, pushing forward the clinical application of MNMs in oral drug delivery.
Collapse
Affiliation(s)
- Shuhao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Chaoran Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Wanting Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Xiufang Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China.
| |
Collapse
|
46
|
Qi Y, Jin M, Li Q, Wu Q, Liao Z, Wei M, Fan X, Yang Q, Tian X, Giuseppe B, Luo L. Chitooligosaccharide reconstitutes intestinal mucus layer to improve oral absorption of water-soluble drugs. J Control Release 2023; 360:831-841. [PMID: 37481213 DOI: 10.1016/j.jconrel.2023.07.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
Intestinal mucus is a complex natural hydrogel barrier with unique physical properties that impede the absorption of various oral drugs. Both washout from the upper water layer and the physical resistance of the mucus layer particularly affect bioavailability of, especially, highly water-soluble molecules. One potential strategy for designing pharmaceutical formulations is to add absorption enhancers (AEs). However, there are few reports of AEs that work on mucus and their underlying mechanisms, leading to imprecise application. In this study, we investigated chitooligosaccharide (COS) as a safe, low-cost, and effective oral drug AE. We revealed the hydrodynamic law of interaction between COS and the intestinal mucus layer, which was associated with absorption benefiting mucus structural reconstruction. Based on this, we designed a translational strategy to improve the bioavailability of a group of soluble oral drugs by drinking COS solution before administration. Moreover, this research is expected to expand its application scenario by reducing drug dosage such as avoiding gastro-intestinal irritation and slowing veterinary antibiotic resistance.
Collapse
Affiliation(s)
- Yiming Qi
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ming Jin
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qing Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qinghua Wu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhiqian Liao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Menghao Wei
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinyi Fan
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qianzhan Yang
- Analytical Instruments Department, Analytical Applications Center, Shimadzu (China) Co., Ltd. Chongqing Branch, Chongqing 404100, China
| | - Xiaohe Tian
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Battaglia Giuseppe
- Department of Chemistry and Institute for the Physics of Living Systems, University College London, London WC1H0AJ, United Kingdom
| | - Lei Luo
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
47
|
Zhang B, Zhu L, Pan H, Cai L. Biocompatible smart micro/nanorobots for active gastrointestinal tract drug delivery. Expert Opin Drug Deliv 2023; 20:1427-1441. [PMID: 37840310 DOI: 10.1080/17425247.2023.2270915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION Oral delivery is the most commonly used route of drug administration owing to good patient compliance. However, the gastrointestinal (GI) tract contains multiple physiological barriers that limit the absorption efficiency of conventional passive delivery systems resulting in a low drug concentration reaching the diseased sites. Micro/nanorobots can convert energy to self-propulsive force, providing a novel platform to actively overcome GI tract barriers for noninvasive drug delivery and treatment. AREAS COVERED In this review, we first describe the microenvironments and barriers in the different compartments of the GI tract. Afterward, the applications of micro/nanorobots to overcome GI tract barriers for active drug delivery are highlighted and discussed. Finally, we summarize and discuss the challenges and future prospects of micro/nanorobots for further clinical applications. EXPERT OPINION Micro/nanorobots with the ability to autonomously propel themselves and to load, transport, and release payloads on demand are ideal carriers for active oral drug delivery. Although there are many challenges to be addressed, micro/nanorobots have great potential to introduce a new era of drug delivery for precision therapy.
Collapse
Affiliation(s)
- Baozhen Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lizhen Zhu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
48
|
Li Y, Liu X, Zheng Y, Zhang Y, Li Z, Cui Z, Jiang H, Zhu S, Wu S. Ultrasmall Cortex Moutan Nanoclusters for the Therapy of Pneumonia and Colitis. Adv Healthc Mater 2023; 12:e2300402. [PMID: 36898770 DOI: 10.1002/adhm.202300402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 03/12/2023]
Abstract
Infectious pneumonia and colitis are hard to be treated due to tissue infection, mucosal immune disorders, and dysbacteriosis. Although conventional nanomaterials can eliminate infection, they also damage normal tissues and intestinal flora. Herein, this work reports bactericidal nanoclusters formed through self-assembly for efficient treatment of infectious pneumonia and enteritis. The ultrasmall (about 2.3 nm) cortex moutan nanoclusters (CMNCs) has excellent antibacterial, antiviral, and immune regulation activity. The formation of nanoclusters is analyzed from the molecular dynamics mainly through the binding between polyphenol structures through hydrogen bonding and ππ stacking interaction. CMNCs have enhanced tissue and mucus permeability ability compared with natural CM. CMNCs precisely targeted bacteria due to polyphenol-rich surface structure and inhibited broad spectrum of bacteria. Besides, they killed H1N1 virus mainly through the inhibition of the neuraminidase. These CMNCs are effective in treating infectious pneumonia and enteritis relative to natural CM. In addition, they can be used for adjuvant colitis treatment by protecting colonic epithelium and altering the composition of gut microbiota. Therefore, CMNCs showed excellent application and clinical translation prospects in the treatment of immune and infectious diseases.
