1
|
Yuan H, Xiao P, Wang F, Guo C, Pan S, Jiang M, Hou S, Sun Y, Wang Y, Zhang Y, Yin T, He H, Gou J, Tang X. Linoleic acid co-administration promotes oral delivery of exenatide-loaded butyrate-decorated nanocapsules. J Control Release 2025; 382:113744. [PMID: 40246242 DOI: 10.1016/j.jconrel.2025.113744] [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: 02/05/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Epithelial cell entrance and trans-epithelial transport are two essential processes that directly affect the efficacy of oral delivery of nanocarriers. Herein, a hydroxyethyl starch-based nanocapsule dual decorated with butyrate and octadecylamine (ODA) was first constructed to enhance transporter-mediated endocytosis and trans-epithelial transport, while reducing exenatide (EXT) loss during absorption. The epithelial barrier was then treated with linoleic acid (LA), which functioned as a cell membrane fluidity regulator. This treatment further improved oral delivery efficiency by lowering the energy cost of endocytosis through fluidizing of the cell membrane and increasing monocarboxylate transporter 1 (MCT1) expression on cell surfaces. The findings revealed that LA upregulated MCT1 expression by 3.26-fold, increased the cellular uptake of nanocapsules co-modified with butyrate and ODA by 4.52-fold, decreased ATP consumption for uptake in LA-pretreated Caco-2 cells to only 18.64 % of that in untreated Caco-2 cells, and increased their transcellular transport by 1.72-fold in a Caco-2/HT29-MTX-E12 co-culture monolayer. Therefore, the oral administration of EXT-loaded Bu-PEG-ODA NCs with LA significantly enhanced the oral bioavailability of EXT (Bu-PEG-ODA NCs group: 10.10 %, Bu-PEG-ODA NCs + LA group: 14.84 %), leading to a significant hypoglycemic effect with a 16.06 % relative pharmacological availability. This dosing strategy exhibited efficacious blood glucose control and pancreatic function recovery capabilities in the type 2 diabetes rat model. This study presents a unique co-optimization strategy based on two key processes in the oral absorption of nanocarriers, yielding significant advancements in the oral bioavailability of nanomedicines and improving their therapeutic efficacy.
Collapse
Affiliation(s)
- Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Peifu Xiao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Fan Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Chen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Shu Pan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Mai Jiang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Shicheng Hou
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yunong Sun
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yibo Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
2
|
Wang Y, Ni M, Huang M, Xing L, Liu X, Jia F, Huang Y. Regulation of nanoparticle exocytosis direction via receptors transfer: A novel strategy to enhance therapeutic efficacy of semaglutide. Int J Pharm 2025; 674:125439. [PMID: 40064382 DOI: 10.1016/j.ijpharm.2025.125439] [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: 12/22/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Coumaric acid (CA) is a typical nutrient required in relatively high quantities by the body. It has been proved CA could specifically bind to monocarboxylate Transporter-1 (MCT-1) receptors, a transporter protein expressed on the surface of intestinal epithelial cells, to facilitate its cellular uptake. Although our preliminary research demonstrated semaglutide (SEM) loaded CA modified nanoparticles (SEM@CNP) could improve the absorption of SEM to some extent, the oral bioavailability still remained suboptimal owing to the lysosomal degradation. To address this issue, INF-7 (peptide chain GLFEAIEGFIENGWEGMIDGWYG) and chloroquine (CQ), two lysosomal escape agents (LEAs) with different mechanisms of action, were incorporated with SEM@CNP for oral delivery (SEM@CNP + INF-7, SEM@CNP + CQ). In type II diabetes mice models, SEM@CNP + CQ effectively inhibited postprandial glucose rise with a relative pharmacological bioavailability of 20.63 ± 2.99 %, 1.73 times higher than SEM@CNP (11.90 ± 4.56 %). Mechanistic studies revealed that: 1) after adding LEAs, the exocytosis preference of nanoparticles was altered, tending towards basolateral exocytosis apparently. Regulated exocytosis directionality was linked to the spatial redistribution of MCT-1 receptors. 2) among the two LEAs, CQ demonstrated superior efficacy compared to INF-7. This superiority was attributed to the earlier onset of action and more pronounced degree of membrane disruption induced by CQ. This research provided new insights for the design of oral delivery systems for peptidic drugs.
Collapse
Affiliation(s)
- Yating Wang
- 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
| | - Mingjie Ni
- 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
| | - Minyi 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
| | - Liyun Xing
- 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
| | - Xi Liu
- 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
| | - Fuya Jia
- 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
| | - 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
|
3
|
Zhang Y, Jia X, Wang Y, Zheng Q. Caveolin-1-mediated LDL transcytosis across endothelial cells in atherosclerosis. Atherosclerosis 2025; 402:119113. [PMID: 39914325 DOI: 10.1016/j.atherosclerosis.2025.119113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/20/2024] [Accepted: 01/26/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis is widely recognized as a chronic inflammatory disease of the arterial wall characterized by the progressive accumulation of lipids, inflammatory cells, and fibrous material in the subendothelial space of large arteries. The occurrence and pathogenesis of atherosclerosis are intricately linked to the deposition of low-density lipoprotein (LDL) in the arterial wall. LDL must cross the intact endothelium to reach the subendothelial space, with caveolin-1 assuming a crucial role in this process. Caveolin-1 is a 21-24 kDa membrane protein located in caveolae and highly expressed in endothelial cells. Previous investigations have demonstrated the pivotal role of caveolin-1 in fostering atherosclerosis through its modulation of membrane trafficking, cholesterol metabolism, and cellular signaling. However, how caveolin-1 regulates LDL transcytosis across endothelial cells in atherosclerosis remains unclear. We provide a comprehensive overview of recent research on the interplay between caveolin-1 and atherosclerosis, with a specific focus on elucidating the role of caveolin-1 in mediating LDL transcytosis across endothelial cells. This review furnishes theoretical foundations supporting the pivotal role of caveolin-1 in both the inception and progression of atherosclerosis. It underscores the prospective viability of caveolin-1 as a new therapeutic target for atherosclerosis and introduces novel perspectives for treatment strategies in the early stages of atherosclerosis.
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Xiong Jia
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China.
| |
Collapse
|
4
|
Song T, Yuan L, Wang J, Li W, Sun Y. Advances in the transport of oral nanoparticles in gastrointestinal tract. Colloids Surf B Biointerfaces 2025; 245:114321. [PMID: 39423764 DOI: 10.1016/j.colsurfb.2024.114321] [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: 08/28/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Biological barriers in the gastrointestinal tract (GIT) prevent oral absorption of insoluble drugs. Recently, significant progress has been made in the development of various nanoparticles (NPs) designed to enhance the efficacy of oral drugs. However, the mechanism underlying the intracellular transport of NPs remains unclear, and there are still limitations to improving the oral bioavailability of drugs. This article reviews the challenges faced in the absorption of oral NPs, proposes strategies to overcome these barriers, and discusses the future prospects.
Collapse
Affiliation(s)
- Tingting Song
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Lu Yuan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Jie Wang
- Department of Pharmacy, Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated to Qingdao University, Qingdao 266033, China
| | - Wenjing Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
5
|
Romero-Carmona CE, Chávez-Corona JI, Lima E, Cortés H, Quintanar-Guerrero D, Bernad-Bernad MJ, Ramos-Martínez I, Peña-Corona SI, Sharifi-Rad J, Leyva-Gómez G. Nanoparticle and microparticle-based systems for enhanced oral insulin delivery: A systematic review and meta-analysis. J Nanobiotechnology 2024; 22:802. [PMID: 39734205 DOI: 10.1186/s12951-024-03045-8] [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: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/31/2024] Open
Abstract
Diabetes mellitus (DM) prevalence is rising worldwide. Current therapies comprising subcutaneous insulin injections can cause adverse effects such as lipodystrophy, local reactions like redness and swelling, fluid retention, and allergic reactions. Nanoparticle carriers for oral insulin are groundbreaking compared to existing methods because they are non-invasive treatments, showing operational convenience, controlled release profile, and ability to simulate the physiological delivery route into the bloodstream. These systems improve patient adherence and have demonstrated the potential to lower blood glucose levels in DM. We present a systematic review and meta-analysis aimed at compiling relevant data to pave the way for developing innovative nano- and microparticles for the oral delivery of insulin. Our analysis of 85 articles revealed that the diminution of glucose levels is not proportional to the administered insulin dosage, which ranged from 1 to 120 International Units (IU). The meta-analysis data indicated that 25 IU of encapsulated porcine insulin did not produce a statistically significant outcome (p = 0.93). In contrast, a dosage of 30 IU was efficacious in eliciting an optimal hypoglycemic effect compared to excipient controls. Parameters such as a high degree of encapsulation (~ 90%), particle size (200-400 nm), and polydispersity index (0.086-0.3) are all associated with lower blood glucose levels. These parameters were also significant in the linear regression analysis. Among the excipients employed, chitosan emerged as a prevalent excipient in formulations due to its biocompatible and biodegradable properties and its ability to establish stable polymeric matrices. Even though oral insulin administration is a promising therapeutic method, it cannot guarantee preclinical safety and therapeutic efficacy yet in regulating glucose levels in diabetic conditions.
