1
|
Cureno Hernandez KE, Lee J, Kim S, Cartwright Z, Herrera-Alonso M. Boronic acid-mediated mucin/surface interactions of zwitterionic polymer brushes. SOFT MATTER 2025; 21:3125-3136. [PMID: 40171575 DOI: 10.1039/d4sm01502a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Mucus is a substance that acts as a protective barrier, shielding tissues from infections caused by viruses and bacteria. Recent studies highlight the advantages of transmucosal drug delivery compared to traditional delivery methods. However, external particles in mucus struggle to penetrate its deeper layers and are often eliminated by mucus clearance mechanisms, hindering effective drug delivery. To gain a deeper understanding of how material surfaces interact with mucus, we grafted brushes of poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) onto silica surfaces, followed by the straightforward installation of a terminal boronic acid moiety (3-phenylboronic acid, APBA). The modification process was carried out following a surface-initiated activator regenerated by electron transfer atom transfer radical polymerization (SI-ARGET ATRP), a method known for its effectiveness in producing well-defined grafted polymers. After conjugation of APBA, we studied the effects of surface chemistry on properties such as pH-sensitivity and mucin adsorption. The surfaces modified with the zwitterionic polymer showed no mucin interaction regardless of system pH. However, all the surfaces containing the boronic acid showed boronic acid-sialic acid interactions, particularly at lower pH values. The insights gained from this study will enhance our understanding of the interactions between the zwitterionic PMPC and the boronic acid APBA with mucins, laying the groundwork for future chemical modifications of particle surfaces aimed at modulating their transport through mucus.
Collapse
Affiliation(s)
- Karla E Cureno Hernandez
- School of Materials Science and Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA.
| | - Jeonghun Lee
- School of Materials Science and Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA.
| | - Sunghoon Kim
- School of Materials Science and Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA.
| | - Zach Cartwright
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA
| | - Margarita Herrera-Alonso
- School of Materials Science and Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA.
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523, USA
| |
Collapse
|
2
|
Jansook P, Sigurdsson HH, Loftsson T. A look to the future: cyclodextrins and cyclodextrin-based drug delivery to the retina. Expert Opin Drug Deliv 2025:1-18. [PMID: 40105773 DOI: 10.1080/17425247.2025.2482049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Retinal diseases are a leading cause of vision loss, affecting millions of people worldwide. Current treatment options are based on invasive methods such as intravitreal injections. Therefore, there is a need for alternative therapeutic strategies that are both effective and more patient-friendly. AREAS COVERED Topical drug delivery has gained attention as a preferred noninvasive approach, although it is hindered by several ocular barriers. Cyclodextrin (CD)-based nanoparticles have emerged as a promising strategy to overcome these limitations by enhancing drug permeability in the posterior segment of the eye. This review discusses the potential of CDs as enabling pharmaceutical excipients, their role in improving ocular drug bioavailability, and provides examples of CD-based eye drop formulations currently under development or undergoing clinical trials. Also, the role of CDs as active pharmaceutical agents in ophthalmology is discussed. EXPERT OPINION CD-based nanoparticle eye drops present a promising solution and have shown clinical success. CDs are approved pharmaceutical excipients for eye drop formulations and can act as active pharmaceutical ingredients for the treatment of inherent retinal diseases. Future innovations in hybrid CD-based delivery systems and integration of novel therapeutic compounds could provide more efficient and targeted treatment options for retinal diseases.
Collapse
Affiliation(s)
- Phatsawee Jansook
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
- Cyclodextrin Application and Nanotechnology-Based Delivery Systems Research Unit, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Hákon H Sigurdsson
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Thorsteinn Loftsson
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
3
|
Zhang F, Tan M, Hu ZE, Zhang YT, Qi XW, Che YT, Li J, Zhang S, Li BJ. A hyaluronic acid-modified cyclodextrin self-assembly system for the delivery of β-carotene in the treatment of dry eye disease. Int J Biol Macromol 2025; 287:138428. [PMID: 39647723 DOI: 10.1016/j.ijbiomac.2024.138428] [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: 06/17/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Dry eye disease (DED) is a multifactorial ocular disease, the core mechanism of which is the tear film instability caused by ocular oxidative stress damage and inflammation. Although various pharmaceutical agents are available for DED treatment, their effectiveness is often limited by the eyes' unique biological barriers, and the long-term use of steroid hormones can lead to several adverse effects. This study reported a nano-supramolecular delivery system consisting of a polycyclodextrin (PCD), hyaluronic acid (HA) and the natural compound β-carotene (BC) for the DED treatment. Our findings indicate that the HA/PCD@BC eye drops effectively distribute on the ocular surface, retain BC, and significantly enhance the corneal penetration of BC. The excellent biocompatibility of HA/PCD@BC was demonstrated through viability testing on different cell lines, the Draize eye test, as well as the hematoxylin-eosin staining (H&E) sections of cornea and conjunctiva. Both in vitro oxidative stress assays and in vivo DED model evaluations demonstrated that the HA/PCD@BC delivery system significantly reduced abnormal oxidative stress levels on the ocular surface, inhibited the secretion of inflammatory factors, and increased the secretion of tear film stabilizing mucin. These effects collectively improved pathological changes in eye tissues and minimized damage to the ocular surface. It is of particular importance to note that HA/PCD@BC eye drops showed superior efficacy in comparison to cyclosporine A (CsA), an FDA-approved first-line drug. To sum up, the HA/PCD@BC nanodelivery system provides a natural, safe and effective therapeutic strategy for the treatment of DED and various ocular diseases.
Collapse
Affiliation(s)
- Fuzhong Zhang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Tan
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zu-E Hu
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ye-Tao Zhang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu-Wei Qi
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Ting Che
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Li
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Sichuan University, Chengdu 610065, China
| | - Sheng Zhang
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Sichuan University, Chengdu 610065, China.
| | - Bang-Jing Li
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Hazt B, Read DJ, Harlen OG, Poon WCK, O'Connell A, Sarkar A. Mucoadhesion across scales: Towards the design of protein-based adhesives. Adv Colloid Interface Sci 2024; 334:103322. [PMID: 39489118 DOI: 10.1016/j.cis.2024.103322] [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: 03/26/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Mucoadhesion is a special case of bioadhesion in which a material adheres to soft mucosal tissues. This review elucidates our current understanding of mucoadhesion across length, time, and energy scales by focusing on relevant structural features of mucus. We highlight the importance of both covalent and non-covalent interactions that can be tailored to maximize mucoadhesive interactions, particularly concerning proteinaceous mucoadhesives, which have been explored only to a limited extent so far in the literature. In particular, we highlight the importance of thiol groups, hydrophobic moieties, and charged species inherent to proteins as key levers to fine tune mucoadhesive performance. Some aspects of protein surface modification by grafting specific functional groups or coupling with polysaccharides to influence mucoadhesive performance are examined. Insights from this review offer a physicochemical roadmap to inform the development of biocompatible, protein-based mucoadhesive systems that can fulfil dual roles for both adhesion and delivery of actives, enabling the fabrication of advanced biomedical, nutritional and allied soft material technologies.
Collapse
Affiliation(s)
- Bianca Hazt
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK
| | - Daniel J Read
- School of Mathematics, University of Leeds, LS2 9JT, UK
| | | | - Wilson C K Poon
- School of Physics and Astronomy, University of Edinburgh, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - Adam O'Connell
- Polymer Science Platform, Reckitt Benckiser Healthcare (UK) Ltd, Dansom Lane S, Hull, HU8 7DS, UK
| | - Anwesha Sarkar
- Food Colloids and Bioprocessing Group, School of Food Science and Nutrition, University of Leeds, LS2 9JT, UK.
| |
Collapse
|
5
|
Xue R, Wu H, Li S, Pu N, Wei D, Zhao N, Cui Y, Li H, Song Z, Tao Y. Biodegradable microspheres come into sight: A promising biomaterial for delivering drug to the posterior segment of the eyeball. Mater Today Bio 2024; 27:101126. [PMID: 38994470 PMCID: PMC11237977 DOI: 10.1016/j.mtbio.2024.101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/23/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Posterior segment disease acts as a major cause of irreversible visual impairments. Successful treatment of posterior segment disease requires the efficient delivery of therapeutic substances to the targeted lesion. However, the complex ocular architecture makes the bioavailability of topically applied drugs extremely low. Invasive delivery approaches like intravitreal injection may cause adverse complications. To enhance the efficiency, several biomedical engineering systems have been developed to increase the penetration efficiency and improve the bioavailability of drugs at the posterior segments. Advantageously, biodegradable microspheres are found to deliver the therapeutic agents in a controlled fashion. The microspheres prepared from novel biomaterials can realize the prolonged release at the posterior segment with minimum side effects. Moreover, it will be degraded automatically into products that are non-toxic to the human body without the necessity of secondary operation to remove the residual polymer matrix. Additionally, biodegradable microspheres have decent thermoplasticity, adjustable hydrophilicity, controlled crystallinity, and high tensile strength, which make them suitable for intraocular delivery. In this review, we introduce the latest advancements in microsphere production technology and elaborate on the biomaterials that are used to prepare microspheres. We discuss systematically the pharmacological characteristics of biodegradable microspheres and compare their potential advantages and limitations in the treatment of posterior segment diseases. These findings would enrich our knowledge of biodegradable microspheres and cast light into the discovery of effective biomaterials for ocular drug delivery.
Collapse
Affiliation(s)
- Rongyue Xue
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Hao Wu
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Siyu Li
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Ning Pu
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Dong Wei
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Na Zhao
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Yongheng Cui
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Haoyan Li
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
- College of Medicine, Zhengzhou university, Zhengzhou, China
| | - Zongming Song
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Ye Tao
- Department of Ophthalmology, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| |
Collapse
|
6
|
Esih H, Mezgec K, Billmeier M, Malenšek Š, Benčina M, Grilc B, Vidmar S, Gašperlin M, Bele M, Zidarn M, Zupanc TL, Morgan T, Jordan I, Sandig V, Schrödel S, Thirion C, Protzer U, Wagner R, Lainšček D, Jerala R. Mucoadhesive film for oral delivery of vaccines for protection of the respiratory tract. J Control Release 2024; 371:179-192. [PMID: 38795814 DOI: 10.1016/j.jconrel.2024.05.041] [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: 02/05/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
The delivery of vaccines plays a pivotal role in influencing the strength and longevity of the immune response and controlling reactogenicity. Mucosal immunization, as compared to parenteral vaccination, could offer greater protection against respiratory infections while being less invasive. While oral vaccination has been presumed less effective and believed to target mainly the gastrointestinal tract, trans-buccal delivery using mucoadhesive films (MAF) may allow targeted delivery to the mucosa. Here we present an effective strategy for mucosal delivery of several vaccine platforms incorporated in MAF, including DNA plasmids, viral vectors, and lipid nanoparticles incorporating mRNA (mRNA/LNP). The mRNA/LNP vaccine formulation targeting SARS-CoV-2 as a proof of concept remained stable within MAF consisting of slowly releasing water-soluble polymers and an impermeable backing layer, facilitating enhanced penetration into the oral mucosa. This formulation elicited antibody and cellular responses comparable to the intramuscular injection, but also induced the production of mucosal IgAs, highlighting its efficacy, particularly for use as a booster vaccine and the potential advantage for protection against respiratory infections. The MAF vaccine preparation demonstrates significant advantages, such as efficient delivery, stability, and simple noninvasive administration with the potential to alleviate vaccine hesitancy.
