1
|
Li C, Zhang J, Yao X, Huang Y, Zhang Y, Yang W. Red blood cell membrane-camouflaged nanocarriers for the delivery of piperlongumine to treat triple-negative breast cancer. Biomed Mater 2025; 20:035034. [PMID: 40328285 DOI: 10.1088/1748-605x/add4da] [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: 12/11/2024] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
The application of the conventional drugs for triple-negative breast cancer (TNBC) treatment in chemotherapy is limited due to their intrinsic drawbacks such as short drug half-life, lack of tumor selectivity and systemic toxicity. Herein, an effective nanoparticle drug delivery system (NDDS) of red blood cell (RBC) membrane-camouflaged piperlongumine (PL)-loaded iron oxide (Fe3O4) magnetic nanoparticles (Fe3O4-PL@RBC) was rationally designed as an effective drug delivery platform forin vivoTNBC treatment. The Fe3O4-PL@RBC showed considerable cytotoxicity against MDA-MB-231 cells, inducing intracellular accumulation of reactive oxygen species, mitochondrial dysfunction and apoptosis. Furthermore, transcriptomic analyses and western blotting analysis demonstrated that the Fe3O4-PL@RBC induced apoptosis through the inhibition of PI3K/AKT/mTOR pathway and downregulation of Bcl-2 protein. In MDA-MB-231 tumor models, the RBC membrane coating in Fe3O4-PL@RBC effectively prolonged the circulation time and sufficient enrichment at the tumor sites. And the Fe3O4-PL@RBC significantly inhibited tumor growth with good biosafety. This study provides guidance for the rational design of effective Fe3O4-based NDDS for TNBC treatment.
Collapse
Affiliation(s)
- Chenxi Li
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Jiaxin Zhang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Yuxin Huang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Yichen Zhang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| |
Collapse
|
2
|
Pal S, Rakshit T, Saha S, Jinagal D. Glucose-Responsive Materials for Smart Insulin Delivery: From Protein-Based to Protein-Free Design. ACS MATERIALS AU 2025; 5:239-252. [PMID: 40093833 PMCID: PMC11907299 DOI: 10.1021/acsmaterialsau.4c00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 03/19/2025]
Abstract
Over the last four decades, glucose-responsive materials have emerged as promising candidates for developing smart insulin delivery systems, offering an alternative approach to treating diabetes. These materials replicate the pancreas's natural "closed loop" insulin secretion function by detecting changes in blood glucose levels and releasing insulin accordingly. This perspective highlights the evolution of glucose-responsive materials from protein-based materials, such as glucose oxidase (GOx), and glucose-binding proteins, such as concanavalin A (ConA), to protein-free materials, including phenylboronic acid (PBA) and their applications in smart insulin delivery. We first describe protein-based glucose-responsive systems that depend on different macromolecules, including enzymes and proteins, that interact directly with glucose to promote insulin release. However, these systems encounter significant stability, scalability, and immunogenicity challenges. In contrast, protein-free systems include hydrogels, nanogels/microgels, and microneedle patches, offering long-term stability and storability. In this direction, we discuss the design principles, mechanisms of glucose/pH sensitivity, and the disintegration of both protein-based and protein-free systems into different glucose environments. Finally, we outline the key challenges, potential solutions, and prospects for developing smart insulin delivery systems.
Collapse
Affiliation(s)
- Suchetan Pal
- Department
of Bioscience and Biomedical Engineering, Indian Institute of Technology-Bhilai, Durg, 491002, CG India
- Department
of Chemistry, Indian Institute of Technology-Bhilai, Durg, 491002, CG India
| | - Tatini Rakshit
- Department
of Chemistry, Shiv Nadar Institution of
Eminence, Greater
Noida, 201314, UP India
| | - Sunita Saha
- Department
of Chemistry, Indian Institute of Technology-Bhilai, Durg, 491002, CG India
| | - Dharmesh Jinagal
- Department
of Chemistry, Indian Institute of Technology-Bhilai, Durg, 491002, CG India
| |
Collapse
|
3
|
Liang J, Bai M, Bi Y, Jian X, Wang S, Jiang S, Zhao Y, Ma W, Yin S, Zhang W. Heyndrickxia coagulans spore-based nanoparticle generator for improved oral insulin delivery and hypoglycemic therapy. J Control Release 2025; 378:103-115. [PMID: 39657890 DOI: 10.1016/j.jconrel.2024.12.008] [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: 08/12/2024] [Revised: 11/15/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Due to the two major physiological barriers restricted by mucus penetration and epithelia transport, oral insulin therapy using nano-delivery system remains challenging. Heyndrickxia coagulans spores can survive the harsh conditions of gastrointestinal tract (GIT), and penetrate in the mucus through germination to probiotics with their amphipathic proteinaceous coat shedding in the gut epithelium, which makes it possible to be functionalized with hydrophilic peptide/protein and form nanoparticles (NPs) in vivo. Inspired by the natural physiological properties of spores, novel deoxycholic acid-modified Heyndrickxia coagulans spores loaded with insulin (DA-Spore/Ins) as the generators of autonomous bio-based nanoparticles were designed to solve these absorption barriers to enhance oral insulin delivery. The DA-Spore/Ins delivery system achieved preferable drug protection and rapid mucus penetration through its germination in the intestinal microenvironment. Meanwhile, DA-Spore/Ins NPs could be spontaneously formed by the self-assembly of the disintegrated DA-covalently amphipathic protein coat and the hydrophilic protein/peptides drug. This can efficiently transport through the epithelial cells through the bile acid pathway. In vivo studies indicated that DA-Spore/Ins delivery system achieved an oral relative bioavailability of 15.1 % and superior hypoglycemic effect in type I diabetic rats characterized by good biocompatibility. These studies suggested that the intrinsic biological characteristics of Heyndrickxia coagulans spore-based nanogenerators rendered their promising application in oral insulin or other protein drug therapy.
Collapse
Affiliation(s)
- Jinying Liang
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory for Epigenetic Molecular Pharmacology, Xinxiang 453003, China.
| | - Mengxin Bai
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; People's Hospital of Kaifeng, Kaifeng 475002, China
| | - Yarong Bi
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiangjie Jian
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Siyan Wang
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Shang Jiang
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory for Epigenetic Molecular Pharmacology, Xinxiang 453003, China
| | - Weiwei Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Shaoping Yin
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenli Zhang
- Development of pharmaceutics, China Pharmaceutical University, Nangjing 210009, China.
| |
Collapse
|
4
|
Ma J, Li X, Wang C. The Application of Nanomaterials in the Treatment of Pancreatic-Related Diseases. Int J Mol Sci 2024; 25:13158. [PMID: 39684868 DOI: 10.3390/ijms252313158] [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: 10/13/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic diseases, typically including pancreatic cancer, pancreatitis, and diabetes, pose enormous threats to people's lives and health. To date, therapeutics with high therapeutic efficacy and low side effects are still challenging. With the development of nanotechnology, nanomaterials have successfully been applied in pancretic disease treatment. Here, we first introduce the diversity of nanomaterials and the effects of their different physicochemical properties on pancreatic function. Following this, we analyze the potential of nanomaterials to enhance pancreatic targeting by overcoming the challenges of traditional delivery methods through surface modifications, structural adjustments, and optimized drug loading. Then, we introduce the application of structurally optimized nanomaterials to pancreatic-related diseases. For instance, on pancreatic cancer (as drug delivery platforms, for the promotion of radiation therapy, and as multifunctional tools), pancreatitis (as drug delivery systems, anti-inflammatory and anti-fibrotic agents), and diabetes (as insulin delivery carriers, for protecting pancreatic β cells, and for improving insulin resistance). Through analysis of the progress of current research, we summarize how nanomaterials can enhance treatment efficacy while minimizing side effects. Finally, we look forward to the prospects of nanomaterials in pancreatic disease treatment.
Collapse
Affiliation(s)
- Jing Ma
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Li
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Ji K, Wei X, Kahkoska AR, Zhang J, Zhang Y, Xu J, Wei X, Liu W, Wang Y, Yao Y, Huang X, Mei S, Liu Y, Wang S, Zhao Z, Lu Z, You J, Xu G, Shen Y, Buse JB, Wang J, Gu Z. An orally administered glucose-responsive polymeric complex for high-efficiency and safe delivery of insulin in mice and pigs. NATURE NANOTECHNOLOGY 2024; 19:1880-1891. [PMID: 39223256 PMCID: PMC11646558 DOI: 10.1038/s41565-024-01764-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
Contrary to current insulin formulations, endogenous insulin has direct access to the portal vein, regulating glucose metabolism in the liver with minimal hypoglycaemia. Here we report the synthesis of an amphiphilic diblock copolymer comprising a glucose-responsive positively charged segment and polycarboxybetaine. The mixing of this polymer with insulin facilitates the formation of worm-like micelles, achieving highly efficient absorption by the gastrointestinal tract and the creation of a glucose-responsive reservoir in the liver. Under hyperglycaemic conditions, the polymer triggers a rapid release of insulin, establishing a portal-to-peripheral insulin gradient-similarly to endogenous insulin-for the safe regulation of blood glucose. This insulin formulation exhibits a dose-dependent blood-glucose-regulating effect in a streptozotocin-induced mouse model of type 1 diabetes and controls the blood glucose at normoglycaemia for one day in non-obese diabetic mice. In addition, the formulation demonstrates a blood-glucose-lowering effect for one day in a pig model of type 1 diabetes without observable hypoglycaemia, showing promise for the safe and effective management of type 1 diabetes.
Collapse
Affiliation(s)
- Kangfan Ji
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xiangqian Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juan Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yang Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jianchang Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xinwei Wei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Wei Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yuejun Yao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Xuehui Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Shaoqian Mei
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Yun Liu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Shiqi Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Zhengjie Zhao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Ziyi Lu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiahuan You
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Guangzheng Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - John B Buse
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Lee JH, Reischl S, Walter RL, Vieregge V, Weber MC, Xu R, Chen H, Cira K, Kasajima A, Friess H, Neumann PA, Kamaly N. Orally delivered biodegradable targeted inflammation resolving pectin-coated nanoparticles induce anastomotic healing post intestinal surgery. Sci Rep 2024; 14:29253. [PMID: 39587209 PMCID: PMC11589105 DOI: 10.1038/s41598-024-80886-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Targeted perioperative therapeutics supporting anastomotic healing during colitis are an urgent medical need. This study aimed to develop and evaluate a novel nanoparticle (NP)-based drug delivery system for improving anastomotic healing in Inflammatory bowel disease (IBD) patients following surgery. We developed pectin-coated polymeric NPs encapsulating the inflammation-resolving peptide Ac2-26. These NPs are designed to survive gastric passage, facilitate localized release in the colon via microbial pectinase degradation, and bind to the intestinal wound through collagen IV targeting. We investigated these NPs in a murine surgical model combining intestinal anastomosis with preoperative colitis induction. Perioperative administration of pectin-chitosan coated NPs containing Ac2-26 (P-C-Col IV-Ac2-26-NP) reduced colitis activity postoperatively. Macroscopic wound closure improved, as evaluated by endoscopy and intraabdominal adhesion scoring. Microscopic analysis revealed an improved semiquantitative healing score in the treatment group. This proof-of-concept study demonstrates that novel P-C-Col IV-Ac2-26-NP could be a promising and clinically feasible perioperative treatment strategy for IBD patients undergoing intestinal surgery. The targeted delivery system shows potential for enhancing anastomotic healing and reducing postoperative complications in this IBD patient population.