Collapse
Affiliation(s)
- Carlos E Romero-Carmona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
| | - Juan I Chávez-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
- Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Universidad Nacional Autónoma de México-FESC, Campus 1, 54714, Cuautitlán Izcalli, Mexico
| | - Enrique Lima
- Laboratorio de Fisicoquímica y Reactividad de Superficies (LaFReS), Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), 14389, Ciudad de Mexico, Mexico
| | - David Quintanar-Guerrero
- Laboratorio de Investigación y Posgrado en Tecnología Farmacéutica, Universidad Nacional Autónoma de México-FESC, Campus 1, 54714, Cuautitlán Izcalli, Mexico
| | - María J Bernad-Bernad
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
| | - Iván Ramos-Martínez
- Unidad de Micología, Departamento de Microbiología-Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico.
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico, Mexico.
| |
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
|
Yu Y, Ni M, Zheng Y, Huang Y. Airway epithelial-targeted nanoparticle reverses asthma in inhalation therapy. J Control Release 2024; 367:223-234. [PMID: 38272396 DOI: 10.1016/j.jconrel.2024.01.044] [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: 09/19/2023] [Revised: 12/26/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Despite extensive research on corticosteroids for treating asthma, their short residence time in the lungs has limited their therapeutic effects in vivo. Nanoparticles have been widely investigated for inhaled drug delivery due to their potential benefits in prolonging drugs' residence time in the lungs. However, the retention of nanoparticles may be limited by mucus and ciliated epithelium clearance mechanisms in the airway. Herein, we anchored a neonatal-Fc-receptor-targeted peptide (FcBP) onto "mucus-penetrating" polyethylene glycol (PEG) nanoparticles (PEG-NP). Interestingly, the mucus-permeability of PEG-NP was not impaired by FcBP-functionalization. Moreover, FcBP modification enhanced cellular internalization and exocytosis via specific receptor-mediated processes, which subsequently ameliorated transepithelial transport and prolonged pulmonary retention. Importantly, after loading dexamethasone, FcBP-functionalization could effectively help nanoparticles cross the airway epithelial layer and be endocytosed by inflammatory cells, resulting in a marked decrease in inflammatory cytokines. Finally, FcBP modification significantly enhanced the therapeutic effect of dexamethasone-loaded nanoparticles in asthma mice. This study demonstrates that FcBP-functionalized PEG-NP can overcome multiple obstacles in the airway to prolong the pulmonary retention of drugs, providing a promising strategy for inhalation therapy.
Collapse
Affiliation(s)
- Yinglan Yu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Mingjie Ni
- 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
| | - Yaxian Zheng
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu 610031, Sichuan, China; Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, 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
|
8
|
Chen J, Escoffre JM, Romito O, Iazourene T, Presset A, Roy M, Potier Cartereau M, Vandier C, Wang Y, Wang G, Huang P, Bouakaz A. Enhanced macromolecular substance extravasation through the blood-brain barrier via acoustic bubble-cell interactions. ULTRASONICS SONOCHEMISTRY 2024; 103:106768. [PMID: 38241945 PMCID: PMC10825521 DOI: 10.1016/j.ultsonch.2024.106768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/01/2024] [Accepted: 01/14/2024] [Indexed: 01/21/2024]
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis, regulates influx and efflux transport, and provides protection to the brain tissue. Ultrasound (US) and microbubble (MB)-mediated blood-brain barrier opening is an effective and safe technique for drug delivery in-vitro and in-vivo. However, the exact mechanism underlying this technique is still not fully elucidated. The aim of the study is to explore the contribution of transcytosis in the BBB transient opening using an in-vitro model of BBB. Utilizing a diverse set of techniques, including Ca2+ imaging, electron microscopy, and electrophysiological recordings, our results showed that the combined use of US and MBs triggers membrane deformation within the endothelial cell membrane, a phenomenon primarily observed in the US + MBs group. This deformation facilitates the vesicles transportation of 500 kDa fluorescent Dextran via dynamin-/caveolae-/clathrin- mediated transcytosis pathway. Simultaneously, we observed increase of cytosolic Ca2+ concentration, which is related with increased permeability of the 500 kDa fluorescent Dextran in-vitro. This was found to be associated with the Ca2+-protein kinase C (PKC) signaling pathway. The insights provided by the acoustically-mediated interaction between the microbubbles and the cells delineate potential mechanisms for macromolecular substance permeability.
Collapse
Affiliation(s)
- Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Zhejiang, China; Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | - Oliver Romito
- Inserm UMR 1069 Nutrition, Croissance et Cancer (N2C), Faculté de Médecine, Université de Tours, F-37032, France
| | - Tarik Iazourene
- Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Antoine Presset
- Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Marie Roy
- Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Marie Potier Cartereau
- Inserm UMR 1069 Nutrition, Croissance et Cancer (N2C), Faculté de Médecine, Université de Tours, F-37032, France
| | - Christophe Vandier
- Inserm UMR 1069 Nutrition, Croissance et Cancer (N2C), Faculté de Médecine, Université de Tours, F-37032, France
| | - Yahua Wang
- Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Zhejiang, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Zhejiang, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| | - Ayache Bouakaz
- Inserm UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| |
Collapse
|
9
|
Razmjooei M, Hosseini SMH, Yousefi G, Golmakani MT, Eskandari MH. Exploiting Apical Sodium-Dependent Bile Acid Transporter (ASBT)-Mediated Endocytosis with Multi-Functional Deoxycholic Acid Grafted Alginate Amide Nanoparticles as an Oral Insulin Delivery System. Pharm Res 2024; 41:335-353. [PMID: 38114803 DOI: 10.1007/s11095-023-03641-7] [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: 08/30/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE Oral administration of insulin is a potential candidate for managing diabetes. However, it is obstructed by the gastrointestinal tract barriers resulting in negligible oral bioavailability. METHODS This investigation presents a novel nanocarrier platform designed to address these challenges. In this regard, the process involved amination of sodium alginate by ethylene diamine, followed by its conjugation with deoxycholic acid. RESULTS The resulting DCA@Alg@INS nanocarrier revealed a significantly high insulin loading content of 63.6 ± 1.03% and encapsulation efficiency of 87.6 ± 3.84%, with a particle size of 206 nm and zeta potentials of -3 mV. In vitro studies showed sustained and pH-dependent release profiles of insulin from nanoparticles. In vitro cellular studies, confocal laser scanning microscopy and flow cytometry analysis confirmed the successful attachment and internalization of DCA@Alg@INS nanoparticles in Caco-2 cells. Furthermore, the DCA@Alg@INS demonstrated a superior capacity for cellular uptake and permeability coefficient relative to the insulin solution, exhibiting sixfold and 4.94-fold enhancement, respectively. According to the uptake mechanism studies, the results indicated that DCA@Alg@INS was mostly transported through an energy-dependent active pathway since the uptake of DCA@Alg@INS by cells was significantly reduced in the presence of NaN3 by ~ 92% and at a low temperature of 4°C by ~ 94%. CONCLUSIONS Given the significance of administering insulin through oral route, deoxycholic acid-modified alginate nanoparticles present a viable option to surmount various obstacles presented by the gastrointestinal.
Collapse
Affiliation(s)
- Maryam Razmjooei
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | | | - Gholamhossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Taghi Golmakani
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Mohammad Hadi Eskandari
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran.
| |
Collapse
|
10
|
Yao L, Liu Q, Lei Z, Sun T. Development and challenges of antimicrobial peptide delivery strategies in bacterial therapy: A review. Int J Biol Macromol 2023; 253:126819. [PMID: 37709236 DOI: 10.1016/j.ijbiomac.2023.126819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The escalating global prevalence of antimicrobial resistance poses a critical threat, prompting concerns about its impact on public health. This predicament is exacerbated by the acute shortage of novel antimicrobial agents, a scarcity attributed to the rapid surge in bacterial resistance. This review delves into the realm of antimicrobial peptides, a diverse class of compounds ubiquitously present in plants and animals across various natural organisms. Renowned for their intrinsic antibacterial activity, these peptides provide a promising avenue to tackle the intricate challenge of bacterial resistance. However, the clinical utility of peptide-based drugs is hindered by limited bioavailability and susceptibility to rapid degradation, constraining efforts to enhance the efficacy of bacterial infection treatments. The emergence of nanocarriers marks a transformative approach poised to revolutionize peptide delivery strategies. This review elucidates a promising framework involving nanocarriers within the realm of antimicrobial peptides. This paradigm enables meticulous and controlled peptide release at infection sites by detecting dynamic shifts in microenvironmental factors, including pH, ROS, GSH, and reactive enzymes. Furthermore, a glimpse into the future reveals the potential of targeted delivery mechanisms, harnessing inflammatory responses and intricate signaling pathways, including adenosine triphosphate, macrophage receptors, and pathogenic nucleic acid entities. This approach holds promise in fortifying immunity, thereby amplifying the potency of peptide-based treatments. In summary, this review spotlights peptide nanosystems as prospective solutions for combating bacterial infections. By bridging antimicrobial peptides with advanced nanomedicine, a new therapeutic era emerges, poised to confront the formidable challenge of antimicrobial resistance head-on.