Collapse
Affiliation(s)
- Hana Esih
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Klemen Mezgec
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Martina Billmeier
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia
| | - Blaž Grilc
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Sara Vidmar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mirjana Gašperlin
- University of Ljubljana, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ljubljana 1000, Slovenia
| | - Marjan Bele
- Department of Materials Chemistry, National Institute of Chemistry, Ljubljana 1000, Slovenia
| | - Mihaela Zidarn
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | | | - Tina Morgan
- University Clinic of Pulmonary and Allergic Diseases Golnik, Golnik, Slovenia
| | - Ingo Jordan
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Volker Sandig
- Applied Science & Technologies, ProBioGen AG, Berlin, Germany
| | - Silke Schrödel
- SIRION Biotech GmbH, Am Klopferspitz 19, 82152 Martinsried, Germany
| | | | - Ulrike Protzer
- Institute of Virology, School of Medicine, Technical University of Munich, Helmholtz Zentrum München, Munich, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology & Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany; Institute of Clinical Microbiology & Hygiene, University Hospital, Regensburg, Germany
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, 1000 Ljubljana, Slovenia.
| |
Collapse
|
7
|
Faria MJ, González-Méijome JM, Real Oliveira MECD, Carracedo G, Lúcio M. Recent advances and strategies for nanocarrier-mediated topical therapy and theranostic for posterior eye disease. Adv Drug Deliv Rev 2024; 210:115321. [PMID: 38679293 DOI: 10.1016/j.addr.2024.115321] [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: 02/16/2024] [Revised: 04/08/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
Posterior eye disorders, such as age-related macular degeneration, diabetic retinopathy, and glaucoma, have a significant impact on human quality of life and are the primary cause of age-related retinal diseases among adults. There is a pressing need for innovative topical approaches to treat posterior eye disorders, as current methods often rely on invasive procedures with inherent risks. Limited success was attained in the realm of topical ophthalmic delivery through non-invasive means. Additionally, there exists a dearth of literature that delves into the potential of this approach for drug delivery and theranostic purposes, or that offers comprehensive design strategies for nanocarrier developers to surmount the significant physiological ocular barriers. This review offers a thorough and up-to-date state-of-the-art overview of 40 studies on therapeutic loaded nanocarriers and theranostic devices that, to the best of our knowledge, represent all successful works that reached posterior eye segments through a topical non-invasive administration. Most importantly, based on the successful literature studies, this review provides a comprehensive summary of the potential design strategies that can be implemented during nanocarrier development to overcome each ocular barrier.
Collapse
Affiliation(s)
- Maria João Faria
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - José M González-Méijome
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal; CEORLab - Clinical and Experimental Optometry Research Lab, Centre of Physics, Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - M Elisabete C D Real Oliveira
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| | - Gonzalo Carracedo
- Department of Optometry and Vision, Faculty of Optics and Optometry, University Complutense of Madrid, C/Arcos de Jalon 118, Madrid 28037, Spain.
| | - Marlene Lúcio
- Centre of Physics of Minho and Porto Universities (CF-UM-UP), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal; CBMA - Centre of Molecular and Environmental Biology, Department of Biology, Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.
| |
Collapse
|
8
|
Wu J, Fu K, Hou C, Wang Y, Ji C, Xue F, Ren J, Dai J, Barr JJ, Tang F. Bacteriophage defends murine gut from Escherichia coli invasion via mucosal adherence. Nat Commun 2024; 15:4764. [PMID: 38834561 DOI: 10.1038/s41467-024-48560-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Bacteriophage are sophisticated cellular parasites that can not only parasitize bacteria but are increasingly recognized for their direct interactions with mammalian hosts. Phage adherence to mucus is known to mediate enhanced antimicrobial effects in vitro. However, little is known about the therapeutic efficacy of mucus-adherent phages in vivo. Here, using a combination of in vitro gastrointestinal cell lines, a gut-on-a-chip microfluidic model, and an in vivo murine gut model, we demonstrated that a E. coli phage, øPNJ-6, provided enhanced gastrointestinal persistence and antimicrobial effects. øPNJ-6 bound fucose residues, of the gut secreted glycoprotein MUC2, through domain 1 of its Hoc protein, which led to increased intestinal mucus production that was suggestive of a positive feedback loop mediated by the mucus-adherent phage. These findings extend the Bacteriophage Adherence to Mucus model into phage therapy, demonstrating that øPNJ-6 displays enhanced persistence within the murine gut, leading to targeted depletion of intestinal pathogenic bacteria.
Collapse
Affiliation(s)
- Jiaoling Wu
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Kailai Fu
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Chenglin Hou
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Yuxin Wang
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Chengyuan Ji
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Feng Xue
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Jianluan Ren
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
| | - Jianjun Dai
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China.
- School of Pharmacy, China Pharmaceutical University; Engineering Research Center for Anti-infective Drug Discovery, Ministry of Education (ERCADD), Nanjing, China.
| | - Jeremy J Barr
- School of Biological Sciences, Monash University, Victoria, Australia.
| | - Fang Tang
- College of Veterinary Medicine, Nanjing Agricultural University; Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing, China.
| |
Collapse
|
9
|
Zhao L, Arias SL, Zipfel W, Brito IL, Yeo J. Coarse-grained modeling and dynamics tracking of nanoparticles diffusion in human gut mucus. Int J Biol Macromol 2024; 267:131434. [PMID: 38614182 DOI: 10.1016/j.ijbiomac.2024.131434] [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: 10/29/2023] [Revised: 02/23/2024] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
The gastrointestinal (GI) tract's mucus layer serves as a critical barrier and a mediator in drug nanoparticle delivery. The mucus layer's diverse molecular structures and spatial complexity complicates the mechanistic study of the diffusion dynamics of particulate materials. In response, we developed a bi-component coarse-grained mucus model, specifically tailored for the colorectal cancer environment, that contained the two most abundant glycoproteins in GI mucus: Muc2 and Muc5AC. This model demonstrated the effects of molecular composition and concentration on mucus pore size, a key determinant in the permeability of nanoparticles. Using this computational model, we investigated the diffusion rate of polyethylene glycol (PEG) coated nanoparticles, a widely used muco-penetrating nanoparticle. We validated our model with experimentally characterized mucus pore sizes and the diffusional coefficients of PEG-coated nanoparticles in the mucus collected from cultured human colorectal goblet cells. Machine learning fingerprints were then employed to provide a mechanistic understanding of nanoparticle diffusional behavior. We found that larger nanoparticles tended to be trapped in mucus over longer durations but exhibited more ballistic diffusion over shorter time spans. Through these discoveries, our model provides a promising platform to study pharmacokinetics in the GI mucus layer.
Collapse
Affiliation(s)
- Liming Zhao
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| | - Sandra L Arias
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Warren Zipfel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA.
| | - Jingjie Yeo
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
10
|
Jiang Y, Fu X, Shao M, Chang W, Zhang H, Liu Z. Eyedrop delivery of therapeutic proteins with zwitterionic polymers to treat dry age-related macular degeneration. Biomaterials 2024; 305:122429. [PMID: 38150770 DOI: 10.1016/j.biomaterials.2023.122429] [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: 08/31/2023] [Revised: 11/23/2023] [Accepted: 12/08/2023] [Indexed: 12/29/2023]
Abstract
In clinics, therapeutic proteins are commonly used to treat retinal diseases through intraocular injection, the treatment which suffers from rather low patient compliance. Topical administration (e.g. eye-drops) of large molecule drugs remains a major challenge due to the presence of various barriers in the eye. In this study, zwitterion-grafted chitosan (CS-ZW) was developed and then self-assembled with protein therapeutics including adalimumab (ADA) or catalase (CAT) for the treatment of dry age-related macular degeneration (dAMD) via topical eyedrops. Since CS-ZW can cross the mucus layer and open the tight junctions between epithelial cells, their delivered therapeutic proteins can be shuttled across the ocular barriers to reach the diseased site in the fundus. CS-ZW/ADA eyedrops delivering ADA to bind TNF-α in the fundus achieved a similar therapeutic effect to intravitreal ADA injection in a mouse dAMD model. In addition, the therapeutic effect was further improved by combining eyedrop formulations of CS-ZW/ADA and CS-ZW/CAT, the latter of which can clear reactive oxygen species (ROS) in the lesion to further assist dAMD treatment. Our work provides a simple and effective delivery vehicle that can non-invasively treat fundus diseases such as dAMD, showing potential advantages in reducing side effects associated with intraocular injection and improving patient compliance.
Collapse
Affiliation(s)
- Yutong Jiang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Xuehui Fu
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ming Shao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Wanwan Chang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China; Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa 999078, China
| | - Han Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| |
Collapse
|
11
|
Aldeeb MME, Wilar G, Suhandi C, Elamin KM, Wathoni N. Nanosuspension-Based Drug Delivery Systems for Topical Applications. Int J Nanomedicine 2024; 19:825-844. [PMID: 38293608 PMCID: PMC10824615 DOI: 10.2147/ijn.s447429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Nanosuspensions have garnered recent attention as a promising strategy for mitigating the bioavailability challenges of hydrophobic drugs, particularly those characterized by poor solubility in both aqueous and organic environments. Addressing solubility issues associated with poorly water-soluble drugs has largely resolved the need to enhance drug absorption and bioavailability. As mucosal formulations and topical administration progress in the future, nanosuspension drug delivery, straightforward formulation techniques, and versatile applications will continue to be subjects of interest. Nanosuspensions have undergone extensive scrutiny in preparation for topical applications, encompassing ocular, pulmonary, and dermal usage. Among the numerous methods aimed at improving cutaneous application, nanocrystals represent a relatively recent yet profoundly intriguing approach. Despite the increasing availability of various nanosuspension products, primarily designed for oral administration, only a limited number of studies have explored skin permeability and drug accumulation in the context of nanosuspensions. Nevertheless, the scant published research unequivocally underscores the potential of this approach for enhancing cutaneous bioavailability, particularly for active ingredients with low to medium solubility. Nanocrystals exhibit increased skin adhesiveness in addition to heightened saturation solubility and dissolution rate, thereby augmenting cutaneous distribution. The article provides a comprehensive overview of nanosuspensions for topical application. The methodology employed is robust, with a well-defined experimental design; however, the limited sample size raises concerns about the generalizability of the findings. While the results demonstrate promising outcomes in terms of enhanced drug delivery, the discussion falls short of addressing certain limitations. Additionally, the references largely focus on recent studies, but a more diverse inclusion of historical perspectives could offer a more holistic view of the subject.