Collapse
Affiliation(s)
- Jong Hyun Lee
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Stefan Reischl
- School of Medicine, Department of Surgery, Technical University of Munich, Munich, Germany
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Robert Leon Walter
- School of Medicine, Department of Surgery, Technical University of Munich, Munich, Germany
| | - Vincent Vieregge
- School of Medicine, Department of Surgery, Technical University of Munich, Munich, Germany
| | - Marie-Christin Weber
- School of Medicine, Department of Surgery, Technical University of Munich, Munich, Germany
| | - Runxin Xu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Hao Chen
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Kamacay Cira
- School of Medicine, Department of Surgery, Technical University of Munich, Munich, Germany
| | - Atsuko Kasajima
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Institute of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | | | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK.
| |
Collapse
|
7
|
Chu H, Xue J, Yang Y, Zheng H, Luo D, Li Z. Advances of Smart Stimulus-Responsive Microneedles in Cancer Treatment. SMALL METHODS 2024; 8:e2301455. [PMID: 38148309 DOI: 10.1002/smtd.202301455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/09/2023] [Indexed: 12/28/2023]
Abstract
Microneedles (MNs) have emerged as a highly promising technology for delivering drugs via the skin. They provide several benefits, including high drug bioavailability, non-invasiveness, painlessness, and high safety. Traditional strategies for intravenous delivery of anti-tumor drugs have risks of systemic toxicity and easy development of drug resistance, while MN technology facilitates precise delivery and on-demand release of drugs in local tissues. In addition, by further combining with stimulus-responsive materials, the construction of smart stimulus-responsive MNs can be achieved, which can respond to specific physical/chemical stimuli from the internal or external environment, thereby further improving the accuracy of tumor treatment and reducing toxicity to surrounding tissues/cells. This review systematically summarizes the classification, materials, and reaction mechanisms of stimulus-responsive MNs, outlines the benefits and challenges of various types of MNs, and details their application and latest progress in cancer treatment. Finally, the development prospects of smart MNs in tumor treatment are also discussed, bringing inspiration for future precision treatment of tumors.
Collapse
Affiliation(s)
- Huaqing Chu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Jiangtao Xue
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuan Yang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| |
Collapse
|
8
|
Feng S, Zhang Y, Hou C, Liu Y, Gao Y, Song Y, Luo J. A temperature-responsive dual-hormone foam nanoengine improves rectal absorptivity of insulin-pramlintide for diabetes treatment. SCIENCE ADVANCES 2024; 10:eadn8695. [PMID: 39196940 PMCID: PMC11352908 DOI: 10.1126/sciadv.adn8695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/24/2024] [Indexed: 08/30/2024]
Abstract
Despite the therapeutic benefits of insulin-pramlintide dual-hormone therapy in diabetes, its application potential has been limited due to a lack of efficient delivery routes. Here, we developed a temperature-responsive dual-hormone foam nanoengine (HormFoam) and combined it with a customized spraying device to further construct an in situ foam-generating system for improving the rectal bioavailability of dual-hormone therapy. To support rapid clinical translation, a continuous microfluidic preparation for HormFoam was proposed, including the power unit of perfluorocarbon nanodroplets and the pharmaceutical components Pluronic F127-functionalized liposomal insulin and pramlintide. We found that HormFoam could consistently generate foams to drive drugs forward after rectal administration, which enhanced intestinal distribution and mucosa absorption, leading to systemic codelivery of insulin-pramlintide. HormFoam reproduced the physiology of endocrine pancreas for glycemic control and induced body weight loss while reversing metabolic disorders in diabetic mice with good biosafety. Therefore, HormFoam represents a state-of-the-art dual-hormone regimen with the potential to address unmet needs in diabetes management.
Collapse
Affiliation(s)
- Shujun Feng
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Chunyuan Hou
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yuta Liu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yujun Song
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China
| | - Jun Luo
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
9
|
Cheng Y, Yang J, Han S, Lu Y. Near-Infrared Triggered Biodegradable Microneedle Patch for Controlled Macromolecule Drug Release. Macromol Biosci 2024; 24:e2400105. [PMID: 38591155 DOI: 10.1002/mabi.202400105] [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: 03/07/2024] [Revised: 04/05/2024] [Indexed: 04/10/2024]
Abstract
Transdermal drug delivery of macromolecule drugs attracts significant attention due to the advantage of convenience and biocompatibility. However, the practical usage of it is limited by the low delivery efficiency and poor drug absorption. To develop an efficient, safe, and controllable transdermal delivery method, the near-infrared (NIR) triggered calcium sulfate and gelatin biodegradable composite microneedle (MN) patches are developed. The MN patches are fabricated by polydimethylsiloxane (PDMS) molds, and the structure data can be adjusted by changing the molds. Such an MN patch can release both macro and micro molecule drugs. After loading with photothermal converter IR780, which can transfer energy of light to heat, the release of macromolecule drugs in MNs can be controlled by applying NIR irradiation. The control effect can be enhanced by spraying 1-tetradecanol (TD) coating and optimizing the ratio (weight) of gelatin and calcium sulfate to 2:6. Besides, the MN patch can deliver drugs through the skin barrier, and the process can be controlled by NIR. Moreover, the insulin-loaded MN patch exhibits some therapeutic effects on healthy mice. This work suggests that biodegradable MNs can achieve controllable drug delivery and potentially be applied in individual treatment via transdermal ingestion.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Junzhu Yang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Sanyang Han
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
10
|
Fuchs S, Caserto JS, Liu Q, Wang K, Shariati K, Hartquist CM, Zhao X, Ma M. A Glucose-Responsive Cannula for Automated and Electronics-Free Insulin Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403594. [PMID: 38639424 PMCID: PMC11223976 DOI: 10.1002/adma.202403594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Automated delivery of insulin based on continuous glucose monitoring is revolutionizing the way insulin-dependent diabetes is treated. However, challenges remain for the widespread adoption of these systems, including the requirement of a separate glucose sensor, sophisticated electronics and algorithms, and the need for significant user input to operate these costly therapies. Herein, a user-centric glucose-responsive cannula is reported for electronics-free insulin delivery. The cannula-made from a tough, elastomer-hydrogel hybrid membrane formed through a one-pot solvent exchange method-changes permeability to release insulin rapidly upon physiologically relevant varying glucose levels, providing simple and automated insulin delivery with no additional hardware or software. Two prototypes of the cannula are evaluated in insulin-deficient diabetic mice. The first cannula-an ends-sealed, subcutaneously inserted prototype-normalizes blood glucose levels for 3 d and controls postprandial glucose levels. The second, more translational version-a cannula with the distal end sealed and the proximal end connected to a transcutaneous injection port-likewise demonstrates tight, 3-d regulation of blood glucose levels when refilled twice daily. This proof-of-concept study may aid in the development of "smart" cannulas and next-generation insulin therapies at a reduced burden-of-care toll and cost to end-users.
Collapse
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Julia S. Caserto
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca NY, 14853, USA
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Kecheng Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Chase M. Hartquist
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
11
|
Zhou J, Ma H, Guan M, Feng J, Dong X, Wei Y, Zhang T. Anti-inflammatory Fucoidan-ConA oral insulin nanosystems for smart blood glucose regulation. Int J Pharm 2024; 659:124250. [PMID: 38777304 DOI: 10.1016/j.ijpharm.2024.124250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/24/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024]
Abstract
The smart oral administration Insulin device has the potential to improve glycemic management. It can reduce the risk of hypoglycemia associated with exogenous Insulin (INS) therapy while also avoiding many of the disadvantages associated with subcutaneous injections. Furthermore, diabetes mellitus (DM) is an endocrine illness characterized by inflammation, and it is critical to minimize the amount of inflammatory markers in diabetic patients while maintaining average blood glucose. In this study, a responsive nanosystem vitamin B12-Fucoidan-Concanavalin A (VB12-FU-ConA NPs) with anti-inflammatory action was developed for smart oral delivery of Insulin. Con A has high sensitivity and strong specificity as a glucose-responsive material. Fucoidan has anti-inflammatory, immunomodulatory, and hypoglycemic functions, and it can bind to Con A to form a reversible complex. Under high glucose conditions, free glucose competitively binds to Con A, which swells the nanocarrier and promotes Insulin release. Furthermore, in the low pH environment of the gastrointestinal tract, positively charged VB12 and anionic fucoidan bind tightly to protect the Insulin wrapped in the carrier, and VB12 can also bind to intestinal epithelial factors to improve transit rate, thereby promoting INS absorption. In vitro tests showed that the release of nanoparticles in hyperglycemic solutions was significantly higher than the drug release in normoglycemic conditions. Oral delivery of the nanosystems dramatically lowered blood glucose levels in type I diabetic mice (T1DM) during in vivo pharmacodynamics, minimizing the risk of hypoglycemia. Blood glucose levels reached a minimum of 8.1 ± 0.4 mmol/L after 8 h. Administering the nanosystem orally notably decreased the serum levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in diabetic mice. The nano delivery system can be degraded and metabolized in the intestinal tract after being taken orally, demonstrating good biodegradability and biosafety. In conclusion, the present study showed that VB12-FU-ConA nanocarriers are expected to be a novel system for rationalizing blood glucose.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Huili Ma
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Min Guan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junfen Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuxin Wei
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
12
|
Pang Y, Li Y, Chen K, Wu M, Zhang J, Sun Y, Xu Y, Wang X, Wang Q, Ning X, Kong D. Porous Microneedles Through Direct Ink Drawing with Nanocomposite Inks for Transdermal Collection of Interstitial Fluid. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305838. [PMID: 38258379 DOI: 10.1002/smll.202305838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/19/2023] [Indexed: 01/24/2024]
Abstract
Interstitial fluid (ISF) is an attractive alternative to regular blood sampling for health checks and disease diagnosis. Porous microneedles (MNs) are well suited for collecting ISF in a minimally invasive manner. However, traditional methods of molding MNs from microfabricated templates involve prohibitive fabrication costs and fixed designs. To overcome these limitations, this study presents a facile and economical additive manufacturing approach to create porous MNs. Compared to traditional layerwise build sequences, direct ink drawing with nanocomposite inks can define sharp MNs with tailored shapes and achieve vastly improved fabrication efficiency. The key to this fabrication strategy is the yield-stress fluid ink that is easily formulated by dispersing silica nanoparticles into the cellulose acetate polymer solution. As-printed MNs are solidified into interconnected porous microstructure inside a coagulation bath of deionized water. The resulting MNs exhibit high mechanical strength and high porosity. This approach also allows porous MNs to be easily integrated on various substrates. In particular, MNs on filter paper substrates are highly flexible to rapidly collect ISF on non-flat skin sites. The extracted ISF is used for quantitative analysis of biomarkers, including glucose, = calcium ions, and calcium ions. Overall, the developments allow facile fabrication of porous MNs for transdermal diagnosis and therapy.