Collapse
Affiliation(s)
- Yuan Li
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| | - Xiangmei Liu
- School of Life Science and Health Engineering, Hebei University of Technology, Xiping Avenue 5340, Beichen District, Tianjin, 300401, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhaoyang Li
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| | - Zhenduo Cui
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| | - Hui Jiang
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| | - Shengli Zhu
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| | - Shuilin Wu
- School of Materials Science & Engineering, Peking University, Beijing, 100871, China
- The Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, School of Materials Science & Engineering, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
49
|
Xu C, Xu H, Zhu Z, Shi X, Xiao B. Recent advances in mucus-penetrating nanomedicines for oral treatment of colonic diseases. Expert Opin Drug Deliv 2023; 20:1371-1385. [PMID: 37498079 DOI: 10.1080/17425247.2023.2242266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Oral administration is the most common route for treating colonic diseases that present increased incidences in recent years. Colonic mucus is a critical rate-limiting barrier for the accumulation of oral therapeutics in the colonic tissues. To overcome this obstacle, mucus-penetrating nanotherapeutics have been exploited to increase the accumulated amounts of drugs in the diseased sites and improve their treatment outcomes against colonic diseases. AREAS COVERED In this review, we introduce the structure and composition of colonic mucus as well as its impact on the bioavailability of oral drugs. We also introduce various technologies used in the construction of mucus-penetrating nanomedicines (e.g. surface modification of polymers, physical means and biological strategies) and discuss their mechanisms and potential techniques for improving mucus penetration of nanotherapeutics. EXPERT OPINION The mucus barrier is often overlooked in oral drug delivery. The weak mucus permeability of conventional medications greatly lowers drug bioavailability. This challenge can be addressed through physical, chemical and biological technologies. In addition to the reported methods, promising approaches may be discovered through interdisciplinary research that further helps enhance the mucus penetration of nanomedicines.
Collapse
Affiliation(s)
- Cheng Xu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| | - Haiting Xu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoxiao Shi
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| |
Collapse
|
50
|
Yuan H, Guo C, Liu L, Zhao L, Zhang Y, Yin T, He H, Gou J, Pan B, Tang X. Progress and prospects of polysaccharide-based nanocarriers for oral delivery of proteins/peptides. Carbohydr Polym 2023; 312:120838. [PMID: 37059563 DOI: 10.1016/j.carbpol.2023.120838] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/03/2023]
Abstract
The oral route has long been recognized as the most preferred route for drug delivery as it offers high patient compliance and requires minimal expertise. Unlike small molecule drugs, the harsh environment of the gastrointestinal tract and low permeability across the intestinal epithelium make oral delivery extremely ineffective for macromolecules. Accordingly, delivery systems that are rationally constructed with suitable materials to overcome barriers to oral delivery are exceptionally promising. Among the most ideal materials are polysaccharides. Depending on the interaction between polysaccharides and proteins, the thermodynamic loading and release of proteins in the aqueous phase can be realized. Specific polysaccharides (dextran, chitosan, alginate, cellulose, etc.) endow systems with functional properties, including muco-adhesiveness, pH-responsiveness, and prevention of enzymatic degradation. Furthermore, multiple groups in polysaccharides can be modified, which gives them a variety of properties and enables them to suit specific needs. This review provides an overview of different types of polysaccharide-based nanocarriers based on different kinds of interaction forces and the influencing factors in the construction of polysaccharide-based nanocarriers. Strategies of polysaccharide-based nanocarriers to improve the bioavailability of orally administered proteins/peptides were described. Additionally, current restrictions and future trends of polysaccharide-based nanocarriers for oral delivery of proteins/peptides were also covered.
Collapse
Affiliation(s)
- Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lei Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxuan Zhao
- Department of Pharmaceutics, College of Pharmacy Sciences, Jilin University, Changchun 130021, China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bochen Pan
- Center for Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|