Collapse
Affiliation(s)
- Longfukang Yao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Qianying Liu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
11
|
Shang L, Xie Q, Yang C, Kong L, Zhang Z. Extracellular Vesicles Facilitate the Transportation of Nanoparticles within and between Cells for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42378-42394. [PMID: 37658814 DOI: 10.1021/acsami.3c10237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The interaction between nanoparticles and cells is closely associated with the therapeutic effects of nanomedicine. Nanoparticles could be transported among cells, but the process-related mechanism remains to be further explored. In this study, it was found that endocytosed cationic polymer nanoparticles (cNPs) could be excreted in an extracellular vesicle (EV)-coated form (cNP@EVs). It was deduced that cNPs may pass through early endosomes, multivesicular bodies (MVBs), and autophagic MVBs within cells. Moreover, a high level of autophagy facilitated the exocytosis process. Since EVs were the effective vehicles for conveying biological information and substances, cNP@EVs were proved to be efficient forms for the intercellular transportation of nanoparticles and have the potential as efficient biomimetic drug delivery systems. These properties endowed cNP@EVs with deep penetration and enhanced antitumor activity. Our findings provided a proof-of-concept for understanding the transfer process of nanoparticles among cells and may help us to further utilize EV-mediated transportation of nanoparticles, therefore, expanding its clinical application.
Collapse
Affiliation(s)
- Lihuan Shang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
12
|
Cui Z, Cui S, Qin L, An Y, Zhang X, Guan J, Wong TW, Mao S. Comparison of virus-capsid mimicking biologic-shell based versus polymeric-shell nanoparticles for enhanced oral insulin delivery. Asian J Pharm Sci 2023; 18:100848. [PMID: 37881796 PMCID: PMC10594566 DOI: 10.1016/j.ajps.2023.100848] [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: 03/23/2023] [Revised: 08/15/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023] Open
Abstract
Virus-capsid mimicking mucus-permeable nanoparticles are promising oral insulin carriers which surmount intestinal mucus barrier. However, the impact of different virus-capsid mimicking structure remains unexplored. In this study, utilizing biotin grafted chitosan as the main skeleton, virus-mimicking nanoparticles endowed with biologic-shell (streptavidin coverage) and polymeric-shell (hyaluronic acid/alginate coating) were designed with insulin as a model drug by self-assembly processes. It was demonstrated that biologic-shell mimicking nanoparticles exhibited a higher intestinal trans-mucus (>80%, 10 min) and transmucosal penetration efficiency (1.6-2.2-fold improvement) than polymeric-shell counterparts. Uptake mechanism studies revealed caveolae-mediated endocytosis was responsible for the absorption of biologic-shell mimicking nanoparticles whereas polymeric-shell mimicking nanoparticles were characterized by clathrin-mediated pathway with anticipated lysosomal insulin digestion. Further, in vivo hypoglycemic study indicated that the improved effect of regulating blood sugar levels was virus-capsid structure dependent out of which biologic-shell mimicking nanoparticles presented the best performance (5.1%). Although the findings of this study are encouraging, much more work is required to meet the standards of clinical translation. Taken together, we highlight the external structural dependence of virus-capsid mimicking nanoparticles on the muco-penetrating and uptake mechanism of enterocytes that in turn affecting their in vivo absorption, which should be pondered when engineering virus-mimicking nanoparticles for oral insulin delivery.
Collapse
Affiliation(s)
- Zhixiang Cui
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuman Cui
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Qin
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yalin An
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jian Guan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tin Wui Wong
- Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Malaysia
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Malaysia
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
13
|
Ji K, Yao Y, Wei X, Liu W, Zhang J, Liu Y, Zhang Y, Wang J, Gu Z. Material design for oral insulin delivery. MED-X 2023; 1:7. [PMID: 37485249 PMCID: PMC10357414 DOI: 10.1007/s44258-023-00006-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 07/25/2023]
Abstract
Frequent insulin injections remain the primary method for controlling the blood glucose level of individuals with diabetes mellitus but are associated with low compliance. Accordingly, oral administration has been identified as a highly desirable alternative due to its non-invasive nature. However, the harsh gastrointestinal environment and physical intestinal barriers pose significant challenges to achieving optimal pharmacological bioavailability of insulin. As a result, researchers have developed a range of materials to improve the efficiency of oral insulin delivery over the past few decades. In this review, we summarize the latest advances in material design that aim to enhance insulin protection, permeability, and glucose-responsive release. We also explore the opportunities and challenges of using these materials for oral insulin delivery.
Collapse
Affiliation(s)
- Kangfan Ji
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yuejun Yao
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Xinwei Wei
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Wei Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Juan Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yun Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Yang Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058 Hangzhou, China
- Department of General Surgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016 China
- Zhejiang Laboratory of Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121 China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| |
Collapse
|
14
|
Zhang Y, Wang Y, Li X, Nie D, Liu C, Gan Y. Ligand-modified nanocarriers for oral drug delivery: Challenges, rational design, and applications. J Control Release 2022; 352:813-832. [PMID: 36368493 DOI: 10.1016/j.jconrel.2022.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/15/2022]
Abstract
Ligand-modified nanocarriers (LMNCs) specific to their targets have attracted increasing interest for enhanced oral drug delivery in recent decades. Although the design of LMNCs for enhanced endocytosis and improved exposure of the loaded drugs through the oral route has received abundant attention, it remains unclear how the design influences their transcellular process, especially the key factors affecting their functions. This review discusses the extracellular and cellular barriers to orally administered LMNCs in the gastrointestinal (GI) tract and new discoveries regarding the GI protein corona and the sequential transport barriers that impede the preplanned movements of LMNCs after oral administration. Furthermore, innovative progress in considering key factors (including target selection, ligand properties, and other important factors) in the rational design of LMNCs for oral drug delivery is presented. In particular, some factors that endow LMNCs with efficient transcytosis rather than only endocytosis are highlighted. Finally, the prospects of orally administered LMNCs in disease therapy for the enhanced oral/local bioavailability of active pharmaceutical ingredients, as well as emerging delivery routes, such as lymphatic drug delivery and systemic location-specific drug release based on oral transcellular LMNCs, are discussed.
Collapse
Affiliation(s)
- Yaqi Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Nie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chang Liu
- State Key Laboratory of Drug Research, 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, 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.
| |
Collapse
|
15
|
Ren Y, Wu W, Zhang X. The feasibility of oral targeted drug delivery: gut immune to particulates? Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
16
|
Mimicking natural cholesterol assimilation to elevate the oral delivery of liraglutide for type Ⅱ diabetes therapy. Asian J Pharm Sci 2022; 17:653-665. [DOI: 10.1016/j.ajps.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/15/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
|
17
|
Liu W, Han Y, Xin X, Chen L, Liu Y, Liu C, Zhang X, Jin M, Jin J, Gao Z, Huang W. Biomimetic and temporal-controlled nanocarriers with ileum transporter targeting for achieving oral administration of chemotherapeutic drugs. J Nanobiotechnology 2022; 20:281. [PMID: 35705976 PMCID: PMC9199201 DOI: 10.1186/s12951-022-01460-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Oral chemotherapy is preferred for patients with cancer owing to its multiple advantages, including convenience, better patient compliance, and improved safety. Nevertheless, various physical barriers exist in this route that hamper the development of oral chemotherapeutic formulations, including destruction of drugs in the gastrointestinal tract (GIT), low permeability in enterocytes, and short residence time in the intestine. To overcome these limitations, it is necessary to design an efficient oral drug delivery system with high efficacy and improved safety. RESULTS Herein, we designed novel glycocholic acid (GCA)-functionalized double layer nanoparticles (GCA-NPs), which can act via an endogenous pathway and in a temporally controlled manner in the intestine, to enhance the oral bioavailability of hydrophobic chemotherapeutic drugs such as paclitaxel (PTX). GCA-NPs were composed of quercetin (Qu)-modified liposomes (QL) coated with GCA-chitosan oligosaccharide conjugate (GCOS). The GCA-NPs thus prepared showed prolonged intestinal retention time and good GIT stability due to the presence of chitosan oligosaccharide (COS) and enhanced active transportation via intestinal apical sodium-dependent bile acid transporter (ASBT) due to the presence of GCA. GCA-NPs also efficiently inhibited intestinal P-gp induced by Qu. PTX-loaded GCA-NPs (PTX@GCA-NPs) had a particle size of 84 nm and an entrapment efficiency of 98% with good stability. As a result, the oral bioavailability of PTX was increased 19-fold compared to that of oral Taxol® at the same dose. Oral PTX@GCA-NPs displayed superior antitumor efficacy and better safety than Taxol® when administered intravenously. CONCLUSIONS Our novel drug delivery system showed remarkable efficacy in overcoming multiple limitations and is a promising carrier for oral delivery of multiple drugs, which addresses several challenges in oral delivery in the clinical context.