Collapse
Affiliation(s)
- Yushuang Pang
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yanyan Li
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Kerong Chen
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- National Laboratory of Solid State Microstructure, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Ming Wu
- Key Laboratory of High Performance Polymer Materials and Technology of Ministry of Education, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jiaxue Zhang
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yuping Sun
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Yurui Xu
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- National Laboratory of Solid State Microstructure, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Xiaoliang Wang
- Key Laboratory of High Performance Polymer Materials and Technology of Ministry of Education, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qian Wang
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Xinghai Ning
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- National Laboratory of Solid State Microstructure, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Desheng Kong
- College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
13
|
Fouad SA, Ismail AM, Abdel Rafea M, Abu Saied MA, El-Dissouky A. Preparation and Characterization of Chitosan Nanofiber: Kinetic Studies and Enhancement of Insulin Delivery System. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:952. [PMID: 38869577 PMCID: PMC11173695 DOI: 10.3390/nano14110952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/02/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024]
Abstract
Insulin-loaded nanofibers were prepared using chitosan as a natural polymer. The loaded insulin with polyethylene oxide was used for preparing monolayer batch S1. Nanofiber S1 was coated by seven layers of film on both sides to form batch S2 as a sandwich containing Layer A (CS, PEG and PEO) and Layer B (PEG and PEO) using electrospinning apparatus. SEM, TEM and FT-IR techniques were used to confirm the drug loading within the composite nanofibers. The in vitro activity that provided a sustained and controlled release of the drug from the nanofiber batch was studied at different pH values spectrophotometrically using a dialysis method. In batches S1 and S2, the release of insulin from nanofiber proceeds via burst release necessary to produce the desired therapeutic activity, followed by slow step. The rate and the percentage release of insulin in batch S2 are found to be higher at all pH values.
Collapse
Affiliation(s)
- Sarah A. Fouad
- Chemistry Department, Faculty of Science, Alexandria University, Ibrahimia, Alexandria 21321, Egypt; (S.A.F.); (A.E.-D.)
| | - Amel M. Ismail
- Chemistry Department, Faculty of Science, Alexandria University, Ibrahimia, Alexandria 21321, Egypt; (S.A.F.); (A.E.-D.)
| | - M. Abdel Rafea
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - M. A. Abu Saied
- Polymeric Materials Research Department, Advanced Technology and New Materials Research Institute, City of Scientific Research and Technological Applications (SRTA-CITY), New Borg El-Arab, Alexandria 21934, Egypt
| | - Ali El-Dissouky
- Chemistry Department, Faculty of Science, Alexandria University, Ibrahimia, Alexandria 21321, Egypt; (S.A.F.); (A.E.-D.)
| |
Collapse
|
14
|
Attar ES, Jayakumar S, Devarajan PV. Oral In-Situ Nanoplatform with Balanced Hydrophobic-Hydrophilic Property for Transport Across Gastrointestinal Mucosa. AAPS PharmSciTech 2024; 25:113. [PMID: 38750336 DOI: 10.1208/s12249-024-02824-8] [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: 02/22/2024] [Accepted: 04/29/2024] [Indexed: 09/05/2024] Open
Abstract
Transport of oral nanocarriers across the GI epithelium necessitates transport across hydrophilic mucus layer and the hydrophobic epithelium. Based on hydrophobic-hydrophilic balance, Curcumin-Lipomer (lipid-polymer hybrid nanoparticles) comprising hydrophobic stearic acid and hydrophilic Gantrez™ AN 119 (Gantrez) were developed, by a radical in-situ approach, to successfully traverse both barriers. A monophasic preconcentrate (Cur-Pre) comprising Cur (Curcumin), stearic acid, Gantrez and stabilizers, prepared by simple solution, was added to an aqueous phase to instantaneously generate Curcumin-Lipomer (Cur-Lipo) of nanosize and high entrapment efficiency (EE). Cur-Lipo size and EE was optimized by Box-Behnken Design. Cur-Lipomers of varying hydrophobic-hydrophilic property obtained by varying the stearic acid: Gantrez ratio exhibited size in the range 200-400 nm, EE > 95% and spherical morphology as seen in the TEM. A decrease in contact angle and in mucus interaction, evident with increase in Gantrez concentration, indicated an inverse corelation with hydrophilicity, while a linear corelation was observed for mucopenetration and hydrophilicity. Cur-SLN (solid lipid nanoparticles) which served as the hydrophobic reference revealed contact angle > 90°, maximum interaction with mucus and minimal mucopenetration. The ex-vivo permeation study through chicken ileum, revealed maximum permeation with Cur-Lipo1 and comparable and significantly lower permeation of Cur-Lipo1-D and Cur-SLN proposing the importance of balancing the hydrophobic-hydrophilic property of the nanoparticles. A 1.78-fold enhancement in flux of hydrophobic Cur-SLN, with no significant change in permeation of the hydrophilic Cur-Lipomers (p > 0.05) following stripping off the mucosal layer was observed. This reiterated the significance of hydrophobic-hydrophilic balance as a promising strategy to design nanoformulations with superior permeation across the GI barrier.
Collapse
Affiliation(s)
- Esha S Attar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology Matunga (E), Mumbai, 400019, India
| | - S Jayakumar
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - Padma V Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology Matunga (E), Mumbai, 400019, India.
| |
Collapse
|
15
|
Jiang Y, Mi L, Xu X, Hii ARK, Wu Z, Qi X. Urease catalyzed high-density sodium alginate microspheres enable high oral bioavailability of macromolecular drugs. Biomater Sci 2024; 12:1515-1528. [PMID: 38284628 DOI: 10.1039/d3bm01715j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Destruction of insulin caused by the gastric microenvironment and rapid deactivation pose inevitable barriers to oral macromolecular absorption, especially for most peptide and protein drugs. In this study, we developed high-density sodium alginate microspheres composed of magnesium oxide and urease to address these challenges. These microspheres aim to anchor the gastric mucus layer and induce microenvironmental liquefaction, thereby enhancing gastric retention and the protection of insulin. The sedimentation test confirmed the capability of the Ins/Ur/MgO@SA microsphere to rapidly traverse the gastric juice under the influence of gravity. Additionally, the urease immobilized on the Ins/Ur/MgO@SA microspheres catalyzes the hydrolysis of urea in the gastric mucus and promotes the liquefaction of mucus, which is beneficial for microsphere retention. The inclusion of MgO particles and urease, acting as pHM modifiers, helps in adjusting the local pH to avoid gastric acid-induced damage. Subsequently, an in vivo pharmacokinetic experiment verified that the relative bioavailability of the p.o. Ins/Ur/MgO@SA treated group was 15-fold higher than that of the p.o.insulin treated group. Meanwhile, satisfactory blood glucose level (BGL) reduction was observed in diabetic animals. In conclusion, Ins/Ur/MgO@SA microspheres demonstrate high biocompatibility as insulin carriers with prolonged drug release time and increased gastric retention properties, showing a far-reaching strategy for oral macromolecular drug delivery.
Collapse
Affiliation(s)
- Yicheng Jiang
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Li Mi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Xiang Xu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
- King's College London, Institution of Pharmaceutical Science, Franklin Wilkins Building, 150 Stamford St, London SE1 9NH, England, UK
| | - Adric Ru Khiing Hii
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Zhenghong Wu
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
- Industrial Technology Innovation Platform, Zhejiang Center for Safety Study of Drug Substances, Hangzhou 310018, China
| |
Collapse
|
16
|
Gulzar N, Andleeb S, Raza A, Ali S, Liaqat I, Raja SA, Ali NM, Khan R, Awan UA. Acute Toxicity, Anti-diabetic, and Anti-cancerous Potential of Trillium Govanianum-conjugated Silver Nanoparticles in Balb/c Mice. Curr Pharm Biotechnol 2024; 25:1304-1320. [PMID: 37594092 DOI: 10.2174/1389201024666230818124025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/26/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND The current study aimed to develop an economic plant-based therapeutic agent to improve the treatment strategies for diseases at the nano-scale because Cancer and Diabetes mellitus are major concerns in developing countries. Therefore, in vitro and in vivo antidiabetic and anti-cancerous activities of Trillium govanianum conjugated silver nanoparticles were assessed. METHODS In the current study synthesis of silver nanoparticles using Trillium govanianum and characterization were done using a scanning electron microscope, UV-visible spectrophotometer, and FTIR analysis. The in vitro and in vivo anti-diabetic and anti-cancerous potential (200 mg/kg and 400 mg/kg) were carried out. RESULTS It was discovered that Balb/c mice did not show any major alterations during observation of acute oral toxicity when administered orally both TGaqu (1000 mg/kg) and TGAgNPs (1000 mg/kg), and results revealed that 1000 mg/kg is not lethal dose as did not find any abnormalities in epidermal and dermal layers when exposed to TGAgNPs. In vitro studies showed that TGAgNPs could not only inhibit alpha-glucosidase and protein kinases but were also potent against the brine shrimp. Though, a significant reduction in blood glucose levels and significant anti-cancerous effects was recorded when alloxan-treated and CCl4-induced mice were treated with TGAgNPs and TGaqu. CONCLUSION Both in vivo and in vitro studies revealed that TGaqu and TGAgNPs are not toxic at 200 mg/kg, 400 mg/kg, and 1000 mg/kg doses and possess strong anti-diabetic and anti-cancerous effects due to the presence of phyto-constituents. Further, suggesting that green synthesized silver nanoparticles could be used in pharmaceutical industries to develop potent therapeutic agents.
Collapse
Affiliation(s)
- Nazia Gulzar
- Department of Zoology, Microbial Biotechnology Laboratory, University of Azad Jammu and Kashmir, Muzaffarabad, 13100, Pakistan
| | - Saiqa Andleeb
- Department of Zoology, Microbial Biotechnology Laboratory, University of Azad Jammu and Kashmir, Muzaffarabad, 13100, Pakistan
| | - Abida Raza
- PMAS-arid Agriculture University Rawalpindi, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Iram Liaqat
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Sadaf Azad Raja
- Bioscience Department, COMSATS University, Park Road, Chak Shahzad, Islamabad, 44000, Pakistan
| | - Nazish Mazhar Ali
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Rida Khan
- Department of Zoology, Microbial Biotechnology Laboratory, University of Azad Jammu and Kashmir, Muzaffarabad, 13100, Pakistan
| | - Uzma Azeem Awan
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| |
Collapse
|
17
|
Luo D, Ni X, Yang H, Feng L, Chen Z, Bai L. A comprehensive review of advanced nasal delivery: Specially insulin and calcitonin. Eur J Pharm Sci 2024; 192:106630. [PMID: 37949195 DOI: 10.1016/j.ejps.2023.106630] [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: 06/22/2023] [Revised: 10/18/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
Peptide drugs through nasal mucous membrane, such as insulin and calcitonin have been widely used in the medical field. There are always two sides to a coin. One side, intranasal drug delivery can imitate the secretion pattern in human body, having advantages of physiological structure and convenient use. Another side, the low permeability of nasal mucosa, protease environment and clearance effect of nasal cilia hinder the intranasal absorption of peptide drugs. Researchers have taken multiple means to achieve faster therapeutic concentration, lower management dose, and fewer side effects for better nasal preparations. To improve the peptide drugs absorption, various strategies had been explored via the nasal mucosa route. In this paper, we reviewed the achievements of 18 peptide drugs in the past decade about the perspectives of the efficacy, mechanism of enhancing intranasal absorption and safety. The most studies were insulin and calcitonin. As a result, absorption enhancers, nanoparticles (NPs) and bio-adhesive system are the most widely used. Among them, chitosan (CS), cell penetrating peptides (CPPs), tight junction modulators (TJMs), soft NPs and gel/hydrogel are the most promising strategies. Moreover, two or three strategies can be combined to prepare drug vectors. In addition, spray freeze dried (SFD), self-emulsifying nano-system (SEN), and intelligent glucose reaction drug delivery system are new research directions in the future.
