1
|
Southward J, Liu F, Aspinall SR, Okwuosa TC. Exploring the potential of mucoadhesive buccal films in geriatric medicine. Drug Dev Ind Pharm 2025:1-21. [PMID: 39963906 DOI: 10.1080/03639045.2025.2467329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/14/2024] [Accepted: 02/10/2025] [Indexed: 03/08/2025]
Abstract
As the global demographic shifts toward an aging society, the geriatric patient population is steadily increasing. These patients often suffer from comorbidities and require numerous oral medications, which can be especially challenging for dysphagic geriatric patients. Mucoadhesive buccal films (MBFs) seem promising and could reduce pill burden, simplify administration, and enable individualized drug therapy. This review aims to explore the age-related changes in the oral cavity and their impact on MBF delivery, including potential strategies to overcome these age-related barriers to drug delivery. It was observed that aging impacts the oral mucosa as well the properties of the saliva. There are several studies in the application of buccal films including the use of a wide range of permeation enhancers. The 3D printing of buccal films seems to introduce dosing flexibility to buccal film manufacturing.
Collapse
Affiliation(s)
- Jasmine Southward
- Department of Clinical, pharmaceutical and biological sciences, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, UK
| | - Fang Liu
- Department of Clinical, pharmaceutical and biological sciences, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, UK
| | - Sam R Aspinall
- Department of Clinical, pharmaceutical and biological sciences, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, UK
| | - Tochukwu C Okwuosa
- Department of Clinical, pharmaceutical and biological sciences, School of Life and Medical Sciences, University of Hertfordshire, College Lane, Hatfield, UK
| |
Collapse
|
2
|
Feng Z, Han C, Zhang N, Wang Y, Luo G, Gao X. An integrated strategy for deciphering the action mechanism of emplastrum: Prescription analysis- component identification- virtual screening and affinity testing in the case of Yaoshen Gao. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119369. [PMID: 39842747 DOI: 10.1016/j.jep.2025.119369] [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: 11/26/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Emplastrum has a long history of use in the clinical practice of traditional Chinese medicine (TCM), valued for its convenient external application and pronounced therapeutic effects. Traditionally, the emplastrum was composed of numerous herbal medicines. The elucidation of their mechanisms of action are of great importance. YaoShen Gao (YSG), as a traditional example of emplastrum, was composed of more than 20 medicinal herbs. Clinically, YSG has been used to treat benign prostatic hyperplasia (BPH). However, the active components and therapeutic targets of YSG remain unclear, requiring further investigation. AIM OF THE STUDY To establish an integrated strategy to uncover the mechanisms underlying the potential active ingredients and therapeutic targets in complex TCM emplastra, using YSG for BPH treatment as a case study. MATERIALS AND METHODS A BPH rat model was established via castration and testosterone propionate injections. The therapeutic efficacy of YSG was evaluated comprehensively through phenotypic, pathological, physiological, and biochemical analyses. Prescription analysis was conducted based on the principles of "monarch, minister, assistant, and courier," as well as clinical dosage and efficacy. Based on the strategy of representative compounds-single herbal medicine -YSG formulation, the chemical profile of YSG was performed using UPLC-Q Exactive Orbitrap- MS. Network pharmacology identified preliminary targets, while molecular docking and literature mining further narrowed these down. Finally, molecular dynamics simulations and Bio-Layer Interferometry (BLI) were used to validate binding affinities of active components to targets. RESULTS The efficacy indicators demonstrated that treatment of YSG significantly reduced prostate wet weight and prostate index in BPH rats, with notable improvements observed in glandular structure. Additionally, YSG inhibited the expression of inflammatory factors (TNF- α, IL- 8) and fibrosis-related proteins (VEGF, TGF-β). Twelve key herbal medicines were identified by prescription analysis from the 20 herbs in YSG, such as Cistanche deserticola, Epimedium sagittatum and so on. High-resolution mass spectrometry (HR-MS) characterized 125 chemical components, and Venn analysis identified 409 common targets between YSG components and BPH. Subsequently, GO and KEGG analyses revealed that these targets are predominantly involved in protein phosphorylation, cellular components, and key signaling pathways. Protein-protein interaction (PPI) analysis identified 10 key targets, suggesting that the therapeutic effects of YSG on BPH are mediated through 39 active compounds and 12 relevant signaling pathways. Molecular docking analysis identified 14 target-compound pairs, and literature supported their relevance in PI3K/AKT, VEGF, TNF, and TGF-β pathways. Molecular dynamics simulations and BLI further validated the strong interactions between representative target-small molecule pairs like AKT1 with bavachalcone (KD = 46.8 μM) and PIK3R1 with apigenin (KD = 47.9 μM). CONCLUSIONS A systematic strategy for identifying active ingredients and therapeutic targets in complex TCM emplastra was developed. YSG contains active components, including bavachalcone, apigenin, schisandrin C, liquiritigenin, 8-prenyldaidzein, estrone, isopimpinellin, 8-prenylkaempferol, which act on six key targets, such as AKT1 and PIK3R1, to regulate the AKT/PI3K, AGE-RAGE, AR, VEGF, TGF-β, TNF and others signaling pathways. These pathways further modulate cellular proliferation, fibrosis, inflammation, and angiogenesis, thereby effectively contributing to the treatment of BPH.