Collapse
Affiliation(s)
- Mohamed Mahmud E Aldeeb
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Department of Pharmaceutics, Faculty of Pharmacy, Elmergib University, Alkhoms, 40414, Libya
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Khaled M Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| |
Collapse
|
12
|
Ashique S, Mishra N, Mohanto S, Gowda BJ, Kumar S, Raikar AS, Masand P, Garg A, Goswami P, Kahwa I. Overview of processed excipients in ocular drug delivery: Opportunities so far and bottlenecks. Heliyon 2024; 10:e23810. [PMID: 38226207 PMCID: PMC10788286 DOI: 10.1016/j.heliyon.2023.e23810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024] Open
Abstract
Ocular drug delivery presents a unique set of challenges owing to the complex anatomy and physiology of the eye. Processed excipients have emerged as crucial components in overcoming these challenges and improving the efficacy and safety of ocular drug delivery systems. This comprehensive overview examines the opportunities that processed excipients offer in enhancing drug delivery to the eye. By analyzing the current landscape, this review highlights the successful applications of processed excipients, such as micro- and nano-formulations, sustained-release systems, and targeted delivery strategies. Furthermore, this article delves into the bottlenecks that have impeded the widespread adoption of these excipients, including formulation stability, biocompatibility, regulatory constraints, and cost-effectiveness. Through a critical evaluation of existing research and industry practices, this review aims to provide insights into the potential avenues for innovation and development in ocular drug delivery, with a focus on addressing the existing challenges associated with processed excipients. This synthesis contributes to a deeper understanding of the promising role of processed excipients in improving ocular drug delivery systems and encourages further research and development in this rapidly evolving field.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, 474005, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - B.H. Jaswanth Gowda
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK
| | - Shubneesh Kumar
- Department of Pharmaceutics, Bharat Institute of Technology, School of Pharmacy, Meerut 250103, UP, India
| | - Amisha S. Raikar
- Department of Pharmaceutics, PES Rajaram and Tarabai Bandekar College of Pharmacy, Ponda, Goa 403401, India
| | - Priya Masand
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, (MIET), NH-58, Delhi-Roorkee Highway, Meerut, Uttar Pradesh 250005, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, India
| | - Priyanka Goswami
- Department of Pharmacognosy, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar 382355, Gujarat, India
- Maharashtra Educational Society's H.K. College of Pharmacy, Mumbai: 400102.India
| | - Ivan Kahwa
- Department of Pharmacy, Faculty of Medicine, Mbarara University of Science and Technology, P.O Box 1410, Mbarara, Uganda
- Pharm-Bio Technology and Traditional Medicine Centre, Mbarara University of Science and Technology, P. O Box 1410, Mbarara, Uganda
| |
Collapse
|
13
|
Wang X, Ding G, Yang P, Cheng G, Kong W, Xu Z. Teleost Eye Is the Portal of IHNV Entry and Contributes to a Robust Mucosal Immune Response. Int J Mol Sci 2023; 25:160. [PMID: 38203332 PMCID: PMC10778588 DOI: 10.3390/ijms25010160] [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: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The ocular mucosa (OM) is an important and unique part of the vertebrate mucosal immune system. The OM plays an important role in maintaining visual function and defending against foreign antigens or microorganisms, while maintaining a balance between the two through complex regulatory mechanisms. However, the function of ocular mucosal defense against foreign pathogens and mucosal immune response in bony fish are still less studied. To acquire deeper understanding into the mucosal immunity of the OM in teleost fish, we established a study of the immune response of rainbow trout (Oncorhynchus mykiss) infected with the infectious hematopoietic necrosis virus (IHNV). Our findings revealed that IHNV could successfully infiltrate the trout's OM, indicating that the OM could be an important portal for the IHNV. Furthermore, qPCR and RNA-Seq analysis results showed that a large number of immune-related genes were significantly upregulated in the OM of trout with IHNV infection. Critically, the results of our RNA-Seq analysis demonstrated that viral infection triggered a robust immune response, as evidenced by the substantial induction of antiviral, innate, and adaptive immune-related genes in the OM of infected fish, which underscored the essential role of the OM in viral infection. Overall, our findings revealed a previously unknown function of teleost OM in antiviral defense, and provided a theoretical basis for the study of the mucosal immunity of fish.
Collapse
Affiliation(s)
- Xinyou Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China;
| | - Guangyi Ding
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (G.D.); (G.C.); (W.K.)
| | - Peng Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (G.D.); (G.C.); (W.K.)
| | - Gaofeng Cheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (G.D.); (G.C.); (W.K.)
| | - Weiguang Kong
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (G.D.); (G.C.); (W.K.)
| | - Zhen Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (G.D.); (G.C.); (W.K.)
| |
Collapse
|
14
|
Kannan RM, Pitha I, Parikh KS. A new era in posterior segment ocular drug delivery: Translation of systemic, cell-targeted, dendrimer-based therapies. Adv Drug Deliv Rev 2023; 200:115005. [PMID: 37419213 DOI: 10.1016/j.addr.2023.115005] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/16/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Vision impairment and loss due to posterior segment ocular disorders, including age-related macular degeneration and diabetic retinopathy, are a rapidly growing cause of disability globally. Current treatments consist primarily of intravitreal injections aimed at preventing disease progression and characterized by high cost and repeated clinic visits. Nanotechnology provides a promising platform for drug delivery to the eye, with potential to overcome anatomical and physiological barriers to provide safe, effective, and sustained treatment modalities. However, there are few nanomedicines approved for posterior segment disorders, and fewer that target specific cells or that are compatible with systemic administration. Targeting cell types that mediate these disorders via systemic administration may unlock transformative opportunities for nanomedicine and significantly improve patient access, acceptability, and outcomes. We highlight the development of hydroxyl polyamidoamine dendrimer-based therapeutics that demonstrate ligand-free cell targeting via systemic administration and are under clinical investigation for treatment of wet age-related macular degeneration.
Collapse
Affiliation(s)
- Rangaramanujam M Kannan
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Chemical and Biomolecular Engineering and Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Ian Pitha
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kunal S Parikh
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Bioengineering Innovation & Design, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Coco G, Ambrosini G, Poletti S, Meliante LA, Taloni A, Scorcia V, Giannaccare G. Recent advances in drug treatments for dry eye disease. Expert Opin Pharmacother 2023; 24:2059-2079. [PMID: 37804227 DOI: 10.1080/14656566.2023.2269090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/06/2023] [Indexed: 10/09/2023]
Abstract
INTRODUCTION Dry eye disease (DED) is a common ocular condition with a significant impact on patients' quality of life. Conventional treatments include behavioral changes, tear substitutes, and anti-inflammatory agents; however, recent advances in the understanding of DED pathogenesis have opened the way to the development of novel treatment strategies able to target several pathways involved in the onset and persistence of DED. AREAS COVERED Literature search was conducted on PubMed and Scopus around the term 'dry eye disease' and others involving its pathophysiology and therapeutic strategy. The primary focus was on recent drugs approved by FDA or under investigation in phase 3 clinical trials. Google and ClinicalTrials.gov were used for obtaining information about the status of FDA approval and ongoing clinical trials. EXPERT OPINION Due to its multifaced pathogenesis, DED management is often challenging, and patients' needs are frequently unmet. Recently, several novel treatments have been either FDA-approved or studied in late-phase trials. These novel drugs target-specific biological components of the ocular surface and reduce inflammation and ocular pain. Additionally, new drug delivery systems allow for increased bioavailability, improve effective dosing, and minimize ocular side effects. These advances in drug therapies show real promise for better management of DED patients.
Collapse
Affiliation(s)
- Giulia Coco
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giacomo Ambrosini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Silvia Poletti
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Laura Antonia Meliante
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Taloni
- Department of Ophthalmology, University of Magna Græcia, Catanzaro, Italy
| | - Vincenzo Scorcia
- Department of Ophthalmology, University of Magna Græcia, Catanzaro, Italy
| | | |
Collapse
|
16
|
Xie G, Lin S, Wu F, Liu J. Nanomaterial-based ophthalmic drug delivery. Adv Drug Deliv Rev 2023; 200:115004. [PMID: 37433372 DOI: 10.1016/j.addr.2023.115004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/13/2023]
Abstract
The low bioavailability and side effects of conventional drugs for eye disease necessitate the development of efficient drug delivery systems. Accompanying the developments of nanofabrication techniques, nanomaterials have been recognized as promising tools to overcome these challenges due to their flexible and programmable properties. Given the advances achieved in material science, a broad spectrum of functional nanomaterials capable of overcoming various ocular anterior and posterior segment barriers have been explored to satisfy the demands for ocular drug delivery. In this review, we first highlight the unique functions of nanomaterials suitable for carrying and transporting ocular drugs. Then, various functionalization strategies are emphasized to endow nanomaterials with superior performance in enhanced ophthalmic drug delivery. The rational design of several affecting factors is essential for ideal nanomaterial candidates and is depicted as well. Lastly, we introduce the current applications of nanomaterial-based delivery systems in the therapy of different ocular anterior and posterior segment diseases. The limitations of these delivery systems as well as potential solutions are also discussed. This work will inspire innovative design thinking for the development of nanotechnology-mediated strategies for advanced drug delivery and treatment toward ocular diseases.