Collapse
Affiliation(s)
- Dan Luo
- Department of Pharmacy, Shantou Hospital of Traditional Chinese Medicine, Shantou, Guangdong, China
| | - Xiaoqing Ni
- 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, China
| | - Hao Yang
- Power China Chengdu Engineering Corporation Limited, Chengdu, Sichuan, China
| | - Lu Feng
- Department of Emergency, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
| | - Zhaoqun Chen
- Department of Pharmacy, Shantou Hospital of Traditional Chinese Medicine, Shantou, Guangdong, China.
| | - Lan Bai
- 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, China; The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
18
|
de Groen PC. A new, all-encompassing aetiology of type 1 diabetes. Immunology 2024; 171:77-91. [PMID: 37772700 DOI: 10.1111/imm.13700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/10/2023] [Indexed: 09/30/2023] Open
Abstract
The aetiology of type 1 diabetes (T1D) is considered multifactorial with the contribution of the MHC on chromosome 6 being most important. Multiple factors also contribute to the aetiology of colorectal neoplasia, but the final event causing the change from normal mucosa to polyp and from polyp to cancer is due to a single somatic mutation event. Repeated formation of colorectal neoplasia within an at-risk population results in a predictable, tapering, exponential neoplasia distribution. Critical mutations driving colorectal neoplasia formation occur in mutation-prone DNA. These observations led to three hypotheses related to T1D. First, a single somatic mutation within the MHC of antigen presenting cells results in a change in phenotype from normal to T1D. Second, the distribution of additional autoimmune diseases (AAIDs) among persons with T1D adheres to a predictable, tapering, exponential distribution. And third, critical mutations driving development of T1D occur in mutation-prone DNA. To address the hypotheses in an orderly fashion, a new analytical method called genome-wide aetiology analysis (GWEA) consisting of nine steps is presented. All data required for GWEA of T1D are obtained from peer-reviewed publications or publicly available genome and proteome databases. Critical GWEA steps include AAID distribution among persons with T1D, analysis of at-risk HLA loci for mutation-prone DNA, determination of the role of non-MHC genes on GWAS, and verification of human data by cell culture or animal experiments. GWEA results show that distribution of AAID among persons with T1D adheres to a predictable, tapering, exponential distribution. A single, critical, somatic mutation within the epitope-binding groove of at-risk HLA loci alters HLA-insulin-peptide-T-cell-receptor (TCR) complex binding affinity and creates a new pathway that leads to loss of self-tolerance. The at-risk HLA loci, in particular binding pockets P1, P4 and P9, are encoded by mutation-prone DNA: GC-rich DNA sequence and somatic hypermutation hotspots. All other genes on GWAS can but do not have to amplify the new autoimmune pathway by facilitating DNA mutations, changing peptide binding affinity, reducing signal inhibition or augmenting signal intensity. Animal experiments agree with human studies. In conclusion, T1D is caused by a somatic mutation within the epitope-binding groove of an at-risk HLA gene that affects HLA-insulin-peptide-TCR complex binding affinity and initiates an autoimmune pathway. The nature of the peptide that binds to a mutated epitope-binding groove of an at-risk HLA gene determines the type of autoimmune disease that develops, that is, one at-risk HLA locus, multiple autoimmune diseases. Thus, T1D and AAIDs, and therefore common autoimmune diseases, share a similar somatic mutation-based aetiology.
Collapse
Affiliation(s)
- Piet C de Groen
- Division of Gastroenterology, Hepatology & Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
19
|
Sarangi M, Padhi S, Rath G. Non-Invasive Delivery of Insulin for Breaching Hindrances against Diabetes. Crit Rev Ther Drug Carrier Syst 2024; 41:1-64. [PMID: 38608132 DOI: 10.1615/critrevtherdrugcarriersyst.2023048197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Insulin is recognized as a crucial weapon in managing diabetes. Subcutaneous (s.c.) injections are the traditional approach for insulin administration, which usually have many limitations. Numerous alternative (non-invasive) slants through different routes have been explored by the researchers for making needle-free delivery of insulin for attaining its augmented absorption as well as bioavailability. The current review delineating numerous pros and cons of several novel approaches of non-invasive insulin delivery by overcoming many of their hurdles. Primary information on the topic was gathered by searching scholarly articles from PubMed added with extraction of data from auxiliary manuscripts. Many approaches (discussed in the article) are meant for the delivery of a safe, effective, stable, and patient friendly administration of insulin via buccal, oral, inhalational, transdermal, intranasal, ocular, vaginal and rectal routes. Few of them have proven their clinical efficacy for maintaining the glycemic levels, whereas others are under the investigational pipe line. The developed products are comprising of many advanced micro/nano composite technologies and few of them might be entering into the market in near future, thereby garnishing the hopes of millions of diabetics who are under the network of s.c. insulin injections.
Collapse
Affiliation(s)
| | - Sasmita Padhi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, Pin-201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar-751030, Odisha, India
| |
Collapse
|
20
|
Maity B, Moorthy H, Govindaraju T. Glucose-Responsive Self-Regulated Injectable Silk Fibroin Hydrogel for Controlled Insulin Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:49953-49963. [PMID: 37847862 DOI: 10.1021/acsami.3c07060] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Stimuli-responsive drug delivery systems are gaining importance in personalized medicine to deliver therapeutic doses in response to disease-specific stimulation. Pancreas-mimicking glucose-responsive insulin delivery systems offer improved therapeutic outcomes in the treatment of type 1 and advanced stage of type 2 diabetic conditions. Herein, we present a glucose-responsive smart hydrogel platform based on phenylboronic acid-functionalized natural silk fibroin protein for regulated insulin delivery. The modified protein was synergistically self-assembled and cross-linked through β-sheet and phenylboronate ester formation. The dynamic nature of the bonding confers smooth injectability through the needle. The cross-linked hydrogel structures firmly hold the glucose-sensing element and insulin in its pores and contribute to long-term sensing and drug storage. Under hyperglycemic conditions, the hydrogen peroxide generated from the sensing element induces hydrogel matrix degradation by oxidative cleavage, enabling insulin release. In vivo studies in a type 1 diabetic Wistar rat model revealed that the controlled insulin release from the hydrogel restored diabetic glucose level to physiological conditions for 36 h. This work establishes the functional modification of silk fibroin into a glucose-responsive hydrogel platform for regulated and functional insulin delivery application.
Collapse
Affiliation(s)
- Biswanath Maity
- Bioorganic Chemistry Laboratory, New Chemistry Unit, and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru 560064, Karnataka, India
| |
Collapse
|
21
|
Li M, Xia W, Khoong YM, Huang L, Huang X, Liang H, Zhao Y, Mao J, Yu H, Zan T. Smart and versatile biomaterials for cutaneous wound healing. Biomater Res 2023; 27:87. [PMID: 37717028 PMCID: PMC10504797 DOI: 10.1186/s40824-023-00426-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023] Open
Abstract
The global increase of cutaneous wounds imposes huge health and financial burdens on patients and society. Despite improved wound healing outcomes, conventional wound dressings are far from ideal, owing to the complex healing process. Smart wound dressings, which are sensitive to or interact with changes in wound condition or environment, have been proposed as appealing therapeutic platforms to effectively facilitate wound healing. In this review, the wound healing processes and features of existing biomaterials are firstly introduced, followed by summarizing the mechanisms of smart responsive materials. Afterwards, recent advances and designs in smart and versatile materials of extensive applications for cutaneous wound healing were submarined. Finally, clinical progresses, challenges and future perspectives of the smart wound dressing are discussed. Overall, by mapping the composition and intrinsic structure of smart responsive materials to their individual needs of cutaneous wounds, with particular attention to the responsive mechanisms, this review is promising to advance further progress in designing smart responsive materials for wounds and drive clinical translation.
Collapse
Affiliation(s)
- Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Min Khoong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Lujia Huang
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hsin Liang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yun Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiayi Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
22
|
Lin HL, Mohamed Shukri FN, Yih ES, Sha GH, Jing GS, Jin GW, Hoong CW, Ying CQ, Panda BP, Candasamy M, Bhattamisra SK. Newer therapeutic approaches towards the management of diabetes mellitus: an update. Panminerva Med 2023; 65:362-375. [PMID: 31663302 DOI: 10.23736/s0031-0808.19.03655-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Diabetes mellitus is a chronic metabolic condition characterized by an elevation of blood glucose levels, resulting from defects in insulin secretion, insulin action, or both. The prevalence of the disease has been rapidly rising all over the globe at an alarming rate. Despite advances in the management of diabetes mellitus, it remains a growing epidemic that has become a significant public health burden due to its high healthcare costs and its complications. There is no cure has yet been found for the disease, however, treatment modalities include insulin and antidiabetic agents along with lifestyle modifications are still the mainstay of therapy for diabetes mellitus. The treatment spectrum for the management of diabetes mellitus has rapidly developed in recent years, with new class of therapeutics and expanded indications. This article focused on the emerging therapeutic approaches other than the conventional pharmacological therapies, which include stem cell therapy, gene therapy, siRNA, nanotechnology and theranostics.
Collapse
Affiliation(s)
- Heng L Lin
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | | | - Eric S Yih
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Grace H Sha
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Grace S Jing
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Gan W Jin
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Chow W Hoong
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Choong Q Ying
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Bibhu P Panda
- Department of Pharmaceutical Technology, School of Pharmacy, Taylor's University, Lakeside Campus, Subang Jaya, Selangor, Malaysia
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Subrat K Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia -
| |
Collapse
|
23
|
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
|
24
|
Wong AR, Hung A, Yang AWH, Gill H, Lenon GB. Poria cocos compounds targeting neuropeptide Y1 receptor (Y1R) for weight management: A computational ligand- and structure-based study with molecular dynamics simulations identified beta-amyrin acetate as a putative Y1R inhibitor. PLoS One 2023; 18:e0277873. [PMID: 37390097 PMCID: PMC10313034 DOI: 10.1371/journal.pone.0277873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 10/09/2022] [Indexed: 07/02/2023] Open
Abstract
Poria cocos (PC) is a medicinal herb frequently used in weight-loss clinical trials, however the mechanisms by which its compounds target orexigenic receptors including the neuropeptide Y1 receptor (Y1R) remain largely unknown. This study aimed to screen PC compounds for favourable pharmacokinetics profiles and examine their molecular mechanisms targeting Y1R. Forty-three PC compounds were systematically sought from pharmacological databases and docked with Y1R (PDB: 5ZBQ). By comparing the relative binding affinities, pharmacokinetics and toxicity profiles, we hypothesised that compounds designated PC1 3,4-Dihydroxybenzoic acid, PC8 Vanillic acid, PC40 1-(alpha-L-Ribofuranosyl)uracil, could be potential antagonists as they contact major residues Asn283 and Asp287, similar to various potent Y1R antagonists. In addition, PC21 Poricoic acid B, PC22 Poricoic acid G and PC43 16alpha,25-Dihydroxy-24-methylene-3,4-secolanosta-4(28),7,9(11)-triene-3,21-dioic acid, contacting Asn299, Asp104 and Asp200 proximal to the extracellular surface could also interfere with agonist binding by stabilising the extracellular loop (ECL) 2 of Y1R in a closed position. Owing to their selective interaction with Phe302, an important residue in binding of selective Y1R antagonists, PC12 beta-Amyrin acetate, PC26 3-Epidehydrotumulosic acid and PC27 Cerevisterol were proposed as putative antagonists. Following the consensus approach, PC12 beta-Amyrin acetate, PC26 3-Epidehydrotumulosic acid and PC27 Cerevisterol were identified as candidate compounds due to their high affinities (-12.2, -11.0 and -10.8 kcal, respectively), high drug-likeness and low toxicity profiles. Trajectory analyses and energy contributions of PC12-Y1R complex further confirmed their structural stability and favourable binding free energies, highlighting the feasibility and possible development of PC12 beta-Amyrin acetate as a future Y1R inhibitor.