Collapse
Affiliation(s)
- ZhiYang Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Chen Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Na Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ying Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - XiaoYan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
3
|
Lykins WR, Pollet J, White JA, Keegan B, Versteeg L, Strych U, Chen WH, Mohamath R, Ramer-Denisoff G, Reed S, Renshaw C, Beaver S, Gerhardt A, Voigt EA, Tomai MA, Sitrin R, Choy RKM, Cassels FJ, Hotez PJ, Bottazzi ME, Fox CB. Optimizing immunogenicity and product presentation of a SARS-CoV-2 subunit vaccine composition: effects of delivery route, heterologous regimens with self-amplifying RNA vaccines, and lyophilization. Front Immunol 2024; 15:1480976. [PMID: 39737197 PMCID: PMC11683073 DOI: 10.3389/fimmu.2024.1480976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Dozens of vaccines have been approved or authorized internationally in response to the ongoing SARS-CoV-2 pandemic, covering a range of modalities and routes of delivery. For example, mucosal delivery of vaccines via the intranasal (i.n.) route has been shown to improve protective mucosal responses in comparison to intramuscular (i.m.) delivery. As we gain knowledge of the limitations of existing vaccines, it is of interest to understand if changes in product presentation or combinations of multiple vaccine modalities can further improve immunological outcomes. Methods We investigated a commercial-stage SARS-CoV-2 receptor binding domain (RBD) antigen adjuvanted with a clinical-stage TLR-7/8 agonist (3M-052) formulated on aluminum oxyhydroxide (Alum). In a murine immunogenicity model, we compared i.n. and i.m. dosing of the RBD-3M-052-Alum vaccine. We measured the magnitude of antibody responses in serum and lungs, the antibody-secreting cell populations in bone marrow, and antigen-specific cytokine-secreting splenocyte populations. Similarly, we compared different heterologous and homologous prime-boost regimens using the RBD-3M-052-Alum vaccine and a clinical-stage self-amplifying RNA (saRNA) vaccine formulated on a nanostructured lipid carrier (NLC) using the i.m. route alone. Finally, we developed a lyophilized presentation of the RBD-3M-052-Alum vaccine and compared it to the liquid presentation and a heterologous regimen including a previously characterized lyophilized form of the saRNA-NLC vaccine. Results and discussion We demonstrate that i.n. dosing of the RBD-3M-052-Alum vaccine increased IgA titers in the lung by more than 1.5 logs, but induced serum IgG titers 0.8 logs lower, in comparison to i.m. dosing of the same vaccine. We also show that the homologous prime-boost RBD-3M-052-Alum regimen led to the highest serum IgG and bronchial IgA titers, whereas the homologous saRNA-NLC regimen led to the highest splenocyte interferon-γ response. We found that priming with the saRNA-NLC vaccine and boosting with the RBD-3M-052-Alum vaccine led to the most desirable immune outcome of all regimens tested. Finally, we show that the lyophilized RBD-3M-052-Alum vaccine retained its immunological characteristics. Our results demonstrate that the route of delivery and the use of heterologous regimens each separately impacts the resulting immune profile, and confirm that multi-product vaccine regimens can be developed with stabilized presentations in mind.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Female
- Immunogenicity, Vaccine
- Administration, Intranasal
- Freeze Drying
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Adjuvants, Vaccine
- mRNA Vaccines/immunology
- Mice, Inbred BALB C
- Adjuvants, Immunologic/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Injections, Intramuscular
- Humans
Collapse
Affiliation(s)
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | | | - Brian Keegan
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Leroy Versteeg
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Wen-Hsiang Chen
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Raodoh Mohamath
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | - Sierra Reed
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | - Samuel Beaver
- Access to Advanced Health Institute, Seattle, WA, United States
| | - Alana Gerhardt