Collapse
Affiliation(s)
- Wei Liu
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Ying Han
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Xin Xin
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Liqing Chen
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Yanhong Liu
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Chao Liu
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Xintong Zhang
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Mingji Jin
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Jingzhe Jin
- Department of Oncology, The First Hospital of Dandong City, Dandong, Liaoning 118000 People’s Republic of China
| | - Zhonggao Gao
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| | - Wei Huang
- grid.506261.60000 0001 0706 7839State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China ,grid.506261.60000 0001 0706 7839Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050 People’s Republic of China
| |
Collapse
|
18
|
Liu C, Li L, Lyu J, Xiang Y, Chen L, Zhou Z, Huang Y. Split bullets loaded nanoparticles for amplified immunotherapy. J Control Release 2022; 347:199-210. [PMID: 35550911 DOI: 10.1016/j.jconrel.2022.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) play central role in adaptive antitumor immunity, while their function is often hampered by low immunogenicity of tumor tissues and surrounding hostile microenvironment. Herein, a "split bullets" loaded nanoplatform that can bidirectionally injure mitochondria (MT) and endoplasmic reticulum (ER) of tumor cells is developed. After cellular uptake, the released "split bullets" separately target to different subcellular destinations and exert distinct effects on DCs: (1) MT-targeted "bullet" recruits peripheral DCs into tumor sites, due to its capability to trigger adenosine triphosphate release from tumor cells; (2) ER-targeted "bullet" activates tumor-infiltrating DCs, which is attributed to its ability to evoke calreticulin exposure on tumor cells. These effects collectively improve the tropism and reactivity of DCs to tumor-specific antigen in a two-pronged way. As a result of enhanced function of DCs in antigen capture, treatment of the "split bullets" loaded nanoplatform ignites robust immune response to suppress primary melanoma, and establishes systemic immune memory against post-surgical tumor recurrence. Overall, this nanoplatform offers a generalizable approach to boost DCs and augment immunotherapy.
Collapse
Affiliation(s)
- Chendong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiayan Lyu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yucheng Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Liqiang Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhou Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, 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, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
19
|
Liang Y, Ding R, Wang H, Liu L, He J, Tao Y, Zhao Z, Zhang J, Wang A, Sun K, Li Y, Shi Y. Orally administered intelligent self-ablating nanoparticles: a new approach to improve drug cellular uptake and intestinal absorption. Drug Deliv 2022; 29:305-315. [PMID: 35037529 PMCID: PMC8765251 DOI: 10.1080/10717544.2021.2023704] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Oral drug delivery to treat diabetes is being increasingly researched. The mucus and the epithelial cell layers hinder drug delivery. We designed a self-ablating nanoparticle to achieve smart oral delivery to overcome the gastrointestinal barrier. We used the zwitterionic dilauroyl phosphatidylcholine, which exhibits a high affinity toward Oligopeptide transporter 1, to modify poly(lactic-co-glycolic acid) nanoparticles and load hemagglutinin-2 peptide to facilitate its escape from lysosomes. Nanoparticles exhibit a core–shell structure, the lipid layer is degraded by the lysosomes when the nanoparticles are captured by lysosomes, then the inner core of the nanoparticles gets exposed. The results revealed that the self-ablating nanoparticles exhibited higher encapsulation ability than the self-assembled nanoparticles (77% vs 64%) and with better stability. Quantitative cellular uptake, cellular uptake mechanisms, and trans-monolayer cellular were studied, and the results revealed that the cellular uptake achieved using the self-ablating nanoparticles was higher than self-assembling nanoparticles, and the number of uptake pathways via which the self-ablating nanoparticles functioned were higher than the self-assembling nanoparticles. Intestinal mucus permeation, in vivo intestinal circulation, was studied, and the results revealed that the small self-assembling nanoparticles exhibit a good extent of intestinal uptake in the presence of mucus. In vitro flip-flop, intestinal circulation revealed that the uptake of the self-ablating nanoparticles was 1.20 times higher than the self-assembled nanoparticles. Pharmacokinetic study and the pharmacodynamic study showed that the bioavailability and hypoglycemic effect of self-ablating nanoparticles were better than self-assembled nanoparticles.
Collapse
Affiliation(s)
- Yanzi Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Ruihuan Ding
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Huihui Wang
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Lanze Liu
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Jibiao He
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Yuping Tao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Zhenyu Zhao
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Jie Zhang
- School of Life Science, Yantai University, Yantai, P. R. China
| | - Aiping Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P. R. China
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, P. R. China
| | - Yanan Shi
- School of Life Science, Yantai University, Yantai, P. R. China
| |
Collapse
|
20
|
Yu Y, Xing L, Li L, Wu J, He J, Huang Y. Coordination of rigidity modulation and targeting ligand modification on orally-delivered nanoparticles for the treatment of liver fibrosis. J Control Release 2021; 341:215-226. [PMID: 34822908 DOI: 10.1016/j.jconrel.2021.11.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 02/05/2023]
Abstract
Although the individual role of ligand modification or rigidity modulation on oral administration of nanoparticle (NP) has been investigated, how they mutually affect each other remains to be elucidated. Here, we fabricated different rigidity NP with or without surface decoration of FcBP, a neonatal Fc receptor domain-binding peptide. In vitro studies showed that, without FcBP modification, stiff NP had higher transcytosis efficiency across the epithelium than softer NP, due to the different endocytosis mechanisms, intracellular trafficking routes, and exocytosis rate. Notably, after FcBP modification, such difference was narrowed, in a manner that was more favorable for softer NP to "catch up" with stiff NP, suggesting ligand modification was more conducive to exert transcytosis-promoting efficacy on softer NP. In vivo experiments demonstrated that, for ligand-free NP, high rigidity was required for efficient oral absorption and liver distribution. Further FcBP modification decreased that "rigidity threshold", and expanded the feasible rigidity range from stiff NP to softer NP. Upon oral administration, FcBP-modified dexamethasone-loaded softer NP achieved a therapeutic efficacy comparable with stiff NP on alleviating liver fibrosis. Collectively, our study highlighted the necessity of coordinating ligand modification and rigidity modulation for oral drug delivery.
Collapse
Affiliation(s)
- Yinglan Yu
- 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
| | - LiYun Xing
- 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
| | - Lian Li
- 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
| | - Jiawei Wu
- 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
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 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
|
21
|
Durán-Lobato M, López-Estévez AM, Cordeiro AS, Dacoba TG, Crecente-Campo J, Torres D, Alonso MJ. Nanotechnologies for the delivery of biologicals: Historical perspective and current landscape. Adv Drug Deliv Rev 2021; 176:113899. [PMID: 34314784 DOI: 10.1016/j.addr.2021.113899] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
Biological macromolecule-based therapeutics irrupted in the pharmaceutical scene generating a great hope due to their outstanding specificity and potency. However, given their susceptibility to degradation and limited capacity to overcome biological barriers new delivery technologies had to be developed for them to reach their targets. This review aims at analyzing the historical seminal advances that shaped the development of the protein/peptide delivery field, along with the emerging technologies on the lead of the current landscape. Particularly, focus is made on technologies with a potential for transmucosal systemic delivery of protein/peptide drugs, followed by approaches for the delivery of antigens as new vaccination strategies, and formulations of biological drugs in oncology, with special emphasis on mAbs. Finally, a discussion of the key challenges the field is facing, along with an overview of prospective advances are provided.
Collapse
|
22
|
Zhang Y, Xiong M, Ni X, Wang J, Rong H, Su Y, Yu S, Mohammad IS, Leung SSY, Hu H. Virus-Mimicking Mesoporous Silica Nanoparticles with an Electrically Neutral and Hydrophilic Surface to Improve the Oral Absorption of Insulin by Breaking Through Dual Barriers of the Mucus Layer and the Intestinal Epithelium. ACS APPLIED MATERIALS & INTERFACES 2021; 13:18077-18088. [PMID: 33830730 DOI: 10.1021/acsami.1c00580] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Protein and peptide drugs orally suffer from extremely low bioavailability principally for the complicated gastrointestinal environment along with the difficulty of passing through the mucus layer and the underlying epithelium. In our work, we fabricated mesoporous silica nanoparticles with modification groups (MSN-NH2@COOH/CPP5) that effectively penetrated the mucus layer and passed through the intestinal epithelium by mimicking the virus surface. Naked nanoparticles were prepared with inner pores of 6 nm diameter to allow efficient insulin loading and coated with the cationic cell-penetrating KLPVM peptide and the anionic glutaric anhydride to yield hydrophilic MSN-NH2@COOH/CPP5 with a ζ-potential of -0.49 mV. The apparent permeability coefficient of virus-mimicking nanoparticles was 14.61 × 10-5 cm/s. The virus-mimicking nanoparticles showed dramatically lower binding to mucin and faster penetration of the mucus layer than positively charged nanoparticles (MSN@NH2) with a ζ-potential of +35.00 mV. The KLPVM peptide enhanced the uptake of MSN-NH2@COOH/CPP5 by coculturing Caco-2 and E12 cells as an intestinal epithelium model. MSN-NH2@COOH/CPP5 enhanced apical-to-basal transcytosis for being internalized primarily through caveolae-mediated endocytosis. Indeed, for MSN-NH2@COOH/CPP5, the transepithelial transport of the Caco-2 cell monolayer was 2.4-fold higher than MSN@NH2 and 2.0-fold higher than MSN-NH2@COOH. In vitro, loading insulin into nanoparticles maintained the bioactivity of the protein under simulated intestinal conditions. Insulin loaded into MSN-NH2@COOH/CPP5 reduced the diabetic rats' blood glucose level by nearly 50%. The bioavailability of insulin encapsulated in the MSN-NH2@COOH/CPP5 nanoparticles was 2.1-fold more than insulin when administered directly into the jejunum. Nanoparticles with modifications indicated no significant toxicity in in vitro or in vivo preliminary studies. The obstacles of the mucus layer and intestinal epithelium may be effectively conquered by these virus-mimicking nanoparticles for oral delivery of protein and peptide drugs.