Collapse
Affiliation(s)
- Ann Rann Wong
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Andrew Hung
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Angela Wei Hong Yang
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Harsharn Gill
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - George Binh Lenon
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
25
|
Li Z, Xu K, Qin L, Zhao D, Yang N, Wang D, Yang Y. Hollow Nanomaterials in Advanced Drug Delivery Systems: From Single- to Multiple Shells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2203890. [PMID: 35998336 DOI: 10.1002/adma.202203890] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/07/2022] [Indexed: 06/15/2023]
Abstract
Hollow-structured nanomaterials (HSNMs) have attracted increased interest in biomedical fields, owing to their excellent potential as drug delivery systems (DDSs) for clinical applications. Among HSNMs, hollow multi-shelled structures (HoMSs) exhibit properties such as high loading capacity, sequential drug release, and multi-functionalized modification and represent a new class of nanoplatforms for clinical applications. The remarkable properties of HoMS-based DDS can simultaneously satisfy and enhance DDSs for delivering small molecular drugs (e.g., antibiotics, chemotherapy drugs, and imaging agents) and macromolecular drugs (e.g., protein/peptide- and nucleic acid-based drugs). First, the latest research advances in delivering small molecular drugs are summarized and highlight the inherent advantages of HoMS-based DDSs for small molecular drug targeting, combining continuous therapeutic drug delivery and theranostics to optimize the clinical benefit. Meanwhile, the macromolecular drugs DDSs are in the initial development stage and currently offer limited delivery modes. There is a growing need to analyze the deficiency of other HSNMs and integrate the advantages of HSNMs, providing solutions for the safe, stable, and cascade delivery of macromolecular drugs to meet vast treatment requirements. Therefore, the latest advances in HoMS-based DDSs are comprehensively reviewed, mainly focusing on the characteristics, research progress by drug category, and future research prospects.
Collapse
Affiliation(s)
- Zhao Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Ke Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Linlin Qin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Decai Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nailiang Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dan Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804, China
| |
Collapse
|
26
|
Ren L, Jiang L, Ren Q, Lv J, Zhu L, Cheng Y. A light-activated polymer with excellent serum tolerance for intracellular protein delivery. Chem Sci 2023; 14:2046-2053. [PMID: 36845943 PMCID: PMC9945510 DOI: 10.1039/d2sc05848k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/20/2023] [Indexed: 01/22/2023] Open
Abstract
The design of efficient materials for intracellular protein delivery has attracted great interest in recent years; however, most current materials for this purpose are limited by poor serum stability due to the early release of cargoes triggered by abundant serum proteins. Here, we propose a light-activated crosslinking (LAC) strategy to prepare efficient polymers with excellent serum tolerance for intracellular protein delivery. A cationic dendrimer engineered with photoactivatable O-nitrobenzene moieties co-assembles with cargo proteins via ionic interactions, followed by light activation to yield aldehyde groups on the dendrimer and the formation of imine bonds with cargo proteins. The light-activated complexes show high stability in buffer and serum solutions, but dis-assemble under low pH conditions. As a result, the polymer successfully delivers cargo proteins green fluorescent protein and β-galactosidase into cells with maintained bioactivity even in the presence of 50% serum. The LAC strategy proposed in this study provides a new insight to improve the serum stability of polymers for intracellular protein delivery.
Collapse
Affiliation(s)
- Lanfang Ren
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Li Jiang
- School of Biomedical Engineering, Shanghai Jiaotong University Shanghai 200240 China
| | - Qianyi Ren
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Linyong Zhu
- School of Biomedical Engineering, Shanghai Jiaotong University Shanghai 200240 China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University Shanghai 200241 China
| |
Collapse
|
27
|
PLGA-Based Micro/Nanoparticles: An Overview of Their Applications in Respiratory Diseases. Int J Mol Sci 2023; 24:ijms24054333. [PMID: 36901762 PMCID: PMC10002081 DOI: 10.3390/ijms24054333] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are critical areas of medical research, as millions of people are affected worldwide. In fact, more than 9 million deaths worldwide were associated with respiratory diseases in 2016, equivalent to 15% of global deaths, and the prevalence is increasing every year as the population ages. Due to inadequate treatment options, the treatments for many respiratory diseases are limited to relieving symptoms rather than curing the disease. Therefore, new therapeutic strategies for respiratory diseases are urgently needed. Poly (lactic-co-glycolic acid) micro/nanoparticles (PLGA M/NPs) have good biocompatibility, biodegradability and unique physical and chemical properties, making them one of the most popular and effective drug delivery polymers. In this review, we summarized the synthesis and modification methods of PLGA M/NPs and their applications in the treatment of respiratory diseases (asthma, COPD, cystic fibrosis (CF), etc.) and also discussed the research progress and current research status of PLGA M/NPs in respiratory diseases. It was concluded that PLGA M/NPs are the promising drug delivery vehicles for the treatment of respiratory diseases due to their advantages of low toxicity, high bioavailability, high drug loading capacity, plasticity and modifiability. And at the end, we presented an outlook on future research directions, aiming to provide some new ideas for future research directions and hopefully to promote their widespread application in clinical treatment.
Collapse
|
28
|
Sharma K, Sharma KK, Sharma A, Jain R. Peptide-based drug discovery: Current status and recent advances. Drug Discov Today 2023; 28:103464. [PMID: 36481586 DOI: 10.1016/j.drudis.2022.103464] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
The progressive development of peptides from reaction vessels to life-saving drugs via rigorous preclinical and clinical assessments is fascinating. Peptide therapeutics have gained momentum with the evolution of techniques in peptide chemistry, such as microwave irradiation in solid- and solution-phase synthesis, ligation chemistry, recombinant synthesis, and amalgamation with synthetic tools, including metal catalysis. Diverse emerging technologies, such as DNA-encoded libraries (DELs) and display techniques, are changing the status quo in the discovery of peptide therapeutics. In this review, we analyzed US Food and Drug Administration (FDA)-approved peptide drugs and those in clinical trials, highlighting recent advances in peptide-based drug discovery.
Collapse
Affiliation(s)
- Komal Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Krishna K Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Anku Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160 062, India.
| |
Collapse
|
29
|
Nguyen HX, Nguyen CN. Microneedle-Mediated Transdermal Delivery of Biopharmaceuticals. Pharmaceutics 2023; 15:277. [PMID: 36678906 PMCID: PMC9864466 DOI: 10.3390/pharmaceutics15010277] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Transdermal delivery provides numerous benefits over conventional routes of administration. However, this strategy is generally limited to a few molecules with specific physicochemical properties (low molecular weight, high potency, and moderate lipophilicity) due to the barrier function of the stratum corneum layer. Researchers have developed several physical enhancement techniques to expand the applications of the transdermal field; among these, microneedle technology has recently emerged as a promising platform to deliver therapeutic agents of any size into and across the skin. Typically, hydrophilic biomolecules cannot penetrate the skin by passive diffusion. Microneedle insertion disrupts skin integrity and compromises its protective function, thus creating pathways (microchannels) for enhanced permeation of macromolecules. Microneedles not only improve stability but also enhance skin delivery of various biomolecules. Academic institutions and industrial companies have invested substantial resources in the development of microneedle systems for biopharmaceutical delivery. This review article summarizes the most recent research to provide a comprehensive discussion about microneedle-mediated delivery of macromolecules, covering various topics from the introduction of the skin, transdermal delivery, microneedles, and biopharmaceuticals (current status, conventional administration, and stability issues), to different microneedle types, clinical trials, safety and acceptability of microneedles, manufacturing and regulatory issues, and the future of microneedle technology.
Collapse
Affiliation(s)
- Hiep X. Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Chien N. Nguyen
- National Institute of Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi 100000, Vietnam
- Faculty of Pharmaceutics and Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi 100000, Vietnam
| |
Collapse
|
30
|
Zhang S, Ge G, Qin Y, Li W, Dong J, Mei J, Ma R, Zhang X, Bai J, Zhu C, Zhang W, Geng D. Recent advances in responsive hydrogels for diabetic wound healing. Mater Today Bio 2022; 18:100508. [PMID: 36504542 PMCID: PMC9729074 DOI: 10.1016/j.mtbio.2022.100508] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Poor wound healing after diabetes mellitus remains a challenging problem, and its pathophysiological mechanisms have not yet been fully elucidated. Persistent bleeding, disturbed regulation of inflammation, blocked cell proliferation, susceptible infection and impaired tissue remodeling are the main features of diabetic wound healing. Conventional wound dressings, including gauze, films and bandages, have a limited function. They generally act as physical barriers and absorbers of exudates, which fail to meet the requirements of the whol diabetic wound healing process. Wounds in diabetic patients typically heal slowly and are susceptible to infection due to hyperglycemia within the wound bed. Once bacterial cells develop into biofilms, diabetic wounds will exhibit robust drug resistance. Recently, the application of stimuli-responsive hydrogels, also known as "smart hydrogels", for diabetic wound healing has attracted particular attention. The basic feature of this system is its capacities to change mechanical properties, swelling ability, hydrophilicity, permeability of biologically active molecules, etc., in response to various stimuli, including temperature, potential of hydrogen (pH), protease and other biological factors. Smart hydrogels can improve therapeutic efficacy and limit total toxicity according to the characteristics of diabetic wounds. In this review, we summarized the mechanism and application of stimuli-responsive hydrogels for diabetic wound healing. It is hoped that this work will provide some inspiration and suggestions for research in this field.
Collapse
Affiliation(s)
- Siming Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Yi Qin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Jiale Dong
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Jiawei Mei
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Ruixiang Ma
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Xianzuo Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China,Corresponding author.
| | - Weiwei Zhang
- Department of Geriatrics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China,Corresponding author.
| |
Collapse
|
31
|
Shen D, Yu H, Wang L, Feng J, Zhang Q, Pan J, Han Y, Ni Z, Liang R, Uddin MA. Glucose-responsive nanoparticles designed via a molecular-docking-driven method for insulin delivery. J Control Release 2022; 352:527-539. [PMID: 36341933 DOI: 10.1016/j.jconrel.2022.10.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Nocturnal blood glucose regulation was one of the key challenges in diabetic treatments. However, development of the smart insulin complexes with mild and glucose-responsive delivering performances was mostly relied on experience of the senior researchers and numerous confirmation experiments. In this work, a series of bioinspired fatty-acid-modified glucose-responsive insulin-delivering polymeric nanoparticles were designed. The molecular docking technique was utilized to efficiently screen the fatty-acid-derived functional groups. The results provided the basis for polymer functionalization and simplified the optimization experiments. For the optimized formulation (C10MS), insulin-loaded C10MS successfully fulfilled the nocturnal-glycemic-controlling requirement of the diabetic rats with lower occurrence of hypoglycemia than the conventional insulin injection schemes. Such formulation also possessed good biocompatibility with the moderate elimination kinetics in vivo, which matched the demand of bio-safety in the daily treatments. Overall, this work opened up a new path for efficient design of functional polymeric materials.