- Access to Advanced Health Institute, Seattle, WA, United States
| | - Emily A. Voigt
- Access to Advanced Health Institute, Seattle, WA, United States
| | | | | | | | | | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
- Department of Biology, Baylor University, Waco, TX, United States
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, United States
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Christopher B. Fox
- Access to Advanced Health Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
4
|
Dong Z, Zhang L, Li G, Li Y, He H, Lu Y, Wu W, Qi J. Mechanism and performance of choline-based ionic liquids in enhancing nasal delivery of glucagon. J Control Release 2024; 375:812-828. [PMID: 39341285 DOI: 10.1016/j.jconrel.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Proteins and peptides have been increasingly developed as pharmaceuticals owing to their high potency and low side effects. However, their administration routes are confined to injections, such as intra-muscular and intra-venous injections, making patient compliance a challenge. Hence, non-injectable delivery systems are crucial to expanding the clinical use of proteins and peptides. In this context, two choline-based ionic liquids (ILs), namely, choline geranic acid ([Ch][Ger]) and choline citric acid ([Ch][Cit]), have been identified as promising agents for enhancing the permeation and prolonging the retention time of glucagon (GC) after intra-nasal administration. Notably, intra-nasal delivery of GC via ILs (GC/ILs) elicited rapid and smooth reversal of acute hypoglycaemia without leading to rebound hyperglycaemia in type 1 diabetic rats subjected to insulin induction. In addition, ILs could improve the transcellular transport of GC through electrostatic interaction. ILs could also transiently open inter-cellular tight junctions transiently to facilitate the paracellular transport of GC. Safety tests indicated that continuous intra-nasal delivery of ILs led to reversible changes, such as epithelial cell inflammation, goblet cell overgrowth, and impacts on the distribution of nasal cilia. However, these changes could be alleviated by the innate self-repair ability of mucosal epithelial cells. This study highlights the considerable potential of ILs for long-term nasal delivery of biomacromolecules.
Collapse
Affiliation(s)
- Zirong Dong
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Luyu Zhang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Guangyue Li
- College of Chemical Engineering, North China University of Science and Technology, Tangshan 063210, PR China
| | - Yang Li
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Haisheng He
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Yi Lu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Wei Wu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China
| | - Jianping Qi
- School of Pharmacy, Key Laboratory of Smart Drug Delivery of MOE, Fudan University, Shanghai 201203, China.
| |
Collapse
|
5
|
Salehi T, Raeisi Estabragh MA, Salarpour S, Ohadi M, Dehghannoudeh G. Absorption enhancer approach for protein delivery by various routes of administration: a rapid review. J Drug Target 2023; 31:950-961. [PMID: 37842966 DOI: 10.1080/1061186x.2023.2271680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
As bioactive molecules, peptides and proteins are essential in living organisms, including animals and humans. Defects in their function lead to various diseases in humans. Therefore, the use of proteins in treating multiple diseases, such as cancers and hepatitis, is increasing. There are different routes to administer proteins, which have limitations due to their large and hydrophilic structure. Another limitation is the presence of biological and lipophilic membranes that do not allow proteins to pass quickly. There are different strategies to increase the absorption of proteins from these biological membranes. One of these strategies is to use compounds as absorption enhancers. Absorption enhancers are compounds such as surfactants, phospholipids and cyclodextrins that increase protein passage through the biological membrane and their absorption by different mechanisms. This review focuses on using other absorption enhancers and their mechanism in protein administration routes.