Collapse
Affiliation(s)
- Guocheng Xie
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
17
|
Martinez-Carrasco R, Rachagani S, Batra SK, Argüeso P, Fini ME. Roles unveiled for membrane-associated mucins at the ocular surface using a Muc4 knockout mouse model. Sci Rep 2023; 13:13558. [PMID: 37604830 PMCID: PMC10442421 DOI: 10.1038/s41598-023-40491-0] [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: 06/15/2023] [Accepted: 08/11/2023] [Indexed: 08/23/2023] Open
Abstract
Membrane-associated mucins (MAMs) are proposed to play critical roles at the ocular surface; however, in vivo evidence has been lacking. Here we investigate these roles by phenotyping of a Muc4 KO mouse. Histochemical analysis for expression of the beta-galactosidase transgene replacing Muc4 revealed a spiraling ribbon pattern across the corneal epithelium, consistent with centripetal cell migration from the limbus. Depletion of Muc4 compromised transcellular barrier function, as evidenced by an increase in rose bengal staining. In addition, the corneal surface was less smooth, consistent with disruption of tear film stability. While surface cells presented with well-developed microprojections, an increase in the number of cells with fewer microprojections was observed. Moreover, an increase in skin-type keratin K10 and a decrease in transcription factor Pax6 was observed, suggesting an incipient transdifferentiation. Despite this, no evidence of inflammatory dry eye disease was apparent. In addition, Muc4 had no effect on signaling by toll-like receptor Tlr4, unlike reports for MUC1 and MUC16. Results of this study provide the first in vivo evidence for the role of MAMs in transcellular barrier function, tear film stability, apical epithelial cell architecture, and epithelial mucosal differentiation at the ocular surface.
Collapse
Affiliation(s)
- Rafael Martinez-Carrasco
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Satyanarayan Rachagani
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE, USA
- Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pablo Argüeso
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, 02111, USA
- Program in Immunology, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
- Program in Genetics, Molecular & Cellular Biology, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
- Program in Pharmacology & Drug Development, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center and Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Program in Genetics, Molecular & Cellular Biology, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
- Program in Pharmacology & Drug Development, Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|
18
|
De Hoon I, Boukherroub R, De Smedt SC, Szunerits S, Sauvage F. In Vitro and Ex Vivo Models for Assessing Drug Permeation across the Cornea. Mol Pharm 2023. [PMID: 37314950 DOI: 10.1021/acs.molpharmaceut.3c00195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Drug permeation across the cornea remains a major challenge due to its unique and complex anatomy and physiology. Static barriers such as the different layers of the cornea, as well as dynamic aspects such as the constant renewal of the tear film and the presence of the mucin layer together with efflux pumps, all present unique challenges for effective ophthalmic drug delivery. To overcome some of the current ophthalmic drug limitations, the identification and testing of novel drug formulations such as liposomes, nanoemulsions, and nanoparticles began to be considered and widely explored. In the early stages of corneal drug development reliable in vitro and ex vivo alternatives, are required, to be in line with the principles of the 3Rs (Replacement, Reduction, and Refinement), with such methods being in addition faster and more ethical alternatives to in vivo studies. The ocular field remains limited to a handful of predictive models for ophthalmic drug permeation. In vitro cell culture models are increasingly used when it comes to transcorneal permeation studies. Ex vivo models using excised animal tissue such as porcine eyes are the model of choice to study corneal permeation and promising advancements have been reported over the years. Interspecies characteristics must be considered in detail when using such models. This review updates the current knowledge about in vitro and ex vivo corneal permeability models and evaluates their advantages and limitations.
Collapse
Affiliation(s)
- Inès De Hoon
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Sabine Szunerits
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| | - Félix Sauvage
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
19
|
Tampucci S, Monti D, Burgalassi S, Terreni E, Paganini V, Di Gangi M, Chetoni P. Binary Polymeric Surfactant Mixtures for the Development of Novel Loteprednol Etabonate Nanomicellar Eyedrops. Pharmaceuticals (Basel) 2023; 16:864. [PMID: 37375811 DOI: 10.3390/ph16060864] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The treatment of several ocular inflammatory conditions affecting different areas of the ocular globe involves the administration of topical ophthalmic formulations containing corticosteroids. This research was aimed at evaluating the solubilising efficacy of 5.0% w/w of different binary mixtures of commercial amphiphilic polymeric surfactants with the purpose of obtaining nanomicellar solutions containing a high amount of loteprednol etabonate (LE). The selected LE-TPGS/HS nanomicelles, containing 0.253 mg/mL of the drug, had a small size (=13.57 nm) and uniform distribution (Polydispersity Index = 0.271), appeared completely transparent and perfectly filterable through 0.2 μm membrane filter, and remained stable up to 30 days at 4 °C. The critical micellar concentration (CMCTPGS/HS) was 0.0983 mM and the negative value of the interaction parameter between the polymeric-surfactant-building unit (βTPGS/HS = -0.1322) confirmed the ability of the polymeric surfactants to interact, favouring the dissolution of LE into nanomicelles. The disappearance of the endothermic peak of LE in the DSC analysis confirmed the interactions of LE with the polymeric surfactants. LE-TPGS/HS produced in vitro LE which sustained diffusion for 44 h (more than 40% of encapsulated LE). Furthermore, the lack of a significant cytotoxic effect on a sensitive corneal epithelial cell line makes it a candidate for further biological studies.
Collapse
Affiliation(s)
- Silvia Tampucci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Italian Inter-University Center for the Promotion of the 3Rs in Teaching and Research, University of Pisa, 56122 Pisa, Italy
| | - Daniela Monti
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Italian Inter-University Center for the Promotion of the 3Rs in Teaching and Research, University of Pisa, 56122 Pisa, Italy
| | - Susi Burgalassi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Italian Inter-University Center for the Promotion of the 3Rs in Teaching and Research, University of Pisa, 56122 Pisa, Italy
| | | | | | | | - Patrizia Chetoni
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
- Italian Inter-University Center for the Promotion of the 3Rs in Teaching and Research, University of Pisa, 56122 Pisa, Italy
| |
Collapse
|
20
|
Wong KY, Liu Y, Zhou L, Wong MS, Liu J. Mucin-targeting-aptamer functionalized liposomes for delivery of cyclosporin A for dry eye diseases. J Mater Chem B 2023; 11:4684-4694. [PMID: 37161679 DOI: 10.1039/d3tb00598d] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Traditional eye drops are convenient to use; however, their effectiveness is limited by their poor retention time and bioavailability in the eyes due to ocular barriers. Therefore, strategies to enhance ocular drug delivery are required. Herein, we constructed a mucin-1 aptamer-functionalized liposome and loaded it with cyclosporin A, a common ocular drug in eye drops used to treat dry eye diseases (DED). Drug encapsulation slightly reduced the liposome size without changing the surface potential of liposomes. Approximately 90% of the cholesterol-modified aptamers were inserted to the liposomes. We evaluated the cytotoxicity, anti-inflammatory effects, cell permeability regulation, and retention time of liposomes in human corneal epithelial cells under dry eye conditions. These results suggest that the aptamer-functionalized liposomes are more efficient as nanocarriers than non-functionalized liposomes and drug-free liposomes. They restore inflammation levels by 1-fold and remain in the cells for up to 24 h. An in vivo study was also performed in a rat DED model, which demonstrated the efficacy of aptamer-functionalized liposomes in restoring tear production and corneal integrity. The present study demonstrated the capability of aptamer-functionalized liposomes in the delivery of ocular drugs for the management of ocular diseases.
Collapse
Affiliation(s)
- Ka-Ying Wong
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong.
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Yibo Liu
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong.
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Liping Zhou
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P. R. China
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P. R. China
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P. R. China
| | - Man-Sau Wong
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P. R. China
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, P. R. China
| | - Juewen Liu
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong.
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
21
|
Ben-David EA, Habibi M, Haddad E, Sammar M, Angel DL, Dror H, Lahovitski H, Booth AM, Sabbah I. Mechanism of nanoplastics capture by jellyfish mucin and its potential as a sustainable water treatment technology. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 869:161824. [PMID: 36720396 DOI: 10.1016/j.scitotenv.2023.161824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
The accumulation of nanoplastics (NPs) in the environment has raised concerns about their impact on human health and the biosphere. The main aim of this study is to understand the mechanism that governs the capture of NPs by jellyfish mucus extracted from the jellyfish Aurelia sp. (A.a.) and compare the capture/removal efficiency to that of conventional coagulants and mucus from other organisms. The efficacy of A.a mucus to capture polystyrene and acrylic NPs (∼100 nm) from spiked wastewater treatment plant (WWTP) effluent was evaluated. The mucus effect on capture kinetics and destabilization of NPs of different polymer compositions, sizes and concentrations was quantified by means of fluorescent NPs, dynamic light scattering and zeta potential measurements and visualized by scanning electron microscopy. A dosing of A.a. mucus equivalent to protein concentrations of ∼2-4 mg L-1 led to a rapid change in zeta potential from a baseline of -30 mV to values close to 0 mV, indicating a marked change from a stable to a non-stable dispersion leading to a rapid (<10 min) and significant removal of NPs (60 %-90 %) from a stable suspension. The A.a. mucus outperformed all other mucus types (0-37 %) and coagulants (0 %-32 % for ferric chloride; 23-40 % for poly aluminum chlorohydrate), highlighting the potential for jellyfish mucus to be used as bio-flocculant. The results indicate a mucus-particle interaction consisting of adsorption-bridging and "mesh" filtration. Further insight is provided by carbohydrate composition and protein disruption analysis. Total protein disruption resulted in a complete loss of the A.a. mucus capacity to capture NPs, while the breaking of disulfide bonds and protein unfolding resulted in improved capture capacity. The study demonstrates that natural jellyfish mucin can capture and remove NPs in water and wastewater treatment systems more efficiently than conventional coagulants, highlighting the potential for development of a new type of bio-flocculant.
Collapse
Affiliation(s)
- Eric A Ben-David
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Maryana Habibi
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Elias Haddad
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Marei Sammar
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel
| | - Dror L Angel
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | - Hila Dror
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | - Haim Lahovitski
- Department of Maritime Civilizations, and Recanati Institute for Maritime Studies, University of Haifa, Haifa, Israel
| | | | - Isam Sabbah
- Prof. Ephraim Katzir Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel; The Institute of Applied Research, The Galilee Society, Shefa-Amr, Israel.
| |
Collapse
|
22
|
Wang C, Pang Y. Nano-based eye drop: Topical and noninvasive therapy for ocular diseases. Adv Drug Deliv Rev 2023; 194:114721. [PMID: 36773886 DOI: 10.1016/j.addr.2023.114721] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/11/2023]
Abstract
Eye drops are the most accessible therapy for ocular diseases, while inevitably suffering from their lower bioavailability which highly restricts the treatment efficacy. The introduction of nanotechnology has attracted considerable interest as it has advantages over conventional ones such as prolonged ocular surface retention time and enhanced ocular barrier penetrating properties, and achieving higher bioavailability and improved treatment efficacy. This review describes various ocular diseases treated with eye drops as well as the physiological and anatomical ocular barriers faced with through drug administration. It also summarizes the recent advances regarding the utilization of nanotechnology in developing eye drops, and how to optimize the nanocarrier-based ocular drug delivery systems. The prospective future research directions for nano-based eye drops are also discussed here.