Collapse
Affiliation(s)
- Yi Zhang
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Mengting Xiong
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Xiaomin Ni
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jingrou Wang
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Hehui Rong
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Yuqing Su
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Shihui Yu
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Imran Shair Mohammad
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Sharon Shui Yee Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Shatin, New Territories, Hong Kong SAR 999077, P. R. China
| | - Haiyan Hu
- Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
23
|
Liu X, Wu R, Li Y, Wang L, Zhou R, Li L, Xiang Y, Wu J, Xing L, Huang Y. Angiopep-2-functionalized nanoparticles enhance transport of protein drugs across intestinal epithelia by self-regulation of targeted receptors. Biomater Sci 2021; 9:2903-2916. [PMID: 33599658 DOI: 10.1039/d1bm00020a] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ligand-modified nanoparticles (NPs) have been widely used in oral drug delivery systems to promote endocytosis on intestinal epithelia. However, their transcytosis across the intestinal epithelia is still limited. Except for complex intracellular trafficking, recycling again from the apical sides into the intestinal lumen of the endocytosed NPs cannot be ignored. In this study, we modified NP surfaces with angiopep-2 (ANG) that targeted the low-density lipoprotein receptor-related protein 1 (LRP-1) expressed on the intestine to increase both the apical endocytosis and basolateral transcytosis of NPs. Notably, our finding revealed that ANG NPs could increase the apical expression and further basolateral redistribution of LRP-1 on Caco-2 cells, thus generating an apical-to-basolateral absorption pattern. Because of the enhanced transcytosis, insulin loaded ANG NPs possessed much stronger absorption efficiency and induced maximal blood glucose reduction to 61.46% in diabetic rats. Self-regulating the distribution of receptors on polarized intestine cells to promote basolateral transcytosis will provide promising insights for the rational design of oral delivery systems of protein/peptide drugs.
Collapse
Affiliation(s)
- Xi Liu
- 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.
| | - Ruinan Wu
- 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.
| | - Yuting Li
- 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.
| | - Lingling Wang
- 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.
| | - Rui Zhou
- 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.
| | - Lian Li
- 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.
| | - Yucheng Xiang
- 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.
| | - Jiawei Wu
- 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.
| | - Liyun Xing
- 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.
| | - 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
|
24
|
Yang C, He B, Dai W, Zhang H, Zheng Y, Wang X, Zhang Q. The role of caveolin-1 in the biofate and efficacy of anti-tumor drugs and their nano-drug delivery systems. Acta Pharm Sin B 2021; 11:961-977. [PMID: 33996409 PMCID: PMC8105775 DOI: 10.1016/j.apsb.2020.11.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
As one of the most important components of caveolae, caveolin-1 is involved in caveolae-mediated endocytosis and transcytosis pathways, and also plays a role in regulating the cell membrane cholesterol homeostasis and mediating signal transduction. In recent years, the relationship between the expression level of caveolin-1 in the tumor microenvironment and the prognostic effect of tumor treatment and drug treatment resistance has also been widely explored. In addition, the interplay between caveolin-1 and nano-drugs is bidirectional. Caveolin-1 could determine the intracellular biofate of specific nano-drugs, preventing from lysosomal degradation, and facilitate them penetrate into deeper site of tumors by transcytosis; while some nanocarriers could also affect caveolin-1 levels in tumor cells, thereby changing certain biophysical function of cells. This article reviews the role of caveolin-1 in tumor prognosis, chemotherapeutic drug resistance, antibody drug sensitivity, and nano-drug delivery, providing a reference for the further application of caveolin-1 in nano-drug delivery systems.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADC, antibody drug conjugates
- BBB, blood–brain barrier
- Biofate
- CAFs, cancer-associated fibroblasts
- CPT, camptothecin
- CSD, caveolin scaffolding domain
- CTB, cholera toxins B
- Cancer
- Caveolin-1
- Drug resistance
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- ER, endoplasmic reticulum
- ERK, extracellular regulated protein kinases
- FGF2, fibroblast growth factor 2
- GGT, γ-glutamyl transpeptidase
- GPI, glycosylphosphatidylinositol
- HER2, human epidermal growth factor receptor 2
- HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A
- HSA, human serum albumin
- IBC, infiltrating breast cancer
- IR, insulin receptor
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- MSV, multistage nanovectors
- NPs, nanoparticles
- Nano-drug delivery systems
- PC, prostate cancer
- PDGF, platelet-derived growth factor
- PFS, progression free survival
- ROS, reactive oxygen species
- SCLC, small cell lung cancer
- SV40, simian virus 40
- Transcytosis
- cell SMA, styrene maleic acid
Collapse
|
25
|
Yao W, Xu Z, Sun J, Luo J, Wei Y, Zou J. Deoxycholic acid-functionalised nanoparticles for oral delivery of rhein. Eur J Pharm Sci 2021; 159:105713. [PMID: 33453389 DOI: 10.1016/j.ejps.2021.105713] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/09/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022]
Abstract
Rhein (RH) is a candidate for the treatment of kidney diseases. However, clinical application of RH is impeded by low aqueous solubility and oral bioavailability. Deoxycholic acid-conjugated nanoparticles (DNPs) were prepared by ionic interaction for enhancing intestinal absorption by targeting the apical sodium-dependent bile acid transporter in the small intestine. Resultant DNPs showed relatively high entrapment efficiency (90.7 ± 0.73)% and drug-loading efficiency (6.5 ± 0.29)% with a particle size of approximately 190 nm and good overall dispersibility. In vitro release of RH from DNPs exhibited sustained and pH-dependent profiles. Cellular uptake and apparent permeability coefficient (Papp) of the DNPs were 3.25- and 5.05-fold higher than that of RH suspensions, respectively. An in vivo pharmacokinetic study demonstrated significantly enhanced oral bioavailability of RH when encapsulated in DNPs, with 2.40- and 3.33-fold higher Cmax and AUC0-inf compared to RH suspensions, respectively. DNPs are promising delivery platforms for poorly absorbed drugs by oral administration.
Collapse
Affiliation(s)
- Wenjie Yao
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China
| | - Zhishi Xu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China
| | - Jiang Sun
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China
| | - Jingwen Luo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China
| | - Yinghui Wei
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China.
| | - Jiafeng Zou
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, Zhejiang, China
| |
Collapse
|
26
|
Wong CYJ, Al-Salami H, Dass CR. β-Cyclodextrin-containing chitosan-oligonucleotide nanoparticles improve insulin bioactivity, gut cellular permeation and glucose consumption. J Pharm Pharmacol 2021; 73:726-739. [PMID: 33769519 DOI: 10.1093/jpp/rgaa052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/08/2020] [Indexed: 01/25/2023]
Abstract
OBJECTIVES The main objective of the present study was to develop a nanoparticulate drug delivery system that can protect insulin against harsh conditions in the gastrointestinal (GI) tract. The effects of the following employed techniques, including lyophilisation, cross-linking and nanoencapsulation, on the physicochemical properties of the formulation were investigated. METHODS We herein developed a nanocarrier via ionotropic gelation by using positively charged chitosan and negatively charged Dz13Scr. The lyophilised nanoparticles with optimal concentrations of tripolyphosphate (cross-linking agent) and β-cyclodextrin (stabilising agent) were characterised by using physical and cellular assays. KEY FINDINGS The addition of cryoprotectants (1% sucrose) in lyophilisation improved the stability of nanoparticles, enhanced the encapsulation efficiency, and ameliorated the pre-mature release of insulin at acidic pH. The developed lyophilised nanoparticles did not display any cytotoxic effects in C2C12 and HT-29 cells. Glucose consumption assays showed that the bioactivity of entrapped insulin was maintained post-incubation in the enzymatic medium. CONCLUSIONS Freeze-drying with appropriate cryoprotectant could conserve the physiochemical properties of the nanoparticles. The bioactivity of the entrapped insulin was maintained. The prepared nanoparticles could facilitate the permeation of insulin across the GI cell line.
Collapse
Affiliation(s)
- Chun Yuen Jerry Wong
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley,Australia.,Curtin Health Innovation Research Institute, Bentley,Australia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley,Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley,Australia.,Curtin Health Innovation Research Institute, Bentley,Australia
| |
Collapse
|
27
|
Wong CY, Al-Salami H, Dass CR. Fabrication techniques for the preparation of orally administered insulin nanoparticles. J Drug Target 2021; 29:365-386. [PMID: 32876505 DOI: 10.1080/1061186x.2020.1817042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The development of orally administered protein drugs is challenging due to their intrinsic unfavourable features, including large molecular size and poor chemical stability, both of which limit gastrointestinal (GI) absorption efficiency. Nanoparticles can overcome the GI barriers effectively and improve the oral bioavailability of proteins in the GI tract. They possess large surface area to volume ratio, and can facilitate the GI absorption of nanoparticles via the paracellular and transcellular routes. Nanoparticles can be prepared by various fabrication techniques that can encapsulate the fragile therapeutic proteins via hydrophobic bonding and electrostatic interaction. A desirable technique should involve minimal harsh conditions and encapsulate therapeutic proteins with preserved functionalities. The current review examines the characteristics of each preparation technique, and illustrates the examples of insulin-loaded nanoparticles that have been developed in each fabrication method. The following techniques, which include nanoprecipitation, hydrophobic conjugation, flash nanocomplexation, double emulsion, ionotropic gelation, and layer-by-layer adsorption, have been used to formulate ligand-modified nanoparticles for targeted delivery of insulin. Other techniques, including reduction, complex coacervation (polyelectrolyte complexation), hydrophobic ion pairing and emulsion solvent diffusion method, and sol-gel technology, were also discussed in the latter part of the review due to their extensive use in fabrication of insulin nanoparticles. This review also discusses the strategies that have been utilised during the formulation process to improve the stability and bioactivity of therapeutic proteins.