Collapse
Affiliation(s)
- Toktam Salehi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Raeisi Estabragh
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Soodeh Salarpour
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mandana Ohadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Dehghannoudeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Maher S, Geoghegan C, Brayden DJ. Safety of surfactant excipients in oral drug formulations. Adv Drug Deliv Rev 2023; 202:115086. [PMID: 37739041 DOI: 10.1016/j.addr.2023.115086] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
Surfactants are a diverse group of compounds that share the capacity to adsorb at the boundary between distinct phases of matter. They are used as pharmaceutical excipients, food additives, emulsifiers in cosmetics, and as household/industrial detergents. This review outlines the interaction of surfactant-type excipients present in oral pharmaceutical dosage forms with the intestinal epithelium of the gastrointestinal (GI) tract. Many surfactants permitted for human consumption in oral products reduce intestinal epithelial cell viability in vitro and alter barrier integrity in epithelial cell monolayers, isolated GI tissue mucosae, and in animal models. This suggests a degree of mis-match for predicting safety issues in humans from such models. Recent controversial preclinical research also infers that some widely used emulsifiers used in oral products may be linked to ulcerative colitis, some metabolic disorders, and cancers. We review a wide range of surfactant excipients in oral dosage forms regarding their interactions with the GI tract. Safety data is reviewed across in vitro, ex vivo, pre-clinical animal, and human studies. The factors that may mitigate against some of the potentially abrasive effects of surfactants on GI epithelia observed in pre-clinical studies are summarised. We conclude with a perspective on the overall safety of surfactants in oral pharmaceutical dosage forms, which has relevance for delivery system development.
Collapse
Affiliation(s)
- Sam Maher
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland.
| | - Caroline Geoghegan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
7
|
Babu SR, Shekara HH, Sahoo AK, Harsha Vardhan PV, Thiruppathi N, Venkatesh MP. Intranasal nanoparticulate delivery systems for neurodegenerative disorders: a review. Ther Deliv 2023; 14:571-594. [PMID: 37691577 DOI: 10.4155/tde-2023-0019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Neurodegenerative diseases are a significant cause of mortality worldwide, and the blood-brain barrier (BBB) poses a significant challenge for drug delivery. An intranasal route is a prominent approach among the various methods to bypass the BBB. There are different pathways involved in intranasal drug delivery. The drawbacks of this method include mucociliary clearance, enzymatic degradation and poor drug permeation. Novel nanoformulations and intranasal drug-delivery devices offer promising solutions to overcome these challenges. Nanoformulations include polymeric nanoparticles, lipid-based nanoparticles, microspheres, liposomes and noisomes. Additionally, intranasal devices could be utilized to enhance drug-delivery efficacy. Therefore, intranasal drug-delivery systems show potential for treating neurodegenerative diseases through trigeminal or olfactory pathways, which can significantly improve patient outcomes.
Collapse
Affiliation(s)
- Someshbabu Ramesh Babu
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Harshith Hosahalli Shekara
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Ashish Kumar Sahoo
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Pyda Venkata Harsha Vardhan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Nitheesh Thiruppathi
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Madhugiri Prakash Venkatesh
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Faculty of Pharmaceutical Sciences, UCSI University, Kaula Lampur, Malaysia
| |
Collapse
|
8
|
Xu Y, Amakye WK, Xiao G, Liu X, Ren J, Wang M. Intestinal absorptivity-increasing effects of sodium N-[8-(2-hydroxybenzoyl)amino]-caprylate on food-derived bioactive peptide. Food Chem 2023; 401:134059. [PMID: 36095999 DOI: 10.1016/j.foodchem.2022.134059] [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: 06/06/2022] [Revised: 08/20/2022] [Accepted: 08/27/2022] [Indexed: 10/14/2022]
Abstract
Delivering bioactive peptides orally is hampered by poor absorption across the gastrointestinal barrier. Using the walnut-derived peptide PW5, PPKNW, we explored whether coformulation of peptides with absorption enhancer sodium N-[8-(2-hydroxybenzoyl)aminocaprylate] (SNAC) could improve the intestinal absorption of orally-administered bioactive peptides. Herein, the application of SNAC enhanced the absorption efficiency of PW5 in a non-everted gut sac model. Particle size distribution (1 027.8 ± 6.74 nm) and zeta potential (-2.89 ± 0.07 mV) of the PW5-SNAC complex were significantly greater than that of individual PW5 and SNAC. Scanning electron microscopy revealed that SNAC application could aggravate the surface roughness and reduce the compact structure of PW5. It further showed that PW5 and SNAC binds through an endothermic process underpinned by hydrogen bond and van der Waals forces and that SNAC could bound primarily to the internal calyx of PW5. These findings are helpful for the effective delivery of bioactive peptides.