Collapse
Affiliation(s)
- Chuhan Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yan Pang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
23
|
Perez VL, Mah FS, Willcox M, Pflugfelder S. Anti-Inflammatories in the Treatment of Dry Eye Disease: A Review. J Ocul Pharmacol Ther 2023; 39:89-101. [PMID: 36796014 DOI: 10.1089/jop.2022.0133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Inflammation is an important driver of dry eye disease (DED) pathogenesis. An initial insult that results in the loss of tear film homeostasis can initiate a nonspecific innate immune response that leads to a chronic and self-sustaining inflammation of the ocular surface, which results in classic symptoms of dry eye. This initial response is followed by a more prolonged adaptive immune response, which can perpetuate and aggravate inflammation and result in a vicious cycle of chronic inflammatory DED. Effective anti-inflammatory therapies can help patients exit this cycle, and effective diagnosis of inflammatory DED and selection of the most appropriate treatment are therefore key to successful DED management and treatment. This review explores the cellular and molecular mechanisms of the immune and inflammatory components of DED, and examines the evidence base for the use of currently available topical treatment options. These agents include topical steroid therapy, calcineurin inhibitors, T cell integrin antagonists, antibiotics, autologous serum/plasma therapy, and omega-3 fatty acid dietary supplements.
Collapse
Affiliation(s)
- Victor L Perez
- Department of Ophthalmology, Foster Center for Ocular Immunology at Duke Eye Center, Duke University School of Medicine, Durham, North Carolina. USA
| | - Francis S Mah
- Scripps Clinic Torrey Pines, La Jolla, California, USA
| | - Mark Willcox
- School of Optometry and Vision Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - Stephen Pflugfelder
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
24
|
Onugwu AL, Nwagwu CS, Onugwu OS, Echezona AC, Agbo CP, Ihim SA, Emeh P, Nnamani PO, Attama AA, Khutoryanskiy VV. Nanotechnology based drug delivery systems for the treatment of anterior segment eye diseases. J Control Release 2023; 354:465-488. [PMID: 36642250 DOI: 10.1016/j.jconrel.2023.01.018] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/07/2023] [Indexed: 01/17/2023]
Abstract
Diseases affecting the anterior segment of the eye are the primary causes of vision impairment and blindness globally. Drug administration through the topical ocular route is widely accepted because of its user/patient friendliness - ease of administration and convenience. However, it remains a significant challenge to efficiently deliver drugs to the eye through this route because of various structural and physiological constraints that restrict the distribution of therapeutic molecules into the ocular tissues. The bioavailability of topically applied ocular medications such as eye drops is typically less than 5%. Developing novel delivery systems to increase the retention time on the ocular surfaces and permeation through the cornea is one of the approaches adopted to boost the bioavailability of topically administered medications. Drug delivery systems based on nanotechnology such as micelles, nanosuspensions, nanoparticles, nanoemulsions, liposomes, dendrimers, niosomes, cubosomes and nanowafers have been investigated as effective alternatives to conventional ocular delivery systems in treating diseases of the anterior segment of the eye. This review discussed different nanotechnology-based delivery systems that are currently investigated for treating and managing diseases affecting the anterior ocular tissues. We also looked at the challenges in translating these systems into clinical use and the prospects of nanocarriers as a vehicle for the delivery of phytoactive compounds to the anterior segment of the eye.
Collapse
Affiliation(s)
- Adaeze Linda Onugwu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chinekwu Sherridan Nwagwu
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Obinna Sabastine Onugwu
- Department of Pharmacognosy, Enugu State University of Science and Technology, Agbani, Enugu State, Nigeria
| | - Adaeze Chidiebere Echezona
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chinazom Precious Agbo
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Stella Amarachi Ihim
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Enugu State, Nigeria; Pharmacology and Physiology Unit, Department of Science Laboratory Technology, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Prosper Emeh
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Petra Obioma Nnamani
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Anthony Amaechi Attama
- Drug Delivery & Nanomedicines Research Laboratory, Department of Pharmaceutics, University of Nigeria, Nsukka, Enugu State, Nigeria; Department of Pharmaceutics and Pharmaceutical Technology, Enugu State University of Science and Technology, Agbani, Enugu State, Nigeria.
| | - Vitaliy V Khutoryanskiy
- Reading School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AD, United Kingdom.
| |
Collapse
|
25
|
Koganti R, Yadavalli T, Sutar Y, Mallick S, Date A, Shukla D. Topical phenylbutyrate antagonizes NF-κB signaling and resolves corneal inflammation. iScience 2022; 25:105682. [PMID: 36536680 PMCID: PMC9758524 DOI: 10.1016/j.isci.2022.105682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/18/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Chronic inflammation of the immune privileged cornea originating from viral or nonviral conditions results in significant vision loss. Topical corticosteroids are the common treatments for corneal inflammation, but the drugs cause serious and potentially blinding side effects in the long term. Therefore, new standalone and/or synergistic anti-inflammatory therapies with lower side effects are desperately needed. Here, we show that the aromatic fatty acid phenylbutyrate (PBA) acts as a potent inhibitor of inflammation in preclinical ocular-inflammation models. PBA prevents the transcription as well as translation of pro-inflammatory cytokines by LPS and poly(I:C) via persistent inhibition of NF-κB signaling. PBA quickens the resolution of ocular inflammation in mice by decreasing corneal thickness and immune cell infiltration. More importantly, PBA can synergize with the dexamethasone to antagonize NF-κB signaling at lower drug concentrations. Our results demonstrate that PBA therapy exerts previously unreported anti-inflammatory effects in the eye and facilitates corneal healing during persistent inflammation.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
| | - Yogesh Sutar
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
- R. Ken Coit College of Pharmacy, The University of Arizona, Tuscon, AZ 85721, USA
| | - Sudipta Mallick
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
| | - Abhijit Date
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii Hilo, Hilo, HI 96720, USA
- R. Ken Coit College of Pharmacy, The University of Arizona, Tuscon, AZ 85721, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
26
|
Preparation and Evaluation of Modified Chitosan Nanoparticles Using Anionic Sodium Alginate Polymer for Treatment of Ocular Disease. Pharmaceutics 2022; 14:pharmaceutics14122802. [PMID: 36559295 PMCID: PMC9786214 DOI: 10.3390/pharmaceutics14122802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Mucoadhesive nanoparticles offer prolonged drug residence time at the corneal epithelium by adhering to the mucous layer of the eye. Here, in this research investigation, voriconazole-loaded chitosan mucoadhesive nanoparticles (VCZ-MA-NPs) were modified to mucous-penetrating nanoparticles (VCZ-MP-NPs) by coating them with anionic polymer sodium alginate. The ionic gelation method was utilized to prepare mucoadhesive chitosan nanoparticles, which were further coated with sodium alginate to obtain the surface properties essential for mucous penetration. The developed VCZ-MA-NPs and VCZ-MP-NPs were evaluated extensively for physicochemical delineation, as well as in vitro and ex vivo studies. The particle size, polydispersity index, and ζ potential of the VCZ-MA-NPs were discovered to be 116 ± 2 nm, 0.23 ± 0.004, and +16.3 ± 0.9 mV, while the equivalent values for VCZ-MP-NPs were 185 ± 1 nm, 0.20 ± 0.01, and -24 ± 0.9 mV, respectively. The entrapment efficiency and drug loading were obtained as 88.06%±1.29% and 7.27% ± 0.95% for VCZ-MA-NPs and 91.31% ± 1.05% and 10.38% ± 0.87% for VCZ-MP-NPs, respectively. The formulations were found to be stable under different conditions (4 °C, 25 °C, and 40 °C). Chitosan nanoparticles and modified nanoparticles showed a spherical and smooth morphology under electron microscopic imaging. An excised caprine cornea was used for the ex vivo permeation study, exhibiting 58.98% ± 0.54% and 70.02% ± 0.61% drug permeation for VCZ-MA-NPs and VCZ-MP-NPs, respectively. The findings revealed that the mucous-penetrating nanoparticles could effectively pass through the corneal epithelium, thus overcoming the mucous barrier and fungal layer of the eye, which highlights their potential in the treatment of fungal keratitis.
Collapse
|
27
|
Pho T, Champion JA. Surface Engineering of Protein Nanoparticles Modulates Transport, Adsorption, and Uptake in Mucus. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51697-51710. [PMID: 36354361 DOI: 10.1021/acsami.2c14670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Protein nanoparticles have been demonstrated as effective carriers for protein antigens and therapeutics due to properties endowed by their protein composition. They exhibit high protein to carrier yields, biocompatibility, and heterogeneous surface properties. While protein nanoparticles have been delivered via multiple routes, including intranasal, their interactions with mucosal barriers have not been well studied or modified. Biological barriers associated with intranasal delivery consist of viscoelastic mucus that hinders material transport through surface interactions and the underlying epithelium. Herein, we altered protein nanoparticle surface properties and characterized interactions with nasal mucus and the subsequent effects on diffusion, cellular uptake, and immune cell maturation. Ovalbumin protein nanoparticles were used, serving as a model vaccine nanoparticle. Unmodified ovalbumin protein nanoparticles were compared to cationic ovalbumin particles functionalized with amine groups, neutral particles functionalized with polyethylene glycol, and zwitterionic particles coated layer-by-layer (LBL) with chitosan and oligonucleotides. Transport analysis indicated rapid diffusion of polyethylene glycol and LBL-modified ovalbumin nanoparticles in porcine nasal mucus, while cationic particles were mucoadhesive. Cellular uptake in the presence of mucus by epithelial and dendritic cells was highest for particles containing positive charges, both LBL and amine-functionalized. These particles also exhibited the most diverse adsorbed protein corona from nasal fluids. The corona impacted both dendritic cell uptake and maturation, with polyethylene glycol and LBL modifications improving CD86 expression. Altogether, surface modifications on protein-based nanocarriers are shown to facilitate distinctive physical and cellular behavior associated with mucosal delivery.