Collapse
Affiliation(s)
- Chun Y Wong
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Australia.,Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|
28
|
Xing L, Zheng Y, Yu Y, Wu R, Liu X, Zhou R, Huang Y. Complying with the physiological functions of Golgi apparatus for secretory exocytosis facilitated oral absorption of protein drugs. J Mater Chem B 2021; 9:1707-1718. [PMID: 33496710 DOI: 10.1039/d0tb02848g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Intestinal epithelial cells are the primary biological barriers for orally administrated nano-formulations and the delivered protein drugs. Thereinto, besides the cellular uptake, intracellular trafficking pathway and the related exocytosis are of great importance to the trans-epithelial transport of drug-loaded NPs. Herein, inspired by the physiological functions of Golgi apparatus for secreting proteins out of cells, Golgi localization-related amino acid l-cysteine (Cys) was modified on the surface of NPs to see whether and how this modification could guide the Golgi pathway-related transport and facilitate the exocytosis of drug-loaded NPs. Meanwhile, cell-penetrating peptide octa-arginine (R8) was co-modified to increase the cellular uptake. The proportion of R8 and Cys modification was explored to get the best effect of endocytosis and exocytosis of NPs. As a result, 25%R8 + 75%Cys NPs with most Cys modification showed efficient transcytosis with the highest transcytosis/endocytosis ratio (0.87). Interestingly, exocytosis mechanism studies indicated that they trafficked through the Golgi secretory pathway and bypassed lysosomes due to Cys modification. The detailed Golgi position mechanism studies further suggested that the thiol group from Cys was important for mediating Golgi transport. In particular, competitive inhibition studies demonstrated that Cys-modified NPs were more conducive to their exocytosis after being transported through the Golgi secretory pathway. We proved that cargos transported via Golgi apparatus tended to be trafficked out of the cells and avoid degradation, which contributed to the transcytosis of 25%R8 + 75%Cys NPs in vitro. Inspiringly, compared with unmodified NPs, 25%R8 + 75%Cys NPs also exhibited promoted intestinal penetration and oral absorption in vivo. Oral delivery of insulin-loaded 25%R8 + 75%Cys NPs showed stronger hypoglycemic effects in diabetic rats. In summary, this work provides a strategy for complying with the physiological functions of Golgi apparatus for secreting to facilitate the exocytosis of NPs, thus further improving the oral absorption of loaded protein drugs.
Collapse
Affiliation(s)
- Liyun Xing
- 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.
| | - Yaxian Zheng
- 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.
| | - Yinglan Yu
- 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.
| | - Ruinan Wu
- 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.
| | - Xi Liu
- 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.
| | - Rui Zhou
- 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.
| | - 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
|
29
|
Guo S, Liang Y, Liu L, Yin M, Wang A, Sun K, Li Y, Shi Y. Research on the fate of polymeric nanoparticles in the process of the intestinal absorption based on model nanoparticles with various characteristics: size, surface charge and pro-hydrophobics. J Nanobiotechnology 2021; 19:32. [PMID: 33499885 PMCID: PMC7839302 DOI: 10.1186/s12951-021-00770-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The use of drug nanocarriers to encapsulate drugs for oral administration may become an important strategy in addressing the challenging oral absorption of some drugs. In this study-with the premise of controlling single variables-we prepared model nanoparticles with different particle sizes, surface charges, and surface hydrophobicity/hydrophilicity. The two key stages of intestinal nanoparticles (NPs) absorption-the intestinal mucus layer penetration stage and the trans-intestinal epithelial cell stage-were decoupled and analyzed. The intestinal absorption of each group of model NPs was then investigated. RESULTS Differences in the behavioral trends of NPs in each stage of intestinal absorption were found to result from differences in particle properties. Small size, low-magnitude negative charge, and moderate hydrophilicity helped NPs pass through the small intestinal mucus layer more easily. Once through the mucus layer, an appropriate size, positive surface charge, and hydrophobic properties helped NPs complete the process of transintestinal epithelial cell transport. CONCLUSIONS To achieve high drug bioavailability, the basic properties of the delivery system must be suitable for overcoming the physiological barrier of the gastrointestinal tract.
Collapse
Affiliation(s)
- Shiqi Guo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Yanzi Liang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Lanze Liu
- College of Life Science, Yantai University, Yantai, 264005, People's Republic of China
| | - Miaomiao Yin
- China Resources Double-crane Pharmaceutical Co., Ltd., Beijing, China
| | - Aiping Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China.
- State Key Laboratory of Long-acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai, China.
| | - Yanan Shi
- College of Life Science, Yantai University, Yantai, 264005, People's Republic of China.
| |
Collapse
|
30
|
Zizzari AT, Pliatsika D, Gall FM, Fischer T, Riedl R. New perspectives in oral peptide delivery. Drug Discov Today 2021; 26:1097-1105. [PMID: 33497830 DOI: 10.1016/j.drudis.2021.01.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/29/2020] [Accepted: 01/18/2021] [Indexed: 12/27/2022]
Abstract
Owing to their structural diversity, peptides are a unique source of innovative active ingredients. However, their development has been challenging because of their disadvantageous pharmacokinetic (PK) properties. Over the past decade, many attempts have been made to improve the oral bioavailability of peptide drugs. In this review, we highlight the most recent and promising techniques aimed at the improvement of the oral bioavailability of peptides. The most recent findings will influence future approaches of pharmaceutical companies in the development of new, more efficient, and safer orally delivered peptides.
Collapse
Affiliation(s)
- Alessandra T Zizzari
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland
| | - Dimanthi Pliatsika
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland
| | - Flavio M Gall
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland
| | - Thomas Fischer
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland
| | - Rainer Riedl
- Center of Organic and Medicinal Chemistry, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences ZHAW, Einsiedlerstrasse 31, 8820 Wädenswil, Switzerland.
| |
Collapse
|
31
|
Zheng Y, Xing L, Chen L, Zhou R, Wu J, Zhu X, Li L, Xiang Y, Wu R, Zhang L, Huang Y. Tailored elasticity combined with biomimetic surface promotes nanoparticle transcytosis to overcome mucosal epithelial barrier. Biomaterials 2020; 262:120323. [DOI: 10.1016/j.biomaterials.2020.120323] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
|
32
|
Guan N, Liu Z, Zhao Y, Li Q, Wang Y. Engineered biomaterial strategies for controlling growth factors in tissue engineering. Drug Deliv 2020; 27:1438-1451. [PMID: 33100031 PMCID: PMC7594870 DOI: 10.1080/10717544.2020.1831104] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/19/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Growth factors are multi-functional signaling molecules that coordinate multi-stage process of wound healing. During wound healing, growth factors are transmitted to wound environment in a positive and physiologically related way, therefore, there is a broad prospect for studying the mediated healing process through growth factors. However, growth factors (GFs) themselves have disadvantages of instability, short life, rapid inactivation of physiological conditions, low safety and easy degradation, which hinder the clinical use of GFs. Rapid development of delivery strategies for GFs has been trying to solve the instability and insecurity of GFs. Particularly, in recent years, GFs delivered by scaffolds based on biomaterials have become a hotspot in this filed. This review introduces various delivery strategies for growth factors based on new biodegradable materials, especially polysaccharides, which could provide guidance for the development of the delivery strategies for growth factors in clinic.
Collapse
Affiliation(s)
- Na Guan
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
| | - Zhihai Liu
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
| | - Yonghui Zhao
- Qingdao Central Hospital, The Second Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Qiu Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, P. R. China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
33
|
The application of biomacromolecules to improve oral absorption by enhanced intestinal permeability: A mini-review. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.02.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Wong CY, Al-Salami H, Dass CR. Formulation and characterisation of insulin-loaded chitosan nanoparticles capable of inducing glucose uptake in skeletal muscle cells in vitro. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101738] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Enhanced Intestinal Absorption of Insulin by Capryol 90, a Novel Absorption Enhancer in Rats: Implications in Oral Insulin Delivery. Pharmaceutics 2020; 12:pharmaceutics12050462. [PMID: 32443624 PMCID: PMC7284608 DOI: 10.3390/pharmaceutics12050462] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 01/13/2023] Open
Abstract
Labrasol® is a self-emulsifying excipient that contains saturated polyglycolysed C6-C14 glycerides and this additive is known to improve the intestinal absorption of poorly absorbed drugs after oral administration. However, the effects of formulations similar to Labrasol® on the intestinal absorption of poorly absorbed drugs have not been characterized. In this study, we used insulin as a model peptide drug and examined the absorption-enhancing effects of Labrasol® and its related formulations for insulin absorption in rats. The co-administration of Labrasol-related formulations with insulin reduced the blood glucose levels. Among these formulations, Capryol 90 was the most effective additive. Notably, the effect of Capryol 90 was greater at pH 3.0 than at pH 7.0. Additionally, almost no mucosal damage was observed in the presence of these formulations, as these formulations did not affect the activity of lactate dehydrogenase (LDH) and the amount of protein released from the small intestine. In mechanistic studies, Capryol 90 improved the stability of insulin and suppressed the association with insulin under acidic conditions. The loosening of the tight junctions (TJs) could be the underlying mechanism by which Capryol 90 improved intestinal insulin absorption via a paracellular route. These findings suggest that Capryol 90 is an effective absorption enhancer for improving the intestinal absorption of insulin, without inducing serious damage to the intestinal epithelium.