Collapse
Affiliation(s)
- Yongzhao Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - William Kwame Amakye
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Ganhong Xiao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jiaoyan Ren
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Research Institute for Food Nutrition and Human Health, Guangzhou, China.
| | - Min Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
9
|
Abstract
The pentapeptide L-R5 has previously been shown to transiently increase the permeability of nasal epithelial cell layers in vitro, allowing paracellular transport of molecules of up to 4 kDa. Protein kinase C zeta (PKC ζ), a member of a family of serine/threonine kinases was shown to be involved in tight junction modulation induced by L-R5. We show here that the ability of L-R5 to modulate tight junctions is comparable to other permeability enhancers such as bilobalide, latrunculin A or C10. Interaction of the peptide with the target protein occurs via electrostatic interaction, with the presence of positive charges being essential for its functionality. L-R5 is myristoylated to allow quick cell entry and onset of activity. While no epithelial cytotoxicity was detected, the hydrophobic myristoyl rest was shown to cause haemolysis. Taken together, these data show that a structural optimization of L-R5 may be possible, both from a toxicological and an efficacy point of view.
Collapse
Affiliation(s)
- Joël Brunner
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland,CONTACT Gerrit Borchard Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Switzerland
| |
Collapse
|
10
|
Fantini A, Giulio L, Delledonne A, Pescina S, Sissa C, Nicoli S, Santi P, Padula C. Buccal Permeation of Polysaccharide High Molecular Weight Compounds: Effect of Chemical Permeation Enhancers. Pharmaceutics 2022; 15:pharmaceutics15010129. [PMID: 36678758 PMCID: PMC9864332 DOI: 10.3390/pharmaceutics15010129] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 01/04/2023] Open
Abstract
The greatest achievement in the advanced drug delivery field should be the optimization of non-invasive formulations for the delivery of high molecular weight compounds. Peptides, proteins, and other macromolecules can have poor membrane permeation, principally due to their large molecular weight. The aim of this work was to explore the possibility of administering fluorescently labeled dextrans (molecular weight 4-150 kDa) across the buccal mucosa. Permeation experiments across pig esophageal mucosa were carried out using fatty acids and bile salts as penetration enhancers. The data obtained show that it is possible to increase or promote the mucosa permeation of high molecular weight dextrans by using caprylic acid or sodium taurocholate as the chemical enhancers. With these enhancers, dextrans with molecular weight of 70 and 150 kDa, that in passive conditions did not permeate, could cross the mucosa in detectable amounts. FD-70 and FD-150 showed comparable permeability values, despite the molecular weight difference. The results obtained in the present work suggest that the buccal administration of high molecular weight compounds is feasible.
Collapse
Affiliation(s)
- Adriana Fantini
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Luca Giulio
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Andrea Delledonne
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Silvia Pescina
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Cristina Sissa
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy
| | - Sara Nicoli
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Patrizia Santi
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Cristina Padula
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
- Correspondence: ; Tel.: +39-0521-905078
| |
Collapse
|
11
|
Human Lactobacillus Biosurfactants as Natural Excipients for Nasal drug Delivery of Hydrocortisone. Pharmaceutics 2022; 14:pharmaceutics14030524. [PMID: 35335901 PMCID: PMC8952429 DOI: 10.3390/pharmaceutics14030524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
The inclusion of a chemical permeation enhancer in a dosage form is considered an effective approach to improve absorption across the nasal mucosa. Herein we evaluated the possibility of exploiting biosurfactants (BS) produced by Lactobacillus gasseri BC9 as innovative natural excipients to improve nasal delivery of hydrocortisone (HC). BC9-BS ability to improve HC solubility and the BS mucoadhesive potential were investigated using the surfactant at a concentration below and above the critical micelle concentration (CMC). In vitro diffusion studies through the biomimetic membrane PermeaPad® and the same synthetic barrier functionalized with a mucin layer were assessed to determine BC9-BS absorption enhancing properties in the absence and presence of the mucus layer. Lastly, the diffusion study was performed across the sheep nasal mucosa using BC9-BS at a concentration below the CMC. Results showed that BC9-BS was able to interact with the main component of the nasal mucosa, and that it allowed for a greater solubilization and also permeation of the drug when it was employed at a low concentration. Overall, it seems that BC9-BS could be a promising alternative to chemical surfactants in the nasal drug delivery field.