Collapse
Affiliation(s)
- Di Shen
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Jingyi Feng
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Qian Zhang
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Jin Pan
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yin Han
- Zhejiang Institute of Medical Device Testing, Hangzhou 310018, PR China
| | - Zhipeng Ni
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Ruixue Liang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Md Alim Uddin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
32
|
Thirunavukkarasu A, Nithya R, Jeyanthi J. Transdermal drug delivery systems for the effective management of type 2 diabetes mellitus: A review. Diabetes Res Clin Pract 2022; 194:109996. [PMID: 35850300 DOI: 10.1016/j.diabres.2022.109996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022]
Abstract
Type 2 Diabetes mellitus (T2DM) is characterized by either insufficient insulin production or the inability to take it up for the glycemic regulation in the human body. According to WHO reports, T2DM will be the seventh-largest syndrome resulting in mortality by 2030. To tackle this chronic metabolic disorder, the person with diabetes population depends on subcutaneous administration (Sub-Q) of insulin and certain oral hypoglycemic drugs. However, these current invasive practices suffered from painful injections, needle phobia, multiple doses, risk of infection and poor-patient compliance. Hence, the search for a non-invasive and patient-friendly insulin administration system was high in the past decades leading to the development of Transdermal Drug Delivery Systems (TDDS). These can offer rapid and sustained release of therapeutic compounds at controlled rates with no pain during the administration. In recent years, the usage of such TDDS has been increasing at an exponential rate in Type 2 diabetes management. In the present review, the scholarly works on the different modes of TDDS were comprehensively reported chronlogically to appreciate their developments. Conclusively, this review critically identified prevailing research gaps in the current TDDS research and presented potential research hotspots for the prospect development in T2DM management.
Collapse
Affiliation(s)
| | - Rajarathinam Nithya
- Department of Industrial Biotechnology, Government College of Technology, Coimbatore 641013, India.
| | | |
Collapse
|
33
|
Kang H, Zuo Z, Lin R, Yao M, Han Y, Han J. The most promising microneedle device: present and future of hyaluronic acid microneedle patch. Drug Deliv 2022; 29:3087-3110. [PMID: 36151726 PMCID: PMC9518289 DOI: 10.1080/10717544.2022.2125600] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Microneedle patch (MNP) is an alternative to the oral route and subcutaneous injection with unique advantages such as painless administration, good compliance, and fewer side effects. Herein, we report MNP as a prominent strategy for drug delivery to treat local or systemic disease. Hyaluronic acid (HA) has advantageous properties, such as human autologous source, strong water absorption, biocompatibility, and viscoelasticity. Therefore, the Hyaluronic acid microneedle patch (HA MNP) occupies a large part of the MNP market. HA MNP is beneficial for wound healing, targeted therapy of certain specific diseases, extraction of interstitial skin fluid (ISF), and preservation of drugs. In this review, we summarize the benefits of HA and cross-linked HA (x-HA) as an MNP matrix. Then, we introduce the types of HA MNP, delivered substances, and drug distribution. Finally, we focus on the biomedical application of HA MNP as an excellent drug carrier in some specific diseases and the extraction and analysis of biomarkers. We also discuss the future development prospect of HA MNP in transdermal drug delivery systems (TDDS).
Collapse
Affiliation(s)
- Huizhi Kang
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhuo Zuo
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Ru Lin
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Muzi Yao
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Yang Han
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Jing Han
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
34
|
Chen Q, Xiao Z, Wang C, Chen G, Zhang Y, Zhang X, Han X, Wang J, Ye X, Prausnitz MR, Li S, Gu Z. Microneedle Patches Loaded with Nanovesicles for Glucose Transporter-Mediated Insulin Delivery. ACS NANO 2022; 16:18223-18231. [PMID: 36322923 PMCID: PMC10738036 DOI: 10.1021/acsnano.2c05687] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glucose-responsive insulin delivery systems that mimic insulin secretion activity in the pancreas show great potential to improve clinical therapeutic outcomes for people with type 1 and advanced type 2 diabetes. Here, we report a glucose-responsive insulin delivery microneedle (MN) array patch that is loaded with red blood cell (RBC) vesicles or liposome nanoparticles containing glucose transporters (GLUTs) bound with glucosamine-modified insulin (Glu-Insulin). In hyperglycemic conditions, high concentrations of glucose in interstitial fluid can replace Glu-Insulin via a competitive interaction with GLUT, leading to a quick release of Glu-Insulin and subsequent regulation of blood glucose (BG) levels in vivo. To prolong the effective glucose-responsive insulin release from MNs, additional free Glu-Insulin, which serves as "stored insulin", is loaded after RBC vesicles or liposome nanoparticles bound with Glu-Insulin. In the streptozotocin (STZ)-induced type 1 diabetic mouse model, this smart GLUT-based insulin patch can effectively control BG levels without causing hypoglycemia.
Collapse
Affiliation(s)
- Qian Chen
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123
| | - Zhisheng Xiao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Guojun Chen
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Yuqi Zhang
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xudong Zhang
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Xiao Han
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinqiang Wang
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao Ye
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
- Key Laboratory for Diagnosis and Treatment of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
- 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
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, 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
|
35
|
Valdez-Salas B, Castillo-Uribe S, Beltran-Partida E, Curiel-Alvarez M, Perez-Landeros O, Guerra-Balcazar M, Cheng N, Gonzalez-Mendoza D, Flores-Peñaloza O. Recovering Osteoblast Functionality on TiO2 Nanotube Surfaces Under Diabetic Conditions. Int J Nanomedicine 2022; 17:5469-5488. [PMID: 36426372 PMCID: PMC9680990 DOI: 10.2147/ijn.s387386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Titanium (Ti) and its alloys (eg, Ti6Al4V) are exceptional treatments for replacing or repairing bones and damaged surrounding tissues. Although Ti-based implants exhibit excellent osteoconductive performance under healthy conditions, the effectiveness and successful clinical achievements are negatively altered in diabetic patients. Concernedly, diabetes mellitus (DM) contributes to osteoblastic dysfunctionality, altering efficient osseointegration. This work investigates the beneficial osteogenic activity conducted by nanostructured TiO2 under detrimental microenvironment conditions, simulated by human diabetic serum. Methods We evaluated the bone-forming functional properties of osteoblasts on synthesized TiO2 nanotubes (NTs) by anodization and Ti6Al4V non-modified alloy surfaces under detrimental diabetic conditions. To simulate the detrimental environment, MC3T3E-1 preosteoblasts were cultured under human diabetic serum (DS) of two diagnosed and metabolically controlled patients. Normal human serum (HS) was used to mimic health conditions and fetal bovine serum (FBS) as the control culture environment. We characterized the matrix mineralization under the detrimental conditions on the control alloy and the NTs. Moreover, we applied immunofluorescence of osteoblasts differentiation markers on the NTs to understand the bone-expression stimulated by the biochemical medium conditions. Results The diabetic conditions depressed the initial osteoblast growth ability, as evidenced by altered early cell adhesion and reduced proliferation. Nonetheless, after three days, the diabetic damage was suppressed by the NTs, enhancing the osteoblast activity. Therefore, the osteogenic markers of bone formation and the differentiation of osteoblasts were reactivated by the nanoconfigured surfaces. Far more importantly, collagen secretion and bone-matrix mineralization were stimulated and conducted to levels similar to those of the control of FBS conditions, in comparison to the control alloy, which was not able to reach similar levels of bone functionality than the NTs. Conclusion Our study brings knowledge for the potential application of nanostructured biomaterials to work as an integrative platform under the detrimental metabolic status present in diabetic conditions.
Collapse
Affiliation(s)
- Benjamin Valdez-Salas
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| | - Sandra Castillo-Uribe
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| | - Ernesto Beltran-Partida
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
- Correspondence: Ernesto Beltran-Partida, Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Blvd. Benito Juárez y Calle de la Normal, Mexicali, Baja California, C.P. 21280, México, Email
| | - Mario Curiel-Alvarez
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| | - Oscar Perez-Landeros
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| | - Minerva Guerra-Balcazar
- Facultad de Ingeniería, División de Investigación y Posgrado, Universidad Autónoma de Querétaro, Querétaro, México
| | | | - Daniel Gonzalez-Mendoza
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| | - Olivia Flores-Peñaloza
- Laboratorio de Biología Molecular y Cáncer, Instituto de Ingeniería, Universidad Autónoma de Baja California, Mexicali, Baja California, México
| |
Collapse
|
36
|
Safdar R, Thanabalan M. Developments in insulin delivery and potential of chitosan for controlled release application: A review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
37
|
Gao Q, Bai Q, Zheng C, Sun N, Liu J, Chen W, Hu F, Lu T. Application of Metal–Organic Framework in Diagnosis and Treatment of Diabetes. Biomolecules 2022; 12:biom12091240. [PMID: 36139080 PMCID: PMC9496218 DOI: 10.3390/biom12091240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetes-related chronic wounds are often accompanied by a poor wound-healing environment such as high glucose, recurrent infections, and inflammation, and standard wound treatments are fairly limited in their ability to heal these wounds. Metal–organic frameworks (MOFs) have been developed to improve therapeutic outcomes due to their ease of engineering, surface functionalization, and therapeutic properties. In this review, we summarize the different synthesis methods of MOFs and conduct a comprehensive review of the latest research progress of MOFs in the treatment of diabetes and its wounds. State-of-the-art in vivo oral hypoglycemic strategies and the in vitro diagnosis of diabetes are enumerated and different antimicrobial strategies (including physical contact, oxidative stress, photothermal, and related ions or ligands) and provascular strategies for the treatment of diabetic wounds are compared. It focuses on the connections and differences between different applications of MOFs as well as possible directions for improvement. Finally, the potential toxicity of MOFs is also an issue that we cannot ignore.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tingli Lu
- Correspondence: ; Tel.: +86-136-5918-8506
| |
Collapse
|
38
|
Zhu J, Liu W, Zhang B, Zhou D, Fan X, Wang X, Liu X. Carbon Dots Embedded Hybrid Microgel with Phenylboronic Acid as Monomer for Fluorescent Glucose Sensing and Glucose-Triggered Insulin Release at Physiological pH. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3065. [PMID: 36080102 PMCID: PMC9457936 DOI: 10.3390/nano12173065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/14/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
A multifunctional and biocompatible hybrid microgel (poly(VPBA-AAm)-CD) using N, S-doped carbon dots (CDs) and ethylene glycol dimethacrylate (EGDMA) as cross-linking agents, and 4-vinylbenzene boronic acid (VPBA) and acrylamide (AAm) as monomers, was designed in this work. This microgel can be easily prepared by a simple one-pot radical dispersion polymerization of the reactants using a rationally designed hydrogen-bonded complex method. The hybrid microgels were spherical particles with a smooth surface and an average particle size of 234 ± 8 nm. The poly(VPBA-AAm)-CD microgel displayed the glucose-responsive swelling within a clinically concerned range at a physiological pH and could realize the controllable release of insulin. In addition, the release rate of insulin in the hybrid microgel (poly(VPBA-AAm)-CD) could be triggered by glucose concentrations in the solution, and the increasing glucose concentrations can accelerate the insulin release. Further in vitro cytotoxicity studies showed that the microgel had good biocompatibility and no obvious toxicity to the cells. These indicate that the prepared microgel (poly(VPBA-AAm)-CD) may supply a new pattern for the self-regulating therapy of insulin deficiency in diabetes.