Collapse
|
36
|
Wong CY, Al-Salami H, Dass CR. Current status and applications of animal models in pre-clinical development of orally administered insulin-loaded nanoparticles. J Drug Target 2020; 28:882-903. [DOI: 10.1080/1061186x.2020.1759078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Chun Y. Wong
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
| | - Hani Al-Salami
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
- Biotechnology and Drug Development Research Laboratory, Curtin University, Bentley, Australia
| | - Crispin R. Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|
37
|
Durán-Lobato M, Niu Z, Alonso MJ. Oral Delivery of Biologics for Precision Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901935. [PMID: 31222910 DOI: 10.1002/adma.201901935] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/02/2019] [Indexed: 05/23/2023]
Abstract
The emerging field of precision medicine is rapidly growing, fostered by the advances in genome mapping and molecular diagnosis. In general, the translation of these advances into precision treatments relies on the use of biological macromolecules, whose structure offers a high specificity and potency. Unfortunately, due to their complex structure and limited ability to overcome biological barriers, these macromolecules need to be administered via injection. The scientific community has devoted significant effort to making the oral administration of macromolecules plausible thanks to the implementation of drug delivery technologies. Here, an overview of the current situation and future prospects in the field of oral delivery of biologics is provided. Technologies in clinical trials, as well as recent and disruptive delivery systems proposed in the literature for local and systemic delivery of biologics including peptides, antibodies, and nucleic acids, are described. Strategies for the specific targeting of gastrointestinal regions-stomach, small bowel, and colon-cell populations, and internalization pathways, are analyzed. Finally, challenges associated with the clinical translation, future prospects, and identified opportunities for advancement in this field are also discussed.
Collapse
Affiliation(s)
- Matilde Durán-Lobato
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Zhigao Niu
- Riddet Institute, Massey University, Palmerston North, 4442, New Zealand
- Food and Bio-based Products Group, AgResearch Ltd, Palmerston North, 4442, New Zealand
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|
38
|
Wong CY, Al-Salami H, Dass CR. Lyophilisation Improves Bioactivity and Stability of Insulin-Loaded Polymeric-Oligonucleotide Nanoparticles for Diabetes Treatment. AAPS PharmSciTech 2020; 21:108. [PMID: 32215761 DOI: 10.1208/s12249-020-01648-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/21/2020] [Indexed: 12/23/2022] Open
Abstract
The oral bioavailability of therapeutic proteins is limited by the gastrointestinal barriers. Encapsulation of labile proteins into nanoparticles is a promising strategy. In order to improve the stability of nanoparticles, lyophilisation has been used to remove water molecules from the suspension. Although various cryoprotections were employed in the preparation of lyophilised nanoparticles, the selection of cryoprotectant type and concentration in majority of the developed formulation was not justified. In this study, nanoparticles were fabricated by cationic chitosan and anionic Dz13Scr using complex coacervation. The effect of cryoprotectant types (mannitol, sorbitol, sucrose and trehalose) and their concentrations (1, 3, 5, 7, 10% w/v) on physiochemical properties of nanoparticles were measured. Cellular assays were performed to investigate the impact of selected cryoprotectant on cytotoxicity, glucose consumption, oral absorption mechanism and gastrointestinal permeability. The obtained results revealed that mannitol (7% w/v) could produce nanoparticles with small size (313.2 nm), slight positive charge and uniform size distribution. The addition of cryoprotectant could preserve the bioactivity of entrapped insulin and improve the stability of nanoparticles against mechanical stress during lyophilisation. The gastrointestinal absorption of nanoparticles is associated with both endocytic and paracellular pathways. With the use of 7% mannitol, lyophilised nanoparticles induced a significant glucose uptake in C2C12 cells. This work illustrated the importance of appropriate cryoprotectant in conservation of particle physiochemical properties, structural integrity and bioactivity. An incompatible cryoprotectant and inappropriate concentration could lead to cake collapse and formation of heterogeneous particle size populations.
Collapse
|
39
|
Tumor extravasation and infiltration as barriers of nanomedicine for high efficacy: The current status and transcytosis strategy. Biomaterials 2020; 240:119902. [PMID: 32105817 DOI: 10.1016/j.biomaterials.2020.119902] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/15/2020] [Accepted: 02/15/2020] [Indexed: 12/12/2022]
Abstract
Nanotechnology-based drug delivery platforms have been explored for cancer treatments and resulted in several nanomedicines in clinical uses and many in clinical trials. However, current nanomedicines have not met the expected clinical therapeutic efficacy. Thus, improving therapeutic efficacy is the foremost pressing task of nanomedicine research. An effective nanomedicine must overcome biological barriers to go through at least five steps to deliver an effective drug into the cytosol of all the cancer cells in a tumor. Of these barriers, nanomedicine extravasation into and infiltration throughout the tumor are the two main unsolved blockages. Up to now, almost all the nanomedicines are designed to rely on the high permeability of tumor blood vessels to extravasate into tumor interstitium, i.e., the enhanced permeability and retention (EPR) effect or so-called "passive tumor accumulation"; however, the EPR features are not so characteristic in human tumors as in the animal tumor models. Following extravasation, the large size nanomedicines are almost motionless in the densely packed tumor microenvironment, making them restricted in the periphery of tumor blood vessels rather than infiltrating in the tumors and thus inaccessible to the distal but highly malignant cells. Recently, we demonstrated using nanocarriers to induce transcytosis of endothelial and cancer cells to enable nanomedicines to actively extravasate into and infiltrate in solid tumors, which led to radically increased anticancer activity. In this perspective, we make a brief discussion about how active transcytosis can be employed to overcome the difficulties, as mentioned above, and solve the inherent extravasation and infiltration dilemmas of nanomedicines.
Collapse
|
40
|
Wong CY, Al-Salami H, Dass CR. Cellular assays and applied technologies for characterisation of orally administered protein nanoparticles: a systematic review. J Drug Target 2020; 28:585-599. [DOI: 10.1080/1061186x.2020.1726356] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Chun Y. Wong
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
| | - Hani. Al-Salami
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
- Biotechnology and Drug Development Research Laboratory, Curtin University, Bentley, Australia
| | - Crispin R. Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Bentley, Australia
| |
Collapse
|
41
|
Bai Y, Zhou R, Wu L, Zheng Y, Liu X, Wu R, Li X, Huang Y. Nanoparticles with surface features of dendritic oligopeptides as potential oral drug delivery systems. J Mater Chem B 2020; 8:2636-2649. [PMID: 32129375 DOI: 10.1039/c9tb02860a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endowing the NPs with specific surface features of dendritic oligopeptides holds great potential for the oral delivery of peptide/protein drugs.
Collapse
Affiliation(s)
- Yuli Bai
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Rui Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Lei Wu
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Yaxian Zheng
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Xi Liu
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Ruinan Wu
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Xiang Li
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education)
- West China School of Pharmacy
- Sichuan University
- Chengdu 610041
- China
| |
Collapse
|
42
|
Abstract
Protein and peptide therapeutics require parenteral administration, which can be a deterrent to medication adherence. For this reason, there have been extensive efforts to develop alternative delivery strategies, particularly for peptides such as insulin that are used to treat endocrine disorders. Oral delivery is especially desirable, but it faces substantial barriers related to the structural organization and physiological function of the gastrointestinal tract. This article highlights strategies designed to overcome these barriers, including permeation enhancers, inhibitors of gut enzymes, and mucus-penetrating and cell-penetrating peptides. It then focuses on the experience with oral peptides that have reached clinical trials, including insulin, calcitonin, parathyroid hormone and vasopressin, with an emphasis on the advances that have recently led to the landmark approval of an oral formulation of the glucagon-like peptide 1 receptor agonist semaglutide for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
43
|
Wong CY, Luna G, Martinez J, Al-Salami H, Dass CR. Bio-nanotechnological advancement of orally administered insulin nanoparticles: Comprehensive review of experimental design for physicochemical characterization. Int J Pharm 2019; 572:118720. [PMID: 31715357 DOI: 10.1016/j.ijpharm.2019.118720] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Therapeutic proteins are labile macromolecules that are prone to degradation during production, freeze-drying and storage. Recent studies showed that nanoparticles can enhance the stability and oral bioavailability of encapsulated proteins. Several conventional approaches (enzyme inhibitors, mucoadhesive polymers) and novel strategies (surface modification, ligand conjugation, flash nano-complexation, stimuli-responsive drug delivery systems) have been employed to improve the physiochemical properties of nanoparticles such as size, zeta potential, morphology, polydispersity index, drug release kinetics and cell-targeting capacity. However, clinical translation of protein-based nanoparticle is limited due to poor experimental design, protocol non-compliance and instrumentation set-up that do not reflect the physiological conditions, resulting in difficulties in mass production of nanoparticles and waste in research funding. In order to address the above concerns, we conducted a comprehensive review to examine the experimental designs and conditions for physical characterization of protein-based nanoparticles. Reliable and robust characterization is essential to verify the cellular interactions and therapeutic potential of protein-based nanoparticles. Importantly, there are a number of crucial factors, which include sample treatment, analytical method, dispersants, sampling grid, staining, quantification parameters, temperature, drug concentration and research materials, should be taken into careful consideration. Variations in research protocol and unreasonable conditions that are used in optimization of pharmaceutical formulations can have great impact in result interpretation. Last but not least, we reviewed all novel instrumentations and assays that are available to examine mucus diffusion capacity, stability and bioactivity of protein-based nanoparticles. These include circular dichroism, fourier transform infrared spectroscopy, X-ray diffractogram, UV spectroscopy, differential scanning calorimetry, fluorescence spectrum, Förster resonance energy transfer, NMR spectroscopy, Raman spectroscopy, cellular assays and animal models.