Collapse
|
12
|
Rajamma SS, Krishnaswami V, Prabu SL, Kandasamy R. Geophila repens phytosome-loaded intranasal gel with improved nasal permeation for the effective treatment of Alzheimer's disease. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
13
|
Inulin-Based Polymeric Micelles Functionalized with Ocular Permeation Enhancers: Improvement of Dexamethasone Permeation/Penetration through Bovine Corneas. Pharmaceutics 2021; 13:pharmaceutics13091431. [PMID: 34575507 PMCID: PMC8472490 DOI: 10.3390/pharmaceutics13091431] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022] Open
Abstract
Ophthalmic drug delivery is still a challenge due to the protective barriers of the eye. A common strategy to promote drug absorption is the use of ocular permeation enhancers, while an innovative approach is the use of polymeric micelles. In the present work, the two mentioned approaches were coupled by conjugating ocular permeation enhancers (PEG2000, carnitine, creatine, taurine) to an inulin-based co-polymer (INU-EDA-RA) in order to obtain self-assembling biopolymers with permeation enhancer properties for the hydrophobic drug dexamethasone (DEX). Inulin derivatives were properly synthetized, were found to expose about 2% mol/mol of enhancer molecules in the side chain, and resulted able to self-assemble at various concentrations by varying the pH and the ionic strength of the medium. Moreover, the ability of polymeric micelles to load dexamethasone was demonstrated, and size, mucoadhesiveness, and cytocompatibility against HCE cells were evaluated. Furthermore, the efficacy of the permeation enhancer was evaluated by ex vivo permeation studies to determine the performance of the used enhancers, which resulted in PEG2000 > CAR > TAU > CRE, while entrapment ability studies resulted in CAR > TAU > PEG2000 > CRE, both for fluorescent-labelled and DEX-loaded micelles. Finally, an increase in terms of calculated Kp and Ac parameters was demonstrated, compared with the values calculated for DEX suspension.
Collapse
|
14
|
Koutsoviti M, Siamidi A, Pavlou P, Vlachou M. Recent Advances in the Excipients Used for Modified Ocular Drug Delivery. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4290. [PMID: 34361483 PMCID: PMC8347600 DOI: 10.3390/ma14154290] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/04/2022]
Abstract
In ocular drug delivery, maintaining an efficient concentration of the drug in the target area for a sufficient period of time is a challenging task. There is a pressing need for the development of effective strategies for drug delivery to the eye using recent advances in material sciences and novel approaches to drug delivery. This review summarizes the important aspects of ocular drug delivery and the factors affecting drug absorption in the eye including encapsulating excipients (chitosan, hyaluronic acid, poloxamer, PLGA, PVCL-PVA-PEG, cetalkonium chloride, and gelatin) for modified drug delivery.
Collapse
Affiliation(s)
- Melitini Koutsoviti
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (M.K.); (A.S.)
| | - Angeliki Siamidi
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (M.K.); (A.S.)
| | - Panagoula Pavlou
- Department of Biomedical Sciences, Division of Aesthetics and Cosmetic Science, University of West Attica, 28 Ag. Spyridonos Str., 12243 Egaleo, Greece;
| | - Marilena Vlachou
- Department of Pharmacy, Division of Pharmaceutical Technology, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; (M.K.); (A.S.)
| |
Collapse
|
15
|
Pires PC, Fazendeiro AC, Rodrigues M, Alves G, Santos AO. Nose-to-brain delivery of phenytoin and its hydrophilic prodrug fosphenytoin combined in a microemulsion - formulation development and in vivo pharmacokinetics. Eur J Pharm Sci 2021; 164:105918. [PMID: 34174414 DOI: 10.1016/j.ejps.2021.105918] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/25/2021] [Accepted: 06/20/2021] [Indexed: 11/16/2022]
Abstract
Phenytoin is a low aqueous solubility antiepileptic drug, but its phosphate ester prodrug fosphenytoin is soluble, although less permeable. In a previous study, the intranasal administration of aqueous-based formulations of fosphenytoin led to high but delayed phenytoin bioavailability compared to the intravenous route. In this work, we hypothesized that formulating an association of the prodrug fosphenytoin and the drug phenytoin (the active and diffusible form), could result in a faster and/or more effective brain targeting. Hence, nano or microemulsions containing both active drug and prodrug were developed and characterized regarding viscosity, osmolality, pH, mean size and in vitro drug release. Then, in vivo pharmacokinetics of a selected microemulsion containing fosphenytoin and phenytoin was evaluated in mice following intranasal administration and compared with a similar microemulsion containing fosphenytoin only. Both microemulsions led to higher brain drug levels at short time points than previously developed simpler aqueous based fosphenytoin formulations, likely due to the microemulsion's permeation enhancing effect. In addition, having a small amount of phenytoin in the formulation led to an equivalent maximum brain drug concentration and an overall higher absolute bioavailability, with a prolonged drug exposure. Hence, it can be concluded that if there is a need for a fast and prolonged therapeutic effect, a drug/phosphate ester prodrug combination in a microemulsion is ideal, but if a fast effect is all that is needed, having the prodrug alone could be enough, while considering a formulation with permeation enhancing components.