Collapse
Affiliation(s)
- Thomas Pho
- School of Chemical and Biomolecular Engineering, BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia30332-2000, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, BioEngineering Program, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, Georgia30332-2000, United States
| |
Collapse
|
28
|
Vaginal Nanoformulations for the Management of Preterm Birth. Pharmaceutics 2022; 14:pharmaceutics14102019. [PMID: 36297454 PMCID: PMC9611874 DOI: 10.3390/pharmaceutics14102019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 12/01/2022] Open
Abstract
Preterm birth (PTB) is a leading cause of infant morbidity and mortality in the world. In 2020, 1 in 10 infants were born prematurely in the United States. The World Health Organization estimates that a total of 15 million infants are born prematurely every year. Current therapeutic interventions for PTB have had limited replicable success. Recent advancements in the field of nanomedicine have made it possible to utilize the vaginal administration route to effectively and locally deliver drugs to the female reproductive tract. Additionally, studies using murine models have provided important insights about the cervix as a gatekeeper for pregnancy and parturition. With these recent developments, the field of reproductive biology is on the cusp of a paradigm shift in the context of treating PTB. The present review focuses on the complexities associated with treating the condition and novel therapeutics that have produced promising results in preclinical studies.
Collapse
|
29
|
Optimization of Lipid Nanoparticles by Response Surface Methodology to Improve the Ocular Delivery of Diosmin: Characterization and In-Vitro Anti-Inflammatory Assessment. Pharmaceutics 2022; 14:pharmaceutics14091961. [PMID: 36145708 PMCID: PMC9506089 DOI: 10.3390/pharmaceutics14091961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022] Open
Abstract
Diosmin is a flavonoid with a great variety of biological activities including antioxidant and anti-inflammatory ones. Its cytoprotective effect in retinal pigment epithelium cells under high glucose conditions makes it a potential support in the treatment of diabetic retinopathy. Despite its benefits, poor solubility in water reduces its potential for therapeutic use, making it the biggest biopharmaceutical challenge. The design of diosmin-loaded nanocarriers for topical ophthalmic application represents a novelty that has not been yet explored. For this purpose, the response surface methodology (RSM) was used to optimize nanostructured lipid carriers (NLCs), compatible for ocular administration, to encapsulate diosmin and improve its physicochemical issues. NLCs were prepared by a simple and scalable technique: a melt emulsification method followed by ultrasonication. The experimental design was composed of four independent variables (solid lipid concentration, liquid lipid concentration, surfactant concentration and type of solid lipid). The effect of the factors was assessed on NLC size and PDI (responses) by analysis of variance (ANOVA). The optimized formulation was selected according to the desirability function (0.993). Diosmin at two different concentrations (80 and 160 µM) was encapsulated into NLCs. Drug-loaded nanocarriers (D-NLCs) were subjected to a physicochemical and technological investigation revealing a mean particle size of 83.58 ± 0.77 nm and 82.21 ± 1.12 nm, respectively for the D-NLC formulation prepared with diosmin at the concentration of 80 µM or 160 µM, and a net negative surface charge (−18.5 ± 0.60 and −18.0 ± 1.18, respectively for the two batches). The formulations were analyzed in terms of pH (6.5), viscosity, and adjusted for osmolarity, making them more compatible with the ocular environment. Subsequently, stability studies were carried out to assess D-NLC behavior under different storage conditions up to 60 days, indicating a good stability of NLC samples at room temperature. In-vitro studies on ARPE-19 cells confirmed the cytocompatibility of NLCs with retinal epithelium. The effect of D-NLCs was also evaluated in-vitro on a model of retinal inflammation, demonstrating the cytoprotective effect of D-NLCs at various concentrations. RSM was found to be a reliable model to optimize NLCs for diosmin encapsulation.
Collapse
|
30
|
Nan K, Feig VR, Ying B, Howarth JG, Kang Z, Yang Y, Traverso G. Mucosa-interfacing electronics. NATURE REVIEWS. MATERIALS 2022; 7:908-925. [PMID: 36124042 PMCID: PMC9472746 DOI: 10.1038/s41578-022-00477-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/28/2022] [Indexed: 06/15/2023]
Abstract
The surface mucosa that lines many of our organs houses myriad biometric signals and, therefore, has great potential as a sensor-tissue interface for high-fidelity and long-term biosensing. However, progress is still nascent for mucosa-interfacing electronics owing to challenges with establishing robust sensor-tissue interfaces; device localization, retention and removal; and power and data transfer. This is in sharp contrast to the rapidly advancing field of skin-interfacing electronics, which are replacing traditional hospital visits with minimally invasive, real-time, continuous and untethered biosensing. This Review aims to bridge the gap between skin-interfacing electronics and mucosa-interfacing electronics systems through a comparison of the properties and functions of the skin and internal mucosal surfaces. The major physiological signals accessible through mucosa-lined organs are surveyed and design considerations for the next generation of mucosa-interfacing electronics are outlined based on state-of-the-art developments in bio-integrated electronics. With this Review, we aim to inspire hardware solutions that can serve as a foundation for developing personalized biosensing from the mucosa, a relatively uncharted field with great scientific and clinical potential.
Collapse
Affiliation(s)
- Kewang Nan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Vivian R. Feig
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Binbin Ying
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Julia G. Howarth
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Ziliang Kang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Yiyuan Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
31
|
Jackson RM, Hatton CF, Spegarova JS, Georgiou M, Collin J, Stephenson E, Verdon B, Haq IJ, Hussain R, Coxhead JM, Mudhar HS, Wagner B, Hasoon M, Davey T, Rooney P, Khan CMA, Ward C, Brodlie M, Haniffa M, Hambleton S, Armstrong L, Figueiredo F, Queen R, Duncan CJA, Lako M. Conjunctival epithelial cells resist productive SARS-CoV-2 infection. Stem Cell Reports 2022; 17:1699-1713. [PMID: 35750043 PMCID: PMC9222349 DOI: 10.1016/j.stemcr.2022.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/01/2022] Open
Abstract
Conjunctival epithelial cells, which express viral-entry receptors angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine type 2 (TMPRSS2), constitute the largest exposed epithelium of the ocular surface tissue and may represent a relevant viral-entry route. To address this question, we generated an organotypic air-liquid-interface model of conjunctival epithelium, composed of basal, suprabasal, and superficial epithelial cells, and fibroblasts, which could be maintained successfully up to day 75 of differentiation. Using single-cell RNA sequencing (RNA-seq), with complementary imaging and virological assays, we observed that while all conjunctival cell types were permissive to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome expression, a productive infection did not ensue. The early innate immune response to SARS-CoV-2 infection in conjunctival cells was characterised by a robust autocrine and paracrine NF-κB activity, without activation of antiviral interferon signalling. Collectively, these data enrich our understanding of SARS-CoV-2 infection at the human ocular surface, with potential implications for the design of preventive strategies and conjunctival transplantation.
Collapse
Affiliation(s)
- Robert M Jackson
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Catherine F Hatton
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Maria Georgiou
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Emily Stephenson
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Bernard Verdon
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Hardeep-Singh Mudhar
- National Specialist Ophthalmic Pathology Service (NSOPS) Department of Histopathology, E-Floor, Royal Hallamshire Hospital, Sheffield, UK
| | - Bart Wagner
- Electron Microscopy Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Megan Hasoon
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Tracey Davey
- Electron Microscopy Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Paul Rooney
- NHS Blood and Transplant Tissue and Eye Services, Liverpool, UK
| | - C M Anjam Khan
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Chris Ward
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Malcolm Brodlie
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Sophie Hambleton
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Francisco Figueiredo
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK; Department of Ophthalmology, Royal Victoria Infirmary and Newcastle University, Newcastle, UK
| | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| | - Christopher J A Duncan
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
32
|
Khopade AJ, Halder A, Burade V, Pateliya B, Jani K, Patel V, Upadhyay S. Ophthalmic suspension of Brimonidine for sustained delivery using nano-resin/drug complex technique. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Sabri Bens M, Dassamiour S, Hambaba L, Akram Mela M, Sami R, M. Al-Mush AA, Benajiba N, Al Masoudi LM. In silico Investigation and BSA Denaturation Inhibitory Activity of Ethyl Acetate and N-butanol Extracts of Centaurea tougourensis Boiss. and Reut. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1296.1308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
34
|
Mohamed HB, Abd El-Hamid BN, Fathalla D, Fouad EA. Current trends in pharmaceutical treatment of Dry Eye Disease: A review. Eur J Pharm Sci 2022; 175:106206. [PMID: 35568107 DOI: 10.1016/j.ejps.2022.106206] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/02/2023]
Abstract
Dry eye disease (DED), keratoconjunctivitis sicca or dysfunctional tear syndrome, is the most prevalent ophthalmic disease which affects a substantial segment of people worldwide with increasing frequency. It is considered a multifactorial disease of the ocular surface and tear film, characterized by a variation of signs and symptoms. The symptoms range from mild to severe itching, burning, irritation, eye fatigue, and ocular inflammation that may lead to potential damage to the cornea, conjunctiva and even vision loss. Correspondingly, depending on the different manifestations and pathophysiology, the treatment must be tailored specifically to each patient by targeting the specific mechanisms implicated in their disease. Currently, there are several medical products and techniques available or under investigation for the treatment of DED. The present article focused on the pathophysiology of DED, the new diagnostic approach and the recently developed drug delivery systems or devices reducing the progress of the disease and treating the causes.
Collapse
Affiliation(s)
- Hebatallah B Mohamed
- Department of Pharmaceutics, Faculty of Pharmacy, South Valley University, Qena, 83523, Egypt.
| | - Basma N Abd El-Hamid
- Department of Pharmaceutics Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Dina Fathalla
- Department of Pharmaceutics Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Ehab A Fouad
- Department of Pharmaceutics Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
35
|
Cyclodextrins and drug membrane permeation: Thermodynamic considerations. J Pharm Sci 2022; 111:2571-2580. [PMID: 35487262 DOI: 10.1016/j.xphs.2022.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/22/2022]
Abstract
Cyclodextrins are hydrophilic oligosaccharides that can increase aqueous solubility of lipophilic drugs through formation of water-soluble drug/cyclodextrin complexes. Although the complexes are hydrophilic, and as such do not permeate biological membranes, the complexes are known to enhance drug permeation through lipophilic membranes and improve drug bioavailability after, for example, oral administration. However, it is not clear how cyclodextrins enhance the permeation. An artificial biomembrane (PermeaPad®) was used to study the effect of donor medium composition on drug permeation. It was observed that in aqueous solutions the hydrophilic cyclodextrins behave not like disperse systems but rather like organic cosolvents such as ethanol, increasing the solubility without having significant effect on the molecular mobility and ability of lipophilic drug molecules to partition into the lipophilic membrane. Also, that partition of dissolved drug molecules from the aqueous exterior into the membrane is at its maximum when their thermodynamic activity is at its maximum. In other words, that drug flux from aqueous cyclodextrin solutions through lipophilic membranes depends on both the concentration and the thermodynamic activity of dissolved drug. Maximum flux is obtained when both the drug concentration and thermodynamic activity of the dissolved drug molecules are at their maximum value.