Collapse
Affiliation(s)
- Jinhua Zhu
- Correspondence: (J.Z.); (X.L.); Tel.: +86-371-23881589 (J.Z.)
| | | | | | | | | | | | - Xiuhua Liu
- Correspondence: (J.Z.); (X.L.); Tel.: +86-371-23881589 (J.Z.)
| |
Collapse
|
39
|
Gorai B, Vashisth H. Progress in Simulation Studies of Insulin Structure and Function. Front Endocrinol (Lausanne) 2022; 13:908724. [PMID: 35795141 PMCID: PMC9252437 DOI: 10.3389/fendo.2022.908724] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/28/2022] [Indexed: 01/02/2023] Open
Abstract
Insulin is a peptide hormone known for chiefly regulating glucose level in blood among several other metabolic processes. Insulin remains the most effective drug for treating diabetes mellitus. Insulin is synthesized in the pancreatic β-cells where it exists in a compact hexameric architecture although its biologically active form is monomeric. Insulin exhibits a sequence of conformational variations during the transition from the hexamer state to its biologically-active monomer state. The structural transitions and the mechanism of action of insulin have been investigated using several experimental and computational methods. This review primarily highlights the contributions of molecular dynamics (MD) simulations in elucidating the atomic-level details of conformational dynamics in insulin, where the structure of the hormone has been probed as a monomer, dimer, and hexamer. The effect of solvent, pH, temperature, and pressure have been probed at the microscopic scale. Given the focus of this review on the structure of the hormone, simulation studies involving interactions between the hormone and its receptor are only briefly highlighted, and studies on other related peptides (e.g., insulin-like growth factors) are not discussed. However, the review highlights conformational dynamics underlying the activities of reported insulin analogs and mimetics. The future prospects for computational methods in developing promising synthetic insulin analogs are also briefly highlighted.
Collapse
Affiliation(s)
| | - Harish Vashisth
- Department of Chemical Engineering, University of New Hampshire, Durham, NH, United States
| |
Collapse
|
40
|
Wang B, Li X, Wang Y, Mao X, Wang X. Post-translational site-specific protein azidolation with an azido pyridoxal derivative. Chem Commun (Camb) 2022; 58:7408-7411. [PMID: 35695118 DOI: 10.1039/d2cc03051a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An azido pyridoxal derivative (PLAA) was developed as a protein azidolation reagent, with selectivity towards N-terminal glycine. Successful PLAA modification was achieved with small peptides, natural proteins and lab-expressed proteins under mild conditions, offering a unique method for post-synthesis site-specific azidolation of a peptide or protein.
Collapse
Affiliation(s)
- Baochuan Wang
- Institute of Advanced Synthesis, Institute of Chemical Biology and Functional Molecules, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Xun Li
- Institute of Advanced Synthesis, Institute of Chemical Biology and Functional Molecules, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Yiwan Wang
- Institute of Advanced Synthesis, Institute of Chemical Biology and Functional Molecules, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Xianxian Mao
- Institute of Advanced Synthesis, Institute of Chemical Biology and Functional Molecules, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China. .,School of Pharmacy, Jiangsu Health Vocational College, Nanjing 211800, China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, Institute of Chemical Biology and Functional Molecules, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
41
|
Wu T, You X, Chen Z. Hollow Microneedles on a Paper Fabricated by Standard Photolithography for the Screening Test of Prediabetes. SENSORS 2022; 22:s22114253. [PMID: 35684875 PMCID: PMC9185271 DOI: 10.3390/s22114253] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023]
Abstract
Microneedle (MN) is a novel technique of the biomedical engineering field because of its ability to evaluate bioinformation via minimal invasion. One of the urgent requirements for ground-breaking health care monitoring is persistent monitoring. Hollow microneedles are extremely attractive to extract skin interstitial fluid (ISF) for analysis, which makes them perfect for sensing biomarkers and facilitating diagnosis. Nevertheless, its intricate fabrication process has hampered its extensive application. The present research demonstrates an easy one-step preparation approach for hollow MNs on the foundation of the refraction index variations of polyethylene glycol diacrylate (PEGDA) in the process of photopolymerization. The fabricated hollow microneedle exhibited ideal mechanical characteristics to penetrate the skin. Hydrodynamic simulations showed that the liquid was risen in a hollow microneedle by capillary force. Furthermore, a paper-based glucose sensor was integrated with the hollow microneedle. We also observed that the MN array smoothly extracted ISF in vitro and in vivo by capillary action. The outcomes displayed the applicability of the MN patch to persistent blood glucose (GLU) monitoring, diagnosis-related tests for patients and pre-diabetic individuals.
Collapse
Affiliation(s)
- Tianwei Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China;
| | - Xueqiu You
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China;
- School of Information Engineering, Jimei University, Xiamen 361021, China
- Correspondence: (X.Y.); (Z.C.)
| | - Zhong Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, State Key Laboratory of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, China;
- Correspondence: (X.Y.); (Z.C.)
| |
Collapse
|
42
|
Fang J, Huang S, Liu F, He G, Li X, Huang X, Chen HJ, Xie X. Semi-Implantable Bioelectronics. NANO-MICRO LETTERS 2022; 14:125. [PMID: 35633391 PMCID: PMC9148344 DOI: 10.1007/s40820-022-00818-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 06/15/2023]
Abstract
Developing techniques to effectively and real-time monitor and regulate the interior environment of biological objects is significantly important for many biomedical engineering and scientific applications, including drug delivery, electrophysiological recording and regulation of intracellular activities. Semi-implantable bioelectronics is currently a hot spot in biomedical engineering research area, because it not only meets the increasing technical demands for precise detection or regulation of biological activities, but also provides a desirable platform for externally incorporating complex functionalities and electronic integration. Although there is less definition and summary to distinguish it from the well-reviewed non-invasive bioelectronics and fully implantable bioelectronics, semi-implantable bioelectronics have emerged as highly unique technology to boost the development of biochips and smart wearable device. Here, we reviewed the recent progress in this field and raised the concept of "Semi-implantable bioelectronics", summarizing the principle and strategies of semi-implantable device for cell applications and in vivo applications, discussing the typical methodologies to access to intracellular environment or in vivo environment, biosafety aspects and typical applications. This review is meaningful for understanding in-depth the design principles, materials fabrication techniques, device integration processes, cell/tissue penetration methodologies, biosafety aspects, and applications strategies that are essential to the development of future minimally invasive bioelectronics.
Collapse
Affiliation(s)
- Jiaru Fang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Shuang Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Fanmao Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Gen He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China
| | - Xi Xie
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
43
|
De Marchi JGB, Cé R, Onzi G, Alves ACS, Santarém N, Cordeiro da Silva A, Pohlmann AR, Guterres SS, Ribeiro AJ. IgG functionalized polymeric nanoparticles for oral insulin administration. Int J Pharm 2022; 622:121829. [PMID: 35580686 DOI: 10.1016/j.ijpharm.2022.121829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
The oral route is the best way to administer a drug; however, fitting peptide drugs in this route is a major challenge. In insulin cases, less than 0.5% of the administered dose achieves systemic circulation. Oral delivery by nanoparticles can increase insulin permeability across the intestinal epithelium while maintaining its structure and activity until release in the gut. This system can be improved to increase permeability across intestinal cells through active delivery. This study aimed to improve a nanoparticle formulation by promoting functionalization of its surface with immunoglobulin G to increase its absorption by intestinal epithelium. The characterization of formulations showed an adequate size and a good entrapment efficiency. Functionalized nanoparticles led to a desirable increase in insulin release time. Differential scanning calorimetry, infrared spectroscopy and paper chromatography proved the interactions of nanoparticle components. With immunoglobulin G, the nanoparticle size was slightly increased, which did not show aggregate formation. The developed functionalized nanoparticle formulation proved to be adequate to carry insulin and potentially increase its internalization by epithelial gut cells, being a promising alternative to the existing formulations for orally administered low-absorption peptides.
Collapse
Affiliation(s)
- J G B De Marchi
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil; Universidade de Coimbra, Faculdade de Farmácia, Coimbra, Portugal
| | - R Cé
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil; Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90650-001, Brazil
| | - G Onzi
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil
| | - A C S Alves
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil; Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90650-001, Brazil
| | - N Santarém
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - A Cordeiro da Silva
- Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal; i(3)S, IBMC, Rua Alfredo Allen, Porto, Portugal
| | - A R Pohlmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil; Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90650-001, Brazil
| | - S S Guterres
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS 90610-000, Brazil
| | - A J Ribeiro
- Universidade de Coimbra, Faculdade de Farmácia, Coimbra, Portugal; i(3)S, IBMC, Rua Alfredo Allen, Porto, Portugal.
| |
Collapse
|
44
|
Zhang P, Du C, Huang T, Hu S, Bai Y, Li C, Feng G, Gao Y, Li Z, Wang B, Hirvonen JT, Fan J, Santos HA, Liu D. Surface Adsorption-Mediated Ultrahigh Efficient Peptide Encapsulation with a Precise Ratiometric Control for Type 1 and 2 Diabetic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200449. [PMID: 35229498 DOI: 10.1002/smll.202200449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Indexed: 06/14/2023]
Abstract
A surface adsorption strategy is developed to enable the engineering of microcomposites featured with ultrahigh loading capacity and precise ratiometric control of co-encapsulated peptides. In this strategy, peptide molecules (insulin, exenatide, and bivalirudin) are formulated into nanoparticles and their surface is decorated with carrier polymers. This polymer layer blocks the phase transfer of peptide nanoparticles from oil to water and, consequently, realizes ultrahigh peptide loading degree (up to 78.9%). After surface decoration, all three nanoparticles are expected to exhibit the properties of adsorbed polymer materials, which enables the co-encapsulation of insulin, exenatide, and bivalirudin with a precise ratiometric control. After solidification of this adsorbed polymer layer, the release of peptides is synchronously prolonged. With the help of encapsulation, insulin achieves 8 days of glycemic control in type 1 diabetic rats with one single injection. The co-delivery of insulin and exenatide (1:1) efficiently controls the glycemic level in type 2 diabetic rats for 8 days. Weekly administration of insulin and exenatide co-encapsulated microcomposite effectively reduces the weight gain and glycosylated hemoglobin level in type 2 diabetic rats. The surface adsorption strategy sets a new paradigm to improve the pharmacokinetic and pharmacological performance of peptides, especially for the combination of peptides.