Collapse
Affiliation(s)
- Patrícia C Pires
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Ana C Fazendeiro
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Márcio Rodrigues
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Research Unit for Inland Development (UDI-IPG), Polytechnic Institute of Guarda, 6300-559 Guarda, Portugal.
| | - Gilberto Alves
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Adriana O Santos
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
16
|
Absorption-Enhancing Mechanisms of Capryol 90, a Novel Absorption Enhancer, for Improving the Intestinal Absorption of Poorly Absorbed Drugs: Contributions to Trans- or Para-Cellular Pathways. Pharm Res 2020; 37:248. [PMID: 33230672 DOI: 10.1007/s11095-020-02963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE We have previously reported that Capryol 90 improves the intestinal absorption of insulin, a peptide drug, without causing serious damage to the intestinal epithelium. However, the effects of Capryol 90 and its related formulations on the intestinal absorption of other drugs, and their absorption-enhancing mechanisms are still unclear. The aim of this study is to evaluate the effects of Capryol 90 and its related formulations on the intestinal absorption of drugs and elucidate their absorption-enhancing mechanisms. METHODS The intestinal absorption of 5(6)-carboxyfluorescein, fluorescein isothiocyanate-dextrans, and alendronate was evaluated using an in situ closed loop method. Brush border membrane vesicles (BBMVs) were labeled with fluorescent probes, and the fluidity of membrane was evaluated by a fluorescence depolarization method. The expression levels of tight junction (TJ) proteins were measured using a Western blot method and immunofluorescence staining. RESULTS Among the tested excipients, Capryol 90 significantly improved the small and large intestinal absorption of drugs. In mechanistic studies, Capryol 90 increased the membrane fluidity of lipid bilayers in BBMVs. Additionally, Capryol 90 decreased the expression levels of TJ-associated proteins, namely claudin-4, occludin, and ZO-1. CONCLUSIONS Capryol 90 is an effective absorption enhancer for improving the intestinal absorption of poorly absorbed drugs via both transcellular and paracellular pathways.
Collapse
|
17
|
Lam JKW, Cheung CCK, Chow MYT, Harrop E, Lapwood S, Barclay SIG, Wong ICK. Transmucosal drug administration as an alternative route in palliative and end-of-life care during the COVID-19 pandemic. Adv Drug Deliv Rev 2020; 160:234-243. [PMID: 33137363 PMCID: PMC7603972 DOI: 10.1016/j.addr.2020.10.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022]
Abstract
The Coronavirus disease 2019 (COVID-19) pandemic has led to a surge in need for alternative routes of administration of drugs for end of life and palliative care, particularly in community settings. Transmucosal routes include intranasal, buccal, sublingual and rectal. They are non-invasive routes for systemic drug delivery with the possibility of self-administration, or administration by family caregivers. In addition, their ability to offer rapid onset of action with reduced first-pass metabolism make them suitable for use in palliative and end-of-life care to provide fast relief of symptoms. This is particularly important in COVID-19, as patients can deteriorate rapidly. Despite the advantages, these routes of administration face challenges including a relatively small surface area for effective drug absorption, small volume of fluid for drug dissolution and the presence of a mucus barrier, thereby limiting the number of drugs that are suitable to be delivered through the transmucosal route. In this review, the merits, challenges and limitations of each of these transmucosal routes are discussed. The goals are to provide insights into using transmucosal drug delivery to bring about the best possible symptom management for patients at the end of life, and to inspire scientists to develop new delivery systems to provide effective symptom management for this group of patients.
Collapse
Affiliation(s)
- Jenny K W Lam
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region.