Collapse
|
36
|
Loftsson T, Stefánsson E. Aqueous eye drops containing drug/cyclodextrin nanoparticles deliver therapeutic drug concentrations to both anterior and posterior segment. Acta Ophthalmol 2022; 100:7-25. [PMID: 33876553 DOI: 10.1111/aos.14861] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
Using topical application to deliver therapeutic concentrations of drugs to the posterior segment of the eye remains very challenging. As a result, posterior segment diseases are usually treated by intravitreal injection or implant. While topical treatments are commonly used for anterior segment conditions, they sometimes require frequent applications. Eye drop formulations based on γ-cyclodextrin (γCD)-based nanoparticle aggregates were developed, which in animal models and clinical studies deliver therapeutic concentrations of drugs (dorzolamide and dexamethasone) to both anterior and posterior segments of the eye. An early study in humans showed dorzolamide/γCD eye drops could achieve comparable intraocular pressure decreases to commercial dorzolamide eye drops, but with less frequent application. Pilot studies with dexamethasone/γCD eye drops suggested that they could be effective in a range of conditions, including diabetic macular oedema, cystoid macular oedema and vitritis secondary to uveitis, postcataract surgery inflammation and postoperative treatment in trabeculectomy. Phase II studies with similar dexamethasone/γCD nanoparticle eye drops in diabetic macular oedema and postcataract surgery inflammation have recently been completed. This technology has the potential to be used with other classes of drug molecules and to replace or complement invasive treatments, providing safer, non-invasive therapies, particularly for posterior segment conditions, that can be self-administered as eye drops by patients.
Collapse
|
37
|
Venkateswaran N, Bian Y, Gupta PK. Practical Guidance for the Use of Loteprednol Etabonate Ophthalmic Suspension 0.25% in the Management of Dry Eye Disease. Clin Ophthalmol 2022; 16:349-355. [PMID: 35173413 PMCID: PMC8843348 DOI: 10.2147/opth.s323301] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/24/2022] [Indexed: 11/30/2022] Open
Abstract
Dry eye disease (DED) is a prevalent ocular surface disease. Like with any chronic disease, patients with DED can experience episodic flares. There are many existing and upcoming treatments for the chronic treatment of DED, yet treatments for DED flares are limited. Loteprednol etabonate 0.25% is an FDA approved treatment modality for the short-term treatment of the signs and symptoms of DED. This medication is formulated with the customized mucus-penetrating particle (MPP) technology, which has a greater ability to penetrate the ocular surface and more effectively deliver the active steroid to the ocular surface tissues as compared with conventional steroid preparations. There is also increasing utility of loteprednol etabonate 0.25% in the treatment of DED before and/or after cataract or refractive surgery or as induction therapy prior to starting chronic immunomodulatory medication for DED.
Collapse
Affiliation(s)
| | - Yandong Bian
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Preeya K Gupta
- Triangle Eye Consultants, Cary, NC, USA
- Correspondence: Preeya K Gupta, Email
| |
Collapse
|
38
|
How liposomes pave the way for ocular drug delivery after topical administration. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Abstract
INTRODUCTION Retinal diseases are one of the main reasons for vision loss where all available drug treatments are based on invasive drug administration such as intravitreal injections. Despite huge efforts and some promising results in animal models, almost all delivery technologies tested have failed in human trials. There are however examples of clinically effective topical delivery systems such as fast dissolving aqueous eye drop suspensions. AREAS COVERED Six obstacles to topical drug delivery to the eye have been identified and discussed in some details. These obstacles consist of static membrane barriers to drug permeation into the eye, dynamic barriers such as the lacrimal drainage and physiochemical barriers such as low thermodynamic activity. It is explained how and why these obstacles hamper drug permeation and how different technologies, both those that are applied in marketed drug products and those that are under investigation, have addressed these obstacles. EXPERT OPINION The reason that most topical drug delivery systems have failed to deliver therapeutic drug concentrations to the retina is that they do not address physiochemical barriers such as the thermodynamic activity of the permeating drug molecules. Topical drug delivery to the retina has only been successful when the static, dynamic, and physiochemical barriers are addressed simultaneously.
Collapse
Affiliation(s)
- Thorsteinn Loftsson
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
40
|
Yan R, Xu L, Wang Q, Wu Z, Zhang H, Gan L. Cyclosporine A Nanosuspensions for Ophthalmic Delivery: A Comparative Study between Cationic Nanoparticles and Drug-Core Mucus Penetrating Nanoparticles. Mol Pharm 2021; 18:4290-4298. [PMID: 34731571 DOI: 10.1021/acs.molpharmaceut.1c00370] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The effect of mucin on ocular bioavailability depends on the extent to which it acts as a barrier or retention site. Mucus penetrating particles (MPPs) can evade the mucus entrapment and associated rapid clearance, but cationic nanoparticles have high adhesion to the mucosa. Both formulations can prolong the drug residence time on the surface of the eyes. The purpose of this work is to compare the effects of mucoadhesion of cationic nanoparticles and mucous permeability of MPPs on ocular bioavailability. Cationic nanosuspensions and drug-core MPP nanosuspensions were developed using the anti-solvent precipitation method. The results of X-ray diffraction revealed that CsA was amorphous. In vitro mucoadhesion evaluation demonstrated that cationic nanosuspensions enhanced the interaction with pig mucin about 5.0-6.0 fold compared to drug-core MPP nanosuspensions. A mucus permeation study by the transwell diffusion system showed that the Papp values of drug-core MPP nanosuspensions were 5.0-10.0 times higher than those of cationic nanosuspensions. In vivo ocular bioavailability evaluation of those CsA formulations was conducted in rabbits using a conventional nanosuspension as a comparison. The CsA concentrations in the cornea following the administration of a cationic nanosuspension and a drug-core MPP nanosuspension were 13,641.10 ng/g and 11,436.07 ng/g, respectively, significantly higher than that of the conventional nanosuspension (8310.762 ng/g). The results showed that both the cationic and MPP nanosuspensions were able to deliver CsA to anterior ocular tissues in effective therapeutic concentrations (10-20 μg/g) with topical drop instillation. The cationic nanosuspension could achieve relatively higher bioavailability than the MPP nanosuspension. The cationic nanosuspension would be a promising ocular drug delivery system.
Collapse
Affiliation(s)
- Rong Yan
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Lai Xu
- Shanghai InnoStar Bio-tech Company Ltd., Shanghai 200120, China
| | - Qiuhe Wang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zheng Wu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Hua Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Li Gan
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
41
|
Characterisation of Gel-Forming Mucins Produced In Vivo and In Ex Vivo Conjunctival Explant Cultures. Int J Mol Sci 2021; 22:ijms221910528. [PMID: 34638869 PMCID: PMC8508887 DOI: 10.3390/ijms221910528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/26/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
One key element to the health of the ocular surface encompasses the presence of gel-forming mucins in the pre-ocular tear film. Conjunctival goblet cells are specialized epithelial cells that secrete mucins necessary for tear film stability and general homeostasis. Their dysfunction can be linked to a range of ocular surface inflammation disorders and chronic injuries. To obtain new perspectives and angles to tackle mucin deficiency, the need for an accurate evaluation of their presence and corresponding mucin secretion in ex vivo conjunctival cultures has become a requisite. In vitro, goblet cells show a significant decrease in the production and secretion of gel-forming mucins, accompanied by signs of dedifferentiation or transdifferentiation. Explant cultures on laminin-treated CLP-PEG hydrogels can, however, support the production of gel-forming mucins. Together, we challenge the current paradigm to evaluate the presence of cultured goblet cells solely based on their general mucin (MUC) content through imaging analyses, showing the need for additional techniques to assess the functionality of goblet cells. In addition, we broadened the gel-forming mucin profile of in vivo goblet cells with MUC5B and MUC6, while MUC2 and MUC6 is added to the profile of cultured goblet cells.
Collapse
|
42
|
Josyula A, Omiadze R, Parikh K, Kanvinde P, Appell MB, Patel P, Saeed H, Sutar Y, Anders N, He P, McDonnell PJ, Hanes J, Date AA, Ensign LM. An ion-paired moxifloxacin nanosuspension eye drop provides improved prevention and treatment of ocular infection. Bioeng Transl Med 2021; 6:e10238. [PMID: 34589607 PMCID: PMC8459599 DOI: 10.1002/btm2.10238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/08/2021] [Accepted: 06/13/2021] [Indexed: 12/28/2022] Open
Abstract
There are numerous barriers to achieving effective intraocular drug administration, including the mucus layer protecting the ocular surface. For this reason, antibiotic eye drops must be used multiple times per day to prevent and treat ocular infections. Frequent eye drop use is inconvenient for patients, and lack of adherence to prescribed dosing regimens limits treatment efficacy and contributes to antibiotic resistance. Here, we describe an ion-pairing approach used to create an insoluble moxifloxacin-pamoate (MOX-PAM) complex for formulation into mucus-penetrating nanosuspension eye drops (MOX-PAM NS). The MOX-PAM NS provided a significant increase in ocular drug absorption, as measured by the area under the curve in cornea tissue and aqueous humor, compared to Vigamox in healthy rats. Prophylactic and treatment efficacy were evaluated in a rat model of ocular Staphylococcus aureus infection. A single drop of MOX-PAM NS was more effective than Vigamox, and completely prevented infection. Once a day dosing with MOX-PAM NS was similar, if not more effective, than three times a day dosing with Vigamox for treating S. aureus infection. The MOX-PAM NS provided increased intraocular antibiotic absorption and improved prevention and treatment of ocular keratitis, and the formulation approach is highly translational and clinically relevant.