Collapse
Affiliation(s)
- Pei Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Chunyang Du
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Tianhe Huang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuai Hu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuancheng Bai
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Cong Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Guobing Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Yue Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhi Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Baoxun Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| | - Jouni T Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Jin Fan
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen/University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Dongfei Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
45
|
Hu Y, Gao S, Lu H, Ying JY. Acid-Resistant and Physiological pH-Responsive DNA Hydrogel Composed of A-Motif and i-Motif toward Oral Insulin Delivery. J Am Chem Soc 2022; 144:5461-5470. [PMID: 35312303 DOI: 10.1021/jacs.1c13426] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An acid-resistant DNA hydrogel that is stable in an extremely acidic environment with pH as low as 1.2 has not been reported before, largely due to the instability of DNA-hybridized structures. To achieve this, adenine (A)-rich and cytosine (C)-rich oligonucleotides are rationally designed and integrated to form copolymers with acrylamide monomers via free-radical polymerization. In an acidic environment (pH 1.2-6.0), the generated copolymers form a hydrogel state, which is cross-linked by parallel A-motif duplex configurations (pH 1.2-3.0) and quadruplex i-motif structures (pH 4.0-6.0) due to the protonation of A and C bases, respectively. Specifically, the protonated A-rich sequences under pH 1.2-3.0 form a stable parallel A-motif duplex cross-linking unit through reverse Hoogsteen interaction and electrostatic attraction. Hemi-protonated C bases under mildly acidic pH (4.0-6.0) form quadruplex i-motif cross-linking configuration via Hoogsteen interaction. Under physiological pH, both A and C bases deprotonated, resulting in the separation of A-motif and i-motif to A-rich and C-rich single strands, respectively, and thereby the dissociation of the DNA hydrogel into the solution state. The acid-resistant and physiological pH-responsive DNA hydrogel was further developed for oral drug delivery to the hostile acidic environment in the stomach (pH 1.2), duodenum (pH 5.0), and small intestine (pH 7.2), where the drug would be released and absorbed. As a proof of concept, insulin was encapsulated in the DNA hydrogel and orally administered to diabetic rats. In vitro and in vivo studies demonstrated the potential usage of the DNA hydrogel for oral drug delivery.
Collapse
Affiliation(s)
- Yuwei Hu
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Shujun Gao
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Hongfang Lu
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| | - Jackie Y Ying
- NanoBio Lab, Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore.,NanoBio Lab, A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, 31 Biopolis Way, The Nanos, #09-01, Singapore 138669, Singapore
| |
Collapse
|
46
|
Influence of Amidation on the Release Profiles of Insulin Drug from Chitosan-Based Matrices. COATINGS 2022. [DOI: 10.3390/coatings12040465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The present study deals with the comparative analysis of insulin drug release from pure chitosan (CS) and its crosslinked amide derivatives. The objective of this study was to investigate the influence of fatty acid derivatives on the release profiles of insulin drug from CS-based matrices. In order to form cross-linked CS-based amide derivatives, the CS biopolymer was reacted with four different fatty acids with varying amount of unsaturation, including stearic acid (SA), oleic acid (OA), linoleic acid (LA), and linolenic acid (LLA), and then subjected to cross-linking. Following this, the pure CS and cross-linked CS amide derivatives were loaded with insulin drug and were characterized thoroughly by making use of various instrumental techniques such as FTIR, UV–Vis, TGA, HRTEM, DLS, PDI, and zeta potential studies. In addition, the insulin release profiles were studied and compared between pure CS and CS amides at two different pHs, 7.4 and 1.2. Finally, the insulin drug release data was investigated with five different pharmacokinetic models (zero, first, Higuchi, Kersmeyer–Peppas, and Hixson models). From the analysis, the cross-linked CS amides was found to be superior to pure CS, and within the amide derivatives, the one with a high amount of unsaturation, LLA-derived CS biopolymer, was shown to be better for the release of insulin drug by means of the diffusion and dissolution pathways.
Collapse
|
47
|
Gowda BHJ, Ahmed MG, Sahebkar A, Riadi Y, Shukla R, Kesharwani P. Stimuli-Responsive Microneedles as a Transdermal Drug Delivery System: A Demand-Supply Strategy. Biomacromolecules 2022; 23:1519-1544. [PMID: 35274937 DOI: 10.1021/acs.biomac.1c01691] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Microneedles are one of the most prominent approaches capable of physically disrupting the stratum corneum without devastating the deeper tissues to deliver both small molecules and macromolecules into the viable epidermis/dermis for local/systemic effects. Over the past two decades, microneedles have caught the attention of many researchers because of their outstanding advantages over oral and parenteral drug delivery systems such as self-administration, pain-free, steady-plasma concentration maintenance, avoidance of first-pass hepatic biotransformation, and so on. So far, scientists have reported various types of microneedle patches to deliver the loaded therapeutics as soon as the microneedles are inserted into the skin, regardless of the demand for therapeutics to treat a specific condition. This way of drug delivery can lead to potential risks such as poor therapeutic efficacy or drug overdose. The stimuli-responsive microneedles are the most predominant tool to achieve the on-demand/need-based drug delivery, leading to safe and effective treatment. Various natural and synthetic polymers that can undergo significant transitions such as swelling, shrinking, dissolution, or disintegration play a pivotal role in the development of stimuli-responsive microneedles. The current Review provides brief information about the history, emergence, type, and working principles of microneedles. Furthermore, it selectively discusses various exogenous and endogenous stimuli-responsive microneedles along with their mechanism of action involved in treating different disease conditions. Collaterally, the emergence of "closed-loop" combinatorial stimuli-responsive microneedle patches for precise delivery of therapeutics is meticulously canvassed. Subsequently, it covers the patents of different stimuli-responsive microneedles and further highlights the existing challenges and future perspectives concerning clinical application and large-scale production.
Collapse
Affiliation(s)
- B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 1696700, Iran.,School of Medicine, The University of Western Australia, Perth 6009, Australia
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, Uttar Pradesh 226002, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
48
|
Xu X, Ran Y, Huang C, Yin Z. Glucose and H 2O 2 Dual-Responsive Nanocomplex Grafted with Insulin Prodrug for Blood Glucose Regulation. Biomacromolecules 2022; 23:1765-1776. [PMID: 35275618 DOI: 10.1021/acs.biomac.2c00016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although "closed-loop" smart insulin delivery systems have been extensively investigated, the majority of them suffer from low insulin loading efficiency and slow glucose response. Here, we constructed a novel nanocomplex (NC), which was prepared by electrostatic interaction between negatively charged insulin prodrug nanoparticles (NPs) and positively charged polycaprolactone-polyethylenimine (PCL-PEI) micelles. The insulin prodrug was linked to acetalated dextran (AD) via borate ester bonds to form IAD NPs, and glucose oxidase (GOx) was encapsulated in PCL-PEI micelles. The NC was negatively charged with a high insulin grafting rate (0.473 mg/mg), and in vitro experiments revealed that IAD was sensitive to hyperglycemia and H2O2, whereas GOx significantly improved the response to glucose by altering the microenvironment to promote sustained insulin release. Furthermore, compared with free insulin and IAD NPs, subcutaneously injected NCs in diabetic rats had long-term hypoglycemic effects, showing excellent biocompatibility in vitro and in vivo, which had good potential in insulin self-regulation delivery.
Collapse
Affiliation(s)
- Xiaowen Xu
- 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
| | - Yu Ran
- 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
| | - Chengyuan 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
| | - Zongning Yin
- 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
|
49
|
Zou JJ, Wei G, Xiong C, Yu Y, Li S, Hu L, Ma S, Tian J. Efficient oral insulin delivery enabled by transferrin-coated acid-resistant metal-organic framework nanoparticles. SCIENCE ADVANCES 2022; 8:eabm4677. [PMID: 35196087 PMCID: PMC8865763 DOI: 10.1126/sciadv.abm4677] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Oral protein delivery is considered a cutting-edge technology to improve patients' quality of life, offering superior patient compliance and convenience compared with injections. However, oral protein formulation has stagnated because of the instability and inefficient penetration of protein in the gastrointestinal tract. Here, we used acid-resistant metal-organic framework nanoparticles (UiO-68-NH2) to encapsulate sufficient insulin and decorated the exterior with targeting proteins (transferrin) to realize highly efficient oral insulin delivery. The UiO-68-NH2 nanocarrier with proper pore size achieved high insulin loading while protecting insulin from acid and enzymatic degradation. Through receptor-mediated transcellular pathway, the transferrin-coated nanoparticles realized efficient transport across the intestinal epithelium and controlled insulin release under physiological conditions, leading to a notable hypoglycemic effect and a high oral bioavailability of 29.6%. Our work demonstrates that functional metal-organic framework nanoparticles can protect proteins from the gastric environment and overcome the intestinal barrier, thus providing the possibility for oral biomacromolecule delivery.
Collapse
Affiliation(s)
- Jun-Jie Zou
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Gaohui Wei
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Chuxiao Xiong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Yunhao Yu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Sihui Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Liefeng Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
| | - Shengqian Ma
- Department of Chemistry, University of North Texas, Denton, TX 76201, USA
- Corresponding author. (S.M.); (J.T.)
| | - Jian Tian
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071 P. R. China
- Corresponding author. (S.M.); (J.T.)
| |
Collapse
|
50
|
Manasa G, Mascarenhas RJ, Shetti NP, Malode SJ, Mishra A, Basu S, Aminabhavi TM. Skin Patchable Sensor Surveillance for Continuous Glucose Monitoring. ACS APPLIED BIO MATERIALS 2022; 5:945-970. [PMID: 35170319 DOI: 10.1021/acsabm.1c01289] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus is a physiological and metabolic disorder affecting millions of people worldwide, associated with global morbidity, mortality, and financial expenses. Long-term complications can be avoided by frequent, continuous self-monitoring of blood glucose. Therefore, this review summarizes the current state-of-art glycemic control regimes involving measurement approaches and basic concepts. Following an introduction to the significance of continuous glucose sensing, we have tracked the evolution of glucose monitoring devices from minimally invasive to non-invasive methods to present an overview of the spectrum of continuous glucose monitoring (CGM) technologies. The conveniences, accuracy, and cost-effectiveness of the real-time CGM systems (rt-CGMs) are the factors considered for discussion. Transdermal biosensing and drug delivery routes have recently emerged as an innovative approach to substitute hypodermal needles. This work reviews skin-patchable glucose monitoring sensors for the first time, providing specifics of all the major findings in the past 6 years. Skin patch sensors and their progressive form, i.e., microneedle (MN) array sensory and delivery systems, are elaborated, covering self-powered, enzymatic, and non-enzymatic devices. The critical aspects reviewed are material design and assembly techniques focusing on flexibility, sensitivity, selectivity, biocompatibility, and user-end comfort. The review highlights the advantages of patchable MNs' multi-sensor technology designed to maintain precise blood glucose levels and administer diabetes drugs or insulin through a "sense and act" feedback loop. Subsequently, the limitations and potential challenges encountered from the MN array as rt-CGMs are listed. Furthermore, the current statuses of working prototype glucose-responsive "closed-loop" insulin delivery systems are discussed. Finally, the expected future developments and outlooks in clinical applications are discussed.
Collapse
Affiliation(s)
- G Manasa
- Electrochemistry Research Group, Department of Chemistry, St. Joseph's College (Autonomous), Lalbagh Road, Bangalore, Karnataka 560027, India
| | - Ronald J Mascarenhas
- Electrochemistry Research Group, Department of Chemistry, St. Joseph's College (Autonomous), Lalbagh Road, Bangalore, Karnataka 560027, India
| | - Nagaraj P Shetti
- Department of Chemistry, School of Advanced Sciences, KLE Technological University, Vidyanagar, Hubballi, Karnataka 580031, India
| | - Shweta J Malode
- Department of Chemistry, School of Advanced Sciences, KLE Technological University, Vidyanagar, Hubballi, Karnataka 580031, India
| | - Amit Mishra
- Department of Chemical Engineering, Inha University, Incheon 22212, South Korea
| | - Soumen Basu
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| | - Tejraj M Aminabhavi
- Department of Chemistry, School of Advanced Sciences, KLE Technological University, Vidyanagar, Hubballi, Karnataka 580031, India
| |
Collapse
|