| | - Chucky C K Cheung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region; School of Pharmacy, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Michael Y T Chow
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region; Sydney Pharmacy School, Faculty of Medicine and Health, University of Sydney, Australia
| | - Emily Harrop
- Helen and Douglas House, Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Susie Lapwood
- Oxford University Hospitals NHS Foundation Trust, United Kingdom
| | - Stephen I G Barclay
- Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, United Kingdom
| | - Ian C K Wong
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region; Centre for Medicines Optimisation Research and Education (CMORE), Research Department of Practice and Policy, School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
18
|
Adapted nano-carriers for gastrointestinal defense components: surface strategies and challenges. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102277. [DOI: 10.1016/j.nano.2020.102277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/18/2020] [Accepted: 07/18/2020] [Indexed: 12/21/2022]
|
19
|
Donalisio M, Argenziano M, Rittà M, Bastiancich C, Civra A, Lembo D, Cavalli R. Acyclovir-loaded sulfobutyl ether-β-cyclodextrin decorated chitosan nanodroplets for the local treatment of HSV-2 infections. Int J Pharm 2020; 587:119676. [DOI: 10.1016/j.ijpharm.2020.119676] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/20/2022]
|
20
|
Detsi A, Kavetsou E, Kostopoulou I, Pitterou I, Pontillo ARN, Tzani A, Christodoulou P, Siliachli A, Zoumpoulakis P. Nanosystems for the Encapsulation of Natural Products: The Case of Chitosan Biopolymer as a Matrix. Pharmaceutics 2020; 12:E669. [PMID: 32708823 PMCID: PMC7407519 DOI: 10.3390/pharmaceutics12070669] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/12/2022] Open
Abstract
Chitosan is a cationic natural polysaccharide, which has emerged as an increasingly interesting biomaterialover the past few years. It constitutes a novel perspective in drug delivery systems and nanocarriers' formulations due to its beneficial properties, including biocompatibility, biodegradability and low toxicity. The potentiality of chemical or enzymatic modifications of the biopolymer, as well as its complementary use with other polymers, further attract the scientific community, offering improved and combined properties in the final materials. As a result, chitosan has been extensively used as a matrix for the encapsulation of several valuable compounds. In this review article, the advantageous character of chitosan as a matrix for nanosystemsis presented, focusing on the encapsulation of natural products. A five-year literature review is attempted covering the use of chitosan and modified chitosan as matrices and coatings for the encapsulation of natural extracts, essential oils or pure naturally occurring bioactive compounds are discussed.
Collapse
Affiliation(s)
- Anastasia Detsi
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Eleni Kavetsou
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Ioanna Kostopoulou
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Ioanna Pitterou
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Antonella Rozaria Nefeli Pontillo
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Andromachi Tzani
- Department of Chemical Sciences, Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Heroon Polytechniou 9, Zografou Campus, 15780 Athens, Greece; (E.K.); (I.K.); (I.P.); (A.R.N.P.); (A.T.)
| | - Paris Christodoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, Vassileos Constantinou Ave. 48, 116 35 Athens, Greece; (P.C.); (A.S.)
| | - Aristeia Siliachli
- Institute of Chemical Biology, National Hellenic Research Foundation, Vassileos Constantinou Ave. 48, 116 35 Athens, Greece; (P.C.); (A.S.)
- Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece
| | - Panagiotis Zoumpoulakis
- Institute of Chemical Biology, National Hellenic Research Foundation, Vassileos Constantinou Ave. 48, 116 35 Athens, Greece; (P.C.); (A.S.)
- Department of Food Science and Technology, Universisty of West Attica, Ag. Spyridonos Str., Egaleo, 12243 Athens, Greece
| |
Collapse
|
21
|
Prediction of the enhanced insulin absorption across a triple co-cultured intestinal model using mucus penetrating PLGA nanoparticles. Int J Pharm 2020; 585:119516. [DOI: 10.1016/j.ijpharm.2020.119516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/25/2020] [Accepted: 06/05/2020] [Indexed: 01/26/2023]
|
22
|
Knerr PJ, Mowery SA, Finan B, Perez-Tilve D, Tschöp MH, DiMarchi RD. Selection and progression of unimolecular agonists at the GIP, GLP-1, and glucagon receptors as drug candidates. Peptides 2020; 125:170225. [PMID: 31786282 DOI: 10.1016/j.peptides.2019.170225] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023]
Abstract
The continued global growth in the prevalence of obesity coupled with the limited number of efficacious and safe treatment options elevates the importance of innovative pharmaceutical approaches. Combinatorial strategies that harness the metabolic benefits of multiple hormonal mechanisms have emerged at the preclinical and more recently clinical stages of drug development. A priority has been anti-obesity unimolecular peptides that function as balanced, high potency poly-agonists at two or all the cellular receptors for the endocrine hormones glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon. This report reviews recent progress in this area, with emphasis on what the initial clinical results demonstrate and what remains to be addressed.
Collapse
Affiliation(s)
- Patrick J Knerr
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | | | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - Diego Perez-Tilve
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Garching, Germany
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA.
| |
Collapse
|