Collapse
Affiliation(s)
- Chun Y Wong
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Giuseppe Luna
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley 6102, Australia
| | - Jorge Martinez
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley 6102, Australia
| | - Hani Al-Salami
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia; Biotechnology and Drug Development Research Laboratory, Curtin University, Bentley 6102, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia.
| |
Collapse
|
44
|
Song Q, Zheng C, Jia J, Zhao H, Feng Q, Zhang H, Wang L, Zhang Z, Zhang Y. A Probiotic Spore-Based Oral Autonomous Nanoparticles Generator for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1903793. [PMID: 31490587 DOI: 10.1002/adma.201903793] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/22/2019] [Indexed: 05/19/2023]
Abstract
Spores, the dormant life forms of probiotics, can germinate to metabolically active vegetative cells with the disintegration of their hydrophobic protein coat in the intestinal microenvironment, which provides the possibility for the formation of nanoparticles (NPs) in vivo. Inspired by the natural physiological process of spores, herein, an oral autonomous NPs generator is developed to overcome the spatially variable gastrointestinal tract environment and multibiological barriers. Spores modified with deoxycholic acid (DA) and loaded with chemotherapeutic drugs (doxorubicin and sorafenib, DOX/SOR) serve as an autonomous production line of NPs, which can efficaciously protect the drugs passing through the rugged environment of the stomach and furthermore can be transported to the intestinal environment and colonized rapidly. Subsequently, the DOX/SOR/Spore-DA NPs are produced by the autonomous NPs generator in the intestinal regions based on the disintegrated hydrophobic protein and the hydrophilic DA, and they can efficiently penetrate the epithelial cells via the bile acid pathway, increasing basolateral drug release. In vitro and in vivo studies confirm that this biological nanogenerator can autonomously produce substantial NPs in the intestine, providing a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Qingling Song
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Cuixia Zheng
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Jiajia Jia
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Hongjuan Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Qianhua Feng
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Lei Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| | - Yun Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, 450001, China
| |
Collapse
|
45
|
Kong J, Wu K, Ji Y, Chen K, Zhang J, Sun H, Liang Y, Liang W, Chang Y, Cheng J, Tong J, Li J, Xing G, Chen G. Enhanced Bioavailability by Orally Administered Sirolimus Nanocrystals. ACS APPLIED BIO MATERIALS 2019; 2:4612-4621. [DOI: 10.1021/acsabm.9b00695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jianglong Kong
- College of Food Science, ShiHezi University, ShiHezi 832000, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Kai Wu
- State Key Laboratory of Organic−Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ying Ji
- The University of California’s Center for Environmental Implications of Nanotechnology, Los Angeles, California 90095, United States
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jiaxin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Hui Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Yuelan Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Wei Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Yanan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Jenny Cheng
- The University of California’s Center for Environmental Implications of Nanotechnology, Los Angeles, California 90095, United States
| | - Junmao Tong
- College of Food Science, ShiHezi University, ShiHezi 832000, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China
| | - Guogang Chen
- College of Food Science, ShiHezi University, ShiHezi 832000, China
| |
Collapse
|
46
|
Song Q, Jia J, Niu X, Zheng C, Zhao H, Sun L, Zhang H, Wang L, Zhang Z, Zhang Y. An oral drug delivery system with programmed drug release and imaging properties for orthotopic colon cancer therapy. NANOSCALE 2019; 11:15958-15970. [PMID: 31418432 DOI: 10.1039/c9nr03802g] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oral drug delivery systems (ODDSs) have attracted considerable attention in relation to orthotopic colon cancer therapy due to certain popular advantages. Unfortunately, their clinical applications are generally limited by the side-effects caused by systemic drug exposure and poor real-time monitoring capabilities. Inspired by the characteristics of pH changes of the gastrointestinal tract (GIT) and specific enzymes secreted by the colonic microflora, we anchored polyacrylic acid (PAA) and chitosan (CS) on Gd3+-doped mesoporous hydroxyapatite nanoparticles (Gd-MHAp NPs) to realize programmed drug release and magnetic resonance imaging (MRI) at the tumor sites. In particular, the grafted PAA, as a pH-responsive switch, could effect controlled drug release in the colon. Further, CS is functionalized as the enzyme-sensitive moiety, which could be degraded by β-glycosidase in the colon. Gadolinium is a paramagnetic lanthanide element used in chelates, working as a contrast medium agent for an MRI system. Interestingly, after oral administration, CS and PAA could protect the drug-loaded nanoparticles (NPs) against variable physiological conditions in the GIT, allowing the drug to reach the colon tumor sites, preventing premature drug release. Enhanced drug concentrations at the colon tumor sites were achieved via this programmed drug release, which subsequently ameliorated the therapeutic effect. In addition, encapsulating both chemotherapeutic (5-fluorouracil, 5-FU) and targeted therapy drug (gefitinib, Gef) within Gd-MHAp NPs produced a synergistic therapeutic effect. In summary, this study demonstrated that such a novel drug system (Gd-MHAp/5-FU/Gef/CS/PAA NPs) could protect, transport, and program drug release locally within the colonic environment; further, this system exhibited a worthwhile therapeutic effect, providing a promising novel treatment strategy for orthotopic colon cancer.
Collapse
Affiliation(s)
- Qingling Song
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Arora M, Ganugula R, Kumar N, Kaur G, Pellois JP, Garg P, Kumar MNVR. Next-Generation Noncompetitive Nanosystems Based on Gambogic Acid: In silico Identification of Transferrin Receptor Binding Sites, Regulatory Shelf Stability, and Their Preliminary Safety in Healthy Rodents. ACS APPLIED BIO MATERIALS 2019; 2:3540-3550. [PMID: 31440745 PMCID: PMC6705617 DOI: 10.1021/acsabm.9b00419] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A major challenge in drug delivery is to enhance the transport of drugs across biological barriers, such as the small intestine, the blood-brain barrier, and the blood-retinal/ocular barrier, and to effectively reach the site of action while minimizing the systemic impact. In recent years, piggybacking cell surface receptors have been considered a viable strategy for active drug delivery across the biological barriers. However, the ligands used to target drugs to plasma membrane receptors often have to compete against endogenous ligands, thereby limiting their binding to the cell surface and their transport across barriers. To address this problem, gambogic acid (GA) was identified as a noncompetitive ligand specific to the transferrin receptor (TfR), a receptor present on various barriers. However, the binding sites of the GA on TfR remain unknown, an essential step toward establishing structure-activity relationships. In silico binding site prediction tools, blind docking, and molecular docking simulation confirm that the GA binding site on the TfR is independent of the transferrin-bound iron binding sites. The GA-conjugated polyesters were processed into nanoparticles suitable for drug delivery applications that possess excellent storage stability under regulatory conditions. Traditionally, GA has been used as an anticancer compound that warrants safety assessment. The preliminary studies in healthy rodents on 10-repeated oral doses show no adverse effects. This work will generate paradigm shifting, new knowledge in the field of nanomedicines using unique noncompetitive nanosystems that do not compete with endogenous transferrin.
Collapse
Affiliation(s)
- M. Arora
- † Department of Pharmaceutical Sciences, College of Pharmacy, Reynolds Medical Building, Texas A&M University, Mail Stop 1114, College Station, Texas 77843, United States
| | - R. Ganugula
- † Department of Pharmaceutical Sciences, College of Pharmacy, Reynolds Medical Building, Texas A&M University, Mail Stop 1114, College Station, Texas 77843, United States
| | - N. Kumar
- ‡ Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar 160062, Punjab, India
| | - G. Kaur
- † Department of Pharmaceutical Sciences, College of Pharmacy, Reynolds Medical Building, Texas A&M University, Mail Stop 1114, College Station, Texas 77843, United States
| | - J.-P. Pellois
- § Department of Biochemistry and Biophysics, Texas A&M University, Mail Stop 2128, College Station, Texas 77843, United States
| | - P. Garg
- ‡ Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar 160062, Punjab, India
| | - M. N. V. Ravi Kumar
- † Department of Pharmaceutical Sciences, College of Pharmacy, Reynolds Medical Building, Texas A&M University, Mail Stop 1114, College Station, Texas 77843, United States
| |
Collapse
|