Collapse
Affiliation(s)
- Aditya Josyula
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Revaz Omiadze
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kunal Parikh
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Center for Bioengineering Innovation and DesignJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Pranjali Kanvinde
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Matthew B. Appell
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Pratikkumar Patel
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of PharmacyUniversity of Hawaii HiloHawaiiUSA
| | - Hiwa Saeed
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of PharmacyUniversity of Hawaii HiloHawaiiUSA
| | - Yogesh Sutar
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of PharmacyUniversity of Hawaii HiloHawaiiUSA
| | - Nicole Anders
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins UniversityBaltimoreMarylandUSA
| | - Ping He
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins UniversityBaltimoreMarylandUSA
| | - Peter J. McDonnell
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Justin Hanes
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins UniversityBaltimoreMarylandUSA
- Department of Environmental Health SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Abhijit A. Date
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of PharmacyUniversity of Hawaii HiloHawaiiUSA
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of MedicineUniversity of Hawaii ManoaHonoluluHawaiiUSA
| | - Laura M. Ensign
- The Center for Nanomedicine, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Ophthalmology, The Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins UniversityBaltimoreMarylandUSA
- Department of Gynecology and Obstetrics and Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
43
|
Design of ophthalmic micelles loaded with diclofenac sodium: effect of chitosan and temperature on the block-copolymer micellization behaviour. Drug Deliv Transl Res 2021; 12:1488-1507. [PMID: 34258717 DOI: 10.1007/s13346-021-01030-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2021] [Indexed: 10/20/2022]
Abstract
Diclofenac sodium 0.1% is a commonly used NSAID with well-documented clinical efficacy in reducing postoperative inflammation; however, its corneal tolerability and ophthalmic tissue bioavailability require further improvement. Advanced micellar delivery systems composed of block-copolymers and chitosan showing fine balance between the mucoadhesion and mucus permeation, capable to slip through the mucus barrier and adhere to the epithelial ocular surface, may be used to tackle both challenges. The aggregation behaviour of the block-copolymers in the presence of different additives will dramatically influence the quality attributes like particle size, particle size distribution, drug-polymer interaction, zeta potential, drug incorporation, important for the delicate balance among mucoadhesion and permeation, as well as safety and efficacy of the ophthalmic micelles. Therefore, quality by design approach and D-optimal experimental design model were used to create a pool of useful data for the influence of chitosan and the formulation factors on the block copolymer's aggregation behaviour during the development and optimization of Diclofenac loaded Chitosan/Lutrol F127 or F68 micelles. Particle size, polydispersity index, dissolution rate, FTIR and DSC studies, NMR spectroscopy, cytotoxicity, mucoadhesivity, mucus permeation studies, and bioadhesivity were assessed as critical quality attributes. FTIR and DSC studies pointed to the chaotropic effect of chitosan during the micelle aggregation. Mainly, Pluronic F68 micellization behaviour was more dramatically affected by the presence of chitosan, and self-aggregation into larger micelles with high polydispersity index was favoured at higher chitosan concentration. The optimized formulation with highest potential for ophthalmic delivery of diclofenac sodium, good cytotoxicity profile, delicate balance of the mucoadhesivity, and mucus permeation was in the design space of Chitosan/Lutrol F127 micelles.
Collapse
|
44
|
Beckman KA, Katz JA, Majmudar PA, Rips AG, Vaidya NS, Rostov AT. KPI-121 1% for pain and inflammation in ocular surgery. Pain Manag 2021; 12:17-23. [PMID: 34164994 DOI: 10.2217/pmt-2021-0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pain and inflammation are common experiences following ocular surgery and, if uncontrolled, can compromise patients' vision, functioning and quality of life. Corticosteroid drugs are available to manage inflammation and discomfort but have limitations in penetrating the ocular mucus barrier to reach the target ocular tissues. KPI-121 1% (INVELTYS®) is a novel formulation of loteprednol etabonate that employs innovative proprietary technology to deliver nanoparticle-sized mucus-penetrating particles to the cornea. Results from clinical trials demonstrate that KPI-121 1% is effective and well tolerated. KPI-121 1% uses mucopenetrative technology for ophthalmic use and is the only US FDA-approved twice-daily ocular corticosteroid indicated for the treatment of inflammation and pain after ocular surgery.
Collapse
Affiliation(s)
- Kenneth A Beckman
- Comprehensive Eyecare of Central Ohio, Westerville, OH 43082, USA.,Department of Ophthalmology, Ohio State University, Columbus, OH 43201, USA
| | - James A Katz
- The Midwest Center for Sight, Des Plaines, IL 60016, USA
| | | | | | - Neel S Vaidya
- Chicago Cornea Consultants, Highland Park, IL 60035, USA
| | | |
Collapse
|
45
|
Gupta PK, Venkateswaran N. The role of KPI-121 0.25% in the treatment of dry eye disease: penetrating the mucus barrier to treat periodic flares. Ther Adv Ophthalmol 2021; 13:25158414211012797. [PMID: 34017938 PMCID: PMC8114292 DOI: 10.1177/25158414211012797] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/07/2021] [Indexed: 11/28/2022] Open
Abstract
The tear film, which includes mucins that adhere to foreign particles, rapidly clears allergens and pathogens from the ocular surface, protecting the underlying tissues. However, the tear film’s ability to efficiently remove foreign particles during blinking can also pose challenges for topical drug delivery, as traditional eye drops (solutions and suspensions) are cleared from the ocular surface before the drug can penetrate into the conjunctival and corneal epithelium. In the past 15 years, there has been an increase in the development of nanoparticles with specialized coatings that have reduced affinity to mucins and are small enough in size to pass through the mucus barrier. These mucus-penetrating particles (MPPs) have been shown to efficiently penetrate the mucus barrier and reach the ocular surface tissues. Dry eye disease (DED) is a common inflammatory ocular surface disorder that often presents with periodic flares (exacerbations). However, currently approved immunomodulatory treatments for DED are intended for long-term use. Thus, there is a need for effective short-term treatments that can address intermittent flares of DED. Loteprednol etabonate, an ocular corticosteroid, was engineered to break down rapidly after administration to the ocular surface tissues and thereby reduce risks associated with other topical steroids. KPI-121 is an ophthalmic suspension that uses the MPP technology to deliver loteprednol etabonate more efficiently to the ocular tissues, achieving in animal models a 3.6-fold greater penetration of loteprednol etabonate to the cornea than traditional loteprednol etabonate ophthalmic suspensions. In clinical trials, short-term treatment with KPI-121 0.25% significantly reduced signs and symptoms of DED compared with its vehicle (placebo). Recently approved KPI-121 0.25%, with its novel drug delivery design and ease of use, has the potential to effectively treat periodic flares of DED experienced by many patients.
Collapse
Affiliation(s)
- Preeya K Gupta
- Department of Ophthalmology, Duke University Eye Center, 4709 Creekstone Drive, Suite 100, Durham, NC 27703, USA
| | - Nandini Venkateswaran
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Zierden HC, Josyula A, Shapiro RL, Hsueh H, Hanes J, Ensign LM. Avoiding a Sticky Situation: Bypassing the Mucus Barrier for Improved Local Drug Delivery. Trends Mol Med 2021; 27:436-450. [PMID: 33414070 PMCID: PMC8087626 DOI: 10.1016/j.molmed.2020.12.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
The efficacy of drugs administered by traditional routes is limited by numerous biological barriers that preclude reaching the intended site of action. Further, full body systemic exposure leads to dose-limiting, off-target side effects. Topical formulations may provide more efficacious drug and nucleic acid delivery for diseases and conditions affecting mucosal tissues, but the mucus protecting our epithelial surfaces is a formidable barrier. Here, we describe recent advances in mucus-penetrating approaches for drug and nucleic acid delivery to the ocular surface, the female reproductive tract, the gastrointestinal tract, and the airways.
Collapse
Affiliation(s)
- Hannah C. Zierden
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Aditya Josyula
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Rachel L. Shapiro
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Henry Hsueh
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD 21287,Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Laura M. Ensign
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD 21287,Departments Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287,Correspondence: (L.M. Ensign)
| |
Collapse
|
47
|
Challenges and strategies for the delivery of biologics to the cornea. J Control Release 2021; 333:560-578. [PMID: 33857565 DOI: 10.1016/j.jconrel.2021.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/05/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023]
Abstract
Biologics, like peptides, proteins and nucleic acids, have proven to be promising drugs for the treatment of numerous diseases. However, besides the off label use of the monoclonal antibody bevacizumab for the treatment of corneal neovascularization, to date no other biologics for corneal diseases have reached the market. Indeed, delivering biologics in the eye remains a challenge, especially at the level of the cornea. While it appears to be a rather accessible tissue for the administration of drugs, the cornea in fact presents several anatomical barriers to delivery. In addition, also intracellular delivery barriers need to be overcome to achieve a promising therapeutic outcome with biologics. This review outlines efforts that have been reported to successfully deliver biologics into the cornea. Biochemical and physical methods for achieving delivery of biologics in the cornea are discussed, with a critical view on their efficacy in overcoming corneal barriers.
Collapse
|
48
|
Patient-centric design for peptide delivery: Trends in routes of administration and advancement in drug delivery technologies. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2020.100079] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
49
|
Sripetch S, Loftsson T. Topical drug delivery to the posterior segment of the eye: Thermodynamic considerations. Int J Pharm 2021; 597:120332. [PMID: 33540025 DOI: 10.1016/j.ijpharm.2021.120332] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022]
Abstract
Almost all studies on non-invasive topical drug delivery to the eye have emphasized the importance of biological barriers, static membrane barriers such as the cornea and the conjunctiva/sclera and dynamic barriers such as the lacrimal drainage. Hardly any have discussed the importance of the thermodynamic activity of the permeating drug molecules. Most drugs permeate from the eye surface into the eye by passive diffusion where, according to Fick's first law, the drug concentration gradient over the various permeation barriers (e.g., the tear fluid and the lipophilic membrane barriers) is the driving force. At the barrier interphases the dissolved drug molecules must partition from one barrier to another. For example, at the tear-cornea interphase the drug molecules must partition from the aqueous exterior into the lipophilic membrane. The drug partition coefficient between two phases is commonly defined as the equilibrium concentration ratio. However, these are only approximations. The actual driving force in Fick's first law is the gradient of the chemical potential and the equilibrium between two phases is attained when the chemical potential of the drug in one phase is equal to that in the other phase. Here the importance of thermodynamic considerations in topical drug delivery to the eye is reviewed.
Collapse
Affiliation(s)
- Suppakan Sripetch
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland
| | - Thorsteinn Loftsson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, IS-107 Reykjavik, Iceland.
| |
Collapse
|